Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters











Publication year range
1.
Front Mol Neurosci ; 12: 224, 2019.
Article in English | MEDLINE | ID: mdl-31616248

ABSTRACT

The endocannabinoid system (ECS) consists particularly of cannabinoid receptors 1 and 2 (CB1 and CB2), their endogenous ligands, and enzymes that synthesize and degrade their ligands. It acts in a variety of organs and disease states ranging from cancer progression over neuropathic pain to neurodegeneration. Protein components engaged in the signaling, trafficking, and homeostasis machinery of the G-protein coupled CB2, are however largely unknown. It is therefore important to identify further interaction partners to better understand CB2 receptor functions in physiology and pathophysiology. For this purpose, we used an affinity purification and mass spectrometry-based proteomics approach of Strep-HA-CB2 receptor in HEK293 cells. After subtraction of background interactions and protein frequency library assessment we could identify 83 proteins that were classified by the identification of minimally 2 unique peptides as highly probable interactors. A functional protein association network analysis obtained an interaction network with a significant enrichment of proteins functionally involved in protein metabolic process, in endoplasmic reticulum, response to stress but also in lipid metabolism and membrane organization. The network especially contains proteins involved in biosynthesis and trafficking like calnexin, Sec61A, tubulin chains TUBA1C and TUBB2B, TMED2, and TMED10. Six proteins that were only expressed in stable CB2 expressing cells were DHC24, DHRS7, GGT7, HECD3, KIAA2013, and PLS1. To exemplify the validity of our approach, we chose a candidate having a relatively low number of edges in the network to increase the likelihood of a direct protein interaction with CB2 and focused on the scaffold/phagosomal protein p62/SQSTM1. Indeed, we independently confirmed the interaction by co-immunoprecipitation and immunocytochemical colocalization studies. 3D reconstruction of confocal images furthermore showed CB2 localization in close proximity to p62 positive vesicles at the cell membrane. In summary, we provide a comprehensive repository of the CB2 interactome in HEK293 cells identified by a systematic unbiased approach, which can be used in future experiments to decipher the signaling and trafficking complex of this cannabinoid receptor. Future studies will have to analyze the exact mechanism of the p62-CB2 interaction as well as its putative role in disease pathophysiology.

2.
Int J Mol Sci ; 20(15)2019 Aug 01.
Article in English | MEDLINE | ID: mdl-31374821

ABSTRACT

The G-protein coupled cannabinoid receptor 2 (CB2) has been implicated in the regulation of adult neurogenesis in the hippocampus. The contribution of CB2 towards basal levels of proliferation and the number of neural progenitors in the subgranular zone (SGZ) of the dentate gyrus, however, remain unclear. We stained hippocampal brain sections of 16- to 17-week-old wildtype and CB2-deficient mice, for neural progenitor and immature neuron markers doublecortin (DCX) and calretinin (CR) and for the proliferation marker Ki67 and quantified the number of positive cells in the SGZ. The quantification revealed that CB2 deficiency neither altered overall cell proliferation nor the size of the DCX+ or DCX and CR double-positive populations in the SGZ compared to control animals. The results indicate that CB2 might not contribute to basal levels of adult neurogenesis in four-month-old healthy mice. CB2 signaling might be more relevant in conditions where adult neurogenesis is dynamically regulated, such as neuroinflammation.


Subject(s)
Hippocampus/physiology , Neurogenesis , Receptor, Cannabinoid, CB2/genetics , Animals , Cell Proliferation , Doublecortin Protein , Female , Gene Deletion , Hippocampus/cytology , Mice , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neurons/cytology , Neurons/metabolism
3.
J Neurochem ; 135(4): 830-44, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26285062

