Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Eur J Pharmacol ; 934: 175301, 2022 Nov 05.
Article in English | MEDLINE | ID: mdl-36191631

ABSTRACT

In this study we aimed to reduce tau pathology, a hallmark of Alzheimer's Disease (AD), by activating mTOR-dependent autophagy in a transgenic mouse model of tauopathy by long-term dosing of animals with mTOR-inhibitors. Rapamycin treatment reduced the burden of hyperphosphorylated and aggregated pathological tau in the cerebral cortex only when applied to young mice, prior to the emergence of pathology. Conversely, PQR530 which exhibits better brain exposure and superior pharmacokinetic properties, reduced tau pathology even when the treatment started after the onset of pathology. Our results show that dosing animals twice per week with PQR530 resulted in intermittent, rather than sustained target engagement. Nevertheless, this pulse-like mTOR inhibition followed by longer intervals of re-activation was sufficient to reduce tau pathology in the cerebral cortex in P301S tau transgenic mice. This suggests that balanced therapeutic dosing of blood-brain-barrier permeable mTOR-inhibitors can result in a disease-modifying effect in AD and at the same time prevents toxic side effects due to prolonged over activation of autophagy.


Subject(s)
Alzheimer Disease , Animals , Mice , Alzheimer Disease/drug therapy , Alzheimer Disease/pathology , tau Proteins/metabolism , TOR Serine-Threonine Kinases/metabolism , Mice, Transgenic , Brain , Sirolimus/pharmacology , Disease Models, Animal
2.
Knee Surg Sports Traumatol Arthrosc ; 23(8): 2406-2412, 2015 Aug.
Article in English | MEDLINE | ID: mdl-24562698

ABSTRACT

PURPOSE: To compare repaired cartilage with native cartilage, and inter-observer reliability, using T2 mapping at 3 T for assessing cartilage repair in osteochondral defects of the talus after the microfracture technique. METHODS: We enrolled eight females and seven males undergoing arthroscopic microfracture for osteochondral defects of the talus at an average follow-up of 7.9 ± 2.2 years (range 5-13 years). Cartilage tissue was assessed using a 3-T magnetic resonance imaging unit with an 8-channel phased array foot and ankle coil (gradient strength, 50 mT/m; slew rate, 200 T/m/s). T2 maps were then calculated. Three independent boarded specialists evaluated the images, and magnetic resonance observation of cartilage repair tissue scores was used to assess the cartilage and joint status. Clinical results were assessed using the Hannover Scoring System (HSS) for the ankle and the American Orthopaedic Foot and Ankle Society (AOFAS) hind-foot score. RESULTS: No significant mean differences were found between the T2 properties of the repair tissue and those of the native reference cartilage (T2 = 38.6 ± 5.3 ms, range 30.2-55.8 ms vs. 40.3 ± 8.5 ms, range 31.4-59.8 ms, respectively; intra-class correlation coefficient = 0.94; confidence interval 0.84-0.99, P ≤ 0.001). Despite ≥50 % defect filling in all patients, subchondral bone changes were considerable. The HSS at the follow-up revealed a mean score of 87 ± 12 (range 51-97), and the AOFAS-Score was 90 ± 13 (range 59-100). CONCLUSIONS: 3 T T2 maps were similar in repaired and native cartilage with good inter-observer reliability. LEVEL OF EVIDENCE: IV.


