Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 59
Filter
Add more filters

Publication year range
1.
J Med Virol ; 96(6): e29735, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38864313

ABSTRACT

Recently, hepatitis E virus (HEV, Paslahepevirus balayani) particles were detected for the first time in the ejaculate of two chronically infected patients. Since then, we have been able to detect HEV in ejaculate in five further patients, and thus in a total of seven out of nine (78%) chronically infected men (age 36-67 years, median 56 years). In five patients, the HEV RNA concentration was more than 100-fold higher compared to the serum, while in two patients, the viral load was more than 10-fold lower. However, it has remained unclear whether viral particles shed in the ejaculate were infectious, as a previous cell culture model had failed to demonstrate the infectivity. In the current study, we employed an optimized HEV cell culture system based on overconfluent PLC/PRF/5 cells to investigate the infectivity of HEV particles from ejaculate and other body fluids. With this approach, we were able to show for the first time that HEV particles in the ejaculate from several patients were infectious. HEV replicated to high viral loads of 1e9 HEV RNA copies per ml. This indicates that HEV-positive ejaculate could bear a risk of infection for sexual partners.


Subject(s)
Hepatitis E virus , Hepatitis E , RNA, Viral , Viral Load , Humans , Hepatitis E virus/isolation & purification , Middle Aged , Hepatitis E/virology , Male , Adult , Aged , RNA, Viral/analysis , Semen/virology , Virion , Cell Line , Virus Shedding
2.
Hippocampus ; 32(7): 517-528, 2022 07.
Article in English | MEDLINE | ID: mdl-35621370

ABSTRACT

Temporal lobe epilepsy is characterized by hippocampal neuronal death in CA1 and hilus. Dentate gyrus granule cells survive but show dispersion of the compact granule cell layer. This is associated with decrease of the glycoprotein Reelin, which regulates neuron migration and dendrite outgrow. Reelin-deficient (reeler) mice show no layering, their granule cells are dispersed throughout the dentate gyrus. We studied granule cell dendritic orientation and distribution of postsynaptic spines in reeler mice and two mouse models of temporal lobe epilepsy, namely the p35 knockout mice, which show Reelin-independent neuronal migration defects, and mice with unilateral intrahippocampal kainate injection. Granule cells were Golgi-stained and analyzed, using a computerized camera lucida system. Granule cells in naive controls exhibited a vertically oriented dendritic arbor with a small bifurcation angle if positioned proximal to the hilus and a wider dendritic bifurcation angle, if positioned distally. P35 knockout- and kainate-injected mice showed a dispersed granule cell layer, granule cells showed basal dendrites with wider bifurcation angles, which lost position-specific differences. Reeler mice lacked dendritic orientation. P35 knockout- and kainate-injected mice showed increased dendritic spine density in the granule cell layer. Molecular layer dendrites showed a reduced spine density in kainate-injected mice only, whereas in p35 knockouts no reduced spine density was seen. Reeler mice showed a homogenous high spine density. We hypothesize that granule cells migrate in temporal lobe epilepsy, develop new dendrites which show a spread of the dendritic tree, create new spines in areas proximal to mossy fiber sprouting, which is present in p35 knockout- and kainate-injected mice and loose spines on distal dendrites if mossy cell death is present, as it was in kainate-injected mice only. These results are in accordance with findings in epilepsy patients.


Subject(s)
Epilepsy, Temporal Lobe , Animals , Dendrites/metabolism , Dentate Gyrus , Disease Models, Animal , Epilepsy, Temporal Lobe/chemically induced , Epilepsy, Temporal Lobe/metabolism , Humans , Kainic Acid/toxicity , Mice , Mice, Neurologic Mutants , Neurons/metabolism
3.
J Hepatol ; 74(6): 1455-1471, 2021 06.
Article in English | MEDLINE | ID: mdl-33577920

ABSTRACT

The term non-alcoholic fatty liver disease (NAFLD) was originally coined to describe hepatic fat deposition as part of the metabolic syndrome. However, a variety of rare hereditary liver and metabolic diseases, intestinal diseases, endocrine disorders and drugs may underlie, mimic, or aggravate NAFLD. In contrast to primary NAFLD, therapeutic interventions are available for many secondary causes of NAFLD. Accordingly, secondary causes of fatty liver disease should be considered during the diagnostic workup of patients with fatty liver disease, and treatment of the underlying disease should be started to halt disease progression. Common genetic variants in several genes involved in lipid handling and metabolism modulate the risk of progression from steatosis to fibrosis, cirrhosis and hepatocellular carcinoma development in NAFLD, alcohol-related liver disease and viral hepatitis. Hence, we speculate that genotyping of common risk variants for liver disease progression may be equally useful to gauge the likelihood of developing advanced liver disease in patients with secondary fatty liver disease.


