Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Cell Rep ; 42(11): 113394, 2023 11 28.
Article in English | MEDLINE | ID: mdl-37950870

ABSTRACT

The pore-forming S. aureus α-toxin (Hla) contributes to virulence and disease pathogenesis. While high concentrations of toxin induce cell death, neutrophils exhibit relative resistance to lysis, suggesting that the action of Hla may not be solely conferred by lytic susceptibility. Using intravital microscopy, we observed that Hla disrupts neutrophil localization and clustering early in infection. Hla forms a narrow, ion-selective pore, suggesting that Hla may dysregulate calcium or other ions to impair neutrophil function. We found that sub-lytic Hla did not permit calcium influx but caused rapid membrane depolarization. Depolarization decreases the electrogenic driving force for calcium, and concordantly, Hla suppressed calcium signaling in vitro and in vivo and calcium-dependent leukotriene B4 (LTB4) production, a key mediator of neutrophil clustering. Thus, Hla disrupts the early patterning of the neutrophil response to infection, in part through direct impairment of neutrophil calcium signaling. This early mis-localization of neutrophils may contribute to establishment of infection.


Subject(s)
Neutrophils , Staphylococcus aureus , Neutrophils/metabolism , Staphylococcus aureus/metabolism , Calcium/metabolism , Calcium Signaling
2.
iScience ; 26(10): 108034, 2023 Oct 20.
Article in English | MEDLINE | ID: mdl-37854699

ABSTRACT

Neutrophil swarming is a complex coordinated process in which neutrophils sensing pathogen or damage signals are rapidly recruited to sites of infections or injuries. This process involves cooperation between neutrophils where autocrine and paracrine positive-feedback loops, mediated by receptor/ligand pairs including lipid chemoattractants and chemokines, amplify localized recruitment of neutrophils. This review will provide an overview of key pathways involved in neutrophil swarming and then discuss the cell intrinsic and systemic mechanisms by which NADPH oxidase 2 (NOX2) regulates swarming, including modulation of calcium signaling, inflammatory mediators, and the mobilization and production of neutrophils. We will also discuss mechanisms by which altered neutrophil swarming in disease may contribute to deficient control of infections and/or exuberant inflammation. Deeper understanding of underlying mechanisms controlling neutrophil swarming and how neutrophil cooperative behavior can be perturbed in the setting of disease may help to guide development of tools for diagnosis and precision medicine.

3.
J Exp Med ; 219(1)2022 01 03.
Article in English | MEDLINE | ID: mdl-34751735

ABSTRACT

The interaction between neutrophils and endothelial cells is critical for the pathogenesis of vascular inflammation. However, the regulation of neutrophil adhesive function remains not fully understood. Intravital microscopy demonstrates that neutrophil DREAM promotes neutrophil recruitment to sites of inflammation induced by TNF-α but not MIP-2 or fMLP. We observe that neutrophil DREAM represses expression of A20, a negative regulator of NF-κB activity, and enhances expression of pro-inflammatory molecules and phosphorylation of IκB kinase (IKK) after TNF-α stimulation. Studies using genetic and pharmacologic approaches reveal that DREAM deficiency and IKKß inhibition significantly diminish the ligand-binding activity of ß2 integrins in TNF-α-stimulated neutrophils or neutrophil-like HL-60 cells. Neutrophil DREAM promotes degranulation through IKKß-mediated SNAP-23 phosphorylation. Using sickle cell disease mice lacking DREAM, we show that hematopoietic DREAM promotes vaso-occlusive events in microvessels following TNF-α challenge. Our study provides evidence that targeting DREAM might be a novel therapeutic strategy to reduce excessive neutrophil recruitment in inflammatory diseases.