Subject(s)
Arthroplasty, Subchondral , Cartilage/pathology , Cartilage/surgery , Magnetic Resonance Imaging/methods , Talus/pathology , Talus/surgery , Adolescent , Adult , Cartilage/injuries , Female , Follow-Up Studies , Humans , Male , Middle Aged , Reproducibility of Results , Talus/injuries , Young Adult
3.
Exp Transl Stroke Med ; 6(1): 3, 2014 Feb 14.
Article in English | MEDLINE | ID: mdl-24528872

ABSTRACT

BACKGROUND AND PURPOSE: Both the immobilization of the unaffected arm combined with physical therapy (forced arm use, FAU) and voluntary exercise (VE) as model for enriched environment are promising approaches to enhance recovery after stroke. The genomic mechanisms involved in long-term plasticity changes after different means of rehabilitative training post-stroke are largely unexplored. The present investigation explored the effects of these physical therapies on behavioral recovery and molecular markers of regeneration after experimental ischemia. METHODS: 42 Wistar rats were randomly treated with either forced arm use (FAU, 1-sleeve plaster cast onto unaffected limb at 8/10 days), voluntary exercise (VE, connection of a freely accessible running wheel to cage), or controls with no access to a running wheel for 10 days starting at 48 hours after photothrombotic stroke of the sensorimotor cortex. Functional outcome was measured using sensorimotor test before ischemia, after ischemia, after the training period of 10 days, at 3 and 4 weeks after ischemia. Global gene expression changes were assessed from the ipsi- and contralateral cortex and the hippocampus. RESULTS: FAU-treated animals demonstrated significantly improved functional recovery compared to the VE-treated group. Both were superior to cage control. A large number of genes are altered by both training paradigms in the ipsi- and contralateral cortex and the hippocampus. Overall, the extent of changes observed correlated well with the functional recovery obtained. One category of genes overrepresented in the gene set is linked to neuronal plasticity processes, containing marker genes such as the NMDA 2a receptor, PKC ζ, NTRK2, or MAP 1b. CONCLUSIONS: We show that physical training after photothrombotic stroke significantly and permanently improves functional recovery after stroke, and that forced arm training is clearly superior to voluntary running training. The behavioral outcomes seen correlate with patterns and extent of gene expression changes in all brain areas examined. We propose that physical training induces a fundamental change in plasticity-relevant gene expression in several brain regions that enables recovery processes. These results contribute to the debate on optimal rehabilitation strategies, and provide a valuable source of molecular entry points for future pharmacological enhancement of recovery.

4.
J Neurochem ; 128(5): 686-700, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24117625

ABSTRACT

The WWC1 gene has been genetically associated with human episodic memory performance, and its product KIdney/BRAin protein (KIBRA) has been shown to interact with the atypical protein kinase protein kinase M ζ (PKMζ). Although recently challenged, PKMζ remains a candidate postsynaptic regulator of memory maintenance. Here, we show that PKMζ is subject to rapid proteasomal degradation and that KIBRA is both necessary and sufficient to counteract this process, thus stabilizing the kinase and maintaining its function for a prolonged time. We define the binding sequence on KIBRA, a short amino acid motif near the C-terminus. Both hippocampal knock-down of KIBRA in rats and KIBRA knock-out in mice result in decreased learning and memory performance in spatial memory tasks supporting the notion that KIBRA is a player in episodic memory. Interestingly, decreased memory performance is accompanied by decreased PKMζ protein levels. We speculate that the stabilization of synaptic PKMζ protein levels by KIBRA may be one mechanism by which KIBRA acts in memory maintenance. KIBRA/WWC1 has been genetically associated with human episodic memory. KIBRA has been shown to be post-synaptically localized, but its function remained obscure. Here, we show that KIBRA shields PKMζ, a kinase previously linked to memory maintenance, from proteasomal degradation via direct interaction. KIBRA levels in the rodent hippocampus correlate closely both to spatial memory performance in rodents and to PKMζ levels. Our findings support a role for KIBRA in memory, and unveil a novel function for this protein.


