Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
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 ; 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
8.
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
9.
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
10.
Am J Pathol ; 186(2): 270-84, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26687990

ABSTRACT

The blood-testis barrier (BTB), formed between adjacent Sertoli cells, undergoes extensive remodeling to facilitate the transport of preleptotene spermatocytes across the barrier from the basal to apical compartments of the seminiferous tubules for further development and maturation into spermatozoa. The actin cytoskeleton serves unique structural and supporting roles in this process, but little is known about the role of microtubules and their regulators during BTB restructuring. The large isoform of the cAMP-responsive scaffold protein AKAP9 regulates microtubule dynamics and nucleation at the Golgi. We found that conditional deletion of Akap9 in mice after the initial formation of the BTB at puberty leads to infertility. Akap9 deletion results in marked alterations in the organization of microtubules in Sertoli cells and a loss of barrier integrity despite a relatively intact, albeit more apically localized F-actin and BTB tight junctional proteins. These changes are accompanied by a loss of haploid spermatids due to impeded meiosis. The barrier, however, progressively reseals in older Akap9 null mice, which correlates with a reduction in germ cell apoptosis and a greater incidence of meiosis. However, spermiogenesis remains defective, suggesting additional roles for AKAP9 in this process. Together, our data suggest that AKAP9 and, by inference, the regulation of the microtubule network are critical for BTB function and subsequent germ cell development during spermatogenesis.


Subject(s)
A Kinase Anchor Proteins/metabolism , Blood-Testis Barrier/metabolism , Germ Cells/cytology , Microtubule-Associated Proteins/metabolism , Microtubules/metabolism , Sertoli Cells/cytology , Testis/metabolism , Actin Cytoskeleton/metabolism , Actins/metabolism , Animals , Male , Mice , Spermatogenesis/physiology
11.
Nat Commun ; 6: 10182, 2015 Dec 18.
Article in English | MEDLINE | ID: mdl-26680259

ABSTRACT

The mechanisms driving T cell homing to lymph nodes and migration to tissue are well described but little is known about factors that affect T cell egress from tissues. Here, we generate mice with a T cell-specific deletion of the scaffold protein A kinase anchoring protein 9 (AKAP9) and use models of inflammatory disease to demonstrate that AKAP9 is dispensable for T cell priming and migration into tissues and lymph nodes, but is required for T cell retention in tissues. AKAP9 deficiency results in increased T cell egress to draining lymph nodes, which is associated with impaired T cell re-activation in tissues and protection from organ damage. AKAP9-deficient T cells exhibit reduced microtubule-dependent recycling of TCRs back to the cell surface and this affects antigen-dependent activation, primarily by non-classical antigen-presenting cells. Thus, AKAP9-dependent TCR trafficking drives efficient T cell re-activation and extends their retention at sites of inflammation with implications for disease pathogenesis.


Subject(s)
A Kinase Anchor Proteins/genetics , Cell Movement/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Lymph Nodes/immunology , Lymphocyte Activation/immunology , Microtubule-Associated Proteins/genetics , Nephritis/immunology , Reperfusion Injury/immunology , T-Lymphocytes/immunology , A Kinase Anchor Proteins/immunology , Animals , Antigen-Presenting Cells/immunology , Cell Adhesion/immunology , Cell Migration Assays, Leukocyte , Cells, Cultured , Endosomes , Gene Knockout Techniques , Glomerular Basement Membrane/immunology , In Vitro Techniques , Inflammation , Kidney/blood supply , Kidney/immunology , Mice , Microtubule-Associated Proteins/immunology , Receptors, Antigen, T-Cell , Transendothelial and Transepithelial Migration/immunology
12.
Proc Natl Acad Sci U S A ; 112(46): 14284-9, 2015 Nov 17.
Article in English | MEDLINE | ID: mdl-26540726

