Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
1.
Int J Biol Macromol ; 260(Pt 2): 129357, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38216011

ABSTRACT

Osteoporosis is a prevalent systemic skeletal disorder, particularly affecting postmenopausal women, primarily due to excessive production and activation of osteoclasts. However, the current anti-osteoporotic drugs utilized in clinical practice may lead to certain side effects. Therefore, it is necessary to further unravel the potential mechanisms regulating the osteoclast differentiation and to identify novel targets for osteoporosis treatment. This study revealed the most significant decline in VSIG4 expression among the VSIG family members. VSIG4 overexpression significantly inhibited RANKL-induced osteoclastogenesis and bone resorption function. Mechanistically, both western blot and immunofluorescence assay results demonstrated that VSIG4 overexpression attenuated the expression of osteoclast marker genes and dampened the activation of MAPK and NF-κB signaling pathways. Furthermore, VSIG4 overexpression could inhibit the generation of reactive oxygen species (ROS) and stimulate the expression of Nrf2 along with its downstream antioxidant enzymes via interaction with Keap1. Notably, a potent Nrf2 inhibitor, ML385, could reverse the inhibitory effect of VSIG4 on osteoclast differentiation. In line with these findings, VSIG4 overexpression also mitigated bone loss induced by OVX and attenuated the activation of osteoclasts in vivo. In conclusion, our results suggest that VSIG4 holds promise as a novel target for addressing postmenopausal osteoporosis. This is achieved by suppressing osteoclast formation via enhancing Nrf2-dependent antioxidant response against reactive oxygen species production.


Subject(s)
Osteogenesis , Osteoporosis , Female , Humans , Reactive Oxygen Species/metabolism , Antioxidants/metabolism , Kelch-Like ECH-Associated Protein 1/metabolism , NF-E2-Related Factor 2/metabolism , Osteoclasts , NF-kappa B/metabolism , Osteoporosis/drug therapy , Osteoporosis/metabolism , Cell Differentiation , Receptors, Complement/metabolism , Receptors, Complement/therapeutic use
2.
Curr Opin Nephrol Hypertens ; 26(2): 123-128, 2017 03.
Article in English | MEDLINE | ID: mdl-27977428

ABSTRACT

PURPOSE OF REVIEW: The complement system represents one of the more primitive forms of innate immunity. It has increasingly been found to contribute to pathologies in the native and transplanted kidney. We provide a concise review of the physiology of the complement cascade, and discuss current and upcoming complement-based therapies. RECENT FINDINGS: Current agents in clinical use either bind to complement components directly or prevent complement from binding to antibodies affixed to the endothelial surface. These include C1 esterase inhibitors, anti-C5 mAbs, anti-CD20 mAbs, and proteasome inhibitors. Treatment continues to show efficacy in the atypical hemolytic uremic syndrome and antibody-mediated rejection. Promising agents not currently available include CCX168, TP10, AMY-101, factor D inhibitors, coversin, and compstatin. Several new trials are targeting complement inhibition to treat antineutrophilic cystoplasmic antibody (ANCA)-associated vasculitis, C3 glomerulopathy, thrombotic microangiopathy, and IgA nephropathy. New agents for the treatment of the atypical hemolytic uremic syndrome are also in development. SUMMARY: Complement-based therapies are being considered for targeted therapy in the atypical hemolytic uremic syndrome and antibody-mediated rejection, C3 glomerulopathy, and ANCA-associated vasculitis. A few agents are currently in use as orphan drugs. A number of other drugs are in clinical trials and, overall, are showing promising preliminary results.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Complement System Proteins/metabolism , Aniline Compounds/therapeutic use , Anti-Neutrophil Cytoplasmic Antibody-Associated Vasculitis/drug therapy , Antigens, CD20/immunology , Atypical Hemolytic Uremic Syndrome/drug therapy , Complement Activation/drug effects , Complement C5/antagonists & inhibitors , Complement C5/immunology , Complement Factor D/antagonists & inhibitors , Glomerulonephritis, IGA/drug therapy , Humans , Nipecotic Acids/therapeutic use , Peptides, Cyclic/therapeutic use , Proteasome Inhibitors/therapeutic use , Receptors, Complement/therapeutic use , Thrombotic Microangiopathies/drug therapy
4.
Circulation ; 131(13): 1171-80, 2015 Mar 31.
Article in English | MEDLINE | ID: mdl-25825397