Subject(s)
Chemical and Drug Induced Liver Injury/epidemiology , Endocrine System Diseases/epidemiology , Gastrointestinal Diseases/epidemiology , Genetic Diseases, Inborn/epidemiology , Hepacivirus , Hepatitis C, Chronic/epidemiology , Metabolic Syndrome/epidemiology , Non-alcoholic Fatty Liver Disease/epidemiology , Obesity, Abdominal/epidemiology , Pregnancy Complications/epidemiology , Adult , Child , Comorbidity , Endocrine System Diseases/drug therapy , Female , Gastrointestinal Diseases/diet therapy , Gastrointestinal Diseases/drug therapy , Genetic Diseases, Inborn/diet therapy , Genetic Predisposition to Disease/genetics , Hepatitis C, Chronic/drug therapy , Hepatitis C, Chronic/virology , Humans , Male , Metabolic Syndrome/diet therapy , Metabolic Syndrome/drug therapy , Non-alcoholic Fatty Liver Disease/genetics , Obesity, Abdominal/complications , Obesity, Abdominal/diet therapy , Pregnancy , Risk Factors , Young Adult
4.
Biol Chem ; 402(9): 1021-1032, 2021 08 26.
Article in English | MEDLINE | ID: mdl-34008380

ABSTRACT

This review article summarizes 20 years of our research on hepatic stellate cells within the framework of two collaborative research centers CRC575 and CRC974 at the Heinrich Heine University. Over this period, stellate cells were identified for the first time as mesenchymal stem cells of the liver, and important functions of these cells in the context of liver regeneration were discovered. Furthermore, it was determined that the space of Disse - bounded by the sinusoidal endothelium and hepatocytes - functions as a stem cell niche for stellate cells. Essential elements of this niche that control the maintenance of hepatic stellate cells have been identified alongside their impairment with age. This article aims to highlight previous studies on stellate cells and critically examine and identify open questions and future research directions.


Subject(s)
Hepatic Stellate Cells , Cell Differentiation , Hepatocytes , Humans , Liver , Liver Regeneration , Stem Cell Niche
5.
Arterioscler Thromb Vasc Biol ; 40(10): 2391-2403, 2020 10.
Article in English | MEDLINE | ID: mdl-32787521

ABSTRACT

OBJECTIVE: Reelin, a secreted glycoprotein, was originally identified in the central nervous system, where it plays an important role in brain development and maintenance. In the cardiovascular system, reelin plays a role in atherosclerosis by enhancing vascular inflammation and in arterial thrombosis by promoting platelet adhesion, activation, and thrombus formation via APP (amyloid precursor protein) and GP (glycoprotein) Ib. However, the role of reelin in hemostasis and arterial thrombosis is not fully understood to date. Approach and Results: In the present study, we analyzed the importance of reelin for cytoskeletal reorganization of platelets and thrombus formation in more detail. Platelets release reelin to amplify alphaIIb beta3 integrin outside-in signaling by promoting platelet adhesion, cytoskeletal reorganization, and clot retraction via activation of Rho GTPases RAC1 (Ras-related C3 botulinum toxin substrate) and RhoA (Ras homolog family member A). Reelin interacts with the collagen receptor GP (glycoprotein) VI with subnanomolar affinity, induces tyrosine phosphorylation in a GPVI-dependent manner, and supports platelet binding to collagen and GPVI-dependent RAC1 activation, PLC gamma 2 (1-phosphatidylinositol-4,5-bisphosphate phosphodiesterase gamma-2) phosphorylation, platelet activation, and aggregation. When GPVI was deleted from the platelet surface by antibody treatment in reelin-deficient mice, thrombus formation was completely abolished after injury of the carotid artery while being only reduced in either GPVI-depleted or reelin-deficient mice. CONCLUSIONS: Our study identified a novel signaling pathway that involves reelin-induced GPVI activation and alphaIIb beta3 integrin outside-in signaling in platelets. Loss of both, GPVI and reelin, completely prevents stable arterial thrombus formation in vivo suggesting that inhibiting reelin-platelet-interaction might represent a novel strategy to avoid arterial thrombosis in cardiovascular disease.


Subject(s)
Blood Platelets/enzymology , Carotid Artery Injuries/enzymology , Cell Adhesion Molecules, Neuronal/blood , Extracellular Matrix Proteins/blood , Nerve Tissue Proteins/blood , Neuropeptides/blood , Phospholipase C gamma/blood , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Platelet Membrane Glycoproteins/metabolism , Serine Endopeptidases/blood , Thrombosis/enzymology , rac1 GTP-Binding Protein/blood , rhoA GTP-Binding Protein/blood , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Blood Coagulation , Carotid Artery Injuries/blood , Carotid Artery Injuries/etiology , Cell Adhesion Molecules, Neuronal/deficiency , Cell Adhesion Molecules, Neuronal/genetics , Clot Retraction , Cytoskeleton/enzymology , Disease Models, Animal , Extracellular Matrix Proteins/deficiency , Extracellular Matrix Proteins/genetics , Mice, 129 Strain , Mice, Inbred C3H , Mice, Inbred C57BL , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Platelet Activation , Reelin Protein , Serine Endopeptidases/deficiency , Serine Endopeptidases/genetics , Signal Transduction , Thrombosis/blood , Thrombosis/etiology
6.
Cereb Cortex ; 30(3): 1688-1707, 2020 03 14.
Article in English | MEDLINE | ID: mdl-31667489