Subject(s)
Inflammation/genetics , Kv Channel-Interacting Proteins/genetics , Microvessels/metabolism , Neutrophil Infiltration/genetics , Neutrophils/metabolism , Repressor Proteins/genetics , Animals , Cell Adhesion/drug effects , Gene Expression Regulation , HL-60 Cells , Humans , I-kappa B Kinase/metabolism , Inflammation/metabolism , Kv Channel-Interacting Proteins/metabolism , Mice, Inbred C57BL , Mice, Knockout , Microvessels/pathology , NF-kappa B/metabolism , Neutrophil Infiltration/drug effects , Neutrophils/drug effects , Phosphorylation/drug effects , Repressor Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tumor Necrosis Factor alpha-Induced Protein 3/genetics , Tumor Necrosis Factor alpha-Induced Protein 3/metabolism , Tumor Necrosis Factor-alpha/pharmacology
4.
J Immunol ; 206(12): 3000-3009, 2021 06 15.
Article in English | MEDLINE | ID: mdl-34078711

ABSTRACT

SARS-CoV-2, the virus that has caused the COVID-19 pandemic, robustly activates the host immune system in critically ill patients. Understanding how the virus engages the immune system will facilitate the development of needed therapeutic strategies. In this study, we demonstrate both in vitro and in vivo that the SARS-CoV-2 surface proteins spike (S) and envelope (E) activate the key immune signaling IFN pathway in both human and mouse immune and epithelial cells independent of viral infection and replication. These proteins induce reactive oxidative species generation and increases in human- and murine-specific, IFN-responsive cytokines and chemokines, similar to their upregulation in critically ill COVID-19 patients. Induction of IFN signaling is dependent on canonical but discrepant inflammatory signaling mediators, as the activation induced by S is dependent on IRF3, TBK1, and MyD88, whereas that of E is largely MyD88 independent. Furthermore, these viral surface proteins, specifically E, induced peribronchial inflammation and pulmonary vasculitis in a mouse model. Finally, we show that the organized inflammatory infiltrates are dependent on type I IFN signaling, specifically in lung epithelial cells. These findings underscore the role of SARS-CoV-2 surface proteins, particularly the understudied E protein, in driving cell specific inflammation and their potential for therapeutic intervention.


Subject(s)
Coronavirus Envelope Proteins/immunology , Epithelial Cells/immunology , Inflammation/immunology , Interferon Type I/immunology , Spike Glycoprotein, Coronavirus/immunology , Animals , Cell Line, Tumor , Epithelial Cells/virology , Female , Humans , Male , Mice
5.
Elife ; 102021 05 28.
Article in English | MEDLINE | ID: mdl-34047696

ABSTRACT

Neutrophil responses against pathogens must be balanced between protection and immunopathology. Factors that determine these outcomes are not well-understood. In a mouse model of genital herpes simplex virus-2 (HSV-2) infection, which results in severe genital inflammation, antibody-mediated neutrophil depletion reduced disease. Comparative single-cell RNA-sequencing analysis of vaginal cells against a model of genital HSV-1 infection, which results in mild inflammation, demonstrated sustained expression of interferon-stimulated genes (ISGs) only after HSV-2 infection primarily within the neutrophil population. Both therapeutic blockade of IFNα/ß receptor 1 (IFNAR1) and genetic deletion of IFNAR1 in neutrophils concomitantly decreased HSV-2 genital disease severity and vaginal IL-18 levels. Therapeutic neutralization of IL-18 also diminished genital inflammation, indicating an important role for this cytokine in promoting neutrophil-dependent immunopathology. Our study reveals that sustained type I interferon (IFN) signaling is a driver of pathogenic neutrophil responses and identifies IL-18 as a novel component of disease during genital HSV-2 infection.


Herpes simplex virus (HSV) is a human pathogen that causes genital herpes, an incurable disease that results in recurrent sores and inflammation. Infection with HSV induces a strong antiviral immune response, which results in large numbers of immune cells arriving at these lesions. But while some of these cells help to control viral replication, others might contribute to the inflammation that drives the disease. One of the first immune cells to respond to infection are neutrophils. Although neutrophils are generally protective, especially against bacteria and fungi, they have also been implicated in tissue damage and severe inflammation during viral infections. But what determines whether a neutrophil will help to fight off an infection or increase disease severity is still an open question. To investigate this, Lebratti, Lim et al. studied mice that had been infected with the genital herpes virus HSV-2, which is known to cause significant amounts of inflammation in mice. The experiments revealed that a signaling molecule called type I interferon, which is thought to be antiviral, causes neutrophils at the site of the infection to produce proteins, such as IL-18, which trigger an inflammatory reaction. Lebratti, Lim et al. found that type I interferon and IL-18 had shifting roles during the course of infection. In the early stages, both molecules had a protective effect, confirming results from previous studies. However, as the infection progressed, sustained levels of type I interferon signaling in neutrophils led to excess amounts of IL-18. Lebratti, Lim et al. discovered that blocking interferon signaling or decreasing the levels of IL-18 later during infection unexpectedly reduced the severity of the disease and resulted in less genital tissue damage. Further experiments also showed that mice infected with another genital herpes virus called HSV-1 did not experience sustained levels of type I interferon. This may explain why this virus causes less severe disease in mice. Understanding how the immune system reacts to viruses could reveal new targets for treatments of genital herpes. At the moment, there is little information about IL-18 production during genital herpes in humans. So, the next step is to see whether neutrophils behave in the same way and whether IL-18 can be detected during human disease. It is possible that the same immune components could promote disease in other infections too. If so, this work may help uncover new drug targets for other viral diseases.


