Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
Mol Immunol ; 56(1-2): 91-7, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23685261

ABSTRACT

Preeclampsia is a major obstetric problem defined by new-onset hypertension and proteinuria associated with compromised placental perfusion. Although activation of the complement system is increased in preeclampsia compared to normal pregnancy, it remains unclear whether excess complement activation is a cause or consequence of placental ischemia. Therefore, we hypothesized that complement activation is critical for placental ischemia-induced hypertension. We employed the reduced utero-placental perfusion pressure (RUPP) model of placental ischemia in the rat to induce hypertension in the third trimester and evaluated the effect of inhibiting complement activation with a soluble recombinant form of an endogenous complement regulator, human complement receptor 1 (sCR1; CDX-1135). On day 14 of a 21-day gestation, rats received either RUPP or Sham surgery and 15 mg/kg/day sCR1 or saline intravenously on days 14-18. Circulating complement component 3 decreased and complement activation product C3a increased in RUPP vs. Sham (p<0.05), indicating complement activation had occurred. Mean arterial pressure (MAP) measured on day 19 increased in RUPP vs. Sham rats (109.8±2.8 mmHg vs. 93.6±1.6 mmHg). Treatment with sCR1 significantly reduced elevated MAP in RUPP rats (98.4±3.6 mmHg, p<0.05) and reduced C3a production. Vascular endothelial growth factor (VEGF) decreased in RUPP compared to Sham rats, and the decrease in VEGF was not affected by sCR1 treatment. Thus, these studies have identified a mechanistic link between complement activation and the pregnancy complication of hypertension apart from free plasma VEGF and have identified complement inhibition as a potential treatment strategy for placental ischemia-induced hypertension in preeclampsia.


Subject(s)
Complement Activation/physiology , Hypertension/physiopathology , Ischemia/physiopathology , Placenta/physiopathology , Animals , Arterial Pressure/drug effects , Carotid Arteries/drug effects , Carotid Arteries/physiopathology , Complement Activation/drug effects , Complement C3/metabolism , Dose-Response Relationship, Drug , Female , Humans , Hypertension/blood , Hypertension/metabolism , In Vitro Techniques , Male , Nitroprusside/pharmacology , Placenta/blood supply , Placenta/metabolism , Pre-Eclampsia/blood , Pre-Eclampsia/metabolism , Pre-Eclampsia/physiopathology , Pregnancy , Rats , Rats, Sprague-Dawley , Receptors, Complement/administration & dosage , Time Factors , Uterus/blood supply , Uterus/metabolism , Uterus/physiopathology , Vascular Endothelial Growth Factor A/blood , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-1/blood , Vascular Endothelial Growth Factor Receptor-1/metabolism , Vasodilation/drug effects , Vasodilator Agents/pharmacology
2.
Virol J ; 9: 285, 2012 Nov 23.
Article in English | MEDLINE | ID: mdl-23173749

ABSTRACT

BACKGROUND: Enterovirus 71 (EV71), one of the most important neurotropic EVs, has caused death and long-term neurological sequelae in hundreds of thousands of young children in the Asia-Pacific region in the past decade. The neurological diseases are attributed to infection by EV71 inducing an extensive peripheral and central nervous system (CNS) inflammatory response with abnormal cytokine production and lymphocyte depletion induced by EV71 infection. In the absence of specific antiviral agents or vaccines, an effective immunosuppressive strategy would be valuable to alleviate the severity of the local inflammation induced by EV71 infection. PRESENTATION OF THE HYPOTHESIS: The complement system plays a pivotal role in the inflammatory response. Inappropriate or excessive activation of the complement system results in a severe inflammatory reaction or numerous pathological injuries. Previous studies have revealed that EV71 infection can induce complement activation and an inflammatory response of the CNS. CR2-targeted complement inhibition has been proved to be a potential therapeutic strategy for many diseases, such as influenza virus-induced lung tissue injury, postischemic cerebral injury and spinal cord injury. In this paper, a mouse model is proposed to test whether a recombinant fusion protein consisting of CR2 and a region of Crry (CR2-Crry) is able to specifically inhibit the local complement activation induced by EV71 infection, and to observe whether this treatment strategy can alleviate or even cure the neurogenic inflammation. TESTING THE HYPOTHESIS: CR2-Crry is expressed in CHO cells, and its biological activity is determined by complement inhibition assays. 7-day-old ICR mice are inoculated intracranially with EV71 to duplicate the neurological symptoms. The mice are then divided into two groups, in one of which the mice are treated with CR2-Crry targeted complement inhibitor, and in the other with phosphate-buffered saline. A group of mice deficient in complement C3, the breakdown products of which bind to CR2, are also infected with EV71 virus. The potential bioavailability and efficacy of the targeted complement inhibitor are evaluated by histology, immunofluorescence staining and radiolabeling. IMPLICATIONS OF THE HYPOTHESIS: CR2-Crry-mediated targeting complement inhibition will alleviate the local inflammation and provide an effective treatment for the severe neurological diseases associated with EV71 infection.


Subject(s)
Enterovirus A, Human/pathogenicity , Immunosuppressive Agents/administration & dosage , Neurogenic Inflammation/drug therapy , Receptors, Complement 3d/antagonists & inhibitors , Animals , Biological Products/administration & dosage , Disease Models, Animal , Mice , Mice, Inbred ICR , Receptors, Complement/administration & dosage , Receptors, Complement/genetics , Receptors, Complement 3b , Receptors, Complement 3d/administration & dosage , Receptors, Complement 3d/genetics , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/genetics , Treatment Outcome
3.
Transpl Immunol ; 24(4): 233-7, 2011 May.
Article in English | MEDLINE | ID: mdl-21440065

ABSTRACT

Kidneys recovered from brain-dead donors have inferior outcomes after transplantation compared to kidneys from living donors. Since complement activation plays an important role in renal transplant related injury, targeting complement activation in brain-dead donors might improve renal function after transplantation. Brain death (BD) was induced in Fisher rats by inflation of an epidurally placed balloon catheter and ventilated for 6h. BD animals were treated with soluble complement receptor 1 (sCR1) 1h before or 1h after BD. Kidney transplantation was performed and 7 days after transplantation animals were sacrificed. Plasma creatinine and urea were measured at days 0, 1, 3, 5 and 7 after transplantation. Renal function was significantly better at day 1 after transplantation in recipients receiving a sCR1 pre-treated donor kidney compared to recipients of a non-treated donor graft. Also treatment with sCR1, 1h after the diagnosis of BD, resulted in a better renal function after transplantation. Gene expression of IL-6, IL-1beta and TGF-beta were significantly lower in renal allografts recovered from treated donors. This study shows that targeting complement activation, during BD in the donor, leads to an improved renal function after transplantation in the recipient.


