Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 66
Filter
Add more filters

Publication year range
1.
Mar Policy ; 132: 104646, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34602712

ABSTRACT

The COVID-19 global pandemic and subsequent implementation of measures to reduce contact within the community have affected fisheries worldwide, yet few studies have reported the impacts on recreational fisheries. This study investigates boat-based recreational fishing in Western Australia from March to August 2020, where COVID-19 measures relevant to recreational fishers included various travel restrictions, and social and physical distancing measures. Information from surveys of licensed recreational fishers and fisheries compliance officers, and camera footage from key boat ramps is presented. A lower proportion of Perth metropolitan fishers went fishing compared with regional fishers. Metropolitan fishers also reported fewer days fished and lower participation in demersal and shore-based line fishing than regional fishers. In contrast, compliance officers observed more fishing activity in both metropolitan and regional locations. Fishing plans were mostly affected by travel restrictions with more metropolitan fishers affected compared with regional fishers. Daily recreational vessel retrievals at key boat ramps varied between locations, with metropolitan fishers initially unable to travel to regional centres. There was no decline in vessel retrievals at metropolitan boat ramps during the most rigid restrictions and northern regional boat ramps experienced substantial increases in recreational vessel activity once travel restrictions eased. Studies of this kind highlight the value of utilising established recreational fishing monitoring programmes to provide a responsive and scientific basis for policymakers to address societal behavioural changes associated with atypical events such as COVID-19.

2.
FASEB J ; 33(10): 11060-11071, 2019 10.
Article in English | MEDLINE | ID: mdl-31298935

ABSTRACT

The canonical complement component 5a (C5a) receptor (C5aR) 1 has well-described roles in tumorigenesis but the contribution of the second receptor, C5aR2, is unclear. The present study demonstrates that B16.F0 melanoma cells express mRNA for both C5aR1 and C5aR2 and signal through ERK and p38 MAPKs in response to C5a. Despite this, C5a had no impact on melanoma cell proliferation or migration in vitro. In vivo studies demonstrated that the growth of B16.F0 melanoma tumors was increased in C5aR2-/- mice but reduced in C5aR1-/- mice and wild-type mice treated with a C5aR1 antagonist. Analysis of tumor-infiltrating leukocyte populations showed no significant differences between wild-type and C5aR2-/- mice. Conversely, percentages of myeloid-derived suppressor cells, macrophages, and regulatory T lymphocytes were lower in tumors from C5aR1-/- mice, whereas total (CD3+) T lymphocytes and CD4+ subsets were higher. Analysis of cytokine and chemokine levels also showed plasma IFN-γ was higher and tumor C-C motif chemokine ligand 2 was lower in the absence of C5aR1. The results suggest that C5aR1 signaling supports melanoma growth by promoting infiltration of immunosuppressive leukocyte populations into the tumor microenvironment, whereas C5aR2 has a more restricted but beneficial role in limiting tumor growth. Overall, these data support the potential of C5aR1-inhibitory therapies for melanoma.-Nabizadeh, J. A., Manthey, H. D., Panagides, N., Steyn, F. J., Lee, J. D., Li, X. X., Akhir, F. N. M., Chen, W., Boyle, G. M., Taylor, S. M., Woodruff, T. M., Rolfe, B. E. C5a receptors C5aR1 and C5aR2 mediate opposing pathologies in a mouse model of melanoma.


Subject(s)
Cell Movement , Lymphocytes, Tumor-Infiltrating/immunology , Melanoma/genetics , Receptor, Anaphylatoxin C5a/genetics , Animals , Cell Proliferation , Cells, Cultured , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Complement C5a/immunology , Female , Interferon-gamma/genetics , Interferon-gamma/metabolism , Lymphocytes, Tumor-Infiltrating/pathology , MAP Kinase Signaling System , Male , Melanoma/immunology , Melanoma/pathology , Mice , Mice, Inbred C57BL , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor, Anaphylatoxin C5a/metabolism , Tumor Microenvironment
3.
J Immunol ; 196(11): 4783-92, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27183625