Subject(s)
Herpes Genitalis/virology , Herpesvirus 2, Human/pathogenicity , Immunity, Mucosal , Interferon Type I/metabolism , Interleukin-18/metabolism , Mucous Membrane/virology , Neutrophil Activation , Neutrophils/virology , Vagina/virology , Animals , Antibodies/pharmacology , Chlorocebus aethiops , Disease Models, Animal , Female , Herpes Genitalis/immunology , Herpes Genitalis/metabolism , Herpes Genitalis/prevention & control , Herpesvirus 1, Human/immunology , Herpesvirus 1, Human/pathogenicity , Herpesvirus 2, Human/immunology , Host-Pathogen Interactions , Immunity, Mucosal/drug effects , Mice, Inbred C57BL , Mice, Transgenic , Mucous Membrane/drug effects , Mucous Membrane/innervation , Mucous Membrane/metabolism , Neutrophil Activation/drug effects , Neutrophils/drug effects , Neutrophils/immunology , Neutrophils/metabolism , Receptor, Interferon alpha-beta/antagonists & inhibitors , Receptor, Interferon alpha-beta/metabolism , Signal Transduction , Vagina/drug effects , Vagina/immunology , Vagina/metabolism , Vero Cells
6.
J Trauma Acute Care Surg ; 90(6): 924-934, 2021 06 01.
Article in English | MEDLINE | ID: mdl-34016916

ABSTRACT

BACKGROUND: Forty percent of critically ill trauma patients will develop an infectious complication. Pneumonia is the most common cause of death of trauma patients surviving their initial insult. We previously demonstrated that polytrauma (PT), defined as two or more severe injuries in at least two areas of the body, induces emergency hematopoiesis characterized by accelerated myelopoiesis in the bone marrow and increased myeloid cell frequency in the peripheral tissues. We hypothesized that PT alone induces priming of neutrophils, resulting in hyperactivation upon secondary exposure to bacteria and causing acute lung injury and increased susceptibility to secondary exposure to Pseudomonas aeruginosa pneumonia. METHODS: C57BL/6 mice were subjected to PT consisting of a lower extremity pseudofracture, liver crush injury, and 15% blood-volume hemorrhage. Pneumonia was induced by intratracheal injection of 5 × 106 CFU live P. aeruginosa or 1 × 107 of heat-killed P. aeruginosa (HKPA). For reactive oxygen species (ROS), studies polymorphonuclear neutrophils (PMNs) were isolated by immunomagnetic bead negative selection and stimulated ex-vivo with HKPA. Reactive oxygen species production was measured by immunofluorescence. For histology, lung sections were stained by hematoxylin-eosin and analyzed by a blinded grader. RESULTS: Polytrauma induced persistent changes in immune function at baseline and to secondary infection. Pneumonia after injury resulted in increased mortality (60% vs. 5% p < 0.01). Blood neutrophils from PT mice had higher resting (unstimulated) ROS production than in naive animals (p < 0.02) demonstrating priming of the neutrophils following PT. After intratracheal HKPA injection, bronchoalveolar lavage PMNs from injured mice had higher ROS production compared with naive mice (p < 0.01), demonstrating an overexuberant immunopathologic response of neutrophils following PT. CONCLUSION: Polytrauma primes neutrophils and causes immunopathologic PMN ROS production, increased lung injury and susceptibility to secondary bacterial pneumonia. These results suggest that trauma-induced immune dysfunction can cause immunopathologic response to secondary infection and suggests neutrophil-mediated pulmonary damage as a therapeutic target for posttrauma pneumonia.


Subject(s)
Acute Lung Injury/immunology , Multiple Trauma/complications , Neutrophils/immunology , Pneumonia, Bacterial/immunology , Pseudomonas Infections/immunology , Acute Lung Injury/blood , Acute Lung Injury/microbiology , Acute Lung Injury/pathology , Animals , Disease Models, Animal , Humans , Lung/immunology , Lung/microbiology , Lung/pathology , Male , Mice , Multiple Trauma/blood , Multiple Trauma/diagnosis , Multiple Trauma/immunology , Neutrophils/metabolism , Pneumonia, Bacterial/blood , Pneumonia, Bacterial/microbiology , Pneumonia, Bacterial/pathology , Pseudomonas Infections/blood , Pseudomonas Infections/microbiology , Pseudomonas Infections/pathology , Pseudomonas aeruginosa/immunology , Reactive Oxygen Species/metabolism , Trauma Severity Indices
7.
Am J Physiol Lung Cell Mol Physiol ; 320(5): L892-L902, 2021 05 01.
Article in English | MEDLINE | ID: mdl-33355521

ABSTRACT

Sepsis is a heterogeneous syndrome clinically and biologically, but biomarkers of distinct host response pathways for early prognostic information and testing targeted treatments are lacking. Olfactomedin 4 (OLFM4), a matrix glycoprotein of neutrophil-specific granules, defines a distinct neutrophil subset that may be an independent risk factor for poor outcomes in sepsis. We hypothesized that increased percentage of OLFM4+ neutrophils on sepsis presentation would be associated with mortality. In a single-center, prospective cohort study, we enrolled adults admitted to an academic medical center from the emergency department (ED) with suspected sepsis [identified by 2 or greater systemic inflammatory response syndrome (SIRS) criteria and antibiotic receipt] from March 2016 through December 2017, followed by sepsis adjudication according to Sepsis-3. We collected 200 µL of whole blood within 24 h of admission and stained for the neutrophil surface marker CD66b followed by intracellular staining for OLFM4 quantitated by flow cytometry. The predictors for 60-day mortality were 1) percentage of OLFM4+ neutrophils and 2) OLFM4+ neutrophils at a cut point of ≥37.6% determined by the Youden Index. Of 120 enrolled patients with suspected sepsis, 97 had sepsis and 23 had nonsepsis SIRS. The mean percentage of OLFM4+ neutrophils was significantly increased in both sepsis and nonsepsis SIRS patients who died (P ≤ 0.01). Among sepsis patients with elevated OLFM4+ (≥37.6%), 56% died, compared with 18% with OLFM4+ <37.6% (P = 0.001). The association between OLFM4+ and mortality withstood adjustment for age, sex, absolute neutrophil count, comorbidities, and standard measures of severity of illness (SOFA score, APACHE III) (P < 0.03). In summary, OLFM4+ neutrophil percentage is independently associated with 60-day mortality in sepsis and may represent a novel measure of the heterogeneity of host response to sepsis.


Subject(s)
Granulocyte Colony-Stimulating Factor/blood , Neutrophils/metabolism , Sepsis/blood , Sepsis/mortality , Adult , Aged , Aged, 80 and over , Disease-Free Survival , Female , Humans , Male , Middle Aged , Risk Factors , Sepsis/drug therapy , Survival Rate
8.
Proc Natl Acad Sci U S A ; 117(39): 24403-24414, 2020 09 29.
Article in English | MEDLINE | ID: mdl-32929002

ABSTRACT

Calcium signals are initiated in immune cells by the process of store-operated calcium entry (SOCE), where receptor activation triggers transient calcium release from the endoplasmic reticulum, followed by opening of plasma-membrane calcium-release activated calcium (CRAC) channels. ORAI1, ORAI2, and ORAI3 are known to comprise the CRAC channel; however, the contributions of individual isoforms to neutrophil function are not well understood. Here, we show that loss of ORAI1 partially decreases calcium influx, while loss of both ORAI1 and ORAI2 completely abolishes SOCE. In other immune-cell types, loss of ORAI2 enhances SOCE. In contrast, we find that ORAI2-deficient neutrophils display decreased calcium influx, which is correlated with measurable differences in the regulation of neutrophil membrane potential via KCa3.1. Decreased SOCE in ORAI1-, ORAI2-, and ORAI1/2-deficient neutrophils impairs multiple neutrophil functions, including phagocytosis, degranulation, leukotriene, and reactive oxygen species (ROS) production, rendering ORAI1/2-deficient mice highly susceptible to staphylococcal infection. This study demonstrates that ORAI1 and ORAI2 are the primary components of the neutrophil CRAC channel and identifies subpopulations of neutrophils where cell-membrane potential functions as a rheostat to modulate the SOCE response. These findings have implications for mechanisms that modulate neutrophil function during infection, acute and chronic inflammatory conditions, and cancer.