ABSTRACT

BACKGROUND: Natural IgM antibodies represent a class of innate pattern recognition receptors that recognize danger-associated molecular patterns expressed on stressed or dying cells. They play important roles in tissue homeostasis by disposing of prenecrotic cells and suppressing inflammation. However, ischemic insult leads to a pathogenic level of IgM binding and complement activation, resulting in inflammation and injury. We investigate the role of self-reactive IgM in the unique setting of transplantation where the donor organ undergoes both cold and warm ischemia and global ischemic insult. METHODS AND RESULTS: By transplanting hearts from wild-type donor mice into antibody-deficient mice reconstituted with specific self-reactive IgM monoclonal antibodies, we identified neoepitopes expressed after transplantation and demonstrated a key role for IgM recognition of these epitopes in graft injury. With this information, we developed and characterized a therapeutic strategy that exploited the postischemia recognition system of natural antibodies. On the basis of neoepitope identification, we constructed an anti-annexin IV single-chain antibody (scFv) and an scFv linked to Crry, an inhibitor of C3 activation (scFv-Crry). In an allograft transplantation model in which recipients contain a full natural antibody repertoire, both constructs blocked graft IgM binding and complement activation and significantly reduced graft inflammation and injury. Furthermore, scFv-Crry specifically targeted to the transplanted heart and, unlike complement deficiency, did not affect immunity to infection, an important consideration for immunosuppressed transplant recipients. CONCLUSIONS: We identified pathophysiologically important epitopes expressed within the heart after transplantation and described a novel translatable strategy for targeted complement inhibition that has several advantages over currently available approaches.


Subject(s)
Antibodies, Bispecific/therapeutic use , Antibodies, Monoclonal/therapeutic use , Heart Transplantation/adverse effects , Immunoglobulin M/therapeutic use , Myocardial Reperfusion Injury/prevention & control , Myocardial Reperfusion/adverse effects , Receptors, Complement/therapeutic use , Self Tolerance/immunology , Single-Chain Antibodies/therapeutic use , Animals , Annexin A4/immunology , Antibodies, Bispecific/genetics , Antibodies, Bispecific/immunology , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/immunology , Complement Activation , Epitopes/immunology , Genes, Synthetic , Homeodomain Proteins/genetics , Immunoglobulin M/deficiency , Immunoglobulin M/genetics , Immunoglobulin M/immunology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Myocardial Reperfusion Injury/immunology , Myocardium/immunology , Organ Specificity , Phospholipids/immunology , Receptors, Complement/genetics , Receptors, Complement 3b , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/therapeutic use , Single-Chain Antibodies/genetics , Single-Chain Antibodies/immunology , Transplantation Tolerance
5.
Neurol Res ; 35(9): 976-83, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23815859

ABSTRACT

OBJECTIVE: Complement receptor type 1 (CR1), one of the most potent inhibitors in complement activation, shows a protective effect on cerebral ischemia/reperfusion (CI/R) injury due to its ability to bind C3b and C4b and to inactivate C3/C5 convertases. So far, no study assessed the effect of the first three short consensus repeats (SCR1-3) with low molecular weight, one of the most active functional domains of CR1, binding C4b with a powerful decay-acceleration effect on classical and alternative C3/C5 convertases pathways. Therefore, we aim to assess this effect on CI/R injury in the present study. METHODS: Seventy-five adult male Sprague-Dawley rats were randomly divided into three groups: sham operation group (n = 15), CI/R group (n = 30), and CI/R group treated with CR1-SCR1-3 protein (n = 30). After middle cerebral artery occlusion (MCAO) for 1 hour and reperfusion for 24 hours, neurological motor deficits, cerebral infarct size, and biochemical parameters including myeloperoxidase (MPO), malondialdehyde (MDA), and superoxide dismutase (SOD) were assessed. Meanwhile, tissues in cerebral cortex were collected and processed for western blotting, immunohistochemistry, and HE staining. RESULTS: CR1-SCR1-3 could improve neurological functions in brain with a 26.8% decrease in neurological motor deficit score and could lead to a 63.8% reduction in cerebral infarct size. Besides, pretreatment using CR1-SCR1-3 could prevent neutrophil infiltration and alleviate inflammation severity and subsequent tissue damage. Decreased C4b expression and action, as well as improved morphological changes, were also observed in cerebral tissues of CI/R+CR1-SCR1-3 rats. CONCLUSION: CR1-SCR1-3 protein could possess a neuroprotective effect on acute CI/R injury.


Subject(s)
Infarction, Middle Cerebral Artery/drug therapy , Neuroprotective Agents/therapeutic use , Receptors, Complement/chemistry , Receptors, Complement/therapeutic use , Reperfusion Injury/drug therapy , Animals , Blotting, Western , Brain/drug effects , Brain/metabolism , Brain/pathology , Complement C4b/metabolism , Consensus Sequence , Immunohistochemistry , Infarction, Middle Cerebral Artery/metabolism , Infarction, Middle Cerebral Artery/pathology , Male , Movement Disorders/drug therapy , Movement Disorders/metabolism , Movement Disorders/pathology , Neuroprotective Agents/chemistry , Neuroprotective Agents/metabolism , Neutrophils/drug effects , Neutrophils/physiology , Protein Interaction Domains and Motifs , Random Allocation , Rats , Rats, Sprague-Dawley , Receptors, Complement/isolation & purification , Receptors, Complement/metabolism , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Severity of Illness Index
6.
Neurol Res ; 33(8): 866-74, 2011 Oct.
Article in English | MEDLINE | ID: mdl-22004711