ABSTRACT

The complement peptide C3a is a key component of the innate immune system and a major fragment produced following complement activation. We used a murine model of melanoma (B16-F0) to identify a hitherto unknown role for C3a-C3aR signaling in promoting tumor growth. The results show that the development and growth of B16-F0 melanomas is retarded in mice lacking C3aR, whereas growth of established melanomas can be arrested by C3aR antagonism. Flow cytometric analysis showed alterations in tumor-infiltrating leukocytes in the absence of C3aR. Specifically, neutrophils and CD4(+) T lymphocyte subpopulations were increased, whereas macrophages were reduced. The central role of neutrophils was confirmed by depletion experiments that reversed the tumor inhibitory effects observed in C3aR-deficient mice and returned tumor-infiltrating CD4(+) T cells to control levels. Analysis of the tumor microenvironment showed upregulation of inflammatory genes that may contribute to the enhanced antitumor response observed in C3aR-deficient mice. C3aR deficiency/inhibition was also protective in murine models of BRAF(V600E) mutant melanoma and colon and breast cancer, suggesting a tumor-promoting role for C3aR signaling in a range of tumor types. We propose that C3aR activation alters the tumor inflammatory milieu, thereby promoting tumor growth. Therapeutic inhibition of C3aR may therefore be an effective means to trigger an antitumor response in melanoma and other cancers.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Carcinogenesis/immunology , Melanoma/immunology , Melanoma/pathology , Neutrophils/immunology , Receptors, Complement/immunology , Animals , CD4-Positive T-Lymphocytes/pathology , Cells, Cultured , Female , Melanoma/genetics , Mice , Mice, Inbred BALB C , Mice, Knockout , Neutrophils/pathology , Receptors, Complement/deficiency
4.
J Neurosci ; 35(16): 6517-31, 2015 Apr 22.
Article in English | MEDLINE | ID: mdl-25904802

ABSTRACT

This study investigated the role of the complement activation fragment C5a in secondary pathology following contusive spinal cord injury (SCI). C5ar(-/-) mice, which lack the signaling receptor for C5a, displayed signs of improved locomotor recovery and reduced inflammation during the first week of SCI compared with wild-type mice. Intriguingly, the early signs of improved recovery in C5ar(-/-) mice deteriorated from day 14 onward, with absence of C5aR ultimately leading to poorer functional outcomes, larger lesion volumes, reduced myelin content, and more widespread inflammation at 35 d SCI. Pharmacological blockade of C5aR with a selective antagonist (C5aR-A) during the first 7 d after SCI improved recovery compared with vehicle-treated mice, and this phenotype was sustained up to 35 d after injury. Consistent with observations made in C5ar(-/-) mice, these improvements were, however, lost if C5aR-A administration was continued into the more chronic phase of SCI. Signaling through the C5a-C5aR axis thus appears injurious in the acute period but serves a protective and/or reparative role in the post-acute phase of SCI. Further experiments in bone marrow chimeric mice suggested that the dual and opposing roles of C5aR on SCI outcomes primarily relate to its expression on CNS-resident cells and not infiltrating leukocytes. Additional in vivo and in vitro studies provided direct evidence that C5aR signaling is required during the postacute phase for astrocyte hyperplasia, hypertrophy, and glial scar formation. Collectively, these findings highlight the complexity of the inflammatory response to SCI and emphasize the importance of optimizing the timing of therapeutic interventions.


Subject(s)
Gliosis/physiopathology , Inflammation/physiopathology , Receptor, Anaphylatoxin C5a/physiology , Spinal Cord Injuries/physiopathology , Animals , Astrocytes/physiology , Cell Proliferation/drug effects , Cell Proliferation/physiology , Cells, Cultured , Complement Activation/drug effects , Complement Activation/physiology , Complement C5a/biosynthesis , Female , Gliosis/complications , Gliosis/drug therapy , Gliosis/pathology , Inflammation/complications , Inflammation/drug therapy , Inflammation/pathology , Mice , Mice, Knockout , Motor Activity/drug effects , Motor Activity/physiology , Peptides, Cyclic/pharmacology , Peptides, Cyclic/therapeutic use , Receptor, Anaphylatoxin C5a/antagonists & inhibitors , Receptor, Anaphylatoxin C5a/biosynthesis , Receptor, Anaphylatoxin C5a/genetics , Recovery of Function/physiology , Spinal Cord Injuries/complications , Spinal Cord Injuries/drug therapy , Spinal Cord Injuries/metabolism
5.
Birth Defects Res A Clin Mol Teratol ; 106(3): 201-7, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26900104

ABSTRACT

BACKGROUND: Periconceptional supplementation with folic acid results in a significant reduction in the incidence of neural tube defects (NTDs). Nonetheless, NTDs remain a leading cause of perinatal morbidity and mortality worldwide, and the mechanism(s) by which folate exerts its protective effects are unknown. Homocysteine is an amino acid that accumulates under conditions of folate-deficiency, and is suggested as a risk factor for NTDs. One proposed mechanism of homocysteine toxicity is its accumulation into proteins in a process termed homocysteinylation. METHODS & RESULTS: Herein, we used a folate-deficient diet in pregnant mice to demonstrate that there is: (i) a significant inverse correlation between maternal serum folate levels and serum homocysteine; (ii) a significant positive correlation between serum homocysteine levels and titers of autoantibodies against homocysteinylated protein; and (iii) a significant increase in congenital malformations and NTDs in mice deficient in serum folate. Furthermore, in mice administered the folate-deplete diet before conception, supplementation with folic acid during the gestational period completely rescued the embryos from congenital defects, and resulted in homocysteinylated protein titers at term that are comparable to that of mice administered a folate-replete diet throughout both the pre- and postconception period. These results demonstrate that a low-folate diet that induces NTDs also increases protein homocysteinylation and the subsequent generation of autoantibodies against homocysteinylated proteins. CONCLUSION: These data support the hypotheses that homocysteinylation results in neo-self antigen formation under conditions of maternal folate deficiency, and that this process is reversible with folic acid supplementation.


