141 results on '"Schäfer MK"'
Search Results
2. Replenishment policies for inventories of recoverable items with attrition
- Author
-
Schaefer, MK
- Published
- 1989
- Full Text
- View/download PDF
3. Potassium channel TASK-5 forms functional heterodimers with TASK-1 and TASK-3 to break its silence.
- Author
-
Rinné S, Schick F, Vowinkel K, Schütte S, Krasel C, Kauferstein S, Schäfer MK, Kiper AK, Müller T, and Decher N
- Subjects
- Animals, Humans, Cell Membrane metabolism, HEK293 Cells, Protein Multimerization, Nerve Tissue Proteins genetics, Nerve Tissue Proteins metabolism, Potassium Channels, Tandem Pore Domain metabolism, Potassium Channels, Tandem Pore Domain genetics
- Abstract
TASK-5 (KCNK15) belongs to the acid-sensitive subfamily of two-pore domain potassium (K
2P ) channels, which includes TASK-1 and TASK-3. TASK-5 stands out as K2P channel for which there is no functional data available, since it was reported in 2001 as non-functional and thus "silent". Here we show that TASK-5 channels are indeed non-functional as homodimers, but are involved in the formation of functional channel complexes with TASK-1 and TASK-3. TASK-5 negatively modulates the surface expression of TASK channels, while the heteromeric TASK-5-containing channel complexes located at the plasma membrane are characterized by changes in single-channel conductance, Gq-coupled receptor-mediated channel inhibition, and sensitivity to TASK modulators. The unique pharmacology of TASK-1/TASK-5 heterodimers, affected by a common polymorphism in KCNK15, needs to be carefully considered in the future development of drugs targeting TASK channels. Our observations provide an access to study TASK-5 at the functional level, particularly in malignant cancers associated with KCNK15., (© 2024. The Author(s).)- Published
- 2024
- Full Text
- View/download PDF
4. KCNQ1 is an essential mediator of the sex-dependent perception of moderate cold temperatures.
- Author
-
Kiper AK, Wegner S, Kadala A, Rinné S, Schütte S, Winter Z, Bertoune MAR, Touska F, Matschke V, Wrobel E, Streit AK, Lang F, Schmidt C, Schulze-Bahr E, Schäfer MK, Voelkl J, Seebohm G, Zimmermann K, and Decher N
- Subjects
- Animals, Female, Male, Mice, Action Potentials physiology, Ganglia, Spinal metabolism, Menthol pharmacology, Mice, Inbred C57BL, Mice, Knockout, TRPM Cation Channels metabolism, TRPM Cation Channels genetics, Cold Temperature, KCNQ1 Potassium Channel metabolism, KCNQ1 Potassium Channel genetics, Thermosensing genetics
- Abstract
Low temperatures and cooling agents like menthol induce cold sensation by activating the peripheral cold receptors TRPM8 and TRPA1, cation channels belonging to the TRP channel family, while the reduction of potassium currents provides an additional and/or synergistic mechanism of cold sensation. Despite extensive studies over the past decades to identify the molecular receptors that mediate thermosensation, cold sensation is still not fully understood and many cold-sensitive peripheral neurons do not express the well-established cold sensor TRPM8. We found that the voltage-gated potassium channel KCNQ1 (Kv7.1), which is defective in cardiac LQT1 syndrome, is, in addition to its known function in the heart, a highly relevant and sex-specific sensor of moderately cold temperatures. We found that KCNQ1 is expressed in skin and dorsal root ganglion neurons, is sensitive to menthol and cooling agents, and is highly sensitive to moderately cold temperatures, in a temperature range at which TRPM8 is not thermosensitive. C-fiber recordings from KCNQ1
-/- mice displayed altered action potential firing properties. Strikingly, only male KCNQ1-/- mice showed substantial deficits in cold avoidance at moderately cold temperatures, with a strength of the phenotype similar to that observed in TRPM8-/- animals. While sex-dependent differences in thermal sensitivity have been well documented in humans and mice, KCNQ1 is the first gene reported to play a role in sex-specific temperature sensation. Moreover, we propose that KCNQ1, together with TRPM8, is a key instrumentalist that orchestrates the range and intensity of cold sensation., Competing Interests: Competing interests statement:The authors declare no competing interest.- Published
- 2024
- Full Text
- View/download PDF
5. Popeye domain containing proteins modulate the voltage-gated cardiac sodium channel Nav1.5.
- Author
-
Rinné S, Kiper AK, Jacob R, Ortiz-Bonnin B, Schindler RFR, Fischer S, Komadowski M, De Martino E, Schäfer MK, Cornelius T, Fabritz L, Helker CSM, Brand T, and Decher N
- Abstract
Popeye domain containing (POPDC) proteins are predominantly expressed in the heart and skeletal muscle, modulating the K
2P potassium channel TREK-1 in a cAMP-dependent manner. POPDC1 and POPDC2 variants cause cardiac conduction disorders with or without muscular dystrophy. Searching for POPDC2-modulated ion channels using a functional co-expression screen in Xenopus oocytes, we found POPDC proteins to modulate the cardiac sodium channel Nav1.5. POPDC proteins downregulate Nav1.5 currents in a cAMP-dependent manner by reducing the surface expression of the channel. POPDC2 and Nav1.5 are both expressed in different regions of the murine heart and consistently POPDC2 co-immunoprecipitates with Nav1.5 from native cardiac tissue. Strikingly, the knock-down of popdc2 in embryonic zebrafish caused an increased upstroke velocity and overshoot of cardiac action potentials. The POPDC modulation of Nav1.5 provides a new mechanism to regulate cardiac sodium channel densities under sympathetic stimulation, which is likely to have a functional impact on cardiac physiology and inherited arrhythmias., Competing Interests: L.F. has received institutional research grants and non-financial support from European Union, DFG, British Heart Foundation, Medical Research Council (UK), NIHR, and several biomedical companies. L.F. is listed as inventor on two patents held by the academic employer (Atrial Fibrillation Therapy WO 2015140571, Markers for Atrial Fibrillation WO 2016012783)., (© 2024 The Author(s).)- Published
- 2024
- Full Text
- View/download PDF
6. Contingent Social Interaction Does Not Prevent Habituation towards Playback of Pro-Social 50-kHz Calls: Behavioral Responses and Brain Activation Patterns.
