Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
J Immunol ; 210(1): 19-23, 2023 01 01.
Article in English | MEDLINE | ID: mdl-36454023

ABSTRACT

T cell-independent (TI) B cell responses to nonprotein Ags involve multiple cues from the innate immune system. Neutrophils express complement receptors and activated neutrophils can release BAFF, but mechanisms effectively linking neutrophil activation to TI B cell responses are incompletely understood. Using germline and conditional knockout mice, we found that TI humoral responses involve alternative pathway complement activation and neutrophil-expressed C3a and C5a receptors (C3aR1/C5aR1) that promote BAFF-dependent B1 cell expansion and TI Ab production. Conditional absence of C3aR1/C5aR1 on neutrophils lowered serum BAFF levels, led to fewer Peyer's patch germinal center B cells, reduced germinal center B cells IgA class-switching, and lowered fecal IgA levels. Together, the results indicate that sequential activation of complement on neutrophils crucially supports humoral TI and mucosal IgA responses through upregulating neutrophil production of BAFF.


Subject(s)
B-Lymphocytes , Neutrophils , Mice , Animals , Complement System Proteins/metabolism , Mice, Knockout , Receptors, Complement/metabolism , Immunoglobulin A
2.
Curr Opin Organ Transplant ; 27(5): 399-404, 2022 10 01.
Article in English | MEDLINE | ID: mdl-35857345

ABSTRACT

PURPOSE OF REVIEW: In antibody-mediated allograft rejection, donor-reactive antibodies cause transplant injury in part via complement activation. New mechanistic insights indicate complement also modulates development of humoral immune responses. Herein we review recent data that describes how complement affects antibody formation and we discuss therapeutic implications. RECENT FINDINGS: Extravasating T cells interacting with integrins express and activate intracellular complement that drives immune-metabolic adaptations vital for CD4 + helper cells. Marginal zone B cells can acquire intact major histocompatibility complexes from dendritic cells via complement-dependent trogocytosis for presentation to T cells. Activated B cells in germinal centers receive co-stimulatory signals from T-helper cells. These germinal center B cells undergo coordinate shifts in surface complement regulator expression that permit complement receptor signaling on the germinal center B cells required for affinity maturation. The positively selected, high-affinity B cells can differentiate into plasma cells that produce donor-HLA-reactive antibodies capable of ligating endothelial, among other, graft cells. Subsequent sublytic complement attack can stimulate endothelial cells to activate CD4 + and CD8 + T cells, promoting cellular and humoral rejection. Newly developed complement inhibitors are being tested to prevent/treat transplant rejection. SUMMARY: The complement system influences T-cell, B-cell and endothelial-cell activation, and thereby contributes allograft injury. Emerging therapeutic strategies targeting complement activation have the potential to prevent or abrogate transplant injury and improve transplant outcomes.


Subject(s)
Antibody Formation , Isoantibodies , Complement System Proteins , Endothelial Cells/metabolism , Graft Rejection/prevention & control , Humans
3.
Transplant Cell Ther ; 28(8): 472.e1-472.e11, 2022 08.
Article in English | MEDLINE | ID: mdl-35643350

