Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Biol Direct ; 18(1): 10, 2023 03 15.
Article in English | MEDLINE | ID: mdl-36922848

ABSTRACT

In tumor therapy anti-angiogenic approaches have the potential to increase the efficacy of a wide variety of subsequently or co-administered agents, possibly by improving or normalizing the defective tumor vasculature. Successful implementation of the concept of vascular normalization under anti-angiogenic therapy, however, mandates a detailed understanding of key characteristics and a respective scoring metric that defines an improved vasculature and thus a successful attempt. Here, we show that beyond commonly used parameters such as vessel patency and maturation, anti-angiogenic approaches largely benefit if the complex vascular network with its vessel interconnections is both qualitatively and quantitatively assessed. To gain such deeper insight the organization of vascular networks, we introduce a multi-parametric evaluation of high-resolution angiographic images based on light-sheet fluorescence microscopy images of tumors. We first could pinpoint key correlations between vessel length, straightness and diameter to describe the regular, functional and organized structure observed under physiological conditions. We found that vascular networks from experimental tumors diverted from those in healthy organs, demonstrating the dysfunctionality of the tumor vasculature not only on the level of the individual vessel but also in terms of inadequate organization into larger structures. These parameters proofed effective in scoring the degree of disorganization in different tumor entities, and more importantly in grading a potential reversal under treatment with therapeutic agents. The presented vascular network analysis will support vascular normalization assessment and future optimization of anti-angiogenic therapy.


Subject(s)
Neoplasms , Neovascularization, Pathologic , Humans , Neovascularization, Pathologic/diagnostic imaging , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/pathology , Neoplasms/diagnostic imaging , Neoplasms/drug therapy , Neoplasms/pathology , Immunotherapy , Angiogenesis Inhibitors/pharmacology , Angiogenesis Inhibitors/therapeutic use
3.
PLoS Genet ; 16(11): e1009106, 2020 11.
Article in English | MEDLINE | ID: mdl-33151932

ABSTRACT

Hirschsprung disease (HSCR, OMIM 142623) involves congenital intestinal obstruction caused by dysfunction of neural crest cells and their progeny during enteric nervous system (ENS) development. HSCR is a multifactorial disorder; pathogenetic variants accounting for disease phenotype are identified only in a minority of cases, and the identification of novel disease-relevant genes remains challenging. In order to identify and to validate a potential disease-causing relevance of novel HSCR candidate genes, we established a complementary study approach, combining whole exome sequencing (WES) with transcriptome analysis of murine embryonic ENS-related tissues, literature and database searches, in silico network analyses, and functional readouts using candidate gene-specific genome-edited cell clones. WES datasets of two patients with HSCR and their non-affected parents were analysed, and four novel HSCR candidate genes could be identified: ATP7A, SREBF1, ABCD1 and PIAS2. Further rare variants in these genes were identified in additional HSCR patients, suggesting disease relevance. Transcriptomics revealed that these genes are expressed in embryonic and fetal gastrointestinal tissues. Knockout of these genes in neuronal cells demonstrated impaired cell differentiation, proliferation and/or survival. Our approach identified and validated candidate HSCR genes and provided further insight into the underlying pathomechanisms of HSCR.


Subject(s)
Hirschsprung Disease/genetics , ATP Binding Cassette Transporter, Subfamily D, Member 1/genetics , Animals , Cell Differentiation/genetics , Cell Line , Cell Proliferation/genetics , Cell Survival/genetics , Computer Simulation , Copper-Transporting ATPases/genetics , Disease Models, Animal , Gene Expression Profiling , Gene Knockout Techniques , Humans , Infant , Male , Mice , Protein Inhibitors of Activated STAT/genetics , Sterol Regulatory Element Binding Protein 1/genetics , Exome Sequencing
5.
J Clin Invest ; 130(11): 6064-6079, 2020 11 02.
Article in English | MEDLINE | ID: mdl-32750041

