Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 145
Filter
1.
BMC Cardiovasc Disord ; 24(1): 417, 2024 Aug 10.
Article in English | MEDLINE | ID: mdl-39127656

ABSTRACT

Mutations in fibrillin 1 (FBN1) is the main cause of Marfan syndrome (MFS) with thoracic aortic aneurysm (TAA) as the main complication. Activation of the complement system plays a key role in the formation of thoracic and abdominal aortic aneurysms. However, the role of the complement system in MFS-associated aortic aneurysms remains unclear. In this study, we observed increased levels of complement C3a and C5a in the plasma of MFS patients and mouse, and the increased deposition of the activated complement system product C3b/iC3b was also observed in the elastic fiber rupture zone of 3-month-old MFS mice. The expression of C3a receptor (C3aR) was increased in MFS aortas, and recombinant C3a promoted the expression of cytokines in macrophages. The administration of a C3aR antagonist (C3aRA) attenuated the development of thoracic aortic aneurysms in MFS mice. The increased inflammation response and matrix metalloproteinases activities were also attenuated by C3aRA treatment in MFS mice. Therefore, these findings indicate that the complement C3a/C3aR inhibition alleviates the formation of aortic aneurysm in Marfan syndrome mice.


Subject(s)
Adipokines , Aortic Aneurysm, Thoracic , Complement C3a , Fibrillin-1 , Marfan Syndrome , Receptors, Complement , Animals , Female , Humans , Male , Mice , Adipokines/genetics , Aorta, Thoracic/pathology , Aortic Aneurysm, Thoracic/prevention & control , Aortic Aneurysm, Thoracic/etiology , Aortic Aneurysm, Thoracic/metabolism , Aortic Aneurysm, Thoracic/pathology , Complement C3a/antagonists & inhibitors , Complement C3a/metabolism , Cytokines/metabolism , Disease Models, Animal , Fibrillin-1/genetics , Inflammation Mediators/metabolism , Macrophages/metabolism , Macrophages/drug effects , Marfan Syndrome/complications , Marfan Syndrome/genetics , Marfan Syndrome/drug therapy , Mice, Inbred C57BL , Receptors, Complement/antagonists & inhibitors , Receptors, G-Protein-Coupled , Signal Transduction
2.
BMC Nephrol ; 20(1): 424, 2019 11 21.
Article in English | MEDLINE | ID: mdl-31752725

ABSTRACT

BACKGROUNDS: The aberrant activation of complement system is critically involved in lupus nephropathy. Recent study showed complement C3 inhibitor was effective in the treatment of lupus nephropathy. In this study, we investigate the effect of a novel complement C3 inhibitor, CRIg/FH, in the treatment of lupus nephropathy in MRL/lpr lupus mice. METHODS: We treated MRL/lpr female mice with a dose escalation of CRIg/FH (10, 5 and 2 mg/kg) by intraperitoneal injection twice weekly since 12 weeks age. In addition, MRL/lpr mice treated with intraperitoneal injection of normal saline or oral prednisone, along with C57BL/6 J healthy mice were maintained to serve as controls. We started 8-h urine collection weekly to screen proteinuria by measuring the levels of urine urea/creatinine. Serum samples was collected at week 16 and 20 to measure levels of urea nitrogen, creatinine, and immunological markers (C3, C4, A-ds-DNA) before the mice were sacrificed at 20 weeks age to collect kidneys for histopathological examinations. RESULTS: Overt skin lesions were observed in MRL/lpr mice treated with normal saline, while skin lesion was not observed in CRIg/FH treated MRL/lpr mice. There was no overt proteinuria observed in MRL/lpr mice treated with CRIg/FH. Serum creatinine and BUN levels in MRL/lpr mice was maintained in highest CRIg/FH dose (10 mg/kg twice a week) to be significantly lower than that in prednisone treated MRL/lpr mice at 20 weeks age. In addition, CRIg/FH treatment in MRL/lpr mice results in a significantly elevated serum C3 and C4 levels when compared to prednisone treatment at both 16 and 20 weeks. Furthermore, our study identified that serum level of A-ds-DNA was also significantly lower in CRIg/FH treatment than that in predisone treated MRL/lpr mice. Renal pathology confirmed that kidneys from CRIg/FH treated MRL/lpr mice suffered less from nephritis and complement disposition. CONCLUSION: Our results showed that the complement inhibitor CRIg/FH can protect MRL/lpr mice from lupus nephropathy by preserving renal function and glomerulus complement activation. Our findings support the positive effect of complement inhibitors in the treatment of lupus nephropathy.


Subject(s)
Complement Inactivating Agents/therapeutic use , Lupus Nephritis/drug therapy , Receptors, Complement/antagonists & inhibitors , Animals , Blood Urea Nitrogen , Complement Activation/drug effects , Complement Inactivating Agents/administration & dosage , Creatinine/blood , Female , Glucocorticoids/administration & dosage , Glucocorticoids/therapeutic use , Injections, Intraperitoneal , Lupus Nephritis/blood , Lupus Nephritis/urine , Mice , Mice, Inbred C57BL , Mice, Inbred MRL lpr , Prednisone/administration & dosage , Prednisone/therapeutic use , Proteinuria/drug therapy , Randomized Controlled Trials as Topic
3.
Neural Plast ; 2019: 1071036, 2019.
Article in English | MEDLINE | ID: mdl-31814819