ABSTRACT

Reelin is an extracellular matrix protein, known for its dual role in neuronal migration during brain development and in synaptic plasticity at adult stages. During the perinatal phase, Reelin expression switches from Cajal-Retzius (CR) cells, its main source before birth, to inhibitory interneurons (IN), the main source of Reelin in the adult forebrain. IN-derived Reelin has been associated with schizophrenia and temporal lobe epilepsy; however, the functional role of Reelin from INs is presently unclear. In this study, we used conditional knockout mice, which lack Reelin expression specifically in inhibitory INs, leading to a substantial reduction in total Reelin expression in the neocortex and dentate gyrus. Our results show that IN-specific Reelin knockout mice exhibit normal neuronal layering and normal behavior, including spatial reference memory. Although INs are the major source of Reelin within the adult stem cell niche, Reelin from INs does not contribute substantially to normal adult neurogenesis. While a closer look at the dentate gyrus revealed some unexpected alterations at the cellular level, including an increase in the number of Reelin expressing CR cells, overall our data suggest that Reelin derived from INs is less critical for cortex development and function than Reelin expressed by CR cells.


Subject(s)
Cell Adhesion Molecules, Neuronal/metabolism , Dentate Gyrus/metabolism , Extracellular Matrix Proteins/metabolism , Interneurons/metabolism , Neocortex/metabolism , Nerve Tissue Proteins/metabolism , Serine Endopeptidases/metabolism , Animals , Behavior, Animal/physiology , Cell Movement/physiology , Dentate Gyrus/physiopathology , Hippocampus/metabolism , Interneurons/drug effects , Mice, Inbred C57BL , Mice, Knockout , Neurogenesis/physiology , Neurons/metabolism , Plant Leaves/metabolism , Reelin Protein
7.
Proc Natl Acad Sci U S A ; 114(21): 5533-5538, 2017 05 23.
Article in English | MEDLINE | ID: mdl-28484035

ABSTRACT

Brain development and function depend on the directed and coordinated migration of neurons from proliferative zones to their final position. The secreted glycoprotein Reelin is an important factor directing neuronal migration. Loss of Reelin function results in the severe developmental disorder lissencephaly and is associated with neurological diseases in humans. Reelin signals via the lipoprotein receptors very low density lipoprotein receptor (VLDLR) and apolipoprotein E receptor 2 (ApoER2), but the exact mechanism by which these receptors control cellular function is poorly understood. We report that loss of the signaling scaffold intersectin 1 (ITSN1) in mice leads to defective neuronal migration and ablates Reelin stimulation of hippocampal long-term potentiation (LTP). Knockout (KO) mice lacking ITSN1 suffer from dispersion of pyramidal neurons and malformation of the radial glial scaffold, akin to the hippocampal lamination defects observed in VLDLR or ApoER2 mutants. ITSN1 genetically interacts with Reelin receptors, as evidenced by the prominent neuronal migration and radial glial defects in hippocampus and cortex seen in double-KO mice lacking ITSN1 and ApoER2. These defects were similar to, albeit less severe than, those observed in Reelin-deficient or VLDLR/ ApoER2 double-KO mice. Molecularly, ITSN1 associates with the VLDLR and its downstream signaling adaptor Dab1 to facilitate Reelin signaling. Collectively, these data identify ITSN1 as a component of Reelin signaling that acts predominantly by facilitating the VLDLR-Dab1 axis to direct neuronal migration in the cortex and hippocampus and to augment synaptic plasticity.


Subject(s)
Adaptor Proteins, Vesicular Transport/metabolism , Cell Adhesion Molecules, Neuronal/metabolism , Extracellular Matrix Proteins/metabolism , Hippocampus/metabolism , Nerve Tissue Proteins/metabolism , Neuronal Plasticity , Neurons/physiology , Serine Endopeptidases/metabolism , Adaptor Proteins, Vesicular Transport/genetics , Animals , Cell Movement , LDL-Receptor Related Proteins/genetics , LDL-Receptor Related Proteins/metabolism , Mice, Knockout , Receptors, LDL/metabolism , Receptors, N-Methyl-D-Aspartate/isolation & purification , Receptors, N-Methyl-D-Aspartate/metabolism , Reelin Protein
8.
J Hepatol ; 69(3): 594-602, 2018 09.
Article in English | MEDLINE | ID: mdl-29705238