Subject(s)
Brain Death , Catheterization , Kidney Transplantation , Kidney/drug effects , Receptors, Complement/administration & dosage , Animals , Cells, Cultured , Complement Activation/drug effects , Cytokines/genetics , Cytokines/metabolism , Humans , Inflammation Mediators/metabolism , Kidney/metabolism , Kidney/pathology , Kidney/surgery , Organ Preservation , Rats , Rats, Inbred Strains , Recovery of Function , Tissue and Organ Harvesting
4.
J Med Microbiol ; 57(Pt 12): 1466-1472, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19018015

ABSTRACT

The Candida antigen CR3-RP (complement receptor 3-related protein) is supposed to be a 'mimicry' protein because of its ability to bind antibody directed against the alpha subunit of the mammalian CR3 (CD11b/CD18). This study aimed to (i) investigate the specific humoral isotypic response to immunization with CR3-RP in vivo in a rabbit animal model, and (ii) determine the role of CR3-RP in the adherence of Candida albicans in vitro using the model systems of buccal epithelial cells (BECs) and biofilm formation. The synthetic C. albicans peptide DINGGGATLPQ corresponding to 11 amino-acids of the CR3-RP sequence DINGGGATLPQALXQITGVIT, determined by N-terminal sequencing, was used for immunization of rabbits to obtain polyclonal anti-CR3-PR serum and for subsequent characterization of the humoral isotypic response of rabbits. A significant increase of IgG, IgA and IgM anti-CR3-RP specific antibodies was observed after the third (P<0.01) and the fourth (P<0.001) immunization doses. The elevation of IgA levels suggested peptide immunomodulation of the IgA1 subclass, presumably in coincidence with Candida epithelial adherence. Blocking CR3-RP with polyclonal anti-CR3-RP serum reduced the ability of Candida to adhere to BECs, in comparison with the control, by up to 35 % (P<0.001), and reduced biofilm formation by 28 % (P<0.001), including changes in biofilm thickness and integrity detected by confocal laser scanning microscopy. These properties of CR3-RP suggest that it has potential for future vaccine development.


Subject(s)
Antibodies, Fungal/blood , Antigens, Fungal/immunology , Candida albicans/immunology , Candida albicans/pathogenicity , Cell Adhesion , Disease Models, Animal , Amino Acid Sequence , Animals , Antigens, Fungal/administration & dosage , Antigens, Fungal/chemistry , Biofilms/growth & development , Candida albicans/growth & development , Candida albicans/physiology , Epithelial Cells/microbiology , Fungal Proteins/administration & dosage , Fungal Proteins/chemistry , Fungal Proteins/immunology , Humans , Immunization , Molecular Sequence Data , Mouth Mucosa/cytology , Mouth Mucosa/microbiology , Rabbits , Receptors, Complement/administration & dosage , Receptors, Complement/chemistry , Receptors, Complement/immunology
5.
Eur J Neurosci ; 26(12): 3486-500, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18052971

ABSTRACT

Complement activation triggers inflammation and has been implicated in neurological diseases associated with pain. However, the role of complement in neuropathic pain has not been clearly defined. In this study, we tested whether complement is activated by partial ligation of the rat sciatic nerve, a widely used model of neuropathic pain, and whether complement activation or inhibition in peripheral nerve influences leukocyte recruitment and neuropathic pain. We found that C3 deposition significantly increased from 6 h to 7 days in the injured nerve and was associated with the extent of thermal hyperalgesia and mechanical allodynia. However, no deposition of the membrane attack complex was detected. Complement activation by endoneurial injection of aggregated rat immunoglobulin G into normal sciatic nerve produced significant thermal hyperalgesia and mechanical allodynia of the ipsilateral hindpaw at 2-7 days after injection. This was accompanied by increased deposition of C3 and recruitment of macrophages at 7 days following injection. Complement inhibition using systemic injections of soluble complement receptor 1 (AVANT Immunotherapeutics, Inc., Needham, USA) into rats markedly suppressed C3 deposition and T-cell and macrophage recruitment to the injured nerve, and produced significant alleviation of thermal hyperalgesia and mechanical allodynia. These results demonstrate that C3 activation in the nerve contributes to increased infiltration of inflammatory cells and to neuropathic pain behaviors following peripheral nerve injury. Complement inhibition may be a potential therapeutic treatment for neuropathic pain.