Subject(s)
Carrier Proteins/physiology , Co-Repressor Proteins/physiology , Learning/physiology , Memory/physiology , Protein Kinase C/physiology , Amino Acid Sequence , Animals , Avoidance Learning/physiology , Behavior, Animal/physiology , Blotting, Western , Carrier Proteins/metabolism , Co-Repressor Proteins/metabolism , Dependovirus/genetics , Genetic Complementation Test , Hippocampus/metabolism , Hippocampus/physiology , Immunoprecipitation , Intracellular Signaling Peptides and Proteins , Male , Maze Learning/physiology , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Knockout , Molecular Sequence Data , Phosphoproteins , Polymerase Chain Reaction , Protein Binding , Protein Kinase C/metabolism , Rats , Rats, Wistar , Stereotaxic Techniques
5.
Front Cell Neurosci ; 8: 464, 2014.
Article in English | MEDLINE | ID: mdl-25653590

ABSTRACT

BACKGROUND: Amyotrophic lateral sclerosis (ALS) is an incurable fatal motoneuron disease with a lifetime risk of approximately 1:400. It is characterized by progressive weakness, muscle wasting, and death ensuing 3-5 years after diagnosis. Granulocyte-colony stimulating factor (G-CSF) is a drug candidate for ALS, with evidence for efficacy from animal studies and interesting data from pilot clinical trials. To gain insight into the disease mechanisms and mode of action of G-CSF, we performed gene expression profiling on isolated lumbar motoneurons from SOD1(G93A) mice, the most frequently studied animal model for ALS, with and without G-CSF treatment. RESULTS: Motoneurons from SOD1(G93A) mice present a distinct gene expression profile in comparison to controls already at an early disease stage (11 weeks of age), when treatment was initiated. The degree of deregulation increases at a time where motor symptoms are obvious (15 weeks of age). Upon G-CSF treatment, transcriptomic deregulations of SOD1(G93A) motoneurons were notably restored. Discriminant analysis revealed that SOD1 mice treated with G-CSF has a transcriptom close to presymptomatic SOD1 mice or wild type mice. Some interesting genes modulated by G-CSF treatment relate to neuromuscular function such as CCR4-NOT or Prss12. CONCLUSIONS: Our data suggest that G-CSF is able to re-adjust gene expression in symptomatic SOD1(G93A) motoneurons. This provides further arguments for G-CSF as a promising drug candidate for ALS.

6.
Cell Stem Cell ; 12(2): 204-14, 2013 Feb 07.
Article in English | MEDLINE | ID: mdl-23395445

ABSTRACT

Memory impairment has been associated with age-related decline in adult hippocampal neurogenesis. Although Notch, bone morphogenetic protein, and Wnt signaling pathways are known to regulate multiple aspects of adult neural stem cell function, the molecular basis of declining neurogenesis in the aging hippocampus remains unknown. Here, we show that expression of the Wnt antagonist Dickkopf-1 (Dkk1) increases with age and that its loss enhances neurogenesis in the hippocampus. Neural progenitors with inducible loss of Dkk1 increase their Wnt activity, which leads to enhanced self-renewal and increased generation of immature neurons. This Wnt-expanded progeny subsequently matures into glutamatergic granule neurons with increased dendritic complexity. As a result, mice deficient in Dkk1 exhibit enhanced spatial working memory and memory consolidation and also show improvements in affective behavior. Taken together, our findings show that upregulating Wnt signaling by reducing Dkk1 expression can counteract age-related decrease in neurogenesis and its associated cognitive decline.


Subject(s)
Cognition/physiology , Intercellular Signaling Peptides and Proteins/metabolism , Neurogenesis/physiology , Animals , Cell Differentiation/genetics , Cell Differentiation/physiology , Hippocampus/cytology , Intercellular Signaling Peptides and Proteins/genetics , Memory, Short-Term/physiology , Mice , Neurogenesis/genetics , Neurons/cytology , Neurons/metabolism , Wnt Signaling Pathway/genetics , Wnt Signaling Pathway/physiology
7.
PLoS One ; 7(1): e29880, 2012.
Article in English | MEDLINE | ID: mdl-22253813

ABSTRACT

Granulocyte-colony stimulating factor (G-CSF) improves outcome after experimental SCI by counteracting apoptosis, and enhancing connectivity in the injured spinal cord. Previously we have employed the mouse hemisection SCI model and studied motor function after subcutaneous or transgenic delivery of the protein. To further broaden confidence in animal efficacy data we sought to determine efficacy in a different model and a different species. Here we investigated the effects of G-CSF in Wistar rats using the New York University Impactor. In this model, corroborating our previous data, rats treated subcutaneously with G-CSF over 2 weeks show significant improvement of motor function.


Subject(s)
Granulocyte Colony-Stimulating Factor/pharmacology , Granulocyte Colony-Stimulating Factor/therapeutic use , Motor Activity/drug effects , Recovery of Function/drug effects , Spinal Cord Injuries/drug therapy , Spinal Cord Injuries/physiopathology , Animals , Disease Models, Animal , Female , Mice , Rats , Rats, Wistar , Treatment Outcome
8.
J Neurochem ; 119(1): 165-75, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21812782

ABSTRACT

The stimulation of neurogenesis is an exciting novel therapeutic option for diseases of the central nervous system, ranging from depression to neurodegeneration. One major bottleneck in screening approaches for neurogenesis-inducing compounds is the very demanding in vivo quantification of newborn neurons based on stereological techniques. To effectively develop compounds in this area, novel fast and reliable techniques for quantification of in vivo neurogenesis are needed. In this study, we introduce a flow cytometry-based method for quantifying newly generated neurons in the brain based on the counting of cell nuclei from dissected brain regions. Important steps involve density sedimentation of the cell nuclei, and staining for the proliferation marker bromodeoxy uridine and nuclear cell type markers such as NeuN. We demonstrate the ability of the technique to detect increased neurogenesis in the hippocampus of animals which underwent physical exercise and received fluoxetine treatment.


Subject(s)
Brain/physiology , Neurogenesis/physiology , Animals , Antimetabolites , Brain/drug effects , Bromodeoxyuridine , Cell Count , Cell Nucleus/physiology , Centrifugation, Density Gradient , DNA-Binding Proteins , Flow Cytometry , Fluoxetine/pharmacology , Hippocampus/drug effects , Hippocampus/metabolism , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Motor Activity/physiology , Nerve Tissue Proteins/metabolism , Neurogenesis/drug effects , Nuclear Proteins/metabolism , Reproducibility of Results , Selective Serotonin Reuptake Inhibitors/pharmacology
9.
J Neurochem ; 113(4): 930-42, 2010 May.
Article in English | MEDLINE | ID: mdl-20202082

ABSTRACT

Granulocyte-colony stimulating factor (G-CSF) is a potent hematopoietic factor that drives differentiation of neutrophilic granulocytes. We have recently shown that G-CSF also acts as a neuronal growth factor, protects neurons in vitro and in vivo, and has regenerative potential in various neurological disease models. Spinal cord injury (SCI) following trauma or secondary to skeletal instability is a terrible condition with no effective therapies available at present. In this study, we show that the G-CSF receptor is up-regulated upon experimental SCI and that G-CSF improves functional outcome in a partial dissection model of SCI. G-CSF significantly decreases apoptosis in an experimental partial spinal transsection model in the mouse and increases expression of the anti-apoptotic G-CSF target gene Bcl-X(L). In vitro, G-CSF enhances neurite outgrowth and branching capacity of hippocampal neurons. In vivo, G-CSF treatment results in improved functional connectivity of the injured spinal cord as measured by Mn(2+)-enhanced MRI. G-CSF also increased length of the dorsal corticospinal tract and density of serotonergic fibers cranial to the lesion center. Mice treated systemically with G-CSF as well as transgenic mice over-expressing G-CSF in the CNS exhibit a strong improvement in functional outcome as measured by the BBB score and gridwalk analysis. We show that G-CSF improves outcome after experimental SCI by counteracting apoptosis, and enhancing connectivity in the injured spinal cord. We conclude that G-CSF constitutes a promising and feasible new therapy option for SCI.


Subject(s)
Granulocyte Colony-Stimulating Factor/pharmacology , Nerve Growth Factors/pharmacology , Neuroprotective Agents/pharmacology , Spinal Cord Injuries/drug therapy , Animals , Apoptosis/drug effects , Apoptosis/physiology , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cell Survival/drug effects , Cell Survival/physiology , Cells, Cultured , Disease Models, Animal , Female , Granulocyte Colony-Stimulating Factor/therapeutic use , Growth Cones/drug effects , Growth Cones/physiology , Growth Cones/ultrastructure , Mice , Nerve Growth Factors/therapeutic use , Neurites/drug effects , Neurites/ultrastructure , Neuroprotective Agents/therapeutic use , Pyramidal Tracts/drug effects , Pyramidal Tracts/injuries , Pyramidal Tracts/physiology , Rats , Rats, Wistar , Receptors, Granulocyte Colony-Stimulating Factor/drug effects , Receptors, Granulocyte Colony-Stimulating Factor/metabolism , Recovery of Function/drug effects , Recovery of Function/physiology , Spinal Cord Injuries/metabolism , Spinal Cord Injuries/physiopathology , Treatment Outcome , Up-Regulation/drug effects , Up-Regulation/physiology , bcl-X Protein/drug effects , bcl-X Protein/metabolism
10.
Brain ; 131(Pt 12): 3335-47, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18835867

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a devastating neurodegenerative disease that results in progressive loss of motoneurons, motor weakness and death within 1-5 years after disease onset. Therapeutic options remain limited despite a substantial number of approaches that have been tested clinically. In particular, various neurotrophic factors have been investigated. Failure in these trials has been largely ascribed to problems of insufficient dosing or inability to cross the blood-brain barrier (BBB). We have recently uncovered the neurotrophic properties of the haematopoietic protein granulocyte-colony stimulating factor (G-CSF). The protein is clinically well tolerated and crosses the intact BBB. This study examined the potential role of G-CSF in motoneuron diseases. We investigated the expression of the G-CSF receptor in motoneurons and studied effects of G-CSF in a motoneuron cell line and in the SOD1(G93A) transgenic mouse model. The neurotrophic growth factor was applied both by continuous subcutaneous delivery and CNS-targeted transgenic overexpression. This study shows that given at the stage of the disease where muscle denervation is already evident, G-CSF leads to significant improvement in motor performance, delays the onset of severe motor impairment and prolongs overall survival of SOD1(G93A)tg mice. The G-CSF receptor is expressed by motoneurons and G-CSF protects cultured motoneuronal cells from apoptosis. In ALS mice, G-CSF increased survival of motoneurons and decreased muscular denervation atrophy. We conclude that G-CSF is a novel neurotrophic factor for motoneurons that is an attractive and feasible drug candidate for the treatment of ALS.


Subject(s)
Amyotrophic Lateral Sclerosis/drug therapy , Granulocyte Colony-Stimulating Factor/therapeutic use , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/pathology , Animals , Apoptosis/drug effects , Cells, Cultured , Disease Models, Animal , Disease Progression , Drug Evaluation, Preclinical/methods , Female , Filgrastim , Granulocyte Colony-Stimulating Factor/administration & dosage , Granulocyte Colony-Stimulating Factor/pharmacology , Humans , Infusions, Subcutaneous , Mice , Mice, Inbred C57BL , Mice, Transgenic , Motor Neurons/drug effects , Motor Neurons/metabolism , Mutation , Receptors, Granulocyte Colony-Stimulating Factor/metabolism , Recombinant Proteins , Reverse Transcriptase Polymerase Chain Reaction/methods , Spinal Cord/metabolism , Superoxide Dismutase/genetics , Superoxide Dismutase-1 , Treatment Outcome
11.
J Neurochem ; 92(3): 597-606, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15659229

ABSTRACT

Synaptopodin (SYNPO) is a cytoskeletal protein that is preferentially located in mature dendritic spines, where it accumulates in the spine neck and closely associates with the spine apparatus. Formation of the spine apparatus critically depends on SYNPO. To further determine its molecular action, we screened for cellular binding partners. Using the yeast two-hybrid system and biochemical assays, SYNPO was found to associate with both F-actin and alpha-actinin. Ectopic expression of SYNPO in neuronal and non-neuronal cells induced actin aggregates, thus confirming a cytoplasmic interaction with the actin cytoskeleton. Whereas F-actin association is mediated by a central SYNPO motif, binding to alpha-actinin requires the C-terminal domain. Notably, the alpha-actinin binding domain is also essential for dendritic targeting and postsynaptic accumulation of SYNPO in primary neurons. Taken together, our data suggest that dendritic spine accumulation of SYNPO critically depends on its interaction with postsynaptic alpha-actinin and that SYNPO may regulate spine morphology, motility and function via its distinct modes of association with the actin cytoskeleton.


Subject(s)
Actinin/metabolism , Actins/metabolism , Dendritic Spines/metabolism , Microfilament Proteins/metabolism , Amino Acid Motifs/genetics , Amino Acid Sequence , Animals , Binding Sites/genetics , Binding Sites/physiology , Cells, Cultured , Haplorhini , Humans , Mice , Microfilament Proteins/genetics , Molecular Sequence Data , Protein Binding/physiology , Protein Structure, Tertiary/genetics , Protein Structure, Tertiary/physiology , Rats , Sequence Homology, Amino Acid , Transfection , Two-Hybrid System Techniques
12.
Biochem Biophys Res Commun ; 317(3): 703-7, 2004 May 07.
Article in English | MEDLINE | ID: mdl-15081397

ABSTRACT

WW domain-containing proteins are found in all eukaryotic cells and they are involved in the regulation of a wide variety of cellular functions. We recently identified the neuronal protein KIBRA as novel member of this family of signal transducers. In this report, we describe the identification of protein kinase C (PKC) zeta as a KIBRA-interacting protein. PKCzeta is known to play an important role in synaptic plasticity and memory formation but its specific targets are not well known. Our studies presented here revealed that KIBRA is a novel substrate for PKCzeta and suggest that PKCzeta phosphorylation may regulate the cellular function of KIBRA.


Subject(s)
Protein Kinase C/metabolism , Proteins/metabolism , HeLa Cells , Humans , Intracellular Signaling Peptides and Proteins , Phosphoproteins , Phosphorylation , Substrate Specificity , Two-Hybrid System Techniques
13.
Biochem Biophys Res Commun ; 300(4): 862-7, 2003 Jan 24.
Article in English | MEDLINE | ID: mdl-12559952

ABSTRACT

In a yeast two hybrid screen with the human isoform of Dendrin (KIAA0749), a putative modulator of the postsynaptic cytoskeleton, we isolated a cDNA coding for a novel protein, KIBRA, possessing two amino-terminal WW domains, an internal C2-like domain and a carboxy-terminal glutamic acid-rich stretch. Northern blot analysis revealed that the expression of KIBRA mRNA was predominately found in kidney and brain. In vitro interaction studies revealed that the first KIBRA WW domain binds specifically to PPxY motifs. Transient transfection of monkey kidney cells with constructs encoding Myc-tagged KIBRA displayed a cytoplasmic localization and a perinuclear enrichment of the protein.


Subject(s)
Nerve Tissue Proteins/metabolism , Proteins/metabolism , Amino Acid Sequence , Animals , Cells, Cultured , Chlorocebus aethiops , Humans , Intracellular Signaling Peptides and Proteins , Molecular Sequence Data , Mutagenesis, Site-Directed , Phosphoproteins , Protein Structure, Tertiary , Proteins/chemistry , Proteins/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sequence Alignment , Two-Hybrid System Techniques
SELECTION OF CITATIONS
SEARCH DETAIL
...