Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 77
Filter
1.
Arterioscler Thromb Vasc Biol ; 42(7): 886-902, 2022 07.
Article in English | MEDLINE | ID: mdl-35477279

ABSTRACT

BACKGROUND: The vascular endothelium maintains tissue-fluid homeostasis by controlling the passage of large molecules and fluid between the blood and interstitial space. The interaction of catenins and the actin cytoskeleton with VE-cadherin (vascular endothelial cadherin) is the primary mechanism for stabilizing AJs (adherens junctions), thereby preventing lung vascular barrier disruption. Members of the Rho (Ras homology) family of GTPases and conventional GEFs (guanine exchange factors) of these GTPases have been demonstrated to play important roles in regulating endothelial permeability. Here, we evaluated the role of DOCK4 (dedicator of cytokinesis 4)-an unconventional Rho family GTPase GEF in vascular function. METHODS: We generated mice deficient in DOCK4' used DOCK4 silencing and reconstitution approaches in human pulmonary artery endothelial cells' used assays to evaluate protein localization, endothelial cell permeability, and small GTPase activation. RESULTS: Our data show that DOCK4-deficient mice are viable. However, these mice have hemorrhage selectively in the lung, incomplete smooth muscle cell coverage in pulmonary vessels, increased basal microvascular permeability, and impaired response to S1P (sphingosine-1-phosphate)-induced reversal of thrombin-induced permeability. Consistent with this, DOCK4 rapidly translocates to the cell periphery and associates with the detergent-insoluble fraction following S1P treatment, and its absence prevents S1P-induced Rac-1 activation and enhancement of barrier function. Moreover, DOCK4-silenced pulmonary artery endothelial cells exhibit enhanced basal permeability in vitro that is associated with enhanced Rho GTPase activation. CONCLUSIONS: Our findings indicate that DOCK4 maintains AJs necessary for lung vascular barrier function by establishing the normal balance between RhoA (Ras homolog family member A) and Rac-1-mediated actin cytoskeleton remodeling, a previously unappreciated function for the atypical GEF family of molecules. Our studies also identify S1P as a potential upstream regulator of DOCK4 activity.


Subject(s)
Endothelial Cells , rho GTP-Binding Proteins , Adherens Junctions/metabolism , Animals , Capillary Permeability/physiology , Cells, Cultured , Endothelial Cells/metabolism , Endothelium, Vascular/metabolism , GTPase-Activating Proteins/genetics , GTPase-Activating Proteins/metabolism , Guanine Nucleotide Exchange Factors/genetics , Guanine Nucleotide Exchange Factors/metabolism , Lung/metabolism , Mice , rho GTP-Binding Proteins/metabolism
2.
J Exp Med ; 219(5)2022 05 02.
Article in English | MEDLINE | ID: mdl-35404389

ABSTRACT

Monocytes undergo phenotypic and functional changes in response to inflammatory cues, but the molecular signals that drive different monocyte states remain largely undefined. We show that monocytes acquire macrophage markers upon glomerulonephritis and may be derived from CCR2+CX3CR1+ double-positive monocytes, which are preferentially recruited, dwell within glomerular capillaries, and acquire proinflammatory characteristics in the nephritic kidney. Mechanistically, the transition to immature macrophages begins within the vasculature and relies on CCR2 in circulating cells and TNFR2 in parenchymal cells, findings that are recapitulated in vitro with monocytes cocultured with TNF-TNFR2-activated endothelial cells generating CCR2 ligands. Single-cell RNA sequencing of cocultures defines a CCR2-dependent monocyte differentiation path associated with the acquisition of immune effector functions and generation of CCR2 ligands. Immature macrophages are detected in the urine of lupus nephritis patients, and their frequency correlates with clinical disease. In conclusion, CCR2-dependent functional specialization of monocytes into macrophages begins within the TNF-TNFR2-activated vasculature and may establish a CCR2-based autocrine, feed-forward loop that amplifies renal inflammation.


Subject(s)
Endothelial Cells , Monocytes , Receptors, CCR2 , Receptors, Tumor Necrosis Factor, Type II , Humans , Ligands , Macrophages , Receptors, CCR2/genetics , Receptors, Tumor Necrosis Factor, Type II/genetics
3.
Med ; 2(9): 1050-1071.e7, 2021 09 10.
Article in English | MEDLINE | ID: mdl-34414383

