Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
PLoS One ; 13(8): e0202594, 2018.
Article in English | MEDLINE | ID: mdl-30133517

ABSTRACT

BACKGROUND AND OBJECTIVE: Complement activation as an early and important inflammatory process contributes to multiple organ dysfunction after trauma. We have recently shown that complement inhibition by decay-accelerating factor (DAF) protects brain from blast-overpressure (BOP)-induced damage. This study was conducted to determine the effect of DAF on acute lung injury induced by BOP exposure and to elucidate its possible mechanisms of action. METHODS: Anesthetized adult male Sprague-Daley rats were exposed to BOP (120 kPa) from a compressed air-driven shock tube. Rats were randomly assigned to three experimental groups: 1) Control (no BOP and no DAF treatment), 2) BOP (120 kPa BOP exposure), and 3) BOP followed by treatment with rhDAF (500µg/kg, i.v) at 30 minutes after blast. After a recovery period of 3, 24, or 48 hours, animals were euthanized followed by the collection of blood and tissues at each time point. Samples were subjected to the assessment of cytokines and histopathology as well as for the interaction of high-mobility-group box 1 (HMGB1) protein, NF-κB, receptor for advanced glycation end products (RAGE), C3a, and C3aR. RESULTS: BOP exposure significantly increased in the production of systemic pro- and anti-inflammatory cytokines, and obvious pathological changes as characterized by pulmonary edema, inflammation, endothelial damage and hemorrhage in the lungs. These alterations were ameliorated by early administration of rhDAF. The rhDAF treatment not only significantly reduced the expression levels of HMGB1, RAGE, NF-κB, C3a, and C3aR, but also reversed the interaction of C3a-C3aR and nuclear translocation of HMGB1 in the lungs. CONCLUSIONS: Our findings indicate that early administration of DAF efficiently inhibits systemic and local inflammation, and mitigates blast-induced lung injury. The underlying mechanism might be attributed to its potential modulation of C3a-C3aR-HMGB1-transcriptional factor axis. Therefore, complement and/or HMGB1 may be potential therapeutic targets in amelioration of acute lung injury after blast injury.


Subject(s)
Acute Lung Injury/drug therapy , Blast Injuries/drug therapy , CD55 Antigens/administration & dosage , HMGB1 Protein/genetics , Inflammation/drug therapy , Acute Lung Injury/genetics , Acute Lung Injury/physiopathology , Animals , Blast Injuries/genetics , Blast Injuries/pathology , Complement Activation/drug effects , Complement C3a/antagonists & inhibitors , Disease Models, Animal , Humans , Inflammation/genetics , Inflammation/physiopathology , Lung/drug effects , Lung/metabolism , Lung/physiopathology , NF-kappa B/genetics , Pressure/adverse effects , Rats , Rats, Sprague-Dawley
2.
Genet Mol Res ; 14(3): 7782-92, 2015 Jul 14.
Article in English | MEDLINE | ID: mdl-26214459

ABSTRACT

It is expensive to induce experimental autoimmune myasthenia gravis (EAMG) by active immunity, and difficult to obtain natural acetylcholine receptor (AChR). We sought a new method of inducing EAMG by immunizing rats with artificially synthesized AChR. The AChR mRNA in TE671 cells was extracted and reverse transcribed. The inclusion body was purified and protein concentration was determined, and the EAMG animal model was used for induction. The serum was extracted from rat blood. The antibody titer was determined using enzyme-linked immunosorbant assay (ELISA). The concentration of decay accelerating factor (DAF) in the rat serum was determined by ELISA, and the metabolism of serum rDAF was determined by western blot. We evaluated the inhibition of rDAF by determining the 50% complement hemolysis unit in the rat serum. The extracellular domain (ECD) nucleotide sequence clone produced by polymerase chain reaction was completely consistent with that in the human gene bank; it was induced by isopropyl ß-D-1-thiogalactopyranoside to express the protein after insertion into vector pET16b. Sodium dodecyl sulfate polyacrylamide gel electrophoresis demonstrated that the inclusion body protein was the exact target. The ECD protein was able to bind with mAb35 after dialysis and renaturation, which demonstrated protein activity. The soluble ECD protein was used to immunize rats and obtain the EAMG models. The inhibitory effect of the complement was unsatisfactory owing to high decay rate after rDAF injection into the EAMG models. It is easy to induce the EAMG model by obtaining the AChRTEα1 subunit ECD protein using the substitution method.