ABSTRACT

In humans, deletions/mutations in the CHL1/CALL gene are associated with mental retardation and schizophrenia. Juvenile CHL1-deficient (CHL1(-/-) ) mice have been shown to display abnormally high numbers of parvalbumin-expressing (PV(+) ) hippocampal interneurons and, as adults, display behavioral traits observed in neuropsychiatric disorders. Here, we addressed the question whether inhibitory interneurons and synaptic plasticity in the CHL1(-/-) mouse are affected during brain maturation and in adulthood. We found that hippocampal, but not neocortical, PV(+) interneurons were reduced with age in CHL1(-/-) mice, from a surplus of +27% at 1 month to a deficit of -20% in adulthood compared with wild-type littermates. This loss occurred during brain maturation, correlating with microgliosis and enhanced interleukin-6 expression. In parallel with the loss of PV(+) interneurons, the inhibitory input to adult CA1 pyramidal cells was reduced and a deficit in short- and long-term potentiation developed at CA3-CA1 excitatory synapses between 2 and 9 months of age in CHL1(-/-) mice. This deficit could be abrogated by a GABAA receptor agonist. We propose that region-specific aberrant GABAergic synaptic connectivity resulting from the mutation and a subsequently enhanced synaptic elimination during brain maturation lead to microgliosis, increase in pro-inflammatory cytokine levels, loss of interneurons, and impaired synaptic plasticity. Close homolog of L1-deficient (CHL1(-/-) ) mice have abnormally high numbers of parvalbumin (PV)-expressing hippocampal interneurons in juvenile animals, but in adult animals a loss of these cells is observed. This loss correlates with an increased density of microglia (M), enhanced interleukin-6 (IL6) production and a deficit in short- and long-term potentiation at CA3-CA1 excitatory synapses. Furthermore, adult CHL1(-/-) mice display behavioral traits similar to those observed in neuropsychiatric disorders of humans.


Subject(s)
Aging , Cell Adhesion Molecules/deficiency , Gene Expression Regulation/genetics , Hippocampus/cytology , Interneurons/metabolism , Parvalbumins/metabolism , Animals , Calcium-Binding Proteins/metabolism , Cell Adhesion Molecules/genetics , Cerebellum , Enzyme-Linked Immunosorbent Assay , Excitatory Postsynaptic Potentials/genetics , In Vitro Techniques , Interleukin-3/metabolism , Interleukin-6/metabolism , Mice , Mice, Transgenic , Microfilament Proteins/metabolism , Microscopy, Electron , Patch-Clamp Techniques , Phosphopyruvate Hydratase/metabolism , S100 Proteins/metabolism , Synapses/metabolism , Synapses/ultrastructure
4.
Mol Ther ; 23(6): 993-1002, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25807288

ABSTRACT

Failure of the mammalian central nervous system (CNS) to regenerate effectively after injury leads to mostly irreversible functional impairment. Gold nanoparticles (AuNPs) are promising candidates for drug delivery in combination with tissue-compatible reagents, such as polyethylene glycol (PEG). PEG administration in CNS injury models has received interest for potential therapy, but toxicity and low bioavailability prevents clinical application. Here we show that intraspinal delivery of PEG-functionalized 40-nm-AuNPs at early stages after mouse spinal cord injury is beneficial for recovery. Positive outcome of hind limb motor function was accompanied by attenuated inflammatory response, enhanced motor neuron survival, and increased myelination of spared or regrown/sprouted axons. No adverse effects, such as body weight loss, ill health, or increased mortality were observed. We propose that PEG-AuNPs represent a favorable drug-delivery platform with therapeutic potential that could be further enhanced if PEG-AuNPs are used as carriers of regeneration-promoting molecules.