Subject(s)
Autoantibodies/blood , Blood Proteins/metabolism , Folic Acid Deficiency/complications , Folic Acid/blood , Homocysteine/chemistry , Neural Tube Defects/etiology , Animals , Blood Proteins/immunology , Diet , Dietary Supplements , Disease Models, Animal , Female , Folic Acid/administration & dosage , Folic Acid/immunology , Folic Acid Deficiency/blood , Folic Acid Deficiency/immunology , Folic Acid Deficiency/pathology , Gestational Age , Homocysteine/biosynthesis , Humans , Mice , Mice, Inbred C57BL , Neural Tube Defects/blood , Neural Tube Defects/immunology , Neural Tube Defects/pathology , Pregnancy , Protein Processing, Post-Translational
6.
Proc Natl Acad Sci U S A ; 110(23): 9439-44, 2013 Jun 04.
Article in English | MEDLINE | ID: mdl-23696668

ABSTRACT

C3a is a key complement activation fragment, yet its neutrophil-expressed receptor (C3aR) still has no clearly defined role. In this study, we used a neutrophil-dependent mouse model of intestinal ischemia-reperfusion (IR) injury to explore the role of C3aR in acute tissue injuries. C3aR deficiency worsened intestinal injury, which corresponded with increased numbers of tissue-infiltrating neutrophils. Circulating neutrophils were significantly increased in C3aR(-/-) mice after intestinal ischemia, and C3aR(-/-) mice also mobilized more circulating neutrophils after granulocyte colony-stimulating factor infusion compared with WT mice, indicating a specific role for C3aR in constraining neutrophil mobilization in response to intestinal injury. In support of this role, C3aR(-/-) mice reconstituted with WT bone marrow reversed IR pathology back to WT levels. Complement C5a receptor (C5aR) antagonism in C3aR(-/-) mice also rectified the worsened pathology after intestinal IR injury but had no effect on circulating neutrophils, highlighting the opposing roles of C3a and C5a in disease pathogenesis. Finally, we found that using a potent C3a agonist to activate C3aR in vivo reduced neutrophil mobilization and ameliorated intestinal IR pathology in WT, but not C3aR(-/-), mice. This study identifies a role for C3aR in regulating neutrophil mobilization after acute intestinal injury and highlights C3aR agonism as a potential treatment option for acute, neutrophil-driven pathologies.


Subject(s)
Intestines/immunology , Neutrophils/immunology , Receptors, Complement/immunology , Reperfusion Injury/prevention & control , Animals , Apoptosis/immunology , Cytokines/blood , Hemoglobins/metabolism , Histological Techniques , Intestines/cytology , Mice , Mice, Knockout , Receptors, Complement/genetics , Receptors, Complement/metabolism , Reperfusion Injury/immunology
7.
Stem Cells ; 32(12): 3278-84, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25132103

ABSTRACT

The complement activation product, C5a, is a pivotal member of the innate immune response; however, a diverse number of nonimmune functions are now being ascribed to C5a signaling, including roles during embryonic development. Here, we identify the expression of the C5a precursor protein, C5, as well as the C5a receptors, C5aR and C5L2, in both human embryonic stem cells and human-induced pluripotent stem cells. We show that administration of a physiologically relevant dose of purified human C5a (1 nM) stimulates activation of ERK1/2 and AKT signaling pathways, and is able to promote maintenance of the pluripotent state in the absence of FGF2. C5a also reduced cell loss following dissociation of human pluripotent stem cells. Our results reveal that complement C5a signaling supports human stem cell pluripotency and survival, and thus may play a key role in shaping early human embryonic development.


