Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 67
Filter
1.
Front Immunol ; 14: 1149822, 2023.
Article in English | MEDLINE | ID: mdl-37283747

ABSTRACT

Dysregulated NLRP3 inflammasome activation drives a wide variety of diseases, while endogenous inhibition of this pathway is poorly characterised. The serum protein C4b-binding protein (C4BP) is a well-established inhibitor of complement with emerging functions as an endogenously expressed inhibitor of the NLRP3 inflammasome signalling pathway. Here, we identified that C4BP purified from human plasma is an inhibitor of crystalline- (monosodium urate, MSU) and particulate-induced (silica) NLRP3 inflammasome activation. Using a C4BP mutant panel, we identified that C4BP bound these particles via specific protein domains located on the C4BP α-chain. Plasma-purified C4BP was internalised into MSU- or silica-stimulated human primary macrophages, and inhibited MSU- or silica-induced inflammasome complex assembly and IL-1ß cytokine secretion. While internalised C4BP in MSU or silica-stimulated human macrophages was in close proximity to the inflammasome adaptor protein ASC, C4BP had no direct effect on ASC polymerisation in in vitro assays. C4BP was also protective against MSU- and silica-induced lysosomal membrane damage. We further provide evidence for an anti-inflammatory function for C4BP in vivo, as C4bp-/- mice showed an elevated pro-inflammatory state following intraperitoneal delivery of MSU. Therefore, internalised C4BP is an inhibitor of crystal- or particle-induced inflammasome responses in human primary macrophages, while murine C4BP protects against an enhanced inflammatory state in vivo. Our data suggests C4BP has important functions in retaining tissue homeostasis in both human and mice as an endogenous serum inhibitor of particulate-stimulated inflammasome activation.


Subject(s)
Inflammasomes , NLR Family, Pyrin Domain-Containing 3 Protein , Animals , Humans , Mice , Complement C4b-Binding Protein/metabolism , Inflammasomes/metabolism , Macrophages/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Silicon Dioxide/pharmacology
2.
Article in English | MEDLINE | ID: mdl-32923410

ABSTRACT

Mammalian species contain an internal circadian (i.e., 24-h) clock that is synchronized to the day and night cycles. Large epidemiological studies, which are supported by carefully controlled studies in numerous species, support the idea that chronic disruption of our circadian cycles results in a number of health issues, including obesity and diabetes, defective immune response, and cancer. Here we focus specifically on the role of the complement immune system and its relationship to the internal circadian clock system. While still an incompletely understood area, there is evidence that dysregulated proinflammatory cytokines, complement factors, and oxidative stress can be induced by circadian disruption and that these may feed back into the oscillator at the level of circadian gene regulation. Such a feedback cycle may contribute to impaired host immune response against pathogenic insults. The complement immune system including its activated anaphylatoxins, C3a and C5a, not only facilitate innate and adaptive immune response in chemotaxis and phagocytosis, but they can also amplify chronic inflammation in the host organism. Consequent development of autoimmune disorders, and metabolic diseases associated with additional environmental insults that activate complement can in severe cases, lead to accelerated tissue dysfunction, fibrosis, and ultimately organ failure. Because several promising complement-targeted therapeutics to block uncontrolled complement activation and treat autoimmune diseases are in various phases of clinical trials, understanding fully the circadian properties of the complement system, and the reciprocal regulation by these two systems could greatly improve patient treatment in the long term.


Subject(s)
Circadian Clocks , Anaphylatoxins , Animals , Complement System Proteins , Humans , Immune System , Immunity
3.
FASEB J ; 34(6): 7540-7560, 2020 06.
Article in English | MEDLINE | ID: mdl-32301538

ABSTRACT

The vascular endothelium has been discovered in the past several years to be important in shaping the cellular immune response. During the immune response the vascular endothelium is constantly perturbed by biologically potent molecules, including the complement activation peptides, C3a and C5a. Despite the importance of C3a and C5a in inflammation and immunity, their role in modulating lymphocyte function via activation of vascular endothelial cells is unknown. Accordingly, we investigated the regulated expression of the C3a and C5a receptors (complement anaphylatoxin C3a receptor [C3aR] and complement anaphylatoxin C5a receptor 1 [C5aR1]) on human umbilical vascular endothelial cells (HUVECs) and examined how C3a or C5a activation of HUVECs affects the activation and polarization of lymphatic cells. Our findings demonstrated that C3a and C5a increase C3aR and C5aR1 expression by HUVECs as well as directing their cellular transmigration and spreading through transwell filters. Moreover, C3a- or C5a-stimulated endothelial cells: (1) caused activation of B-lymphoblasts with significant increase in Fas Ligand (CD95L) (FasL), CD69, and IL-R1 expression, and (2) skewed T-lymphoblast cells toward a Th1 subtype, (CD4+ /CCR5+ ) that correlated with significant increase of IFN-γ. Collectively, these data indicate that C3a and C5a signaling is important in the activation and polarization of lymphocytes as they traffic through the vascular endothelium during the immune response.