ABSTRACT

BACKGROUND: T cells control viral infection, promote vaccine durability, and in coronavirus disease 2019 (COVID-19) associate with mild disease. We investigated whether prior measles-mumps-rubella (MMR) or tetanus-diphtheria-pertussis (Tdap) vaccination elicits cross-reactive T cells that mitigate COVID-19. METHODS: Antigen-presenting cells (APC) loaded ex vivo with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), MMR, or Tdap antigens and autologous T cells from COVID-19-convalescent participants, uninfected individuals, and COVID-19 mRNA-vaccinated donors were co-cultured. T cell activation and phenotype were detected by interferon-γ (IFN-γ) enzyme-linked immunospot (ELISpot) assays and flow cytometry. ELISAs (enzyme-linked immunosorbant assays) and validation studies identified the APC-derived cytokine(s) driving T cell activation. TCR clonotyping and single-cell RNA sequencing (scRNA-seq) identified cross-reactive T cells and their transcriptional profile. A propensity-weighted analysis of COVID-19 patients estimated the effects of MMR and Tdap vaccination on COVID-19 outcomes. FINDINGS: High correlation was observed between T cell responses to SARS-CoV-2 (spike-S1 and nucleocapsid) and MMR and Tdap proteins in COVID-19-convalescent and -vaccinated individuals. The overlapping T cell population contained an effector memory T cell subset (effector memory re-expressing CD45RA on T cells [TEMRA]) implicated in protective, anti-viral immunity, and their detection required APC-derived IL-15, known to sensitize T cells to activation. Cross-reactive TCR repertoires detected in antigen-experienced T cells recognizing SARS-CoV-2, MMR, and Tdap epitopes had TEMRA features. Indices of disease severity were reduced in MMR- or Tdap-vaccinated individuals by 32%-38% and 20%-23%, respectively, among COVID-19 patients. CONCLUSIONS: Tdap and MMR memory T cells reactivated by SARS-CoV-2 may provide protection against severe COVID-19. FUNDING: This study was supported by a National Institutes of Health (R01HL065095, R01AI152522, R01NS097719) donation from Barbara and Amos Hostetter and the Chleck Foundation.


Subject(s)
COVID-19 , Measles , Whooping Cough , COVID-19/prevention & control , Humans , Mumps Vaccine , Receptors, Antigen, T-Cell , Rubella Vaccine , SARS-CoV-2 , Spike Glycoprotein, Coronavirus , T-Lymphocytes
4.
Nat Commun ; 12(1): 4791, 2021 08 09.
Article in English | MEDLINE | ID: mdl-34373452

ABSTRACT

Classical dendritic cells (cDC) are professional antigen-presenting cells (APC) that regulate immunity and tolerance. Neutrophil-derived cells with properties of DCs (nAPC) are observed in human diseases and after culture of neutrophils with cytokines. Here we show that FcγR-mediated endocytosis of antibody-antigen complexes or an anti-FcγRIIIB-antigen conjugate converts neutrophils into nAPCs that, in contrast to those generated with cytokines alone, activate T cells to levels observed with cDCs and elicit CD8+ T cell-dependent anti-tumor immunity in mice. Single cell transcript analyses and validation studies implicate the transcription factor PU.1 in neutrophil to nAPC conversion. In humans, blood nAPC frequency in lupus patients correlates with disease. Moreover, anti-FcγRIIIB-antigen conjugate treatment induces nAPCs that can activate autologous T cells when using neutrophils from individuals with myeloid neoplasms that harbor neoantigens or those vaccinated against bacterial toxins. Thus, anti-FcγRIIIB-antigen conjugate-induced conversion of neutrophils to immunogenic nAPCs may represent a possible immunotherapy for cancer and infectious diseases.


Subject(s)
Antigens, Neoplasm/immunology , Neoplasms/immunology , Neutrophils/immunology , Receptors, IgG/genetics , Receptors, IgG/immunology , Adaptor Proteins, Vesicular Transport/genetics , Animals , Antigen Presentation/immunology , Antigen-Antibody Complex , Bone Marrow , CD4-Positive T-Lymphocytes , CD8-Positive T-Lymphocytes , Cell Movement , Cell Proliferation , Cytokines/immunology , Dendritic Cells/immunology , Endocytosis , Humans , Immunity, Innate , Immunotherapy , Lymph Nodes/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Differentiation Factor 88/genetics , Reactive Oxygen Species , Transcriptome
5.
bioRxiv ; 2021 May 04.
Article in English | MEDLINE | ID: mdl-33972940

ABSTRACT

T cells are critical for control of viral infection and effective vaccination. We investigated whether prior Measles-Mumps-Rubella (MMR) or Tetanus-Diphtheria-pertussis (Tdap) vaccination elicit cross-reactive T cells that mitigate COVID-19. Using co-cultures of antigen presenting cells (APC) loaded with antigens and autologous T cells, we found a high correlation between responses to SARS-CoV-2 (Spike-S1 and Nucleocapsid) and MMR and Tdap vaccine proteins in both SARS-CoV-2 infected individuals and individuals immunized with mRNA-based SARS-CoV-2 vaccines. The overlapping T cell population contained effector memory T cells (TEMRA) previously implicated in anti-viral immunity and their activation required APC-derived IL-15. TCR- and scRNA-sequencing detected cross-reactive clones with TEMRA features among the cells recognizing SARS-CoV-2, MMR and Tdap epitopes. A propensity-weighted analysis of 73,582 COVID-19 patients revealed that severe disease outcomes (hospitalization and transfer to intensive care unit or death) were reduced in MMR or Tdap vaccinated individuals by 38-32% and 23-20% respectively. In summary, SARS-CoV-2 re-activates memory T cells generated by Tdap and MMR vaccines, which may reduce disease severity.