Subject(s)
Complement C5a/metabolism , Fibroblast Growth Factor 2/metabolism , Human Embryonic Stem Cells/cytology , Pluripotent Stem Cells/cytology , Human Embryonic Stem Cells/metabolism , Humans , Immunity, Innate/immunology , Inflammation/immunology , Pluripotent Stem Cells/metabolism , Receptor, Anaphylatoxin C5a/metabolism , Signal Transduction/immunology
8.
J Immunol ; 190(7): 3493-9, 2013 Apr 01.
Article in English | MEDLINE | ID: mdl-23420882

ABSTRACT

The complement system is involved in a range of diverse developmental processes, including cell survival, growth, differentiation, and regeneration. However, little is known about the role of complement in embryogenesis. In this study, we demonstrate a novel role for the canonical complement 5a receptor (C5aR) in the development of the mammalian neural tube under conditions of maternal dietary folic acid deficiency. Specifically, we found C5aR and C5 to be expressed throughout the period of neurulation in wild-type mice and localized the expression to the cephalic regions of the developing neural tube. C5aR was also found to be expressed in the neuroepithelium of early human embryos. Ablation of the C5ar1 gene or the administration of a specific C5aR peptide antagonist to folic acid-deficient pregnant mice resulted in a high prevalence of severe anterior neural tube defect-associated congenital malformations. These findings provide a new and compelling insight into the role of the complement system during mammalian embryonic development.


Subject(s)
Folic Acid Deficiency/complications , Neural Tube Defects/etiology , Neural Tube Defects/prevention & control , Receptor, Anaphylatoxin C5a/metabolism , Signal Transduction , Animals , Complement C5/genetics , Complement C5/metabolism , Disease Models, Animal , Embryo, Mammalian/metabolism , Female , Humans , Male , Mice , Mice, Knockout , Morphogenesis/genetics , Neural Tube/embryology , Neural Tube/metabolism , Neural Tube Defects/pathology , Neurulation/genetics , Pregnancy , Protein Transport , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor, Anaphylatoxin C5a/genetics
9.
FASEB J ; 27(3): 855-64, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23239822

ABSTRACT

C5a is the paramount proinflammatory mediator of the complement cascade, and has been previously thought to act only through a single, G-protein-coupled, C5a receptor (C5aR; also termed CD88). In 2000, a second C5a receptor, C5L2 (previously known as GPR77), was discovered; yet, despite 12 yr of intensive research, its biological, or pathophysiological, function is both enigmatic and controversial. Unlike C5aR, this receptor does not couple to G proteins, and early studies promoted the hypothesis that C5L2 functions as a decoy receptor. However, recent data have provided other evidence for more complicated and conflicting interactions between C5L2 and other inflammatory mediators. C5L2 has been recently demonstrated to physically interact with both C5aR and ß-arrestin to negatively regulate C5aR signaling toward an anti-inflammatory manner, and to reduce pathology, in several disease models in vivo. In direct contrast, other groups have demonstrated that C5L2 stimulation caused release of HMGB1 both in vitro and in vivo, and enhanced pathology in sepsis models, suggesting a clear proinflammatory signaling role. These astoundingly contradictory data challenge our precepts and complicate the foundational bases for the possible targeting of C5L2 as a therapeutic option in inflammatory disease. C5L2 may be the great masquerader in complement biology; its function dependent on the cell type, species, and disease context. Because of these unusual and unforeseen complexities, we present the current state of knowledge on C5L2 structure, expression and, most controversially, its putative functions.-Li, R., Coulthard, L.G., Wu, M. C. L., Taylor, S. M., Woodruff, T. M. C5L2: a controversial receptor of complement anaphylatoxin, C5a.


Subject(s)
Complement C5a/metabolism , Receptors, Chemokine/metabolism , Receptors, Complement/metabolism , Sepsis/metabolism , Signal Transduction , Animals , Arrestins/genetics , Arrestins/immunology , Arrestins/metabolism , Complement C5a/genetics , Complement C5a/immunology , Disease Models, Animal , Gene Expression Regulation/genetics , Gene Expression Regulation/immunology , Humans , Protein Structure, Tertiary , Receptor, Anaphylatoxin C5a , Receptors, Chemokine/genetics , Receptors, Chemokine/immunology , Receptors, Complement/genetics , Receptors, Complement/immunology , Sepsis/genetics , Sepsis/immunology , Sepsis/pathology , Sepsis/therapy , beta-Arrestins
10.
J Neuroinflammation ; 10: 119, 2013 Sep 26.
Article in English | MEDLINE | ID: mdl-24067070