- Author
-
Berz A, Pasquini de Souza C, Wöhr M, Steinmüller S, Bruntsch M, Schäfer MK, and Schwarting RKW
- Abstract
Rats, which are highly social animals, are known to communicate using ultrasonic vocalizations (USV) in different frequency ranges. Calls around 50 kHz are related to positive affective states and promote social interactions. Our previous work has shown that the playback of natural 50-kHz USV leads to a strong social approach response toward the sound source, which is related to activation in the nucleus accumbens. In male Wistar rats, the behavioral response habituates, that is, becomes weaker or is even absent, when such playback is repeated several days later, an outcome found to be memory-dependent. Here, we asked whether such habituation is due to the lack of a contingent social consequence after playback in the initial test and whether activation of the nucleus accumbens, as measured by c-fos immunohistochemistry, can still be observed in a retest. To this end, groups of young male Wistar rats underwent an initial 50-kHz USV playback test, immediately after which they were either (1) kept temporarily alone, (2) exposed to a same-sex juvenile, or (3) to their own housing group. One week later, they underwent a retest with playback; this time not followed by social consequences but by brain removal for c-fos immunohistochemistry. Consistent with previous reports, behavioral changes evoked by the initial exposure to 50-kHz USV playback included a strong approach response. In the retest, no such response was found, irrespective of whether rats had experienced a contingent social consequence after the initial test or not. At the neural level, no substantial c-fos activation was found in the nucleus accumbens, but unexpected strong activation was detected in the anterior cingulate cortex, with some of it in GABAergic cells. The c-fos patterns did not differ between groups but cell numbers were individually correlated with behavior, i.e., rats that still approached in response to playback in the retest showed more activation. Together, these data do not provide substantial evidence that the lack of a contingent social consequence after 50-kHz USV playback accounts for approach habituation in the retest. Additionally, there is apparently no substantial activation of the nucleus accumbens in the retest, whereas the exploratory findings in the anterior cingulate cortex indicate that this brain area might be involved when individual rats still approach 50-kHz USV playback.
- Published
- 2022
- Full Text
- View/download PDF
7. Optimized Tuning of Auditory Inner Hair Cells to Encode Complex Sound through Synergistic Activity of Six Independent K + Current Entities.
- Author
-
Dierich M, Altoè A, Koppelmann J, Evers S, Renigunta V, Schäfer MK, Naumann R, Verhulst S, Oliver D, and Leitner MG
- Subjects
- 4-Aminopyridine pharmacology, Animals, CHO Cells, Cricetulus, Hair Cells, Auditory, Inner drug effects, Ion Channel Gating drug effects, Membrane Potentials drug effects, Mice, Inbred C57BL, Protein Subunits metabolism, Hair Cells, Auditory, Inner metabolism, Potassium Channels metabolism, Sound
- Abstract
Auditory inner hair cells (IHCs) convert sound vibrations into receptor potentials that drive synaptic transmission. For the precise encoding of sound qualities, receptor potentials are shaped by K
+ conductances tuning the properties of the IHC membrane. Using patch-clamp and computational modeling, we unravel this membrane specialization showing that IHCs express an exclusive repertoire of six voltage-dependent K+ conductances mediated by Kv 1.8, Kv 7.4, Kv 11.1, Kv 12.1, and BKCa channels. All channels are active at rest but are triggered differentially during sound stimulation. This enables non-saturating tuning over a far larger potential range than in IHCs expressing fewer current entities. Each conductance contributes to optimizing responses, but the combined activity of all channels synergistically improves phase locking and the dynamic range of intensities that IHCs can encode. Conversely, hypothetical simpler IHCs appear limited to encode only certain aspects (frequency or intensity). The exclusive channel repertoire of IHCs thus constitutes an evolutionary adaptation to encode complex sound through multifaceted receptor potentials., Competing Interests: Declaration of Interests The authors declare no competing interests., (Copyright © 2020 The Author(s). Published by Elsevier Inc. All rights reserved.)- Published
- 2020
- Full Text
- View/download PDF
8. Pituitary Adenylate Cyclase-Activating Peptide (PACAP)-Glutamate Co-transmission Drives Circadian Phase-Advancing Responses to Intrinsically Photosensitive Retinal Ganglion Cell Projections by Suprachiasmatic Nucleus.
- Author
-
Lindberg PT, Mitchell JW, Burgoon PW, Beaulé C, Weihe E, Schäfer MK, Eiden LE, Jiang SZ, and Gillette MU
- Abstract
Results from a variety of sources indicate a role for pituitary adenylate cyclase-activating polypeptide (PACAP) in light/glutamate-induced phase resetting of the circadian clock mediated by the retinohypothalamic tract (RHT). Attempts to block or remove PACAP's contribution to clock-resetting have generated phenotypes that differ in their responses to light or glutamate. For example, previous studies of circadian behaviors found that period-maintenance and early-night phase delays are intact in PACAP-null mice, yet there is a consistent deficit in behavioral phase-resetting to light stimulation in the late night. Here we report rodent stimulus-response characteristics of PACAP release from the RHT, and map these to responses of the suprachiasmatic nucleus (SCN) in intact and PACAP-deficient mouse hypothalamus with regard to phase-resetting. SCN of PACAP-null mice exhibit normal circadian rhythms in neuronal activity, but are "blind" to glutamate stimulating phase-advance responses in late night, although not in early night, consistent with previously reported selective lack of late-night light behavioral responsiveness of these mice. Induction of CREB phosphorylation, a hallmark of the light/glutamate response of the SCN, also is absent in SCN-containing ex vivo slices from PACAP-deficient mouse hypothalamus. PACAP replacement to the SCN of PACAP-null mice restored wild-type phase-shifting of firing-rate patterns in response to glutamate applied to the SCN in late night. Likewise, ex vivo SCN of wild-type mice post-orbital enucleation are unresponsive to glutamate unless PACAP also is restored. Furthermore, we demonstrate that the period of efficacy of PACAP at SCN nerve terminals corresponds to waxing of PACAP mRNA expression in ipRGCs during the night, and waning during the day. These results validate the use of PACAP-deficient mice in defining the role and specificity of PACAP as a co-transmitter with glutamate in ipRGC-RHT projections to SCN in phase advancing the SCN circadian rhythm in late night., (Copyright © 2019 Lindberg, Mitchell, Burgoon, Beaulé, Weihe, Schäfer, Eiden, Jiang and Gillette.)
- Published
- 2019
- Full Text
- View/download PDF
9. PAI-1 but Not PAI-2 Gene Deficiency Attenuates Ischemic Brain Injury After Experimental Stroke.
- Author
-
Griemert EV, Recarte Pelz K, Engelhard K, Schäfer MK, and Thal SC
- Subjects
- Animals, Brain Injuries genetics, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Plasminogen Activator Inhibitor 2 genetics, Serpin E2 genetics, Stroke genetics, Brain Injuries metabolism, Brain Injuries prevention & control, Plasminogen Activator Inhibitor 2 deficiency, Serpin E2 deficiency, Stroke metabolism
- Abstract
After stroke, secondary brain damage is influenced by the extent of fibrin clot formation. This is counteracted by the endogenous fibrinolysis. Of major interest are the key players of the fibrinolytic plasminogen activator system including the urokinase plasminogen activator (uPA), the tissue-type plasminogen activator (tPA), and their endogenous inhibitors plasminogen activator inhibitor 1 (PAI-1) and PAI-2. The role of PAI-1 in brain injury is well established, whereas the importance of PAI-2 is unknown at present. The objectives of the present were twofold: first, to characterize the time-dependent cerebral mRNA expression of the plasminogen activator system (PAS) after brain ischemia and second, to investigate the impact of PAI-1 and PAI-2 on brain infarct volume using gene-deficient mice. Adult C57Bl/6J mice were subjected to unilateral transient middle cerebral artery occlusion (MCAO) followed by reperfusion for 3, 24, 72, or 120 h. Quantitative PCR revealed that brain mRNA expression levels of the PAS components, and particularly of PAI-1 (237-fold) and PAI-2 (19-fold), peaked at 24 h after stroke. Accordingly, PAI-1 plasma activity was strongly increased. Brain infarct volume in TTC (2,3,5-triphenyltetrazolium chloride)-stained brain sections was significantly smaller 24 h after MCAO in PAI-1-deficient mice (- 31%), but not in PAI-2-deficient mice (- 6%). Thus, endogenous upregulation of PAI-1, but not of PAI-2, might contribute to increased brain damage after acute ischemic stroke. The present study therefore shows that PAI-2 is induced by brain ischemia, but does not play an important or relevant role for secondary brain damage after brain injury.
- Published
- 2019
- Full Text
- View/download PDF
10. Plasminogen activator inhibitor-1 augments damage by impairing fibrinolysis after traumatic brain injury.
- Author
-
Griemert EV, Schwarzmaier SM, Hummel R, Gölz C, Yang D, Neuhaus W, Burek M, Förster CY, Petkovic I, Trabold R, Plesnila N, Engelhard K, Schäfer MK, and Thal SC
- Subjects
- Animals, Brain drug effects, Brain Injuries, Traumatic drug therapy, Fibrinolysis drug effects, Indoleacetic Acids pharmacology, Indoleacetic Acids therapeutic use, Male, Mice, Mice, Inbred C57BL, Serpin E2 antagonists & inhibitors, Brain metabolism, Brain pathology, Brain Injuries, Traumatic metabolism, Brain Injuries, Traumatic pathology, Fibrinolysis physiology, Serpin E2 metabolism
- Abstract
Objective: Plasminogen activator inhibitor-1 (PAI-1) is the key endogenous inhibitor of fibrinolysis, and enhances clot formation after injury. In traumatic brain injury, dysregulation of fibrinolysis may lead to sustained microthrombosis and accelerated lesion expansion. In the present study, we hypothesized that PAI-1 mediates post-traumatic malfunction of coagulation, with inhibition or genetic depletion of PAI-1 attenuating clot formation and lesion expansion after brain trauma., Methods: We evaluated PAI-1 as a possible new target in a mouse controlled cortical impact (CCI) model of traumatic brain injury. We performed the pharmacological inhibition of PAI-1 with PAI-039 and stimulation by tranexamic acid, and we confirmed our results in PAI-1-deficient animals., Results: PAI-1 mRNA was time-dependently upregulated, with a 305-fold peak 12 hours after CCI, which effectively counteracted the 2- to 3-fold increase in cerebral tissue-type/urokinase plasminogen activator expression. PAI-039 reduced brain lesion volume by 26% at 24 hours and 43% at 5 days after insult. This treatment also attenuated neuronal apoptosis and improved neurofunctional outcome. Moreover, intravital microscopy demonstrated reduced post-traumatic thrombus formation in the pericontusional cortical microvasculature. In PAI-1-deficient mice, the therapeutic effect of PAI-039 was absent. These mice also displayed 13% reduced brain damage compared with wild type. In contrast, inhibition of fibrinolysis with tranexamic acid increased lesion volume by 25% compared with vehicle., Interpretation: This study identifies impaired fibrinolysis as a critical process in post-traumatic secondary brain damage and suggests that PAI-1 may be a central endogenous inhibitor of the fibrinolytic pathway, promoting a procoagulatory state and clot formation in the cerebral microvasculature. Ann Neurol 2019;85:667-680., (© 2019 The Authors. Annals of Neurology published by Wiley Periodicals, Inc. on behalf of American Neurological Association.)
- Published
- 2019
- Full Text
- View/download PDF
11. Neural cell adhesion molecule Negr1 deficiency in mouse results in structural brain endophenotypes and behavioral deviations related to psychiatric disorders.
- Author
-
Singh K, Jayaram M, Kaare M, Leidmaa E, Jagomäe T, Heinla I, Hickey MA, Kaasik A, Schäfer MK, Innos J, Lilleväli K, Philips MA, and Vasar E
- Subjects
- Animals, Brain metabolism, Mental Disorders metabolism, Mice, Mice, Knockout, Behavior, Animal, Brain pathology, Cell Adhesion Molecules, Neuronal genetics, Endophenotypes, Mental Disorders pathology
- Abstract
Neuronal growth regulator 1 (NEGR1) belongs to the immunoglobulin (IgLON) superfamily of cell adhesion molecules involved in cortical layering. Recent functional and genomic studies implicate the role of NEGR1 in a wide spectrum of psychiatric disorders, such as major depression, schizophrenia and autism. Here, we investigated the impact of Negr1 deficiency on brain morphology, neuronal properties and social behavior of mice. In situ hybridization shows Negr1 expression in the brain nuclei which are central modulators of cortical-subcortical connectivity such as the island of Calleja and the reticular nucleus of thalamus. Brain morphological analysis revealed neuroanatomical abnormalities in Negr1
-/- mice, including enlargement of ventricles and decrease in the volume of the whole brain, corpus callosum, globus pallidus and hippocampus. Furthermore, decreased number of parvalbumin-positive inhibitory interneurons was evident in Negr1-/- hippocampi. Behaviorally, Negr1-/- mice displayed hyperactivity in social interactions and impairments in social hierarchy. Finally, Negr1 deficiency resulted in disrupted neurite sprouting during neuritogenesis. Our results provide evidence that NEGR1 is required for balancing the ratio of excitatory/inhibitory neurons and proper formation of brain structures, which is prerequisite for adaptive behavioral profiles. Therefore, Negr1-/- mice have a high potential to provide new insights into the neural mechanisms of neuropsychiatric disorders.- Published