ABSTRACT

OBJECTIVES: Soluble complement receptor type 1 (sCR1), a potent inhibitor of complement activation, has been shown to protect brain cells against cerebral ischemic/reperfusion (CI/R) injury due to its decay-accelerating activity for C3/C5 convertase and co-factor activity for C3b/C4b degradation. However, the effect of short consensus repeats (SCRs) 15-18, one of active domains of sCR1 with high C3b/C4b degradability, has not been demonstrated. Here, we investigated the protective effect of recombinant SCR(15-18) protein in middle cerebral artery occlusion (MCAO)-induced focal CI/R injury. METHODS: Recombinant SCR(15-18) protein was successfully expressed in Escherichia coli and refolded to its optimal bioactivity. Seventy-five Sprague-Dawley rats were randomly assigned into three groups: sham-operated group, CI/R group, and SCR(15-18)+CI/R group pretreated with 20 mg/kg SCR(15-18) protein. After 2 hours of MCAO and subsequent 24 hours of reperfusion, rats were evaluated for neurological deficits and cerebral infarction. Polymorphonuclear leukocyte accumulation, C3b deposition, and morphological changes in cerebral tissue were also estimated. RESULTS: SCR(15-18) pretreatment induced a 20% reduction of infarct size and an improvement of neurological function with 22·2% decrease of neurological deficit scores. Inhibition of cerebral neutrophils infiltration by SCR(15-18) was indicated from the reduction of myeloperoxidase activity in SCR(15-18)+CI/R rats. Decreased C3b deposition and improved morphological changes were also found in cerebral tissue of SCR(15-18)-treated rats. DISCUSSION: Our studies suggest a definitive moderately protective effect of SCR(15-18) against CI/R damage and provide preclinical experimental evidence supporting the possibility of using it as a small anti-complement therapeutic agent for CI/R injury therapy.


Subject(s)
Cerebral Infarction/drug therapy , Complement Activation/drug effects , DNA-(Apurinic or Apyrimidinic Site) Lyase/therapeutic use , Infarction, Middle Cerebral Artery/drug therapy , Neuroprotective Agents/therapeutic use , Peptide Fragments/therapeutic use , Receptors, Complement/therapeutic use , Reperfusion Injury/drug therapy , Animals , Brain/drug effects , Brain/metabolism , Brain/pathology , Cerebral Infarction/pathology , Complement C3b/metabolism , Disease Models, Animal , Humans , Infarction, Middle Cerebral Artery/metabolism , Male , Neuroprotective Agents/pharmacology , Neutrophils , Peptide Fragments/pharmacology , Rats , Recombinant Proteins/pharmacology , Recombinant Proteins/therapeutic use , Reperfusion Injury/metabolism
7.
J Surg Res ; 167(2): e131-6, 2011 May 15.
Article in English | MEDLINE | ID: mdl-19691988

ABSTRACT

Complement activation has been demonstrated to contribute significantly to the expression of IR-induced tissue damage. Each of the three complement pathways, classic, alternative, and lectin, has been implicated in the instigation of tissue pathology. In this study, we used a selective inhibitor of the alternative pathway, that is, a soluble form of complement receptor of the immunoglobulin superfamily (CRIg-Fc) to determine whether it can prevent IR tissue injury. We demonstrate that treatment of C57B1/6 mice prior to mesenteric IR prevents local (intestinal) and remote (lung) injury by limiting deposition of complement and entry of polymorphonuclear cells to the sites of injury. Our results show that CRIg-Fc represents a candidate to limit IR injury as it occurs in various clinical conditions.


Subject(s)
Complement Pathway, Alternative/physiology , Immunoglobulin Fc Fragments/therapeutic use , Intestine, Small/blood supply , Intestine, Small/physiopathology , Receptors, Complement/therapeutic use , Reperfusion Injury/prevention & control , Acute Lung Injury/pathology , Acute Lung Injury/physiopathology , Acute Lung Injury/prevention & control , Animals , B-Lymphocytes/pathology , Cell Movement/drug effects , Complement Pathway, Alternative/drug effects , Immunoglobulin Fc Fragments/pharmacology , Intestine, Small/pathology , Mesentery/blood supply , Mesentery/pathology , Mesentery/physiopathology , Mice , Mice, Inbred C57BL , Models, Animal , Neutrophil Infiltration/drug effects , Recombinant Fusion Proteins/pharmacology , Recombinant Fusion Proteins/therapeutic use , Reperfusion Injury/pathology , Reperfusion Injury/physiopathology , T-Lymphocytes/pathology
9.
Virol J ; 7: 30, 2010 Feb 09.
Article in English | MEDLINE | ID: mdl-20144216