ABSTRACT

BACKGROUND: Components of the innate immune complement system have been implicated in the pathogenesis of amyotrophic lateral sclerosis (ALS); however, a comprehensive examination of complement expression in this disease has not been performed. This study therefore aimed to determine the expression of complement components (C1qB, C4, factor B, C3/C3b, C5 and CD88) and regulators (CD55 and CD59a) in the lumbar spinal cord of hSOD1(G93A) mice during defined disease stages. METHODS: hSOD1(G93A) and wild-type mice were examined at four different ages of disease progression. mRNA and protein expression of complement components and regulators were examined using quantitative PCR, western blotting and ELISA. Localisation of complement components within lumbar spinal cord was investigated using immunohistochemistry. Statistical differences between hSOD1(G93A) and wild-type mice were analysed using a two-tailed t-test at each stage of disease progression. RESULTS: We found several early complement factors increased as disease progressed, whilst complement regulators decreased; suggesting overall increased complement activation through the classical or alternative pathways in hSOD1(G93A) mice. CD88 was also increased during disease progression, with immunolocalisation demonstrating expression on motor neurons and increasing expression on microglia surrounding the regions of motor neuron death. CONCLUSIONS: These results indicate that local complement activation and increased expression of CD88 may contribute to motor neuron death and ALS pathology in the hSOD1(G93A) mouse. Hence, reducing complement-induced inflammation could be an important therapeutic strategy to treat ALS.


Subject(s)
Amyotrophic Lateral Sclerosis/immunology , Complement System Proteins/metabolism , Spinal Cord/immunology , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/pathology , Animals , Blotting, Western , Complement System Proteins/analysis , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Humans , Immunohistochemistry , In Situ Hybridization , Mice , Mice, Inbred C57BL , Mice, Transgenic , Motor Neurons/metabolism , Motor Neurons/pathology , Real-Time Polymerase Chain Reaction , Spinal Cord/metabolism , Spinal Cord/pathology , Superoxide Dismutase/genetics , Superoxide Dismutase-1
11.
Am J Pathol ; 181(5): 1493-503, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23021982

ABSTRACT

The Eph/ephrin receptor-ligand system plays an important role in embryogenesis and adult life, principally by influencing cell behavior through signaling pathways, resulting in modification of the cell cytoskeleton and cell adhesion. There are 10 EphA receptors, and six EphB receptors, distinguished on sequence difference and binding preferences, that interact with the six glycosylphosphatidylinositol-linked ephrin-A ligands and the three transmembrane ephrin-B ligands, respectively. The Eph/ephrin proteins, originally described as developmental regulators that are expressed at low levels postembryonically, are re-expressed after injury to the optic nerve, spinal cord, and brain in fish, amphibians, rodents, and humans. In rodent spinal cord injury, the up-regulation of EphA4 prevents recovery by inhibiting axons from crossing the injury site. Eph/ephrin proteins may be partly responsible for the phenotypic changes to the vascular endothelium in inflammation, which allows fluid and inflammatory cells to pass from the vascular space into the interstitial tissues. Specifically, EphA2/ephrin-A1 signaling in the lung may be responsible for pulmonary inflammation in acute lung injury. A role in T-cell maturation and chronic inflammation (heart failure, inflammatory bowel disease, and rheumatoid arthritis) is also reported. Although there remains much to learn about Eph/ephrin signaling in human disease, and specifically in injury and inflammation, this area of research raises the exciting prospect that novel therapies will be developed that precisely target these pathways.


Subject(s)
Ephrins/metabolism , Inflammation/metabolism , Receptors, Eph Family/metabolism , Signal Transduction , Wounds and Injuries/metabolism , Animals , Humans , Ligands
12.
FASEB J ; 26(9): 3680-90, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22651932

ABSTRACT

C5a receptors are found in the central nervous system (CNS), on both neurons and glia. However, the origin of the C5a, which activates these receptors, is unclear. In the present study, we show that primary cultured mouse cortical neurons constitutively express C5, the precursor of C5a, and express the classical receptor for C5a, CD88. With cell ischemia caused by 12 h glucose deprivation, or oxygen-glucose deprivation (OGD), neurons demonstrated increased apoptosis, up-regulation of CD88, and increased levels of C5a in the media. Exogenous murine C5a (100 nM) added to the neuronal cultures resulted in apoptosis, without affecting cell necrosis. Pretreatment of the cells with the specific CD88 receptor antagonist PMX53 (100 nM) significantly blocked ischemia-induced apoptosis (∼50%), and neurons from CD88(-/-) mice were similarly protected. In a murine model of stroke, using middle cerebral artery occlusion (MCAO), we found that C5a levels in the brain increased; this also occurred in cerebral slice cultures exposed to OGD. CD88(-/-) mice subjected to MCAO had significantly reduced infarct volumes and improved neurological scores. Taken together, our results demonstrate that neurons in the CNS have the capability to generate C5a following ischemic stress, and this has the potential to activate their C5a receptors, with deleterious consequences.