ABSTRACT

BACKGROUND & AIMS: To affect immune response and inflammation, the hepatitis C virus (HCV) substantially influences intercellular communication pathways that are decisive for immune cell recruitment. The present study investigates mechanisms by which HCV modulates chemokine-mediated intercellular communication from infected cells. METHODS: Chemokine expression was studied in HCVcc-infected cell lines or cell lines harbouring a subgenomic replicon, as well as in serum samples from patients. Expression or activity of mediators and signalling intermediates was manipulated using knockdown approaches or specific inhibitors. RESULTS: HCV enhances expression of CXCR2 ligands in its host cell via the induction of epidermal growth factor (EGF) production. Knockdown of EGF or of the p65 subunit of the NF-κB complex results in a substantial downregulation of HCV-induced CXCR2 ligand expression, supporting the involvement of an EGF-dependent mechanism as well as activation of NF-κB. Furthermore, HCV upregulates expression of CXCR2 ligands in response to EGF stimulation via downregulation of the T-cell protein tyrosine phosphatase (TC-PTP [PTPN2]), activation of NF-κB, and enhancement of EGF-inducible signal transduction via MEK1 (MAP2K1). This results in the production of a cytokine/chemokine pattern by the HCV-infected cell that can recruit neutrophils but not monocytes. CONCLUSIONS: These data reveal a novel EGF-dependent mechanism by which HCV influences chemokine-mediated intercellular communication. We propose that this mechanism contributes to modulation of the HCV-induced inflammation and the antiviral immune response. LAY SUMMARY: In most cases hepatitis C virus (HCV) results in chronic infection and persistent viral replication, taking decades until development of overt disease. To achieve such a course, the respective virus must have developed mechanisms to circumvent antiviral response, to modulate the inflammatory response and to utilise the infrastructure of its host with moderate effect on its viability. The present study provides novel data indicating that HCV induces epidermal growth factor production in its host cell, enhancing epidermal growth factor-inducible expression of chemokines that bind to the CXCR2 receptor and recruit neutrophile granulocytes. Importantly, chemokines are critical mediators determining the pattern of immune cells recruited to the site of injury and thereby the local inflammatory and immunological milieu. These data strongly suggest that HCV triggers mechanisms that enable the virus to influence the inflammatory and immunological processes of its host.


Subject(s)
Cell Communication/immunology , Epidermal Growth Factor , Hepacivirus/physiology , Hepatitis C, Chronic , Inflammation , Receptors, Interleukin-8B/immunology , Signal Transduction/immunology , Cell Line , Epidermal Growth Factor/immunology , Epidermal Growth Factor/metabolism , Hepatitis C, Chronic/immunology , Hepatitis C, Chronic/virology , Host Microbial Interactions/immunology , Humans , Immunity, Cellular , Inflammation/immunology , Inflammation/virology , Up-Regulation , Virus Replication/physiology
9.
Development ; 141(23): 4513-25, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25377550

ABSTRACT

Low density lipoprotein receptor-related protein 1 (LRP1) is indispensable for embryonic development. Comparing different genetically engineered mouse models, we found that expression of Lrp1 is essential in the embryo proper. Loss of LRP1 leads to lethal vascular defects with lack of proper investment with mural cells of both large and small vessels. We further demonstrate that LRP1 modulates Gi-dependent sphingosine-1-phosphate (S1P) signaling and integrates S1P and PDGF-BB signaling pathways, which are both crucial for mural cell recruitment, via its intracellular domain. Loss of LRP1 leads to a lack of S1P-dependent inhibition of RAC1 and loss of constraint of PDGF-BB-induced cell migration. Our studies thus identify LRP1 as a novel player in angiogenesis and in the recruitment and maintenance of mural cells. Moreover, they reveal an unexpected link between lipoprotein receptor and sphingolipid signaling that, in addition to angiogenesis during embryonic development, is of potential importance for other targets of these pathways, such as tumor angiogenesis and inflammatory processes.


Subject(s)
Embryonic Development/physiology , Lysophospholipids/metabolism , Neovascularization, Physiologic/physiology , Receptors, LDL/metabolism , Signal Transduction/physiology , Sphingosine/analogs & derivatives , Tumor Suppressor Proteins/metabolism , Animals , Becaplermin , Blotting, Western , Cell Movement/physiology , Genetic Engineering , Human Umbilical Vein Endothelial Cells , Humans , Immunohistochemistry , In Situ Hybridization , Low Density Lipoprotein Receptor-Related Protein-1 , Mice , Microscopy, Electron , Proto-Oncogene Proteins c-sis/metabolism , Real-Time Polymerase Chain Reaction , Sphingosine/metabolism
10.
Virol J ; 14(1): 106, 2017 06 08.
Article in English | MEDLINE | ID: mdl-28595606

ABSTRACT

BACKGROUND: The major resistance-associated substitution for sofosbuvir (S282T) in HCV NS5B causes severe viral fitness costs and rapidly reverts back to prototype in the absence of selection pressure. Accordingly, resistance against sofosbuvir is rarely detected even in patients after treatment failure. CASE PRESENTATION: We report a case of a GT3a infected patient with viral breakthrough under SOF/DCV therapy. At the time of breakthrough the RAS S282T was predominant in NS5B and then rapidly disappeared during follow-up by week 12 after treatment. Interestingly, despite only serine was encoded in position 282 during follow-up, two distinct genetic pathways for reversion were detectable. In 31% of the quasispecies the original codon for serine was present whereas in the majority of the quasispecies an alternative codon was selected. This alternative codon usage was unique for all GT3a isolates from the HCV database and remained detectable as a genetic footprint for prior resistance selection at the RNA level for at least 6 months. CONCLUSIONS: Comparative analyses of viral sequences at the codon level before and after DAA treatment may help to elucidate the patient's history of resistance selection, which is particularly valuable for highly unfit substitutions that are detectable only for a short period of time. If such codon changes increase the risk of re-selection of resistance upon a second exposure to SOF remains to be addressed.