ABSTRACT

Store-operated Ca2+ entry (SOCE) is the major route of Ca2+ influx in platelets. The Ca2+ sensor stromal interaction molecule 1 (STIM1) triggers SOCE by forming punctate structures with the Ca2+ channel Orai1 and the inositol trisphosphate receptor (IP3R), thereby linking the endo-/sarcoplasmic reticulum to the plasma membrane. Here, we identified the BAR domain superfamily member bridging integrator 2 (BIN2) as an interaction partner of STIM1 and IP3R in platelets. Deletion of platelet BIN2 (Bin2fl/fl,Pf4-Cre mice) resulted in reduced Ca2+ store release and Ca2+ influx in response to all tested platelet agonists. These defects were a consequence of impaired IP3R function in combination with defective STIM1-mediated SOC channel activation, while Ca2+ store content and agonist-induced IP3 production were unaltered. This severely defective Ca2+ signaling translated into impaired thrombus formation under flow and a protection of Bin2fl/fl,Pf4-Cre mice in models of arterial thrombosis and stroke. Our results establish BIN2 as a central regulator of platelet activation in thrombosis and thrombo-inflammatory disease settings.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Blood Platelets/metabolism , Calcium Signaling , Thrombosis/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Blood Platelets/pathology , Disease Models, Animal , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Inositol 1,4,5-Trisphosphate Receptors/genetics , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Mice , Mice, Transgenic , Stromal Interaction Molecule 1/genetics , Stromal Interaction Molecule 1/metabolism , Thrombosis/genetics , Thrombosis/pathology
6.
Nat Commun ; 11(1): 1315, 2020 03 11.
Article in English | MEDLINE | ID: mdl-32161256

ABSTRACT

Myocardial ischemia is one of the leading health problems worldwide. Therapy consists of the restitution of coronary perfusion which is followed by myocardial inflammation. Platelet-neutrophil interaction is a crucial process during inflammation, yet its consequences are not fully understood. Here, we show that platelet-neutrophil complexes (PNCs) are increased in patients with acute myocardial infarction and that this is associated with increased levels of neuronal guidance protein semaphorin 7A (SEMA7A). To investigate this further, we injected WT animals with Sema7a and found increased infarct size with increased numbers of PNCs. Experiments in genetically modified animals identify Sema7a on red blood cells to be crucial for this condition. Further studies revealed that Sema7a interacts with the platelet receptor glycoprotein Ib (GPIb). Treatment with anti-Sema7a antibody protected from myocardial tissue injury. In summary, we show that Sema7a binds to platelet GPIb and enhances platelet thrombo-inflammatory activity, aggravating post-ischemic myocardial tissue injury.


Subject(s)
Antigens, CD/metabolism , Myocardial Infarction/pathology , Myocardial Reperfusion Injury/pathology , Platelet Glycoprotein GPIb-IX Complex/metabolism , Semaphorins/metabolism , Thrombosis/pathology , Adolescent , Adult , Aged , Aged, 80 and over , Animals , Antigens, CD/genetics , Antigens, CD/immunology , Blood Platelets/immunology , Blood Platelets/metabolism , Coronary Vessels/pathology , Disease Models, Animal , Erythrocytes/immunology , Erythrocytes/metabolism , Female , GPI-Linked Proteins/genetics , GPI-Linked Proteins/immunology , GPI-Linked Proteins/metabolism , Humans , Male , Mice , Mice, Knockout , Middle Aged , Myocardial Infarction/blood , Myocardial Infarction/immunology , Myocardial Reperfusion Injury/blood , Myocardial Reperfusion Injury/immunology , Myocardium/pathology , Prospective Studies , Semaphorins/genetics , Semaphorins/immunology , Thrombosis/immunology , Young Adult
7.
Platelets ; 31(6): 801-811, 2020 Aug 17.
Article in English | MEDLINE | ID: mdl-31948362

ABSTRACT

Platelets are essential for normal hemostasis; however, pathological conditions can also trigger unwanted platelet activation precipitating thrombosis and ischemic damage of vital organs such as the heart or brain. Glycoprotein (GP)VI- and C-type lectin-like receptor 2 (CLEC-2)-mediated (hem)immunoreceptor tyrosine-based activation motif (ITAM) signaling represents a major pathway for platelet activation. The two members of the Growth-factor receptor-bound protein 2 (Grb2) family of adapter proteins expressed in platelets - Grb2 and Grb2-related adapter protein downstream of Shc (Gads) - are part of the hem(ITAM) signaling cascade by forming an adapter protein complex with linker for activation of T cells (LAT). To date, a possible functional redundancy between these two adapters in platelet activation has not been investigated. We here generated megakaryocyte- and platelet-specific Grb2/Gads double knockout (DKO) mice and analyzed their platelet function in vitro and in vivo. The DKO platelets exhibited virtually abolished (hem)ITAM signaling whereas only partial defects were seen in Grb2 or Gads single-deficient platelets. This was based on impaired phosphorylation of key molecules in the (hem)ITAM signaling cascade and translated into impaired hemostasis and partially defective arterial thrombosis, thereby exceeding the defects in either Grb2 KO or Gads KO mice. Despite this severe (hem)ITAM signaling defect, CLEC-2 dependent regulation of blood-lymphatic vessel separation was not affected in the DKO animals. These results provide direct evidence for critically redundant roles of Grb2 and Gads for platelet function in hemostasis and thrombosis, but not development.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , GRB2 Adaptor Protein/metabolism , Immunoreceptor Tyrosine-Based Activation Motif/genetics , Animals , Humans , Mice , Signal Transduction
8.
Haematologica ; 105(6): 1738-1749, 2020 06.
Article in English | MEDLINE | ID: mdl-31537686