ABSTRACT

Conditioning regimens used for hematopoietic stem cell transplantation (HCT) can escalate the severity of acute T cell-mediated graft-versus-host disease (GVHD) by disrupting gastrointestinal integrity and initiating lipopolysaccharide (LPS)-dependent innate immune cell activation. Activation of the complement cascade has been associated with murine GVHD, and previous work has shown that alternative pathway complement activation can amplify T cell immunity. Whether and how mannan-binding lectin (MBL), a component of the complement system that binds mannose as well as oligosaccharide components of LPS and lipoteichoic acid, affects GVHD is unknown. In this study, we tested the hypothesis that MBL modulates murine GVHD and examined the mechanisms by which it does so. We adoptively transferred C3.SW bone marrow (BM) cells ± T cells into irradiated wild type (WT) or MBL-deficient C57Bl/6 (B6) recipients with or without inhibiting MBL-initiated complement activation using C1-esterase inhibitor (C1-INH). We analyzed the clinical severity of disease expression and analyzed intestinal gene and cell infiltration. In vitro studies assessed MBL expression on antigen-presenting cells (APCs) and compared LPS-induced responses of WT and MBL-deficient APCs. MBL-deficient recipients of donor BM ± T cells exhibited significantly less weight loss over the first 2 weeks post-transplantation weeks compared with B6 controls (P < .05), with similar donor engraftment in the 2 groups. In recipients of C3.SW BM + T cells, the clinical expression of GVHD was less severe (P < .05) and overall survival was better (P < .05) in MBL-deficient mice compared with WT mice. On day-7 post-transplantation, analyses showed that the MBL-deficient recipients exhibited less intestinal IL1b, IL17, and IL12 p40 gene expression (P < .05 for each) and fewer infiltrating intestinal CD11c+, CD11b+, and F4/80+ cells and TCRß+, CD4+, CD4+IL17+, and CD8+ T cells (P < .05 for each). Ovalbumin or allogeneic cell immunizations induced equivalent T cell responses in MBL-deficient and WT mice, demonstrating that MBL-deficiency does not directly impact T cell immunity in the absence of irradiation conditioning. Administration of C1-INH did not alter the clinical expression of GVHD in preconditioned WT B6 recipients, suggesting that MBL amplifies clinical expression of GVHD via a complement-independent mechanism. WT, but not MBL-deficient, APCs express MBL on their surfaces. LPS-stimulated APCs from MBL-deficient mice produced less proinflammatory cytokines (P < .05) and induced weaker alloreactive T cell responses (P < .05) compared with WT APCs. Together, our data show that MBL modulates murine GVHD, likely by amplifying complement-independent, LPS-initiated gastrointestinal inflammation. The results suggest that devising strategies to block LPS/MBL ligation on APCs has the potential to reduce the clinical expression of GVHD.


Subject(s)
Graft vs Host Disease , Inflammation , Mannose-Binding Lectin , Animals , Bone Marrow Transplantation , CD8-Positive T-Lymphocytes , Graft vs Host Disease/genetics , Inflammation/etiology , Inflammation/genetics , Lipopolysaccharides/adverse effects , Lipopolysaccharides/pharmacology , Mannose-Binding Lectin/genetics , Mice , Mice, Inbred C57BL , Transplantation, Homologous
4.
JCI Insight ; 6(19)2021 10 08.
Article in English | MEDLINE | ID: mdl-34473647

ABSTRACT

Herein, we report that Shroom3 knockdown, via Fyn inhibition, induced albuminuria with foot process effacement (FPE) without focal segmental glomerulosclerosis (FSGS) or podocytopenia. Interestingly, knockdown mice had reduced podocyte volumes. Human minimal change disease (MCD), where podocyte Fyn inactivation was reported, also showed lower glomerular volumes than FSGS. We hypothesized that lower glomerular volume prevented the progression to podocytopenia. To test this hypothesis, we utilized unilateral and 5/6th nephrectomy models in Shroom3-KD mice. Knockdown mice exhibited less glomerular and podocyte hypertrophy after nephrectomy. FYN-knockdown podocytes had similar reductions in podocyte volume, implying that Fyn was downstream of Shroom3. Using SHROOM3 or FYN knockdown, we confirmed reduced podocyte protein content, along with significantly increased phosphorylated AMPK, a negative regulator of anabolism. AMPK activation resulted from increased cytoplasmic redistribution of LKB1 in podocytes. Inhibition of AMPK abolished the reduction in glomerular volume and induced podocytopenia in mice with FPE, suggesting a protective role for AMPK activation. In agreement with this, treatment of glomerular injury models with AMPK activators restricted glomerular volume, podocytopenia, and progression to FSGS. Glomerular transcriptomes from MCD biopsies also showed significant enrichment of Fyn inactivation and Ampk activation versus FSGS glomeruli. In summary, we demonstrated the important role of AMPK in glomerular volume regulation and podocyte survival. Our data suggest that AMPK activation adaptively regulates glomerular volume to prevent podocytopenia in the context of podocyte injury.


Subject(s)
Adenylate Kinase/metabolism , Kidney Glomerulus/metabolism , Microfilament Proteins/genetics , Nephrotic Syndrome/genetics , Podocytes/metabolism , Adenylate Kinase/antagonists & inhibitors , Adolescent , Adult , Aged , Albuminuria/genetics , Animals , Cell Size , Cell Survival/genetics , Child , Child, Preschool , Female , Gene Knockdown Techniques , Glomerulonephritis, Membranous/genetics , Glomerulonephritis, Membranous/pathology , Glomerulosclerosis, Focal Segmental/genetics , Glomerulosclerosis, Focal Segmental/pathology , Humans , Hypertrophy , Infant , Kidney Glomerulus/pathology , Male , Mice , Middle Aged , Nephrectomy , Nephrosis, Lipoid/genetics , Nephrosis, Lipoid/pathology , Nephrotic Syndrome/pathology , Podocytes/pathology , Proportional Hazards Models , Proto-Oncogene Proteins c-fyn/genetics , Young Adult
5.
J Am Soc Nephrol ; 32(10): 2542-2560, 2021 10.
Article in English | MEDLINE | ID: mdl-34261755