Subject(s)
Anaphylatoxins/immunology , B-Lymphocytes/immunology , Complement C3a/immunology , Complement C5a/immunology , Peptides/immunology , T-Lymphocytes/immunology , Cells, Cultured , Complement Activation/immunology , Endothelium, Vascular/immunology , Human Umbilical Vein Endothelial Cells , Humans , Inflammation/immunology , Receptor, Anaphylatoxin C5a/immunology , Receptors, Complement/immunology , Signal Transduction/immunology
4.
Diabetes ; 69(1): 83-98, 2020 01.
Article in English | MEDLINE | ID: mdl-31624141

ABSTRACT

The sequelae of diabetes include microvascular complications such as diabetic kidney disease (DKD), which involves glucose-mediated renal injury associated with a disruption in mitochondrial metabolic agility, inflammation, and fibrosis. We explored the role of the innate immune complement component C5a, a potent mediator of inflammation, in the pathogenesis of DKD in clinical and experimental diabetes. Marked systemic elevation in C5a activity was demonstrated in patients with diabetes; conventional renoprotective agents did not therapeutically target this elevation. C5a and its receptor (C5aR1) were upregulated early in the disease process and prior to manifest kidney injury in several diverse rodent models of diabetes. Genetic deletion of C5aR1 in mice conferred protection against diabetes-induced renal injury. Transcriptomic profiling of kidney revealed diabetes-induced downregulation of pathways involved in mitochondrial fatty acid metabolism. Interrogation of the lipidomics signature revealed abnormal cardiolipin remodeling in diabetic kidneys, a cardinal sign of disrupted mitochondrial architecture and bioenergetics. In vivo delivery of an orally active inhibitor of C5aR1 (PMX53) reversed the phenotypic changes and normalized the renal mitochondrial fatty acid profile, cardiolipin remodeling, and citric acid cycle intermediates. In vitro exposure of human renal proximal tubular epithelial cells to C5a led to altered mitochondrial respiratory function and reactive oxygen species generation. These experiments provide evidence for a pivotal role of the C5a/C5aR1 axis in propagating renal injury in the development of DKD by disrupting mitochondrial agility, thereby establishing a new immunometabolic signaling pathway in DKD.


Subject(s)
Complement C5a/physiology , Diabetes Mellitus, Experimental , Diabetic Nephropathies , Kidney/pathology , Mitochondria/metabolism , Animals , Cells, Cultured , Complement C5a/genetics , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Diabetic Nephropathies/genetics , Diabetic Nephropathies/metabolism , Diabetic Nephropathies/pathology , Energy Metabolism/genetics , Fibrosis/genetics , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Rats , Rats, Sprague-Dawley , Receptor, Anaphylatoxin C5a/physiology , Signal Transduction
5.
J Ocul Pharmacol Ther ; 36(1): 65-69, 2020.
Article in English | MEDLINE | ID: mdl-31596637

ABSTRACT

Purpose: Age-related macular degeneration (AMD) is a common disease trending towards epidemic proportions and is a leading cause of irreversible vision loss in people over the age of 65. A pathomechanism of AMD is death and/or dysfunction of retinal pigment epithelial (RPE) cells; RPE loss invariably results in photoreceptor atrophy. Treatment options for AMD are very limited, and include vitamin supplements and lifestyle changes. An exciting potential therapy currently being tested in clinical trials is transplantation of stem cell-derived RPE. Methods: We developed a NIH-registered embryonic stem line (CR-4), and in this study set out to determine if CR4-RPE are tolerated in normal mice and in murine models of retinal degeneration by injecting a bolus of CR4-RPE cells in the subretinal space of immunosuppressed wild-type, Mer mutant (Merkd), and Elovl4 deficient mice. Results: Mice with CR-RPE grafts were monitored daily, were examined routinely using OCT, and histology was prepared and examined at terminal end-points. Based on the parameters of the study, none of the animals with CR-RPE grafts (n=36) experienced any obvious adverse reactions. Conclusions: We conclude that transplanted CR-4 hES-derived RPE cells are well tolerated in immunosuppressed healthy and dystrophic murine retinas.