Subject(s)
Apoptosis , Brain Ischemia/pathology , Complement C5a/biosynthesis , Neurons/metabolism , Animals , Brain Ischemia/metabolism , Cell Line, Tumor , Enzyme-Linked Immunosorbent Assay , Female , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/pathology , Polymerase Chain Reaction , Pregnancy , Receptor, Anaphylatoxin C5a/genetics
13.
Birth Defects Res A Clin Mol Teratol ; 97(9): 602-609, 2013 Sep.
Article in English | MEDLINE | ID: mdl-24078477

ABSTRACT

Periconceptional supplementation with folic acid has led to a significant worldwide reduction in the incidence of neural tube defects (NTDs). However, despite increasing awareness of the benefits of folic acid supplementation and the implementation of food fortification programs in many countries, NTDs continue to be a leading cause of perinatal morbidity and mortality worldwide. Furthermore, there exists a significant subgroup of women who appear to be resistant to the protective effects of folic acid supplementation. The following review addresses emerging clinical and experimental evidence for a role of the immune system in the etiopathogenesis of NTDs, with the aim of developing novel preventative strategies to further reduce the incidence of NTD-affected pregnancies. In particular, recent studies demonstrating novel roles and interactions between innate immune factors such as the complement cascade, neurulation, and folate metabolism are explored.


Subject(s)
Folate Receptors, GPI-Anchored/metabolism , Immunologic Factors/metabolism , Neural Tube Defects/epidemiology , Neural Tube Defects/etiology , Neural Tube Defects/physiopathology , Neurulation/physiology , Pregnancy in Diabetics/immunology , Tetrahydrofolates/metabolism , Anticonvulsants/adverse effects , Autoantibodies/immunology , Chemokine CCL2/immunology , Chemokine CCL2/metabolism , Complement System Proteins/immunology , Complement System Proteins/metabolism , Female , Folate Receptors, GPI-Anchored/immunology , Humans , Immunologic Factors/blood , Neural Tube Defects/prevention & control , Neurulation/immunology , Pregnancy , Risk Factors , Tetrahydrofolates/blood , Valproic Acid/adverse effects
14.
J Neuroinflammation ; 9: 137, 2012 Jun 21.
Article in English | MEDLINE | ID: mdl-22721265

ABSTRACT

The complement system, a major component of the innate immune system, is becoming increasingly recognised as a key participant in physiology and disease. The awareness that immunological mediators support various aspects of both normal central nervous system (CNS) function and pathology has led to a renaissance of complement research in neuroscience. Various studies have revealed particularly novel findings on the wide-ranging involvement of complement in neural development, synapse elimination and maturation of neural networks, as well as the progression of pathology in a range of chronic neurodegenerative disorders, and more recently, neurotraumatic events, where rapid disruption of neuronal homeostasis potently triggers complement activation. The purpose of this review is to summarise recent findings on complement activation and acquired brain or spinal cord injury, i.e. ischaemic-reperfusion injury or stroke, traumatic brain injury (TBI) and spinal cord injury (SCI), highlighting the potential for complement-targeted therapeutics to alleviate the devastating consequences of these neurological conditions.


Subject(s)
Brain Injuries/immunology , Central Nervous System/immunology , Central Nervous System/metabolism , Complement Activation/immunology , Neurodegenerative Diseases/immunology , Spinal Cord Injuries/immunology , Animals , Brain Injuries/metabolism , Brain Injuries/pathology , Central Nervous System/pathology , Humans , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Spinal Cord Injuries/metabolism , Spinal Cord Injuries/pathology
15.
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 µg/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
16.
FASEB J ; 25(7): 2447-55, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21490292

ABSTRACT

The complement C5a receptor, CD88, is present on many of the cells found within human atherosclerotic plaques, but little is known about the role of C5a in atherogenesis. Using real-time PCR, we determined that ApoE(-/-) mice fed a normal diet express more aortic CD88 mRNA compared with controls, and this increase coincides with atherosclerotic lesion development (P<0.001 for 3- vs. 25-wk-old animals). Conversely, mRNA expression of the alternative C5a receptor, C5L2, in aortas of ApoE(-/-) mice, was lower than controls at all time points. Using immunohistochemistry, we confirmed the presence of CD88 on macrophages, smooth muscle cells, and activated endothelial cells in plaques from brachiocephalic arteries. Treatment of ApoE(-/-) mice with a CD88 antagonist (PMX53; 3 mg/kg s.c. 3 ×/wk plus 1 mg/kg/d p.o.) for 25 wk reduced lesion size and lipid content in the plaque by ∼ 40% (P<0.05). Our study provides evidence for a proatherogenic role for C5a and identifies the CD88 antagonist PMX53 as a potential antiatherosclerotic drug.