ABSTRACT

Genetically modified mice have advanced our knowledge on platelets in hemostasis and beyond tremendously. However, mouse models harbor certain limitations, including availability of platelet specific transgenic strains, and off-target effects on other cell types. Transfusion of genetically modified platelets into thrombocytopenic mice circumvents these problems. Additionally, ex vivo treatment of platelets prior to transfusion eliminates putative side effects on other cell types. Thrombocytopenia is commonly induced by administration of anti-platelet antibodies, which opsonize platelets to cause rapid clearance. However, antibodies do not differentiate between endogenous or exogenous platelets, impeding transfusion efficacy. In contrast, genetic depletion with the inducible diphtheria toxin receptor (iDTR) system induces thrombocytopenia via megakaryocyte ablation without direct effects on circulating platelets. We compared the iDTR system with antibody-based depletion methods regarding their utility in platelet transfusion experiments, outlining advantages and disadvantages of both approaches. Antibodies led to thrombocytopenia within two hours and allowed the dose-dependent adjustment of the platelet count. The iDTR model caused complete thrombocytopenia within four days, which could be sustained for up to 11 days. Neither platelet depletion approach caused platelet activation. Only the iDTR model allowed efficient platelet transfusion by keeping endogenous platelet levels low and maintaining exogenous platelet levels over longer time periods, thus providing clear advantages over antibody-based methods. Transfused platelets were fully functional in vivo, and our model allowed examination of transgenic platelets. Using donor platelets from already available genetically modified mice or ex vivo treated platelets, may decrease the necessity of platelet-specific mouse strains, diminishing off-target effects and thereby reducing animal numbers.


Subject(s)
Platelet Count , Platelet Transfusion , Thrombocytopenia , Animals , Blood Platelets , Hemostasis , Mice , Thrombocytopenia/genetics , Thrombocytopenia/therapy
9.
Blood ; 134(25): 2318-2329, 2019 12 19.
Article in English | MEDLINE | ID: mdl-31697813

ABSTRACT

During platelet spreading, the actin cytoskeleton undergoes rapid rearrangement, forming filopodia and lamellipodia. Controversial data have been published on the role of lamellipodia in thrombus formation and stability. The Wiskott-Aldrich syndrome protein-family verprolin-homologous protein (WAVE)-regulatory complex, which has been shown in other cells to drive lamellipodium formation by enhancing actin nucleation via the actin-related protein 2/3 (Arp2/3) complex, is activated by Ras-related C3 botulinum toxin substrate 1 (Rac1) interaction with the WAVE complex subunit cytoplasmic fragile X mental retardation 1-interacting protein 1 (Cyfip1). We analyzed Cyfip1flox/floxPf4-Cre mice to investigate the role of Cyfip1 in platelet function. These mice displayed normal platelet counts and a slight reduction in platelet volume. Activation of mutant platelets was only moderately reduced to all tested agonists as measured by αIIbß3 integrin activation and P-selectin surface exposure. However, lamellipodium formation of mutant platelets was completely abolished on different matrices. Nevertheless, Cyfip1-deficient platelets formed stable thrombi on collagen fibers ex vivo and in 2 models of occlusive arterial thrombosis in vivo. Similarly, the hemostatic function and maintenance of vascular integrity during inflammation of the skin and lung were unaltered in the mutant mice. Investigation of platelet morphology in an induced thrombus under flow revealed that platelets rather form filopodia in the thrombus shell, and are flattened with filopodium-like structures when in direct contact to collagen fibers at the bottom of the thrombus. We provide for the first time direct evidence that platelet lamellipodium formation is not required for stable thrombus formation, and that morphological changes of platelets differ between a static spreading assay and thrombus formation under flow.


Subject(s)
Blood Platelets/metabolism , Pseudopodia/metabolism , Thrombosis/metabolism , Actin-Related Protein 2-3 Complex/genetics , Actin-Related Protein 2-3 Complex/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Blood Platelets/pathology , Female , Male , Mice , Mice, Transgenic , Neuropeptides/genetics , Neuropeptides/metabolism , P-Selectin/genetics , P-Selectin/metabolism , Pseudopodia/genetics , Thrombosis/genetics , Thrombosis/pathology , rac1 GTP-Binding Protein/genetics , rac1 GTP-Binding Protein/metabolism
10.
Sci Rep ; 9(1): 8333, 2019 06 06.
Article in English | MEDLINE | ID: mdl-31171812

ABSTRACT

Zinc (Zn2+) can modulate platelet and coagulation activation pathways, including fibrin formation. Here, we studied the (patho)physiological consequences of abnormal platelet Zn2+ storage and release. To visualize Zn2+ storage in human and mouse platelets, the Zn2+ specific fluorescent dye FluoZin3 was used. In resting platelets, the dye transiently accumulated into distinct cytosolic puncta, which were lost upon platelet activation. Platelets isolated from Unc13d-/- mice, characterized by combined defects of α/δ granular release, showed a markedly impaired Zn2+ release upon activation. Platelets from Nbeal2-/- mice mimicking Gray platelet syndrome (GPS), characterized by primarily loss of the α-granule content, had strongly reduced Zn2+ levels, which was also confirmed in primary megakaryocytes. In human platelets isolated from patients with GPS, Hermansky-Pudlak Syndrome (HPS) and Storage Pool Disease (SPD) altered Zn2+ homeostasis was detected. In turbidity and flow based assays, platelet-dependent fibrin formation was impaired in both Nbeal2-/- and Unc13d-/- mice, and the impairment could be partially restored by extracellular Zn2+. Altogether, we conclude that the release of ionic Zn2+ store from secretory granules upon platelet activation contributes to the procoagulant role of Zn2+ in platelet-dependent fibrin formation.


Subject(s)
Blood Platelets/cytology , Blood Proteins/genetics , Membrane Proteins/genetics , Platelet Storage Pool Deficiency/genetics , Zinc/metabolism , Adolescent , Adult , Animals , Blood Coagulation , Child , Cytosol/metabolism , Female , Fibrin/chemistry , Gray Platelet Syndrome/genetics , Healthy Volunteers , Hermanski-Pudlak Syndrome/genetics , Homeostasis , Humans , Male , Mice , Mice, Knockout , Microscopy, Confocal , Microscopy, Fluorescence , Nephelometry and Turbidimetry , Platelet Activation
11.
Blood ; 133(25): 2696-2706, 2019 06 20.
Article in English | MEDLINE | ID: mdl-30952674

ABSTRACT

Maintenance of tumor vasculature integrity is indispensable for tumor growth and thus affects tumor progression. Previous studies have identified platelets as major regulators of tumor vascular integrity, as their depletion selectively rendered tumor vessels highly permeable and caused massive intratumoral hemorrhage. While these results established platelets as potential targets for antitumor therapy, their depletion is not a treatment option due to their essential role in hemostasis. Thus, a detailed understanding of how platelets safeguard vascular integrity in tumors is urgently demanded. Here, we show for the first time that functional inhibition of glycoprotein VI (GPVI) on the platelet surface with an antibody (JAQ1) F(ab)2 fragment rapidly induces tumor hemorrhage and diminishes tumor growth similar to complete platelet depletion while not inducing systemic bleeding complications. The intratumor bleeding and tumor growth arrest could be reverted by depletion of Ly6G+ cells, confirming them to be responsible for the induction of bleeding and necrosis within the tumor. In addition, JAQ1 F(ab)2-mediated GPVI inhibition increased intratumoral accumulation of coadministered chemotherapeutic agents, such as Doxil and paclitaxel, thereby resulting in a profound antitumor effect. In summary, our findings identify platelet GPVI as a key regulator of vascular integrity specifically in growing tumors and could serve as a basis for the development of antitumor strategies based on the interference with platelet function.