Subject(s)
Human Embryonic Stem Cells/cytology , Macular Degeneration/therapy , Retinal Pigment Epithelium/cytology , Animals , Disease Models, Animal , Eye Proteins/metabolism , Humans , Macular Degeneration/metabolism , Membrane Proteins/deficiency , Membrane Proteins/metabolism , Mice , Mice, Knockout
6.
Mol Immunol ; 118: 91-98, 2020 02.
Article in English | MEDLINE | ID: mdl-31862673

ABSTRACT

The purpose of this study was to identify a membrane-bound complement inhibitor that could be overexpressed on retinal pigment epithelial cells (RPE) providing a potential therapy for age-related macular degeneration (AMD). This type of therapy may allow replacement of damaged RPE with cells that are able to limit complement activation in the retina. Complement Receptor 1 (CR1) is a membrane-bound complement inhibitor commonly found on erythrocytes and immune cells. In this study, QPCR and flow cytometry data demonstrated that CR1 is not well-expressed by RPE, indicating that its overexpression may provide extra protection from complement activation. To screen CR1 for this ability, a stable CR1-expressing ARPE19 line was created using a combination of antibiotic selection and FACS. Cell-based assays were used to demonstrate that addition of CR1 inhibited deposition of complement proteins C3b and C6 on the transfected line. In the end, this study identifies CR1 as a complement inhibitor that may be overexpressed on stem cell-derived RPE to create a potential "enhanced" cell therapy for AMD. A combination cell/complement therapy may create transplantable RPE better suited to avoid complement-mediated lysis and limit chronic inflammation in the retina.


Subject(s)
Epithelial Cells/immunology , Macular Degeneration/immunology , Receptors, Complement 3b/immunology , Retina/immunology , Retinal Pigment Epithelium/immunology , Retinal Pigments/immunology , Cell Line , Complement Activation/immunology , Complement C3b/immunology , Complement C6/immunology , Erythrocytes/immunology , Humans
7.
J Immunol ; 203(10): 2701-2711, 2019 11 15.
Article in English | MEDLINE | ID: mdl-31597707

ABSTRACT

Infection with Listeria monocytogenes is acquired through ingestion of contaminated foods and may lead to systemic infection and possible death, with an overall 20% mortality rate. Our previous work using C5aR1-/- mice and C3aR-/- mice demonstrated that C5aR1 and C3aR both play powerful anti-inflammatory and prosurvival roles during systemic infection with L. monocytogenes In our current study, we have examined the role of the third anaphylatoxin receptor, C5aR2, in the host immune response to systemic L. monocytogenes infection. C5aR2-/- mice had significantly lower bacterial burdens in the spleens and livers on both day 1 and 3 postinfection compared with C5aR2+/+ mice. The decreased bacterial burdens in the C5aR2-/- mice correlated with less liver damage and with improved survival of CD4+ and CD8+ T cells in the spleen on day 3 postinfection compared with C5aR2+/+ mice. C5aR2-/- mice also produced significantly less G-CSF, IL-6, and MCP-1 in the serum, spleen, and liver on day 1 postinfection compared with C5aR2+/+ mice. C5aR2-/- and C5aR2+/+ mice produced similar amounts of IFN-γ in their spleens on day 1 postinfection. Purified naive splenocytes from C5aR2-/- mice produced significantly more IFN-γ and IL-12p70 during in vitro infection with L. monocytogenes compared with splenocytes from C5aR2+/+ mice in an NF-κB-dependent manner. Induction of IL-12 and IFN-γ early during infection with L. monocytogenes is protective to the host, and we believe this innate increased ability to produce more IL-12 and IFN-γ provided early protection to the C5aR2-/- mice.