Subject(s)
Antiviral Agents/therapeutic use , Hepatitis C, Chronic/drug therapy , Hepatitis C, Chronic/virology , Mutation, Missense , Sofosbuvir/therapeutic use , Treatment Failure , Viral Nonstructural Proteins/genetics , Amino Acid Substitution , Female , Humans , Middle Aged , Suppression, Genetic
12.
Proc Natl Acad Sci U S A ; 111(17): 6431-6, 2014 Apr 29.
Article in English | MEDLINE | ID: mdl-24737893

ABSTRACT

Large glycosylating toxins are major virulence factors of various species of pathogenic Clostridia. Prototypes are Clostridium difficile toxins A and B, which cause antibiotics-associated diarrhea and pseudomembranous colitis. The current model of the toxins' action suggests that receptor binding is mediated by a C-terminal domain of combined repetitive oligopeptides (CROP). This model is challenged by the glycosylating Clostridium perfringens large cytotoxin (TpeL toxin) that is devoid of the CROP domain but still intoxicates cells. Using a haploid genetic screen, we identified LDL receptor-related protein 1 (LRP1) as a host cell receptor for the TpeL toxin. LRP1-deficient cells are not able to take up TpeL and are not intoxicated. Expression of cluster IV of LRP1 is sufficient to rescue toxin uptake in these cells. By plasmon resonance spectroscopy, a KD value of 23 nM was determined for binding of TpeL to LRP1 cluster IV. The C terminus of TpeL (residues 1335-1779) represents the receptor-binding domain (RBD) of the toxin. RBD-like regions are conserved in all other clostridial glycosylating toxins preceding their CROP domain. CROP-deficient C. difficile toxin B is toxic to cells, depending on the RBD-like region (residues 1349-1811) but does not interact with LRP1. Our data indicate the presence of a second, CROP-independent receptor-binding domain in clostridial glycosylating toxins and suggest a two-receptor model for the cellular uptake of clostridial glycosylating toxins.


Subject(s)
Bacterial Toxins/metabolism , Clostridium perfringens/metabolism , Low Density Lipoprotein Receptor-Related Protein-1/metabolism , Animals , Bacterial Toxins/chemistry , Cell Membrane/metabolism , Embryo, Mammalian/cytology , Endocytosis , Fibroblasts/metabolism , Genetic Testing , Glycosylation , Haploidy , HeLa Cells , Humans , Mice , Models, Biological , Protein Binding , Protein Structure, Tertiary
13.
Cereb Cortex ; 25(10): 3446-57, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25085881

ABSTRACT

Cajal-Retzius (CR) cells are essential for cortical development and lamination. These pioneer neurons arise from distinct progenitor sources, including the cortical hem and the ventral pallium at pallium-subpallium boundary (PSB). CXCR4, the canonical receptor for the chemokine CXCL12, controls the superficial location of hem-derived CR cells. However, recent studies showed that CXCR7, a second CXCL12 receptor, is also expressed in CR cells at early developmental stages. We thus investigated the role of CXCR7 during CR cell development using multiple loss-of-function approaches. Cxcr7 gene inactivation led to aberrant localization of Reelin-positive cells within the pallium. In addition, Cxcr7(-/-) mice were characterized by significant accumulation of ectopic CR cells in the lateral part of the dorsal pallium compared with Cxcr4 knockout mice. Loss-of-function approaches, using either gene targeting or pharmacological receptor inhibition, reveal that CXCR7 and CXCR4 act both in CR positioning. Finally, conditional Cxcr7 deletion in cells derived from Dbx1-expressing progenitors indicates an essential role of CXCR7 in controlling the positioning of a subpopulation of PSB-derived CR cells. Our data demonstrate that CXCR7 has a role in the positioning of hem and PSB-derived CR cells, CXCL12 regulating CR cell subpial localization through the combined action of CXCR4 and CXCR7.


Subject(s)
Cell Movement , Cerebral Cortex/embryology , Neurons/physiology , Receptors, CXCR/metabolism , Animals , Cell Adhesion Molecules, Neuronal/metabolism , Cerebral Cortex/cytology , Cerebral Cortex/metabolism , Extracellular Matrix Proteins/metabolism , Mice , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Receptors, CXCR4/metabolism , Reelin Protein , Serine Endopeptidases/metabolism , Signal Transduction
14.
Cereb Cortex ; 24(11): 3046-58, 2014 Nov.
Article in English | MEDLINE | ID: mdl-23803971

ABSTRACT

GABAergic inhibitory interneurons (IN) represent a heterogeneous population with different electrophysiological, morphological, and molecular properties. The correct balance between interneuronal subtypes is important for brain function and is impaired in several neurological and psychiatric disorders. Here we show the data of 123 molecularly and electrophysiologically characterized neurons of juvenile rat barrel cortex acute slices, 48 of which expressed Reelin (Reln). Reln mRNA was exclusively detected in Gad65/67-positive cells but was found in interneuronal subtypes in different proportions: all cells of the adapting-Somatostatin (SST) cluster expressed Reln, whereas 63% of the adapting-neuropeptide Y (NPY, 50% of the fast-spiking Parvalbumin (PVALB), and 27% of the adapting/bursting-Vasoactive Intestinal Peptide (VIP) cluster were Reln-positive. Silhouette analysis revealed a high impact of the parameter Reln on cluster quality. By analyzing the co-localization of RELN immunoreactivity with those of different IN-markers, we found that RELN is produced layer-independently in SST-, NPY-, and NOS1-expressing INs, whereas co-localization of RELN and VIP was mostly absent. Of note, RELN co-localized with PVALB, predominantly in INs of layers IV/V (>30%). Our findings emphasize RELN's role as an important IN-marker protein and provide a basis for the functional characterization of Reln-expressing INs and its role in the regulation of inhibitory IN networks.


