Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
Add more filters










Publication year range
1.
Am J Transplant ; 13(9): 2483-6, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23919410

ABSTRACT

Strongyloides stercoralis hyperinfection in an immunocompromised host has a high mortality rate but may initially present with nonspecific pulmonary and gastrointestinal symptoms. Donor-derived S. stercoralis by kidney transplantation is an uncommon diagnosis and difficult to prove. We report two renal allograft recipients on different immunosuppressive maintenance regimens that developed strongyloidiasis after transplantation from the same donor. Recipient 1 presented with a small bowel obstruction. Larvae were demonstrated on a duodenal biopsy and isolated from gastric, pulmonary, and stool samples. Serologic testing for S. stercoralis was negative at a referral laboratory but positive at the Centers for Disease Control. The patient's hospital course was complicated by a hyperinfection syndrome requiring subcutaneous ivermectin due to malabsorption. Recipient 1 survived but the allograft failed. Recipient 2 had larvae detected in stool samples after complaints of diarrhea and was treated. On retrospective testing for S. stercoralis, pretransplant serum collected from the donor and Recipient 1 was positive and negative, respectively. Donor-derived strongyloidiasis by renal transplantation is a preventable disease that may be affected by the immunosuppressive maintenance regimen. Subcutaneous ivermectin is an option in the setting of malabsorption. Finally, routine screening for S. stercoralis infection in donors from endemic areas may prevent future complications.


Subject(s)
Immunocompromised Host , Kidney Transplantation/adverse effects , Strongyloidiasis/transmission , Adult , Albendazole/therapeutic use , Animals , Ganciclovir/analogs & derivatives , Ganciclovir/therapeutic use , Humans , Ivermectin/administration & dosage , Ivermectin/therapeutic use , Malabsorption Syndromes , Male , Middle Aged , Strongyloides stercoralis , Strongyloidiasis/diagnosis , Strongyloidiasis/drug therapy , Tissue Donors , Transplantation, Homologous/adverse effects , Valganciclovir
2.
Mucosal Immunol ; 5(1): 19-29, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21937985

ABSTRACT

Neisseria gonorrhoeae is a common bacterial sexually transmitted infection. Like all Gram-negative bacteria, the outer membrane of the gonococcus is rich in endotoxin, a known ligand for Toll-like receptor (TLR)4. However, the role of endotoxin and that of its cognate receptor TLR4 in the mucosal response to acute gonococcal infection in the genital tract of women is unclear. To test this, we examined the course of infection after vaginal inoculation of N. gonorrhoeae in mice carrying the Lps(d) mutation in Tlr4, which renders them unresponsive to endotoxin. Although there was no difference in the duration of colonization, Lps(d) mice had a significantly higher peak bacterial burden which coincided with a massive polymorphonuclear cell influx and concomitant upregulation of a subset of inflammatory cytokine and chemokine markers. Notably, infected Lps(d) mice showed a decrease in interleukin-17, suggesting that Th17 responses are more dependent on TLR4 signaling in vivo. Defective polymorphonuclear cell-mediated and complement-independent serum killing of gonococci in Lps(d) mice was also observed and may account for the increased bacterial burden. This is the first in vivo evidence that TLR4-regulated factors modulate early inflammatory responses to gonococcal infection in the female reproductive tract and control bacterial replication.


Subject(s)
Gonorrhea/immunology , Neisseria gonorrhoeae/physiology , Th17 Cells/metabolism , Toll-Like Receptor 4/metabolism , Animals , Cell Movement/genetics , Cell Movement/immunology , Cells, Cultured , Cytokines/genetics , Cytokines/metabolism , Cytotoxicity, Immunologic/genetics , Cytotoxicity, Immunologic/immunology , Disease Models, Animal , Female , Gonorrhea/genetics , Humans , Inflammation Mediators/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Mutant Strains , Mutation/genetics , Neisseria gonorrhoeae/pathogenicity , Signal Transduction/genetics , Signal Transduction/immunology , Th17 Cells/immunology , Th17 Cells/pathology , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/immunology , Up-Regulation
3.
J Synchrotron Radiat ; 8(Pt 2): 311-3, 2001 Mar 01.
Article in English | MEDLINE | ID: mdl-11512763

ABSTRACT

XAFS and x-ray diffraction (XRD) are complementary structure determination techniques. The combination of XAFS and XRD can be used to determine the complete crystal structure when diffraction can not be refined. This is often the case at high pressures or high temperatures where there is limited access to the samples and energy dispersive x-ray diffraction is used. A new method to determine the atomic positions within the unit cell using EXAFS data with the programs RUNFIT and MKFIT is described. These programs systematically produce and test models for the XAFS data that are consistent with the diffraction results. The programs were written to solve the structure of two intermediate high pressure phases of AgCl, and are distributed with a working example.