ABSTRACT

Worldwide, millions of individuals suffer an ischemic stroke each year, causing major disability, especially in the elderly, where stroke is the number one cause of disability. However, to date, no effective therapy exists that targets the functional recovery after stroke. After necrosis, neuroinflammation is a common feature of the acute stroke and a major obstacle to tissue restoration. In the lesioned area, the dying neurons release chemotactic signals, such as fractalkine/CX3CL1, which evoke "eat-me" signals that are recognized by microglia expressing complement C3a receptor (C3aR), resulting in phagocytosis of the dying but still viable neurons, known as secondary phagocytosis. Using a mouse model of stroke and two-photon microscopy, we aimed to attenuate poststroke phagocytosis of the dying but still viable neurons by using SB 290157, an antagonist of C3aR. We found that intracortical administration of SB 290157 reduced the number of inflammatory microglial cells expressing ED1 and Iba1 antigens at the lesion site. We could show, in vivo, that two days after a needle-induced cortical lesion there were less microglial cells present around the injury site, displaying less high-order branches and an increase in the lower order ones, suggesting an attenuated phagocytic phenotype in treated animals as compared with controls. We conclude that the C3aR antagonist, SB 290157, may be used in the future to limit the neuronal death by limiting secondary phagocytosis after stroke.


Subject(s)
Arginine/analogs & derivatives , Benzhydryl Compounds/administration & dosage , Microglia/drug effects , Neurons/drug effects , Receptors, Complement/antagonists & inhibitors , Stroke/metabolism , Trifluoroacetic Acid/administration & dosage , Animals , Arginine/administration & dosage , Disease Models, Animal , Mice , Microglia/metabolism , Microglia/pathology , Neurons/metabolism , Neurons/pathology , Phagocytosis/drug effects , Recovery of Function/drug effects , Stroke/pathology
4.
Am J Transplant ; 18(10): 2417-2428, 2018 10.
Article in English | MEDLINE | ID: mdl-29504277

ABSTRACT

Donor brain death (BD) is an inherent part of lung transplantation (LTx) and a key contributor to ischemia-reperfusion injury (IRI). Complement activation occurs as a consequence of BD in other solid organ Tx and exacerbates IRI, but the role of complement in LTx has not been investigated. Here, we investigate the utility of delivering nebulized C3a receptor antagonist (C3aRA) pretransplant to BD donor lungs in order to reduce post-LTx IRI. BD was induced in Balb/c donors, and lungs nebulized with C3aRA or vehicle 30Ā minutes prior to lung procurement. Lungs were then cold stored for 18Ā hours before transplantation into C57Bl/6 recipients. Donor lungs from living donors (LD) were removed and similarly stored. At 6Ā hours and 5Ā days post-LTx, recipients of BD donor lungs had exacerbated IRI and acute rejection (AR), respectively, compared to recipients receiving LD lungs, as determined by increased histopathological injury, immune cells, and cytokine levels. A single pretransplant nebulized dose of C3aRA to the donor significantly reduced IRI as compared to vehicle-treated BD donors, and returned IRI and AR grades to that seen following LD LTx. These data demonstrate a role for complement inhibition in the amelioration of IRI post-LTx in the context of donor BD.


Subject(s)
Brain Death/physiopathology , Graft Rejection/prevention & control , Lung Transplantation/adverse effects , Receptors, Complement/antagonists & inhibitors , Reperfusion Injury/prevention & control , Tissue Donors , Administration, Inhalation , Animals , Graft Rejection/etiology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Reperfusion Injury/etiology
5.
Article in Zh | MEDLINE | ID: mdl-28511297

ABSTRACT

Objective: To explore the effect of complement C3 a-C3a receptor in the kidney immune inju-ry in trichloroethylene-sensitized mice by using C3a receptor specific antagonist C3aRA and discuss the patho-genesis of kidney injury in occupational dermatitis medicamentosa-like of trichloroethylene (ODMLT) . Methods: 42 female 6~8 weeks old BALB/c mice of specific pathogen free were randomly divided into blank control group (5) , solvent control group (5) , TCE treatment group (16) and TCE+C3aRA treatment group (16) . The TCE treat-ment group and TCE+C3aRA treatment group were further divided into the sensitized group and the non-sensi-tized group according to the skin sensitization test score. Renal function was detected by biochemical detection kit; expression of C3aR in kidney tissue was detected by qPCR; expression of IL-1Ɵ and TNF-α protein were de-tected by immunohistochemical. Results: Compared with solvent control group and corresponding non-sensitized group, CRE and BUN in TCE sensitized group and TCE + C3aRA sensitized group were significantly increased (P<0.05) . Compared with TCE sensitized group, CRE and BUN in TCE+C3aRA sensitized group were signifi-cantly decreased (P<0.05) . Compared with solvent control group and TCE non-sensitized group, the expression level of C3aR gene in kidney tissue in TCE sensitized group was significantly increased (P<0.05) . There was a large number of IL-1Ɵ and TNF-α protein expression in kidney tissue in TCE sensitized group and TCE+C3aRA sensitized group. Compared with the TCE sensitized group, the expression level of IL-1Ɵ and TNF-α protein in kidney tissue in TCE+C3aRA sensitized group was significantly decreased (P<0.05) . Conclusion: C3a-C3aR may be involved in the kidney immune injury in TCE sensitized mice, C3aRA has a protective effect on the kid-ney immune injury in TCE sensitized mice.


