Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 64
Filter
1.
Nat Immunol ; 22(9): 1093-1106, 2021 09.
Article in English | MEDLINE | ID: mdl-34282331

ABSTRACT

Neutrophils display distinct gene expression patters depending on their developmental stage, activation state and tissue microenvironment. To determine the transcription factor networks that shape these responses in a mouse model, we integrated transcriptional and chromatin analyses of neutrophils during acute inflammation. We showed active chromatin remodeling at two transition stages: bone marrow-to-blood and blood-to-tissue. Analysis of differentially accessible regions revealed distinct sets of putative transcription factors associated with control of neutrophil inflammatory responses. Using ex vivo and in vivo approaches, we confirmed that RUNX1 and KLF6 modulate neutrophil maturation, whereas RELB, IRF5 and JUNB drive neutrophil effector responses and RFX2 and RELB promote survival. Interfering with neutrophil activation by targeting one of these factors, JUNB, reduced pathological inflammation in a mouse model of myocardial infarction. Therefore, our study represents a blueprint for transcriptional control of neutrophil responses in acute inflammation and opens possibilities for stage-specific therapeutic modulation of neutrophil function in disease.


Subject(s)
Chromatin Assembly and Disassembly/genetics , Inflammation/immunology , Neutrophils/immunology , Transcriptional Activation/genetics , Animals , CHO Cells , Cell Line , Core Binding Factor Alpha 2 Subunit/metabolism , Cricetulus , Female , Interferon Regulatory Factors/metabolism , Kruppel-Like Factor 6/metabolism , Mice , Mice, Inbred C57BL , Myocardial Infarction/immunology , Myocardial Infarction/pathology , Regulatory Factor X Transcription Factors/metabolism , Transcription Factor RelB/metabolism , Transcription Factors/metabolism , Transcription, Genetic/genetics
2.
Nature ; 631(8021): 645-653, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38987596

ABSTRACT

Platelet homeostasis is essential for vascular integrity and immune defence1,2. Although the process of platelet formation by fragmenting megakaryocytes (MKs; thrombopoiesis) has been extensively studied, the cellular and molecular mechanisms required to constantly replenish the pool of MKs by their progenitor cells (megakaryopoiesis) remains unclear3,4. Here we use intravital imaging to track the cellular dynamics of megakaryopoiesis over days. We identify plasmacytoid dendritic cells (pDCs) as homeostatic sensors that monitor the bone marrow for apoptotic MKs and deliver IFNα to the MK niche triggering local on-demand proliferation and maturation of MK progenitors. This pDC-dependent feedback loop is crucial for MK and platelet homeostasis at steady state and under stress. pDCs are best known for their ability to function as vigilant detectors of viral infection5. We show that virus-induced activation of pDCs interferes with their function as homeostatic sensors of megakaryopoiesis. Consequently, activation of pDCs by SARS-CoV-2 leads to excessive megakaryopoiesis. Together, we identify a pDC-dependent homeostatic circuit that involves innate immune sensing and demand-adapted release of inflammatory mediators to maintain homeostasis of the megakaryocytic lineage.


Subject(s)
Dendritic Cells , Homeostasis , Megakaryocytes , Thrombopoiesis , Animals , Female , Humans , Male , Mice , Apoptosis , Blood Platelets/cytology , Bone Marrow , Cell Lineage , Cell Proliferation , Dendritic Cells/immunology , Dendritic Cells/cytology , Feedback, Physiological , Immunity, Innate , Intravital Microscopy , Megakaryocytes/cytology , Megakaryocytes/immunology , Mice, Inbred C57BL , SARS-CoV-2/immunology , COVID-19/immunology , COVID-19/physiopathology , COVID-19/virology
3.
Nat Immunol ; 14(1): 41-51, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23179077

ABSTRACT

Coordinated navigation within tissues is essential for cells of the innate immune system to reach the sites of inflammatory processes, but the signals involved are incompletely understood. Here we demonstrate that NG2(+) pericytes controlled the pattern and efficacy of the interstitial migration of leukocytes in vivo. In response to inflammatory mediators, pericytes upregulated expression of the adhesion molecule ICAM-1 and released the chemoattractant MIF. Arteriolar and capillary pericytes attracted and interacted with myeloid leukocytes after extravasating from postcapillary venules, 'instructing' them with pattern-recognition and motility programs. Inhibition of MIF neutralized the migratory cues provided to myeloid leukocytes by NG2(+) pericytes. Hence, our results identify a previously unknown role for NG2(+) pericytes as an active component of innate immune responses, which supports the immunosurveillance and effector function of extravasated neutrophils and macrophages.


Subject(s)
Intercellular Adhesion Molecule-1/metabolism , Intramolecular Oxidoreductases/metabolism , Leukocytes/immunology , Macrophage Migration-Inhibitory Factors/metabolism , Pericytes/immunology , Receptors, Pattern Recognition/immunology , Antibodies, Blocking/pharmacology , Arterioles/immunology , Capillaries/immunology , Cell Communication/drug effects , Cell Movement/drug effects , Cell Movement/immunology , Cells, Cultured , Humans , Immunity, Innate , Inflammation Mediators/metabolism , Intercellular Adhesion Molecule-1/genetics , Intramolecular Oxidoreductases/genetics , Intramolecular Oxidoreductases/immunology , Leukocytes/drug effects , Macrophage Migration-Inhibitory Factors/genetics , Macrophage Migration-Inhibitory Factors/immunology , Neutrophil Activation/drug effects , Up-Regulation/drug effects , Venules/immunology
4.
Blood ; 141(6): 645-658, 2023 02 09.
Article in English | MEDLINE | ID: mdl-36223592

ABSTRACT

The mechanisms of coordinated changes in proteome composition and their relevance for the differentiation of neutrophil granulocytes are not well studied. Here, we discover 2 novel human genetic defects in signal recognition particle receptor alpha (SRPRA) and SRP19, constituents of the mammalian cotranslational targeting machinery, and characterize their roles in neutrophil granulocyte differentiation. We systematically study the proteome of neutrophil granulocytes from patients with variants in the SRP genes, HAX1, and ELANE, and identify global as well as specific proteome aberrations. Using in vitro differentiation of human induced pluripotent stem cells and in vivo zebrafish models, we study the effects of SRP deficiency on neutrophil granulocyte development. In a heterologous cell-based inducible protein expression system, we validate the effects conferred by SRP dysfunction for selected proteins that we identified in our proteome screen. Thus, SRP-dependent protein processing, intracellular trafficking, and homeostasis are critically important for the differentiation of neutrophil granulocytes.


Subject(s)
Induced Pluripotent Stem Cells , Proteome , Animals , Humans , Zebrafish , Human Genetics , Mammals , Adaptor Proteins, Signal Transducing
5.
J Immunol ; 208(6): 1445-1455, 2022 03 15.
Article in English | MEDLINE | ID: mdl-35181637

ABSTRACT

Plasmacytoid dendritic cells (pDCs) display an increased abundance in visceral adipose tissue (VAT) of humans with obesity. In the current study, we set out to decipher the molecular mechanisms of their recruitment to VAT and the functional relevance of this process. We observed increased pDC numbers in murine blood, liver, spleen, and VAT after feeding a high-fat diet (HFD) for 3 wk when compared with a standard diet. pDCs were enriched in fat-associated lymphoid clusters representing highly specific lymphoid regions within VAT. HFD led to an enlargement of fat-associated lymphoid clusters with an increased density and migratory speed of pDCs as shown by intravital multiphoton microscopy. For their recruitment into VAT, pDCs employed P-selectin with E-selectin and L-selectin being only critical in response to HFD, indicating that the molecular cues underlying pDC trafficking were dependent on the nutritional state. Subsequent recruitment steps required α4ß1 and α4ß7 integrins and engagement of CCR7. Application of fingolimod (FTY720) abrogated egress of pDCs from VAT, indicating the involvement of sphingosine-1-phosphate in this process. Furthermore, HFD altered pDC functions by promoting their activation and type 1 IFN expression. Blocking pDC infiltration into VAT prevented weight gain and improved glucose tolerance during HFD. In summary, a HFD fundamentally alters pDC biology by promoting their trafficking, retention, and activation in VAT, which in turn seems to regulate metabolism.


Subject(s)
Diet, High-Fat , Intra-Abdominal Fat , Adipose Tissue , Animals , Dendritic Cells , Intra-Abdominal Fat/metabolism , Mice , Phenotype
6.
Eur J Immunol ; 52(5): 760-769, 2022 05.
Article in English | MEDLINE | ID: mdl-35099066

ABSTRACT

Signaling via ß2 integrins (CD11/CD18) as well as TCRs and BCRs involves similar pathways. However, the activation of the same signaling molecule can result in opposing effects. One such example is the hematopoietic progenitor kinase 1 (HPK1), which negatively regulates T and B cell activation but enforces neutrophil adhesion via ß2 integrins. This difference may be defined by specific HPK1 interacting networks in different leukocyte subsets which have already been described in the adaptive immune system. Here, we set out to identify interacting proteins of HPK1 in neutrophil-like differentiated HL-60 cells exposed to immobilized fibrinogen and left nonactivated or Mn2+ -activated to allow ß2 integrin-dependent adhesion. Co-IP experiments followed by mass spectrometry led to the identification of 115 HPK1-interacting proteins. A total of 58 proteins were found only in nonactivated cells and 39 proteins only in Mn2+ -activated adherent cells. From these results, we decoded a pre-existing signaling cluster of HPK1 in nonactivated cells encompassing proteins essential for ß2 integrin-mediated signaling during neutrophil trafficking, namely DNAX-activation protein 12 (DAP12), spleen tyrosine kinase (Syk), and Rac1. Thus, our study provides novel insights into the complex architecture of the signaling processes during neutrophil activation and the complex signaling profile of HPK1 in leukocytes.


Subject(s)
Proteomics , Receptors, Antigen, T-Cell , Humans , Immunity, Innate , Integrins/metabolism , Protein Serine-Threonine Kinases
7.
J Immunol ; 206(2): 432-445, 2021 01 15.
Article in English | MEDLINE | ID: mdl-33310871

ABSTRACT

Although neutrophils play important roles in immunity and inflammation, their analysis is strongly hindered by their short-lived and terminally differentiated nature. Prior studies reported conditional immortalization of myeloid progenitors using retroviral expression of an estrogen-dependent fusion protein of the HoxB8 transcription factor. This approach allowed the long-term culture of mouse myeloid progenitors (HoxB8 progenitors) in estrogen-containing media, followed by differentiation toward neutrophils upon estrogen withdrawal. Although several reports confirmed the in vitro functional responsiveness of the resulting differentiated cells (HoxB8 neutrophils), little is known about their capacity to perform in vivo neutrophil functions. We have addressed this issue by an in vivo transplantation approach. In vitro-generated HoxB8 neutrophils showed a neutrophil-like phenotype and were able to perform conventional neutrophil functions, like respiratory burst, chemotaxis, and phagocytosis. The i.v. injection of HoxB8 progenitors into lethally irradiated recipients resulted in the appearance of circulating donor-derived HoxB8 neutrophils. In vivo-differentiated HoxB8 neutrophils were able to migrate to the inflamed peritoneum and to phagocytose heat-killed Candida particles. The reverse passive Arthus reaction could be induced in HoxB8 chimeras but not in irradiated, nontransplanted control animals. Repeated injection of HoxB8 progenitors also allowed us to maintain stable circulating HoxB8 neutrophil counts for several days. Injection of arthritogenic K/B×N serum triggered robust arthritis in HoxB8 chimeras, but not in irradiated, nontransplanted control mice. Taken together, our results indicate that HoxB8 progenitor-derived neutrophils are capable of performing various in vivo neutrophil functions, providing a framework for using the HoxB8 system for the in vivo analysis of neutrophil function.


Subject(s)
Adenoviridae/genetics , Genetic Vectors/genetics , Homeodomain Proteins/metabolism , Inflammation/immunology , Myeloid Progenitor Cells/immunology , Neutrophils/immunology , Animals , Cell Differentiation , Cell Line, Transformed , Chemotaxis , Gene Expression Regulation , Homeodomain Proteins/genetics , Mice , Mice, Inbred C57BL , Signal Transduction , Transgenes/genetics
8.
J Am Soc Nephrol ; 31(2): 257-278, 2020 02.
Article in English | MEDLINE | ID: mdl-31932472

ABSTRACT

BACKGROUND: Mononuclear phagocytes (MPs), including macrophages, monocytes, and dendritic cells (DCs), are phagocytic cells with important roles in immunity. The developmental origin of kidney DCs has been highly debated because of the large phenotypic overlap between macrophages and DCs in this tissue. METHODS: We used fate mapping, RNA sequencing, flow cytometry, confocal microscopy, and histo-cytometry to assess the origin and phenotypic and functional properties of renal DCs in healthy kidney and of DCs after cisplatin and ischemia reperfusion-induced kidney injury. RESULTS: Adult kidney contains at least four subsets of MPs with prominent Clec9a-expression history indicating a DC origin. We demonstrate that these populations are phenotypically, functionally, and transcriptionally distinct from each other. We also show these kidney MPs exhibit unique age-dependent developmental heterogeneity. Kidneys from newborn mice contain a prominent population of embryonic-derived MHCIInegF4/80hiCD11blow macrophages that express T cell Ig and mucin domain containing 4 (TIM-4) and MER receptor tyrosine kinase (MERTK). These macrophages are replaced within a few weeks after birth by phenotypically similar cells that express MHCII but lack TIM-4 and MERTK. MHCII+F4/80hi cells exhibit prominent Clec9a-expression history in adulthood but not early life, indicating additional age-dependent developmental heterogeneity. In AKI, MHCIInegF4/80hi cells reappear in adult kidneys as a result of MHCII downregulation by resident MHCII+F4/80hi cells, possibly in response to prostaglandin E2 (PGE2). RNA sequencing further suggests MHCII+F4/80hi cells help coordinate the recruitment of inflammatory cells during renal injury. CONCLUSIONS: Distinct developmental programs contribute to renal DC and macrophage populations throughout life, which could have important implications for therapies targeting these cells.


Subject(s)
Dendritic Cells/immunology , Kidney/immunology , Macrophages/immunology , Nephritis/immunology , Acute Kidney Injury/immunology , Age Factors , Animals , CD11b Antigen/analysis , CX3C Chemokine Receptor 1/analysis , Calcium-Binding Proteins/analysis , Cisplatin/pharmacology , Histocompatibility Antigens Class II/analysis , Kidney/drug effects , Kidney/metabolism , Lectins, C-Type/analysis , Mice , Mice, Inbred C57BL , Receptors, G-Protein-Coupled/analysis , Receptors, Immunologic/analysis
9.
Blood ; 131(17): 1887-1898, 2018 04 26.
Article in English | MEDLINE | ID: mdl-29487067

ABSTRACT

Neutrophil extravasation and interstitial migration are important steps during the recruitment of neutrophils to sites of inflammation. In the present study, we addressed the functional importance of the unconventional class I myosin 1f (Myo1f) for neutrophil trafficking during acute inflammation. In contrast to leukocyte rolling and adhesion, the genetic absence of Myo1f severely compromised neutrophil extravasation into the inflamed mouse cremaster tissue when compared with Myo1f+/+ mice as studied by intravital microscopy. Similar results were obtained in experimental models of acute peritonitis and acute lung injury. In contrast to 2-dimensional migration, which occurred independently of Myo1f, Myo1f was indispensable for neutrophil migration in 3-dimensional (3D) environments, that is, transmigration and migration in collagen networks as it regulated squeezing and dynamic deformation of the neutrophil nucleus during migration through physical barriers. Thus, we provide evidence for an important role of Myo1f in neutrophil trafficking during inflammation by specifically regulating neutrophil extravasation and migration in 3D environments.


Subject(s)
Abdominal Muscles/metabolism , Acute Lung Injury/metabolism , Cell Movement , Myosin Type I/metabolism , Neutrophil Infiltration , Neutrophils/metabolism , Peritonitis/metabolism , Abdominal Muscles/pathology , Acute Disease , Acute Lung Injury/genetics , Acute Lung Injury/pathology , Animals , Disease Models, Animal , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Mice , Mice, Knockout , Myosin Type I/genetics , Neutrophils/pathology , Peritonitis/genetics , Peritonitis/pathology
10.
J Immunol ; 201(6): 1748-1764, 2018 09 15.
Article in English | MEDLINE | ID: mdl-30068598

ABSTRACT

Neutrophils are the first leukocytes to arrive at sites of injury during the acute inflammatory response. To maintain the polarized morphology during migration, nonmuscle myosins class II are essential, but studies using genetic models to investigate the role of Myh9 for neutrophil migration were missing. In this study, we analyzed the functional role of Myh9 on neutrophil trafficking using genetic downregulation of Myh9 in Vav-iCre+/Myh9wt/fl mice because the complete knockout of Myh9 in the hematopoietic system was lethal. Migration velocity and Euclidean distance were significantly diminished during mechanotactic migration of Vav-iCre+/Myh9wt/fl neutrophils compared with Vav-iCre-/Myh9wt/fl control neutrophils. Similar results were obtained for transmigration and migration in confined three-dimensional environments. Stimulated emission depletion nanoscopy revealed that a certain threshold of Myh9 was required to maintain proper F-actin dynamics in the front of the migrating cell. In laser-induced skin injury and in acute peritonitis, reduced Myh9 expression in the hematopoietic system resulted in significantly diminished neutrophil extravasation. Investigation of bone marrow chimeric mice in the peritonitis model revealed that the migration defect was cell intrinsic. Expression of Myh9-EGFP rescued the Myh9-related defects in two-dimensional and three-dimensional migration of Hoxb8-SCF cell-derived neutrophils generated from fetal liver cells with a Myh9 knockdown. Live cell imaging provided evidence that Myh9 was localized in branching lamellipodia and in the uropod where it may enable fast neutrophil migration. In summary, the severe migration defects indicate an essential and fundamental role of Myh9 for neutrophil trafficking in innate immunity.


Subject(s)
Cell Movement/immunology , Immunity, Innate , Neutrophil Infiltration , Neutrophils/immunology , Nonmuscle Myosin Type IIA/immunology , Pseudopodia/immunology , Actins/genetics , Actins/immunology , Animals , Cell Movement/genetics , Mice , Mice, Transgenic , Myosin Heavy Chains , Neutrophils/pathology , Nonmuscle Myosin Type IIA/genetics , Peritonitis/genetics , Peritonitis/immunology , Peritonitis/pathology , Pseudopodia/genetics , Skin/immunology , Skin/injuries , Skin/pathology
11.
Blood ; 130(7): 847-858, 2017 08 17.
Article in English | MEDLINE | ID: mdl-28615221

ABSTRACT

Trafficking of polymorphonuclear neutrophils (PMNs) during inflammation critically depends on the ß2 integrins lymphocyte function-associated antigen 1 (LFA-1) (CD11a/CD18) and macrophage-1 antigen (CD11b/CD18). Here, we identify coronin 1A (Coro1A) as a novel regulator of ß2 integrins that interacts with the cytoplasmic tail of CD18 and is crucial for induction of PMN adhesion and postadhesion events, including adhesion strengthening, spreading, and migration under flow conditions. Transition of PMN rolling to firm adhesion critically depends on Coro1A by regulating the accumulation of high-affinity LFA-1 in focal zones of adherent cells. Defective integrin affinity regulation in the genetic absence of Coro1A impairs leukocyte adhesion and extravasation in inflamed cremaster muscle venules in comparison with control animals. In a Helicobacter pylori mouse infection model, PMN infiltration into the gastric mucosa is dramatically reduced in Coro1A-/- mice, resulting in an attenuated gastric inflammation. Thus, Coro1A represents an important novel player in integrin biology, with key functions in PMN trafficking during innate immunity.


Subject(s)
4-Butyrolactone/analogs & derivatives , CD18 Antigens/metabolism , Cell Movement , Immunity, Innate , Neutrophils/cytology , Neutrophils/metabolism , 4-Butyrolactone/metabolism , Actins/metabolism , Animals , Calcium Signaling , Cell Adhesion , Gastritis/immunology , Gastritis/microbiology , Gastritis/pathology , Helicobacter pylori/physiology , Lymphocyte Function-Associated Antigen-1/metabolism , Macrophage-1 Antigen/metabolism , Mice, Inbred C57BL , Receptors, G-Protein-Coupled/metabolism , Rheology
12.
Circ Res ; 120(1): 99-109, 2017 Jan 06.
Article in English | MEDLINE | ID: mdl-27777247

ABSTRACT

RATIONALE: The AMP-activated protein kinase (AMPK) is stimulated by hypoxia, and although the AMPKα1 catalytic subunit has been implicated in angiogenesis, little is known about the role played by the AMPKα2 subunit in vascular repair. OBJECTIVE: To determine the role of the AMPKα2 subunit in vascular repair. METHODS AND RESULTS: Recovery of blood flow after femoral artery ligation was impaired (>80%) in AMPKα2-/- versus wild-type mice, a phenotype reproduced in mice lacking AMPKα2 in myeloid cells (AMPKα2ΔMC). Three days after ligation, neutrophil infiltration into ischemic limbs of AMPKα2ΔMC mice was lower than that in wild-type mice despite being higher after 24 hours. Neutrophil survival in ischemic tissue is required to attract monocytes that contribute to the angiogenic response. Indeed, apoptosis was increased in hypoxic neutrophils from AMPKα2ΔMC mice, fewer monocytes were recruited, and gene array analysis revealed attenuated expression of proangiogenic proteins in ischemic AMPKα2ΔMC hindlimbs. Many angiogenic growth factors are regulated by hypoxia-inducible factor, and hypoxia-inducible factor-1α induction was attenuated in AMPKα2-deficient cells and accompanied by its enhanced hydroxylation. Also, fewer proteins were regulated by hypoxia in neutrophils from AMPKα2ΔMC mice. Mechanistically, isocitrate dehydrogenase expression and the production of α-ketoglutarate, which negatively regulate hypoxia-inducible factor-1α stability, were attenuated in neutrophils from wild-type mice but remained elevated in cells from AMPKα2ΔMC mice. CONCLUSIONS: AMPKα2 regulates α-ketoglutarate generation, hypoxia-inducible factor-1α stability, and neutrophil survival, which in turn determine further myeloid cell recruitment and repair potential. The activation of AMPKα2 in neutrophils is a decisive event in the initiation of vascular repair after ischemia.


Subject(s)
AMP-Activated Protein Kinases/physiology , Apoptosis/physiology , Hypoxia-Inducible Factor 1, alpha Subunit/biosynthesis , Ischemia/metabolism , Neutrophils/metabolism , Animals , Blood Vessels/metabolism , Blood Vessels/pathology , Endothelial Cells/metabolism , Endothelial Cells/pathology , Hindlimb/blood supply , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Ischemia/pathology , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic
13.
Arterioscler Thromb Vasc Biol ; 38(5): 1007-1019, 2018 05.
Article in English | MEDLINE | ID: mdl-29567680

ABSTRACT

OBJECTIVE: Cardiovascular diseases and depression are the leading causes of disability in Western countries. Clinical data on potential cardiovascular effects of serotonin reuptake inhibitors (SSRIs), the most commonly used antidepressant drugs, are controversial. In addition to blocking serotonin reuptake transporter in the brain, SSRIs deplete the major peripheral serotonin (5-hydroxytryptamine [5-HT]) storage by inhibiting serotonin reuptake transporter-mediated uptake in platelets. In this study, we aimed to investigate the effect of chronic SSRI intake on the development of atherosclerosis. APPROACH AND RESULTS: Treatment of apolipoprotein E-deficient mice with the SSRI fluoxetine for 2, 4, or 16 weeks increased atherosclerotic lesion formation, with most pronounced effect during early plaque development. Intravital microscopy of carotid arteries revealed enhanced myeloid cell adhesion on fluoxetine treatment. Mechanistically, we found that fluoxetine augmented vascular permeability and increased chemokine-induced integrin-binding activity of circulating leukocytes. In vitro stimulation of murine blood demonstrated that fluoxetine, but not 5-HT, could directly promote ß1 and ß2 integrin activation provided C-C motif chemokine ligand 5 was also present. Similar effects were observed with the SSRI escitalopram. Enhanced C-C motif chemokine ligand 5-induced integrin activation by fluoxetine was also confirmed in a human neutrophil-like cell line. In contrast to the proatherogenic properties of fluoxetine, pharmacological inhibition of the peripheral 5-HT synthesizing enzyme tryptophan hydroxylase 1 did not promote atherosclerosis, suggesting that the proatherogenic effect of fluoxetine occurs independent of peripheral 5-HT depletion. CONCLUSIONS: SSRI intake may promote atherosclerosis and therefore potentially increase the risk for acute cardiovascular events by a mechanism that is independent of 5-HT depletion.


Subject(s)
Aorta/drug effects , Aortic Diseases/chemically induced , Atherosclerosis/chemically induced , Carotid Arteries/drug effects , Carotid Artery Diseases/chemically induced , Fluoxetine/toxicity , Plaque, Atherosclerotic , Selective Serotonin Reuptake Inhibitors/toxicity , Animals , Aorta/metabolism , Aorta/pathology , Aortic Diseases/blood , Aortic Diseases/genetics , Aortic Diseases/pathology , Atherosclerosis/blood , Atherosclerosis/genetics , Atherosclerosis/pathology , Blood Platelets/drug effects , Blood Platelets/metabolism , CD18 Antigens/blood , Capillary Permeability/drug effects , Carotid Arteries/metabolism , Carotid Arteries/pathology , Carotid Artery Diseases/blood , Carotid Artery Diseases/genetics , Carotid Artery Diseases/pathology , Cell Adhesion/drug effects , Chemokine CCL5/blood , Disease Models, Animal , Disease Progression , Drug Administration Schedule , Fluoxetine/administration & dosage , HEK293 Cells , HL-60 Cells , Humans , Integrin beta1/blood , Male , Mice, Inbred C57BL , Mice, Knockout, ApoE , Myeloid Cells/drug effects , Myeloid Cells/metabolism , Serotonin/blood , Selective Serotonin Reuptake Inhibitors/administration & dosage , Signal Transduction , Time Factors
14.
Eur J Clin Invest ; 48 Suppl 2: e12966, 2018 Nov.
Article in English | MEDLINE | ID: mdl-29896791

ABSTRACT

BACKGROUND: Neutrophil recruitment during acute inflammation critically depends on the spatial and temporal regulation of ß2 integrins (CD11/CD18). This regulation occurs by inside-out and outside-in signalling via interaction of cytoplasmic proteins with the intracellular domains of the integrin α- and ß-subunits. The underlying molecular mechanisms regulating ß2 integrins in neutrophils are still incompletely understood. AIM: This review provides a comprehensive overview of our current knowledge on proteins interacting with the cytoplasmic tail of CD18, the conserved ß-subunit of ß2 integrins, their regulation and their functional importance for neutrophil trafficking during acute inflammation. RESULTS: A total of 22 proteins including Talin, Kindlin 3 and Coronin 1A have been reported to interact with the CD18 cytoplasmic tail. Here, proteins binding to the cytoplasmic domain of CD18 in experiments using purified, recombinant proteins or peptides in, for example, pull-down assays, are defined as direct interactors. Proteins that have been shown to interact with the cytoplasmic domain of CD18 using whole cell lysates in, for example, pull-down experiments are claimed as interacting proteins without evidence for direct interaction. In summary, ß2 integrin activation and signalling depend on a specific subset of proteins interacting with CD18 and their precise regulation. If disturbed, profound defects of neutrophil recruitment and activation become evident compromising the innate immune response. CONCLUSIONS: The knowledge of proteins interacting with ß2 integrins and their regulation during neutrophil trafficking does not only improve our basic understanding of innate immunity but may pave the way to novel therapeutic strategies in the treatment of inflammatory diseases.


Subject(s)
CD18 Antigens/physiology , Neutrophils/physiology , Cell Adhesion/physiology , Cell Movement/physiology , Humans , Neutrophil Infiltration/physiology , Protein Binding/physiology , Proteins/physiology , Signal Transduction/physiology
15.
Circulation ; 134(16): 1176-1188, 2016 10 18.
Article in English | MEDLINE | ID: mdl-27660294

ABSTRACT

BACKGROUND: Therapeutic targeting of arterial leukocyte recruitment in the context of atherosclerosis has been disappointing in clinical studies. Reasons for such failures include the lack of knowledge of arterial-specific recruitment patterns. Here we establish the importance of the cathepsin G (CatG) in the context of arterial myeloid cell recruitment. METHODS: Intravital microscopy of the carotid artery, the jugular vein, and cremasteric arterioles and venules in Apoe-/-and CatG-deficient mice (Apoe-/-Ctsg-/-) was used to study site-specific myeloid cell behavior after high-fat diet feeding or tumor necrosis factor stimulation. Atherosclerosis development was assessed in aortic root sections after 4 weeks of high-fat diet, whereas lung inflammation was assessed after inhalation of lipopolysaccharide. Endothelial deposition of CatG and CCL5 was quantified in whole-mount preparations using 2-photon and confocal microscopy. RESULTS: Our observations elucidated a crucial role for CatG during arterial leukocyte adhesion, an effect not found during venular adhesion. Consequently, CatG deficiency attenuates atherosclerosis but not acute lung inflammation. Mechanistically, CatG is immobilized on arterial endothelium where it activates leukocytes to firmly adhere engaging integrin clustering, a process of crucial importance to achieve effective adherence under high-shear flow. Therapeutic neutralization of CatG specifically abrogated arterial leukocyte adhesion without affecting myeloid cell adhesion in the microcirculation. Repetitive application of CatG-neutralizing antibodies permitted inhibition of atherogenesis in mice. CONCLUSIONS: Taken together, these findings present evidence of an arterial-specific recruitment pattern centered on CatG-instructed adhesion strengthening. The inhibition of this process could provide a novel strategy for treatment of arterial inflammation with limited side effects.


Subject(s)
Arteries , Cathepsin G/metabolism , Chemotaxis , Myeloid Cells/metabolism , Venules , Animals , Atherosclerosis/drug therapy , Atherosclerosis/etiology , Atherosclerosis/metabolism , Atherosclerosis/pathology , Biomarkers , Cathepsin G/antagonists & inhibitors , Cathepsin G/genetics , Cell Adhesion/genetics , Chemokine CCL5/genetics , Chemokine CCL5/metabolism , Chemotaxis/genetics , Chemotaxis/immunology , Disease Models, Animal , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Humans , Integrins/metabolism , Leukocyte Rolling , Mice , Mice, Knockout , Microcirculation , Myeloid Cells/immunology , Protein Binding , Shear Strength
16.
Blood ; 125(26): 4069-77, 2015 Jun 25.
Article in English | MEDLINE | ID: mdl-25795918

ABSTRACT

Platelet aggregation at sites of vascular injury is not only essential for hemostasis, but may also cause acute ischemic disease states such as myocardial infarction or stroke. The hemi-immunoreceptor tyrosine-based activation motif-containing C-type lectinlike receptor 2 (CLEC-2) mediates powerful platelet activation through a Src- and spleen tyrosine kinase (Syk)-dependent tyrosine phosphorylation cascade. Thereby, CLEC-2 not only contributes to thrombus formation and stabilization but also plays a central role in blood-lymphatic vessel development, tumor metastasis, and prevention of inflammatory bleeding, making it a potential pharmacologic target to modulate these processes. We have previously shown that injection of the anti-CLEC-2 antibody, INU1, results in virtually complete immunodepletion of platelet CLEC-2 in mice, which is, however, preceded by a severe transient thrombocytopenia thereby limiting its potential therapeutic use. The mechanisms underlying this targeted CLEC-2 downregulation have remained elusive. Here, we show that INU1-induced CLEC-2 immunodepletion occurs through Src-family kinase-dependent receptor internalization in vitro and in vivo, presumably followed by intracellular degradation. In mice with platelet-specific Syk deficiency, INU1-induced CLEC-2 internalization/degradation was fully preserved whereas the associated thrombocytopenia was largely prevented. These results show for the first time that CLEC-2 can be downregulated from the platelet surface through internalization in vitro and in vivo and that this can be mechanistically uncoupled from the associated antibody-induced thrombocytopenia.