ABSTRACT

BACKGROUND: Although high-affinity IgG auto- and alloantibodies are important drivers of kidney inflammation that can result in ESKD, therapeutic approaches that effectively reduce such pathogenic antibodies remain elusive. Erythropoietin (EPO) has immunomodulatory functions, but its effects on antibody production are unknown. METHODS: We assessed the effect and underlying mechanisms of EPO/EPO receptor (EPOR) signaling on primary and secondary, T cell-dependent and T-independent antibody formation using in vitro culture systems, murine models of organ transplantation and lupus nephritis, and mice conditionally deficient for the EPOR expressed on T cells or B cells. RESULTS: In wild-type mice, recombinant EPO inhibited primary, T cell-dependent humoral immunity to model antigens and strong, polyclonal stimuli, but did not alter T-independent humoral immune responses. EPO also significantly impaired secondary humoral immunity in a potent allogeneic organ transplant model system. The effects required T cell, but not B cell, expression of the EPOR and resulted in diminished frequencies of germinal center (GC) B cells and T follicular helper cells (TFH). In vitro and in vivo experiments showed that EPO directly prevented TFH differentiation and function via a STAT5-dependent mechanism that reduces CD4+ T cell expression of Bcl6. In lupus models, EPO reduced TFH, GC B cells, and autoantibody production, and abrogated autoimmune glomerulonephritis, demonstrating clinical relevance. In vitro studies verified that EPO prevents differentiation of human TFH cells. CONCLUSIONS: Our findings newly demonstrate that EPO inhibits TFH-dependent antibody formation, an observation with potential implications for treating antibody-mediated diseases, including those of the kidney.


Subject(s)
Antibody Formation/drug effects , Cell Differentiation/drug effects , Erythropoietin/pharmacology , Immunity, Humoral/drug effects , T Follicular Helper Cells/physiology , Animals , B-Lymphocytes/immunology , CD4 Lymphocyte Count , Cells, Cultured , Erythropoietin/genetics , Erythropoietin/metabolism , Female , Humans , Male , Mice , Phosphorylation , Receptors, Erythropoietin/metabolism , STAT5 Transcription Factor/metabolism , Signal Transduction , T Follicular Helper Cells/immunology , T Follicular Helper Cells/metabolism , T-Lymphocytes, Regulatory/immunology
6.
Nat Immunol ; 22(6): 757-768, 2021 06.
Article in English | MEDLINE | ID: mdl-34031614

ABSTRACT

Maturation of B cells within germinal centers (GCs) generates diversified B cell pools and high-affinity B cell antigen receptors (BCRs) for pathogen clearance. Increased receptor affinity is achieved by iterative cycles of T cell-dependent, affinity-based B cell positive selection and clonal expansion by mechanisms hitherto incompletely understood. Here we found that, as part of a physiologic program, GC B cells repressed expression of decay-accelerating factor (DAF/CD55) and other complement C3 convertase regulators via BCL6, but increased the expression of C5b-9 inhibitor CD59. These changes permitted C3 cleavage on GC B cell surfaces without the formation of membrane attack complex and activated C3a- and C5a-receptor signals required for positive selection. Genetic disruption of this pathway in antigen-activated B cells by conditional transgenic DAF overexpression or deletion of C3a and C5a receptors limited the activation of mechanistic target of rapamycin (mTOR) in response to BCR-CD40 signaling, causing premature GC collapse and impaired affinity maturation. These results reveal that coordinated shifts in complement regulation within the GC provide crucial signals underlying GC B cell positive selection.


Subject(s)
B-Lymphocytes/immunology , Complement Activation , Complement C3a/metabolism , Complement C5a/metabolism , Germinal Center/immunology , Animals , Animals, Genetically Modified , B-Lymphocytes/metabolism , CD55 Antigens/genetics , CD55 Antigens/metabolism , CD59 Antigens/metabolism , Cell Line, Tumor , Clonal Hematopoiesis/immunology , Germinal Center/cytology , Germinal Center/metabolism , Humans , Lymphocyte Activation , Mice , Palatine Tonsil/cytology , Palatine Tonsil/pathology , Proto-Oncogene Proteins c-bcl-6/metabolism , Receptor, Anaphylatoxin C5a/genetics , Receptor, Anaphylatoxin C5a/metabolism , Receptors, Antigen, B-Cell/metabolism , Receptors, Complement/genetics , Receptors, Complement/metabolism , Signal Transduction/immunology , TOR Serine-Threonine Kinases/metabolism
7.
8.
J Am Soc Nephrol ; 29(11): 2641-2657, 2018 11.
Article in English | MEDLINE | ID: mdl-30341149

ABSTRACT

BACKGROUND: We previously showed that the presence of a CKD-associated locus in SHROOM3 in a donor kidney results in increased expression of SHROOM3 (an F-actin-binding protein important for epithelial morphogenesis, via rho-kinase [ROCK] binding); this facilitates TGF-b signaling and allograft fibrosis. However, other evidence suggests Shroom3 may have a protective role in glomerular development. METHODS: We used human data, Shroom3 knockdown podocytes, and inducible shRNA-mediated knockdown mice to study the role of Shroom3 in adult glomeruli. RESULTS: Expression data from the Nephroseq database showed glomerular and nonglomerular SHROOM3 had opposing associations with renal function in CKD biopsy samples. In human allografts, homozygosity at rs17319721, the SHROOM3 locus linked with lower GFR, was associated with reduced albuminuria by 2 years after transplant. Although our previous data showed reduced renal fibrosis with tubular Shroom3 knockdown, this study found that glomerular but not tubular Shroom3 knockdown induced albuminuria. Electron microscopy revealed diffuse foot process effacement, and glomerular RNA-sequencing showed enrichment of tyrosine kinase signaling and podocyte actin cytoskeleton pathways in knockdown mice. Screening SHROOM3-interacting proteins identified FYN (a src-kinase) as a candidate.We confirmed the interaction of endogenous SHROOM3 with FYN in human podocytes via a critical Src homology 3-binding domain, distinct from its ROCK-binding domain. Shroom3-Fyn interaction was required in vitro and in vivo for activation of Fyn kinase and downstream nephrin phosphorylation in podocytes. SHROOM3 knockdown altered podocyte morphology, cytoskeleton, adhesion, and migration. CONCLUSIONS: We demonstrate a novel mechanism that may explain SHROOM3's dichotomous associations in glomerular versus nonglomerular compartments in CKD.


Subject(s)
Albuminuria/metabolism , Kidney Transplantation , Kidney/metabolism , Membrane Proteins/metabolism , Microfilament Proteins/metabolism , Proto-Oncogene Proteins c-fyn/metabolism , Actin Cytoskeleton/metabolism , Adolescent , Adult , Aged , Albuminuria/genetics , Albuminuria/pathology , Allografts , Animals , Child , Child, Preschool , Enhancer Elements, Genetic , Female , Gene Knockdown Techniques , Glomerular Filtration Rate/genetics , Homozygote , Humans , Kidney/pathology , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Microfilament Proteins/chemistry , Microfilament Proteins/deficiency , Microfilament Proteins/genetics , Middle Aged , Phosphorylation , Podocytes/metabolism , Podocytes/pathology , Polymorphism, Single Nucleotide , Proto-Oncogene Proteins c-fyn/chemistry , RNA, Small Interfering/genetics , Renal Insufficiency, Chronic/genetics , Renal Insufficiency, Chronic/metabolism , Renal Insufficiency, Chronic/surgery , Signal Transduction , Young Adult , src Homology Domains
9.
Ann Rheum Dis ; 75(6): 1236-45, 2016 06.
Article in English | MEDLINE | ID: mdl-26245757