6.
Cell Rep ; 35(7): 109142, 2021 05 18.
Article in English | MEDLINE | ID: mdl-34010642

ABSTRACT

The interaction of the human FcγRIIA with immune complexes (ICs) promotes neutrophil activation and thus must be tightly controlled to avoid damage to healthy tissue. Here, we demonstrate that a fungal-derived soluble ß-1,3/1,6-glucan binds to the glycosphingolipid long-chain lactosylceramide (LacCer) to reduce FcγRIIA-mediated recruitment to immobilized ICs under flow, a process requiring high-affinity FcγRIIA-immunoglobulin G (IgG) interactions. The inhibition requires Lyn phosphorylation of SHP-1 phosphatase and the FcγRIIA immunotyrosine-activating motif. ß-glucan reduces the effective 2D affinity of FcγRIIA for IgG via Lyn and SHP-1 and, in vivo, inhibits FcγRIIA-mediated neutrophil recruitment to intravascular IgG deposited in the kidney glomeruli in a glycosphingolipid- and Lyn-dependent manner. In contrast, ß-glucan did not affect FcγR functions that bypass FcγR affinity for IgG. In summary, we have identified a pathway for modulating the 2D affinity of FcγRIIA for ligand that relies on LacCer-Lyn-SHP-1-mediated inhibitory signaling triggered by ß-glucan, a previously described activator of innate immunity.


Subject(s)
Glycosphingolipids/metabolism , Ligands , Receptors, IgG/metabolism , Humans , Signal Transduction
7.
Nat Commun ; 10(1): 4097, 2019 09 10.
Article in English | MEDLINE | ID: mdl-31506438

ABSTRACT

Phenotypic variation of quantitative traits is orchestrated by a complex interplay between the environment (e.g. diet) and genetics. However, the impact of gene-environment interactions on phenotypic traits mostly remains elusive. To address this, we feed 1154 mice of an autoimmunity-prone intercross line (AIL) three different diets. We find that diet substantially contributes to the variability of complex traits and unmasks additional genetic susceptibility quantitative trait loci (QTL). By performing whole-genome sequencing of the AIL founder strains, we resolve these QTLs to few or single candidate genes. To address whether diet can also modulate genetic predisposition towards a given trait, we set NZM2410/J mice on similar dietary regimens as AIL mice. Our data suggest that diet modifies genetic susceptibility to lupus and shifts intestinal bacterial and fungal community composition, which precedes clinical disease manifestation. Collectively, our study underlines the importance of including environmental factors in genetic association studies.


Subject(s)
Crosses, Genetic , Diet , Genes , Genetic Association Studies , Quantitative Trait, Heritable , Animals , Animals, Outbred Strains , Antibodies, Antinuclear/genetics , Bacteria/growth & development , Biodiversity , Female , Fungi/growth & development , Genetic Predisposition to Disease , Lupus Nephritis/genetics , Lupus Nephritis/immunology , Male , Mice , Microbiota , Physical Chromosome Mapping , Quantitative Trait Loci/genetics , Spleen/metabolism , Transcriptome/genetics , Whole Genome Sequencing
8.
Ann Rheum Dis ; 78(2): 228-237, 2019 02.
Article in English | MEDLINE | ID: mdl-30459279