ABSTRACT

Three genes, CCM1, CCM2, and CCM3, interact genetically and biochemically and are mutated in cerebral cavernous malformations (CCM). A recently described member of this CCM family of proteins, CCM2-like (CCM2L), has high homology to CCM2. Here we show that its relative expression in different tissues differs from that of CCM2 and, unlike CCM2, the expression of CCM2L in endothelial cells is regulated by density, flow, and statins. In vitro, both CCM2L and CCM2 bind MEKK3 in a complex with CCM1. Both CCM2L and CCM2 interfere with MEKK3 activation and its ability to phosphorylate MEK5, a downstream target. The in vivo relevance of this regulation was investigated in zebrafish. A knockdown of ccm2l and ccm2 in zebrafish leads to a more severe "big heart" and circulation defects compared with loss of function of ccm2 alone, and also leads to substantial body axis abnormalities. Silencing of mekk3 rescues the big heart and body axis phenotype, suggesting cross-talk between the CCM proteins and MEKK3 in vivo. In endothelial cells, CCM2 deletion leads to activation of ERK5 and a transcriptional program that are downstream of MEKK3. These findings suggest that CCM2L and CCM2 cooperate to regulate the activity of MEKK3.


Subject(s)
Carrier Proteins/metabolism , MAP Kinase Kinase Kinase 3/metabolism , MAP Kinase Signaling System/physiology , Transcription, Genetic/physiology , Zebrafish Proteins/metabolism , Zebrafish/embryology , Animals , Cardiomegaly/embryology , Cardiomegaly/genetics , Carrier Proteins/genetics , Endothelial Cells/cytology , Endothelial Cells/metabolism , Enzyme Activation/physiology , Gene Knockdown Techniques , MAP Kinase Kinase Kinase 3/genetics , Mitogen-Activated Protein Kinase 7/genetics , Mitogen-Activated Protein Kinase 7/metabolism , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Zebrafish/genetics , Zebrafish Proteins/genetics
13.
Cell Rep ; 10(10): 1655-1664, 2015 Mar 17.
Article in English | MEDLINE | ID: mdl-25772353

ABSTRACT

Leukocyte CD18 integrins increase their affinity for ligand by transmitting allosteric signals to and from their ligand-binding αI domain. Mechanical forces induce allosteric changes that paradoxically slow dissociation by increasing the integrin/ligand bond lifetimes, referred to as catch bonds. Mac-1 formed catch bonds with its ligands. However, a Mac-1 gene (ITGAM) coding variant (rs1143679, R77H), which is located in the ß-propeller domain and is significantly associated with systemic lupus erythematosus risk, exhibits a marked impairment in 2D ligand affinity and affinity maturation under mechanical force. Targeted mutations and activating antibodies reveal that the failure in Mac-1 R77H allostery is rescued by induction of cytoplasmic tail separation and full integrin extension. These findings demonstrate roles for R77, and the ß-propeller in which it resides, in force-induced allostery relay and integrin bond stabilization. Defects in these processes may have pathological consequences, as the Mac-1 R77H variant is associated with increased susceptibility to lupus.

14.
Annu Rev Pathol ; 9: 181-218, 2014.
Article in English | MEDLINE | ID: mdl-24050624

ABSTRACT

Neutrophils and neutrophil-like cells are the major pathogen-fighting immune cells in organisms ranging from slime molds to mammals. Central to their function is their ability to be recruited to sites of infection, to recognize and phagocytose microbes, and then to kill pathogens through a combination of cytotoxic mechanisms. These include the production of reactive oxygen species, the release of antimicrobial peptides, and the recently discovered expulsion of their nuclear contents to form neutrophil extracellular traps. Here we discuss these primordial neutrophil functions, which also play key roles in tissue injury, by providing details of neutrophil cytotoxic functions and congenital disorders of neutrophils. In addition, we present more recent evidence that interactions between neutrophils and adaptive immune cells establish a feed-forward mechanism that amplifies pathologic inflammation. These newly appreciated contributions of neutrophils are described in the setting of several inflammatory and autoimmune diseases.