Subject(s)
Disease Resistance/immunology , Host Microbial Interactions/immunology , Listeria monocytogenes/immunology , Listeriosis/immunology , Receptor, Anaphylatoxin C5a/metabolism , Animals , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Survival/immunology , Complement C5a/metabolism , Interferon-gamma/metabolism , Interleukin-12/metabolism , Listeriosis/microbiology , Liver/immunology , Liver/microbiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptor, Anaphylatoxin C5a/genetics , Spleen/immunology , Spleen/microbiology , Spleen/pathology
8.
Virulence ; 10(1): 677-694, 2019 12.
Article in English | MEDLINE | ID: mdl-31274379

ABSTRACT

The complement system is pivotal in the defense against invasive disease caused by Neisseria meningitidis (Nme, meningococcus), particularly via the membrane attack complex. Complement activation liberates the anaphylatoxins C3a and C5a, which activate three distinct G-protein coupled receptors, C3aR, C5aR1 and C5aR2 (anaphylatoxin receptors, ATRs). We recently discovered that C5aR1 exacerbates the course of the disease, revealing a downside of complement in Nme sepsis. Here, we compared the roles of all three ATRs during mouse nasal colonization, intraperitoneal infection and human whole blood infection with Nme. Deficiency of complement or ATRs did not alter nasal colonization, but significantly affected invasive disease: Compared to WT mice, the disease was aggravated in C3ar-/- mice, whereas C5ar1-/- and C5ar2-/- mice showed increased resistance to meningococcal sepsis. Surprisingly, deletion of either of the ATRs resulted in lower cytokine/chemokine responses, irrespective of the different susceptibilities of the mice. This was similar in ex vivo human whole blood infection using ATR inhibitors. Neutrophil responses to Nme were reduced in C5ar1-/- mouse blood. Upon stimulation with C5a plus Nme, mouse macrophages displayed reduced phosphorylation of ERK1/2, when C5aR1 or C5aR2 were ablated or inhibited, suggesting that both C5a-receptors prime an initial macrophage response to Nme. Finally, in vivo blockade of C5aR1 alone (PMX205) or along with C5aR2 (A8Δ71-73) resulted in ameliorated disease, whereas neither antagonizing C3aR (SB290157) nor its activation with a "super-agonist" peptide (WWGKKYRASKLGLAR) demonstrated a benefit. Thus, C5aR1 and C5aR2 augment disease pathology and are interesting targets for treatment, whereas C3aR is protective in experimental meningococcal sepsis.


Subject(s)
Meningococcal Infections/immunology , Neisseria meningitidis/immunology , Receptor, Anaphylatoxin C5a/immunology , Receptors, Complement/immunology , Anaphylatoxins/immunology , Animals , Chemokines/immunology , Cytokines/immunology , Humans , Macrophages/immunology , Macrophages/microbiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neisseria meningitidis/pathogenicity , Neutrophils/immunology , Neutrophils/microbiology , Receptor, Anaphylatoxin C5a/genetics , Receptors, Complement/genetics , Sepsis
9.
Stem Cell Res ; 39: 101508, 2019 08.
Article in English | MEDLINE | ID: mdl-31357114

ABSTRACT

Monosomy of chromosome X is associated with high prenatal mortality of female embryos and severe developmental abnormalities of patients born with Turner's syndrome (45,XO). The CDMLe012-A-1 human embryonic stem cell (hESC) line, derived from a day six blastocyst with a normal 46,XX female karyotype spontaneously lost an X-chromosome during cell culture. This 45,XO karyotype was stably maintained for more than 55 passages. Since the CDMLe012-A-1 cells express pluripotent stem cell markers and differentiate into cells derived from the three germ layers, the cell line represents a stable, pluripotent stem cell model of Turner's syndrome.


Subject(s)
Human Embryonic Stem Cells/cytology , Human Embryonic Stem Cells/metabolism , Turner Syndrome/genetics , Blastocyst/cytology , Blastocyst/metabolism , Cell Differentiation/genetics , Cell Differentiation/physiology , Female , Humans , Karyotype , Karyotyping , Pregnancy
10.
JCI Insight ; 52019 05 21.
Article in English | MEDLINE | ID: mdl-31112138

ABSTRACT

Alteration of innate immune cells in the lungs can promote loss of peripheral tolerance that leads to autoimmune responses in cigarette smokers. Development of autoimmunity in smokers with emphysema is also strongly linked to the expansion of autoreactive T helper (Th) cells expressing interferon gamma (Th1), and interleukin 17A (Th17). However, the mechanisms responsible for enhanced self-recognition and reduced immune tolerance in smoker with emphysema remain less clear. Here we show that C1q, a component of the complement protein 1 complex (C1), is downregulated in lung CD1a+ antigen presenting cells (APCs) isolated from emphysematous human, and mouse lung APCs after chronic cigarette smoke exposure. C1q potentiated the function of APCs to differentiate CD4+ T cells to Tregs, while it inhibited Th17 cell development and proliferation. Mice deficient in C1q that were exposed to chronic smoke exhibited exaggerated lung inflammation marked by increased Th17 cells, while reconstitution of C1q in the lungs enhanced Tregs abundance, dampened smoke-induced lung inflammation, and reversed established emphysema. Our findings demonstrate that cigarette smoke-mediated loss of C1q could play a key role in reduced peripheral tolerance, which could be explored to treat emphysema.