ABSTRACT

OBJECTIVE: Immune complexes (ICs) play a critical role in the pathology of autoimmune diseases. The aim of this study was to generate and characterise a first-in-class anti-FcγRIIA antibody (Ab) VIB9600 (previously known as MEDI9600) that blocks IgG immune complex-mediated cellular activation for clinical development. METHODS: VIB9600 was humanised and optimised from the IV.3 Ab. Binding affinity and specificity were determined by Biacore and ELISA. Confocal microscopy, Flow Cytometry-based assays and binding competition assays were used to assess the mode of action of the antibody. In vitro cell-based assays were used to demonstrate suppression of IC-mediated inflammatory responses. In vivo target suppression and efficacy was demonstrated in FcγRIIA-transgenic mice. Single-dose pharmacokinetic (PK)/pharmacodynamic study multiple dose Good Laboratory Practice (GLP) toxicity studies were conducted in non-human primates. RESULTS: We generated a humanised effector-deficient anti-FcγRIIA antibody (VIB9600) that potently blocks autoantibody and IC-mediated proinflammatory responses. VIB9600 suppresses FcγRIIA activation by blocking ligand engagement and by internalising FcγRIIA from the cell surface. VIB9600 inhibits IC-induced type I interferons from plasmacytoid dendritic cells (involved in SLE), antineutrophil cytoplasmic antibody (ANCA)-induced production of reactive oxygen species by neutrophils (involved in ANCA-associated vasculitis) and IC-induced tumour necrosis factor α and interleukin-6 production (involved in rheumatoid arthritis). In FcγRIIA transgenic mice, VIB9600 suppressed antiplatelet antibody-induced thrombocytopaenia, acute anti-GBM Ab-induced nephritis and anticollagen Ab-induced arthritis. VIB9600 also exhibited favourable PK and safety profiles in cynomolgus monkey studies. CONCLUSIONS: VIB9600 is a specific humanised antibody antagonist of FcγRIIA with null effector function that warrants further clinical development for the treatment of IC-mediated diseases.


Subject(s)
Antibodies, Anti-Idiotypic/pharmacology , Antigen-Antibody Complex/drug effects , Autoimmune Diseases/drug therapy , Immunologic Factors/pharmacology , Receptors, IgG/immunology , Animals , Antibodies, Antineutrophil Cytoplasmic/immunology , Antigen-Antibody Complex/immunology , Autoimmune Diseases/immunology , Dendritic Cells/immunology , Humans , Immunoglobulin G/immunology , Interleukin-6/immunology , Macaca fascicularis , Mice , Mice, Transgenic , Neutrophils/immunology , Reactive Oxygen Species/immunology , Tumor Necrosis Factor-alpha/immunology
9.
Curr Opin Rheumatol ; 31(2): 193-200, 2019 03.
Article in English | MEDLINE | ID: mdl-30540580

ABSTRACT

PURPOSE OF REVIEW: Systemic lupus erythematosus (SLE) is a multiorgan autoimmune disease characterized by IgG-autoantibodies to nuclear antigens that can deposit in the kidney and trigger lupus nephritis. Neutrophils accumulate in the kidneys of patients with proliferative LUPUS NEPHRITIS and neutrophil products and a subset of granulocytes, called low-density granulocytes (LDG) may contribute to lupus nephritis pathogenesis. Here, we will discuss recent studies implicating neutrophils in the pathogenesis of human SLE nephritis and then examine studies that provide mechanistic insights into how these cells are recruited to the glomerulus following immune complex deposition and how their products may promote lupus nephritis. RECENT FINDINGS: SLE patients display unique blood transcriptional signatures linked to Type I interferon and myeloblast differentiation, which could help stratify lupus nephritis progression. Multiphoton intravital microscopy of kidney glomerular capillaries revealed a role for neutrophil FcγRs in the rapid capture of neutrophils following immune complex deposition. The view that reduced degradation of neutrophil extracellular traps (NETS) contributes to lupus nephritis progression, is now challenged by experimental data in lupus-prone mice that genetically fail to produce NETS but still are afflicted. SUMMARY: A greater understanding of the neutrophil dependent mechanisms that promote lupus nephritis may potentially inform on newer therapeutic options that target neutrophil accumulation and reactivity in the nephritic kidney.


Subject(s)
Autoantibodies/immunology , Autoimmunity , Lupus Nephritis/immunology , Neutrophils/immunology , Animals , Extracellular Traps/immunology , Humans
10.
Nat Commun ; 9(1): 5058, 2018 11 29.
Article in English | MEDLINE | ID: mdl-30498196

ABSTRACT

Vascular-deposited IgG immune complexes promote neutrophil recruitment, but how this process is regulated is still unclear. Here we show that the CD18 integrin Mac-1, in its bent state, interacts with the IgG receptor FcγRIIA in cis to reduce the affinity of FcγRIIA for IgG and inhibit FcγRIIA-mediated neutrophil recruitment under flow. The Mac-1 rs1143679 lupus-risk variant reverses Mac-1 inhibition of FcγRIIA, as does a Mac-1 ligand and a mutation in Mac-1's ligand binding αI-domain. Sialylated complex glycans on FcγRIIA interact with the αI-domain via divalent cations, and this interaction is required for FcγRIIA inhibition by Mac-1. Human neutrophils deficient in CD18 integrins exhibit augmented FcγRIIA-dependent recruitment to IgG-coated endothelium. In mice, CD18 integrins on neutrophils dampen IgG-mediated neutrophil accumulation in the kidney. In summary, cis interaction between sialylated FcγRIIA and the αI-domain of Mac-1 alters the threshold for IgG-mediated neutrophil recruitment. A disruption of this interaction may increase neutrophil influx in autoimmune diseases.