- 2019
- Full Text
- View/download PDF
12. The VAMP-associated protein VAPB is required for cardiac and neuronal pacemaker channel function.
- Author
-
Silbernagel N, Walecki M, Schäfer MK, Kessler M, Zobeiri M, Rinné S, Kiper AK, Komadowski MA, Vowinkel KS, Wemhöner K, Fortmüller L, Schewe M, Dolga AM, Scekic-Zahirovic J, Matschke LA, Culmsee C, Baukrowitz T, Monassier L, Ullrich ND, Dupuis L, Just S, Budde T, Fabritz L, and Decher N
- Subjects
- Animals, Carrier Proteins physiology, Embryo, Nonmammalian cytology, Embryo, Nonmammalian physiology, Female, HeLa Cells, Humans, Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels genetics, Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels metabolism, Mice, Mice, Knockout, Neurons cytology, Rats, Rats, Sprague-Dawley, Vesicular Transport Proteins, Xenopus laevis, Zebrafish, Heart physiology, Ion Channel Gating, Membrane Proteins physiology, Neurons physiology, Pacemaker, Artificial
- Abstract
Hyperpolarization-activated cyclic nucleotide-gated (HCN) channels encode neuronal and cardiac pacemaker currents. The composition of pacemaker channel complexes in different tissues is poorly understood, and the presence of additional HCN modulating subunits was speculated. Here we show that vesicle-associated membrane protein-associated protein B (VAPB), previously associated with a familial form of amyotrophic lateral sclerosis 8, is an essential HCN1 and HCN2 modulator. VAPB significantly increases HCN2 currents and surface expression and has a major influence on the dendritic neuronal distribution of HCN2. Severe cardiac bradycardias in VAPB-deficient zebrafish and VAPB
-/- mice highlight that VAPB physiologically serves to increase cardiac pacemaker currents. An altered T-wave morphology observed in the ECGs of VAPB-/- mice supports the recently proposed role of HCN channels for ventricular repolarization. The critical function of VAPB in native pacemaker channel complexes will be relevant for our understanding of cardiac arrhythmias and epilepsies, and provides an unexpected link between these diseases and amyotrophic lateral sclerosis.-Silbernagel, N., Walecki, M., Schäfer, M.-K. H., Kessler, M., Zobeiri, M., Rinné, S., Kiper, A. K., Komadowski, M. A., Vowinkel, K. S., Wemhöner, K., Fortmüller, L., Schewe, M., Dolga, A. M., Scekic-Zahirovic, J., Matschke, L. A., Culmsee, C., Baukrowitz, T., Monassier, L., Ullrich, N. D., Dupuis, L., Just, S., Budde, T., Fabritz, L., Decher, N. The VAMP-associated protein VAPB is required for cardiac and neuronal pacemaker channel function.- Published
- 2018
- Full Text
- View/download PDF
13. NEGR1 and FGFR2 cooperatively regulate cortical development and core behaviours related to autism disorders in mice.
- Author
-
Szczurkowska J, Pischedda F, Pinto B, Managò F, Haas CA, Summa M, Bertorelli R, Papaleo F, Schäfer MK, Piccoli G, and Cancedda L
- Subjects
- Animals, Autism Spectrum Disorder metabolism, Behavior, Animal physiology, Cell Adhesion Molecules, Neuronal metabolism, Cell Membrane metabolism, Cell Movement, Cerebral Cortex growth & development, Dendritic Spines physiology, Disease Models, Animal, Down-Regulation, HEK293 Cells, Humans, MAP Kinase Signaling System physiology, Mice, Mice, Inbred C57BL, Mice, Knockout, Neurogenesis, Receptor, Fibroblast Growth Factor, Type 2 metabolism, Signal Transduction physiology, Autism Spectrum Disorder physiopathology, Cell Adhesion Molecules, Neuronal physiology, Receptor, Fibroblast Growth Factor, Type 2 physiology
- Abstract
Autism spectrum disorders are neurodevelopmental conditions with diverse aetiologies, all characterized by common core symptoms such as impaired social skills and communication, as well as repetitive behaviour. Cell adhesion molecules, receptor tyrosine kinases and associated downstream signalling have been strongly implicated in both neurodevelopment and autism spectrum disorders. We found that downregulation of the cell adhesion molecule NEGR1 or the receptor tyrosine kinase fibroblast growth factor receptor 2 (FGFR2) similarly affects neuronal migration and spine density during mouse cortical development in vivo and results in impaired core behaviours related to autism spectrum disorders. Mechanistically, NEGR1 physically interacts with FGFR2 and modulates FGFR2-dependent extracellular signal-regulated kinase (ERK) and protein kinase B (AKT) signalling by decreasing FGFR2 degradation from the plasma membrane. Accordingly, FGFR2 overexpression rescues all defects due to Negr1 knockdown in vivo. Negr1 knockout mice present phenotypes similar to Negr1-downregulated animals. These data indicate that NEGR1 and FGFR2 cooperatively regulate cortical development and suggest a role for defective NEGR1-FGFR2 complex and convergent downstream ERK and AKT signalling in autism spectrum disorders.
- Published
- 2018
- Full Text
- View/download PDF
14. Sensory neuropathy in progressive motor neuronopathy (pmn) mice is associated with defects in microtubule polymerization and axonal transport.
- Author
-
Schäfer MK, Bellouze S, Jacquier A, Schaller S, Richard L, Mathis S, Vallat JM, and Haase G
- Subjects
- Animals, Axons metabolism, Axons pathology, Cells, Cultured, Disease Models, Animal, Embryo, Mammalian, Ganglia, Spinal cytology, Mice, Mice, Inbred C57BL, Mice, Neurologic Mutants, Mice, Transgenic, Microtubules genetics, Microtubules ultrastructure, Molecular Chaperones genetics, Motor Neuron Disease genetics, Motor Neuron Disease pathology, Mutation, Missense genetics, Neurons metabolism, Neurons pathology, Neurons ultrastructure, Phrenic Nerve pathology, Phrenic Nerve ultrastructure, Polymerization, Sural Nerve metabolism, Sural Nerve ultrastructure, Axonal Transport genetics, Microtubules metabolism, Motor Neuron Disease complications, Peripheral Nervous System Diseases etiology, Peripheral Nervous System Diseases pathology, Sural Nerve pathology
- Abstract
Motor neuron diseases such as amyotrophic lateral sclerosis (ALS) are now recognized as multi-system disorders also involving various non-motor neuronal cell types. The precise extent and mechanistic basis of non-motor neuron damage in human ALS and ALS animal models remain however unclear. To address this, we here studied progressive motor neuronopathy (pmn) mice carrying a missense loss-of-function mutation in tubulin binding cofactor E (TBCE). These mice manifest a particularly aggressive form of motor axon dying back and display a microtubule loss, similar to that induced by human ALS-linked TUBA4A mutations. Using whole nerve confocal imaging of pmn × thy1.2-YFP16 fluorescent reporter mice and electron microscopy, we demonstrate axonal discontinuities, bead-like spheroids and ovoids in pmn suralis nerves indicating prominent sensory neuropathy. The axonal alterations qualitatively resemble those in phrenic motor nerves but do not culminate in the loss of myelinated fibers. We further show that the pmn mutation decreases the level of TBCE, impedes microtubule polymerization in dorsal root ganglion (DRG) neurons and causes progressive loss of microtubules in large and small caliber suralis axons. Live imaging of axonal transport using GFP-tagged tetanus toxin C-fragment (GFP-TTC) demonstrates defects in microtubule-based transport in pmn DRG neurons, providing a potential explanation for the axonal alterations in sensory nerves. This study unravels sensory neuropathy as a pathological feature of mouse pmn, and discusses the potential contribution of cytoskeletal defects to sensory neuropathy in human motor neuron disease., (© 2016 International Society of Neuropathology.)