Subject(s)
Antigen-Presenting Cells/metabolism , Cigarette Smoking/adverse effects , Complement C1q/metabolism , Emphysema/immunology , Th17 Cells/immunology , Adult , Aged , Animals , Antigen-Presenting Cells/immunology , Autoimmunity , Case-Control Studies , Cell Differentiation/immunology , Cell Proliferation , Cells, Cultured , Cigarette Smoking/immunology , Coculture Techniques , Complement C1q/genetics , Complement C1q/immunology , Disease Models, Animal , Down-Regulation/immunology , Emphysema/pathology , Female , Gene Expression Profiling , Gene Knockdown Techniques , Humans , Immune Tolerance , Lung/cytology , Lung/immunology , Lung/pathology , Lymphocyte Activation , Male , Mice , Mice, Knockout , Middle Aged , Primary Cell Culture , Smoke/adverse effects , T-Lymphocytes, Regulatory/immunology , Tissue Array Analysis , Tobacco Products/adverse effects
12.
Mol Neurodegener ; 12(1): 66, 2017 09 18.
Article in English | MEDLINE | ID: mdl-28923083

ABSTRACT

BACKGROUND: Pharmacologic inhibition of C5aR1, a receptor for the complement activation proinflammatory fragment, C5a, suppressed pathology and cognitive deficits in Alzheimer's disease (AD) mouse models. To validate that the effect of the antagonist was specifically via C5aR1 inhibition, mice lacking C5aR1 were generated and compared in behavior and pathology. In addition, since C5aR1 is primarily expressed on cells of the myeloid lineage, and only to a lesser extent on endothelial cells and neurons in brain, gene expression in microglia isolated from adult brain at multiple ages was compared across all genotypes. METHODS: C5aR1 knock out mice were crossed to the Arctic AD mouse model, and characterized for pathology and for behavior performance in a hippocampal dependent memory task. CX3CR1GFP and CCR2RFP reporter mice were bred to C5aR1 sufficient and knockout wild type and Arctic mice to enable sorting of microglia (GFP-positive, RFP-negative) isolated from adult brain at 2, 5, 7 and 10 months of age followed by RNA-seq analysis. RESULTS: A lack of C5aR1 prevented behavior deficits at 10 months, although amyloid plaque load was not altered. Immunohistochemical analysis showed no CCR2+ monocytes/macrophages near the plaques in the Arctic brain with or without C5aR1. Microglia were sorted from infiltrating monocytes (GFP and RFP-positive) for transcriptome analysis. RNA-seq analysis identified inflammation related genes as differentially expressed, with increased expression in the Arctic mice relative to wild type and decreased expression in the Arctic/C5aR1KO relative to Arctic. In addition, phagosomal-lysosomal gene expression was increased in the Arctic mice relative to wild type but further increased in the Arctic/C5aR1KO mice. A decrease in neuronal complexity was seen in hippocampus of 10 month old Arctic mice at the time that correlates with the behavior deficit, both of which were rescued in the Arctic/C5aR1KO. CONCLUSIONS: These data are consistent with microglial polarization in the absence of C5aR1 signaling reflecting decreased induction of inflammatory genes and enhancement of degradation/clearance pathways, which is accompanied by preservation of CA1 neuronal complexity and hippocampal dependent cognitive function. These results provide links between microglial responses and loss of cognitive performance and, combined with the previous pharmacological approach to inhibit C5aR1 signaling, support the potential of this receptor as a novel therapeutic target for AD in humans.


Subject(s)
Alzheimer Disease/metabolism , Hippocampus/metabolism , Inflammation/metabolism , Microglia/metabolism , Receptor, Anaphylatoxin C5a/metabolism , Alzheimer Disease/pathology , Animals , Cognition , Hippocampus/pathology , Humans , Inflammation/pathology , Mice , Mice, Knockout , Microglia/pathology , Receptor, Anaphylatoxin C5a/deficiency , Signal Transduction/physiology
13.
Stem Cell Res ; 18: 37-40, 2017 01.
Article in English | MEDLINE | ID: mdl-28395800

ABSTRACT

The CR-4 human embryonic stem cell line was derived from the inner cell mass of a developing blastocyst. This cell line has been adapted to grow in feeder-free conditions and is especially well-suited for differentiation to retinal pigment epithelium. The line demonstrates a normal human 46,XX female karyotype. Pluripotency was assessed through qRT-PCR for expression of NANOG, OCT-4, and SOX-2. A teratoma assay was performed and results were positive for all three germ layers. Testing for Mycoplasma was negative.