Subject(s)
Macrophage-1 Antigen/metabolism , Neutrophils/metabolism , Receptors, IgG/metabolism , Animals , Basement Membrane/metabolism , Endothelium/metabolism , Glycosylation , HEK293 Cells , Humans , Immunoglobulin G/metabolism , Jurkat Cells/metabolism , Macrophage-1 Antigen/chemistry , Male , Mice , Nephritis/metabolism , Protein Structure, Secondary , Receptors, IgG/chemistry
11.
Nat Med ; 24(2): 232-238, 2018 02.
Article in English | MEDLINE | ID: mdl-29309057

ABSTRACT

Rhabdomyolysis is a serious syndrome caused by skeletal muscle injury and the subsequent release of breakdown products from damaged muscle cells into systemic circulation. The muscle damage most often results from strenuous exercise, muscle hypoxia, medications, or drug abuse and can lead to life-threatening complications, such as acute kidney injury (AKI). Rhabdomyolysis and the AKI complication can also occur during crush syndrome, an emergency condition that commonly occurs in victims of natural disasters, such as earthquakes, and man-made disasters, such as wars and terrorism. Myoglobin released from damaged muscle is believed to trigger renal dysfunction in this form of AKI. Recently, macrophages were implicated in the disease pathogenesis of rhabdomyolysis-induced AKI, but the precise molecular mechanism remains unclear. In the present study, we show that macrophages released extracellular traps (ETs) comprising DNA fibers and granule proteins in a mouse model of rhabdomyolysis. Heme-activated platelets released from necrotic muscle cells during rhabdomyolysis enhanced the production of macrophage extracellular traps (METs) through increasing intracellular reactive oxygen species generation and histone citrullination. Here we report, for the first time to our knowledge, this unanticipated role for METs and platelets as a sensor of myoglobin-derived heme in rhabdomyolysis-induced AKI. This previously unknown mechanism might be targeted for treatment of the disease. Finally, we found a new therapeutic tool for prevention of AKI after rhabdomyolysis, which might rescue some sufferers of this pathology.


Subject(s)
Acute Kidney Injury/genetics , Crush Syndrome/genetics , Platelet Activation/genetics , Rhabdomyolysis/genetics , Acute Kidney Injury/etiology , Acute Kidney Injury/pathology , Animals , Citrullination/genetics , Crush Syndrome/etiology , Crush Syndrome/pathology , DNA/genetics , DNA/metabolism , Disease Models, Animal , Extracellular Traps/genetics , Extracellular Traps/metabolism , Heme/metabolism , Histones/metabolism , Humans , Macrophages/metabolism , Macrophages/pathology , Mice , Muscle, Skeletal/injuries , Muscle, Skeletal/pathology , Myoglobin/genetics , Reactive Oxygen Species/metabolism , Rhabdomyolysis/complications , Rhabdomyolysis/pathology , Secretory Vesicles/genetics
12.
J Clin Invest ; 127(10): 3810-3826, 2017 Oct 02.
Article in English | MEDLINE | ID: mdl-28891817

ABSTRACT

The kidney glomerular capillaries are frequent sites of immune complex deposition and subsequent neutrophil accumulation in post-infectious and rapidly progressive glomerulonephritis. However, the mechanisms of neutrophil recruitment remain enigmatic, and there is no targeted therapeutic to avert this proximal event in glomerular inflammation. The uniquely human activating Fc receptor FcγRIIA promotes glomerular neutrophil accumulation and damage in anti-glomerular basement membrane-induced (anti-GBM-induced) glomerulonephritis when expressed on murine neutrophils. Here, we found that neutrophils are directly captured by immobilized IgG antibodies under physiological flow conditions in vitro through FcγRIIA-dependent, Abl/Src tyrosine kinase-mediated F-actin polymerization. Biophysical measurements showed that the lifetime of FcγRIIA-IgG bonds increased under mechanical force in an F-actin-dependent manner, which could enable the capture of neutrophils under physiological flow. Kidney intravital microscopy revealed that circulating neutrophils, which were similar in diameter to glomerular capillaries, abruptly arrested following anti-GBM antibody deposition via neutrophil FcγRIIA and Abl/Src kinases. Accordingly, inhibition of Abl/Src with bosutinib reduced FcγRIIA-mediated glomerular neutrophil accumulation and renal injury in experimental, crescentic anti-GBM nephritis. These data identify a pathway of neutrophil recruitment within glomerular capillaries following IgG deposition that may be targeted by bosutinib to avert glomerular injury.