Subject(s)
Neuralgia/etiology , Sciatic Nerve/injuries , Animals , Behavior, Animal/drug effects , Complement Activation , Complement C3/antagonists & inhibitors , Complement C3/metabolism , Hyperalgesia/chemically induced , Hyperesthesia/chemically induced , Immunoglobulin G/administration & dosage , Immunoglobulin G/pharmacology , Injections , Leukocytes , Ligation , Macrophages/pathology , Male , Neuralgia/psychology , Rats , Rats, Wistar , Receptors, Complement/administration & dosage , Sciatic Nerve/drug effects , Sciatic Nerve/metabolism , Sciatic Nerve/pathology , T-Lymphocytes/pathology , Time Factors
6.
J Med Primatol ; 36(6): 375-80, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17976043

ABSTRACT

BACKGROUND: Soluble complement receptor-1 (sCR1), a potent complement inhibitor, confers neuroprotection in a murine stroke model. Additional neuroprotective benefit is achieved by sLe x-glycosylation of sCR1. In an effort to translate sCR1-sLe x to clinical trials, we evaluated this agent in a primate stroke model. METHODS: Adult male baboons randomly received either sCR1-sLe x or vehicle. Stroke volume was assessed on day 3, and neurological examinations were conducted daily. Complement activity (CH50) was measured at 30 minute, 2, 6, 12 hour, 3, and 10 days post-ischemia. RESULTS: The experiment was terminated prematurely following an interim analysis. In a preliminary cohort (n = 3 per arm), infarct volume was greater in the treated animals. No difference in neurological score was found between groups. CH50 levels were significantly reduced in the sCR1sLe x-treated groups. A hypotensive response was also observed in animals treated with sCR1-sLe x. Conclusions Further work is necessary to explain the hypotensive response observed in primates prior to further clinical development of sCR1-sLe x.


Subject(s)
Disease Models, Animal , Neuroprotective Agents/administration & dosage , Papio anubis , Receptors, Complement/administration & dosage , Reperfusion Injury/prevention & control , Stroke/prevention & control , Animals , Brain Ischemia/prevention & control , Cerebral Infarction/prevention & control , Complement Hemolytic Activity Assay/veterinary , Drug Evaluation, Preclinical , Male , Random Allocation , Time Factors , Treatment Outcome
7.
J Immunol ; 177(10): 7266-74, 2006 Nov 15.
Article in English | MEDLINE | ID: mdl-17082645

ABSTRACT

The mechanisms that contribute to inflammatory damage following ischemic stroke are poorly characterized, but studies indicate a role for both complement and P-selectin. In this study, we show that compared with wild-type mice, C3-deficient mice showed significant improvement in survival, neurological deficit, and infarct size at 24 h after middle cerebral artery occlusion and reperfusion. Furthermore, P-selectin protein expression was undetectable in the cerebral microvasculature of C3-deficient mice following reperfusion, and there was reduced neutrophil influx, reduced microthrombus formation, and increased blood flow postreperfusion in C3-deficient mice. We further investigated the use of a novel complement inhibitory protein in a therapeutic paradigm. Complement receptor 2 (CR2)-Crry inhibits complement activation at the C3 stage and targets to sites of complement activation. Treatment of normal mice with CR2-Crry at 30 min postreperfusion resulted in a similar level of protection to that seen in C3-deficient mice in all of the above-measured parameters. The data demonstrate an important role for complement in cerebrovascular thrombosis, inflammation, and injury following ischemic stroke. P-selectin expression in the cerebrovasculature, which is also implicated in cerebral ischemia and reperfusion injury, was shown to be distal to and dependent on complement activation. Data also show that a CR2-targeted approach of complement inhibition provides appropriate bioavailability in cerebral injury to enable complement inhibition at a dose that does not significantly affect systemic levels of serum complement activity, a potential benefit for stroke patients where immunosuppression would be undesirable due to significantly increased susceptibility to lung infection.


Subject(s)
Brain Ischemia/metabolism , Complement C3/antagonists & inhibitors , Complement C3/physiology , P-Selectin/biosynthesis , Animals , Apoptosis/genetics , Apoptosis/immunology , Brain Ischemia/etiology , Brain Ischemia/genetics , Brain Ischemia/therapy , Cerebrovascular Circulation/immunology , Complement C3/deficiency , Complement C3/genetics , Complement Inactivator Proteins/administration & dosage , Complement Inactivator Proteins/physiology , Complement Inactivator Proteins/therapeutic use , Infarction, Middle Cerebral Artery/genetics , Infarction, Middle Cerebral Artery/metabolism , Infarction, Middle Cerebral Artery/pathology , Infarction, Middle Cerebral Artery/therapy , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophil Infiltration/genetics , Neutrophil Infiltration/immunology , P-Selectin/physiology , Receptors, Complement/administration & dosage , Receptors, Complement/metabolism , Receptors, Complement/physiology , Receptors, Complement 3b , Receptors, Complement 3d/administration & dosage , Receptors, Complement 3d/physiology , Receptors, Complement 3d/therapeutic use , Reperfusion Injury/etiology , Reperfusion Injury/genetics , Reperfusion Injury/metabolism , Reperfusion Injury/therapy , Survival Analysis
8.
Am J Pathol ; 169(3): 1039-47, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16936276

ABSTRACT

Initiation of an inflammatory cascade following traumatic spinal cord injury (SCI) is thought to cause secondary injury and to adversely impact functional recovery, although the mechanisms involved are not well defined. We report on the dynamics of complement activation and deposition in the mouse spinal cord following traumatic injury, the role of complement in the development of SCI, and the characterization of a novel targeted complement inhibitor. Following traumatic injury, mice deficient in C3 had a significantly improved locomotor score when compared with wild-type controls, and analysis of their spinal cords revealed significantly more tissue sparing, with significantly less necrosis, demyelination, and neutrophil infiltration. Wild-type mice were also treated with CR2-Crry, a novel inhibitor of complement activation that targets to sites of C3 deposition. A single intravenous injection of CR2-Crry 1 hour after traumatic injury improved functional outcome and pathology to an extent similar to that seen in C3-deficient animals. CR2-Crry specifically targeted to the injured spinal cord in a distribution pattern corresponding to that seen for deposited C3. As immunosuppression is undesirable in patients following SCI, targeted CR2-Crry may provide appropriate bioavailability to treat SCI at a dose that does not significantly affect systemic levels of serum complement activity.