Subject(s)
Apolipoproteins E/deficiency , Atherosclerosis/metabolism , Complement C5a/metabolism , Receptor, Anaphylatoxin C5a/metabolism , Animals , Aorta/drug effects , Aorta/metabolism , Aorta/pathology , Apolipoproteins E/genetics , Atherosclerosis/genetics , Atherosclerosis/prevention & control , Brachiocephalic Trunk/drug effects , Brachiocephalic Trunk/metabolism , Brachiocephalic Trunk/pathology , Cholesterol/blood , Complement C5a/antagonists & inhibitors , Endothelial Cells/metabolism , Female , Immunohistochemistry , Lipids/analysis , Lipids/blood , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocytes, Smooth Muscle/metabolism , Peptides, Cyclic/pharmacology , Receptor, Anaphylatoxin C5a/antagonists & inhibitors , Receptor, Anaphylatoxin C5a/genetics , Receptors, Chemokine/genetics , Receptors, Chemokine/metabolism , Reverse Transcriptase Polymerase Chain Reaction
17.
Biology (Basel) ; 11(10)2022 Oct 20.
Article in English | MEDLINE | ID: mdl-36290440

ABSTRACT

The management of human-shark interactions can benefit from the implementation of effective shark hazard mitigation measures. A Shark-Management-Alert-in-Real-Time (SMART) drumline trial in the Capes region of Western Australia was instigated after several serious incidents involving surfers and white sharks (Carcharodon carcharias). The project aimed to determine whether white sharks (target species), which were relocated after capture, remained offshore using satellite and acoustic tagging. Over a 27-month period, 352 fish were caught, 55% of which comprised tiger sharks (Galeocerdo cuvier). Ninety-one percent of animals were released alive in good condition. Only two white sharks were caught; both were relocated ≥ 1 km offshore before release and moved immediately further offshore after capture, remaining predominately in offshore waters for the duration of their 54-day and 186-day tag deployments. Our results confirm that desirable animal welfare outcomes can be achieved using SMART drumlines when response times are minimised. The low target catches and the detection of 24 other tagged white sharks within the study area supported the decision to cease the trial. Our results reiterate there is no simple remedy for dealing with the complexities of shark hazards and reinforce the importance of trialing mitigation measures under local conditions.

18.
J Cardiovasc Pharmacol ; 58(5): 479-86, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21753735

ABSTRACT

The anaphylatoxin C5a generated by activation of the innate immunity complement system is a potent inflammatory peptide mediator through the G-protein-coupled receptor C5aR (CD88) present in immune-inflammatory cells, including monocytes, macrophages, neutrophils, T cells, and mast cells. Inflammatory cells infiltrate and initiate the development of fibrosis in the chronically hypertensive heart. In this study, we have investigated whether treatment with a selective C5aR antagonist prevents cardiovascular remodeling in deoxycorticosterone acetate (DOCA)-salt hypertensive rats. Control and DOCA-salt rats were treated with PMX53 (AcF-[OPdChaWR], 1 mg·kg·d oral gavage) for 32 days; structural and functional changes in cardiovascular system were determined. DOCA-salt hypertension increased leukocyte extravasation into ventricular tissue, increasing collagen deposition and ventricular stiffness; PMX53 treatment attenuated these changes, thereby improving cardiac function. Further, treatment with PMX53 suppressed an increased expression of C5aR in the left ventricle from DOCA-salt rats, consistent with the reduced infiltration of inflammatory cells. Vascular endothelial dysfunction in thoracic aortic rings was attenuated by PMX53 treatment, but systolic blood pressure was unchanged in DOCA-salt rats. In the heart, PMX53 treatment attenuated inflammatory cell infiltration, fibrosis, and ventricular stiffness, indicating that C5aR is critically involved in ventricular remodeling by regulating inflammatory responses in the hypertensive heart.


Subject(s)
Desoxycorticosterone/pharmacology , Endomyocardial Fibrosis/prevention & control , Hypertension/complications , Inflammation/prevention & control , Peptides, Cyclic/therapeutic use , Receptor, Anaphylatoxin C5a/antagonists & inhibitors , Acetylcholine/pharmacology , Animals , Aorta, Thoracic/drug effects , Aorta, Thoracic/physiopathology , Blood Pressure/drug effects , Cardiac Output/drug effects , Cicatrix/pathology , Cicatrix/prevention & control , Collagen/metabolism , Coronary Circulation/drug effects , Echocardiography , Endomyocardial Fibrosis/etiology , Endomyocardial Fibrosis/metabolism , Endomyocardial Fibrosis/pathology , Gene Expression/drug effects , Heart/drug effects , Heart/physiopathology , Heart Ventricles/metabolism , Heart Ventricles/pathology , Heart Ventricles/physiopathology , Hypertension/chemically induced , Hypertrophy, Left Ventricular/pathology , Hypertrophy, Left Ventricular/prevention & control , Inflammation/etiology , Inflammation/pathology , Leukocytes, Mononuclear/pathology , Male , Mast Cells/pathology , Myocardium/pathology , Nitroprusside/pharmacology , Norepinephrine/pharmacology , Peptides, Cyclic/pharmacology , Rats , Rats, Wistar , Receptor, Anaphylatoxin C5a/metabolism , Vasoconstriction/drug effects , Vasodilation/drug effects , Ventricular Dysfunction/physiopathology , Ventricular Dysfunction/prevention & control
19.
J Immunol ; 183(2): 1375-83, 2009 Jul 15.
Article in English | MEDLINE | ID: mdl-19561098