- Published
- 2017
- Full Text
- View/download PDF
15. Osteointegration and Resorption of Intravertebral and Extravertebral Calcium Phosphate Cement.
- Author
-
Klein R, Tetzlaff R, Weiss C, Schäfer MK, Tanner M, Wiedenhöfer B, Grafe I, Meeder PJ, Noeldge G, Nawroth PP, and Kasperk C
- Subjects
- Adult, Aged, Body Weight, Female, Follow-Up Studies, Humans, Magnetic Resonance Imaging, Male, Middle Aged, Movement, Osteoporosis, Osteoporotic Fractures complications, Osteoporotic Fractures diagnostic imaging, Pain etiology, Pain surgery, Tomography Scanners, X-Ray Computed, Treatment Outcome, Visual Analog Scale, Bone Cements therapeutic use, Calcium Phosphates therapeutic use, Kyphoplasty methods, Osteoporotic Fractures surgery
- Abstract
Study Design: Eleven patients with painful osteoporotic vertebral fractures who underwent kyphoplasty using calcium phosphate (CaP) cement were followed up for 1 week, 1, 2, and 3 years in a monocentric, nonrandomized, noncontrolled retrospective trial., Objective: This study investigates long-term radiomorphologic features of intraosseous CaP cement implants and of extraosseous CaP cement leakages for up to 3 years after implantation by kyphoplasty., Summary of Background Data: Kyphoplasty is frequently used for the treatment of painful osteoporotic fractures. Of the materials available, CaP is frequently used as a filling material. Resorption of this material is frequently observed, although clinical outcome is comparable with other cements., Methods: Kyphoplasty utilizing CaP cement was performed in 11 patients with painful osteoporotic vertebral fractures. All patients received a pharmacological antiosteoporosis treatment consisting of calcium, vitamin D, and a standard dose of oral bisphosphonates. Radiomorphologic measurements, pain, and mobility were assessed., Results: Intraosseous and extraosseous CaP cement volumes decreased significantly over 3 years. However, vertebral stability as determined by a constant vertebral body height and the sagittal index was not impaired. Pain improved significantly 2 years after implantation and the mobility scores 1 year after kyphoplasty at least until the third year., Conclusions: Intravertebral CaP cement implants are resorbed slowly over time without jeopardizing stability and clinical outcomes most likely because of a slowly progressing osseous replacement. Extraosseous CaP cement material because of leakages during the kyphoplasty procedure is almost completely resorbed as early as 2 years after the leakage occurred. Therefore, CaP cement is an important alternative to PMMA-based cement materials utilized for kyphoplasty of osteoporotic vertebral fractures.
- Published
- 2017
- Full Text
- View/download PDF
16. Sex-Dependent Regulation of Aromatase-Mediated Synaptic Plasticity in the Basolateral Amygdala.
- Author
-
Bender RA, Zhou L, Vierk R, Brandt N, Keller A, Gee CE, Schäfer MK, and Rune GM
- Subjects
- Animals, Basolateral Nuclear Complex drug effects, Excitatory Postsynaptic Potentials drug effects, Excitatory Postsynaptic Potentials physiology, Female, Letrozole, Male, Mice, Mice, Inbred C57BL, Neuronal Plasticity drug effects, Nitriles pharmacology, Organ Culture Techniques, Rats, Triazoles pharmacology, Aromatase physiology, Aromatase Inhibitors pharmacology, Basolateral Nuclear Complex physiology, Neuronal Plasticity physiology, Sex Characteristics
- Abstract
The basolateral amygdala (BLA) integrates sensory input from cortical and subcortical regions, a function that requires marked synaptic plasticity. Here we provide evidence that cytochrome P450 aromatase (AROM), the enzyme converting testosterone to 17β-estradiol (E2), contributes to the regulation of this plasticity in a sex-specific manner. We show that AROM is expressed in the BLA, particularly in the basolateral nucleus (BL), in male and female rodents. Systemic administration of the AROM inhibitor letrozole reduced spine synapse density in the BL of adult female mice but not in the BL of male mice. Similarly, in organotypic corticoamygdalar slice cultures from immature rats, treatment with letrozole significantly reduced spine synapses in the BL only in cultures derived from females. In addition, letrozole sex-specifically altered synaptic properties in the BL: in acute slices from juvenile (prepubertal) female rats, wash-in of letrozole virtually abolished long-term potentiation (LTP), whereas it did not prevent the generation of LTP in the slices from males. Together, these data indicate that neuron-derived E2 modulates synaptic plasticity in rodent BLA sex-dependently. As protein expression levels of AROM, estrogen and androgen receptors did not differ between males and females and were not sex-specifically altered by letrozole, the findings suggest sex-specific mechanisms of E2 signaling. SIGNIFICANCE STATEMENT The basolateral amygdala (BLA) is a key structure of the fear circuit. This research reveals a sexually dimorphic regulation of synaptic plasticity in the BLA involving neuronal aromatase, which produces the neurosteroid 17β-estradiol (E2). As male and female neurons in rodent BLA responded differently to aromatase inhibition both in vivo and in vitro , our findings suggest that E2 signaling in BLA neurons is regulated sex-dependently, presumably via mechanisms that have been established during sexual determination. These findings could be relevant for the understanding of sex differences in mood disorders and of the side effects of cytochrome P450 aromatase inhibitors, which are frequently used for breast cancer therapy., (Copyright © 2017 the authors 0270-6474/17/371532-14$15.00/0.)
- Published
- 2017
- Full Text
- View/download PDF
17. Progranulin protects against exaggerated axonal injury and astrogliosis following traumatic brain injury.
- Author
-
Menzel L, Kleber L, Friedrich C, Hummel R, Dangel L, Winter J, Schmitz K, Tegeder I, and Schäfer MK
- Subjects
- Animals, Animals, Newborn, Astrocytes drug effects, Astrocytes pathology, Axons metabolism, Blood-Brain Barrier pathology, Calcium-Binding Proteins metabolism, Cells, Cultured, Cytokines genetics, Cytokines metabolism, Disease Models, Animal, Gene Expression drug effects, Gene Expression genetics, Gene Expression Regulation drug effects, Gene Expression Regulation genetics, Gliosis pathology, Granulins, Intercellular Signaling Peptides and Proteins genetics, Intercellular Signaling Peptides and Proteins metabolism, Lipopolysaccharides pharmacology, Mice, Mice, Inbred C57BL, Mice, Transgenic, Microfilament Proteins metabolism, Nerve Tissue Proteins metabolism, Nervous System Diseases etiology, Nervous System Diseases pathology, Progranulins, Axons pathology, Brain Injuries, Traumatic complications, Brain Injuries, Traumatic pathology, Gliosis etiology, Gliosis prevention & control, Intercellular Signaling Peptides and Proteins therapeutic use
- Abstract
In response to traumatic brain injury (TBI) microglia/macrophages and astrocytes release inflammatory mediators with dual effects on secondary brain damage progression. The neurotrophic and anti-inflammatory glycoprotein progranulin (PGRN) attenuates neuronal damage and microglia/macrophage activation in brain injury but mechanisms are still elusive. Here, we studied histopathology, neurology and gene expression of inflammatory markers in PGRN-deficient mice (Grn
-/- ) 24 h and 5 days after experimental TBI. Grn-/- mice displayed increased perilesional axonal injury even though the overall brain tissue loss and neurological consequences were similar to wild-type mice. Brain inflammation was elevated in Grn-/- mice as reflected by increased transcription of pro-inflammatory cytokines TNFα, IL-1β, IL-6, and decreased transcription of the anti-inflammatory cytokine IL-10. However, numbers of Iba1+ microglia/macrophages and immigrated CD45+ leukocytes were similar at perilesional sites while determination of IgG extravasation suggested stronger impairment of blood brain barrier integrity in Grn-/- compared to wild-type mice. Most strikingly, Grn-/- mice displayed exaggerated astrogliosis 5 days after TBI as demonstrated by anti-GFAP immunohistochemistry and immunoblot. GFAP+ astrocytes at perilesional sites were immunolabelled for iNOS and TNFα suggesting that pro-inflammatory activation of astrocytes was attenuated by PGRN. Accordingly, recombinant PGRN (rPGRN) attenuated LPS- and cytokine-evoked iNOS and TNFα mRNA expression in cultured astrocytes. Moreover, intracerebroventricular administration of rPGRN immediately before trauma reduced brain damage and neurological deficits, and restored normal levels of cytokine transcription, axonal injury and astrogliosis 5 days after TBI in Grn-/- mice. Our results show that endogenous and recombinant PGRN limit axonal injury and astrogliosis and suggest therapeutic potential of PGRN in TBI. GLIA 2017;65:278-292., (© 2016 Wiley Periodicals, Inc.)- Published
- 2017
- Full Text
- View/download PDF
18. L1 syndrome diagnosis complemented with functional analysis of L1CAM variants located to the two N-terminal Ig-like domains.
- Author
-
Christaller WA, Vos Y, Gebre-Medhin S, Hofstra RM, and Schäfer MK
- Subjects
- Amino Acid Sequence, Binding Sites genetics, Cell Communication genetics, Cell Membrane metabolism, Endoplasmic Reticulum metabolism, Family Health, Female, Genetic Diseases, X-Linked diagnosis, Genetic Diseases, X-Linked metabolism, HEK293 Cells, Humans, Immunoblotting, Immunoglobulin Domains genetics, Intellectual Disability diagnosis, Intellectual Disability metabolism, Male, Microscopy, Confocal, Neural Cell Adhesion Molecule L1 metabolism, Pedigree, Sequence Homology, Amino Acid, Spastic Paraplegia, Hereditary diagnosis, Spastic Paraplegia, Hereditary metabolism, Genetic Diseases, X-Linked genetics, Genetic Predisposition to Disease genetics, Intellectual Disability genetics, Mutation, Missense, Neural Cell Adhesion Molecule L1 genetics, Spastic Paraplegia, Hereditary genetics
- Abstract
L1CAM gene mutations cause neurodevelopmental disorders collectively termed L1 syndrome. Insufficient information about L1CAM variants complicates clinical prognosis, genetic diagnosis and genetic counseling. We combined clinical data, in silico effect predictions and functional analysis of four L1CAM variants, p.I37N, p.T38M, p.M172I and p.D202Y, located to the two N-terminal Ig-like domains present in five families with symptoms of L1 syndrome. Software tools predicted destabilizing effects of p.I37N and p.D202Y but results for p.T38M and p.M172I were inconsistent. Cell surface expression of mutant proteins L1-T38M, L1-M172I and L1-D202Y was normal. Conversely, L1-I37N accumulated in the endoplasmic reticulum (ER) and showed temperature-sensitive protein maturation suggesting that p.I37N induces protein misfolding. L1CAM-mediated cell-cell aggregation was severely impaired by L1CAM variants p.I37N, p.M172I and p.D202Y but was preserved by the variant p.T38M. Our experimental data indicate that protein misfolding and accumulation in the ER affect function of the L1CAM variant p.I37N whereas the variants p.M172I and p.D202Y impair homophilic interaction at the cell surface., (© 2016 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd.)
- Published
- 2017
- Full Text
- View/download PDF
19. Down-regulation of neuronal L1 cell adhesion molecule expression alleviates inflammatory neuronal injury.
- Author
-
Menzel L, Paterka M, Bittner S, White R, Bobkiewicz W, van Horssen J, Schachner M, Witsch E, Kuhlmann T, Zipp F, and Schäfer MK
- Subjects
- Aged, Animals, Axons drug effects, Axons pathology, Coculture Techniques, Disease Models, Animal, Down-Regulation drug effects, Encephalomyelitis, Autoimmune, Experimental chemically induced, Female, HEK293 Cells, Humans, Male, Mice, Mice, Inbred C57BL, Mice, Transgenic, Middle Aged, Myelin Proteolipid Protein pharmacology, Myelin-Oligodendrocyte Glycoprotein pharmacology, Nerve Tissue Proteins genetics, Nerve Tissue Proteins metabolism, Neural Cell Adhesion Molecule L1 genetics, Neurons drug effects, Peptide Fragments pharmacology, Synapsins genetics, Synapsins metabolism, T-Lymphocytes drug effects, T-Lymphocytes pathology, Down-Regulation physiology, Encephalomyelitis, Autoimmune, Experimental metabolism, Encephalomyelitis, Autoimmune, Experimental pathology, Neural Cell Adhesion Molecule L1 metabolism, Neurons metabolism, T-Lymphocytes physiology
- Abstract
In multiple sclerosis (MS), the immune cell attack leads to axonal injury as a major cause for neurological disability. Here, we report a novel role of the cell adhesion molecule L1 in the crosstalk between the immune and nervous systems. L1 was found to be expressed by CNS axons of MS patients and human T cells. In MOG
35-55 -induced murine experimental neuroinflammation, CD4+ T cells were associated with degenerating axons in the spinal cord, both expressing L1. However, neuronal L1 expression in the spinal cord was reduced, while levels of the transcriptional repressor REST (RE1-Silencing Transcription Factor) were up-regulated. In PLP139-151 -induced relapsing-remitting neuroinflammation, L1 expression was low at the peak stage of disease, reached almost normal levels in the remission stage, but decreased again during disease relapse indicating adaptive expression regulation of L1. In vitro, activated CD4+ T cells caused contact-dependent down-regulation of L1, up-regulation of its repressor REST and axonal injury in co-cultured neurons. T cell adhesion to neurons and axonal injury were prevented by an antibody blocking L1 suggesting that down-regulation of L1 ameliorates neuroinflammation. In support of this hypothesis, antibody-mediated blocking of L1 in C57BL/6 mice as well as neuron-specific depletion of L1 in synapsinCre × L1fl/fl mice reduces disease severity and axonal pathology despite unchanged immune cell infiltration of the CNS. Our data suggest that down-regulation of neuronal L1 expression is an adaptive process of neuronal self-defense in response to pro-inflammatory T cells, thereby alleviating immune-mediated axonal injury.- Published
- 2016
- Full Text
- View/download PDF
20. Plasticity Related Gene 3 (PRG3) overcomes myelin-associated growth inhibition and promotes functional recovery after spinal cord injury.