Subject(s)
Calcium Signaling , Calcium/immunology , Neutrophils/immunology , ORAI1 Protein/immunology , ORAI2 Protein/immunology , Animals , Female , Intermediate-Conductance Calcium-Activated Potassium Channels/genetics , Intermediate-Conductance Calcium-Activated Potassium Channels/immunology , Male , Mice , Mice, Inbred C57BL , ORAI1 Protein/genetics , ORAI2 Protein/genetics
9.
Blood ; 135(12): 891-903, 2020 03 19.
Article in English | MEDLINE | ID: mdl-31951647

ABSTRACT

Leukocyte reduced NADP (NADPH) oxidase plays a key role in host defense and immune regulation. Genetic defects in NADPH oxidase result in chronic granulomatous disease (CGD), characterized by recurrent bacterial and fungal infections and aberrant inflammation. Key drivers of hyperinflammation induced by fungal cell walls in CGD are still incompletely defined. In this study, we found that CGD (CYBB-) neutrophils produced higher amounts of leukotriene B4 (LTB4) in vitro after activation with zymosan or immune complexes, compared with wild-type (WT) neutrophils. This finding correlated with increased calcium influx in CGD neutrophils, which was restrained in WT neutrophils by the electrogenic activity of NADPH oxidase. Increased LTB4 generation by CGD neutrophils was also augmented by paracrine cross talk with the LTB4 receptor BLT1. CGD neutrophils formed more numerous and larger clusters in the presence of zymosan in vitro compared with WT cells, and the effect was also LTB4- and BLT1-dependent. In zymosan-induced lung inflammation, focal neutrophil infiltrates were increased in CGD compared with WT mice and associated with higher LTB4 levels. Inhibiting LTB4 synthesis or antagonizing the BLT1 receptor after zymosan challenge reduced lung neutrophil recruitment in CGD to WT levels. Thus, LTB4 was the major driver of excessive neutrophilic lung inflammation in CGD mice in the early response to fungal cell walls, likely by a dysregulated feed-forward loop involving amplified neutrophil production of LTB4. This study identifies neutrophil LTB4 generation as a target of NADPH oxidase regulation, which could potentially be exploited therapeutically to reduce excessive inflammation in CGD.


Subject(s)
Cell Wall/immunology , Fungi/immunology , NADPH Oxidases/genetics , NADPH Oxidases/metabolism , Neutrophil Infiltration/genetics , Neutrophils/metabolism , Receptors, Leukotriene B4/metabolism , Animals , Calcium , Disease Models, Animal , Disease Susceptibility , Mice , Mycoses/genetics , Mycoses/immunology , Mycoses/metabolism , Mycoses/microbiology , Neutrophil Infiltration/immunology , Neutrophils/immunology , Neutrophils/pathology , Oxidation-Reduction , Oxidative Stress , Protein Binding , Signal Transduction
10.
Cell Calcium ; 78: 56-65, 2019 03.
Article in English | MEDLINE | ID: mdl-30641250

ABSTRACT

Calcium is a major intracellular signaling messenger in innate immune cells. Similar to other immune cell subsets, the majority of calcium entry into innate immune cells is induced by cell surface receptors that stimulate store-operated calcium entry through calcium-release activated calcium (CRAC) channels. Since the molecular description of the STIM family of calcium sensors and the ORAI family of CRAC channel proteins, the majority of studies support a dominant role for these proteins in calcium signaling in innate cells. In reviewing the literature on CRAC channel function in innate cells, several general themes emerge. All innate cells express multiple members of the STIM and ORAI family members, however the ratio and relative contribution of individual isoforms changes depending on the cell type and activation state of the cell. It is evident that study of functional roles for STIM molecules is clearly ahead of studies of specific ORAI family members in all innate cell types, and that studies of CRAC channels in innate cells are not nearly as advanced as studies in lymphocytes. However, taken together, evidence from both STIM calcium sensors and ORAI channels in innate cells indicates that deficiency of STIM and ORAI proteins tends not to affect the development of any innate cell lineage, but certainly affects their function, in particular activation of the neutrophil oxidase and mast cell activation via IgE receptors. Furthermore, there are clearly hints that therapeutic targeting of CRAC channels in innate cells offers a new approach to various inflammatory and allergic diseases.