Subject(s)
Complement Activation/drug effects , Complement C3/antagonists & inhibitors , Receptors, Complement 3d/administration & dosage , Receptors, Complement/administration & dosage , Recombinant Fusion Proteins/administration & dosage , Spinal Cord Injuries/drug therapy , Animals , Complement C3/deficiency , Complement C3/metabolism , Demyelinating Diseases/drug therapy , Demyelinating Diseases/metabolism , Demyelinating Diseases/pathology , Female , Humans , Mice , Mice, Knockout , Necrosis/drug therapy , Necrosis/metabolism , Necrosis/pathology , Neutrophil Infiltration/drug effects , Receptors, Complement/genetics , Receptors, Complement 3b , Receptors, Complement 3d/genetics , Recombinant Fusion Proteins/genetics , Spinal Cord Injuries/metabolism , Spinal Cord Injuries/pathology
9.
Exp Neurol ; 199(2): 454-64, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16545803

ABSTRACT

The complement system represents an important mediator of neuroinflammation in traumatic brain injury. We have previously shown that transgenic mice with central nervous system-targeted overexpression of Crry, a potent murine complement inhibitor at the level of C3 convertases, are protected from complement-mediated neuropathological sequelae in brain-injured mice. This knowledge was expanded in the present study to a pharmacological approach by the use of a recombinant Crry molecule (termed Crry-Ig) which was recently made available in a chimeric form fused to the non-complement fixing mouse IgG1 Fc region. In a standardized model of closed head injury in mice, the systemic injection of 1 mg Crry-Ig at 1 h and 24 h after trauma resulted in a significant neurological improvement for up to 7 days, as compared to vehicle-injected control mice (P < 0.05, repeated measures ANOVA). Furthermore, the extensive neuronal destruction seen in the hippocampal CA3/CA4 sublayers in head-injured mice with vehicle injection only was shown to be preserved - to a similar extent as in "sham"-operated mice - by the posttraumatic injection of Crry-Ig. Real-time RT-PCR analysis revealed that the post-treatment with Crry-Ig resulted in a significant up-regulation of candidate neuroprotective genes in the injured hemisphere (Bcl-2, C1-Inh, CD55, CD59), as compared to the vehicle control group (P < 0.01, unpaired Student's t test). Increased intracerebral Bcl-2 expression by Crry-Ig treatment was furthermore confirmed at the protein level by Western blot analysis. These data suggest that pharmacological complement inhibition represents a promising approach for attenuation of neuroinflammation and secondary neurodegeneration after head injury.


Subject(s)
Brain Injuries , Brain/drug effects , Gene Expression/drug effects , Neurons/drug effects , Neuroprotective Agents/administration & dosage , Receptors, Complement/administration & dosage , Animals , Behavior, Animal/drug effects , Behavior, Animal/physiology , Blotting, Western/methods , Brain/metabolism , Brain/pathology , Brain Injuries/drug therapy , Brain Injuries/pathology , Brain Injuries/physiopathology , Cell Survival/drug effects , Complement System Proteins/genetics , Complement System Proteins/metabolism , Disease Models, Animal , Immunohistochemistry/methods , Male , Mice , Mice, Inbred C57BL , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Psychomotor Performance/drug effects , RNA, Messenger/metabolism , Receptors, Complement 3b , Reverse Transcriptase Polymerase Chain Reaction/methods , Time Factors , Trauma Severity Indices
11.
J Immunol ; 174(9): 5750-7, 2005 May 01.
Article in English | MEDLINE | ID: mdl-15843577

ABSTRACT

In glomerular diseases of diverse etiologies, dysfunction of the glomerular barrier to protein passage results in proteinuria, and proteinuria is considered an independent risk factor that plays a direct role in inflammation, interstitial fibrosis, and renal failure. The mechanism by which proteinuria leads to nephrotoxic injury is unclear, but a role for complement in mediating interstitial damage appears likely. We describe a strategy for Ag-specific targeting of complement inhibitors using a single chain Ab fragment and show that complement inhibitors targeted to the tubular epithelium protect against tubulointerstitial injury and renal dysfunction in a rat model of puromycin-induced nephrosis. The targeting of systemically administered complement inhibitors markedly enhanced their efficacy and obviated the need to systemically inhibit complement, thus reducing the risk of compromising host defense and immune homeostasis. Targeted inhibition of complement activation by Crry, and of membrane attack complex (MAC) formation by CD59 was equally therapeutic, demonstrating that the MAC plays a key role in proteinuria-induced tubulointerstitial injury. CD59 activity was dependent on its being targeted to the site of complement activation, and this is the first report of specific inhibition of the MAC in vivo after systemic administration of inhibitor. The data establish the MAC is a valid target for pharmaceutical intervention in proteinuric disorders and provide an approach to investigate the role of the MAC in complement-dependent disease under clinically relevant conditions.


Subject(s)
CD59 Antigens/metabolism , Complement Inactivator Proteins/metabolism , Complement Membrane Attack Complex/physiology , Kidney Tubules, Proximal/immunology , Kidney Tubules, Proximal/pathology , Nephrotic Syndrome/immunology , Nephrotic Syndrome/prevention & control , Receptors, Complement/metabolism , Animals , Antigens, Surface , CD59 Antigens/administration & dosage , Complement Inactivator Proteins/chemistry , Complement Inactivator Proteins/pharmacokinetics , Disease Models, Animal , Drug Delivery Systems/methods , Female , Kidney Tubules, Proximal/metabolism , Nephritis, Interstitial/immunology , Nephritis, Interstitial/pathology , Nephritis, Interstitial/prevention & control , Nephrotic Syndrome/pathology , Organ Specificity/immunology , Rats , Rats, Sprague-Dawley , Receptors, Cell Surface , Receptors, Complement/administration & dosage , Receptors, Complement/blood , Receptors, Complement/chemistry , Recombinant Proteins/administration & dosage , Recombinant Proteins/blood , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacokinetics
12.
Circulation ; 110(11 Suppl 1): II274-9, 2004 Sep 14.
Article in English | MEDLINE | ID: mdl-15364875