Subject(s)
Cell Adhesion Molecules, Neuronal/metabolism , Extracellular Matrix Proteins/metabolism , Interneurons/physiology , Nerve Tissue Proteins/metabolism , Neural Inhibition/physiology , Serine Endopeptidases/metabolism , Somatosensory Cortex/cytology , Animals , Animals, Newborn , Cell Adhesion Molecules, Neuronal/genetics , Cell Count , Cluster Analysis , Extracellular Matrix Proteins/genetics , Membrane Potentials/physiology , Nerve Tissue Proteins/genetics , Patch-Clamp Techniques , RNA, Messenger/metabolism , Rats , Rats, Wistar , Reelin Protein , Serine Endopeptidases/genetics , gamma-Aminobutyric Acid/metabolism
15.
J Biol Chem ; 288(30): 21909-23, 2013 Jul 26.
Article in English | MEDLINE | ID: mdl-23760271

ABSTRACT

The lipoprotein receptor LRP1 is essential in neurons of the central nervous system, as was revealed by the analysis of conditional Lrp1-deficient mouse models. The molecular basis of its neuronal functions, however, is still incompletely understood. Here we show by immunocytochemistry, electron microscopy, and postsynaptic density preparation that LRP1 is located postsynaptically. Basal and NMDA-induced phosphorylation of the transcription factor cAMP-response element-binding protein (CREB) as well as NMDA target gene transcription are reduced in LRP1-deficient neurons. In control neurons, NMDA promotes γ-secretase-dependent release of the LRP1 intracellular domain (LRP1-ICD). However, pull-down and chromatin immunoprecipitation (ChIP) assays showed no direct interaction between the LRP1-ICD and either CREB or target gene promoters. On the other hand, NMDA-induced degradation of the postsynaptic scaffold protein PSD-95 was impaired in the absence of LRP1, whereas its ubiquitination was increased, indicating that LRP1 influences the composition of postsynaptic protein complexes. Accordingly, NMDA-induced internalization of the AMPA receptor subunit GluA1 was impaired in LRP1-deficient neurons. These results show a role of LRP1 in the regulation and turnover of synaptic proteins, which may contribute to the reduced dendritic branching and to the neurological phenotype observed in the absence of LRP1.


Subject(s)
Guanylate Kinases/metabolism , Membrane Proteins/metabolism , Receptors, LDL/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Signal Transduction/physiology , Tumor Suppressor Proteins/metabolism , Amyloid Precursor Protein Secretases/metabolism , Animals , Blotting, Western , Cell Survival/genetics , Cell Survival/physiology , Cells, Cultured , Disks Large Homolog 4 Protein , Embryo, Mammalian/cytology , Female , Gene Expression/drug effects , Low Density Lipoprotein Receptor-Related Protein-1 , Male , Mice , Mice, Knockout , Mice, Transgenic , N-Methylaspartate/metabolism , N-Methylaspartate/pharmacology , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Protein Binding , Protein Subunits/metabolism , Receptors, LDL/deficiency , Receptors, LDL/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Signal Transduction/genetics , Synapses/drug effects , Synapses/metabolism , Synapses/physiology , Synaptosomes/metabolism , Tumor Suppressor Proteins/deficiency , Tumor Suppressor Proteins/genetics
16.
Glia ; 61(8): 1347-63, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23828756

ABSTRACT

During dentate gyrus development, the early embryonic radial glial scaffold is replaced by a secondary glial scaffold around birth. In contrast to neocortical and early dentate gyrus radial glial cells, these postnatal glial cells are severely altered with regard to position and morphology in reeler mice lacking the secreted protein Reelin. In this study, we focus on the functional impact of these defects. Most radial glial cells throughout the nervous system serve as scaffolds for migrating neurons and precursor cells for both neurogenesis and gliogenesis. Precursor cell function has been demonstrated for secondary radial glial cells but the exact function of these late glial cells in granule cell migration and positioning is not clear. No data exist concerning the interplay between granule neurons and late radial glial cells during dentate gyrus development. Herein, we show that despite the severe morphological defects in the reeler dentate gyrus, the precursor function of secondary radial glial cells is not impaired during development in reeler mice. In addition, selective ablation of Disabled-1, an intracellular adaptor protein essential for Reelin signaling, in neurons but not in glial cells allowed us to distinguish effects of Reelin signaling on radial glial cells from possible secondary effects based on defective granule cells positioning.