Subject(s)
Coated Materials, Biocompatible/pharmacology , Drug Delivery Systems/methods , Gold/pharmacology , Metal Nanoparticles/chemistry , Polyethylene Glycols/pharmacology , Spinal Cord Injuries/drug therapy , Animals , Coated Materials, Biocompatible/chemistry , Disease Models, Animal , Female , Gold/chemistry , Mice , Mice, Inbred C57BL , Polyethylene Glycols/chemistry , Recovery of Function/drug effects
5.
Neurobiol Dis ; 56: 104-15, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23639788

ABSTRACT

Alzheimer's disease (AD) is a devastating neurodegenerative disorder and the most common cause of elderly dementia. In an effort to contribute to the potential of molecular approaches to reduce degenerative processes we have tested the possibility that the neural adhesion molecule L1 ameliorates some characteristic cellular and molecular parameters associated with the disease in a mouse model of AD. Three-month-old mice overexpressing mutated forms of amyloid precursor protein and presenilin-1 under the control of a neuron-specific promoter received an injection of adeno-associated virus encoding the neuronal isoform of full-length L1 (AAV-L1) or, as negative control, green fluorescent protein (AAV-GFP) into the hippocampus and occipital cortex. Four months after virus injection, the mice were analyzed for histological and biochemical parameters of AD. AAV-L1 injection decreased the Aß plaque load, levels of Aß42, Aß42/40 ratio and astrogliosis compared with AAV-GFP controls. AAV-L1 injected mice also had increased densities of inhibitory synaptic terminals on pyramidal cells in the hippocampus when compared with AAV-GFP controls. Numbers of microglial cells/macrophages were similar in both groups, but numbers of microglial cells/macrophages per plaque were increased in AAV-L1 injected mice. To probe for a molecular mechanism that may underlie these effects, we analyzed whether L1 would directly and specifically interact with Aß. In a label-free binding assay, concentration dependent binding of the extracellular domain of L1, but not of the close homolog of L1 to Aß40 and Aß42 was seen, with the fibronectin type III homologous repeats 1-3 of L1 mediating this effect. Aggregation of Aß42 in vitro was reduced in the presence of the extracellular domain of L1. The combined observations indicate that L1, when overexpressed in neurons and glia, reduces several histopathological hallmarks of AD in mice, possibly by reduction of Aß aggregation. L1 thus appears to be a candidate molecule to ameliorate the pathology of AD, when applied in therapeutically viable treatment schemes.


Subject(s)
Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Neural Cell Adhesion Molecule L1/metabolism , Neural Cell Adhesion Molecule L1/therapeutic use , Alzheimer Disease/pathology , Animals , Blotting, Western , Brain/pathology , Dependovirus/genetics , Enzyme-Linked Immunosorbent Assay , Gliosis/pathology , Green Fluorescent Proteins , Hippocampus/metabolism , Hippocampus/pathology , Humans , Immunohistochemistry , Male , Mice , Mice, Transgenic , Microglia/drug effects , Occipital Lobe/metabolism , Occipital Lobe/pathology , Plaque, Amyloid/pathology , Protein Binding , Pyramidal Cells/drug effects , Receptors, CCR2/metabolism , Tissue Fixation
6.
PLoS One ; 8(4): e61299, 2013.
Article in English | MEDLINE | ID: mdl-23585889

ABSTRACT

Dysregulation of the proteolytic processing of amyloid precursor protein by γ-secretase and the ensuing generation of amyloid-ß is associated with the pathogenesis of Alzheimer's disease. Thus, the identification of amyloid precursor protein binding proteins involved in regulating processing of amyloid precursor protein by the γ-secretase complex is essential for understanding the mechanisms underlying the molecular pathology of the disease. We identified calreticulin as novel amyloid precursor protein interaction partner that binds to the γ-secretase cleavage site within amyloid precursor protein and showed that this Ca(2+)- and N-glycan-independent interaction is mediated by amino acids 330-344 in the C-terminal C-domain of calreticulin. Co-immunoprecipitation confirmed that calreticulin is not only associated with amyloid precursor protein but also with the γ-secretase complex members presenilin and nicastrin. Calreticulin was detected at the cell surface by surface biotinylation of cells overexpressing amyloid precursor protein and was co-localized by immunostaining with amyloid precursor protein and presenilin at the cell surface of hippocampal neurons. The P-domain of calreticulin located between the N-terminal N-domain and the C-domain interacts with presenilin, the catalytic subunit of the γ-secretase complex. The P- and C-domains also interact with nicastrin, another functionally important subunit of this complex. Transfection of amyloid precursor protein overexpressing cells with full-length calreticulin leads to a decrease in amyloid-ß42 levels in culture supernatants, while transfection with the P-domain increases amyloid-ß40 levels. Similarly, application of the recombinant P- or C-domains and of a synthetic calreticulin peptide comprising amino acid 330-344 to amyloid precursor protein overexpressing cells result in elevated amyloid-ß40 and amyloid-ß42 levels, respectively. These findings indicate that the interaction of calreticulin with amyloid precursor protein and the γ-secretase complex regulates the proteolytic processing of amyloid precursor protein by the γ-secretase complex, pointing to calreticulin as a potential target for therapy in Alzheimer's disease.