Subject(s)
Aniline Compounds/pharmacology , Glomerulonephritis/immunology , Immunoglobulin G/immunology , Kidney Glomerulus/immunology , Neutrophils/immunology , Nitriles/pharmacology , Quinolines/pharmacology , Receptors, IgG/immunology , Animals , Capillaries/immunology , Capillaries/pathology , Glomerulonephritis/genetics , Glomerulonephritis/pathology , HL-60 Cells , Humans , Kidney Glomerulus/pathology , Mice , Mice, Knockout , Neutrophils/pathology , Proto-Oncogene Proteins c-abl/antagonists & inhibitors , Proto-Oncogene Proteins c-abl/genetics , Proto-Oncogene Proteins c-abl/immunology , Receptors, IgG/genetics , src-Family Kinases/antagonists & inhibitors , src-Family Kinases/immunology
13.
JCI Insight ; 2(10)2017 May 18.
Article in English | MEDLINE | ID: mdl-28515361

ABSTRACT

Though recent reports suggest that neutrophil extracellular traps (NETs) are a source of antigenic nucleic acids in systemic lupus erythematosus (SLE), we recently showed that inhibition of NETs by targeting the NADPH oxidase complex via cytochrome b-245, ß polypeptide (cybb) deletion exacerbated disease in the MRL.Faslpr lupus mouse model. While these data challenge the paradigm that NETs promote lupus, it is conceivable that global regulatory properties of cybb and cybb-independent NETs confound these findings. Furthermore, recent reports indicate that inhibitors of peptidyl arginine deiminase, type IV (Padi4), a distal mediator of NET formation, improve lupus in murine models. Here, to clarify the contribution of NETs to SLE, we employed a genetic approach to delete Padi4 in the MRL.Faslpr model and used a pharmacological approach to inhibit PADs in both the anti-glomerular basement membrane model of proliferative nephritis and a human-serum-transfer model of SLE. In contrast to prior inhibitor studies, we found that deletion of Padi4 did not ameliorate any aspect of nephritis, loss of tolerance, or immune activation. Pharmacological inhibition of PAD activity had no effect on end-organ damage in inducible models of glomerulonephritis. These data provide a direct challenge to the concept that NETs promote autoimmunity and target organ injury in SLE.

14.
J Leukoc Biol ; 101(2): 493-505, 2017 02.
Article in English | MEDLINE | ID: mdl-27965383

ABSTRACT

CD47 is known to play an important role in CD4+ T cell homeostasis. We recently reported a reduction in mice deficient in the Cd47 gene (Cd47-/-) CD4+ T cell adhesion and transendothelial migration (TEM) in vivo and in vitro as a result of impaired expression of high-affinity forms of LFA-1 and VLA-4 integrins. A prior study concluded that Cd47-/- mice were resistant to experimental autoimmune encephalomyelitis (EAE) as a result of complete failure in CD4+ T cell activation after myelin oligodendrocyte glycoprotein peptide 35-55 aa (MOG35-55) immunization. As the prior EAE study was published before our report, authors could not have accounted for defects in T cell integrin function as a mechanism to protect Cd47-/- in EAE. Thus, we hypothesized that failure of T cell activation involved defects in LFA-1 and VLA-4 integrins. We confirmed that Cd47-/- mice were resistant to MOG35-55-induced EAE. Our data, however, supported a different mechanism that was not a result of failure of CD4+ T cell activation. Instead, we found that CD4+ T cells in MOG35-55-immunized Cd47-/- mice were activated, but clonal expansion contracted within 72 h after immunization. We used TCR crosslinking and mitogen activation in vitro to investigate the underlying mechanism. We found that naïve Cd47-/- CD4+ T cells exhibited a premature block in proliferation and survival because of impaired activation of LFA-1, despite effective TCR-induced activation. These results identify CD47 as an important regulator of LFA-1 and VLA-4 integrin-adhesive functions in T cell proliferation, as well as recruitment, and clarify the roles played by CD47 in MOG35-55-induced EAE.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD47 Antigen/metabolism , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/prevention & control , Lymphocyte Function-Associated Antigen-1/metabolism , Adoptive Transfer , Animals , Antibodies, Monoclonal/pharmacology , Antigen Presentation/drug effects , Antigen Presentation/immunology , CD28 Antigens/metabolism , CD4-Positive T-Lymphocytes/drug effects , Cell Adhesion/drug effects , Cell Proliferation/drug effects , Chemokines/pharmacology , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Humans , Immunization , Integrin alpha4beta1/metabolism , Intercellular Adhesion Molecule-1/metabolism , Lymph Nodes/drug effects , Lymph Nodes/pathology , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Mice, Inbred C57BL , Myelin-Oligodendrocyte Glycoprotein/immunology , Receptors, Antigen, T-Cell/metabolism , Vascular Cell Adhesion Molecule-1/metabolism
15.
Nat Commun ; 7: 12993, 2016 Sep 29.
Article in English | MEDLINE | ID: mdl-27680869

ABSTRACT

Inducible cAMP early repressor (ICER) has been described as a transcriptional repressor isoform of the cAMP response element modulator (CREM). Here we report that ICER is predominantly expressed in Th17 cells through the IL-6-STAT3 pathway and binds to the Il17a promoter, where it facilitates the accumulation of the canonical enhancer RORγt. In vitro differentiation from naive ICER/CREM-deficient CD4+ T cells to Th17 cells is impaired but can be rescued by forced overexpression of ICER. Consistent with a role of Th17 cells in autoimmune and inflammatory diseases, ICER/CREM-deficient B6.lpr mice are protected from developing autoimmunity. Similarly, both anti-glomerular basement membrane-induced glomerulonephritis and experimental encephalomyelitis are attenuated in ICER/CREM-deficient mice compared with their ICER/CREM-sufficient littermates. Importantly, we find ICER overexpressed in CD4+ T cells from patients with systemic lupus erythematosus. Collectively, our findings identify a unique role for ICER, which affects both organ-specific and systemic autoimmunity in a Th17-dependent manner.

16.
EBioMedicine ; 10: 204-15, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27453322

ABSTRACT

Neutrophils are central players in the innate immune system. They generate neutrophil extracellular traps (NETs), which protect against invading pathogens but are also associated with the development of autoimmune and/or inflammatory diseases and thrombosis. Here, we report that lactoferrin, one of the components of NETs, translocated from the cytoplasm to the plasma membrane and markedly suppressed NETs release. Furthermore, exogenous lactoferrin shrunk the chromatin fibers found in released NETs, without affecting the generation of oxygen radicals, but this failed after chemical removal of the positive charge of lactoferrin, suggesting that charge-charge interactions between lactoferrin and NETs were required for this function. In a model of immune complex-induced NET formation in vivo, intravenous lactoferrin injection markedly reduced the extent of NET formation. These observations suggest that lactoferrin serves as an intrinsic inhibitor of NETs release into the circulation. Thus, lactoferrin may represent a therapeutic lead for controlling NETs release in autoimmune and/or inflammatory diseases.


Subject(s)
Extracellular Traps/metabolism , Inflammation/immunology , Inflammation/metabolism , Lactoferrin/metabolism , Neutrophils/metabolism , Amino Acids , Cell Line , Cell Membrane/metabolism , Gene Silencing , Histones/metabolism , Humans , Inflammation/genetics , Lactoferrin/chemistry , Lactoferrin/genetics , Leukocyte Elastase/metabolism , Protein Transport , Proteolysis , RNA Interference , Reactive Oxygen Species/metabolism
17.
J Leukoc Biol ; 100(3): 581-8, 2016 09.
Article in English | MEDLINE | ID: mdl-26965632

ABSTRACT

The C-type lectin receptor dectin-1 and the integrin Mac-1 have key roles in controlling fungal infection. Here, we demonstrate that dectin-1- and Mac-1-induced activation of protein kinase Cδ in neutrophils, independent of the Card9 adaptor, is required for reactive oxygen species production and for intracellular killing upon Candida albicans uptake. Protein kinase Cδ was also required for zymosan-induced cytokine generation in neutrophils. In macrophages, protein kinase Cδ deficiency prevented fungi-induced reactive oxygen species generation but had no effect on activation of TGF-ß-activated kinase-1, an effector of Card9, or nuclear factor κB activation, nor did it affect phagolysosomal maturation, autophagy, or intracellular C. albicans killing. In vivo, protein kinase Cδ-deficient mice were highly susceptible to C. albicans and Aspergillus fumigatus infection, which was partially rescued with adoptively transferred wild-type neutrophils. Thus, protein kinase Cδ activation downstream of dectin-1 and Mac-1 has an important role in neutrophil, but not macrophage, functions required for host defense against fungal pathogens.


Subject(s)
Aspergillosis/immunology , Candidiasis/immunology , Macrophages/immunology , Neutrophils/immunology , Protein Kinase C-delta/physiology , Animals , Aspergillosis/metabolism , Aspergillosis/microbiology , Aspergillus fumigatus/immunology , CARD Signaling Adaptor Proteins/physiology , Candida albicans/immunology , Candidiasis/metabolism , Candidiasis/microbiology , Cytokines/metabolism , Female , Lectins, C-Type/metabolism , Macrophage-1 Antigen/metabolism , Macrophages/metabolism , Macrophages/microbiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/metabolism , Neutrophils/metabolism , Neutrophils/microbiology , Phagocytosis , Reactive Oxygen Species/metabolism , Signal Transduction , Transforming Growth Factor beta
18.
Nat Commun ; 7: 10828, 2016 Mar 04.
Article in English | MEDLINE | ID: mdl-26940548