Subject(s)
Cell Adhesion Molecules, Neuronal/deficiency , Dentate Gyrus/metabolism , Ependymoglial Cells/physiology , Extracellular Matrix Proteins/deficiency , Mutation , Nerve Tissue Proteins/deficiency , Serine Endopeptidases/deficiency , Signal Transduction/genetics , Animals , Animals, Newborn , Cell Adhesion Molecules, Neuronal/biosynthesis , Cells, Cultured , Dentate Gyrus/growth & development , Extracellular Matrix Proteins/biosynthesis , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Mice, Neurologic Mutants , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Phenotype , Recombinant Proteins/biosynthesis , Reelin Protein , Serine Endopeptidases/biosynthesis , Stem Cells/metabolism
17.
Cell Tissue Res ; 354(3): 887-9, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23963646

ABSTRACT

We have previously reported that apolipoprotein E (apoE), a protein component of very-low-density lipoproteins (VLDL) and high-density lipoproteins and a potent plasma-borne atheroprotective factor, exerts anti-inflammatory activity in macrophages by switching the activation profile from M1 ("classic") to M2 ("alternative") in a process involving signaling via low-density lipoprotein receptor (LDLR) family members including the VLDL receptor (VLDLR) or apoE receptor-2 (apoER2). The present study was undertaken to investigate whether LDLR-related protein 1 (LRP-1), another member of the LDLR family and a ubiquitously expressed multifunctional cell surface receptor, modulates M1→M2 conversion in murine macrophages. We investigate bone marrow or peritoneal macrophages isolated from wild-type C57/Bl6 mice or mice with conditional inactivation of the LRP-1 gene in the myeloid lineage for the expression of polarization markers. Our results suggest that the deficiency of LRP-1 down-regulates M2 marker expression in macrophages, while enhancing the macrophage response to M1 stimuli. To our knowledge, this is the first demonstration that LRP-1 affects macrophage polarization and promotes the development of an anti-inflammatory M2 functional phenotype.


Subject(s)
Macrophages/metabolism , Receptors, LDL/biosynthesis , Tumor Suppressor Proteins/biosynthesis , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Gene Silencing , Low Density Lipoprotein Receptor-Related Protein-1 , Macrophages/cytology , Macrophages, Peritoneal/cytology , Macrophages, Peritoneal/metabolism , Mice , Mice, Inbred C57BL , Phenotype , Receptors, LDL/genetics , Signal Transduction , Tumor Suppressor Proteins/genetics
18.
J Neurosci ; 31(7): 2352-60, 2011 Feb 16.
Article in English | MEDLINE | ID: mdl-21325502

ABSTRACT

The extracellular matrix molecule Reelin is known to control neuronal migration during development. Recent evidence suggests that it also plays a role in the maturation of postsynaptic dendrites and spines as well as in synaptic plasticity. Here, we aimed to address the question whether Reelin plays a role in presynaptic structural organization and function. Quantitative electron microscopic analysis of the number of presynaptic boutons in the stratum radiatum of hippocampal region CA1 did not reveal differences between wild-type animals and Reelin-deficient reeler mutant mice. However, additional detailed analysis showed that the number of presynaptic vesicles was significantly increased in CA1 synapses of reeler mutants. To test the hypothesis that vesicle fusion is altered in reeler, we studied proteins known to control transmitter release. SNAP25, a protein of the soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) complex, was found to be significantly reduced in reeler mutants, whereas other SNARE complex proteins remained unaltered. Addition of recombinant Reelin to organotypic slice cultures of reeler hippocampi substantially rescued not only SNAP25 protein expression levels but also the number of vesicles per bouton area indicating a role for Reelin in presynaptic functions. Next, we analyzed paired-pulse facilitation, a presynaptic mechanism associated with transmitter release, and observed a significant decrease at CA1 synapses of reeler mutants when compared with wild-type animals. Together, these novel findings suggest a role for Reelin in modulating presynaptic release mechanisms.


Subject(s)
Cell Adhesion Molecules, Neuronal/physiology , Extracellular Matrix Proteins/physiology , Nerve Tissue Proteins/physiology , Neurotransmitter Agents/metabolism , Presynaptic Terminals/metabolism , Serine Endopeptidases/physiology , Action Potentials/drug effects , Action Potentials/genetics , Animals , Animals, Newborn , Antibodies/pharmacology , CA1 Region, Hippocampal/cytology , Cell Adhesion Molecules, Neuronal/deficiency , Cell Adhesion Molecules, Neuronal/immunology , Cell Adhesion Molecules, Neuronal/pharmacology , Cell Line, Transformed , Clathrin/metabolism , Culture Media, Conditioned/pharmacology , Electron Microscope Tomography/methods , Enzyme Inhibitors/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/genetics , Extracellular Matrix Proteins/deficiency , Extracellular Matrix Proteins/immunology , Extracellular Matrix Proteins/pharmacology , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Green Fluorescent Proteins/genetics , Humans , In Vitro Techniques , Integrin beta1/metabolism , LDL-Receptor Related Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation/genetics , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/immunology , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/pharmacology , Patch-Clamp Techniques , Presynaptic Terminals/ultrastructure , R-SNARE Proteins/metabolism , Receptors, LDL/genetics , Reelin Protein , Serine Endopeptidases/deficiency , Serine Endopeptidases/immunology , Serine Endopeptidases/pharmacology , Synapses/drug effects , Synapses/metabolism , Synapses/ultrastructure , Synaptic Vesicles/metabolism , Synaptic Vesicles/ultrastructure , Synaptosomal-Associated Protein 25/metabolism , Transfection/methods
19.
Arterioscler Thromb Vasc Biol ; 31(5): 1160-8, 2011 May.
Article in English | MEDLINE | ID: mdl-21350196