Subject(s)
Amyloid Precursor Protein Secretases/genetics , Amyloid beta-Protein Precursor/metabolism , Membrane Glycoproteins/genetics , Presenilin-1/genetics , Amino Acid Sequence , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/metabolism , Animals , Binding Sites , CHO Cells , Cricetinae , Gene Expression Regulation , Hippocampus/cytology , Hippocampus/metabolism , Humans , Membrane Glycoproteins/metabolism , Mice , Molecular Sequence Data , Neurons/cytology , Neurons/metabolism , Peptide Fragments/metabolism , Presenilin-1/metabolism , Protein Binding , Protein Structure, Tertiary , Proteolysis , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Signal Transduction
7.
Exp Neurol ; 237(2): 274-85, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22868200

ABSTRACT

It is widely accepted that the immune system plays important functional roles in regeneration after injury to the spinal cord. Immune response towards injury involves a complex interplay of immune system cells, such as neutrophils, macrophages and microglia, T- and B-lymphocytes. We investigated the influence of the lymphocyte component of the immune system on the locomotor outcome of severe spinal cord injury in a genetic mouse model of immune suppression. Transgenic mice lacking mature T- and B-lymphocytes due to the recombination activating gene 2 gene deletion (RAG2-/- mice) were subjected to severe compression of the lower thoracic spinal cord, with the wild-type mice of the same inbred background serving as controls. According to both the Basso Mouse Scale score and single frame motion analysis, the RAG2-/- mice showed improved recovery in comparison to control mice at six weeks after injury. Better locomotor function was associated with enhanced catecholaminergic and cholinergic reinnervation of the spinal cord caudal to injury and increased axonal regrowth/sprouting at the site of injury. Myelination of axons in the ventral column measured as g-ratio was more extensive in RAG2-/- than in control mice 6weeks after injury. Additionally, the number of microglia/macrophages was decreased in the lumbar spinal cord of RAG2-/- mice after injury, whereas the number of astrocytes was increased compared with controls. We conclude that T- and B-lymphocytes restrict functional recovery from spinal cord injury by increasing numbers of microglia/macrophages as well as decreasing axonal sprouting and myelination.


Subject(s)
B-Lymphocytes/immunology , Nerve Regeneration/immunology , Recovery of Function/immunology , Spinal Cord Injuries/immunology , T-Lymphocytes/immunology , Animals , Blotting, Western , Disease Models, Animal , Female , Immunohistochemistry , Mice , Mice, Inbred C57BL , Mice, Transgenic , Motor Activity/physiology , Spinal Cord Injuries/pathology
8.
J Biol Chem ; 287(21): 17161-17175, 2012 May 18.
Article in English | MEDLINE | ID: mdl-22431726