Subject(s)
Software , Spectrometry, X-Ray Emission/methods , X-Ray Diffraction/methods , Crystallization , Molecular Structure
4.
J Immunol ; 167(4): 2257-67, 2001 Aug 15.
Article in English | MEDLINE | ID: mdl-11490013

ABSTRACT

Down-regulation of cell surface expression of Toll-like receptor (TLR) 4 following LPS stimulation has been suggested to underlie endotoxin tolerance. In this study, we examined whether overexpression of TLR2 or TLR4 would affect the ability of cells to become tolerant to LPS or the mycobacterial components, arabinose-capped lipoarabinomannan (LAM) and soluble tuberculosis factor (STF). To this end, Chinese hamster ovary/CD14 cells stably transfected with a NF-kappaB-dependent reporter construct, endothelial leukocyte adhesion molecule CD25 (the 3E10 clone), were engineered to overexpress either human TLR2 or TLR4. Transfected TLRs exhibited proper signaling functions, as evidenced by increased LPS responsiveness of 3E10/TLR4 cells and acquisition of sensitivity to TLR2-specific ligands upon transfection of TLR2 into TLR2-negative 3E10 cells. Pretreatment of cells with LPS, LAM, or STF did not modulate TLR2 or TLR4 cell surface expression. Following LPS exposure, 3E10, 3E10/TLR2, and 3E10/TLR4 cells exhibited comparable decreases in LPS-mediated NF-kappaB activation and mitogen-activated protein (MAP) kinase phosphorylation. Likewise, LPS pretreatment profoundly inhibited LPS-induced NF-kappaB translocation in Chinese hamster ovary cells that concomitantly overexpressed human TLR4 and myeloid differentiation protein-2 (MD-2), but failed to modulate TLR4 or MD-2 cell surface expression. Pretreatment of 3E10/TLR2 cells with LAM or STF decreased their NF-kappaB responses induced by subsequent stimulation with these substances or LPS. Conversely, prior exposure of 3E10/TLR2 cells to LPS led to hyporesponsiveness to LPS, LAM, and STF, indicating that LPS and mycobacterial products induce cross-tolerance. Thus, tolerance to LPS and mycobacterial components cannot be attributed solely to a decrease in TLR/MD-2 expression levels, suggesting inhibition of expression or function of other signaling intermediates.


Subject(s)
Antigens, Bacterial/immunology , CHO Cells/immunology , Drosophila Proteins , Immune Tolerance , Lipopolysaccharide Receptors/biosynthesis , Lipopolysaccharides/immunology , Membrane Glycoproteins/biosynthesis , Mycobacterium tuberculosis/immunology , Receptors, Cell Surface/biosynthesis , Animals , Antigens, Surface/biosynthesis , Antigens, Surface/genetics , CHO Cells/metabolism , Cell Line , Clone Cells , Cricetinae , Enzyme Activation/genetics , Enzyme Activation/immunology , Humans , Immune Tolerance/genetics , Lipopolysaccharide Receptors/genetics , Lipopolysaccharides/antagonists & inhibitors , Lymphocyte Antigen 96 , Membrane Glycoproteins/genetics , Membrane Proteins/biosynthesis , Membrane Proteins/genetics , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/antagonists & inhibitors , NF-kappa B/metabolism , Receptors, Cell Surface/genetics , Toll-Like Receptor 2 , Toll-Like Receptor 4 , Toll-Like Receptors , Transfection
5.
Infect Immun ; 69(4): 2230-6, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11254578

ABSTRACT

The recent isolation of a lipopolysaccharide (LPS)-deficient mutant of Neisseria meningitidis has allowed us to explore the roles of other gram-negative cell wall components in the host response to infection. The experiments in this study were designed to examine the ability of this mutant strain to activate cells. Although it was clearly less potent than the parental strain, we found the LPS-deficient mutant to be a capable inducer of the inflammatory response in monocytic cells, inducing a response similar to that seen with Staphylococcus aureus. Cellular activation by the LPS mutant was related to expression of CD14, a high-affinity receptor for LPS and other microbial products, as well as Toll-like receptor 2, a member of the Toll family of receptors recently implicated in host responses to gram-positive bacteria. In contrast to the parental strain, the synthetic LPS antagonist E5564 did not inhibit the LPS-deficient mutant. We conclude that even in the absence of LPS, the gram-negative cell wall remains a potent inflammatory stimulant, utilizing signaling pathways independent of those involved in LPS signaling.


Subject(s)
Bacterial Proteins/physiology , Drosophila Proteins , Lipid A/analogs & derivatives , Lipopolysaccharides/toxicity , Membrane Glycoproteins/physiology , Membrane Proteins/physiology , Neisseria meningitidis/immunology , Receptors, Cell Surface/physiology , Animals , CHO Cells , Cell Wall/physiology , Cricetinae , Humans , Lipid A/pharmacology , Lipopolysaccharide Receptors/physiology , Mutation , Toll-Like Receptor 2 , Toll-Like Receptors , Tumor Necrosis Factor-alpha/metabolism
6.
Infect Immun ; 68(12): 6770-6, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11083794

ABSTRACT

Lipopolysaccharide (LPS) and related bacterial products can be recognized by host inflammatory cells in a particulate, bacterium-bound form, as well as in various soluble, released forms. In the present study we have compared the mechanisms used by LPS, detoxified LPS (DLPS), and mannuronic acid polymers (M-polymers), in solution or covalently linked to particles, in stimulating monocytes to tumor necrosis factor (TNF) production. The addition of recombinant LPS binding protein (LBP) and/or soluble CD14 (sCD14) enhanced the production of TNF from monocytes stimulated with soluble LPS, DLPS, or M-polymer, but did not affect the response to M-polymer or DLPS attached to particles. Treatment of monocytes with antibody to CD14, CD18, or CD11b showed that CD14, but not CR3 (CD11b/CD18), mediated monocyte TNF production in response to the soluble antigens. In contrast, anti-CD14, anti-CD11b and anti-CD18 monoclonal antibodies all inhibited the response to the particulate stimuli. On the other hand, B975, a synthetic analog of Rhodobacter capsulatus lipid A, completely abrogated the monocyte TNF response induced by LPS but did not affect the TNF induction by DLPS or M-polymer, either in soluble or particulate forms. These data demonstrate that the engagement of immune receptors by bacterial products such as LPS, DLPS, and M-polymer is dependent upon the presentation form of their constituent carbohydrates, and that factors such as aggregation state, acylation, carbohydrate chain length, and solid versus liquid phase of bacterial ligands influence the mechanisms used by cells in mediating proinflammatory responses.


Subject(s)
Acute-Phase Proteins , Alginates/pharmacology , CD18 Antigens/physiology , Lipopolysaccharide Receptors/physiology , Lipopolysaccharides/pharmacology , Membrane Glycoproteins , Animals , CHO Cells , Carrier Proteins/physiology , Cricetinae , Glucuronic Acid , Hexuronic Acids , Macrophage-1 Antigen/physiology , Monocytes/drug effects , Tumor Necrosis Factor-alpha/biosynthesis
7.
J Immunol ; 165(8): 4272-80, 2000 Oct 15.
Article in English | MEDLINE | ID: mdl-11035061

ABSTRACT

Gram-negative bacteria and the LPS constituent of their outer membranes stimulate the release of inflammatory mediators believed to be responsible for the clinical manifestations of septic shock. The GPI-linked membrane protein, CD14, initiates the signaling cascade responsible for the induction of this inflammatory response by LPS. In this paper, we report the generation and characterization of CD14-null mice in which the entire coding region of CD14 was deleted. As expected, LPS failed to elicit TNF-alpha and IL-6 production in macrophages taken from these animals, and this loss in responsiveness is associated with impaired activation of both the NF-kappaB and the c-Jun N-terminal mitogen-activated protein kinase pathways. The binding and uptake of heat-killed Escherichia coli, measured by FACS analysis, did not differ between CD14-null and wild-type macrophages. However, in contrast to the findings with LPS, whole E. coli stimulated similar levels of TNF-alpha release from CD14-null and wild-type macrophages at a dose of 10 bioparticles per cell. This effect was dose dependent, and at lower bacterial concentrations CD14-deficient macrophages produced significantly less TNF-alpha than wild type. Approximately half of this CD14-independent response appeared to be mediated by CD11b/CD18, as demonstrated by receptor blockade using neutrophil inhibitory factor. An inhibitor of phagocytosis, cytochalasin B, abrogated the induction of TNF-alpha in CD14-deficient macrophages by E. coli. These data indicate that CD14 is essential for macrophage responses to free LPS, whereas other receptors, including CD11b/CD18, can compensate for the loss of CD14 in response to whole bacteria.


Subject(s)
Escherichia coli/immunology , Gene Deletion , Lipopolysaccharide Receptors/genetics , Lipopolysaccharides/immunology , Macrophages, Peritoneal/immunology , Macrophages, Peritoneal/microbiology , Animals , Bacterial Adhesion/genetics , Bacterial Adhesion/immunology , Bacterial Outer Membrane Proteins/physiology , CD18 Antigens/physiology , Cell Line , Cytokines/biosynthesis , Escherichia coli/physiology , Female , Lipopolysaccharide Receptors/blood , Macrophage Activation/genetics , Macrophage-1 Antigen/physiology , Macrophages, Peritoneal/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutagenesis, Insertional , Phagocytosis/immunology , Signal Transduction/immunology
8.
J Clin Invest ; 105(4): 497-504, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10683379

ABSTRACT

Lipopolysaccharide (LPS) is the main inducer of shock and death in Gram-negative sepsis. Recent evidence suggests that LPS-induced signal transduction begins with CD14-mediated activation of 1 or more Toll-like receptors (TLRs). The lipid A analogues lipid IVa and Rhodobacter sphaeroides lipid A (RSLA) exhibit an uncommon species-specific pharmacology. Both compounds inhibit the effects of LPS in human cells but display LPS-mimetic activity in hamster cells. We transfected human TLR4 or human TLR2 into hamster fibroblasts to determine if either of these LPS signal transducers is responsible for the species-specific pharmacology. RSLA and lipid IVa strongly induced NF-kappaB activity and IL-6 release in Chinese hamster ovary fibroblasts expressing CD14 (CHO/CD14), but these compounds antagonized LPS antagonists in CHO/CD14 fibroblasts that overexpressed human TLR4. No such antagonism occurred in cells overexpressing human TLR2. We cloned TLR4 from hamster macrophages and found that human THP-1 cells expressing the hamster TLR4 responded to lipid IVa as an LPS mimetic, as if they were hamster in origin. Hence, cells heterologously overexpressing TLR4 from different species acquired a pharmacological phenotype with respect to recognition of lipid A substructures that corresponded to the species from which the TLR4 transgene originated. These data suggest that TLR4 is the central lipid A-recognition protein in the LPS receptor complex.


Subject(s)
Drosophila Proteins , Lipopolysaccharide Receptors/metabolism , Lipopolysaccharides/metabolism , Membrane Glycoproteins/metabolism , Receptors, Cell Surface/metabolism , Animals , CHO Cells , Cricetinae , Glycolipids/metabolism , Humans , Ligands , Lipid A/analogs & derivatives , Lipid A/metabolism , Lipopolysaccharide Receptors/genetics , Lipopolysaccharides/antagonists & inhibitors , Macrophages/cytology , Macrophages/metabolism , Membrane Glycoproteins/genetics , Molecular Mimicry , Receptors, Cell Surface/genetics , Recombinant Proteins/metabolism , Rhodobacter sphaeroides , Signal Transduction , Species Specificity , Toll-Like Receptor 2 , Toll-Like Receptor 4 , Toll-Like Receptors
9.
J Endotoxin Res ; 6(5): 411-5, 2000.
Article in English | MEDLINE | ID: mdl-11521065

ABSTRACT

The inflammatory response to bacterial infections plays an important role in the detection and elimination of invading micro-organisms. Various components of the bacterial cell wall are capable of activating this pro-inflammatory response. In the case of Gram-negative bacteria, lipopolysaccharide (LPS) is the dominant trigger, although other bacterial factors are also capable of activating this systemic inflammatory response. Recently, Toll-like receptors (TLRs) have been implicated in host responses to bacterial pathogens. Specifically, TLR4 mediates LPS responses while TLR2 plays a broader role in the recognition of a variety of bacteria and bacterial antigens. The experiments in this study were designed to examine the role of Gram-negative cell wall components, other than LPS, and their cellular receptors in the host response to infection using an LPS-deficient mutant of Neisseria meningitidis. Although less potent than the parental strain, we found the LPS-deficient mutant to be a capable inducer of the inflammatory response in a variety of cell types. Moreover, cellular activation by this mutant required expression of CD14 and TLR2.


Subject(s)
Drosophila Proteins , Lipopolysaccharides/immunology , Membrane Glycoproteins/immunology , Neisseria meningitidis/immunology , Receptors, Cell Surface/immunology , Animals , CHO Cells , Cell Wall/immunology , Cricetinae , Humans , In Vitro Techniques , Inflammation Mediators/immunology , Leukocytes, Mononuclear/immunology , Lipopolysaccharide Receptors/genetics , Membrane Glycoproteins/genetics , Mutation , Neisseria meningitidis/genetics , Receptors, Cell Surface/genetics , Toll-Like Receptor 2 , Toll-Like Receptor 4 , Toll-Like Receptors , Transfection
10.
J Biol Chem ; 274(47): 33419-25, 1999 Nov 19.
Article in English | MEDLINE | ID: mdl-10559223

ABSTRACT

Toll-like receptors (TLRs) 2 and 4 are signal transducers for lipopolysaccharide, the major proinflammatory constituent in the outer membrane of Gram-negative bacteria. We observed that membrane lipoproteins/lipopeptides from Borrelia burgdorferi, Treponema pallidum, and Mycoplasma fermentans activated cells heterologously expressing TLR2 but not those expressing TLR1 or TLR4. These TLR2-expressing cells were also stimulated by living motile B. burgdorferi, suggesting that TLR2 recognition of lipoproteins is relevant to natural Borrelia infection. Importantly, a TLR2 antibody inhibited bacterial lipoprotein/lipopeptide-induced tumor necrosis factor release from human peripheral blood mononuclear cells, and TLR2-null Chinese hamster macrophages were insensitive to lipoprotein/lipopeptide challenge. The data suggest a role for the native protein in cellular activation by these ligands. In addition, TLR2-dependent responses were seen using whole Mycobacterium avium and Staphylococcus aureus, demonstrating that this receptor can function as a signal transducer for a wide spectrum of bacterial products. We conclude that diverse pathogens activate cells through TLR2 and propose that this molecule is a central pattern recognition receptor in host immune responses to microbial invasion.


Subject(s)
Bacterial Proteins/metabolism , Drosophila Proteins , Lipoproteins/metabolism , Membrane Glycoproteins/metabolism , Receptors, Cell Surface/metabolism , Animals , Borrelia burgdorferi Group/metabolism , CHO Cells , Cricetinae , Humans , Mycobacterium avium/metabolism , Mycoplasma fermentans/metabolism , Protein Binding , Toll-Like Receptor 1 , Toll-Like Receptor 2 , Toll-Like Receptor 4 , Toll-Like Receptors , Treponema pallidum/metabolism
11.
J Immunol ; 163(1): 1-5, 1999 Jul 01.
Article in English | MEDLINE | ID: mdl-10384090

ABSTRACT

Invasive infection with Gram-positive and Gram-negative bacteria often results in septic shock and death. The basis for the earliest steps in innate immune response to Gram-positive bacterial infection is poorly understood. The LPS component of the Gram-negative bacterial cell wall appears to activate cells via CD14 and Toll-like receptor (TLR) 2 and TLR4. We hypothesized that Gram-positive bacteria might also be recognized by TLRs. Heterologous expression of human TLR2, but not TLR4, in fibroblasts conferred responsiveness to Staphylococcus aureus and Streptococcus pneumoniae as evidenced by inducible translocation of NF-kappaB. CD14 coexpression synergistically enhanced TLR2-mediated activation. To determine which components of Gram-positive cell walls activate Toll proteins, we tested a soluble preparation of peptidoglycan prepared from S. aureus. Soluble peptidoglycan substituted for whole organisms. These data suggest that the similarity of clinical response to invasive infection by Gram-positive and Gram-negative bacteria is due to bacterial recognition via similar TLRs.


Subject(s)
Drosophila Proteins , Gram-Positive Bacteria/immunology , Membrane Glycoproteins/immunology , Receptors, Cell Surface/immunology , Animals , Bacterial Proteins , CHO Cells , Cell Wall/immunology , Cell Wall/metabolism , Cricetinae , Drosophila melanogaster/immunology , Gram-Positive Bacteria/pathogenicity , Gram-Positive Bacterial Infections/immunology , Humans , Immunity, Innate , Lipopolysaccharides/immunology , Membrane Glycoproteins/biosynthesis , Membrane Glycoproteins/genetics , NF-kappa B/genetics , Peptidoglycan/immunology , Peptidoglycan/isolation & purification , Receptors, Cell Surface/biosynthesis , Receptors, Cell Surface/genetics , Recombinant Fusion Proteins/biosynthesis , Staphylococcus aureus/immunology , Streptococcus pneumoniae/immunology , Streptolysins/metabolism , Toll-Like Receptor 2 , Toll-Like Receptor 4 , Toll-Like Receptors , Transfection/immunology
12.
Infect Dis Clin North Am ; 13(2): 341-53, vii, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10340170

ABSTRACT

The ability of a host to sense invasion by a pathogenic organism, and to respond appropriately to control infection, is paramount to survival. To that end, an array of receptors and binding proteins has evolved as part of the innate immune system to detect Gram-negative bacteria. This article reviews the role of CD14, other LPS binding proteins, and the Toll family of receptors in the innate recognition of bacterial lipopolysaccharide.


Subject(s)
Drosophila Proteins , Lipopolysaccharide Receptors/physiology , Lipopolysaccharides/toxicity , Animals , CD11 Antigens/physiology , CD18 Antigens/physiology , Endotoxins/antagonists & inhibitors , Humans , Lipid A/pharmacology , Lipopolysaccharide Receptors/metabolism , Lipopolysaccharide Receptors/pharmacology , Lipopolysaccharides/antagonists & inhibitors , Membrane Glycoproteins/physiology , Mice , Mice, Knockout/genetics , Receptors, Cell Surface/physiology , Shock, Septic/etiology , Signal Transduction , Toll-Like Receptors
13.
J Biol Chem ; 274(20): 13993-8, 1999 May 14.
Article in English | MEDLINE | ID: mdl-10318811

ABSTRACT

The activation of phagocytes by lipopolysaccharide (LPS) has been implicated in the pathogenesis of Gram-negative sepsis. Although the interaction between CD14 and LPS is a key event in the signaling cascade, the molecular mechanism by which cellular activation occurs remains obscure. We hypothesized that the main function of CD14 was to bind LPS and transfer it to a second receptor, which then initiates the subsequent signal for cellular activation. Thus, surface binding of LPS to the cell membrane would be the critical step that CD14 carries out. To test this hypothesis, we examined the activity of two other proteins known to bind LPS, lipopolysaccharide-binding protein and bactericidal/permeability-increasing protein. We found that when these normally soluble proteins were expressed in Chinese hamster ovary-K1 fibroblasts as glycosylphosphatidylinositol-anchored proteins, both could substitute for CD14 in initiating LPS signaling. Pharmacological studies with synthetic lipid A analogues demonstrated that these surface expressed LPS-binding proteins had characteristics that were qualitatively identical to membrane CD14. These data support the hypothesis that a receptor distinct from CD14 functions as the actual signal transducer and suggest that surface binding of LPS to the cell membrane is the crucial first step for initiating downstream signaling events.


Subject(s)
Lipopolysaccharide Receptors/metabolism , Lipopolysaccharides/pharmacology , Membrane Proteins/metabolism , Signal Transduction , Amino Acid Sequence , Animals , Antimicrobial Cationic Peptides , Base Sequence , Blood Bactericidal Activity , Blood Proteins/metabolism , CD11 Antigens/metabolism , CHO Cells , Cricetinae , Fibroblasts/drug effects , Fibroblasts/metabolism , Gram-Negative Bacteria/metabolism , Humans , Molecular Sequence Data
14.
J Immunol ; 161(10): 5413-20, 1998 Nov 15.
Article in English | MEDLINE | ID: mdl-9820516

ABSTRACT

The activation of phagocytes by the lipid A moiety of LPS has been implicated in the pathogenesis of Gram-negative sepsis. While two LPS receptors, CD14 and CD11/CD18, have been associated with cell signaling, details of the LPS signal transduction cascade remain obscure. CD14, which exists as a GPI-anchored and a soluble protein, lacks cytoplasmic-signaling domains, suggesting that an ancillary molecule is required to activate cells. The CD11/CD18 integrins are transmembrane proteins. Like CD14, they are capable of mediating LPS-induced cellular activation when expressed on the surface of hamster fibroblasts Chinese hamster ovary (CHO)-K1. The observation that a cytoplasmic deletion mutant is still capable of activating transfected CHO-K1 argues that CD11/CD18 also utilizes an associated signal transducer. We sought to identify further similarities between the signaling systems utilized by CD14 and CD11/CD18. LPS-binding protein, which transfers LPS to CD14, enhanced both LPS-induced cellular activation and binding of Gram-negative bacteria in CD11/CD18-transfected CHO-K1, thus implying that LPS-binding protein can also transfer LPS to CD11/CD18. When synthetic lipid A analogues were analyzed for their ability to function as LPS agonists, or antagonists, in the CHO transfectants, we found the effects were identical regardless of which LPS receptor was expressed. This supports the hypothesis that a receptor distinct from CD14 and CD11/CD18 is responsible for discriminating between the lipid A of LPS and the LPS antagonists. We propose that this receptor, which is the target of the LPS antagonists, functions as the true signal transducer in LPS-induced cellular activation for both CD14 and CD11/CD18.


Subject(s)
Acute-Phase Proteins , CD11 Antigens/physiology , CD18 Antigens/physiology , Lipid A/physiology , Lipopolysaccharide Receptors/physiology , Membrane Glycoproteins , Signal Transduction/immunology , Animals , CD11 Antigens/genetics , CHO Cells , Carrier Proteins/physiology , Cell Line , Cricetinae , Gram-Negative Bacteria/immunology , Humans , Lipid A/analogs & derivatives , Lipopolysaccharides/antagonists & inhibitors , Lipopolysaccharides/chemistry , Lipopolysaccharides/metabolism , Lipopolysaccharides/pharmacology , Macrophage Activation/drug effects , Mice , Mycobacterium/immunology , Peptidoglycan/pharmacology , Transfection/drug effects , Transfection/immunology , Tumor Cells, Cultured
15.
J Immunol ; 161(6): 3001-9, 1998 Sep 15.
Article in English | MEDLINE | ID: mdl-9743364

ABSTRACT

Gram-negative bacterial LPS is a potent activator of inflammatory responses. The binding of LPS to CD14 initiates signal transduction; however, the molecular processes immediately following this event remain unclear. We engineered an LPS-inducible fibroblast reporter cell line to facilitate the use of molecular genetic techniques to study the LPS signaling pathway. A plasmid containing the human Tac Ag cDNA under transcriptional control of the human E selectin promoter was cotransfected into Chinese hamster ovary (CHO)-K1 cells together with a CD14 expression plasmid. A cell line was obtained, 3E10, which upregulated expression of Tac following stimulation with LPS. Pools of mutagenized cells were exposed to LPS and then labeled with anti-Tac mAb. Cells that failed to up-regulate Tac expression were enriched by flow cytometry. Thirty clonal mutant cell lines were identified that continued to express CD14 and bind LPS, but failed to express Tac or translocate nuclear factor-kappaB (NF-kappaB) following LPS exposure. TNF-alpha-treated mutant cells continued to express Tac and translocate NF-kappaB. An analysis of LPS-induced NF-kappaB activity in heterokaryons derived from polyethylene glycol-fused cell lines indicated that recessive mutations in genes encoding components of the LPS signaling pathway accounted for the signaling defects. To date, two complementation groups have been identified from 11 cell lines analyzed. These data demonstrate that the TNF-alpha signaling pathway diverges from the LPS pathway early in the signal-transduction cascade despite similarities in LPS- and TNF-alpha-induced responses. Identification of the genes affected in these mutant reporter cells should identify heretofore-elusive components of the LPS signaling cascade.


Subject(s)
Acute-Phase Proteins , Endotoxins/genetics , Genes, Reporter/immunology , Lipopolysaccharides/immunology , Membrane Glycoproteins , Mutagenesis/immunology , Signal Transduction/genetics , Tumor Necrosis Factor-alpha/genetics , Animals , Biological Transport/immunology , Blotting, Northern , CHO Cells , Carrier Proteins/physiology , Cell Culture Techniques/methods , Cell Fusion/genetics , Cell Fusion/immunology , Cell Line , Cell Separation , Cricetinae , Endotoxins/deficiency , Flow Cytometry , Genetic Complementation Test , Humans , Lipopolysaccharides/metabolism , Lipopolysaccharides/pharmacology , NF-kappa B/metabolism , Phenotype , Receptors, Interleukin-2/biosynthesis , Receptors, Interleukin-2/drug effects , Receptors, Interleukin-2/genetics , Signal Transduction/immunology , Transfection/immunology , Tumor Necrosis Factor-alpha/pharmacology
16.
J Immunol ; 160(9): 4535-42, 1998 May 01.
Article in English | MEDLINE | ID: mdl-9574560

ABSTRACT

This study was undertaken to evaluate the role of CD14 and complement receptors type 3 (CR3) and 4 (CR4) in mediating TNF release and NF-kappaB activation induced by LPS and cell wall preparations from group B streptococci type III (GBS). LPS and GBS caused TNF secretion from human monocytes in a CD14-dependent manner, and soluble CD14, LPS binding protein, or their combination potentiated both LPS- and GBS-induced activities. Blocking of either CD14 or CD18, the common beta-subunit of CR3 and CR4, decreased GBS-induced TNF release, while LPS-mediated TNF production was inhibited by anti-CD14 mAb only. Chinese hamster ovary cell transfectants (CHO) that express human CD14 (CHO/CD14) responded to both LPS and GBS with NF-kappaB translocation, which was inhibited by anti-CD14 mAb and enhanced by LPS binding protein. While LPS showed fast kinetics of NF-kappaB activation in CHO/CD14 cells, a slower NF-kappaB response was induced by GBS. LPS also activated NF-kappaB in CHO cells transfected with either human CR3 or CR4 cDNA, although responses were delayed and weaker than those of CHO/CD14 cells. In contrast to LPS, GBS failed to induce NF-kappaB in CHO/CR3 or CHO/CR4 cells. Both C3H/OuJ (Lps[n]) and C3H/HeJ (Lps[d]) mouse peritoneal macrophages responded to GBS with TNF production and NF-kappaB translocation, whereas LPS was active only in C3H/OuJ macrophages. Thus, LPS and GBS differentially involve CD14 and CR3 or CR4 for signaling NF-kappaB activation in CHO cells and TNF release in human monocytes, and engage a different set of receptors and/or intracellular signaling pathways in mouse macrophages.


Subject(s)
Lipopolysaccharide Receptors/immunology , Lipopolysaccharides/immunology , Macrophage-1 Antigen/immunology , NF-kappa B/immunology , Streptococcus agalactiae/immunology , Tumor Necrosis Factor-alpha/immunology , Animals , CHO Cells , Cell Wall/immunology , Cricetinae , Humans , Lipopolysaccharides/pharmacology , Mice , Signal Transduction/immunology
18.
J Immunol ; 159(1): 433-8, 1997 Jul 01.
Article in English | MEDLINE | ID: mdl-9200483

ABSTRACT

Ligand binding to integrins activates intracellular signaling pathways that coordinate and regulate a variety of cellular responses. There is evidence to suggest that the cytoplasmic tails play a key role in several of these signaling events. We sought to determine whether the beta2 integrin complement receptor type 3 (CR3; CD11b/CD18), a receptor for LPS, could initiate an intracellular signal in the absence of its cytoplasmic domains. Expression of full length CR3 in a Chinese hamster ovary-K1 fibroblast line enabled serum-independent translocation of nuclear factor-kappaB in response to binding LPS. Unexpectedly, a cell line expressing a mutated form of CR3 deficient in the cytoplasmic domains was also competent for transmitting a signal in response to LPS. In contrast, phagocytosis of whole Gram-negative bacteria and iC3b-coated erythrocytes took place only with a full length receptor. Thus, while full length CR3 is necessary for productive phagocytic signals, LPS activation does not require the cytoplasmic domains. CR3 may function to activate cells by presenting LPS to a downstream signal transducer.