Subject(s)
Blood Platelets/metabolism , Lectins, C-Type/metabolism , Platelet Activation/physiology , Animals , Antibodies, Monoclonal/pharmacology , Blotting, Western , Down-Regulation , Flow Cytometry , Fluorescent Antibody Technique , Intracellular Signaling Peptides and Proteins/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Protein-Tyrosine Kinases/metabolism , Syk Kinase , Thrombocytopenia/chemically induced
17.
Arterioscler Thromb Vasc Biol ; 36(6): 1247-53, 2016 06.
Article in English | MEDLINE | ID: mdl-27102960

ABSTRACT

OBJECTIVE: Ischemic stroke, which is mainly caused by thromboembolic occlusion of brain arteries, is the second leading cause of death and disability worldwide with limited treatment options. The platelet collagen receptor glycoprotein VI (GPVI) is a key player in arterial thrombosis and a critical determinant of stroke outcome, making its signaling pathway an attractive target for pharmacological intervention. The spleen tyrosine kinase (Syk) is an essential signaling mediator downstream of not only GPVI but also other platelet and immune cell receptors. We sought to assess whether Syk might be an effective antithrombotic target. APPROACH AND RESULTS: We demonstrate that mice lacking Syk in platelets specifically are protected from arterial thrombus formation and ischemic stroke but display unaltered hemostasis. Furthermore, we show that mice treated with the novel, selective, and orally bioavailable Syk inhibitor BI1002494 were protected in a model of arterial thrombosis and had smaller infarct sizes and a significantly better neurological outcome 24 hours after transient middle cerebral artery occlusion, also when BI1002494 was administered therapeutically, that is, after ischemia. CONCLUSIONS: These results provide direct evidence that pharmacological Syk inhibition might provide a safe therapeutic strategy to prevent arterial thrombosis and to limit infarct progression in acute stroke.


Subject(s)
Arterial Occlusive Diseases/prevention & control , Blood Platelets/drug effects , Fibrinolytic Agents/administration & dosage , Hemostasis/drug effects , Infarction, Middle Cerebral Artery/prevention & control , Protein Kinase Inhibitors/administration & dosage , Syk Kinase/antagonists & inhibitors , Thrombosis/prevention & control , Administration, Oral , Animals , Arterial Occlusive Diseases/blood , Arterial Occlusive Diseases/enzymology , Arterial Occlusive Diseases/genetics , Blood Platelets/enzymology , Brain/drug effects , Brain/enzymology , Brain/pathology , Disease Models, Animal , Dose-Response Relationship, Drug , Genotype , Infarction, Middle Cerebral Artery/blood , Infarction, Middle Cerebral Artery/enzymology , Infarction, Middle Cerebral Artery/genetics , Mice, Inbred C57BL , Mice, Knockout , Motor Activity/drug effects , Phenotype , Signal Transduction/drug effects , Syk Kinase/blood , Syk Kinase/deficiency , Syk Kinase/genetics , Thrombosis/blood , Thrombosis/enzymology , Thrombosis/genetics , Time Factors
18.
Blood ; 123(12): 1887-96, 2014 Mar 20.
Article in English | MEDLINE | ID: mdl-24458438

ABSTRACT

Emerging evidence suggests a role of the cytokine midkine (MK) in inflammation. In this study, its functional relevance for recruitment of polymorphonuclear neutrophils (PMNs) during acute inflammation was investigated. Intravital microscopy and histologic analysis of tumor necrosis factor-α-stimulated cremaster muscle venules revealed severely compromised leukocyte adhesion and extravasation in MK(-/-) mice compared with MK(+/+) animals. Systemic administration of recombinant MK completely rescued the adhesion defect in MK(-/-) mice. In a hind limb ischemia model, leukocyte accumulation in MK(-/-) mice was significantly diminished compared with MK(+/+) animals. However, MK did not lead to an inflammatory activation of PMNs or endothelial cells suggesting that it does not serve as classical proinflammatory cytokine. Unexpectedly, immobilized MK mediated PMN adhesion under static and flow conditions, whereas PMN-derived MK was dispensable for the induction of adhesion. Furthermore, adhesion strengthening remained unaffected by MK. Flow cytometry revealed that immobilized, but not soluble MK, significantly promoted the high affinity conformation of ß2 integrins of PMNs. Blocking studies of low-density lipoprotein receptor-related protein 1 (LRP1) suggested that LRP1 may act as a receptor for MK on PMNs. Thus, MK seems to support PMN adhesion by promoting the high affinity conformation of ß2 integrins, thereby facilitating PMN trafficking during acute inflammation.