Subject(s)
Complement C3a/metabolism , Kidney/pathology , Receptors, Complement/metabolism , Trichloroethylene/toxicity , Animals , Female , Kidney/drug effects , Kidney/metabolism , Mice , Mice, Inbred BALB C , Receptors, Complement/antagonists & inhibitors
6.
Biochem Biophys Res Commun ; 476(4): 467-474, 2016 08 05.
Article in English | MEDLINE | ID: mdl-27255994

ABSTRACT

CD93, also known as the complement component C1q receptor (C1qRp), has been reported to promote the progression of some cancer types. However, the expression and physiological significance of CD93 in nasopharyngeal carcinoma (NPC) remain largely elusive. In this study, we first examined the expression of CD93 in NPC and experimentally manipulated its expression. We observed that vascular CD93 expression is elevated in NPC and is correlated with T classification, N classification, distant metastasis, clinical stage and poor prognosis (all PĀ <Ā 0.05). In addition, overexpression of CD93 promoted angiogenesis inĀ vitro. What's more, we found that CD93 was highly expressed in NPC tissues and cells, and the regulation of CD93 on cell proliferation was determined by cell counting kit (CCK)-8 assay and cell cycle analyses. Our findings provide unique insight into the pathogenesis of NPC and underscore the need to explore novel therapeutic targets such as CD93 to improve NPC treatment.


Subject(s)
Membrane Glycoproteins/metabolism , Nasopharyngeal Neoplasms/blood supply , Nasopharyngeal Neoplasms/immunology , Receptors, Complement/metabolism , Carcinoma , Cell Cycle , Cell Line, Tumor , Cell Proliferation , Female , Gene Expression , Gene Knockdown Techniques , Human Umbilical Vein Endothelial Cells , Humans , Immunohistochemistry , Male , Membrane Glycoproteins/antagonists & inhibitors , Membrane Glycoproteins/genetics , Middle Aged , Nasopharyngeal Carcinoma , Nasopharyngeal Neoplasms/pathology , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/immunology , RNA, Small Interfering/genetics , Receptors, Complement/antagonists & inhibitors , Receptors, Complement/genetics
7.
J Biol Chem ; 289(21): 14534-46, 2014 May 23.
Article in English | MEDLINE | ID: mdl-24711457

ABSTRACT

Age-related macular degeneration (AMD) is a complex disease caused by genetic and environmental factors, including genetic variants in complement components and smoking. Smoke exposure leads to oxidative stress, complement activation, endoplasmic reticulum (ER) stress, and lipid dysregulation, which have all been proposed to be associated with AMD pathogenesis. Here we examine the effects of smoke exposure on the retinal pigment epithelium (RPE). Mice were exposed to cigarette smoke or filtered air for 6 months. RPE cells grown as stable monolayers were exposed to 5% cigarette smoke extract (CSE). Effects of smoke were determined by biochemical, molecular, and histological measures. Effects of the alternative pathway (AP) of complement and complement C3a anaphylatoxin receptor signaling were analyzed using knock-out mice or specific inhibitors. ER stress markers were elevated after smoke exposure in RPE of intact mice, which was eliminated in AP-deficient mice. To examine this relationship further, RPE monolayers were exposed to CSE. Short term smoke exposure resulted in production and release of complement C3, the generation of C3a, oxidative stress, complement activation on the cell membrane, and ER stress. Long term exposure to CSE resulted in lipid accumulation, and secretion. All measures were reversed by blocking C3a complement receptor (C3aR), alternative complement pathway signaling, and antioxidant therapy. Taken together, our results provide clear evidence that smoke exposure results in oxidative stress and complement activation via the AP, resulting in ER stress-mediated lipid accumulation, and further suggesting that oxidative stress and complement act synergistically in the pathogenesis of AMD.


Subject(s)
Complement Activation/physiology , Endoplasmic Reticulum Stress/physiology , Lipids/analysis , Oxidative Stress/physiology , Retinal Pigment Epithelium/metabolism , Smoke , Acetylcysteine/pharmacology , Animals , Arginine/analogs & derivatives , Arginine/pharmacology , Benzhydryl Compounds/pharmacology , Blotting, Western , Cells, Cultured , Complement Activation/drug effects , Complement Factor B/genetics , Complement Factor B/metabolism , Complement Pathway, Alternative/drug effects , Complement Pathway, Alternative/physiology , Endoplasmic Reticulum Chaperone BiP , Endoplasmic Reticulum Stress/drug effects , Free Radical Scavengers/pharmacology , Heat-Shock Proteins/genetics , Humans , Lipid Metabolism , Macular Degeneration/genetics , Macular Degeneration/metabolism , Macular Degeneration/physiopathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Nicotine/pharmacology , Oxidative Stress/drug effects , Receptors, Complement/antagonists & inhibitors , Receptors, Complement/metabolism , Retinal Pigment Epithelium/cytology , Retinal Pigment Epithelium/drug effects , Reverse Transcriptase Polymerase Chain Reaction , Nicotiana/chemistry , Transcription Factor CHOP/genetics
8.
J Immunol ; 190(8): 4215-25, 2013 Apr 15.
Article in English | MEDLINE | ID: mdl-23479227

ABSTRACT

During experimental sepsis, excessive generation of the anaphylatoxin C5a results in reduction of the C5a receptor (C5aR) on neutrophils. These events have been shown to result in impaired innate immunity. However, the regulation and fate of C5aR on neutrophils during sepsis are largely unknown. In contrast to 30 healthy volunteers, 60 patients in septic shock presented evidence of complement activation with significantly increased serum levels of C3a, C5a, and C5b-9. In the septic shock group, the corresponding decrease in complement hemolytic activity distinguished survivors from nonsurvivors. Neutrophils from patients in septic shock exhibited decreased C5aR expression, which inversely correlated with serum concentrations of C-reactive protein (CRP) and clinical outcome. In vitro exposure of normal neutrophils to native pentameric CRP led to a dose- and time-dependent loss of C5aR expression on neutrophils, whereas the monomeric form of CRP, as well as various other inflammatory mediators, failed to significantly alter C5aR levels on neutrophils. A circulating form of C5aR (cC5aR) was detected in serum by immunoblotting and a flow-based capture assay, suggestive of an intact C5aR molecule. Levels of cC5aR were significantly enhanced during septic shock, with serum levels directly correlating with lethality. The data suggest that septic shock in humans is associated with extensive complement activation, CRP-dependent loss of C5aR on neutrophils, and appearance of cC5aR in serum, which correlated with a poor outcome. Therefore, cC5aR may represent a new sepsis marker to be considered in tailoring individualized immune-modulating therapy.