Subject(s)
Human Embryonic Stem Cells/cytology , Retinal Pigment Epithelium/cytology , Animals , Blastocyst/cytology , Cell Differentiation , Cell Line , Female , Human Embryonic Stem Cells/metabolism , Humans , Karyotype , Mice , Mice, Inbred NOD , Mice, SCID , Microscopy, Fluorescence , Phagocytosis , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/transplantation , Teratoma/metabolism , Teratoma/pathology , Transcription Factors/genetics , Transcription Factors/metabolism
14.
J Immunol ; 198(8): 3237-3244, 2017 04 15.
Article in English | MEDLINE | ID: mdl-28275134

ABSTRACT

Listeria monocytogenes is an intracellular Gram-positive bacterium that induces expression of type I IFNs (IFN-α/IFN-ß) during infection. These cytokines are detrimental to the host during infection by priming leukocytes to undergo L. monocytogenes-mediated apoptosis. Our previous studies showed that C5aR1-/- and C3aR-/- mice are highly susceptible to L. monocytogenes infection as a result of increased IFN-ß-mediated apoptosis of major leukocyte cell populations, including CD4+ and CD8+ T cells. However, the mechanisms by which C3a and C5a modulate IFN-ß expression during L. monocytogenes infection were not examined in these initial investigations. Accordingly, we report in this article that C5a and C3a suppress IFN-ß production in response to L. monocytogenes via cyclic di-AMP (c-di-AMP), a secondary messenger molecule of L. monocytogenes, in J774A.1 macrophage-like cells and in bone marrow-derived dendritic cells (BMDCs). Moreover, C5a and C3a suppress IFN-ß production by acting through their respective receptors, because no inhibition was seen in C5aR1-/- or C3aR-/- BMDCs, respectively. C5a and C3a suppress IFN-ß production in a manner that is dependent on Bruton's tyrosine kinase, p38 MAPK, and TANK-binding kinase 1 (TBK1), as demonstrated by the individual use of Bruton's tyrosine kinase, p38 MAPK, and TBK1 inhibitors. Pretreatment of cells with C5a and C3a reduced the expression of the IFN-ß signaling molecules DDX41, STING, phosphorylated TBK1, and phosphorylated p38 MAPK in wild-type BMDCs following treatment with c-di-AMP. Collectively, these data demonstrate that C3a and C5a, via direct signaling through their specific receptors, suppress IFN-ß expression by modulation of a distinct innate cytosolic surveillance pathway involving DDX41, STING, and other downstream molecular targets of L. monocytogenes-generated c-di-AMP.


Subject(s)
Complement C3a/immunology , Complement C5a/immunology , Immunity, Innate/immunology , Interferon-beta/immunology , Listeriosis/immunology , Signal Transduction/immunology , Animals , Blotting, Western , Complement C3a/metabolism , Complement C5a/metabolism , Cyclic AMP , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Interferon-beta/biosynthesis , Listeria monocytogenes , Listeriosis/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout
15.
Brain ; 140(2): 353-369, 2017 02.
Article in English | MEDLINE | ID: mdl-27956400

ABSTRACT

Ischaemic stroke induces endogenous repair processes that include proliferation and differentiation of neural stem cells and extensive rewiring of the remaining neural connections, yet about 50% of stroke survivors live with severe long-term disability. There is an unmet need for drug therapies to improve recovery by promoting brain plasticity in the subacute to chronic phase after ischaemic stroke. We previously showed that complement-derived peptide C3a regulates neural progenitor cell migration and differentiation in vitro and that C3a receptor signalling stimulates neurogenesis in unchallenged adult mice. To determine the role of C3a-C3a receptor signalling in ischaemia-induced neural plasticity, we subjected C3a receptor-deficient mice, GFAP-C3a transgenic mice expressing biologically active C3a in the central nervous system, and their respective wild-type controls to photothrombotic stroke. We found that C3a overexpression increased, whereas C3a receptor deficiency decreased post-stroke expression of GAP43 (P < 0.01), a marker of axonal sprouting and plasticity, in the peri-infarct cortex. To verify the translational potential of these findings, we used a pharmacological approach. Daily intranasal treatment of wild-type mice with C3a beginning 7 days after stroke induction robustly increased synaptic density (P < 0.01) and expression of GAP43 in peri-infarct cortex (P < 0.05). Importantly, the C3a treatment led to faster and more complete recovery of forepaw motor function (P < 0.05). We conclude that C3a-C3a receptor signalling stimulates post-ischaemic neural plasticity and intranasal treatment with C3a receptor agonists is an attractive approach to improve functional recovery after ischaemic brain injury.