Subject(s)
CD55 Antigens/therapeutic use , Myasthenia Gravis, Autoimmune, Experimental/drug therapy , Neuroprotective Agents/therapeutic use , Animals , CD55 Antigens/administration & dosage , CD55 Antigens/blood , Disease Models, Animal , Electrophoresis, Polyacrylamide Gel , Enzyme-Linked Immunosorbent Assay , Female , Humans , Injections, Intravenous , Myasthenia Gravis, Autoimmune, Experimental/blood , Myasthenia Gravis, Autoimmune, Experimental/pathology , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/pharmacology , Protein Renaturation/drug effects , Protein Structure, Tertiary , Rats, Inbred Lew , Recombinant Proteins/metabolism , Solubility
3.
Muscle Nerve ; 49(5): 749-56, 2014 May.
Article in English | MEDLINE | ID: mdl-24037951

ABSTRACT

INTRODUCTION: The site of pathology in myasthenia gravis (MG) is the neuromuscular junction (NMJ). Our goal was to determine the ability to direct complement inhibition to the NMJ. METHODS: A single-chain antibody directed against the alpha subunit of the acetylcholine receptor was synthesized (scFv-35) and coupled to decay-accelerating factor (DAF, scFv-35-DAF). scFv-35-DAF was tested in a passive model of experimentally acquired MG. RESULTS: Administration of scFv-35-DAF to mice deficient in intrinsic complement inhibitors produced no weakness despite confirmation of its localization to the NMJ and no evidence of tissue destruction related to complement activation. Rats with experimentally acquired MG treated with scFV-35-DAF showed less weakness and a reduction of complement deposition. CONCLUSIONS: We demonstrate a method to effectively target a therapeutic agent to the NMJ.


Subject(s)
CD55 Antigens/administration & dosage , Drug Delivery Systems/methods , Myasthenia Gravis, Autoimmune, Experimental/drug therapy , Neuromuscular Junction , Receptors, Cholinergic , Single-Chain Antibodies/therapeutic use , Animals , Mice , Myasthenia Gravis, Autoimmune, Experimental/immunology , Rats , Single-Chain Antibodies/immunology
4.
Acta Neuropathol Commun ; 1: 52, 2013 Aug 16.
Article in English | MEDLINE | ID: mdl-24252631

ABSTRACT

BACKGROUND: Blast-induced neurotrauma (BINT) is the signature life threatening injury of current military casualties. Neuroinflammation is a key pathological occurrence of secondary injury contributing to brain damage after blast injury. We have recently demonstrated that blast-triggered complement activation and cytokine release are associated with BINT. Here, we evaluated if administration of the complement inhibitor recombinant human decay-accelerating factor (rhDAF) is beneficial on neuroinflammation and neurodegeneration in a rat model of moderate BINT. Administration of rhDAF after exposure to moderate blast overpressure (BOP, 120 kPa) mitigated brain injury characterized by neuronal degeneration. rhDAF treatment reduced complement hemolytic activity at 3 hours and tissue complement deposition at 3, 24, and 48 hours as well as systemic and local cytokine release at 24 hours post BOP. Furthermore, rhDAF protected blood-brain barrier (BBB) integrity and reduced cytotoxic edema. Interaction between complement cleavage component, C3a and C3a receptor and tau phosphorylation were also attenuated in rhDAF treated animals at 3 and 24 hours after BOP. These novel findings suggest early complement targeted inhibition as a new therapeutic strategy to decrease neuroinflammation and neurodegeneration after blast TBI. RESULT: Administration of rhDAF after exposure to moderate blast overpressure (BOP, 120 kPa) mitigated brain injury characterized by neuronal degeneration. rhDAF treatment reduced complement hemolytic activity at 3 hours and tissue complement deposition at 3, 24, and 48 hours as well as systemic and local cytokine release at 24 hours post BOP. Furthermore, rhDAF protected blood-brain barrier (BBB) integrity and reduced cytotoxic edema. Interaction between complement cleavage component, C3a and C3a receptor and tau phosphorylation were also attenuated in rhDAF treated animals at 3 and 24 hours after BOP. CONCLUSION: These novel findings suggest early complement targeted inhibition as a new therapeutic strategy to decrease neuroinflammation and neurodegeneration after blast TBI.


Subject(s)
Blast Injuries/complications , Brain Injuries/drug therapy , Brain Injuries/etiology , CD55 Antigens/administration & dosage , Neuroprotective Agents/administration & dosage , Animals , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/physiopathology , Brain Edema/drug therapy , Brain Edema/etiology , Brain Edema/physiopathology , Brain Injuries/physiopathology , Cytokines/metabolism , Disease Models, Animal , Humans , Male , Nerve Degeneration/drug therapy , Nerve Degeneration/etiology , Neuroimmunomodulation/drug effects , Pressure , Random Allocation , Rats, Sprague-Dawley , Time Factors , Treatment Outcome
5.
J Surg Res ; 167(2): e103-15, 2011 May 15.
Article in English | MEDLINE | ID: mdl-20189588

ABSTRACT

BACKGROUND: C-reactive protein (CRP) is an acute pro-inflammatory mediator that has been demonstrated to enhance ischemia/reperfusion (IR) injury by virtue of activating the complement system. CRP is able to interact with complement proteins such as C1q, complement factor H, and C4b-binding protein. Since complement activation is central in the expression of tissue injury following IR, we have investigated the effects of human decay-accelerating factor (DAF), a complement inhibitor, on CRP-potentiated complement activation and tissue injury in mice subjected to mesenteric IR. MATERIALS AND METHODS: Male C57B1/6 mice were allocated into eight groups: (1) Sham-operated group without IR injury; (2) CRP+Sham group; (3) IR group; (4) CRP+IR group; (5) DAF group; (6) CRP+DAF group; (7) IR+DAF group, and (8) CRP+IR+DAF group. Intestinal and lung injury, neutrophil infiltration, myeloperoxidase (MPO) expression, complement component deposition, and interleukin-6 (IL-6) production were assessed for each treatment group of mice. RESULTS: We report that administration of DAF significantly attenuates the CRP-enhanced intestinal injury as well as remote lung damages following acute mesenteric IR in mice, while DAF inhibits complement activation, suppresses neutrophil infiltration, and reduces IL-6 production. CONCLUSIONS: Our study suggests that inhibition complement activation with DAF may prove useful for the treatment of post-ischemic inflammatory injuries associated with an increased production of CRP.


Subject(s)
C-Reactive Protein/metabolism , CD55 Antigens/therapeutic use , Mesentery/metabolism , Mesentery/physiopathology , Reperfusion Injury/metabolism , Reperfusion Injury/prevention & control , Animals , CD55 Antigens/administration & dosage , CD55 Antigens/pharmacology , Complement C5a/metabolism , Complement Membrane Attack Complex/metabolism , Injections, Intravenous , Interleukin-6/metabolism , Male , Mesentery/blood supply , Mice , Mice, Inbred C57BL , Models, Animal , Neutrophil Infiltration/drug effects , Peroxidase/metabolism , Reperfusion Injury/physiopathology
6.
J Immunol ; 177(8): 5558-66, 2006 Oct 15.
Article in English | MEDLINE | ID: mdl-17015743

ABSTRACT

Decay-accelerating factor (DAF) and complement receptor 1-related gene/protein y (Crry) are two membrane-bound complement regulators on murine erythrocytes that inhibit C3/C5 convertases. Previously, we found that Crry- but not DAF-deficient erythrocytes were susceptible to alternative pathway complement-mediated elimination in vivo. To determine whether it is a unique activity or a higher level expression of Crry makes it indispensable on murine erythrocytes, we over-expressed DAF on Crry-deficient (Crry(-/-)) erythrocytes by retroviral vector-mediated DAF gene transduction of bone marrow stem cells. DAF retrovirus-transduced erythrocytes expressed 846 +/- 127 DAF molecules/cell (DAF(high)) compared with 249 +/- 94 DAF molecules/cell (DAF(low)) and 774 +/- 135 Crry molecules/cell on control mouse erythrocytes. DAF(high)-Crry(-/-) erythrocytes were significantly more resistant than either DAF(low)-Crry(-/-), DAF(-/-) -Crry(+/+) or wild-type erythrocytes to classical pathway complement-mediated C3 deposition in vitro. Furthermore, increased DAF expression rescued Crry(-/-) erythrocytes from acute alternative pathway complement attack in vivo. Notably, long term monitoring revealed that DAF(high)-Crry(-/-) erythrocytes were still more susceptible than wild-type erythrocytes to complement-mediated elimination as they had a shorter half-life in complement-sufficient mice but survived equally well in complement-deficient mice. These results suggest that both a high level expression and a more potent anti-alternative pathway complement activity of Crry contributed to its indispensable role on murine erythrocytes. Additionally, they demonstrate the feasibility of using stem cell gene therapy to correct membrane complement regulator deficiency on blood cells in vivo.


Subject(s)
CD55 Antigens/administration & dosage , Complement Pathway, Alternative , Erythrocytes , Receptors, Complement/deficiency , Stem Cells/metabolism , Transduction, Genetic/methods , Animals , Bone Marrow Cells , CD55 Antigens/therapeutic use , Erythrocytes/immunology , Erythrocytes/metabolism , Erythrocytes/pathology , Genetic Therapy/methods , Half-Life , Mice , Mice, Knockout , Receptors, Complement/immunology , Receptors, Complement 3b , Retroviridae
7.
Blood ; 92(11): 4439-45, 1998 Dec 01.
Article in English | MEDLINE | ID: mdl-9834251

ABSTRACT

Hemolytic anemia is a major feature of paroxysmal nocturnal hemoglobinuria (PNH). Intravascular red blood cell (RBC) destruction is caused by increased sensitivity of the abnormal erythrocyte to complement-mediated lysis, due to the GPI absence of a membrane-bound glycosylphosphatidylinositol (GPI)-linked protein, which functions as an inhibitor of reactive lysis (CD59). Both in vivo and in vitro models have suggested the feasibility of cell-to-cell transfer of GPI proteins, and patients with hemolysis could potentially benefit from transfer of CD59 to their deficient erythrocytes. We studied the ability of RBC components prepared from outdated packed RBC collections, as well as high-density lipoprotein (HDL) preparations, rich in CD55 and CD59, to promote protein transfer, as assessed by flow cytometry, immunoblotting, and susceptibility to complement-mediated lysis. By flow cytometry, CD55 and CD59 were present on RBC-derived microvesicles that stained with an antiglycophorin antibody Ab; in addition, soluble CD59 and CD55 were detected by immunoblot in soluble fractions eluated from RBC units stored for more than 35 days, but not in fresh blood. Both commercial HDL preparations and those prepared in our laboratory contained CD55 and CD59, as assayed by immunoblot. When RBC that were deficient (GPI)-anchored protein, obtained from five patients, with PNH were incubated with HDL preparations for 2 to 4 hours, there was significant transfer of both proteins to the cell surface, as demonstrated by flow cytometry. Washed RBC microvesicles, prepared by ultrasonification, also mediated transfer of GPI-linked proteins to deficient RBC. Pretreatment of microvesicles, RBC eluate preparations, and HDL with phosphatidylinositol-specific, phospholipase C, abrogated protein transfer to deficient cells, indicating that increased cell-associated CD55 and CD59 levels were related to insertion of the intact GPI moiety, rather than to simple adhesion. PNH RBC that were exposed to HDL, RBC eluate preparations, or microvesicles demonstrated decreased in vitro complement-mediated hemolysis in the Ham test. Transfer of GPI-linked proteins from soluble preparations containing CD55 and CD59 to PNH erythrocytes is feasible and may have clinical utility.


Subject(s)
CD55 Antigens/administration & dosage , CD59 Antigens/administration & dosage , Drug Delivery Systems , Erythrocytes/metabolism , Hemoglobinuria, Paroxysmal/blood , CD55 Antigens/immunology , CD55 Antigens/metabolism , CD55 Antigens/therapeutic use , CD59 Antigens/immunology , CD59 Antigens/metabolism , CD59 Antigens/therapeutic use , Erythrocytes/immunology , Erythrocytes/pathology , Flow Cytometry , Glycosylphosphatidylinositols , Hemoglobinuria, Paroxysmal/drug therapy , Humans , Lipoproteins, HDL
SELECTION OF CITATIONS
SEARCH DETAIL
...