Subject(s)
Calcium Release Activated Calcium Channels/metabolism , Hypersensitivity/immunology , Hypersensitivity/metabolism , Immunity, Innate/immunology , Inflammation/immunology , Inflammation/metabolism , Animals , Calcium/metabolism , Humans
11.
Blood ; 130(13): 1565-1577, 2017 09 28.
Article in English | MEDLINE | ID: mdl-28724541

ABSTRACT

Neutrophils are key effector cells of the innate immune system. Calcium-dependent signaling pathways initiated by store-operated calcium entry (SOCE) are known to regulate neutrophil activation; however, the precise mechanism of this process remains unclear. STIM1 and STIM2 are calcium-sensing molecules that link calcium depletion of the endoplasmic reticulum with opening of plasma membrane calcium channels. Although a role for STIM1 in neutrophil SOCE and activation has been established, the function of STIM2 is unknown. Here we use mice with conditional ablation of Stim1 and/or Stim2 to investigate the role of STIM2 in neutrophil activation. We demonstrate that loss of STIM2 results in decreased SOCE, particularly at lower doses of agonists. Reactive oxygen species (ROS) production, degranulation, and phagocytosis are normal in the absence of STIM2, suggesting STIM1 is the dominant calcium sensor required for classical short-term neutrophil responses. However, neutrophil cytokine production required STIM2, but not STIM1, at least in part as a result of redox regulation of cytokine gene expression. In vivo loss of STIM2 results in lower cytokine levels and protection from mortality in a mouse model of systemic inflammatory response syndrome. These data, combined with previous studies focusing on STIM1, define distinct but cooperative functions for STIM1 and STIM2 in modulating neutrophil bactericidal and cytokine responses.


Subject(s)
Calcium/metabolism , Cytokines/biosynthesis , Neutrophil Activation , Stromal Interaction Molecule 1/physiology , Stromal Interaction Molecule 2/physiology , Animals , Calcium Channels/metabolism , Mice , Oxidation-Reduction , Stromal Interaction Molecule 1/immunology , Stromal Interaction Molecule 2/immunology
12.
J Leukoc Biol ; 98(4): 497-502, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25714804

ABSTRACT

Calcium signals in neutrophils are initiated by a variety of cell-surface receptors, including formyl peptide and other GPCRs, FcRs, and integrins. The predominant pathway by which calcium enters immune cells is termed SOCE, whereby plasma membrane CRAC channels allow influx of extracellular calcium into the cytoplasm when intracellular ER stores are depleted. The identification of 2 key families of SOCE regulators, STIM calcium "sensors" and ORAI calcium channels, has allowed for genetic manipulation of SOCE pathways and provided valuable insight into the molecular mechanism of calcium signaling in immune cells, including neutrophils. This review focuses on our current knowledge of the molecules involved in neutrophil SOCE and how study of these molecules has further informed our understanding of the role of calcium signaling in neutrophil activation.


Subject(s)
Calcium Signaling/immunology , Neutrophil Activation/immunology , Neutrophils/immunology , Animals , Humans
13.
Blood ; 123(14): 2238-49, 2014 Apr 03.
Article in English | MEDLINE | ID: mdl-24493668