Subject(s)
Brain Ischemia/drug therapy , Complement C3a/therapeutic use , Neuronal Plasticity/drug effects , Administration, Intranasal , Animals , Brain Infarction/drug therapy , Brain Infarction/etiology , Brain Ischemia/complications , Brain Ischemia/diagnostic imaging , Brain Ischemia/genetics , CD11b Antigen/genetics , CD11b Antigen/metabolism , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Disease Models, Animal , Functional Laterality/drug effects , Functional Laterality/genetics , GAP-43 Protein/metabolism , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Glial Fibrillary Acidic Protein/metabolism , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neuronal Plasticity/genetics , Psychomotor Performance/drug effects , Recovery of Function/drug effects , Recovery of Function/genetics , S100 Calcium Binding Protein beta Subunit/metabolism , Synapsins/metabolism , Vesicular Glutamate Transport Protein 1/metabolism
16.
Biomol Ther (Seoul) ; 25(2): 130-139, 2017 Mar 01.
Article in English | MEDLINE | ID: mdl-27737526

ABSTRACT

CXCR5+ T follicular helper (Tfh) cells are associated with aberrant autoantibody production in patients with antibody-mediated autoimmune diseases including lupus. Follicular regulatory T (Tfr) cells expressing CXCR5 and Bcl6 have been recently identified as a specialized subset of Foxp3+ regulatory T (Treg) cells that control germinal center reactions. In this study, we show that retroviral transduction of CXCR5 gene in Foxp3+ Treg cells induced a stable expression of functional CXCR5 on their surface. The Cxcr5-transduced Treg cells maintained the expression of Treg cell signature genes and the suppressive activity. The expression of CXCR5 as well as Foxp3 in the transduced Treg cells appeared to be stable in vivo in an adoptive transfer experiment. Moreover, Cxcr5-transduced Treg cells preferentially migrated toward the CXCL13 gradient, leading to an effective suppression of antibody production from B cells stimulated with Tfh cells. Therefore, our results demonstrate that enforced expression of CXCR5 onto Treg cells efficiently induces Tfr cell-like properties, which might be a promising cellular therapeutic approach for the treatment of antibody-mediated autoimmune diseases.

18.
Immunobiology ; 221(12): 1407-1417, 2016 12.
Article in English | MEDLINE | ID: mdl-27476791

ABSTRACT

Listeria monocytogenes is a leading cause of foodborne-illness associated mortality that has attracted considerable attention in recent years due to several significant outbreaks. It has also served as a model organism for the study of intracellular pathogens. For these reasons the host response to L. monocytogenes has long been the subject of investigation. A potent innate and adaptive immune response is required for containment and clearance of L. monocytogenes. However, some elements of this response, such as type 1 interferons, can be detrimental to the host. Recent studies have revealed novel functions for the complement system, an ancient arm of innate immunity, in this process. Here we review the role of complement in the host response to L. monocytogenes.


Subject(s)
Complement System Proteins/immunology , Listeria monocytogenes/immunology , Listeriosis/immunology , Adaptive Immunity , Animals , Complement Activation , Host-Pathogen Interactions , Humans , Immunity, Innate , Interferon Type I/metabolism
19.
PLoS Pathog ; 12(6): e1005678, 2016 06.
Article in English | MEDLINE | ID: mdl-27304426

ABSTRACT

Spores of Bacillus anthracis, the causative agent of anthrax, are known to persist in the host lungs for prolonged periods of time, however the underlying mechanism is poorly understood. In this study, we demonstrated that BclA, a major surface protein of B. anthracis spores, mediated direct binding of complement factor H (CFH) to spores. The surface bound CFH retained its regulatory cofactor activity resulting in C3 degradation and inhibition of downstream complement activation. By comparing results from wild type C57BL/6 mice and complement deficient mice, we further showed that BclA significantly contributed to spore persistence in the mouse lungs and dampened antibody responses to spores in a complement C3-dependent manner. In addition, prior exposure to BclA deletion spores (ΔbclA) provided significant protection against lethal challenges by B. anthracis, whereas the isogenic parent spores did not, indicating that BclA may also impair protective immunity. These results describe for the first time an immune inhibition mechanism of B. anthracis mediated by BclA and CFH that promotes spore persistence in vivo. The findings also suggested an important role of complement in persistent infections and thus have broad implications.