Subject(s)
Neutrophils/immunology , Neutrophils/metabolism , Receptors, Complement/blood , Shock, Septic/blood , Shock, Septic/immunology , Adult , Aged , Animals , Female , Humans , Male , Mice , Mice, Inbred C57BL , Rats , Rats, Sprague-Dawley , Receptor, Anaphylatoxin C5a , Receptors, Complement/antagonists & inhibitors , Shock, Septic/mortality , Survival
9.
Adv Exp Med Biol ; 865: 57-74, 2015.
Article in English | MEDLINE | ID: mdl-26306443

ABSTRACT

The complement system is a network of interacting fluid-phase and cell surface-associated molecules that trigger, amplify, and regulate immune and inflammatory signaling pathways. Dysregulation of this finely balanced network can destabilize host-microbe homeostasis and cause inflammatory tissue damage. Evidence from clinical and animal model-based studies suggests that complement is implicated in the pathogenesis of periodontitis, a polymicrobial community-induced chronic inflammatory disease that destroys the tooth-supporting tissues. This review discusses molecular mechanisms of complement involvement in the dysbiotic transformation of the periodontal microbiome and the resulting destructive inflammation, culminating in loss of periodontal bone support. These mechanistic studies have additionally identified potential therapeutic targets. In this regard, interventional studies in preclinical models have provided proof-of-concept for using complement inhibitors for the treatment of human periodontitis.


Subject(s)
Bacteroidaceae Infections/drug therapy , Complement Inactivating Agents/therapeutic use , Complement System Proteins/metabolism , Dysbiosis/drug therapy , Periodontitis/drug therapy , Receptors, Complement/antagonists & inhibitors , Animals , Bacteroidaceae Infections/immunology , Bacteroidaceae Infections/microbiology , Bacteroidaceae Infections/pathology , Complement Activation/drug effects , Disease Models, Animal , Dysbiosis/immunology , Dysbiosis/microbiology , Dysbiosis/pathology , Host-Pathogen Interactions/drug effects , Humans , Macaca fascicularis , Mice , Peptides, Cyclic/therapeutic use , Periodontitis/immunology , Periodontitis/microbiology , Periodontitis/pathology , Porphyromonas gingivalis/immunology , Porphyromonas gingivalis/pathogenicity , Pyridones/therapeutic use , Receptors, Complement/immunology , Receptors, Complement/metabolism
10.
FASEB J ; 27(2): 822-31, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23118029

ABSTRACT

Mammalian survival depends on metabolizing nutrients, storing energy, and combating infection. Complement activation in blood triggers energy-depleting immune responses to fight infections. Here we identify surprising energy-conserving roles for complement proteins C5a and C3a and their receptors, C5aR and C3aR, roles that are contraindicated in complement biology. Rats fed a high-carbohydrate high-fat diet developed obesity, visceral adiposity, adipose inflammation, glucose/insulin intolerance, and cardiovascular dysfunction that correlated with increased plasma C3a, adipose C5aR, and C3aR. These in vivo changes were dramatically attenuated by receptor-selective antagonists of either C5aR (5 mg/kg/d p.o.) or C3aR (30 mg/kg/d p.o.), which both reduced proinflammatory adipokines and altered expression of inflammatory genes in adipose tissue. In vitro C5a and C3a (100 nM) exhibited novel insulin-like effects on 3T3-L1 adipocytes, promoting energy conservation by increasing glucose and fatty acid uptake while inhibiting cAMP signaling and lipolysis, and induced PGE(2) release from macrophages, effects all blocked by each respective antagonist (10 ĀµM). These studies reveal important new links between complement signaling and metabolism, highlight new complement functions on adipocytes and in adipose tissue, demonstrate how aberrant immune responses may exacerbate obesity and metabolic dysfunction, and show that targeting C3aR or C5aR with antagonists is a new strategy for treating metabolic dysfunction.


Subject(s)
Obesity/prevention & control , Receptor, Anaphylatoxin C5a/antagonists & inhibitors , Receptors, Complement/antagonists & inhibitors , 3T3-L1 Cells , Adipocytes/drug effects , Adipocytes/immunology , Adipocytes/metabolism , Animals , Arginine/analogs & derivatives , Arginine/pharmacology , Benzhydryl Compounds/pharmacology , Diet, High-Fat/adverse effects , Dietary Carbohydrates/administration & dosage , Dietary Carbohydrates/adverse effects , Inflammation/immunology , Inflammation/prevention & control , Lipid Metabolism/drug effects , Lipid Metabolism/immunology , Macrophages/drug effects , Macrophages/immunology , Macrophages/metabolism , Male , Metabolic Diseases/etiology , Metabolic Diseases/immunology , Metabolic Diseases/metabolism , Metabolic Diseases/prevention & control , Mice , Obesity/etiology , Obesity/immunology , Obesity/metabolism , Peptides, Cyclic/pharmacology , Rats , Rats, Wistar , Signal Transduction/drug effects , Signal Transduction/immunology
11.
J Immunol ; 189(10): 4797-805, 2012 Nov 15.
Article in English | MEDLINE | ID: mdl-23041570