Subject(s)
Immunoglobulin Fab Fragments/pharmacology , Neoplasms, Experimental/pathology , Platelet Membrane Glycoproteins/antagonists & inhibitors , Animals , Female , Hemorrhage/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neovascularization, Pathologic
12.
Nat Med ; 25(4): 641-655, 2019 04.
Article in English | MEDLINE | ID: mdl-30936549

ABSTRACT

Non-alcoholic fatty liver disease ranges from steatosis to non-alcoholic steatohepatitis (NASH), potentially progressing to cirrhosis and hepatocellular carcinoma (HCC). Here, we show that platelet number, platelet activation and platelet aggregation are increased in NASH but not in steatosis or insulin resistance. Antiplatelet therapy (APT; aspirin/clopidogrel, ticagrelor) but not nonsteroidal anti-inflammatory drug (NSAID) treatment with sulindac prevented NASH and subsequent HCC development. Intravital microscopy showed that liver colonization by platelets depended primarily on Kupffer cells at early and late stages of NASH, involving hyaluronan-CD44 binding. APT reduced intrahepatic platelet accumulation and the frequency of platelet-immune cell interaction, thereby limiting hepatic immune cell trafficking. Consequently, intrahepatic cytokine and chemokine release, macrovesicular steatosis and liver damage were attenuated. Platelet cargo, platelet adhesion and platelet activation but not platelet aggregation were identified as pivotal for NASH and subsequent hepatocarcinogenesis. In particular, platelet-derived GPIbα proved critical for development of NASH and subsequent HCC, independent of its reported cognate ligands vWF, P-selectin or Mac-1, offering a potential target against NASH.


Subject(s)
Blood Platelets/metabolism , Liver Neoplasms/blood , Liver Neoplasms/drug therapy , Non-alcoholic Fatty Liver Disease/blood , Non-alcoholic Fatty Liver Disease/drug therapy , Platelet Glycoprotein GPIb-IX Complex/metabolism , Animals , Blood Platelets/drug effects , Body Weight/drug effects , Cytokines/metabolism , Cytoplasmic Granules/drug effects , Cytoplasmic Granules/metabolism , Endothelium/drug effects , Endothelium/metabolism , Hepatocytes/drug effects , Hepatocytes/pathology , Humans , Hyaluronan Receptors/metabolism , Hyaluronic Acid/metabolism , Kupffer Cells/drug effects , Kupffer Cells/metabolism , Liver/drug effects , Liver/metabolism , Liver/pathology , Mice, Transgenic , Platelet Aggregation/drug effects , Platelet Aggregation Inhibitors/pharmacology , Platelet Count
13.
Stroke ; 50(2): 478-486, 2019 02.
Article in English | MEDLINE | ID: mdl-30566040

ABSTRACT

Background and Purpose- Acetylsalicylic acid and clopidogrel are the 2 main antithrombotic drugs for secondary prevention in patients with ischemic stroke (IS) without indication for anticoagulation. Because of their limited efficacy and potential side effects, novel antiplatelet agents are urgently needed. Cilostazol, a specific phosphodiesterase (PDE)-3 inhibitor, protected from IS in clinical studies comprising mainly Asian populations. Nevertheless, the detailed mechanistic role of PDE-3 inhibitors in IS pathophysiology is hardly understood. In this project, we analyzed the efficacy and pathophysiologic mechanisms of a novel and only recently described PDE-3 inhibitor (substance V) in a mouse model of focal cerebral ischemia. Methods- Focal cerebral ischemia was induced by transient middle cerebral artery occlusion in 6- to 8-week-old male C57Bl/6 wild-type mice receiving substance V or vehicle 1 hour after ischemia induction. Infarct volumes and functional outcomes were assessed between day 1 and day 7, and findings were validated by magnetic resonance imaging. Blood-brain barrier damage, as well as the extent of local inflammatory response and cell death, was determined. Results- Inhibition of PDE-3 by pharmacological blockade with substance V significantly reduced infarct volumes and improved neurological outcome on day 1 and 7 after experimental cerebral ischemia. Reduced blood-brain barrier damage, attenuated brain tissue inflammation, and decreased local cell death could be identified as potential mechanisms. PDE-3 inhibitor treatment did neither increase the number of intracerebral hemorrhages nor affect platelet function. Conclusions- The novel PDE-3 inhibitor substance V protected mice from IS independent from platelet function. Pharmaceutical inactivation of PDE-3 might become a promising therapeutic approach to combat IS via inhibition of thromboinflammatory mechanisms and stabilization of the blood-brain barrier.


Subject(s)
Blood Platelets/metabolism , Blood-Brain Barrier/metabolism , Brain Ischemia/prevention & control , Cilostazol/pharmacology , Phosphodiesterase 3 Inhibitors/pharmacology , Stroke/prevention & control , Animals , Blood Platelets/pathology , Blood-Brain Barrier/pathology , Brain Ischemia/metabolism , Brain Ischemia/pathology , Disease Models, Animal , Male , Mice , Stroke/metabolism , Stroke/pathology
14.
Blood ; 129(12): 1702-1706, 2017 03 23.
Article in English | MEDLINE | ID: mdl-28077416

ABSTRACT

Platelets maintain hemostasis after injury, but also during inflammation. Recent studies have shown that platelets prevent inflammatory bleeding through (hem) immunoreceptor tyrosine-based activation motif-dependent mechanisms irrespective of aggregation during skin and lung inflammation. Although the exact mechanisms underlying this process remain unknown, it was speculated that mediators released from platelet granules might be involved. Maintaining cerebral hemostasis during stroke treatment is of high clinical relevance because hemorrhage may aggravate the disease state and increase mortality. Although it was shown that platelets help maintain hemostasis in the ischemic brain, their exact contribution remains ill defined. Here we show that Unc13d-/- /Nbeal2-/- mice, which lack platelet α- and dense-granule secretion, show no signs of hemorrhage in models of skin or lung inflammation. In stark contrast, lack of platelet granule release resulted in impaired hemostasis in the ischemic brain after transient middle cerebral artery occlusion leading to increased intracranial hemorrhage and mortality. Our results reveal for the first time that platelet granule constituents are essential for maintenance of hemostasis during thrombo-inflammatory brain infarction but not experimental inflammation of the skin or lung, thereby uncovering vascular bed-specific differences in the prevention of inflammatory bleeding.


Subject(s)
Blood Platelets/metabolism , Brain Ischemia/pathology , Cerebral Hemorrhage/prevention & control , Animals , Brain Ischemia/blood , Cerebral Hemorrhage/blood , Hemorrhage/pathology , Hemostasis , Infarction, Middle Cerebral Artery , Inflammation/blood , Inflammation/pathology , Lung/pathology , Mice , Secretory Vesicles/physiology , Skin/pathology
15.
J Immunol ; 194(9): 4422-30, 2015 May 01.
Article in English | MEDLINE | ID: mdl-25801434

ABSTRACT

The proinflammatory microenvironment in the respiratory airway induces maturation of both resident and infiltrating dendritic cells (DCs) upon influenza A virus (IAV) infection. This results in upregulation of antiviral pathways as well as modulation of endocytic processes, which affect the susceptibility of DCs to IAV infection. Therefore, it is highly relevant to understand how IAV interacts with and infects mature DCs. To investigate how different subsets of human myeloid DCs (MDCs) involved in tissue inflammation are affected by inflammatory stimulation during IAV infection, we stimulated primary blood MDCs and inflammatory monocyte-derived DCs (MDDCs) with TLR ligands, resulting in maturation. Interestingly, MDDCs but not MDCs were protected against IAV infection after LPS (TLR4) stimulation. In contrast, stimulation with TLR7/8 ligand protected MDCs but not MDDCs from IAV infection. The reduced susceptibility to IAV infection correlated with induction of type I IFNs. We found that differential expression of TLR4, TRIF, and MyD88 in the two MDC subsets regulated the ability of the cells to enter an antiviral state upon maturation. This difference was functionally confirmed using small interfering RNA and inhibitors. Our data show that different human MDC subsets may play distinct roles during IAV infection, as their capacity to induce type I IFNs is dependent on TLR-specific maturation, resulting in differential susceptibility to IAV infection.


Subject(s)
Dendritic Cells/metabolism , Influenza A virus/immunology , Influenza, Human/immunology , Influenza, Human/metabolism , Myeloid Cells/metabolism , Toll-Like Receptors/metabolism , Adaptor Proteins, Vesicular Transport/genetics , Adaptor Proteins, Vesicular Transport/metabolism , Dendritic Cells/drug effects , Dendritic Cells/immunology , Dendritic Cells/virology , Gene Knockdown Techniques , Humans , Influenza, Human/genetics , Interferon Type I/biosynthesis , Lipopolysaccharides/immunology , Lipopolysaccharides/pharmacology , Myeloid Cells/drug effects , Myeloid Cells/immunology , Myeloid Cells/virology , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , Poly I-C/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism , Toll-Like Receptors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...