ABSTRACT

BACKGROUND: Influenza is a respiratory disease that seriously threatens human health. In fact, influenza virus itself does not make critical contribution to mortality induced by influenza, but "cytokine storm" produced by the excessive immune response triggered by the virus can result in inflammatory reaction of lung tissues and fatal lung tissue injury, and thus increase influenza mortality. Therefore, besides antiviral drugs, immunosuppression drugs should also be included in infection treatment. PRESENTATION OF THE HYPOTHESIS: Complement is the center of inflammatory reaction. If complement system is over activated, the body will have strong inflammatory reaction or tissue injury, resulting in pathological process. Many studies have proved that, inflammatory injury of lung tissues caused by influenza virus is closely related to complement activation. Therefore, inhibiting complement activation can significantly reduce inflammatory injury in lung tissues. As complement is both a physiological defense and pathological damage medium, systematic inhibition may result in side effects including infection. Therefore, we design targeting complement inhibitors for complement activation sites, i.e. with CR2 as targeting vector, complement inhibitors like CD59 and Crry are targeted to inflammatory sites to specially inhibit the complement activation in local injury, thus local inflammatory reaction is inhibited. TESTING THE HYPOTHESIS: CR2-CD59 and CR2-Crry targeting complement inhibitors are fusion-expressed, and their biological activity is examined via in vivo and in vitro tests. CR2 targeting complement inhibitors are used to treat mouse influenza viral pneumonia model, with PBS treatment group as the control. The survival and lung tissue injury of the mice is observed and the effect of CR2 targeting complement inhibitors on pneumonia induced by influenza virus is evaluated. IMPLICATIONS OF THE HYPOTHESIS: CR2 targeting complement inhibitors are expected to be ideal drugs for viral pneumonia.


Subject(s)
Immunosuppressive Agents/therapeutic use , Influenza, Human/immunology , Influenza, Human/pathology , Lung/pathology , Orthomyxoviridae/immunology , Receptors, Complement 3d/antagonists & inhibitors , Animals , Biological Products/genetics , Biological Products/therapeutic use , CD59 Antigens/genetics , CD59 Antigens/therapeutic use , Humans , Influenza, Human/drug therapy , Lung/virology , Mice , Mice, Inbred BALB C , Orthomyxoviridae Infections/drug therapy , Orthomyxoviridae Infections/pathology , Receptors, Complement/genetics , Receptors, Complement/therapeutic use , Receptors, Complement 3b , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/therapeutic use , Survival Analysis
10.
Spinal Cord ; 48(2): 105-11, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19736560

ABSTRACT

STUDY DESIGN: Spinal cord injured rat model, treated with soluble complement receptor type 1 (sCR1). SETTING: Experimental Animal Department of China Medical University, Shenyang, China. OBJECTIVES: Soluble CR1 is a powerful inhibitor of complement activation. In this study, we investigate the effectiveness of sCR1 on spinal cord injury (SCI) in rats. METHODS: Spinal cord injury was induced in Sprague-Dawley rats. Three experimental groups were examined; the sCR1 group was administered sCR1 at 1 h after the SCI, whereas the control group was administered saline at 1 h after SCI and the sham group underwent a sham operation without SCI or administration. The expressions of C9 and CD59 in the injured spinal cords were evaluated by immunohistochemistry, and numbers of positive cells counted. Furthermore, myeloperoxidase (MPO) activity and motor function were evaluated in each group. RESULTS: At all postoperative time points, the numbers of C9- and CD59-positive cells in the sCR1 group were reduced compared with the control group and MPO activity was significantly decreased compared with both other groups. Moreover, the Basso, Beattie and Bresnahan score for the sCR1 group was significantly improved as compared with that of the control group after 7 days postoperatively. CONCLUSION: Soluble CR1 decreases inflammation reactions by inhibiting activation of the complement system and improves motor function after acute SCI.


Subject(s)
Complement Activation/physiology , Motor Neurons/physiology , Receptors, Complement/therapeutic use , Spinal Cord Injuries/drug therapy , Spinal Cord Injuries/physiopathology , Animals , CD59 Antigens/physiology , Complement C9/physiology , Models, Animal , Motor Neurons/pathology , Peroxidase/physiology , Rats , Rats, Sprague-Dawley , Recovery of Function/physiology , Spinal Cord Injuries/pathology , Treatment Outcome
11.
Neurobiol Dis ; 34(3): 450-6, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19281846