ABSTRACT

BACKGROUND: This study was undertaken to determine whether soluble human complement receptor type 1 (TP10), a potent inhibitor of complement activation, would reduce morbidity and mortality in high-risk patients undergoing cardiac surgery on cardiopulmonary bypass (CPB). METHODS: This was a randomized multicenter, prospective, placebo-controlled, double-blind study in which 564 high-risk patients undergoing cardiac surgery on CPB received an intravenous bolus of TP10 (1, 3, 5, 10 mg/kg) or placebo immediately before CPB. The primary endpoint was the composite events of death, myocardial infarction (MI), prolonged (> or =24 hours) intra-aortic balloon pump support (IABP), and prolonged intubation. RESULTS: TP10 significantly inhibited complement activity after 10 to 15 minutes of CPB and this inhibition persisted for 3 days postoperatively. However, there was no difference in the primary endpoint between the 2 groups (33.7% placebo versus 31.4% TP10; P=0.31). The primary composite endpoint was, however, reduced in all male TP10 patients by 30% (P=0.025). TP10 reduced the incidence of death or MI in males by 36% (P=0.026), the incidence of death or MI in CABG males by 43% (P=0.043) and the need for prolonged IABP support in male CABG and valve patients by 100% (P=0.019). There was, however, no improvement seen in female TP10 patients. There were no significant differences in adverse events between the groups. CONCLUSIONS: TP10 effectively inhibits complement activation during CPB; however, this was not associated with an improvement in the primary endpoint of the study. Nevertheless, TP10 did significantly decrease the incidence of mortality and MI in male patients.


Subject(s)
Cardiac Surgical Procedures , Cardiopulmonary Bypass/adverse effects , Complement Activation/drug effects , Myocardial Ischemia/prevention & control , Receptors, Complement/therapeutic use , Aged , Cardiac Surgical Procedures/mortality , Complement C3a/biosynthesis , Complement Membrane Attack Complex/biosynthesis , Double-Blind Method , Female , Humans , Infections/epidemiology , Injections, Intravenous , Intra-Aortic Balloon Pumping/statistics & numerical data , Length of Stay/statistics & numerical data , Male , Middle Aged , Myocardial Infarction/diagnosis , Myocardial Infarction/epidemiology , Myocardial Infarction/etiology , Myocardial Ischemia/etiology , Myocardial Ischemia/immunology , Prospective Studies , Receptors, Complement/administration & dosage , Sex Factors , Solubility , Treatment Outcome
13.
Pain ; 110(1-2): 299-309, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15275780

ABSTRACT

In inflammatory neuropathy, immune activation near intact peripheral nerves induces mechanical allodynia. The identity of the peripheral immune product(s) that lead to these changes in pain behavior is unknown. The present series of studies utilized the sciatic inflammatory neuropathy (SIN) model to examine this question. Here, inflammatory neuropathy is created by injecting an immune activator (zymosan) around one sciatic nerve via an indwelling catheter. Our prior studies demonstrated that peri-sciatic zymosan activated macrophages and neutrophils to release proinflammatory cytokines and reactive oxygen species (ROS). In addition, zymosan is a classical activator of the complement cascade. Thus the present series of experiments examined whether any of these inflammatory mediators are involved in the initial induction of SIN-induced ipsilateral or bilateral allodynias. Peri-sciatic injection of selective inhibitors/antagonists revealed that a number of immune products are early mediators of the resultant allodynias, including proinflammatory cytokines (tumor necrosis factor, interleukin-1, and interleukin-6), ROS, and complement. Thus these immune-derived substances can markedly alter sensory nerve function at mid-axon.


Subject(s)
Complement System Proteins , Cytokines/metabolism , Neuralgia/immunology , Reactive Oxygen Species/metabolism , Sciatic Neuropathy/metabolism , Animals , Antibodies/administration & dosage , Behavior, Animal , Carrier Proteins/therapeutic use , Catalase/administration & dosage , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Interactions , Functional Laterality , Hyperalgesia/chemically induced , Hyperalgesia/immunology , Interleukin 1 Receptor Antagonist Protein , Interleukin-6/immunology , Male , Neuralgia/drug therapy , Neuralgia/etiology , Pain Measurement , Physical Stimulation , Rats , Rats, Sprague-Dawley , Receptors, Complement/administration & dosage , Receptors, Tumor Necrosis Factor/therapeutic use , Receptors, Tumor Necrosis Factor, Type I , Sciatic Nerve/immunology , Sciatic Nerve/pathology , Sciatic Neuropathy/chemically induced , Sciatic Neuropathy/immunology , Sialoglycoproteins/administration & dosage , Superoxide Dismutase/administration & dosage , Tumor Necrosis Factor Decoy Receptors , Zymosan/toxicity
14.
Br J Pharmacol ; 142(4): 756-64, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15159277

ABSTRACT

1. Complement activation is implicated in the pathogenesis of intestinal ischaemia-reperfusion injury (I/R), although the relative importance of individual complement components is unclear. A C3a receptor antagonist N(2)-[(2,2-diphenylethoxy)acetyl]-l-arginine (C3aRA) has been compared with a C5a receptor antagonist (C5aRA), AcF-[OPdChaWR], in a rat model of intestinal I/R. 2. C3aRA (IC(50)=0.15 microm) and C5aRA (IC(50)=0.32 microm) bound selectively to human polymorphonuclear leukocyte (PMN) C3a and C5a receptors, respectively. Effects on circulating neutrophils and blood pressure in the rat were also assessed. 3. Anaesthetised rats, subjected to intestinal ischaemia (30 min) and reperfusion (120 min), were administered intravenously with either (A) the C3aRA (0.1-1.0 mg x kg(-1)); the C5aRA (1.0 mg x kg(-1)); the C3aRA+C5aRA (each 1.0 mg x kg(-1)); or vehicle, 45 min prior, or (B) the C3aRA (1.0 mg x kg(-1)) or vehicle, 120 min prior to reperfusion. 4. The C3aRA and C5aRA, administered 45 min prior to reperfusion, displayed similar efficacies at ameliorating several disease markers (increased oedema, elevated ALT levels and mucosal damage) of rat intestinal I/R. The combination drug treatment did not result in greater injury reduction than either antagonist alone. However, doses of the C3aRA (0.01-10 mg x kg(-1)) caused transient neutropaenia, and the highest dose (10 mg x kg(-1)) also caused a rapid and transient hypertension. 5. The C3aRA (1.0 mg x kg(-1)), delivered 120 min prior to reperfusion to remove the global effect of C3aRA-induced neutrophil sequestration, did not attenuate the markers of intestinal I/R, despite persistent C3aR antagonism at this time. 6. C3aR antagonism does not appear to be responsible for the anti-inflammatory actions of this C3aRA in intestinal I/R in the rat. Instead, C3aRA-mediated global neutrophil tissue sequestration during ischaemia and early reperfusion may account for the protective effects observed.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Arginine/analogs & derivatives , Arginine/therapeutic use , Benzhydryl Compounds/therapeutic use , Disease Models, Animal , Intestinal Mucosa/pathology , Membrane Proteins/antagonists & inhibitors , Peptides, Cyclic/therapeutic use , Receptor, Anaphylatoxin C5a/antagonists & inhibitors , Receptors, Complement/antagonists & inhibitors , Reperfusion Injury/drug therapy , Alanine Transaminase/blood , Animals , Anti-Inflammatory Agents/administration & dosage , Anti-Inflammatory Agents/chemical synthesis , Arginine/administration & dosage , Arginine/chemical synthesis , Australia , Benzhydryl Compounds/administration & dosage , Benzhydryl Compounds/chemical synthesis , Dose-Response Relationship, Drug , Drug Combinations , Female , Humans , Injections, Intravenous , Intestinal Mucosa/drug effects , Intestinal Mucosa/ultrastructure , Membrane Proteins/administration & dosage , Membrane Proteins/therapeutic use , Neutropenia/chemically induced , Neutropenia/complications , Neutrophils/drug effects , Neutrophils/metabolism , Peptides, Cyclic/administration & dosage , Peptides, Cyclic/chemical synthesis , Rats , Rats, Wistar , Receptor, Anaphylatoxin C5a/administration & dosage , Receptor, Anaphylatoxin C5a/therapeutic use , Receptors, Complement/administration & dosage , Receptors, Complement/therapeutic use , Reperfusion Injury/complications , Reperfusion Injury/metabolism , Time Factors
15.
Am J Respir Crit Care Med ; 168(11): 1333-41, 2003 Dec 01.
Article in English | MEDLINE | ID: mdl-14500265

ABSTRACT

Studies in murine models have suggested the involvement of the complement anaphylatoxins (C3a and C5a) in the development of allergic asthma. We investigated the effects of inhibiting complement activation after sensitization but before allergen challenge on the development of allergic airway inflammation and airway hyperresponsiveness. To prevent complement activation, we used a recombinant soluble form of the mouse membrane complement inhibitor complement receptor-related gene y (Crry) fused to the IgG1 hinge, CH2 and CH3 domains (Crry-Ig), which has decay-accelerating activity for both the classic and alternative pathways of complement as well as cofactor activity for factor I-mediated cleavage of C3b and C4b. C57BL/6 mice were sensitized (Days 1 and 14) and challenged (Days 24-26) with ovalbumin. Crry-Ig was administered after allergen sensitization either as an intraperitoneal injection or by nebulization before allergen challenge. Crry-Ig significantly prevented the development of airway hyperresponsiveness, decreased airway and lung eosinophilia as well as the numbers of lung lymphocytes, decreased levels of interleukin (IL)-4, IL-5, and IL-13 in bronchoalveolar lavage fluid and decreased serum ovalbumin-specific IgE and IgG1. These results suggest that prevention of complement activation may have a therapeutic role in the treatment of allergic airway inflammation and asthma in sensitized individuals.


Subject(s)
Complement Pathway, Alternative/drug effects , Complement Pathway, Classical/drug effects , Immunoglobulin G/administration & dosage , Proteins/administration & dosage , Receptors, Complement/administration & dosage , Respiratory Hypersensitivity/drug therapy , Animals , Bronchial Provocation Tests , Disease Models, Animal , Female , Inflammation/drug therapy , Mice , Mice, Inbred C57BL , Receptors, Complement 3b
16.
Microvasc Res ; 62(3): 284-92, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11678631

ABSTRACT

Recent observations provide evidence that complement is implicated as an important factor in the pathophysiology of ischemia/reperfusion injury (IRI). Here, we assessed the effects of complement inhibition on hepatic microcirculation by in vivo microscopy (IVM) using a rat model of warm hepatic ischemia clamping the left pedicle for 70 min. Ten animals received the physiological complement regulator soluble complement receptor type 1 (sCR1) intravenously 1 min prior to reperfusion. Controls were given an equal amount of Ringer's solution (n = 10). Microvascular perfusion and leukocyte adhesion were studied 30 to 100 min after reperfusion by IVM. Microvascular perfusion in hepatic sinusoids was significantly improved in the sCR1 group (80.6 +/- 0.6% of all observed sinusoids were perfused [sCR1] vs 67.3 +/- 1.2% [controls]). The number of adherent leukocytes was reduced in sinusoids (49.9 +/- 3.4 [sCR1] vs 312.3 +/- 14.2 in controls [adherent leukocytes per square millimeter of liver surface]; P < 0.001) as well as in postsinusoidal venules after sCR1 treatment (230.9 +/- 21.7 [sCR1] vs 1906.5 +/- 93.5 [controls] [adherent leukocytes per square millimeter of endothelial surface]; P < 0.001). Reflecting reduced hepatocyte injury, liver transaminases were decreased significantly upon sCR1 treatment compared to controls. Our results provide further evidence that complement plays a decisive role in warm hepatic IRI. Therefore, we conclude that complement inhibition by sCR1 is effective as a therapeutical approach to reduce microcirculatory disorders after reperfusion following warm organ ischemia.