ABSTRACT

The functions of the cell adhesion molecule L1 in the developing and adult nervous system are triggered by homophilic and heterophilic interactions that stimulate signal transductions that activate cellular responses. Here, we show that stimulation of signaling by function-triggering L1 antibodies or L1-Fc leads to serine protease-dependent cleavage of full-length L1 at the plasma membrane and generation of a sumoylated transmembrane 70-kDa fragment comprising the intracellular and transmembrane domains and part of the extracellular domain. The 70-kDa transmembrane fragment is transported from the plasma membrane to a late endosomal compartment, released from endosomal membranes into the cytoplasm, and transferred from there into the nucleus by a pathway that depends on importin and chromatin-modifying protein 1. Mutation of the sumoylation site at Lys(1172) or of the nuclear localization signal at Lys(1147) abolished L1-stimulated generation or nuclear import of the 70-kDa fragment, respectively. Nuclear import of the 70-kDa fragment may activate cellular responses in parallel or in association with phosphorylation-dependent signaling pathways. Alterations in the levels of the 70-kDa fragment during development and in the adult after spinal cord injury or in a mouse model of Alzheimer disease suggest that this fragment is functionally implicated in development, regeneration, neurodegeneration, tumorigenesis, and possibly synaptic plasticity in the mature nervous system.


Subject(s)
Cell Nucleus/metabolism , Nerve Tissue Proteins/metabolism , Neural Cell Adhesion Molecule L1/metabolism , Signal Transduction , Sumoylation , Active Transport, Cell Nucleus/genetics , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Animals , Cell Nucleus/genetics , Disease Models, Animal , Endosomes/genetics , Endosomes/metabolism , HEK293 Cells , Humans , Intracellular Membranes/metabolism , Mice , Mutation , Neural Cell Adhesion Molecule L1/genetics , Nuclear Localization Signals/genetics , Nuclear Localization Signals/metabolism , Protein Structure, Tertiary , Spinal Cord/embryology , Spinal Cord/metabolism , Spinal Cord Injuries/genetics , Spinal Cord Injuries/metabolism
9.
Anat Rec (Hoboken) ; 292(12): 1882-92, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19943341

ABSTRACT

Magnetic resonance imaging (MRI) is the only noninvasive technique that provides structural information on both cell loss and metabolic changes. After reviewing all the results obtained in clinical studies, reliable biomarkers in neurological diseases are still lacking. Diffusional MRI, MR spectroscopy, and the assessment of regional atrophy are promising approaches, but they cannot be simultaneously used on a single patient. Thus, for further research progress, reliable animal models are needed. To this aim, we have used the clinical MRI to assess neurodegenerative processes in the hSOD-1(G93A) ALS rat model and in the trimethyltin (TMT)-treated model of Alzheimer's-like disease. T2-weighted (T2W) hyperintensive neurodegenerative foci were found in the brainstem of the ALS rat with apparent lateral ventricle dilation (T1W-hypointensity vs. T2W-hyperintensity). Degenerative processes in these areas were also confirmed by confocal images of GFAP-positive astrogliosis. MRI after i.v.i. of magnetic anti-CD4 antibodies indicated an accumulation of inflammatory cells near dilated ventricles. TMT-treated rats also revealed the dilation of lateral ventricles. Expected deterioration in the hippocampus was not observed by clinical MRI, but immunocytochemistry could reveal significant redistribution of macro- and microglia in this structure. In both models, Gd-DTPA contrast revealed a compromised blood brain barrier that may serve as the passage for inflammatory immune cells in the vicinity of dilated lateral ventricles. Moreover, in both models the midbrain region of the dorsal hippocampus was the target of BBB compromise, thus revealing a potentially vulnerable point that can be the primary target of neurodegeneration in the central nervous system.


Subject(s)
Alzheimer Disease/pathology , Amyotrophic Lateral Sclerosis/pathology , Brain/pathology , Magnetic Resonance Imaging/methods , Alzheimer Disease/metabolism , Alzheimer Disease/physiopathology , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/physiopathology , Animals , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/pathology , Blood-Brain Barrier/physiopathology , Brain/metabolism , Brain/physiopathology , Chemotaxis, Leukocyte/immunology , Disease Models, Animal , Encephalitis/metabolism , Encephalitis/pathology , Encephalitis/physiopathology , Gliosis/metabolism , Gliosis/pathology , Gliosis/physiopathology , Humans , Nerve Degeneration/metabolism , Nerve Degeneration/pathology , Nerve Degeneration/physiopathology , Rats
10.
Gen Physiol Biophys ; 28 Spec No: 212-8, 2009.
Article in English | MEDLINE | ID: mdl-19893103

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a devastating neurological disorder affecting upper and lower motoneurons. The transgenic ALS rat model (hSOD-1(G93A)) was used for magnetic resonance imaging (MRI) study using a low field wide bore magnet. T2-weighted hyperintensities were observed in the brainstem, rubrospinal tract and vagus motor nuclei with prominent lateral ventricle and cerebral aqueduct enlargements. These changes could be observed already in presymptomatic animals. T2*-weighted MRI with magnetically labeled antibodies (against CD4) revealed lymphocyte infiltration in the brainstem-midbrain region corresponding to the areas of dilated lateral ventricles. Confocal imaging revealed reactive astroglia in these areas. Thus, with the use of wide bore MRI new sites of neurodegeneration and inflammation were revealed in the hSOD-1(G93A) rat model.


Subject(s)
Amyotrophic Lateral Sclerosis/complications , Brain Diseases/diagnosis , Brain Diseases/pathology , Tissue Survival , Amyotrophic Lateral Sclerosis/pathology , Animals , Brain Diseases/complications , Disease Models, Animal , Disease Progression , Humans , Magnetic Resonance Imaging , Microscopy, Confocal , Rats , Rats, Sprague-Dawley
11.
J Comp Neurol ; 513(5): 496-510, 2009 Apr 10.
Article in English | MEDLINE | ID: mdl-19226508

ABSTRACT

Several L1-related adhesion molecules, expressed in a well-coordinated temporospatial pattern during development, are important for fine tuning of specific cerebellar circuitries. We tested the hypothesis that CHL1, the close homologue of L1, abundantly expressed in the developing and adult cerebellum, is also required for normal cerebellar histogenesis. We found that constitutive ablation of CHL1 in mice caused significant loss (20-23%) of Purkinje and granule cells in the mature 2-month-old cerebellum. The ratio of stellate/basket interneurons to Purkinje cells was abnormally high (+38%) in CHL1-deficient (CHL1-/-) mice compared with wild-type (CHL1+/+) littermates, but the gamma-aminobutyric acid (GABA)ergic synaptic inputs to Purkinje cell bodies and dendrites were normal, as were numbers of Golgi interneurons, microglia, astrocytes, and Bergmann glia. Purkinje cell loss occurred before the first postnatal week and was associated with enhanced apoptosis, presumably as a consequence of CHL1 deficiency in afferent axons. In contrast, generation of granule cells, as indicated by in vivo analyses of cell proliferation and death, was unaffected in 1-week-old CHL1-/- mice, but numbers of migrating granule cells in the molecular layer were increased. This increase was likely related to retarded cell migration because CHL1-/- granule cells migrated more slowly than CHL1+/+ cells in vitro, and Bergmann glial processes guiding migration in vivo expressed CHL1 in wild-type mice. Granule cell deficiency in adult CHL1-/- mice appeared to result from decreased precursor cell proliferation after the first postnatal week. Our results indicate that CHL1 promotes Purkinje and granule cell survival and granule cell migration during cerebellar development.


Subject(s)
Cell Adhesion Molecules/physiology , Cell Movement/physiology , Cerebellum/anatomy & histology , Cerebellum/growth & development , Neurons/physiology , Purkinje Cells/physiology , Animals , Animals, Newborn , Cell Adhesion Molecules/deficiency , Cell Adhesion Molecules/genetics , Cell Count/methods , Cell Death/physiology , Cell Differentiation/physiology , Cells, Cultured , Cerebellum/cytology , Dendrites/metabolism , Dendrites/physiology , Embryo, Mammalian , Female , Immunohistochemistry , Interneurons/metabolism , Interneurons/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neuroglia/metabolism , Neuroglia/physiology , Neurons/metabolism , Polymerase Chain Reaction , Pregnancy , Purkinje Cells/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL