Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Shock ; 45(1): 33-9, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26679472

ABSTRACT

Hemorrhagic shock resulting from blood loss directs the majority of the blood to the vital organs, dramatically reducing blood flow to the intestines and resulting in damage and inflammation. The excessive intestinal inflammatory response includes pro-inflammatory cytokines and complement activation, although the mechanism is not clear. Toll-like receptors play a vital role in the innate immune response and toll-like receptor 2 (TLR2) is required for intestinal ischemia/reperfusion-induced injury. We hypothesized that TLR2 plays an integral role in the intestinal inflammatory response after hemorrhage and subjected C57Bl/6 wild-type and Tlr2(-/-) mice to atraumatic loss of ∼30% total blood volume. Two hours after blood removal, the intestinal injury and inflammation were assessed. We demonstrate that compared with wild-type control mice, Tlr2(-/-) mice sustain less intestinal damage and inflammation. Importantly, TLR2 regulated eicosanoid and complement activation and IL-12 and TNFα secretions, indicating interactions between TLR2 and complement in response to significant blood loss.


Subject(s)
Complement System Proteins/immunology , Inflammation Mediators/immunology , Inflammation/immunology , Shock, Hemorrhagic/immunology , Toll-Like Receptor 2/immunology , Animals , Complement Activation/immunology , Inflammation/etiology , Inflammation/pathology , Intestines/blood supply , Intestines/pathology , Male , Mice, Inbred C57BL , Mice, Knockout , Regional Blood Flow , Shock, Hemorrhagic/complications , Toll-Like Receptor 2/deficiency
2.
J Leukoc Biol ; 98(5): 791-804, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26216936

ABSTRACT

Multiple pathologic conditions, including hemorrhage, tumor angiogenesis, and ischemia-reperfusion events, will result in hypoxia and subsequent reperfusion. Previous studies have analyzed the lipid changes within whole tissues and indicated that ischemia-reperfusion altered tissue and cellular phospholipids. Using an in vitro cell culture model of hypoxia and reoxygenation, we examined the endothelial lipid changes. We hypothesized that phospholipid scramblase 1, a protein that regulates bilayer asymmetry, is involved in altering the phospholipids of endothelial cells during hypoxia, a component of ischemia, leading to ß2-glycoprotein I and IgM binding and subsequent lipid-mediated, inflammatory responses. We have completed the first comprehensive study of steady-state phospholipid scramblase 1 mRNA levels, protein expression, and activity under conditions of hypoxia and reoxygenation. Phospholipid scramblase 1 regulates phosphatidylserine exposure in response to oxygen stress, leading to ß2-glycoprotein I and IgM binding and lipid-mediated, inflammatory responses.


Subject(s)
Endothelium, Vascular/metabolism , Phospholipid Transfer Proteins/metabolism , Reperfusion Injury/metabolism , Vasculitis/metabolism , beta 2-Glycoprotein I/metabolism , Animals , Cell Hypoxia , Cell Line , Endothelium, Vascular/pathology , Immunoglobulin M/genetics , Immunoglobulin M/metabolism , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Mice , Phospholipid Transfer Proteins/genetics , Reperfusion Injury/genetics , Reperfusion Injury/pathology , Vasculitis/genetics , Vasculitis/pathology , beta 2-Glycoprotein I/genetics
3.
J Immunol ; 194(3): 1190-8, 2015 Feb 01.
Article in English | MEDLINE | ID: mdl-25539820

ABSTRACT

In multiple clinical conditions, including trauma and hemorrhage, reperfusion magnifies ischemic tissue damage. Ischemia induces expression of multiple neoantigens, including lipid alterations that are recognized by the serum protein, ß2-glycoprotein I (ß2-GPI). During reperfusion, binding of ß2-GPI by naturally occurring Abs results in an excessive inflammatory response that may lead to death. As ß2-GPI is critical for intestinal ischemia/reperfusion (IR)-induced tissue damage and TLR2 is one of the proposed receptors for ß2-GPI, we hypothesized that IR-induced intestinal damage and inflammation require TLR2. Using TLR2(-/-) mice, we demonstrate that TLR2 is required for IR-induced mucosal damage, as well as complement activation and proinflammatory cytokine production. In response to IR, TLR2(-/-) mice have increased serum ß2-GPI compared with wild-type mice, but ß2-GPI is not deposited on ischemic intestinal tissue. In addition, TLR2(-/-) mice also did not express other novel Ags, suggesting a sequential response. Unlike other TLRs, TLR2(-/-) mice lacked the appropriate Ab repertoire to induce intestinal IR tissue damage or inflammation. Together, these data suggest that, in addition to the inflammatory response, IR-induced injury requires TLR2 for naturally occurring Ab production.


Subject(s)
Antibodies/immunology , Inflammation/immunology , Inflammation/metabolism , Intestinal Mucosa/metabolism , Intestines/immunology , Reperfusion Injury/immunology , Reperfusion Injury/metabolism , Toll-Like Receptor 2/metabolism , Animals , Antibodies/blood , Complement System Proteins/immunology , Cytokines/biosynthesis , Disease Models, Animal , Eicosanoids/biosynthesis , Immunoglobulin M/blood , Immunoglobulin M/immunology , Inflammation/genetics , Inflammation/pathology , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Intestines/pathology , Mice , Mice, Knockout , Reperfusion Injury/genetics , Toll-Like Receptor 2/genetics , beta 2-Glycoprotein I/immunology , beta 2-Glycoprotein I/metabolism
4.
Am J Clin Exp Immunol ; 1(2): 124-135, 2012.
Article in English | MEDLINE | ID: mdl-23205322

ABSTRACT

The mortality rate due to intestinal ischemia/reperfusion (IR) remains at 60-80%. As toll-like receptor (TLR) 4 has been shown to be critical for IR injury in several organs, including the intestine, and TLR9 is necessary for IR-induced damage of the liver, we investigated the hypothesis that TLR9 is involved in intestinal IR-induced damage. Wildtype (C57Bl/6) and TLR9(-/-) mice were subjected to intestinal IR or Sham treatment. Several markers of damage and inflammation were assessed, including mucosal injury, eicosanoid production, cytokine secretion and complement deposition. Although IR-induced injury was not altered, PGE(2) production was decreased in TLR9(-/-) mice. Attenuated PGE(2) production was not due to differences in percentage of lipids or COX-2 transcription. The data indicate that TLR9 is not required for IR-induced injury or inflammation of the intestine.

5.
J Immunol ; 189(10): 5047-56, 2012 Nov 15.
Article in English | MEDLINE | ID: mdl-23034168

ABSTRACT

Intestinal ischemic events, which are followed by reperfusion, induce significant tissue damage and frequently result in multiple organ failure, with >70% mortality. Upon reperfusion, excessive inflammation leads to exacerbated tissue damage. Previous studies indicated that binding of the serum protein, ß2-glycoprotein I, to the endothelium initiates a cascade of inflammatory molecules that is required for damage. We hypothesized that peptides derived from the binding domain (domain V) of ß2-glycoprotein I would attenuate ischemia/reperfusion-induced damage and inflammation in a therapeutic manner. Using a mouse model of intestinal ischemia/reperfusion, we administered peptides either prior to ischemia or at clinically relevant time points during reperfusion and evaluated intestinal tissue damage and inflammation after 2 h of reperfusion. We demonstrate that multiple peptides attenuate injury and inflammation in a dose-dependent manner and, perhaps more significantly, are efficacious when administered up to 30 min after the onset of reperfusion. In addition, an all D-amino acid retro-inverso peptide was biologically active. Thus, the ß2-glycoprotein I-derived peptides attenuate injury and inflammation when administered in a therapeutic manner in intestinal ischemia/reperfusion injury.


Subject(s)
Intestines/immunology , Peptides/pharmacology , Reperfusion Injury/immunology , Reperfusion Injury/prevention & control , beta 2-Glycoprotein I/pharmacology , Animals , Disease Models, Animal , Endothelium, Vascular/immunology , Endothelium, Vascular/pathology , Inflammation/immunology , Inflammation/pathology , Inflammation/prevention & control , Intestines/pathology , Mice , Peptides/immunology , Protein Structure, Tertiary , Reperfusion Injury/pathology , Time Factors , beta 2-Glycoprotein I/immunology
6.
Mol Immunol ; 52(3-4): 207-16, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22750067

ABSTRACT

Intestinal ischemia-reperfusion (IR)-induced injury results from a complex cascade of inflammatory components. In the mouse model of intestinal IR, the serum protein, ß2-glycoprotein I (ß2-GPI) binds to the cell surface early in the cascade. The bound ß2-GPI undergoes a conformational change which exposes a neoantigen recognized by naturally occurring antibodies and initiates the complement cascade. We hypothesized that providing additional antigen with exogenous ß2-GPI would alter IR-induced tissue injury. Administration of human but not mouse ß2-GPI attenuated IR-induced tissue damage and prostaglandin E(2) production indicating a physiological difference between ß2-GPI isolated from the two species. To investigate whether structural features were responsible for this physiological difference, we compared the chemical, physical and biochemical properties of the two proteins. Despite possessing 76% amino acid identity and 86% sequence homology, we found that mouse ß2-GPI differs from the human protein in size, carbohydrate chain location, heterogeneity and secondary structural content. These data suggest that the structural differences result in mouse Ab recognition of soluble human but not mouse ß2-GPI and attenuated IR-induced injury. We conclude that caution should be exercised in interpreting results obtained by using human ß2-GPI in a mouse model.


Subject(s)
Inflammation/immunology , Intestines/immunology , Reperfusion Injury/immunology , beta 2-Glycoprotein I/chemistry , beta 2-Glycoprotein I/immunology , Amino Acid Sequence , Animals , Cell Membrane/metabolism , Dinoprostone/biosynthesis , Homeodomain Proteins/genetics , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Protein Structure, Secondary , Proteomics , Reperfusion Injury/drug therapy , Sequence Alignment , beta 2-Glycoprotein I/metabolism
7.
J Immunol ; 186(3): 1755-62, 2011 Feb 01.
Article in English | MEDLINE | ID: mdl-21187447

ABSTRACT

Intestinal ischemia-reperfusion (IR)-induced damage requires complement receptor 2 (CR2) for generation of the appropriate natural Ab repertoire. Pathogenic Abs recognize neoantigens on the ischemic tissue, activate complement, and induce intestinal damage. Because C3 cleavage products act as ligands for CR2, we hypothesized that CR2(hi) marginal zone B cells (MZBs) require C3 for generation of the pathogenic Abs. To explore the ability of splenic CR2(+) B cells to generate the damaging Ab repertoire, we adoptively transferred either MZBs or follicular B cells (FOBs) from C57BL/6 or Cr2(-/-) mice into Rag-1(-/-) mice. Adoptive transfer of wild type CR2(hi) MZBs but not CR2(lo) FOBs induced significant damage, C3 deposition, and inflammation in response to IR. In contrast, similarly treated Rag-1(-/-) mice reconstituted with either Cr2(-/-) MZB/B1 B cells (B1Bs) or FOBs lacked significant intestinal damage and displayed limited complement activation. To determine whether C3 cleavage products are critical in CR2-dependent Ab production, we evaluated the ability of the natural Ab repertoire of C3(-/-) mice to induce damage in response to IR. Infusion of C3(-/-) serum into Cr2(-/-) mice restored IR-induced tissue damage. Furthermore, Rag-1(-/-) mice sustained significant damage after infusion of Abs from C3(-/-) but not Cr2(-/-) mice. Finally, adoptive transfer of MZBs from C3(-/-) mice into Rag-1(-/-) mice resulted in significant tissue damage and inflammation. These data indicate that CR2 expression on MZBs is sufficient to induce the appropriate Abs required for IR-induced tissue damage and that C3 is not critical for generation of the pathogenic Abs.


Subject(s)
Autoantibodies/biosynthesis , B-Lymphocyte Subsets/immunology , Complement C3/physiology , Receptors, Complement 3d/physiology , Spleen/immunology , Adoptive Transfer , Animals , Autoantibodies/therapeutic use , B-Lymphocyte Subsets/pathology , B-Lymphocyte Subsets/transplantation , Cells, Cultured , Complement C3/deficiency , Homeodomain Proteins/genetics , Immunophenotyping , Intestinal Mucosa/blood supply , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Complement 3d/biosynthesis , Receptors, Complement 3d/deficiency , Reperfusion Injury/immunology , Reperfusion Injury/pathology , Reperfusion Injury/therapy , Spleen/metabolism , Spleen/pathology
8.
Exp Physiol ; 96(2): 104-13, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21056969

ABSTRACT

Ischaemia-reperfusion-induced intestinal injury requires both Toll-like receptor 4 (TLR4) signalling through myeloid differentiation primary response gene (88) (MyD88) and complement activation. As a common Gram-negative intestinal pathogen, Helicobacter hepaticus signals through TLR4 and upregulates the complement inhibitor, decay accelerating factor (DAF; CD55). Since ischaemia-reperfusion (IR) injury is complement dependent, we hypothesized that Helicobacter infection may alter IR-induced intestinal damage. Infection increased DAF transcription and subsequently decreased complement activation in response to IR without altering intestinal damage in wild-type mice. Ischaemia-reperfusion induced similar levels of DAF mRNA expression in uninfected wild-type, MyD88(-/-) or TIR-domain-containing adaptor-inducing interferon-ß (Trif)-deficient mice. However, during infection, IR-induced DAF transcription was significantly attenuated in Trif-deficient mice. Likewise, IR-induced intestinal damage, complement component 3 deposition and prostaglandin E(2) production were attenuated in Helicobacter-infected, Trif-deficient but not MyD88(-/-) mice. While infection attenuated IR-induced cytokine production in wild-type and MyD88(-/-) mice, there was no further decrease in Trif-deficient mice. These data indicate distinct roles for MyD88 and Trif in IR-induced inflammation and suggest that chronic, undetected infections, such as Helicobacter, alter the use of the adaptor proteins to induce damage.


Subject(s)
Adaptor Proteins, Vesicular Transport/metabolism , Helicobacter Infections/metabolism , Intestinal Diseases/metabolism , Myeloid Differentiation Factor 88/metabolism , Reperfusion Injury/metabolism , Signal Transduction , Animals , Mice , Mice, Inbred C57BL , Mice, Knockout
9.
J Immunol ; 185(10): 6168-78, 2010 Nov 15.
Article in English | MEDLINE | ID: mdl-20956350

ABSTRACT

Reperfusion of ischemic tissue induces significant tissue damage in multiple conditions, including myocardial infarctions, stroke, and transplantation. Although not as common, the mortality rate of mesenteric ischemia/reperfusion (IR) remains >70%. Although complement and naturally occurring Abs are known to mediate significant damage during IR, the target Ags are intracellular molecules. We investigated the role of the serum protein, ß2-glycoprotein I as an initiating Ag for Ab recognition and ß2-glycoprotein I (ß2-GPI) peptides as a therapeutic for mesenteric IR. The time course of ß2-GPI binding to the tissue indicated binding and complement activation within 15 min postreperfusion. Treatment of wild-type mice with peptides corresponding to the lipid binding domain V of ß2-GPI blocked intestinal injury and inflammation, including cellular influx and cytokine and eicosanoid production. The optimal therapeutic peptide (peptide 296) contained the lysine-rich region of domain V. In addition, damage and most inflammation were also blocked by peptide 305, which overlaps with peptide 296 but does not contain the lysine-rich, phospholipid-binding region. Importantly, peptide 296 retained efficacy after replacement of cysteine residues with serine. In addition, infusion of wild-type serum containing reduced levels of anti-ß2-GPI Abs into Rag-1(-/-) mice prevented IR-induced intestinal damage and inflammation. Taken together, these data suggest that the serum protein ß2-GPI initiates the IR-induced intestinal damage and inflammatory response and as such is a critical therapeutic target for IR-induced damage and inflammation.


Subject(s)
Inflammation/metabolism , Mesentery/metabolism , Reperfusion Injury/metabolism , beta 2-Glycoprotein I/metabolism , Animals , Immunohistochemistry , Immunoprecipitation , Inflammation/immunology , Intestinal Mucosa/metabolism , Mesentery/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Reperfusion Injury/pathology
10.
Mol Immunol ; 48(1-3): 356-64, 2010.
Article in English | MEDLINE | ID: mdl-20800895

ABSTRACT

Innate immune responses including TLR4 and complement activation are required for mesenteric ischemia/reperfusion (IR)-induced tissue damage. We examined the regulation of TLR4 and complement activation in a mouse model of intestinal IR. Intestinal IR-induced C3 deposition in a TLR4 dependent manner. In addition, in wild-type but not TLR4 deficient mice, IR significantly increased C3 and Factor B (FB) mRNA expression within the intestine. To further examine the role of TLR4 and complement, we administered the complement inhibitor, CR2-Crry, to target local complement activation in wild-type C57Bl/10, and TLR4 deficient B10/ScN mice. TLR4 deficient mice sustained less damage and inflammation after IR than wild-type mice, but administration of CR2-Crry did not further reduce tissue damage. In contrast, CR2-Crry treatment of wild-type mice was accompanied by a reduction in complement activation and in C3 and FB transcription in response to IR. CR2-Crry also significantly decreased intestinal IL-6 and IL-12p40 production in both the wild-type and TLR4 deficient mice. These data indicate that TLR4 regulates extrahepatic complement production while complement regulates TLR4-mediated cytokine production during intestinal IR.


Subject(s)
Complement Activation/immunology , Inflammation/immunology , Intestines/immunology , Reperfusion Injury/immunology , Toll-Like Receptor 4/immunology , Animals , Enzyme-Linked Immunosorbent Assay , Inflammation/metabolism , Intestinal Mucosa/metabolism , Intestines/injuries , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Reperfusion Injury/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Toll-Like Receptor 4/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...