ABSTRACT

OBJECTIVES: Gout is a highly inflammatory but self-limiting joint disease induced by the precipitation of monosodium urate (MSU) crystals. While it is well established that inflammasome activation by MSU mediates acute inflammation, little is known about the mechanism controlling its spontaneous resolution. The aim of this study was to analyse the role of neutrophil-derived microvesicles (PMN-Ecto) in the resolution of acute gout. METHODS: PMN-Ecto were studied in a murine model of MSU-induced peritonitis using C57BL/6, MerTK(-/-) and C5aR(-/-) mice. The peritoneal compartment was assessed for the number of infiltrating neutrophils (PMN), neutrophil microvesicles (PMN-Ecto), cytokines (interleukin-1ß, TGFß) and complement factors (C5a). Human PMN-Ecto were isolated from exudates of patients undergoing an acute gouty attack and functionally tested in vitro. RESULTS: C5a generated after the injection of MSU primed the inflammasome for IL-1ß release. Neutrophils infiltrating the peritoneum in response to C5a released phosphatidylserine (PS)-positive PMN-Ecto early on in the course of inflammation. These PMN-Ecto in turn suppressed C5a priming of the inflammasome and consequently inhibited IL-1ß release and neutrophil influx. PMN-Ecto-mediated suppression required surface expression of the PS-receptor MerTK and could be reproduced using PS-expressing liposomes. In addition, ectosomes triggered the release of TGFß independent of MerTK. TGFß, however, was not sufficient to control acute MSU-driven inflammation in vivo. Finally, PMN-Ecto from joint aspirates of patients with gouty arthritis had similar anti-inflammatory properties. CONCLUSIONS: PMN-Ecto-mediated control of inflammasome-driven inflammation is a compelling concept of autoregulation initiated early on during PMN activation in gout.


Subject(s)
Cell-Derived Microparticles/physiology , Complement C5a/immunology , Gout/pathology , Inflammasomes/physiology , Neutrophils/physiology , Acute Disease , Animals , Cell-Derived Microparticles/transplantation , Cells, Cultured , Complement C5a/biosynthesis , Cytokines/metabolism , Gout/immunology , Gout/metabolism , Humans , Inflammasomes/immunology , Inflammasomes/metabolism , Liposomes/metabolism , Mice, Inbred C57BL , Neutrophil Activation/physiology , Neutrophil Infiltration/physiology , Peritonitis/immunology , Peritonitis/pathology , Phosphatidylserines/metabolism
10.
Arterioscler Thromb Vasc Biol ; 34(2): 313-20, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24311376

ABSTRACT

OBJECTIVE: Transfusion of aged blood has been associated with increased morbidity and mortality in critically ill patients. During storage, erythrocytes release increasing numbers of microvesicles (red blood cell-derived microvesicles [RBC-MV]). We hypothesized that RBC-MV mediate some of the deleterious effects of aged blood transfusions. APPROACH AND RESULTS: We established a murine transfusion model using RBC-MV purified from aged mouse erythrocytes. Injection of RBC-MV into healthy mice had no effect. However, they aggravated pulmonary leukocyte sequestration and peripheral blood leukopenia induced by lipopolysaccharides. Lipopolysaccharide-induced proinflammatory cytokines were significantly increased in plasma after RBC-MV injection. These effects were not seen in C5aR-deficient mice. In vitro, RBC-MV bound C3 fragments after incubation with plasma but failed to bind immunoglobulins, C1q, or mannose-binding lectin. Preventing thrombin generation inhibited complement activation in vitro and in vivo and reversed the proinflammatory effects of RBC-MV in lipopolysaccharide-primed mice. Finally, the RBC-MV-induced phenotype was recapitulated using phosphatidylserine-expressing liposomes, suggesting that surface expression of phosphatidylserine by RBC-MV was mechanistically involved. CONCLUSIONS: These results point toward a thrombin-dependent mechanism of complement activation by RBC-MV independent of the classical, lectin, or alternative pathway. Besides identifying RBC-MV as potential mediators of transfusion-related morbidity, our findings may be relevant for other inflammatory disorders involving intravascular microvesicle release, for example, sickle cell disease or thrombotic microangiopathy.


Subject(s)
Cell-Derived Microparticles/immunology , Complement Activation , Erythrocyte Transfusion/adverse effects , Erythrocytes/immunology , Inflammation/immunology , Thrombin/immunology , Animals , Cell-Derived Microparticles/metabolism , Complement C3/immunology , Complement C3/metabolism , Erythrocytes/metabolism , Genotype , Inflammation/blood , Inflammation/chemically induced , Leukopenia/blood , Leukopenia/immunology , Lipopolysaccharides , Liposomes , Lung/immunology , Lung/metabolism , Mice , Mice, 129 Strain , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Models, Animal , Neutrophil Infiltration , Peptide Fragments/immunology , Peptide Fragments/metabolism , Phenotype , Phosphatidylserines/immunology , Phosphatidylserines/metabolism , Receptor, Anaphylatoxin C5a/deficiency , Receptor, Anaphylatoxin C5a/genetics , Thrombin/metabolism , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...