ABSTRACT

The complement anaphylatoxins C3a, C5a, and desarginated C5a (C5a(desArg)) play critical roles in the induction of inflammation and the modulation of innate and acquired immune responses after binding to their G protein-coupled receptors, C3a receptor and C5a receptor (C5aR). The role of C5a(desArg) in inducing cell activation has been often neglected, because the affinity of C5a(desArg) for C5aR has been reported to be much lower than that of C5a. We have used a novel label-free cellular assay to reassess the potential of C5a(desArg) to induce activation of transfected and primary immune cells. Our results indicate that physiological levels of C5a(desArg) induce significant levels of cell activation that are even higher than those achieved by stimulating cells with analogous concentrations of C5a. Such activation was strictly dependent on C5aR, because it was completely abrogated by PMX-53, a C5aR antagonist. Pharmacological inhibition of specific G proteins located downstream of C5aR indicated differential involvement of G(α) proteins upon C5aR engagement by C5a or C5a(desArg). Further, mass spectrometric characterization of plasma-derived C5a and C5a(desArg) provided important insight into the posttranslational modification pattern of these anaphylatoxins, which includes glycosylation at Asn(64) and partial cysteinylation at Cys(27). Although the context-specific physiological contribution of C5a(desArg) has to be further explored, our data suggest that C5a(desArg) acts as a key molecule in the triggering of local inflammation as well as the maintenance of blood surveillance and homeostatic status.


Subject(s)
Biological Assay/methods , Complement C3/immunology , Complement C5a/immunology , Receptors, Complement/immunology , Animals , Cell Line, Tumor , Complement C3/analysis , Complement C3/genetics , Complement C5a/analysis , Complement C5a/genetics , GTP-Binding Protein alpha Subunits/genetics , GTP-Binding Protein alpha Subunits/immunology , Humans , Peptides, Cyclic/pharmacology , Rats , Receptor, Anaphylatoxin C5a , Receptors, Complement/antagonists & inhibitors , Receptors, Complement/genetics
12.
J Immunol ; 188(11): 5694-705, 2012 Jun 01.
Article in English | MEDLINE | ID: mdl-22539791

ABSTRACT

Allergen-specific IgE plays an essential role in the pathogenesis of allergic asthma. Although there has been increasing evidence suggesting the involvement of IL-17 in the disease, the relationship between IL-17 and IgE-mediated asthmatic responses has not yet been defined. In this study, we attempted to elucidate the contribution of IL-17 to an IgE-mediated late-phase asthmatic response and airway hyperresponsiveness (AHR). BALB/c mice passively sensitized with an OVA-specific IgE mAb were challenged with OVA intratracheally four times. The fourth challenge caused a late-phase increase in airway resistance associated with elevated levels of IL-17(+)CD4(+) cells in the lungs. Multiple treatments with a C3a receptor antagonist or anti-C3a mAb during the challenges inhibited the increase in IL-17(+)CD4(+) cells. Meanwhile, a single treatment with the antagonist or the mAb at the fourth challenge suppressed the late-phase increase in airway resistance, AHR, and infiltration by neutrophils in bronchoalveolar lavage fluid. Because IL-17 production in the lungs was significantly repressed by both treatments, the effect of an anti-IL-17 mAb was examined. The late-phase increase in airway resistance, AHR, and infiltration by neutrophils in bronchoalveolar lavage fluid was inhibited. Furthermore, an anti-Gr-1 mAb had a similar effect. Collectively, we found that IgE mediated the increase of IL-17(+)CD4(+) cells in the lungs caused by repeated Ag challenges via C3a. The mechanisms leading to the IgE-mediated late-phase asthmatic response and AHR are closely associated with neutrophilic inflammation through the production of IL-17 induced by C3a.


Subject(s)
Asthma/immunology , Asthma/pathology , Bronchial Hyperreactivity/immunology , Complement C3a/physiology , Immunoglobulin E/physiology , Interleukin-17/physiology , Neutrophils/immunology , Neutrophils/pathology , Animals , Antibodies, Monoclonal/physiology , Asthma/metabolism , Bronchial Hyperreactivity/pathology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/pathology , Complement C3a/antagonists & inhibitors , Complement C3a/immunology , Inflammation Mediators/antagonists & inhibitors , Inflammation Mediators/immunology , Inflammation Mediators/physiology , Interleukin-17/biosynthesis , Interleukin-17/immunology , Male , Mice , Mice, Inbred BALB C , Receptors, Complement/antagonists & inhibitors , Time Factors
13.
Clin Exp Hypertens ; 36(1): 58-63, 2014.
Article in English | MEDLINE | ID: mdl-23713944

ABSTRACT

Vascular smooth muscle cells (VSMCs) derived from spontaneously hypertensive rats (SHR) show exaggerated growth with a synthetic phenotype and angiotensin II (Ang II) production associated with increased production of complement (C3). We hypothesized that C3 is involved in the growth of mesangial cells (MCs) from hypertensive rats. We examined the effects of a C3a receptor inhibitor on proliferation, phenotype and Ang II generation in MCs from stroke prone-spontaneously hypertensive rats (SHR)-SP, SHR and Wistar-Kyoto (WKY) rats. Expression of C3 and C3a receptor were evaluated by immunohistochemical staining of the renal cortex. We examined the effects of the C3a inhibitor, SB290157, on proliferation, the expression of phenotype-marker mRNAs and Ang II production in cells from SHR-SP, SHR and WKY rats. Immunostaining of C3 was stronger in SHR and SHRSP glomeruli. MCs from SHR-SP and SHR abundantly express pre-pro C3 mRNA. SB290157 significantly inhibited basal DNA synthesis and proliferation of MCs from SHR-SP and SHR. Expression of osteopontin mRNA in MCs from SHR-SP and SHR was decreased with SB290157 treatment, whereas MC basal expression of α-SMA mRNA was decreased. SB290157 significantly decreased the production of Ang II in MCs from SHR-SP and SHR. Endogenous C3a promotes exaggerated growth with a synthetic phenotype and the production of Ang II in MCs from SHR-SP and SHR. The C3 and C3a receptor system may primarily be involved in the pathogenesis of renal remodeling in hypertensive rats.


Subject(s)
Complement C3a/metabolism , Hypertension/metabolism , Mesangial Cells/metabolism , Receptors, Complement/metabolism , Actins/genetics , Angiotensin II/biosynthesis , Animals , Arginine/analogs & derivatives , Arginine/pharmacology , Benzhydryl Compounds/pharmacology , Cell Proliferation/drug effects , Complement C3a/genetics , Gene Expression/drug effects , Hypertension/complications , Hypertension/pathology , Kidney Cortex/metabolism , Kidney Cortex/pathology , Male , Mesangial Cells/drug effects , Mesangial Cells/pathology , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Osteopontin/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Receptors, Complement/antagonists & inhibitors , Stroke/etiology , Stroke/metabolism , Stroke/pathology
14.
Brain Res Bull ; 213: 110986, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38810789

ABSTRACT

Cerebral ischemia-reperfusion injury (CIRI), a prevalent stroke-related complication, can lead to severe brain damage. Inflammation is a crucial factor in CIRI pathogenesis, and the complement component 3a receptor (C3aR) could be a key mediator in the post-CIRI inflammatory cascade. In this study, the role of C3aR in CIRI was investigated utilizing a middle cerebral artery occlusion (MCAO) model in C3aR knockout (KO) mice. Magnetic resonance imaging (MRI) and neurofunctional assessments revealed that C3aR KO mice exhibited significantly diminished cerebral infarction and improved neurological impairments. Consequently, the focus shifted to searching for a small molecule antagonist of C3aR. JR14a, a new potent thiophene antagonist of C3aR, was injected intraperitoneally into mice 1-h post-MCAO model implementation. The mass spectrometry (MS) results indicated the ability of JR14a to penetrate the blood-brain barrier. Subsequent TTC staining and neurofunctional assessments revealed the efficacy of JR14a in reducing cerebral infarct volume and neurological impairment following MCAO. In addition, immunofluorescence (IF) and immunohistochemistry (IHC) demonstrated attenuated microglial activation, neutrophil infiltration, and blood-brain barrier disruption by JR14a in the MCAO model. Furthermore, enzyme-linked immunosorbent assay (ELISA) and Western blotting supported the role of JR14a in downregulating the expression levels of C3aR, tumor necrosis factor-alpha (TNF-α), and interleukin-6 (IL-6), as well as the phosphorylation of p65. In conclusion, the findings suggested that C3aR could be a potential therapeutic target for CIRI, and JR14a emerged as a promising treatment candidate.


Subject(s)
Infarction, Middle Cerebral Artery , Mice, Knockout , Neuroinflammatory Diseases , Reperfusion Injury , Animals , Reperfusion Injury/drug therapy , Reperfusion Injury/metabolism , Infarction, Middle Cerebral Artery/drug therapy , Mice , Male , Neuroinflammatory Diseases/drug therapy , Neuroinflammatory Diseases/metabolism , Mice, Inbred C57BL , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Receptors, Complement/antagonists & inhibitors , Receptors, Complement/metabolism , Brain Ischemia/drug therapy , Brain Ischemia/metabolism , Disease Models, Animal , Microglia/drug effects , Microglia/metabolism , Thiophenes/pharmacology , Thiophenes/therapeutic use , Neuroprotective Agents/pharmacology , Inflammation/drug therapy , Inflammation/metabolism
15.
Adv Exp Med Biol ; 735: 55-81, 2013.
Article in English | MEDLINE | ID: mdl-23402019

ABSTRACT

For the last two decades, there had been remarkable advancement in understanding the role of complement regulatory proteins in autoimmune disorders and importance of complement inhibitors as therapeutics. Systemic lupus erythematosus is a prototype of systemic autoimmune disorders. The disease, though rare, is potentially fatal and afflicts women at their reproductive age. It is a complex disease with multiorgan involvement, and each patient presents with a different set of symptoms. The diagnosis is often difficult and is based on the diagnostic criteria set by the American Rheumatology Association. Presence of antinuclear antibodies and more specifically antidouble-stranded DNA indicates SLE. Since the disease is multifactorial and its phenotypes are highly heterogeneous, there is a need to identify multiple noninvasive biomarkers for SLE. Lack of validated biomarkers for SLE disease activity or response to treatment is a barrier to the efficient management of the disease, drug discovery, as well as development of new therapeutics. Recent studies with gene knockout mice have suggested that membrane-bound complement regulatory proteins (CRPs) may critically determine the sensitivity of host tissues to complement injury in autoimmune and inflammatory disorders. Case-controlled and followup studies carried out in our laboratory suggest an intimate relation between the level of DAF, MCP, CR1, and CD59 transcripts and the disease activity in SLE. Based on comparative evaluation of our data on these four membrane-bound complement regulatory proteins, we envisaged CR1 and MCP transcripts as putative noninvasive disease activity markers and the respective proteins as therapeutic targets for SLE. Following is a brief appraisal on membrane-bound complement regulatory proteins DAF, MCP, CR1, and CD59 as biomarkers and therapeutic targets for SLE.


Subject(s)
Complement System Proteins/drug effects , Complement System Proteins/physiology , Lupus Erythematosus, Systemic/drug therapy , Membrane Proteins/drug effects , Membrane Proteins/physiology , Animals , Biomarkers , CD55 Antigens/physiology , CD59 Antigens/drug effects , CD59 Antigens/physiology , Humans , Membrane Cofactor Protein/antagonists & inhibitors , Membrane Cofactor Protein/physiology , Receptors, Complement/antagonists & inhibitors , Receptors, Complement/physiology
16.
Biol Reprod ; 86(6): 190, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22441801

ABSTRACT

Human preterm and term parturition is associated with inflammatory cascades in the uteroplacental unit. Activation of the complement cascade releases potent proinflammatory mediators, including the anaphylatoxin C5a, which exerts its biological effects through its receptors, C5AR (also known as CD88) and C5L2, official symbol GPR77. To date, there are few data available on the role of C5a and CD88 in human pregnancy, so the aim of this study was to determine the effect of C5a and CD88 on some key inflammatory pathways involved in human parturition. Placental tissue samples were obtained from normal pregnancies at the time of Cesarean section. Human placental and fetal membranes were incubated in the absence (basal control) or presence of 0.5 Āµg/ml (~60 nM) human recombinant C5a for 24 h. Concentrations of proinflammatory cytokines, prostaglandins, and 8-isoprostane (a marker of oxidative stress) were quantified by ELISA and secretory matrix metalloproteinases (MMPs) activity by zymography. NFKB DNA binding activity and NFKBIA (IkappaB-alpha; inhibitor of NFKB) protein degradation were analyzed by ELISA and Western blotting, respectively. In the presence of C5a, proinflammatory cytokines (IL6 and IL8), cyclooxygenase (COX)-2; official symbol PTGS2) expression, and subsequent prostaglandin (PGE(2) and PGF(2alpha)), MMP9 enzyme production, and NFKB DNA activation were all significantly increased. The C5a-induced prolabor responses were significantly reduced by treatment with the selective CD88 antagonist PMX53 and the NFKB inhibitor BAY 11-7082. We conclude that C5a upregulates prolabor mediators in human gestational tissues via CD88-mediated NFKB activation.


Subject(s)
Complement C5a/metabolism , Parturition/metabolism , Placenta/metabolism , Receptors, Complement/metabolism , Cytokines/metabolism , Dinoprost/analogs & derivatives , Dinoprost/metabolism , Female , Humans , Lipopolysaccharides , NF-kappa B/metabolism , Peptide Hydrolases/metabolism , Peptides, Cyclic , Pregnancy , Prostaglandins/metabolism , Receptor, Anaphylatoxin C5a , Receptors, Complement/antagonists & inhibitors
17.
Lung ; 190(2): 189-98, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22037793

ABSTRACT

BACKGROUND: Bronchial asthma is an inflammatory disease of the airways. Thrombin-activatable fibrinolysis inhibitor (TAFI) is a carboxypeptidase that besides inhibiting fibrinolysis, also regulates inflammatory processes. The only validated substrate known for TAFI is fibrin. In the present study we evaluated the role of TAFI in bronchial asthma by comparing the development of allergic bronchial asthma between wild-type (WT) and TAFI-deficient mice (KO). METHODS: Asthmatic inflammation was induced by sensitization and challenge with ovalbumin in WT (WT/OVA) and TAFI KO (KO/OVA) mice. WT mice (WT/SAL) and TAFI KO (KO/SAL) were used as controls. Cytokines, markers of inflammation, and coagulation were measured in bronchoalveolar lavage fluid (BALF). RESULTS: Airway hyperresponsiveness was worse in KO/OVA mice than in WT/OVA mice or control mice. Markers of lung injury were significantly increased in BALF from KO/OVA mice compared to WT/OVA mice. Airway hyperresponsiveness and the BALF concentrations of IL-5 and osteopontin were significantly increased in KO/OVA mice compared to WT/OVA mice. Treatment of WT/OVA and KO/OVA mice with a C5a receptor antagonist significantly decreased hyperresponsiveness along with the BALF concentrations of total protein and C5a compared to untreated asthmatic mice. CONCLUSION: The results of this study suggest that TAFI plays a protective role in the pathogenesis of allergic inflammation probably by inhibiting the complement system.


Subject(s)
Asthma/immunology , Bronchial Hyperreactivity/immunology , Carboxypeptidase B2/metabolism , Airway Resistance , Animals , Asthma/enzymology , Biomarkers/chemistry , Blood Coagulation , Bronchoalveolar Lavage Fluid/chemistry , Bronchoalveolar Lavage Fluid/immunology , Carboxypeptidase B2/deficiency , Complement C5a/immunology , Fibrinolysis , Interleukin-5/analysis , Interleukin-5/metabolism , L-Lactate Dehydrogenase/blood , Lung Injury/immunology , Lung Injury/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Osteopontin/metabolism , Ovalbumin/immunology , Plasminogen Activator Inhibitor 1/metabolism , Receptors, Complement/antagonists & inhibitors
18.
Anticancer Res ; 42(3): 1207-1215, 2022 Mar.
Article in English | MEDLINE | ID: mdl-35220210

ABSTRACT

BACKGROUND: We aimed to clarify the role of complement C3a and its receptor C3aR in progression of pancreatic ductal adenocarcinoma (PDAC). MATERIALS AND METHODS: We evaluated the serum levels of C3 and C3a in patients with PDAC. C3aR expression in tissue was assessed using a tissue microarray. To confirm the protumoral effects of C3a in PDAC, we conducted in vitro experiments using PDAC cell lines (Panc-1 and MiaPaca-2) that exhibit high C3aR expression. RESULTS: Serum levels of both C3 and C3a were higher in 26 patients with PDAC than in 28 nontumor-bearing controls. In the tissue microarray, we observed increased expression of C3aR in PDAC cells, especially in cases with metastatic lesions. In vitro experiments showed that C3a facilitated tumor cell proliferation, migration and invasion by activating the extracellular-regulated kinase signaling pathway and inducing epithelial-to-mesenchymal transition. Inhibition of the C3a-C3aR axis by pharmacological blockade and short-hairpin RNA-mediated knockdown of C3aR alleviated its protumoral effect. CONCLUSION: These findings provide a new approach for the development of treatments targeting the C3a-C3aR axis.