Subject(s)
Adaptive Immunity/immunology , Infections/immunology , Inflammation/immunology , Neutrophils/immunology , Animals , Extracellular Matrix/immunology , Homeostasis/immunology , Humans , Neutrophil Activation/immunology , Neutrophil Infiltration/immunology , Phagocytosis/immunology , Reactive Oxygen Species/immunology
15.
Immunity ; 38(5): 1025-37, 2013 May 23.
Article in English | MEDLINE | ID: mdl-23623383

ABSTRACT

Endothelial-dependent mechanisms of mononuclear cell influx are not well understood. We showed that acute stimulation of murine microvascular endothelial cells expressing the tumor necrosis factor receptors TNFR1 and TNFR2 with the soluble cytokine TNF led to CXCR3 chemokine generation. The TNF receptors signaled through interferon regulatory factor-1 (IRF1) to induce interferon-ß (IFN-ß) and subsequent autocrine signaling via the type I IFN receptor and the transcription factor STAT1. Both TNFR2 and TNFR1 were required for IRF1-IFNß signaling and, in human endothelial cells TNFR2 expression alone induced IFN-ß signaling and monocyte recruitment. In vivo, TNFR1 was required for acute renal neutrophil and monocyte influx after systemic TNF treatment, whereas the TNFR2-IRF1-IFN-ß autocrine loop was essential only for macrophage accumulation. In a chronic model of proliferative nephritis, IRF1 and renal-expressed TNFR2 were essential for sustained macrophage accumulation. Thus, our data identify a pathway in endothelial cells that selectively recruits monocytes during a TNF-induced inflammatory response.


Subject(s)
Interferon Regulatory Factor-1/metabolism , Interferon-beta/metabolism , Monocytes/immunology , Receptors, Tumor Necrosis Factor, Type II/metabolism , Tumor Necrosis Factor-alpha/immunology , Animals , Autocrine Communication/immunology , Cells, Cultured , Endothelial Cells/metabolism , Humans , Inflammation/immunology , Interferon Regulatory Factor-1/genetics , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocytes/metabolism , Nephritis/metabolism , Neutrophils/metabolism , Receptor, Interferon alpha-beta/metabolism , Receptors, CXCR3/biosynthesis , Receptors, Tumor Necrosis Factor, Type I/biosynthesis , Receptors, Tumor Necrosis Factor, Type II/biosynthesis , STAT1 Transcription Factor/metabolism , Tumor Necrosis Factor-alpha/metabolism
16.
J Am Soc Nephrol ; 23(3): 507-15, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22266663

ABSTRACT

Levels of proinflammatory cytokines associate with risk for developing type 2 diabetes but whether chronic inflammation contributes to the development of diabetic complications, such as ESRD, is unknown. In the 1990s, we recruited 410 patients with type 2 diabetes for studies of diabetic nephropathy and recorded their characteristics at enrollment. During 12 years of follow-up, 59 patients developed ESRD (17 per 1000 patient-years) and 84 patients died without ESRD (24 per 1000 patient-years). Plasma markers of systemic inflammation, endothelial dysfunction, and the TNF pathway were measured in the study entry samples. Of the examined markers, only TNF receptors 1 and 2 (TNFR1 and TNFR2) associated with risk for ESRD. These two markers were highly correlated, but ESRD associated more strongly with TNFR1. The cumulative incidence of ESRD for patients in the highest TNFR1 quartile was 54% after 12 years but only 3% for the other quartiles (P<0.001). In Cox proportional hazard analyses, TNFR1 predicted risk for ESRD even after adjustment for clinical covariates such as urinary albumin excretion. Plasma concentration of TNFR1 outperformed all tested clinical variables with regard to predicting ESRD. Concentrations of TNFRs moderately associated with death unrelated to ESRD. In conclusion, elevated concentrations of circulating TNFRs in patients with type 2 diabetes at baseline are very strong predictors of the subsequent progression to ESRD in subjects with and without proteinuria.


Subject(s)
Diabetes Mellitus, Type 2/complications , Diabetic Nephropathies/epidemiology , Diabetic Nephropathies/etiology , Kidney Failure, Chronic/epidemiology , Kidney Failure, Chronic/etiology , Receptors, Tumor Necrosis Factor, Type II/blood , Receptors, Tumor Necrosis Factor, Type I/blood , Adult , Aged , Biomarkers/blood , Cohort Studies , Diabetic Nephropathies/physiopathology , Disease Progression , Female , Follow-Up Studies , Humans , Incidence , Kidney Failure, Chronic/physiopathology , Male , Middle Aged , Predictive Value of Tests , Proportional Hazards Models , Retrospective Studies , Risk Factors , Signal Transduction/physiology , Survival Rate
17.
J Am Soc Nephrol ; 23(3): 516-24, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22266664