ABSTRACT

Decreased activity of cAMP responsive element-binding protein (CREB) is thought to contribute to the death of striatal medium spiny neurons in Huntington's disease (HD). Therefore, therapies that increase levels of activated CREB, may be effective in fighting neurodegeneration in HD. In this study, we sought to determine whether the phosphodiesterase type 10 (PDE10A) inhibitor TP10 exerts a neuroprotective effect in an excitotoxic model of HD. Rats were surgically administered with quinolinic acid into striatum and subsequently treated with TP10 daily for two or eight weeks. After 2 weeks of TP10 treatment, striatal lesion size was 52% smaller and the surviving cell number was several times higher than in the vehicle-treated group. These beneficial effects of TP10 were maintained through 8 weeks. TP10 treatment also increased significantly the levels of activated CREB in the striatal spiny neurons, which is hypothesized to be a contributing mechanism for the neuroprotective effect. Our findings suggest PDE10A inhibition as a novel neuroprotective approach to the treatment of HD and confirm the importance of phosphodiesterase inhibition in fighting the disease.


Subject(s)
Huntington Disease/chemically induced , Huntington Disease/drug therapy , Neuroprotective Agents/therapeutic use , Phosphoric Diester Hydrolases/metabolism , Receptors, Complement/therapeutic use , Animals , Brain-Derived Neurotrophic Factor/metabolism , Cell Survival/drug effects , Cerebral Cortex/drug effects , Cerebral Cortex/pathology , Corpus Striatum/drug effects , Corpus Striatum/pathology , Cyclic AMP Response Element-Binding Protein/metabolism , Disease Models, Animal , Enzyme Inhibitors/therapeutic use , Huntington Disease/pathology , Male , Neurons/drug effects , Neurons/physiology , Phosphorylation , Quinolinic Acid , Rats , Rats, Wistar
12.
Am J Pathol ; 172(4): 1043-52, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18349134

ABSTRACT

Complement activation is a crucial early event in Wallerian degeneration. In this study we show that treatment of rats with soluble complement receptor 1 (sCR1), an inhibitor of all complement pathways, blocked both systemic and local complement activation after crush injury of the sciatic nerve. Deposition of membrane attack complex (MAC) in the nerve was inhibited, the nerve was protected from axonal and myelin breakdown at 3 days after injury, and macrophage infiltration and activation was strongly reduced. We show that both classical and alternative complement pathways are activated after acute nerve trauma. Inhibition of the classical pathway by C1 inhibitor (Cetor) diminished, but did not completely block, MAC deposition in the injured nerve, blocked myelin breakdown, inhibited macrophage infiltration, and prevented macrophage activation at 3 days after injury. However, in contrast to sCR1 treatment, early signs of axonal degradation were visible in the nerve, linking MAC deposition to axonal damage. We conclude that sCR1 protects the nerve from early axon loss after injury and propose complement inhibition as a potential therapy for the treatment of diseases in which axon loss is the main cause of disabilities.


Subject(s)
Axons/drug effects , Axons/pathology , Neuroprotective Agents/pharmacology , Peripheral Nerves/drug effects , Peripheral Nerves/pathology , Receptors, Complement/therapeutic use , Animals , Complement Activation/drug effects , Complement Pathway, Alternative/drug effects , Humans , Macrophage Activation/drug effects , Macrophages/drug effects , Macrophages/pathology , Male , Myelin Sheath/metabolism , Nerve Crush , Peripheral Nerves/ultrastructure , Rats , Receptors, Complement/metabolism , Wallerian Degeneration/pathology
13.
Cardiovasc Res ; 76(3): 482-93, 2007 Dec 01.
Article in English | MEDLINE | ID: mdl-17825275

ABSTRACT

OBJECTIVES: Membrane-targeted application of complement inhibitors may ameliorate ischemia/reperfusion (I/R) injury by directly targeting damaged cells. We investigated whether Mirococept, a membrane-targeted, myristoylated peptidyl construct derived from complement receptor 1 (CR1) could attenuate I/R injury following acute myocardial infarction in pigs. METHODS: In a closed-chest pig model of acute myocardial infarction, Mirococept, the non-tailed derivative APT154, or vehicle was administered intracoronarily into the area at risk 5 min pre-reperfusion. Infarct size, cardiac function and inflammatory status were evaluated. RESULTS: Mirococept targeted damaged vasculature and myocardium, significantly decreasing infarct size compared to vehicle, whereas APT154 had no effect. Cardioprotection correlated with reduced serum troponin I and was paralleled by attenuated local myocardial complement deposition and tissue factor expression. Myocardial apoptosis (TUNEL-positivity) was also reduced with the use of Mirococept. Local modulation of the pro-inflammatory and pro-coagulant phenotype translated to improved left ventricular end-diastolic pressure, ejection fraction and regional wall motion post-reperfusion. CONCLUSIONS: Local modification of a pro-inflammatory and pro-coagulant environment after regional I/R injury by site-specific application of a membrane-targeted complement regulatory protein may offer novel possibilities and insights into potential treatment strategies of reperfusion-induced injury.