Subject(s)
Carcinoma, Pancreatic Ductal/enzymology , Complement C3/metabolism , Epithelial-Mesenchymal Transition , Extracellular Signal-Regulated MAP Kinases/metabolism , Pancreatic Neoplasms/enzymology , Receptors, Complement/metabolism , Aged , Aged, 80 and over , Arginine/analogs & derivatives , Arginine/pharmacology , Benzhydryl Compounds/pharmacology , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Cell Movement , Cell Proliferation , Complement Inactivating Agents/pharmacology , Enzyme Activation , Epithelial-Mesenchymal Transition/drug effects , Female , Gene Expression Regulation, Neoplastic , Humans , Male , Middle Aged , Neoplasm Invasiveness , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Receptors, Complement/antagonists & inhibitors , Receptors, Complement/genetics , Signal Transduction
19.
Am J Respir Cell Mol Biol ; 45(4): 834-42, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21421909

ABSTRACT

The widely used herbicide, paraquat (PQ), is highly toxic and claims thousands of lives from both accidental and voluntary ingestion. The pathological mechanisms of PQ poisoning-induced acute lung injury (ALI) are not well understood, and the role of complement in PQ-induced ALI has not been elucidated. We developed and characterized a mouse model of PQ-induced ALI and studied the role of complement in the pathogenesis of PQ poisoning. Intraperitoneal administration of PQ caused dose- and time-dependent lung damage and mortality, with associated inflammatory response. Within 24 hours of PQ-induced ALI, there was significantly increased expression of the complement proteins, C1q and C3, in the lung. Expression of the anaphylatoxin receptors, C3aR and C5aR, was also increased. Compared with wild-type mice, C3-deficient mice survived significantly longer and displayed significantly reduced lung inflammation and pathology after PQ treatment. Similar reductions in PQ-induced inflammation, pathology, and mortality were recorded in mice treated with the C3 inhibitors, CR2-Crry, and alternative pathway specific CR2-fH. A similar therapeutic effect was also observed by treatment with either C3a receptor antagonist or a blocking C5a receptor monoclonal antibody. Together, these studies indicate that PQ-induced ALI is mediated through receptor signaling by the C3a and C5a complement activation products that are generated via the alternative complement pathway, and that complement inhibition may be an effective clinical intervention for postexposure treatment of PQ-induced ALI.


Subject(s)
Acute Lung Injury/prevention & control , Anti-Inflammatory Agents/pharmacology , Complement Activation/drug effects , Lung/drug effects , Paraquat , Acute Lung Injury/chemically induced , Acute Lung Injury/genetics , Acute Lung Injury/immunology , Acute Lung Injury/pathology , Animals , Antibodies, Monoclonal/pharmacology , Complement Activation/genetics , Complement C1q/metabolism , Complement C3/antagonists & inhibitors , Complement C3/genetics , Complement C3/metabolism , Disease Models, Animal , Lung/immunology , Lung/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptor, Anaphylatoxin C5a/antagonists & inhibitors , Receptor, Anaphylatoxin C5a/metabolism , Receptors, Complement/antagonists & inhibitors , Receptors, Complement/metabolism , Recombinant Fusion Proteins/pharmacology , Signal Transduction/drug effects , Time Factors
20.
Mol Pharmacol ; 79(6): 1005-13, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21441599

ABSTRACT

Human mast cells express the G protein coupled receptor (GPCR) for C5a (CD88). Previous studies indicated that C5a could cause mast cell degranulation, at least in part, via a mechanism similar to that proposed for basic neuropeptides such as substance P, possibly involving Mas-related gene 2 (MrgX2). We therefore sought to more clearly define the receptor specificity for C5a-induced mast cell degranulation. We found that LAD2, a human mast cell line, and CD34(+) cell-derived primary mast cells express functional MrgX1 and MrgX2 but the immature human mast cell line HMC-1 does not. A potent CD88 antagonist, PMX-53 (10 nM) inhibited C5a-induced Ca(2+) mobilization in HMC-1 cells, but at higher concentrations (≥30 nM) it caused degranulation in LAD2 mast cells, CD34(+) cell-derived mast cells, and RBL-2H3 cells stably expressing MrgX2. PMX-53 did not, however, activate RBL-2H3 cells expressing MrgX1. Although C5a induced degranulation in LAD2 and CD34(+) cell-derived mast cells, it did not activate RBL-2H3 cells expressing MrgX1 or MrgX2. Replacement of Trp with Ala and Arg with dArg abolished the ability of PMX-53 to inhibit C5a-induced Ca(2+) mobilization in HMC-1 cells and to cause degranulation in RBL-2H3 cells expressing MrgX2. These findings demonstrate that C5a does not use MrgX1 or MrgX2 for mast cell degranulation. Moreover, it reveals the novel finding that PMX-53 functions as a potent CD88 antagonist and a low-affinity agonist for MrgX2. Furthermore, Trp and Arg residues are required for the ability of PMX53 to act as both a CD88 antagonist and a MrgX2 agonist.


Subject(s)
Mast Cells/drug effects , Nerve Tissue Proteins/agonists , Peptides, Cyclic/pharmacology , Receptors, Complement/antagonists & inhibitors , Receptors, G-Protein-Coupled/agonists , Receptors, Neuropeptide/agonists , Cell Differentiation , Cell Line , Cells, Cultured , Complement C5a/antagonists & inhibitors , Humans , Receptor, Anaphylatoxin C5a , Receptors, G-Protein-Coupled/metabolism , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL