Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 99
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 181(5): 1080-1096.e19, 2020 05 28.
Article in English | MEDLINE | ID: mdl-32380006

ABSTRACT

Environmental signals shape host physiology and fitness. Microbiota-derived cues are required to program conventional dendritic cells (cDCs) during the steady state so that they can promptly respond and initiate adaptive immune responses when encountering pathogens. However, the molecular underpinnings of microbiota-guided instructive programs are not well understood. Here, we report that the indigenous microbiota controls constitutive production of type I interferons (IFN-I) by plasmacytoid DCs. Using genome-wide analysis of transcriptional and epigenetic regulomes of cDCs from germ-free and IFN-I receptor (IFNAR)-deficient mice, we found that tonic IFNAR signaling instructs a specific epigenomic and metabolic basal state that poises cDCs for future pathogen combat. However, such beneficial biological function comes with a trade-off. Instructed cDCs can prime T cell responses against harmless peripheral antigens when removing roadblocks of peripheral tolerance. Our data provide fresh insights into the evolutionary trade-offs that come with successful adaptation of vertebrates to their microbial environment.


Subject(s)
Dendritic Cells/immunology , Interferon Type I/immunology , Microbiota/immunology , Adaptive Immunity/immunology , Adaptive Immunity/physiology , Animals , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/microbiology , Female , Male , Mice , Mice, Inbred C57BL , Microbiota/physiology , Receptor, Interferon alpha-beta/metabolism , Signal Transduction/immunology
2.
Cell ; 183(5): 1282-1297.e18, 2020 11 25.
Article in English | MEDLINE | ID: mdl-33098771

ABSTRACT

Classically considered short-lived and purely defensive leukocytes, neutrophils are unique in their fast and moldable response to stimulation. This plastic behavior may underlie variable and even antagonistic functions during inflammation or cancer, yet the full spectrum of neutrophil properties as they enter healthy tissues remains unexplored. Using a new model to track neutrophil fates, we found short but variable lifetimes across multiple tissues. Through analysis of the receptor, transcriptional, and chromatin accessibility landscapes, we identify varying neutrophil states and assign non-canonical functions, including vascular repair and hematopoietic homeostasis. Accordingly, depletion of neutrophils compromised angiogenesis during early age, genotoxic injury, and viral infection, and impaired hematopoietic recovery after irradiation. Neutrophils acquired these properties in target tissues, a process that, in the lungs, occurred in CXCL12-rich areas and relied on CXCR4. Our results reveal that tissues co-opt neutrophils en route for elimination to induce programs that support their physiological demands.


Subject(s)
Cell Lineage , Neutrophils/metabolism , Organ Specificity , Animals , Chromatin/metabolism , Female , Hematopoiesis , Intestines/blood supply , Lung/blood supply , Male , Mice, Inbred C57BL , Neovascularization, Physiologic , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism , Receptors, CXCR4/metabolism , Single-Cell Analysis , Transcription, Genetic , Transcriptome/genetics
3.
Circ Res ; 134(10): e93-e111, 2024 May 10.
Article in English | MEDLINE | ID: mdl-38563147

ABSTRACT

BACKGROUND: Endothelial activation promotes the release of procoagulant extracellular vesicles and inflammatory mediators from specialized storage granules. Endothelial membrane exocytosis is controlled by phosphorylation. We hypothesized that the absence of PTP1B (protein tyrosine phosphatase 1B) in endothelial cells promotes venous thromboinflammation by triggering endothelial membrane fusion and exocytosis. METHODS: Mice with inducible endothelial deletion of PTP1B (End.PTP1B-KO) underwent inferior vena cava ligation to induce stenosis and venous thrombosis. Primary endothelial cells from transgenic mice and human umbilical vein endothelial cells were used for mechanistic studies. RESULTS: Vascular ultrasound and histology showed significantly larger venous thrombi containing higher numbers of Ly6G (lymphocyte antigen 6 family member G)-positive neutrophils in mice with endothelial PTP1B deletion, and intravital microscopy confirmed the more pronounced neutrophil recruitment following inferior vena cava ligation. RT2 PCR profiler array and immunocytochemistry analysis revealed increased endothelial activation and adhesion molecule expression in primary End.PTP1B-KO endothelial cells, including CD62P (P-selectin) and VWF (von Willebrand factor). Pretreatment with the NF-κB (nuclear factor kappa B) kinase inhibitor BAY11-7082, antibodies neutralizing CD162 (P-selectin glycoprotein ligand-1) or VWF, or arginylglycylaspartic acid integrin-blocking peptides abolished the neutrophil adhesion to End.PTP1B-KO endothelial cells in vitro. Circulating levels of annexin V+ procoagulant endothelial CD62E+ (E-selectin) and neutrophil (Ly6G+) extracellular vesicles were also elevated in End.PTP1B-KO mice after inferior vena cava ligation. Higher plasma MPO (myeloperoxidase) and Cit-H3 (citrullinated histone-3) levels and neutrophil elastase activity indicated neutrophil activation and extracellular trap formation. Infusion of End.PTP1B-KO extracellular vesicles into C57BL/6J wild-type mice most prominently enhanced the recruitment of endogenous neutrophils, and this response was blunted in VWF-deficient mice or by VWF-blocking antibodies. Reduced PTP1B binding and tyrosine dephosphorylation of SNAP23 (synaptosome-associated protein 23) resulting in increased VWF exocytosis and neutrophil adhesion were identified as mechanisms, all of which could be restored by NF-κB kinase inhibition using BAY11-7082. CONCLUSIONS: Our findings show that endothelial PTP1B deletion promotes venous thromboinflammation by enhancing SNAP23 phosphorylation, endothelial VWF exocytosis, and neutrophil recruitment.


Subject(s)
Exocytosis , Mice, Knockout , Protein Tyrosine Phosphatase, Non-Receptor Type 1 , Venous Thrombosis , von Willebrand Factor , Animals , Protein Tyrosine Phosphatase, Non-Receptor Type 1/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 1/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 1/deficiency , Humans , Mice , von Willebrand Factor/metabolism , von Willebrand Factor/genetics , Venous Thrombosis/metabolism , Venous Thrombosis/genetics , Venous Thrombosis/pathology , Human Umbilical Vein Endothelial Cells/metabolism , Inflammation/metabolism , Inflammation/genetics , Mice, Inbred C57BL , Neutrophils/metabolism , Endothelial Cells/metabolism , Cells, Cultured , Vena Cava, Inferior/metabolism , Vena Cava, Inferior/pathology , Male , Neutrophil Infiltration , NF-kappa B/metabolism
4.
Proc Natl Acad Sci U S A ; 120(40): e2215421120, 2023 10 03.
Article in English | MEDLINE | ID: mdl-37756334

ABSTRACT

Externalized histones erupt from the nucleus as extracellular traps, are associated with several acute and chronic lung disorders, but their implications in the molecular pathogenesis of interstitial lung disease are incompletely defined. To investigate the role and molecular mechanisms of externalized histones within the immunologic networks of pulmonary fibrosis, we studied externalized histones in human and animal bronchoalveolar lavage (BAL) samples of lung fibrosis. Neutralizing anti-histone antibodies were administered in bleomycin-induced fibrosis of C57BL/6 J mice, and subsequent studies used conditional/constitutive knockout mouse strains for TGFß and IL-27 signaling along with isolated platelets and cultured macrophages. We found that externalized histones (citH3) were significantly (P < 0.01) increased in cell-free BAL fluids of patients with idiopathic pulmonary fibrosis (IPF; n = 29) as compared to healthy controls (n = 10). The pulmonary sources of externalized histones were Ly6G+CD11b+ neutrophils and nonhematopoietic cells after bleomycin in mice. Neutralizing monoclonal anti-histone H2A/H4 antibodies reduced the pulmonary collagen accumulation and hydroxyproline concentration. Histones activated platelets to release TGFß1, which signaled through the TGFbRI/TGFbRII receptor complex on LysM+ cells to antagonize macrophage-derived IL-27 production. TGFß1 evoked multiple downstream mechanisms in macrophages, including p38 MAPK, tristetraprolin, IL-10, and binding of SMAD3 to the IL-27 promotor regions. IL-27RA-deficient mice displayed more severe collagen depositions suggesting that intact IL-27 signaling limits fibrosis. In conclusion, externalized histones inactivate a safety switch of antifibrotic, macrophage-derived IL-27 by boosting platelet-derived TGFß1. Externalized histones are accessible to neutralizing antibodies for improving the severity of experimental pulmonary fibrosis.


Subject(s)
Idiopathic Pulmonary Fibrosis , Interleukin-27 , Humans , Mice , Animals , Mice, Inbred C57BL , Histones , Blood Platelets , Idiopathic Pulmonary Fibrosis/chemically induced , Idiopathic Pulmonary Fibrosis/genetics
5.
Eur J Immunol ; 53(5): e2250339, 2023 05.
Article in English | MEDLINE | ID: mdl-36959687

ABSTRACT

Polyphosphates are highly conserved, linear polymers of monophosphates that reside in all living cells. Bacteria produce long chains containing hundreds to thousands of phosphate units, which can interfere with host defense to infection. Here, we report that intratracheal long-chain polyphosphate administration to C57BL/6J mice resulted in the release of proinflammatory cytokines and influx of Ly6G+ polymorphonuclear neutrophils in the bronchoalveolar lavage fluid causing a disruption of the physiologic endothelial-epithelial small airway barrier and histologic signs of lung injury. Polyphosphate-induced effects were attenuated after neutrophil depletion in mice. In isolated murine neutrophils, long-chain polyphosphates modulated cytokine release induced by lipopolysaccharides (LPS) from Gram-negative bacteria or lipoteichoic acid from Gram-positive bacteria. In addition, long-chain polyphosphates induced immune evasive effects in human neutrophils. In detail, long-chain polyphosphates downregulated CD11b and curtailed the phagocytosis of Escherichia coli particles by neutrophils. Polyphosphates modulated the migration capacity by inducing CD62L shedding resulting in CD62Llow and CD11blow neutrophils. The release of IL-8 induced by LPS was also significantly reduced. Pharmacologic blockade of PI3K with wortmannin antagonized long-chain polyphosphate-induced effects on LPS-induced IL-8 release. In conclusion, polyphosphates govern immunomodulation in murine and human neutrophils, suggesting polyphosphates as a therapeutic target for bacterial infections to restore innate immune defense.


Subject(s)
Lipopolysaccharides , Neutrophils , Humans , Mice , Animals , Lipopolysaccharides/pharmacology , Polyphosphates/pharmacology , Interleukin-8 , Mice, Inbred C57BL , Cytokines , Bronchoalveolar Lavage Fluid , Escherichia coli , Immunomodulation , Lung
6.
J Immunol ; 208(8): 2008-2018, 2022 04 15.
Article in English | MEDLINE | ID: mdl-35354611

ABSTRACT

IL-27 is a heterodimeric IL-12 family cytokine formed by noncovalent association of the promiscuous EBI3 subunit and selective p28 subunit. IL-27 is produced by mononuclear phagocytes and unfolds pleiotropic immune-modulatory functions through ligation to IL-27 receptor α (IL-27RA). Although IL-27 is known to contribute to immunity and to limit inflammation after various infections, its relevance for host defense against multicellular parasites is still poorly defined. Here, we investigated the role of IL-27 during infection with the soil-transmitted hookworm, Nippostrongylus brasiliensis, in its early host intrapulmonary life cycle. IL-27(p28) was detectable in bronchoalveolar lavage fluid of C57BL/6J wild-type mice on day 1 after s.c. inoculation. IL-27RA expression was most abundant on lung-invading γδ T cells. Il27ra-/- mice showed increased lung parasite burden together with aggravated pulmonary hemorrhage and higher alveolar total protein leakage as a surrogate for epithelial-vascular barrier disruption. Conversely, injections of recombinant mouse (rm)IL-27 into wild-type mice reduced lung injury and parasite burden. In multiplex screens, higher airway accumulations of IL-6, TNF-α, and MCP-3 (CCL7) were observed in Il27ra-/- mice, whereas rmIL-27 treatment showed a reciprocal effect. Importantly, γδ T cell numbers in airways were enhanced by endogenous or administered IL-27. Further analysis revealed a direct antihelminthic function of IL-27 on γδ T cells as adoptive intratracheal transfer of rmIL-27-treated γδ T cells during primary N. brasiliensis lung infection conferred protection in mice. In summary, this report demonstrates protective functions of IL-27 to control the early lung larval stage of hookworm infection.


Subject(s)
Hookworm Infections , Interleukin-27 , Animals , Interleukins , Lung , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Antigen, T-Cell, gamma-delta
7.
Amino Acids ; 54(10): 1339-1356, 2022 Oct.
Article in English | MEDLINE | ID: mdl-35451695

ABSTRACT

The essential amino acid tryptophan (Trp) is metabolized by gut commensals, yielding in compounds that affect innate immune cell functions directly, but also acting on the aryl hydrocarbon receptor (AHR), thus regulating the maintenance of group 3 innate lymphoid cells (ILCs), promoting T helper 17 (TH17) cell differentiation, and interleukin-22 production. In addition, microbiota-derived Trp metabolites have direct effects on the vascular endothelium, thus influencing the development of vascular inflammatory phenotypes. Indoxyl sulfate was demonstrated to promote vascular inflammation, whereas indole-3-propionic acid and indole-3-aldehyde had protective roles. Furthermore, there is increasing evidence for a contributory role of microbiota-derived indole-derivatives in blood pressure regulation and hypertension. Interestingly, there are indications for a role of the kynurenine pathway in atherosclerotic lesion development. Here, we provide an overview on the emerging role of gut commensals in the modulation of Trp metabolism and its influence in cardiovascular disease development.


Subject(s)
Cardiovascular Diseases , Microbiota , Humans , Tryptophan/metabolism , Immunity, Innate , Lymphocytes , Indoles/metabolism , Inflammation
8.
Int J Mol Sci ; 23(12)2022 Jun 09.
Article in English | MEDLINE | ID: mdl-35742876

ABSTRACT

The biocompatibility of carrier nanomaterials in blood is largely hampered by their activating or inhibiting role on the clotting system, which in many cases prevents safe intravascular application. Here, we characterized an aqueous colloidal ethyl hydroxyethyl cellulose (EHEC) solution and tested its effect on ex vivo clot formation, platelet aggregation, and activation by thromboelastometry, aggregometry, and flow cytometry. We compared the impact of EHEC solution on platelet aggregation with biocompatible materials used in transfusion medicine (the plasma expanders gelatin polysuccinate and hydroxyethyl starch). We demonstrate that the EHEC solution, in contrast to commercial products exhibiting Newtonian flow behavior, resembles the shear-thinning behavior of human blood. Similar to established nanomaterials that are considered biocompatible when added to blood, the EHEC exposure of resting platelets in platelet-rich plasma does not enhance tissue thromboplastin- or ellagic acid-induced blood clotting, or platelet aggregation or activation, as measured by integrin αIIbß3 activation and P-selectin exposure. Furthermore, the addition of EHEC solution to adenosine diphosphate (ADP)-stimulated platelet-rich plasma does not affect the platelet aggregation induced by this agonist. Overall, our results suggest that EHEC may be suitable as a biocompatible carrier material in blood circulation and for applications in flow-dependent diagnostics.


Subject(s)
Platelet Aggregation , Polymers , Adenosine Diphosphate/pharmacology , Blood Platelets , Cellulose/pharmacology , Humans , Platelet Function Tests/methods , Polymers/pharmacology
9.
Blood ; 134(14): 1119-1131, 2019 10 03.
Article in English | MEDLINE | ID: mdl-31434703

ABSTRACT

Antiphospholipid antibodies (aPLs) with complex lipid and/or protein reactivities cause complement-dependent thrombosis and pregnancy complications. Although cross-reactivities with coagulation regulatory proteins contribute to the risk for developing thrombosis in patients with antiphospholipid syndrome, the majority of pathogenic aPLs retain reactivity with membrane lipid components and rapidly induce reactive oxygen species-dependent proinflammatory signaling and tissue factor (TF) procoagulant activation. Here, we show that lipid-reactive aPLs activate a common species-conserved TF signaling pathway. aPLs dissociate an inhibited TF coagulation initiation complex on the cell surface of monocytes, thereby liberating factor Xa for thrombin generation and protease activated receptor 1/2 heterodimer signaling. In addition to proteolytic signaling, aPLs promote complement- and protein disulfide isomerase-dependent TF-integrin ß1 trafficking that translocates aPLs and NADPH oxidase to the endosome. Cell surface TF pathway inhibitor (TFPI) synthesized by monocytes is required for TF inhibition, and disabling TFPI prevents aPL signaling, indicating a paradoxical prothrombotic role for TFPI. Myeloid cell-specific TFPI inactivation has no effect on models of arterial or venous thrombus development, but remarkably prevents experimental aPL-induced thrombosis in mice. Thus, the physiological control of TF primes monocytes for rapid aPL pathogenic signaling and thrombosis amplification in an unexpected crosstalk between complement activation and coagulation signaling.


Subject(s)
Antibodies, Antiphospholipid/immunology , Monocytes/immunology , Thromboplastin/immunology , Thrombosis/immunology , Animals , Blood Coagulation , Cells, Cultured , Female , Humans , Lipoproteins/immunology , Male , Mice, Inbred C57BL , Monocytes/pathology , Signal Transduction , Thrombosis/blood , Thrombosis/pathology
10.
Arterioscler Thromb Vasc Biol ; 40(9): 2279-2292, 2020 09.
Article in English | MEDLINE | ID: mdl-32611241

ABSTRACT

OBJECTIVE: Recruitment of neutrophils and formation of neutrophil extracellular traps (NETs) contribute to lethality in acute mesenteric infarction. To study the impact of the gut microbiota in acute mesenteric infarction, we used gnotobiotic mouse models to investigate whether gut commensals prime the reactivity of neutrophils towards formation of neutrophil extracellular traps (NETosis). Approach and Results: We applied a mesenteric ischemia-reperfusion (I/R) injury model to germ-free (GF) and colonized C57BL/6J mice. By intravital imaging, we quantified leukocyte adherence and NET formation in I/R-injured mesenteric venules. Colonization with gut microbiota or monocolonization with Escherichia coli augmented the adhesion of leukocytes, which was dependent on the TLR4 (Toll-like receptor-4)/TRIF (TIR-domain-containing adapter-inducing interferon-ß) pathway. Although neutrophil accumulation was decreased in I/R-injured venules of GF mice, NETosis following I/R injury was significantly enhanced compared with conventionally raised mice or mice colonized with the minimal microbial consortium altered Schaedler flora. Also ex vivo, neutrophils from GF and antibiotic-treated mice showed increased LPS (lipopolysaccharide)-induced NETosis. Enhanced TLR4 signaling in GF neutrophils was due to elevated TLR4 expression and augmented IRF3 (interferon regulatory factor-3) phosphorylation. Likewise, neutrophils from antibiotic-treated conventionally raised mice had increased NET formation before and after ischemia. Increased NETosis in I/R injury was abolished in conventionally raised mice deficient in the TLR adaptor TRIF. In support of the desensitizing influence of enteric LPS, treatment of GF mice with LPS via drinking water diminished LPS-induced NETosis in vitro and in the mesenteric I/R injury model. CONCLUSIONS: Collectively, our results identified that the gut microbiota suppresses NETing neutrophil hyperreactivity in mesenteric I/R injury, while ensuring immunovigilance by enhancing neutrophil recruitment.


Subject(s)
Extracellular Traps/metabolism , Gastrointestinal Microbiome , Mesenteric Ischemia/metabolism , Mesentery/blood supply , Neutrophil Infiltration , Neutrophils/metabolism , Reperfusion Injury/metabolism , Venules/metabolism , Animals , Bacillus subtilis/pathogenicity , Cell Adhesion , Cells, Cultured , Disease Models, Animal , Escherichia coli/pathogenicity , Extracellular Traps/microbiology , Female , Germ-Free Life , Host-Pathogen Interactions , Leukocyte Rolling , Leukocytes/metabolism , Leukocytes/microbiology , Male , Mesenteric Ischemia/microbiology , Mesenteric Ischemia/pathology , Mice, Inbred C57BL , Mice, Knockout , Reperfusion Injury/microbiology , Reperfusion Injury/pathology , Signal Transduction , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism , Venules/microbiology , Venules/pathology
11.
Subcell Biochem ; 94: 437-464, 2020.
Article in English | MEDLINE | ID: mdl-32189311

ABSTRACT

Von Willebrand factor (VWF) and coagulation factor VIII (FVIII) circulate as a complex in plasma and have a major role in the hemostatic system. VWF has a dual role in hemostasis. It promotes platelet adhesion by anchoring the platelets to the subendothelial matrix of damaged vessels and it protects FVIII from proteolytic degradation. Moreover, VWF is an acute phase protein that has multiple roles in vascular inflammation and is massively secreted from Weibel-Palade bodies upon endothelial cell activation. Activated FVIII on the other hand, together with coagulation factor IX forms the tenase complex, an essential feature of the propagation phase of coagulation on the surface of activated platelets. VWF deficiency, either quantitative or qualitative, results in von Willebrand disease (VWD), the most common bleeding disorder. The deficiency of FVIII is responsible for Hemophilia A, an X-linked bleeding disorder. Here, we provide an overview on the role of the VWF-FVIII interaction in vascular physiology.


Subject(s)
Cardiovascular Physiological Phenomena , Factor VIII/metabolism , von Willebrand Factor/metabolism , Factor VIII/chemistry , Hemophilia A/metabolism , Humans , von Willebrand Diseases/metabolism , von Willebrand Factor/chemistry
12.
Proc Natl Acad Sci U S A ; 115(27): 7087-7092, 2018 07 03.
Article in English | MEDLINE | ID: mdl-29925597

ABSTRACT

Airborne fungal pathogens, predominantly Aspergillus fumigatus, can cause severe respiratory tract diseases. Here we show that in environments, fungal spores can already be decorated with nanoparticles. Using representative controlled nanoparticle models, we demonstrate that various nanoparticles, but not microparticles, rapidly and stably associate with spores, without specific functionalization. Nanoparticle-spore complex formation was enhanced by small nanoparticle size rather than by material, charge, or "stealth" modifications and was concentration-dependently reduced by the formation of environmental or physiological biomolecule coronas. Assembly of nanoparticle-spore surface hybrid structures affected their pathobiology, including reduced sensitivity against defensins, uptake into phagocytes, lung cell toxicity, and TLR/cytokine-mediated inflammatory responses. Following infection of mice, nanoparticle-spore complexes were detectable in the lung and less efficiently eliminated by the pulmonary immune defense, thereby enhancing A. fumigatus infections in immunocompromised animals. Collectively, self-assembly of nanoparticle-fungal complexes affects their (patho)biological identity, which may impact human health and ecology.


Subject(s)
Aspergillus fumigatus/immunology , Cytokines/immunology , Lung/immunology , Nanoparticles , Pulmonary Aspergillosis/immunology , Spores, Fungal/immunology , A549 Cells , Animals , Humans , Lung/pathology , Mice , Protein Corona/immunology , Pulmonary Aspergillosis/pathology , THP-1 Cells
14.
Platelets ; 31(8): 981-988, 2020 Nov 16.
Article in English | MEDLINE | ID: mdl-31814487

ABSTRACT

The exploration of thrombotic mechanisms relies on the application of blood collection methods from laboratory mice with a minimal pre-activation of platelets and the clotting system. So far, very little is known on how the blood collection method and the anticoagulant used influence pre-activation of mouse platelets and coagulation. To determine the most suitable blood collection method, we systematically compared blood collection by heart puncture, Vena cava puncture, and puncture of the retro-orbital vein plexus and the use of citrate, heparin, and EDTA as frequently used anticoagulants with regard to platelet activation and whole blood clotting parameters. The activation of platelet-rich plasma diluted in Tyrode's buffer was analyzed by flow cytometry, analyzing the exposure of P-selectin and activated integrin αIIbß3. Clotting of whole blood was profiled by thrombelastometry. Puncture of the retro-orbital vein plexus by plastic capillaries is not superior in terms of blood volume and platelet pre-activation, whereas heart puncture and Vena cava puncture resulted in similarly high blood volumes. Cardiac puncture and Vena cava puncture did not result in pre-activated platelets with citrate as an anticoagulant, but the use of EDTA resulted in increased levels of integrin αIIbß3 activation. Puncture of the retro-orbital vein plexus by plastic capillaries resulted in increased platelet integrin αIIbß3 activation, which could be prevented by soaking with citrate or coating with heparin. Further, activation of coagulation in citrated whole blood by puncture of the retro-orbital vein plexus using a blunt plastic capillary was observed by thromboelastometry. The use of citrate is the optimal anticoagulant in mouse platelet assays. Blood collections from the heart or Vena cava represent reliable alternatives to retro-orbital puncture of the vein plexus to avoid pre-activation of platelets and coagulation.


Subject(s)
Anticoagulants/therapeutic use , Blood Coagulation Tests/methods , Platelet Function Tests/methods , Animals , Anticoagulants/pharmacology , Humans , Mice , Mice, Inbred C57BL
15.
Int J Mol Sci ; 21(19)2020 Sep 28.
Article in English | MEDLINE | ID: mdl-32998468

ABSTRACT

The commensal microbiota is a recognized enhancer of arterial thrombus growth. While several studies have demonstrated the prothrombotic role of the gut microbiota, the molecular mechanisms promoting arterial thrombus growth are still under debate. Here, we demonstrate that germ-free (GF) mice, which from birth lack colonization with a gut microbiota, show diminished static deposition of washed platelets to type I collagen compared with their conventionally raised (CONV-R) counterparts. Flow cytometry experiments revealed that platelets from GF mice show diminished activation of the integrin αIIbß3 (glycoprotein IIbIIIa) when activated by the platelet agonist adenosine diphosphate (ADP). Furthermore, washed platelets from Toll-like receptor-2 (Tlr2)-deficient mice likewise showed impaired static deposition to the subendothelial matrix component type I collagen compared with wild-type (WT) controls, a process that was unaffected by GPIbα-blockade but influenced by von Willebrand factor (VWF) plasma levels. Collectively, our results indicate that microbiota-triggered steady-state activation of innate immune pathways via TLR2 enhances platelet deposition to subendothelial matrix molecules. Our results link host colonization status with the ADP-triggered activation of integrin αIIbß3, a pathway promoting platelet deposition to the growing thrombus.


Subject(s)
Adenosine Diphosphate/pharmacology , Blood Platelets/drug effects , Collagen Type I/genetics , Platelet Glycoprotein GPIIb-IIIa Complex/genetics , Thrombosis/microbiology , von Willebrand Factor/genetics , Animals , Arteries/metabolism , Arteries/pathology , Blood Platelets/immunology , Blood Platelets/pathology , Cell Adhesion/drug effects , Collagen Type I/immunology , Female , Gastrointestinal Microbiome/immunology , Gene Expression , Germ-Free Life , Humans , Immunity, Innate , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Platelet Glycoprotein GPIIb-IIIa Complex/agonists , Platelet Glycoprotein GPIIb-IIIa Complex/immunology , Platelet Glycoprotein GPIb-IX Complex/genetics , Platelet Glycoprotein GPIb-IX Complex/immunology , Primary Cell Culture , Symbiosis/immunology , Thrombosis/genetics , Thrombosis/immunology , Thrombosis/pathology , Toll-Like Receptor 2/deficiency , Toll-Like Receptor 2/genetics , Toll-Like Receptor 2/immunology , von Willebrand Factor/immunology
16.
Eur J Immunol ; 48(4): 564-575, 2018 04.
Article in English | MEDLINE | ID: mdl-29230812