ABSTRACT

Acute peritonitis is a frequent medical condition that can trigger severe sepsis as a life-threatening complication. Neutrophils are first-responders in infection but recruitment mechanisms to the abdominal cavity remain poorly defined. Here, we demonstrate that high endothelial venules (HEVs) of the greater omentum constitute a main entry pathway in TNFα-, Escherichia coli (E. coli)- and caecal ligation and puncture-induced models of inflammation. Neutrophil transmigration across HEVs is faster than across conventional postcapillary venules and requires a unique set of adhesion receptors including peripheral node addressin, E-, L-selectin and Mac-1 but not P-selectin or LFA-1. Omental milky spots readily concentrate intra-abdominal E. coli where macrophages and recruited neutrophils collaborate in phagocytosis and killing. Inhibition of the omental neutrophil response exacerbates septic progression of peritonitis. This data identifies HEVs as a clinically relevant vascular recruitment site for neutrophils in acute peritonitis that is indispensable for host defence against early systemic bacterial spread and sepsis.


Subject(s)
Neutrophils/physiology , Omentum/blood supply , Peritonitis/immunology , Sepsis/immunology , Venules/physiology , Animals , Antigens, Ly/genetics , Antigens, Ly/metabolism , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Escherichia coli , Escherichia coli Infections/immunology , Female , Gene Expression Regulation , Genes, Transgenic, Suicide , Male , Mice , Mice, Knockout , Tumor Necrosis Factor-alpha/metabolism
19.
Trends Mol Med ; 22(2): 151-163, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26778189

ABSTRACT

Kidney inflammation is a major contributor to progressive renal injury, leading to glomerulonephritis (GN) and chronic kidney disease. We review recent advances in our understanding of leukocyte accumulation in the kidney, emphasizing key chemokines involved in GN. We discuss features of renal inflammation such as the evolving concept of immune cell plasticity. We also describe certain aspects of organ-specific tissue microenvironments in shaping immune cell responses, as well as the current knowledge of how regulatory T lymphocytes impact on other immune effector cell populations to control inflammation. It is clear that present and future research in these areas may contribute to the development of novel targeted therapeutics, with the hope of alleviating the burden of end-stage renal disease (ESRD).


Subject(s)
Glomerulonephritis/drug therapy , Immunity, Innate/drug effects , Immunologic Factors/therapeutic use , Kidney Failure, Chronic/prevention & control , Renal Insufficiency, Chronic/drug therapy , Cell Movement/drug effects , Cellular Microenvironment/drug effects , Cellular Microenvironment/immunology , Cytokines/genetics , Cytokines/immunology , Gene Expression Regulation , Glomerulonephritis/immunology , Glomerulonephritis/pathology , Humans , Inflammation , Kidney/drug effects , Kidney/immunology , Kidney/pathology , Kidney Failure, Chronic/immunology , Kidney Failure, Chronic/pathology , Renal Insufficiency, Chronic/immunology , Renal Insufficiency, Chronic/pathology , Signal Transduction , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/pathology , Th1 Cells/drug effects , Th1 Cells/immunology , Th1 Cells/pathology
20.
Immunol Rev ; 269(1): 175-93, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26683153

ABSTRACT

Mac-1 (CD11b/CD18) is a ß2 integrin classically regarded as a pro-inflammatory molecule because of its ability to promote phagocyte cytotoxic functions and enhance the function of several effector molecules such as FcγR, uPAR, and CD14. Nevertheless, recent reports have revealed that Mac-1 also plays significant immunoregulatory roles, and genetic variants in ITGAM, the gene that encodes CD11b, confer risk for the autoimmune disease systemic lupus erythematosus (SLE). This has renewed interest in the physiological roles of this integrin and raised new questions on how its seemingly opposing biological functions may be regulated. Here, we provide an overview of the CD18 integrins and how their activation may be regulated as this may shed light on how the opposing roles of Mac-1 may be elicited. We then discuss studies that exemplify Mac-1's pro-inflammatory versus regulatory roles particularly in the context of IgG immune complex-mediated inflammation. This includes a detailed examination of molecular mechanisms that could explain the risk-conferring effect of rs1143679, a single nucleotide non-synonymous Mac-1 polymorphism associated with SLE.


Subject(s)
CD11b Antigen/metabolism , Immune Complex Diseases/immunology , Lupus Erythematosus, Systemic/immunology , Animals , CD11b Antigen/genetics , Genetic Predisposition to Disease , Humans , Immunomodulation , Phagocytosis , Polymorphism, Genetic , Risk
SELECTION OF CITATIONS
SEARCH DETAIL
...