Subject(s)
CD18 Antigens/physiology , Inflammation/physiopathology , Intercellular Signaling Peptides and Proteins/physiology , Neutrophils/physiology , Animals , CD11 Antigens/physiology , CD18 Antigens/genetics , Cell Adhesion/immunology , Cell Adhesion/physiology , Cytokines/immunology , Cytokines/physiology , Humans , Inflammation/immunology , Inflammation/pathology , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/immunology , Low Density Lipoprotein Receptor-Related Protein-1/immunology , Low Density Lipoprotein Receptor-Related Protein-1/physiology , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Midkine , Nerve Growth Factors/genetics , Nerve Growth Factors/immunology , Nerve Growth Factors/physiology , Neutrophils/immunology , Neutrophils/pathology , Receptors, LDL/immunology , Receptors, LDL/physiology , Tumor Suppressor Proteins/immunology , Tumor Suppressor Proteins/physiology
19.
Blood ; 121(20): 4184-94, 2013 May 16.
Article in English | MEDLINE | ID: mdl-23460610

ABSTRACT

Recruitment of polymorphonuclear neutrophils (PMNs) to sites of acute inflammation critically depends on ß2 integrins (CD11/CD18). Recently, the mammalian actin-binding protein 1 (mAbp1) was identified as an important adaptor protein regulating PMN trafficking downstream of ß2 integrins. Here, we show that mAbp1 constitutively co-immunoprecipitated with hematopoietic progenitor kinase 1 (HPK1) in neutrophil-like differentiated HL-60 (dHL-60) cells. HPK1 was enriched at the lamellipodium of polarized dHL-60 cells, where it colocalized with mAbp1 and actin. Functional analysis of PMNs from HPK1-deficient mice showed that HPK1 was critical for CXCL1-induced lymphocyte function-associated antigen 1 (LFA-1)-mediated PMN adhesion to ICAM-1 under flow conditions. Accordingly, CXCL1-mediated induction of high-affinity LFA-1 required HPK1, but macrophage antigen 1 (Mac-1) affinity regulation was independent of HPK1. Intravital microscopy of the mouse cremaster muscle confirmed the defect of CXCL1-induced leukocyte adhesion in HPK1-deficient mice. Furthermore, ß2 integrin-mediated post-adhesion processes-adhesion strengthening, spreading, and directed mechanotactic crawling of PMNs under flow conditions-involved HPK1 in vitro and in vivo. Upon intrascrotal administration of tumor necrosis factor α (TNF-α), PMN adhesion and extravasation were severely compromised in HPK1-deficient mice. In summary, our results indicate that HPK1 is critically involved in LFA-1-mediated PMN trafficking during acute inflammation.


Subject(s)
Acute-Phase Reaction/genetics , Inflammation/genetics , Lymphocyte Function-Associated Antigen-1/physiology , Neutrophil Infiltration/genetics , Protein Serine-Threonine Kinases/physiology , Acute Disease , Animals , Cell Adhesion/genetics , Cell Adhesion/immunology , Cells, Cultured , HL-60 Cells , Humans , Inflammation/immunology , Lymphocyte Function-Associated Antigen-1/genetics , Lymphocyte Function-Associated Antigen-1/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Protein Serine-Threonine Kinases/genetics
20.
Blood ; 119(21): 4981-91, 2012 May 24.
Article in English | MEDLINE | ID: mdl-22411867

ABSTRACT

Dasatinib is a tyrosine kinase inhibitor used to treat imatinib-resistant chronic myeloid leukemia and Philadelphia chromosome-positive acute lymphoblastic leukemia. At present, little is known about how dasatinib influences nonmalignant cells. In the present study, we tested the effect of dasatinib on functional responses of normal mature human neutrophils. Dasatinib completely blocked integrin- and Fc-receptor-mediated neutrophil functions, with the lowest IC(50) values below 10nM under serum-free conditions. Dasatinib caused a partial inhibition of neutrophil responses triggered by G-protein-coupled receptors and had a moderate effect on neutrophil responses triggered by microbial compounds. Whereas dasatinib inhibited neutrophil chemotaxis under static conditions in 2 dimensions, it did not affect migration under flow conditions or in 3-dimensional environments. Dasatinib did not have any major effect on phagocytosis or killing of bacteria by neutrophils. Adhesion of human neutrophils in the presence of whole serum was significantly inhibited by 50-100nM dasatinib, which corresponds to the reported serum concentrations in dasatinib-treated patients. Finally, ex vivo adhesion of mouse peripheral blood neutrophils was strongly reduced after oral administration of 5 mg/kg of dasatinib. Those results suggest that dasatinib treatment may affect the proinflammatory functions of mature neutrophils and raise the possibility that dasatinib-related compounds may provide clinical benefit in neutrophil-mediated inflammatory diseases.


Subject(s)
Inflammation/prevention & control , Neutrophils/drug effects , Pyrimidines/pharmacology , Thiazoles/pharmacology , Animals , Anti-Inflammatory Agents/pharmacology , Antineoplastic Agents/pharmacology , Cell Adhesion/drug effects , Cell Adhesion/immunology , Cell Respiration/drug effects , Cells, Cultured , Dasatinib , Down-Regulation/drug effects , Drug Evaluation, Preclinical , Humans , Inflammation/metabolism , Inflammation Mediators/antagonists & inhibitors , Inflammation Mediators/metabolism , Inflammation Mediators/physiology , Mice , Mice, Inbred C57BL , Neutrophil Activation/drug effects , Neutrophil Activation/physiology , Neutrophils/immunology , Neutrophils/metabolism , Neutrophils/physiology , Respiratory Burst/drug effects , Respiratory Burst/physiology
SELECTION OF CITATIONS
SEARCH DETAIL