ABSTRACT

The stromal-interacting molecule 1 (STIM1) is a potent sensor of intracellular calcium, which in turn regulates entry of external calcium through plasma membrane channels to affect immune cell activation. Although the contribution of STIM1 to calcium signaling in lymphocytes has been well studied, the role of this protein in neutrophil-mediated inflammation and host defense is unknown. We report that STIM1-deficient murine neutrophils show loss of store-operated calcium entry (SOCE) in response to both soluble ligands that activate G-proteins as well as Fcγ-receptor or integrin ligation that activates tyrosine kinase signaling. This results in modest defects in phagocytosis and degranulation responses but a profound block in superoxide production by the phagocyte oxidase. We trace the primary intracellular target of calcium to be protein kinase C isoforms α and ß (PKCα and PKCß), which in turn phosphorylate subunits of the oxidase leading to superoxide production. In vivo the loss of SOCE in stim1(-/-) chimeric mice results in marked susceptibility to bacterial infections but also protection from tissue injury in hepatic ischemia/reperfusion injury. These results demonstrate the critical role of STIM1-mediated SOCE and define major protein targets of calcium signaling in neutrophil activation during inflammatory disease.


Subject(s)
Immunity/genetics , Inflammation/genetics , Membrane Glycoproteins/physiology , Oxidoreductases/metabolism , Phagocytes/enzymology , Animals , Calcium/metabolism , Calcium Channels , Calcium Signaling/genetics , Cells, Cultured , Enzyme Activation , Inflammation/metabolism , Liver/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Reperfusion Injury/genetics , Reperfusion Injury/metabolism , Stromal Interaction Molecule 1
14.
J Immunol ; 178(7): 4606-14, 2007 Apr 01.
Article in English | MEDLINE | ID: mdl-17372019

ABSTRACT

The Src homology 2 domain-containing leukocyte phosphoprotein of 76 kDa (SLP-76) is an adaptor molecule critical for immunoreceptor and integrin signaling in multiple hemopoietic lineages. We showed previously that SLP-76 is required for neutrophil function in vitro, including integrin-induced adhesion and production of reactive oxygen intermediates, and to a lesser extent, FcgammaR-induced calcium flux and reactive oxygen intermediate production. It has been difficult to determine whether SLP-76 regulates neutrophil responses in vivo, because Slp-76(-/-) mice exhibit marked defects in thymocyte and vascular development, as well as platelet and mast cell function. To circumvent these issues, we generated mice with targeted loss of SLP-76 expression within myeloid cells. Neutrophils obtained from these animals failed to respond to integrin activation in vitro, similar to Slp-76(-/-) cells. Despite these abnormalities, SLP-76-deficient neutrophils migrated normally in vivo in response to Staphylococcus aureus infection and efficiently cleared micro-organisms. Interestingly, SLP-76-deficient neutrophils did not induce a robust inflammatory response in the localized Shwartzman reaction. Collectively, these data suggest that disruption of integrin signaling via loss of SLP-76 expression differentially impairs neutrophil functions in vivo, with preservation of migration and killing of S. aureus but reduction in LPS-induced tissue damage and vascular injury.


Subject(s)
Abscess/immunology , Adaptor Proteins, Signal Transducing/physiology , Neutrophils/immunology , Phosphoproteins/physiology , Shwartzman Phenomenon/immunology , Staphylococcal Infections/immunology , Staphylococcus aureus , Abscess/microbiology , Abscess/pathology , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Movement/genetics , Integrases/genetics , Integrins/genetics , Integrins/metabolism , Mice , Mice, Mutant Strains , Myeloid Cells/immunology , Phosphoproteins/genetics , Phosphoproteins/metabolism , Shwartzman Phenomenon/microbiology , Shwartzman Phenomenon/pathology , Signal Transduction , Staphylococcal Infections/pathology
15.
Mol Cell Biol ; 26(18): 6936-49, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16943434

ABSTRACT

Syk tyrosine kinase and Src homology 2 (SH2) domain-containing leukocyte-specific phosphoprotein of 76 kDa (SLP-76) are signaling mediators activated downstream of immunoreceptor tyrosine-based activation motif (ITAM)-containing immunoreceptors and integrins. While the signaling cascades descending from integrins are similar to immunoreceptors, the mechanism of Syk activation and SLP-76 recruitment remains unclear. We used an in vivo structure-function approach to study the requirements for the domains of Syk and SLP-76 in immunoreceptor and integrin signaling. We found that both SH2 domains and the kinase domain of Syk are required for immunoreceptor-dependent signaling and cellular response via integrins. While the Gads-binding domain of SLP-76 is needed for immunoreceptor signaling, it appears dispensable for integrin signaling. Syk and SLP-76 also are required for initiating and/or maintaining separation between the blood and lymphatic vasculature. Therefore, we correlated the signaling requirement of the various domains of Syk and SLP-76 to their requirement in regulating vascular separation. Our data suggest ITAMs are required in Syk-dependent integrin signaling, demonstrate the separation of the structural features of SLP-76 to selectively support immunoreceptor versus integrin signaling, and provide evidence that the essential domains of SLP-76 for ITAM signals are those which most efficiently support separation between lymphatic and blood vessels.


Subject(s)
Hematopoietic System/cytology , Hematopoietic System/metabolism , Integrins/metabolism , Phosphoproteins/metabolism , Signal Transduction , Adaptor Proteins, Signal Transducing/metabolism , Animals , Blood Vessels/cytology , Blood Vessels/pathology , Blood Vessels/physiology , CHO Cells , Cell Membrane/metabolism , Cells, Cultured , Cricetinae , Humans , Intracellular Signaling Peptides and Proteins/deficiency , Mice , Mutation/genetics , Phosphoproteins/deficiency , Phosphotyrosine/metabolism , Protein Binding , Protein Structure, Tertiary , Protein-Tyrosine Kinases/deficiency , Receptors, Immunologic/metabolism , Syk Kinase , src Homology Domains
16.
Mol Cell Biol ; 24(24): 10923-32, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15572693

ABSTRACT

PML-retinoic acid receptor alpha (RARalpha) regulated adaptor molecule 1 (PRAM-1) is an intracellular adaptor molecule that is upregulated during the induced granulocytic differentiation of promyelocytic leukemic cells and during normal human myelopoiesis. This report describes the generation of PRAM-1-deficient mice and an analysis of the function of this adaptor in neutrophil differentiation and mature neutrophil function. We demonstrate here that neutrophil differentiation is not impaired in PRAM-1-deficient mice and that PRAM-1-deficient neutrophils function normally following engagement of Fcgamma receptors. In contrast, mature PRAM-1-null neutrophils exhibit significant defects in adhesion-dependent reactive oxygen intermediate production and degranulation. Surprisingly, other integrin-dependent responses, such as cell spreading and activation of several signaling pathways, are normal. Together, these findings demonstrate the uncoupling of key integrin-dependent responses in the absence of PRAM-1 and show this adaptor to be critical for select integrin functions in neutrophils.


Subject(s)
Integrins/metabolism , Neutrophils/metabolism , Proteins/metabolism , Adaptor Proteins, Signal Transducing , Amino Acid Sequence , Animals , Calcium/metabolism , Cell Degranulation , Gene Expression , Gene Targeting , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Sequence Data , Neutrophils/cytology , Neutrophils/immunology , Opsonin Proteins , Phagocytosis , Precipitin Tests , Protein Structure, Tertiary , Proteins/chemistry , Proteins/genetics , Respiratory Burst/immunology , Sequence Homology, Amino Acid , Staphylococcus aureus
17.
Immunity ; 19(5): 761-9, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14614862

ABSTRACT

While the contribution of intracellular adaptor proteins to lymphocyte activation has been well studied, the function of these molecules in innate immune effector cells such as neutrophils has not been extensively addressed. Here we demonstrate a critical role for the adaptor molecule SH2 domain-containing leukocyte-specific phosphoprotein of 76 kDa (SLP-76) in FcgammaR and integrin signaling. Stimulation of these receptors induces tyrosine phosphorylation and cytoplasmic relocalization of SLP-76 in freshly isolated murine neutrophils. Neutrophils lacking SLP-76 demonstrate decreased FcgammaR-induced calcium flux and reactive oxygen intermediate (ROI) production in response to immune complex stimulation. More dramatically, SLP-76-/- neutrophils fail to produce ROI, spread, or activate critical downstream regulators in response to integrin ligation. These results provide genetic evidence for a critical role of SLP-76 in the regulation of neutrophil function.


Subject(s)
Integrins/metabolism , Neutrophils/metabolism , Phosphoproteins/metabolism , Receptors, IgG/metabolism , Signal Transduction/physiology , Adaptor Proteins, Signal Transducing , Animals , Cell Membrane/metabolism , Gene Targeting , Mice , Neutrophils/ultrastructure , Phosphoproteins/deficiency , Phosphoproteins/genetics , Phosphorylation , Tyrosine/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...