ABSTRACT

Alzheimer's disease (AD) is an age-related dementia, characterized by amyloid plaques, neurofibrillary tangles, neuroinflammation, and neuronal loss in the brain. Components of the complement system, known to produce a local inflammatory reaction, are associated with the plaques and tangles in AD brain, and thus a role for complement-mediated inflammation in the acceleration or progression of disease has been proposed. A complement activation product, C5a, is known to recruit and activate microglia and astrocytes in vitro by activation of a G protein-coupled cell-surface C5aR. Here, oral delivery of a cyclic hexapeptide C5a receptor antagonist (PMX205) for 2-3 mo resulted in substantial reduction of pathological markers such as fibrillar amyloid deposits (49-62%) and activated glia (42-68%) in two mouse models of AD. The reduction in pathology was correlated with improvements in a passive avoidance behavioral task in Tg2576 mice. In 3xTg mice, PMX205 also significantly reduced hyperphosphorylated tau (69%). These data provide the first evidence that inhibition of a proinflammatory receptor-mediated function of the complement cascade (i.e., C5aR) can interfere with neuroinflammation and neurodegeneration in AD rodent models, suggesting a novel therapeutic target for reducing pathology and improving cognitive function in human AD patients.


Subject(s)
Alzheimer Disease/drug therapy , Nerve Degeneration/prevention & control , Peptides, Cyclic/pharmacology , Receptor, Anaphylatoxin C5a/antagonists & inhibitors , Receptors, Complement/antagonists & inhibitors , Alzheimer Disease/pathology , Alzheimer Disease/physiopathology , Animals , Disease Models, Animal , Inflammation/prevention & control , Mice , Neurofibrillary Tangles/drug effects , Neurofibrillary Tangles/pathology , Neuroglia/pathology , Peptides, Cyclic/administration & dosage , Peptides, Cyclic/therapeutic use , Personality Disorders/prevention & control , Plaque, Amyloid/drug effects , Plaque, Amyloid/pathology
20.
J Neurochem ; 113(2): 389-401, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20132482

ABSTRACT

Alzheimer's disease (AD), a progressive neurodegenerative disease characterized by the accumulation of amyloid-beta protein and neuronal loss, is the leading cause of age-related dementia in the world today. The disease is also associated with neuroinflammation, robust activation of astrocytes and microglia, and evidence of activation of the complement system, localized with both fibrillar amyloid-beta (fAbeta) plaques and tangles. The observations are consistent with a complement-dependent component of AD progression. We have previously shown that inhibition of the major complement receptor for C5a (CD88) with the antagonist PMX205 results in a significant reduction in pathology in two mouse models of AD. To further characterize the role of complement in AD-related neuroinflammation, we examined the age- and disease-associated expression of CD88 in brain of transgenic mouse models of AD and the influence of PMX205 on the presence of various complement activation products using flow cytometry, western blot, and immunohistochemistry. CD88 was found to be up-regulated in microglia, in the immediate vicinity of amyloid plaques. While thioflavine plaque load and glial recruitment is significantly reduced after treatment with PMX205, C1q remains co-localized with fAbeta plaques and C3 is still expressed by the recruited astrocytes. Thus, with PMX205, potentially beneficial activities of these early complement components may remain intact, while detrimental activities resulting from C5a-CD88 interaction are inhibited. This further supports the targeted inhibition of specific complement mediated activities as an approach for AD therapy.


Subject(s)
Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Gene Expression Regulation/genetics , Microglia/metabolism , Receptor, Anaphylatoxin C5a/metabolism , Statistics as Topic/methods , Age Factors , Amyloid beta-Protein Precursor/genetics , Animals , Animals, Newborn , Cerebral Cortex/pathology , Disease Models, Animal , Flow Cytometry/methods , Gene Expression Regulation/drug effects , Glial Fibrillary Acidic Protein/metabolism , Hippocampus/pathology , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Mutation/genetics , Neurons/metabolism , Peptides, Cyclic/pharmacology , Receptor, Anaphylatoxin C5a/antagonists & inhibitors , Receptor, Anaphylatoxin C5a/deficiency
SELECTION OF CITATIONS
SEARCH DETAIL