Subject(s)
Complement C1 Inactivator Proteins/pharmacology , Complement C1s/physiology , Ischemia/physiopathology , Liver Circulation/physiology , Microcirculation/drug effects , Receptors, Complement/physiology , Reperfusion Injury/physiopathology , Animals , Cell Adhesion/physiology , Endothelium, Vascular/cytology , Endothelium, Vascular/physiology , Hemodynamics , Injections, Intravenous , Ischemia/pathology , Laser-Doppler Flowmetry/instrumentation , Leukocytes/physiology , Liver Circulation/drug effects , Male , Microcirculation/physiopathology , Microscopy, Fluorescence , Rats , Rats, Wistar , Receptors, Complement/administration & dosage , Temperature , Time Factors , Venules/physiopathology , Video Recording
17.
Curr Opin Investig Drugs ; 2(3): 364-71, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11575706

ABSTRACT

AVANT Immunotherapeutics is developing TP-10, a recombinant soluble complement receptor type 1 (sCR1), for the potential treatment of reperfusion injury (following surgery, ischemic disease and organ transplantation), organ rejection, acute inflammatory injury to the lungs and autoimmune diseases [348669]. TP-10 has been awarded Orphan Drug status from the FDA for the prevention and reduction of adult respiratory distress syndrome (ARDS) and as a treatment for infants undergoing cardiac surgery [180849], [359588]. A placebo-controlled phase II trial, conducted at approximately 30 sites in the US and involving approximately 600 adult patients undergoing cardiac surgery utilizing cardiopulmonary bypass, was initiated in November 2000. This safety and efficacy study was designed to assess the ability of TP-10 to mitigate the injury to the heart, brain and other organs that occurs when patients are placed on cardiopulmonary bypass circuits, thus potentially improving postoperative outcomes [391437]. In September 2000, the company was planning a double-blind, placebo controlled phase IIb trial in infants undergoing cardiac surgery; AVANT expected to initiated in 30 infants in January 2001 [395086]. The data from this trial will enable the company to further define its clinical endpoints before inititating a pivotal phase III trial in 2001 [382529]. A phase I/II trial of TP-10 involving 15 infants, under 12 months of age, undergoing cardiac surgery for congenital heart defects was initiated by the company in September 1999. The trial will evaluate the ability of TP-10 to mitigate the injury to the heart and other organs when patients are placed on cardiopulmonary bypass circuits [340602]. Enrollment was complete by January 2000 [352458]. Phase I safety trials of TP-10, including studies in adult patients at risk for adult respiratory distress syndrome (ARDS), adult patients with first-time myocardial infarction (heart attack), and pediatric patients undergoing cardiac surgery demonstrated that TP-10 is well tolerated. However, after completion, in December 1997, of a phase IIa trial in nine patients with ARDS, AVANT decided to cease development for this indication. TP-10 was licensed to Novartis AG for use in xeno- and allotransplantation in July 1999. Extensive animal studies have shown TP-10 to have potential in a wide variety of complement-mediated conditions, including organ transplantation, multiple sclerosis, rheumatoid arthritis and lupus [238093]. Early work demonstrated favorable results in animal models of reperfusion injury [180849] and hyperacute xenograft rejection in guinea pig to rat and pig to primate organ transplants [191552]. AVANT has received Notices of Allowance (July 1998) from the USPTO for three separate patent applications covering pharmaceutical compositions of TP-10, methods of purification and methods of certain TP-10 glycoforms for treating diseases or disorders resulting from inappropriate complement activation [291776]. In January 1999, the company was awarded US-05856297 which covers pharmaceutical compositions of TP-10. US-05856300 was also awarded covering compositions and methods of producing the drug [312267].


Subject(s)
Complement Inactivator Proteins/pharmacology , Drugs, Investigational , Receptors, Complement/administration & dosage , Acute-Phase Reaction/prevention & control , Animals , Clinical Trials, Phase I as Topic , Clinical Trials, Phase II as Topic , Complement Inactivator Proteins/administration & dosage , Complement Inactivator Proteins/therapeutic use , Disease Models, Animal , Drug Evaluation , Drug Evaluation, Preclinical , Forecasting , Humans , Injections, Intravenous , Patents as Topic , Receptors, Complement/immunology , Receptors, Complement/therapeutic use
18.
Clin Immunol ; 99(2): 222-31, 2001 May.
Article in English | MEDLINE | ID: mdl-11318594

ABSTRACT

C1q and the outer envelope protein of HIV, gp120, have several structural and functional similarities. Therefore, it is plausible to assume that proteins that are able to interact with C1q may also interact with isolated gp120 as well as the whole HIV-1 virus. Based on this hypothesis, we studied the potential ability of the recombinant form of the 33-kDa protein, which binds to the globular "heads" of C1q (gC1q-R/p33), to inhibit the growth of different HIV-1 strains in cell cultures. gC1q-R/p33 was found to effectively and dose-dependently inhibit the production of one T-lymphotropic (X4) and one macrophage-tropic (R5) strain in human T cell lines (MT-4 and H9) and human monocyte-derived macrophage cultures, respectively. At a concentration range of 5-25 microg/ml, gC1q-R caused a marked and prolonged suppression of virus production. The extent of inhibition was enhanced when gC1q-R was first incubated with and then removed from the target cell cultures before virus infection, compared to that when the cells were infected with gC1q-R-HIV mixtures. The extent of inhibition was comparable to that of the Leu3a anti-CD4 antibody. Addition of gC1q-R to the cell cultures on day 1 or 2 after infection induced markedly less inhibition of HIV-1 growth than pretreatment of the cells just before or together with the infective HIV strains. In ELISA experiments, gC1q-R did not bind to a solid-phase recombinant gp120 while strong and dose-dependent binding of gC1q-R to solid-phase CD4 was observed. Our present findings indicate that gC1q-R is an effective inhibitor of HIV-1 infection, which prevents viral entry by blocking the interaction between CD4 and gp120. Since gC1q-R is a human protein, it is most probably not antigenic in humans. It would seem logical, therefore, to consider gC1q-R or its fragments involved in the CD4 binding as potential therapeutic agents.