Subject(s)
Cell Membrane/physiology , Myocardial Reperfusion Injury/prevention & control , Peptide Fragments/therapeutic use , Receptors, Complement/therapeutic use , Animals , Apoptosis/physiology , Cell Membrane/drug effects , Disease Models, Animal , Dose-Response Relationship, Drug , Heart Rate/physiology , Myocardial Infarction/pathology , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/physiopathology , Peptide Fragments/pharmacology , Stroke Volume/physiology , Swine , Troponin I/blood , Ventricular Function, Left/physiology
14.
Circulation ; 116(11 Suppl): I83-8, 2007 Sep 11.
Article in English | MEDLINE | ID: mdl-17846331

ABSTRACT

BACKGROUND: TP10, a potent inhibitor of complement activation during cardiopulmonary bypass (CPB) has been shown to significantly reduce the incidence of death and myocardial infarction (MI) in high-risk male patients undergoing cardiac surgery. However, the effect of TP10 in females was undefined because of the limited number of females studied. To examine the possibility of a gender effect, this phase 2 multi-center trial was undertaken to determine whether TP10 would also limit ischemic damage in a larger sample size of high-risk females undergoing cardiac surgery on cardiopulmonary bypass (CPB). METHODS AND RESULTS: This prospective, double-blind, placebo-controlled, multi-center trial involved 297 high-risk (urgent surgery, CABG + Valve, reoperations, ejection fraction <30%) female patients randomized to receive a 5 mg/kg dose of TP10 (n=150) or placebo (n=147) as a 30-minute intravenous infusion before surgery. The primary end point was the incidence of death or MI at 28 days after surgery. Complement activation was assessed by levels of CH50 and SC5b-9 during and after CPB. TP10 was well tolerated and there were no differences in the safety profiles of the 2 groups. Although TP10 effectively suppressed complement activation (at 2 hours after CPB CH50 (mean+SD % change from baseline) 50+/-17% placebo versus 4+/-14% TP10; P=0.0001; SC5b-9 (ng/mL) 917+/-1067 placebo versus 204+/-79 TP10; P=0.0001), there was no difference in the primary end point between the groups (17% placebo versus 21% TP10; P=0.2550). CONCLUSIONS: The benefits of TP10 appear to be gender-related. and mechanisms other than complement activation may be responsible for myocardial injury in high-risk female patients during cardiac surgery on CPB.


Subject(s)
Cardiac Surgical Procedures , Complement System Proteins/metabolism , Receptors, Complement/therapeutic use , Sex Characteristics , Aged , Cardiac Surgical Procedures/methods , Double-Blind Method , Female , Humans , Male , Middle Aged
15.
Nat Clin Pract Nephrol ; 3(5): 277-86, 2007 May.
Article in English | MEDLINE | ID: mdl-17457361

ABSTRACT

The fact that the complement system is activated during immune-complex glomerular disease has been known for nearly 50 years. Detection of complement deposition in the glomerulus using immunochemistry has become an important element of the histological analysis of renal biopsies, and is key to the diagnosis of many types of glomerulonephritis. In recent years it has become evident that complement activation is involved in the pathogenesis of other types of renal disease; complement activation is implicated in transplant injury, atypical hemolytic uremic syndrome and progressive tubulointerstitial fibrosis. Emergence of this evidence has provided insight into how these diseases develop, and has yielded useful diagnostic tools and potential targets for therapeutic intervention. Clinicians have, by using plasma-based therapies, unknowingly treated abnormalities of the complement system in renal patients for many years. Advances in antibody and protein technologies have led to the development of complement inhibitors that have been used in phase III clinical studies. More-specific agents and applications are likely to be developed over the coming years and are discussed in this Review.


Subject(s)
Complement Activation , Complement System Proteins/immunology , Kidney Diseases/immunology , Animals , Complement System Proteins/genetics , Humans , Kidney Diseases/genetics , Kidney Transplantation , Receptors, Complement/therapeutic use
16.
J Am Soc Nephrol ; 18(5): 1508-15, 2007 May.
Article in English | MEDLINE | ID: mdl-17389734

ABSTRACT

Renal fibrosis is the final common pathway of most progressive renal diseases. C5 was recently identified as a risk factor for liver fibrosis. This study investigated the role of C5 in the development of renal tubulointerstitial fibrosis by (1) induction of renal fibrosis in wild-type and C5(-/-) mice by unilateral ureteral ligation (UUO) and (2) investigation of the effects of a C5a receptor antagonist (C5aRA) in UUO. In C5(-/-) mice, when compared with wild-type controls, markers of renal fibrosis (Sirius Red, type I collagen, fibronectin, alpha-smooth muscle actin, vimentin, and infiltrating macrophages) were significantly reduced on day 5 of UUO. On day 10, fibronectin mRNA and protein expression were still reduced in the C5(-/-) mice. Cortical mRNA of all PDGF isoforms and of TGF-beta(1) (i.e., central mediators of renal disease) were significantly reduced in C5(-/-) mice when compared with controls. Renal tubular cell expression of the C5aR was sparse in normal cortex but markedly upregulated after UUO. Treatment of wild-type UUO mice with C5aRA also led to a significant reduction of cortical Sirius Red staining, fibronectin protein expression, and PDGF-B mRNA expression on day 5. Neither genetic C5 deficiency nor C5aRA treatment caused any histologic changes in the nonobstructed kidneys. In cultured murine cortical tubular cells, C5a stimulated production of TGF-beta(1), and this was inhibited by C5aRA. Using a combined genetic and pharmacologic approach, C5, in particular C5a, is identified as a novel profibrotic factor in renal disease and as a potential new therapeutic target.