ABSTRACT

Elevated plasma concentrations of TNF receptors 1 and 2 (TNFR1 and TNFR2) predict development of ESRD in patients with type 2 diabetes without proteinuria, suggesting these markers may contribute to the pathogenesis of renal decline. We investigated whether circulating markers of the TNF pathway determine GFR loss among patients with type 1 diabetes. We followed two cohorts comprising 628 patients with type 1 diabetes, normal renal function, and no proteinuria. Over 12 years, 69 patients developed estimated GFR less than 60 mL/min per 1.73 m(2) (16 per 1000 person-years). Concentrations of TNFR1 and TNFR2 were strongly associated with risk for early renal decline. Renal decline was associated only modestly with total TNFα concentration and appeared unrelated to free TNFα. The cumulative incidence of estimated GFR less than 60 mL/min per 1.73 m(2) for patients in the highest TNFR2 quartile was 60% after 12 years compared with 5%-19% in the remaining quartiles. In Cox proportional hazards analysis, patients with TNFR2 values in the highest quartile were threefold more likely to experience renal decline than patients in the other quartiles (hazard ratio, 3.0; 95% confidence interval, 1.7-5.5). The risk associated with high TNFR1 values was slightly less than that associated with high TNFR2 values. TNFR levels were unrelated to baseline free TNFα level and remained stable over long periods within an individual. In conclusion, early GFR loss in patients with type 1 diabetes without proteinuria is strongly associated with circulating TNF receptor levels but not TNFα levels (free or total).


Subject(s)
Diabetes Mellitus, Type 1/complications , Kidney Diseases/epidemiology , Kidney Diseases/etiology , Kidney Failure, Chronic/epidemiology , Kidney Failure, Chronic/etiology , Receptors, Tumor Necrosis Factor, Type II/blood , Receptors, Tumor Necrosis Factor, Type I/blood , Adult , Biomarkers/blood , Chronic Disease , Cohort Studies , Female , Follow-Up Studies , Glomerular Filtration Rate/physiology , Humans , Kidney/physiopathology , Kidney Diseases/physiopathology , Kidney Failure, Chronic/physiopathology , Male , Middle Aged , Multivariate Analysis , Predictive Value of Tests , Proportional Hazards Models , Retrospective Studies , Risk Factors , Severity of Illness Index , Signal Transduction/physiology
18.
Cell Host Microbe ; 10(6): 603-15, 2011 Dec 15.
Article in English | MEDLINE | ID: mdl-22177564

ABSTRACT

Resistance to fungal infections is attributed to engagement of host pattern-recognition receptors, notably the ß-glucan receptor Dectin-1 and the integrin Mac-1, which induce phagocytosis and antifungal immunity. However, the mechanisms by which these receptors coordinate fungal clearance are unknown. We show that upon ligand binding, Dectin-1 activates Mac-1 to also recognize fungal components, and this stepwise process is critical for neutrophil cytotoxic responses. Both Mac-1 activation and Dectin-1- and Mac-1-induced neutrophil effector functions require Vav1 and Vav3, exchange factors for RhoGTPases. Mac-1- or Vav1,3-deficient mice have increased susceptibility to systemic candidiasis that is not due to impaired neutrophil recruitment but defective intracellular killing of C. albicans yeast forms, and Mac-1 or Vav1,3 reconstitution in hematopoietic cells restores resistance. Our results demonstrate that antifungal immunity depends on Dectin-1-induced activation of Mac-1 functions that is coordinated by Vav proteins, a pathway that may localize cytotoxic responses of circulating neutrophils to infected tissues.


Subject(s)
Candida albicans/immunology , Candidiasis/immunology , Lectins, C-Type/immunology , Macrophage-1 Antigen/immunology , Neutrophils/immunology , Proto-Oncogene Proteins c-vav/immunology , Receptors, Immunologic/immunology , Animals , Candida albicans/physiology , Candidiasis/genetics , Candidiasis/microbiology , Female , Humans , Lectins, C-Type/genetics , Macrophage-1 Antigen/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophils/microbiology , Phagocytosis , Proto-Oncogene Proteins c-vav/genetics , Receptors, Immunologic/genetics , Signal Transduction
19.
Arthritis Rheum ; 63(2): 467-78, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21280001

ABSTRACT

OBJECTIVE: Rheumatoid arthritis culminates in joint destruction that, in mouse models of disease, is supported by innate immune molecules, including Fcγ receptors (FcγR) and complement. However, these findings may not be predictive of the outcome in humans, given the structural differences between murine and human activating FcγR on neutrophils, a prominent component of joint exudates. The aim of this study was to examine the role of human neutrophil FcγRIIa in the development of arthritis and probe the underlying mechanism by which FcγRIIa initiates disease. METHODS: K/BxN mouse serum transfer-induced arthritis was examined in mice expressing human FcγRIIa on neutrophils but lacking their own activating FcγR (γ-chain-deficient mice). The role of mast cells, complement (C3 and C5a), and CD18 integrins in FcγRIIa-initiated disease was examined using cell reconstitution approaches, inhibitors, and functional blocking antibodies, respectively. Crosstalk between the complement receptor C5aR and FcγRIIa on neutrophils was evaluated in vitro. RESULTS: The expression of human FcγRIIa on neutrophils was sufficient to restore susceptibility to K/BxN serum-induced neutrophil recruitment, synovitis, and bone destruction in γ-chain-deficient mice. Joint inflammation was robust and proceeded even in the absence of mast cells and vascular permeability, features shown to contribute to disease in wild-type mice. Neutrophil recruitment was dependent on the presence of a CD18 integrin, lymphocyte function-associated antigen 1, and C5aR. In addition, C5aR significantly enhanced FcγRIIa-mediated phagocytosis and oxidative burst in vitro. CONCLUSION: Human and murine activating FcγR on neutrophils are not functionally equivalent, and in humans, they may play a primary role in arthritis. Crosstalk between neutrophil C5aR and FcγRIIa is essential for disease progression, thus highlighting a new aspect of complement during the effector phase of inflammatory arthritis.


Subject(s)
Arthritis, Experimental/immunology , Neutrophils/immunology , Receptor, Anaphylatoxin C5a/immunology , Receptors, IgG/immunology , Adoptive Transfer , Animals , Arthritis, Experimental/metabolism , Bone Marrow Transplantation , Disease Models, Animal , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophil Infiltration , Neutrophils/metabolism , Phagocytosis/immunology , Receptor Cross-Talk/immunology , Receptor, Anaphylatoxin C5a/metabolism , Receptors, IgG/metabolism , Species Specificity , Synovitis/immunology , Synovitis/metabolism
20.
Blood ; 117(2): 708-18, 2011 Jan 13.
Article in English | MEDLINE | ID: mdl-20952690

ABSTRACT

Adhesive forces at endothelial cell-cell borders maintain vascular integrity. cAMP enhances barrier properties and controls cellular processes through protein kinase A bound to A-kinase anchoring proteins (AKAPs). It also activates exchange protein directly activated by cAMP (Epac1), an exchange factor for Ras-related protein 1 (Rap1) GTPases that promotes cadherin- and integrin-mediated adhesion through effects on the actin cytoskeleton. We demonstrate that AKAP9 facilitates the microtubule polymerization rate in endothelial cells, interacts with Epac1, and is required for Epac1-stimulated microtubule growth. AKAP9 is not required for maintaining barrier properties under steady-state conditions. Rather, it is essential when the cell is challenged to make new adhesive contacts, as is the case when Epac activation enhances barrier function through a mechanism that, surprisingly, requires integrin adhesion at cell-cell contacts. In the present study, defects in Epac-induced responses in AKAP9-silenced cells were evident despite an intact Epac-induced increase in Rap activation, cortical actin, and vascular endothelial-cadherin adhesion. We describe a pathway that integrates Epac-mediated signals with AKAP9-dependent microtubule dynamics to coordinate integrins at lateral borders.


Subject(s)
A Kinase Anchor Proteins/metabolism , Cell Adhesion/physiology , Cytoskeletal Proteins/metabolism , Endothelial Cells/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Microtubules/metabolism , Blotting, Western , Cell Separation , Flow Cytometry , Humans , Immunohistochemistry , Immunoprecipitation , Microscopy, Confocal , Microscopy, Fluorescence , Microtubules/ultrastructure , Protein Isoforms , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...