Subject(s)
Anthrax/immunology , Complement Factor H/immunology , Immune Tolerance/immunology , Membrane Glycoproteins/immunology , Spores, Bacterial/immunology , Animals , Anthrax/metabolism , Bacillus anthracis/immunology , Bacillus anthracis/metabolism , Complement Factor H/metabolism , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred C57BL , Spores, Bacterial/metabolism
20.
Stem Cells Transl Med ; 4(10): 1234-45, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26285657

ABSTRACT

UNLABELLED: Human embryonic stem cells (hESCs) are a promising source of cells for tissue regeneration, yet histoincompatibility remains a major challenge to their clinical application. Because the human leukocyte antigen class I (HLA-I) molecules are the primary mediators of immune rejection, we hypothesized that cells derived from a hESC line lacking HLA-I expression could be transplanted without evoking a robust immune response from allogeneic recipients. In the present study, we used the replacement targeting strategy to delete exons 2 and 3 of ß2-microglobulin on both gene alleles in hESCs. Because ß2-microglobulin serves as the HLA-I light chain, disruption of the ß2-microglobulin gene led to complete HLA-I deficiency on the cell surface of hESCs and their derivatives. Therefore, these cells were resistant to CD8+ T-cell-mediated destruction. Although interferon-γ (IFN-γ) treatment significantly induced ß2-microglobulin expression, promoting CD8+ T cell-mediated killing of control hESCs and their derivatives, CD8+ T-cell-mediated cytotoxicity was barely observed with ß2-microglobulin-null hESCs and their derivatives treated with IFN-γ. This genetic manipulation to disrupt HLA-I expression did not affect the self-renewal capacity, genomic stability, or pluripotency of hESCs. Despite being relatively sensitive to natural killer (NK) cell-mediated killing due to the lack of HLA-I expression, when transplanted into NK cell-depleted immunocompetent mice, ß2-microglobulin-null hESCs developed into tumors resembling those derived from control hESCs in severe combined immunodeficiency mice. These results demonstrate that ß2-microglobulin-null hESCs significantly reduce immunogenicity to CD8+ T cells and might provide a renewable source of cells for tissue regeneration without the need for HLA matching in the future. SIGNIFICANCE: This study reports the generation of a novel ß2-microglobulin (B2M)-/- human embryonic stem cell (hESC) line. Differentiated mature cells from this line do not express cell surface human leukocyte antigen molecules even after interferon-γ stimulation and are resistant to alloreactive CD8+ T cells. Moreover, this B2M-/- hESC line contains no off-target integration or cleavage events, is devoid of stable B2M mRNA, exhibits a normal karyotype, and retains its self-renewal capacity, genomic stability, and pluripotency. Although B2M-/- hESC-derived cells are more susceptible to natural killer (NK) cells, murine transplantation studies have indicated that they are, overall, much less immunogenic than normal hESCs. Thus, these data show for the first time that, in vivo, the advantages provided by B2M-/- hESC-derived cells in avoiding CD8+ T-cell killing appear significantly greater than any disadvantage caused by increased susceptibility to NK cells.


Subject(s)
Gene Knockdown Techniques , Human Embryonic Stem Cells/immunology , beta 2-Microglobulin/genetics , Alleles , Animals , CD8-Positive T-Lymphocytes/immunology , Cell Self Renewal , Cell Survival , Cell Transplantation/adverse effects , Cytotoxicity, Immunologic , Exons/genetics , Gene Expression Profiling , Genetic Vectors , Genomic Instability , Graft Rejection/prevention & control , HLA Antigens , Heterografts , Histocompatibility , Human Embryonic Stem Cells/cytology , Human Embryonic Stem Cells/transplantation , Humans , Interferon-gamma/pharmacology , Killer Cells, Natural/immunology , Mice , Mice, SCID , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/immunology , Pluripotent Stem Cells/transplantation , Sequence Deletion , Teratoma/etiology , Teratoma/immunology , beta 2-Microglobulin/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...