Subject(s)
Complement C5/genetics , Disease Models, Animal , Kidney Diseases/genetics , Kidney Diseases/pathology , Kidney/pathology , Animals , Cells, Cultured , Complement C5a/genetics , Complement C5a/metabolism , Fibrosis , Gene Expression Regulation , Humans , Kidney Diseases/drug therapy , Kidney Diseases/metabolism , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Platelet-Derived Growth Factor/genetics , Platelet-Derived Growth Factor/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism , Receptors, Complement/therapeutic use , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism , Ureteral Obstruction/drug therapy , Ureteral Obstruction/genetics , Ureteral Obstruction/pathology , Wound Healing/drug effects
17.
Zhonghua Xin Xue Guan Bing Za Zhi ; 35(11): 1037-40, 2007 Nov.
Article in Chinese | MEDLINE | ID: mdl-18269827

ABSTRACT

OBJECTIVE: To investigate the protective effects of recombinant SCR15-18 domain of human soluble complement receptor type 1 (sCR1-SCR-15-18) in rats underwent myocardial ischemia and reperfusion (I/R). METHODS: Sprague-Dawley rats were randomly divided into three groups (n = 12 each group): sham (SO); 30 min ischemia/3h reperfusion (I/R) and I/R plus sCR1-SCR15-18 (15 mg/kg before I/R, sCR1). Serum LDH, CK and cardiac myeloperoxidase (MPO) activity were measured. Infarct size, myocardial histopathological changes and myocardial C3c were also compared among groups. RESULTS: Infarct size [(16.1 +/- 3.3)% vs. (22.9 +/- 3.0)%, infarct zone/left ventricular mass, P < 0. 05] and CK [(2532.5 +/- 597.1) U/L vs. (3400.9 +/- 534.9) U/L, P < 0. 05] and LDH [(5436.2 +/- 611.3) U/L vs. (6572.0 +/- 476.3) U/L, P < 0. 05] as well as MPO activity in infarct zone [(0.81 +/- 0.14) U/g vs. (1.12 +/- 0.13) U/g, P < 0.05] were significantly decreased post sCR1 compared to I/R group. sCR1 also significantly attenuated histological myocardial injury and reduced the deposition of C3c in infarct zone. CONCLUSION: sCR1-SCR15-18 protein exerts cardioprotective effects in this rat I/R model.


Subject(s)
Myocardial Reperfusion Injury/prevention & control , Receptors, Complement/therapeutic use , Animals , Creatine Kinase/metabolism , Disease Models, Animal , Humans , L-Lactate Dehydrogenase/metabolism , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Peroxidase/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Complement 3b/metabolism , Recombinant Proteins/therapeutic use
18.
Expert Rev Cardiovasc Ther ; 4(5): 649-54, 2006 Sep.
Article in English | MEDLINE | ID: mdl-17081086

ABSTRACT

Cardiopulmonary bypass (CPB) for cardiac surgery or lung transplantation initiates a systemic inflammatory response characterized by increased vascular permeability, generalized edema, abnormal lung function and oxygenation and impaired ventricular function. This post-CPB syndrome significantly contributes to postoperative morbidity and mortality. Activation of complement during CPB is a key component that initiates and augments this process. TP10, soluble complement receptor 1, is a novel complement inhibitor that is a potent inhibitor of C3 and C5 convertases, blocking activation of the complement cascade at the nexus of all three complement pathways. Recent controlled trials in humans have demonstrated that TP10 effectively inhibits complement activation during CPB. In high-risk adult patients, TP10 decreases the incidence of mortality and myocardial infarction in males but not in females following cardiac surgery. TP10 is also well tolerated and protects vascular function in infants undergoing CPB. In addition, TP10 leads to early extubation in adult lung transplant recipients. TP10 is currently positioned for clinical development in a male-only indication of cardiac surgery on CPB.


Subject(s)
Cardiopulmonary Bypass/adverse effects , Cardiovascular Diseases/etiology , Cardiovascular Diseases/prevention & control , Complement Inactivating Agents/therapeutic use , Receptors, Complement/therapeutic use , Complement Inactivating Agents/adverse effects , Complement Inactivating Agents/pharmacokinetics , Humans , Lung/drug effects , Lung/physiopathology , Lung Transplantation , Treatment Outcome
19.
J Thorac Cardiovasc Surg ; 129(2): 423-8, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15678055

ABSTRACT

OBJECTIVE: Complement activation has been shown to play a significant role in ischemia-reperfusion injury after lung transplantation. TP-10 (soluble complement receptor 1 inhibitor) inhibits the activation of complement by inactivating C3a and C5a convertases. This was a clinical trial of TP-10 to reduce ischemia-reperfusion injury in lung transplantation. METHODS: In a randomized, double-blinded, multicenter, placebo-controlled trial, 59 patients from four lung transplant programs received TP-10 (10 mg/kg, n = 28) or placebo (n = 31) before reperfusion. This dose achieved 90% complement inhibition for 24 hours, and activity had returned toward normal by 72 hours. RESULTS: At 24 hours, 14 of 28 patients in the TP-10 group (50%) were extubated, whereas only 6 of 31 patients in the placebo group (19%) were (P = .01). The total times on the ventilator and in the intensive care unit both tended to be shorter in the TP-10 group, but these differences did not achieve statistical significance. Among patients requiring cardiopulmonary bypass (n = 5 in placebo group and n = 7 in TP-10 group), the mean duration of mechanical ventilation was reduced by 11 days in the TP-10 group (10.6 +/- 5.0 days vs 21.5 +/- 5.9 days in placebo group, P = .2). Operative deaths, incidences of infection and rejection, and length of hospital stay were not significantly different between the two groups. CONCLUSIONS: Short-term complement inhibition with TP-10 led to early extubation in a significantly higher proportion of lung transplant recipients. The effect of TP-10 was greater among patients undergoing cardiopulmonary bypass, with a large reduction in ventilator days. Complement inhibition thus significantly decreases the duration of mechanical ventilation and could be useful in improving the outcome of lung transplant recipients.


Subject(s)
Lung Transplantation , Postoperative Complications/etiology , Postoperative Complications/prevention & control , Reperfusion Injury/etiology , Reperfusion Injury/prevention & control , Adolescent , Adult , Aged , Biomarkers/blood , Cardiopulmonary Bypass , Complement Inactivator Proteins/antagonists & inhibitors , Complement Inactivator Proteins/therapeutic use , Complement System Proteins/drug effects , Complement System Proteins/metabolism , Double-Blind Method , Female , Graft Rejection/etiology , Graft Rejection/mortality , Graft Rejection/prevention & control , Humans , Length of Stay , Lung Diseases/mortality , Lung Diseases/surgery , Male , Middle Aged , North America , Oxygen Consumption/drug effects , Postoperative Complications/mortality , Receptors, Complement/antagonists & inhibitors , Receptors, Complement/therapeutic use , Reperfusion Injury/mortality , Respiration, Artificial , Surgical Wound Infection/etiology , Surgical Wound Infection/mortality , Surgical Wound Infection/prevention & control , Survival Analysis , Treatment Outcome
20.
Chin J Traumatol ; 8(1): 49-53, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15676091

ABSTRACT

OBJECTIVE: To determine the effects of recombinant soluble complement receptor type I (sCR1) on the immune inflammatory reaction in acute spinal cord injury tissue of rats and its protective effects. METHODS: SD rat models of acute spinal cord injury were prepared by modified Allen's method. The motor function of the rat lower extremities in sCR1 group and normal saline (NS) group was evaluated by the tiltboard experiment at 12 h, 1 d, 3 d, 7 d, and 14 d. The neutrophil infiltration and C3c positive expression were observed. The myeloperoxidase activity was assessed in the injury tissue at 12 h, 1 d, 3 d, 7 d, and 14 d after injury in the two groups. RESULTS: The motor function of rat in sCR1 group at 3 d, 7 d, and 14 d was obviously better than that in NS group (P<0.01, P<0.01, P<0.01). C3c positive expression in sCR1 group at each time point after injury was obviously less than that in NS group (P<0.01). The myeloperoxidase activity in sCR1 group at each time point after injury was obviously less than that in NS group (P<0.01). CONCLUSIONS: Recombinant soluble complement receptor type I (sCR1) can lessen the immune inflammatory reaction in acute spinal cord injury tissue and relieve secondary spinal cord injury by inhibiting the activation of the complement system.


Subject(s)
Receptors, Complement/therapeutic use , Recombinant Proteins/therapeutic use , Spinal Cord Injuries/drug therapy , Animals , Disease Models, Animal , Immunohistochemistry , Inflammation , Peroxidase/biosynthesis , Random Allocation , Rats , Rats, Sprague-Dawley , Spinal Cord Injuries/enzymology , Spinal Cord Injuries/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...