Subject(s)
CD18 Antigens/metabolism , Lipopolysaccharides/pharmacology , Signal Transduction/drug effects , Amino Acid Sequence , Animals , CD18 Antigens/genetics , CHO Cells , Cricetinae , Humans , Lipopolysaccharides/metabolism , Molecular Sequence Data , Mutation
19.
Infect Immun ; 65(3): 931-5, 1997 Mar.
Article in English | MEDLINE | ID: mdl-9038299

ABSTRACT

Previously, we demonstrated that monoclonal antibodies (MAb) directed against any of the three defined complement receptors (CR) for the third component of complement (CR1, CR3, and CR4) profoundly inhibited the binding of serum-opsonized Cryptococcus neoformans to monocyte-derived macrophages. These studies suggested either that a synergistic interaction between multiple CR was required for optimal binding of C. neoformans or that the MAb were exerting nonspecific effects (such as receptor coassociation). In the present studies, we took a novel approach to dissecting out the contributions of individual receptors to binding of a microbial pathogen. Chinese hamster ovary (CHO) cells stably transfected with human CR1, CR3, or CR4 were challenged with serum-opsonized C. neoformans. We found that CHO cells transfected with any of the three receptors bound C. neoformans, with the avidity of binding to CR3 being the greatest followed in decreasing order by CR1 and CR4. Following binding of C. neoformans to transfected CHO cells, most organisms remained surface attached only, although for each receptor a significant percentage (18.5 to 27.3%) of C. neoformans was internalized. Both C. neoformans and sheep erythrocytes that were selectively opsonized with the fragments of the third component of complement, C3b and iC3b, were bound preferentially by CHO cells transfected with CR1 and CR3, respectively. These data establish CR1, CR3, and CR4 as receptors independently capable of binding C. neoformans opsonized with fragments of C3. Moreover, our study demonstrates the usefulness of transfected cell lines as a powerful tool for identifying the contribution of individual receptors to the binding of a microbial pathogen.


Subject(s)
Cryptococcus neoformans/immunology , Receptors, Complement/physiology , Animals , Antibodies, Monoclonal/immunology , CHO Cells , Cricetinae , Humans , Mice , Phagocytosis , Receptors, Complement/genetics , Transfection
20.
J Exp Med ; 186(12): 2051-6, 1997 Dec 15.
Article in English | MEDLINE | ID: mdl-9396775

ABSTRACT

Gram-negative bacterial lipopolysaccharide (LPS) stimulates phagocytic leukocytes by interacting with the cell surface protein CD14. Cellular responses to LPS are markedly potentiated by the LPS-binding protein (LBP), a lipid-transfer protein that binds LPS aggregates and transfers LPS monomers to CD14. LBP also transfers LPS to lipoproteins, thereby promoting the neutralization of LPS. LBP present in normal plasma has been shown to enhance the LPS responsiveness of cells in vitro. The role of LBP in promoting LPS responsiveness in vivo was tested in LBP-deficient mice produced by gene targeting in embryonic stem cells. Whole blood from LBP-deficient animals was 1,000-fold less responsive to LPS as assessed by the release of tumor necrosis factor (TNF)-alpha. Blood from gene-targeted mice was devoid of immunoreactive LBP, essentially incapable of transferring LPS to CD14 in vitro, and failed to support cellular responses to LPS. These activities were restored by the addition of exogenous recombinant murine LBP to the plasma. Despite these striking in vitro findings, no significant differences in TNF-alpha levels were observed in plasma from wild-type and LBP-deficient mice injected with LPS. These data suggest the presence of an LBP-independent mechanism for responding to LPS. These LBP knockout mice may provide a tool for discovering the nature of the presumed second mechanism for transferring LPS to responsive cells.


Subject(s)
Acute-Phase Proteins/genetics , Carrier Proteins/physiology , Gene Deletion , Lipopolysaccharides/pharmacology , Membrane Glycoproteins , Animals , Carrier Proteins/genetics , Chimera , In Vitro Techniques , Kidney/chemistry , Kidney/drug effects , Lipopolysaccharide Receptors/metabolism , Liver/chemistry , Liver/drug effects , Mice , Mice, Knockout , Mice, Mutant Strains , Tumor Necrosis Factor-alpha/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...