ABSTRACT

OBJECTIVE: Apolipoprotein E (apoE) exerts potent antiinflammatory effects. Here, we investigated the effect of apoE on the functional phenotype of macrophages. METHODS AND RESULTS: Human apoE receptors very-low-density lipoprotein receptor (VLDL-R) and apoE receptor-2 (apoER2) were stably expressed in RAW264.7 mouse macrophages. In these cells, apoE downregulated markers of the proinflammatory M1 phenotype (inducible nitric oxide synthase, interleukin [IL]-12, macrophage inflammatory protein-1α) but upregulated markers of the antiinflammatory M2 phenotype (arginase I, SOCS3, IL-1 receptor antagonist [IL-1RA]). In addition, M1 macrophage responses (migration, generation of reactive oxygen species, antibody-dependent cell cytotoxicity, phagocytosis), as well as poly(I:C)- or interferon-γ-induced production of proinflammatory cytokines; cyclooxygenase-2 expression; and activation of nuclear factor-κB, IκB, and STAT1, were suppressed in VLDL-R- or apoER2-expressing cells. Conversely, the suppression of the M2 phenotype and the enhanced response to poly(I:C) were observed in apoE-producing bone marrow macrophages derived from VLDL-R-deficient mice but not wild-type or low-density lipoprotein receptor-deficient mice. The modulatory effects of apoE on macrophage polarization were inhibited in apoE receptor-expressing RAW264.7 cells exposed to SB220025, a p38 mitogen-activated protein kinase inhibitor, and PP1, a tyrosine kinase inhibitor. Accordingly, apoE induced tyrosine kinase-dependent activation of p38 mitogen-activated protein kinase in VLDL-R- or apoER2-expressing macrophages. Under in vivo conditions, apoE-/- mice transplanted with apoE-producing wild-type bone marrow showed increased plasma IL-1RA levels, and peritoneal macrophages of transplanted animals were shifted to the M2 phenotype (increased IL-1RA production and CD206 expression). CONCLUSIONS: ApoE signaling via VLDL-R or apoER2 promotes macrophage conversion from the proinflammatory M1 to the antiinflammatory M2 phenotype. This effect may represent a novel antiinflammatory activity of apoE.


Subject(s)
Apolipoproteins E/metabolism , Inflammation/prevention & control , Macrophages/metabolism , Animals , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Bone Marrow Transplantation , Cell Line , Female , Genotype , Inflammation/immunology , Inflammation/metabolism , Inflammation Mediators/metabolism , Interferon-gamma/metabolism , LDL-Receptor Related Proteins/deficiency , LDL-Receptor Related Proteins/genetics , Macrophages/drug effects , Macrophages/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Poly I-C/pharmacology , Protein Kinase Inhibitors/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/metabolism , Receptors, LDL/deficiency , Receptors, LDL/genetics , Signal Transduction , Time Factors , Transfection , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/metabolism
20.
Front Immunol ; 13: 1045498, 2022.
Article in English | MEDLINE | ID: mdl-36439181

ABSTRACT

Background and aims: There is growing interest in T cell-based immune therapies for a functional cure of chronic HBV infection including check-point inhibition, T cell-targeted vaccines or TCR-grafted effector cells. All these approaches depend on recognition of HLA class I-presented viral peptides. The HBV core region 18-27 is an immunodominant target of CD8+ T cells and represents the prime target for T cell-based therapies. Here, a high-resolution analysis of the core18-27 specific CD8+ T cell and the selected escape pathways was performed. Methods: HLA class I typing and viral sequence analyses were performed for 464 patients with chronic HBV infection. HBV-specific CD8+ T-cell responses against the prototype and epitope variants were characterized by flow cytometry. Results: Consistent with promiscuous presentation of the core18-27 epitope, antigen-specific T cells were detected in patients carrying HLA-A*02:01, HLA-B*35:01, HLA-B*35:03 or HLA-B*51:01. Sequence analysis confirmed reproducible selection pressure on the core18-27 epitope in the context of these alleles. Interestingly, the selected immune escape pathways depend on the presenting HLA-class I-molecule. Although cross-reactive T cells were observed, some epitope variants achieved functional escape by impaired TCR-interaction or disturbed antigen processing. Of note, selection of epitope variants was exclusively observed in HBeAg negative HBV infection and here, detection of variants associated with significantly greater magnitude of the CD8 T cell response compared to absence of variants. Conclusion: The core18-27 epitope is highly variable and under heavy selection pressure in the context of different HLA class I-molecules. Some epitope variants showed evidence for impaired antigen processing and reduced presentation. Viruses carrying such escape substitutions will be less susceptible to CD8+ T cell responses and should be considered for T cell-based therapy strategies.


Subject(s)
CD8-Positive T-Lymphocytes , Hepatitis B virus , Humans , Alleles , Hepatitis B virus/genetics , HLA-B Antigens/genetics , Epitopes , Receptors, Antigen, T-Cell/genetics
SELECTION OF CITATIONS
SEARCH DETAIL