ABSTRACT

Commensal gut microbiota have recently been implicated in cardiovascular disease (CVD) and cerebrovascular disease. Atherosclerotic plaque formation depends on the colonization status of the host. In addition to host nutrition and the related microbiota-dependent metabolic changes, activation of innate immune pathways triggers the development of atherosclerosis and supports arterial thrombosis. Gnotobiotic mouse models have uncovered that activation of Toll-like receptor-2 by gut microbial ligands supports von Willebrand factor-integrin mediated platelet deposition to the site of vascular injury. Depending on nutritional factors, the microbiota-derived choline-metabolite trimethylamine N-oxide (TMAO) increases atherosclerotic plaque size, triggers prothrombotic platelet function and promotes arterial thrombus growth. Hence, the composition of the commensal microbiota is an emerging risk factor for CVD. Here, we provide an overview on microbiota-dependent pathomechanisms that drive the development of CVD and arterial thrombosis.


Subject(s)
Atherosclerosis/pathology , Cardiovascular Diseases/etiology , Cerebrovascular Disorders/etiology , Gastrointestinal Microbiome/physiology , Plaque, Atherosclerotic/pathology , Thrombosis/pathology , Animals , Atherosclerosis/immunology , Blood Platelets/metabolism , Humans , Immunity, Innate/immunology , Infant , Infant, Newborn , Integrins/immunology , Methylamines/metabolism , Mice , Plaque, Atherosclerotic/microbiology , Risk Factors , Symbiosis , Thrombosis/microbiology , Toll-Like Receptor 2/metabolism , von Willebrand Factor/immunology
17.
Blood ; 129(16): 2291-2302, 2017 04 20.
Article in English | MEDLINE | ID: mdl-28223279

ABSTRACT

Expanding evidence indicates multiple interactions between the hemostatic system and innate immunity, and the coagulation and complement cascades. Here we show in a tissue factor (TF)-dependent model of flow restriction-induced venous thrombosis that complement factors make distinct contributions to platelet activation and fibrin deposition. Complement factor 3 (C3) deficiency causes prolonged bleeding, reduced thrombus incidence, thrombus size, fibrin and platelet deposition in the ligated inferior vena cava, and diminished platelet activation in vitro. Initial fibrin deposition at the vessel wall over 6 hours in this model was dependent on protein disulfide isomerase (PDI) and TF expression by myeloid cells, but did not require neutrophil extracellular trap formation involving peptidyl arginine deiminase 4. In contrast to C3-/- mice, C5-deficient mice had no apparent defect in platelet activation in vitro, and vessel wall platelet deposition and initial hemostasis in vivo. However, fibrin formation, the exposure of negatively charged phosphatidylserine (PS) on adherent leukocytes, and clot burden after 48 hours were significantly reduced in C5-/- mice compared with wild-type controls. These results delineate that C3 plays specific roles in platelet activation independent of formation of the terminal complement complex and provide in vivo evidence for contributions of complement-dependent membrane perturbations to prothrombotic TF activation on myeloid cells.


Subject(s)
Blood Platelets/immunology , Complement C3/genetics , Complement C5/genetics , Hemostasis/immunology , Thrombosis/immunology , Vena Cava, Inferior/immunology , Animals , Blood Platelets/pathology , Complement Activation , Complement C3/metabolism , Complement C5/metabolism , Extracellular Traps/genetics , Extracellular Traps/immunology , Fibrin/genetics , Fibrin/immunology , Gene Expression , Humans , Hydrolases/genetics , Hydrolases/immunology , Immunity, Innate , Leukocytes/immunology , Leukocytes/pathology , Ligation , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Cells/immunology , Myeloid Cells/pathology , Phosphatidylserines/metabolism , Platelet Activation/immunology , Protein Disulfide-Isomerases/genetics , Protein Disulfide-Isomerases/immunology , Protein-Arginine Deiminase Type 4 , Thromboplastin/genetics , Thromboplastin/immunology , Thrombosis/blood , Thrombosis/genetics , Thrombosis/pathology , Vena Cava, Inferior/metabolism , Vena Cava, Inferior/pathology
18.
Blood ; 130(4): 542-553, 2017 07 27.
Article in English | MEDLINE | ID: mdl-28572286