Subject(s)
Complement C1q/metabolism , HIV-1/drug effects , HIV-1/physiology , Hyaluronan Receptors , Membrane Glycoproteins , Receptors, Complement/administration & dosage , Receptors, Complement/metabolism , Anti-HIV Agents/chemistry , Anti-HIV Agents/pharmacology , Binding Sites , CD4 Antigens/metabolism , Carrier Proteins , Cell Line , Complement C1q/chemistry , HIV Envelope Protein gp120/metabolism , HIV Infections/drug therapy , HIV Infections/immunology , HIV Infections/virology , HIV-1/pathogenicity , Humans , Macrophages/drug effects , Macrophages/immunology , Macrophages/virology , Mitochondrial Proteins , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Solubility , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/virology , Virus Replication/drug effects
19.
Crit Care Med ; 28(9): 3149-54, 2000 Sep.
Article in English | MEDLINE | ID: mdl-11008973

ABSTRACT

OBJECTIVE: To determine the safety, pharmacokinetics, biological effects, and immunogenicity of recombinant soluble complement receptor 1 (TP10) in patients with acute lung injury (ALI) and acute respiratory distress syndrome (ARDS). DESIGN: Open label, ascending dosage, phase I trial. SETTING: Two academic teaching hospitals. PATIENTS: A total of 24 patients diagnosed with ALI/ARDS. INTERVENTION: A single, 30-min intravenous infusion of 0.1, 0.3, 1, 3, or 10 mg/kg TP10. MEASUREMENTS AND MAIN RESULTS: Serum levels of TP10 increased in proportion to the dose. Mean variable estimates (+/-SD) were half-life of disposition 69.7 +/- 39.7 hrs, plasma clearance 2.39 +/- 1.32 mL/hr/kg, and volume of distribution 190.6 +/- 135.0 mL/kg. Inhibition of complement activity, measured by CH50, was significant for the interaction of dose and time (p = .024). The C3a levels demonstrated a trend for dose which did not reach statistical significance (p = .090) and soluble C5b-9 levels were significant only for dose (p = .023). As expected by the proposed physiologic mechanism, C4a levels were not affected by TP10, dose, or time. The overall mortality rate was 33%. Neither the type nor the frequency rate of specific adverse events were substantially different between dose groups. Seven adverse events in four patients were thought to be possibly related to TP10. CONCLUSIONS: TP10 has a half-life of approximately 70 hrs and at doses > or =1 mg/kg, significantly inhibits complement activity at the levels of C3 and C5 in patients with ALI/ARDS. Complement inhibition was more prolonged over time with TP10 doses of 3 and 10 mg/kg. TP10 appears to be safe at the doses tested. Further studies will be required to completely assess the impact of TP10 on pathophysiology and clinical outcome in patients with ALI/ARDS.


Subject(s)
Lung Injury , Receptors, Complement/administration & dosage , Recombinant Proteins/administration & dosage , Respiratory Distress Syndrome/drug therapy , Adult , Aged , Antibodies/blood , Critical Care , Dose-Response Relationship, Drug , Female , Half-Life , Humans , Infusions, Intravenous , Lung/immunology , Male , Middle Aged , Receptors, Complement/immunology , Recombinant Proteins/adverse effects , Recombinant Proteins/immunology , Respiratory Distress Syndrome/immunology , Respiratory Distress Syndrome/mortality , Survival Rate
20.
Science ; 285(5427): 595-9, 1999 Jul 23.
Article in English | MEDLINE | ID: mdl-10417391

ABSTRACT

Glycoprotein adhesion receptors such as selectins contribute to tissue injury in stroke. Ischemic neurons strongly expressed C1q, which may target them for complement-mediated attack or C1qRp-mediated clearance. A hybrid molecule was used to simultaneously inhibit both complement activation and selectin-mediated adhesion. The extracellular domain of soluble complement receptor-1 (sCR1) was sialyl Lewis x glycosylated (sCR1sLex) to inhibit complement activation and endothelial-platelet-leukocyte interactions. sCR1 and sCR1sLex colocalized to ischemic cerebral microvessels and C1q-expressing neurons, inhibited neutrophil and platelet accumulation, and reduced cerebral infarct volumes. Additional benefit was conferred by sialyl Lewis x glycosylation of the unmodified parent sCR1 molecule.


Subject(s)
Cerebrovascular Disorders/drug therapy , Ischemic Attack, Transient/drug therapy , Neuroprotective Agents/therapeutic use , Oligosaccharides/therapeutic use , Receptors, Complement/therapeutic use , Animals , Blood Platelets/physiology , Cell Adhesion , Cerebral Cortex/blood supply , Cerebral Cortex/immunology , Cerebral Cortex/metabolism , Cerebral Infarction/drug therapy , Cerebrovascular Circulation , Cerebrovascular Disorders/immunology , Cerebrovascular Disorders/physiopathology , Complement Activation , Complement C1q/metabolism , Glycosylation , Humans , Ischemic Attack, Transient/immunology , Ischemic Attack, Transient/physiopathology , Leukocytes/physiology , Mice , Neurons/immunology , Neurons/metabolism , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/adverse effects , Neuroprotective Agents/metabolism , Neutrophils/physiology , Oligosaccharides/administration & dosage , Oligosaccharides/adverse effects , Oligosaccharides/metabolism , Platelet Adhesiveness , Receptors, Complement/administration & dosage , Receptors, Complement/metabolism , Reperfusion Injury/drug therapy , Reperfusion Injury/immunology , Reperfusion Injury/metabolism , Selectins/metabolism , Sialyl Lewis X Antigen , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...