ABSTRACT

The symbiotic gut microbiota play pivotal roles in host physiology and the development of cardiovascular diseases, but the microbiota-triggered pattern recognition signaling mechanisms that impact thrombosis are poorly defined. In this article, we show that germ-free (GF) and Toll-like receptor-2 (Tlr2)-deficient mice have reduced thrombus growth after carotid artery injury relative to conventionally raised controls. GF Tlr2-/- and wild-type (WT) mice were indistinguishable, but colonization with microbiota restored a significant difference in thrombus growth between the genotypes. We identify reduced plasma levels of von Willebrand factor (VWF) and reduced VWF synthesis, specifically in hepatic endothelial cells, as a critical factor that is regulated by gut microbiota and determines thrombus growth in Tlr2-/- mice. Static platelet aggregate formation on extracellular matrix was similarly reduced in GF WT, Tlr2-/- , and heterozygous Vwf+/- mice that are all characterized by a modest reduction in plasma VWF levels. Defective platelet matrix interaction can be restored by exposure to WT plasma or to purified VWF depending on the VWF integrin binding site. Moreover, administration of VWF rescues defective thrombus growth in Tlr2-/- mice in vivo. These experiments delineate an unexpected pathway in which microbiota-triggered TLR2 signaling alters the synthesis of proadhesive VWF by the liver endothelium and favors platelet integrin-dependent thrombus growth.


Subject(s)
Gastrointestinal Microbiome , Liver/metabolism , Signal Transduction , Thrombosis/metabolism , Toll-Like Receptor 2/metabolism , von Willebrand Factor/biosynthesis , Animals , Blood Platelets/metabolism , Blood Platelets/pathology , Germ-Free Life , Liver/pathology , Mice , Mice, Knockout , Platelet Aggregation/genetics , Thrombosis/genetics , Thrombosis/pathology , Toll-Like Receptor 2/genetics , von Willebrand Factor/genetics
19.
Opt Express ; 27(18): 25731-25748, 2019 Sep 02.
Article in English | MEDLINE | ID: mdl-31510440

ABSTRACT

We introduce a passively-aligned, flexure-tuned cavity optomechanical system in which a membrane is positioned microns from one end mirror of a Fabry-Perot optical cavity. By displacing the membrane through gentle flexure of its silicon supporting frame (i.e., to ∼80 m radius of curvature (ROC)), we gain access to the full range of available optomechanical couplings, finding also that the optical spectrum exhibits none of the abrupt discontinuities normally found in "membrane-in-the-middle" (MIM) systems. More aggressive flexure (3 m ROC) enables >15 µm membrane travel, milliradian tilt tuning, and a wavelength-scale (1.64 ± 0.78 µm) membrane-mirror separation. We also provide a complete set of analytical expressions for this system's leading-order dispersive and dissipative optomechanical couplings. Notably, this system can potentially generate orders of magnitude larger linear dissipative or quadratic dispersive strong coupling parameters than is possible with a MIM system. Additionally, it can generate the same purely quadratic dispersive coupling as a MIM system, but with significantly suppressed linear dissipative back-action (and force noise).

20.
Arterioscler Thromb Vasc Biol ; 38(10): 2318-2326, 2018 10.
Article in English | MEDLINE | ID: mdl-29903735

ABSTRACT

Objective- To investigate the effect of gut microbiota and diet on atherogenesis. Approach and Results- Here, we investigated the interaction between the gut microbiota and diet on atherosclerosis by feeding germ-free or conventionally raised Apoe-/- mice chow or Western diet alone or supplemented with choline (which is metabolized by the gut microbiota and host enzymes to trimethylamine N-oxide) for 12 weeks. We observed smaller aortic lesions and lower plasma cholesterol levels in conventionally raised mice compared with germ-free mice on a chow diet; these differences were not observed in mice on a Western diet. Choline supplementation increased plasma trimethylamine N-oxide levels in conventionally raised mice but not in germ-free mice. However, this treatment did not affect the size of aortic lesions or plasma cholesterol levels. Gut microbiota composition was analyzed by sequencing of 16S rRNA genes. As expected, the global community structure and relative abundance of many taxa differed between mice fed chow or a Western diet. Choline supplementation had minor effects on the community structure although the relative abundance of some taxa belonging to Clostridiales was altered. Conclusions- In conclusion, the impact of the gut microbiota on atherosclerosis is dietary dependent and is associated with plasma cholesterol levels. Furthermore, the microbiota was required for trimethylamine N-oxide production from dietary choline, but this process could not be linked to increased atherosclerosis in this model.


Subject(s)
Aortic Diseases/microbiology , Atherosclerosis/microbiology , Bacteria/metabolism , Choline/administration & dosage , Diet, Western , Dietary Supplements , Gastrointestinal Microbiome , Intestines/microbiology , Mice, Knockout, ApoE , Animal Feed , Animals , Aortic Diseases/blood , Aortic Diseases/genetics , Aortic Diseases/prevention & control , Atherosclerosis/blood , Atherosclerosis/genetics , Atherosclerosis/prevention & control , Bacteria/genetics , Bacteria/growth & development , Cholesterol/blood , Choline/metabolism , Disease Models, Animal , Male , Methylamines/metabolism , Mice, Inbred C57BL , Ribotyping
SELECTION OF CITATIONS
SEARCH DETAIL