- Author
-
Broggini T, Schnell L, Ghoochani A, Mateos JM, Buchfelder M, Wiendieck K, Schäfer MK, Eyupoglu IY, and Savaskan NE
- Subjects
- Animals, Axons metabolism, Mice, Mice, Transgenic, Neurites metabolism, Neurons metabolism, Phosphoric Monoester Hydrolases genetics, Spinal Cord Injuries genetics, Myelin Sheath metabolism, Phosphoric Monoester Hydrolases metabolism, Recovery of Function physiology, Spinal Cord Injuries metabolism
- Abstract
The Plasticity Related Gene family covers five, brain-specific, transmembrane proteins (PRG1-5, also termed LPPR1-5) that operate in neuronal plasticity during development, aging and brain trauma. Here we investigated the role of the PRG family on axonal and filopodia outgrowth. Comparative analysis revealed the strongest outgrowth induced by PRG3 (LPPR1). During development, PRG3 is ubiquitously located at the tip of neuronal processes and at the plasma membrane and declines with age. In utero electroporation of PRG3 induced dendritic protrusions and accelerated spine formations in cortical pyramidal neurons. The neurite growth promoting activity of PRG3 requires RasGRF1 (RasGEF1/Cdc25) mediated downstream signaling. Moreover, in axon collapse assays, PRG3-induced neurites resisted growth inhibitors such as myelin, Nogo-A (Reticulon/RTN-4), thrombin and LPA and impeded the RhoA-Rock-PIP5K induced neurite repulsion. Transgenic adult mice with constitutive PRG3 expression displayed strong axonal sprouting distal to a spinal cord lesion. Moreover, fostered PRG3 expression promoted complex motor-behavioral recovery compared to wild type controls as revealed in the Schnell swim test (SST). Thus, PRG3 emerges as a developmental RasGRF1-dependent conductor of filopodia formation and axonal growth enhancer. PRG3-induced neurites resist brain injury-associated outgrowth inhibitors and contribute to functional recovery after spinal cord lesions. Here, we provide evidence that PRG3 operates as an essential neuronal growth promoter in the nervous system. Maintaining PRG3 expression in aging brain may turn back the developmental clock for neuronal regeneration and plasticity., Competing Interests: The authors declare no competing financial conflict of interests.
- Published
- 2016
- Full Text
- View/download PDF
21. l-DOPA-induced dyskinesia is associated with a deficient numerical downregulation of striatal tyrosine hydroxylase mRNA-expressing neurons.
- Author
-
Klietz M, Keber U, Carlsson T, Chiu WH, Höglinger GU, Weihe E, Schäfer MK, and Depboylu C
- Subjects
- Animals, Corpus Striatum metabolism, Corpus Striatum pathology, Disease Models, Animal, Down-Regulation drug effects, Dyskinesia, Drug-Induced pathology, Enkephalins metabolism, Male, Medial Forebrain Bundle, Mice, Inbred C57BL, Neuronal Plasticity, Neurons metabolism, Neurons pathology, Oxidopamine, Protein Precursors metabolism, RNA, Messenger metabolism, Antiparkinson Agents adverse effects, Corpus Striatum drug effects, Dyskinesia, Drug-Induced metabolism, Levodopa adverse effects, Neurons drug effects, Tyrosine 3-Monooxygenase metabolism
- Abstract
l-3,4-Dihydroxyphenylalanine (l-DOPA) is the therapeutic gold standard in Parkinson's disease. However, most patients develop debilitating abnormal involuntary movements termed l-DOPA-induced dyskinesia (LID) as therapy-complicating side effects. The underlying mechanisms of LID pathogenesis are still not fully understood. Recent evidence suggests an involvement of striatal tyrosine hydroxylase (TH) protein-expressing neurons, as they are capable of endogenously producing l-DOPA and possibly dopamine. The aim of this study was to elucidate changes of TH transcription in the striatum and nucleus accumbens that occur under experimental conditions of LID. Mice with a unilateral 6-hydroxydopamine-induced lesion of the medial forebrain bundle were treated daily with l-DOPA for 15days to provoke dyskinesia. In situ hybridization analysis revealed a significant numerical decrease of TH mRNA-positive neurons in the striatum and nucleus accumbens of mice not exhibiting LID, whereas dyskinetic animals failed to show this reduction of TH transcription. Interestingly, similar changes were observed in intact non-deafferentiated striata, demonstrating an l-DOPA-responsive transcriptional TH regulation independently from nigrostriatal lesion severity. Consolidation with our previous study on TH protein level (Keber et al., 2015) impressively highlights that LID is associated with both a deficient downregulation of TH transcription and an excessive translation of TH protein in intrastriatal neurons. As TH protein levels in comparison to mRNA levels showed a stronger correlation with development and severity of LID, antidyskinetic treatment strategies should focus on translational and posttranslational modulations of TH as a promising target., (Copyright © 2016 IBRO. Published by Elsevier Ltd. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
22. Lack of NG2 exacerbates neurological outcome and modulates glial responses after traumatic brain injury.
- Author
-
Huang C, Sakry D, Menzel L, Dangel L, Sebastiani A, Krämer T, Karram K, Engelhard K, Trotter J, and Schäfer MK
- Subjects
- Animals, Antigens genetics, Arginase metabolism, Blood-Brain Barrier metabolism, Brain pathology, Brain Injuries pathology, Calcium-Binding Proteins metabolism, Capillary Permeability physiology, Cell Count, Cells, Cultured, Chemokine CXCL13 metabolism, Cohort Studies, Disease Models, Animal, Gliosis metabolism, Gliosis pathology, Leukocyte Common Antigens metabolism, Mice, Inbred C57BL, Mice, Knockout, Microfilament Proteins metabolism, Neuroglia pathology, Proteoglycans genetics, RNA, Messenger metabolism, Severity of Illness Index, Antigens metabolism, Brain metabolism, Brain Injuries metabolism, Neuroglia metabolism, Proteoglycans metabolism
- Abstract
Traumatic brain injury (TBI) is a major cause of death and disability. The underlying pathophysiology is characterized by secondary processes including neuronal death and gliosis. To elucidate the role of the NG2 proteoglycan we investigated the response of NG2-knockout mice (NG2-KO) to TBI. Seven days after TBI behavioral analysis, brain damage volumetry and assessment of blood brain barrier integrity demonstrated an exacerbated response of NG2-KO compared to wild-type (WT) mice. Reactive astrocytes and expression of the reactive astrocyte and neurotoxicity marker Lcn2 (Lipocalin-2) were increased in the perilesional brain tissue of NG2-KO mice. In addition, microglia/macrophages with activated morphology were increased in number and mRNA expression of the M2 marker Arg1 (Arginase 1) was enhanced in NG2-KO mice. While TBI-induced expression of pro-inflammatory cytokine genes was unchanged between genotypes, PCR array screening revealed a marked TBI-induced up-regulation of the C-X-C motif chemokine 13 gene Cxcl13 in NG2-KO mice. CXCL13, known to attract immune cells to the inflamed brain, was expressed by activated perilesional microglia/macrophages seven days after TBI. Thirty days after TBI, NG2-KO mice still exhibited more pronounced neurological deficits than WT mice, up-regulation of Cxcl13, enhanced CD45+ leukocyte infiltration and a relative increase of activated Iba-1+/CD45+ microglia/macrophages. Our study demonstrates that lack of NG2 exacerbates the neurological outcome after TBI and associates with abnormal activation of astrocytes, microglia/macrophages and increased leukocyte recruitment to the injured brain. These findings suggest that NG2 may counteract neurological deficits and adverse glial responses in TBI., (© 2015 Wiley Periodicals, Inc.)
- Published
- 2016
- Full Text
- View/download PDF
23. Posttraumatic Propofol Neurotoxicity Is Mediated via the Pro-Brain-Derived Neurotrophic Factor-p75 Neurotrophin Receptor Pathway in Adult Mice.
- Author
-
Sebastiani A, Granold M, Ditter A, Sebastiani P, Gölz C, Pöttker B, Luh C, Schaible EV, Radyushkin K, Timaru-Kast R, Werner C, Schäfer MK, Engelhard K, Moosmann B, and Thal SC
- Subjects
- Animals, Blood Pressure, Caspase 3 biosynthesis, Cell Death, Gait, Heart Rate, Immunoassay, Mice, Mice, Inbred C57BL, RNA, Messenger biosynthesis, Receptor, Nerve Growth Factor antagonists & inhibitors, Spectrin metabolism, Brain Injuries drug therapy, Brain Injuries physiopathology, Brain-Derived Neurotrophic Factor biosynthesis, Propofol pharmacology, Receptor, Nerve Growth Factor metabolism
- Abstract
Objectives: The gamma-aminobutyric acid modulator propofol induces neuronal cell death in healthy immature brains by unbalancing neurotrophin homeostasis via p75 neurotrophin receptor signaling. In adulthood, p75 neurotrophin receptor becomes down-regulated and propofol loses its neurotoxic effect. However, acute brain lesions, such as traumatic brain injury, reactivate developmental-like programs and increase p75 neurotrophin receptor expression, probably to foster reparative processes, which in turn could render the brain sensitive to propofol-mediated neurotoxicity. This study investigates the influence of delayed single-bolus propofol applications at the peak of p75 neurotrophin receptor expression after experimental traumatic brain injury in adult mice., Design: Randomized laboratory animal study., Setting: University research laboratory., Subjects: Adult C57BL/6N and nerve growth factor receptor-deficient mice., Interventions: Sedation by IV propofol bolus application delayed after controlled cortical impact injury., Measurements and Main Results: Propofol sedation at 24 hours after traumatic brain injury increased lesion volume, enhanced calpain-induced αII-spectrin cleavage, and increased cell death in perilesional tissue. Thirty-day postinjury motor function determined by CatWalk (Noldus Information Technology, Wageningen, The Netherlands) gait analysis was significantly impaired in propofol-sedated animals. Propofol enhanced pro-brain-derived neurotrophic factor/brain-derived neurotrophic factor ratio, which aggravates p75 neurotrophin receptor-mediated cell death. Propofol toxicity was abolished both by pharmacologic inhibition of the cell death domain of the p75 neurotrophin receptor (TAT-Pep5) and in mice lacking the extracellular neurotrophin binding site of p75 neurotrophin receptor., Conclusions: This study provides first evidence that propofol sedation after acute brain lesions can have a deleterious impact and implicates a role for the pro-brain-derived neurotrophic factor-p75 neurotrophin receptor pathway. This observation is important as sedation with propofol and other compounds with GABA receptor activity are frequently used in patients with acute brain pathologies to facilitate sedation or surgical and interventional procedures.
- Published
- 2016
- Full Text
- View/download PDF
24. Proneurotrophin Binding to P75 Neurotrophin Receptor (P75ntr) Is Essential for Brain Lesion Formation and Functional Impairment after Experimental Traumatic Brain Injury.
- Author
-
Sebastiani A, Gölz C, Werner C, Schäfer MK, Engelhard K, and Thal SC
- Subjects
- Animals, Behavior, Animal, Brain Injuries pathology, Brain Injuries physiopathology, Disease Models, Animal, Female, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Protein Binding, RNA, Messenger metabolism, Receptors, Nerve Growth Factor antagonists & inhibitors, Receptors, Nerve Growth Factor genetics, Brain Injuries drug therapy, Brain Injuries metabolism, Cell Death drug effects, Receptors, Nerve Growth Factor metabolism, Signal Transduction drug effects
- Abstract
Traumatic brain injury (TBI) initiates an excessive mediator release of e.g. neurotrophins, which promote neuronal survival, differentiation, and modulate synaptic plasticity. Paradoxically, mature forms of neurotrophins promote neuronal survival, whereas unprocessed forms of neurotrophins induce cell death through p75 neurotrophin receptor (p75NTR) signaling. p75NTR is widely expressed during synaptogenesis and is subsequently downregulated in adulthood. Repair mechanisms after acute cerebral insults can reactivate its expression. Therefore, the influence of p75NTR on secondary brain damage was addressed. mRNA levels of p75NTR and its ligands were quantified in brain tissue up to 7 days after experimental TBI (controlled cortical impact; CCI). Brain damage, motor function and inflammatory marker gene expression were determined in mice lacking the proneurotrophin-binding site of the p75NTR protein (NGFR(-/-)) and wild type littermates (NGFR(+/+)) 24 h and 5 days after CCI. In addition, the effect of TAT-Pep5 (pharmacological inhibitor of the intracellular p75NTR death domain) on lesion volume was evaluated 24 h after insult. p75NTR mRNA levels were induced nine-fold by TBI. In NGFR(-/-) mice, lesion volume was reduced by 29% at 24 h and by 21% 5 days after CCI. Motor coordination was significantly improved 24 h after trauma compared with the wild type. Pharmacological inhibition of the p75NTR signaling reduced lesion volume by 18%. The present study presents first time evidence that genetic mutation of the neurotrophin interaction site of p75NTR strongly limits post-traumatic cell death. In addition, we revealed pharmacological targeting of the intracellular p75NTR cell death domain as a promising approach to limit acute brain damage.
- Published
- 2015
- Full Text
- View/download PDF
25. Striatal tyrosine hydroxylase-positive neurons are associated with L-DOPA-induced dyskinesia in hemiparkinsonian mice.
- Author
-
Keber U, Klietz M, Carlsson T, Oertel WH, Weihe E, Schäfer MK, Höglinger GU, and Depboylu C
- Subjects
- Amphetamine pharmacology, Animals, Antiparkinson Agents adverse effects, Disease Models, Animal, Dyskinesia, Drug-Induced etiology, Levodopa adverse effects, Male, Medial Forebrain Bundle drug effects, Medial Forebrain Bundle injuries, Mice, Mice, Inbred C57BL, Oxidopamine toxicity, Parkinson Disease drug therapy, Parkinson Disease etiology, Phosphopyruvate Hydratase metabolism, Proto-Oncogene Proteins c-fos metabolism, Serotonin Plasma Membrane Transport Proteins metabolism, Statistics, Nonparametric, Corpus Striatum pathology, Dyskinesia, Drug-Induced pathology, Functional Laterality physiology, Neurons metabolism, Tyrosine 3-Monooxygenase metabolism
- Abstract
L-3,4-Dihydroxyphenylalanine (L-DOPA) is the therapeutic gold standard in Parkinson's disease. However, long-term treatment is complicated by the induction of debilitating abnormal involuntary movements termed L-DOPA-induced dyskinesias (LIDs). Until today the underlying mechanisms of LID pathogenesis are not fully understood. The aim of this study was to reveal new factors, which may be involved in the induction of LID. We have focused on the expression of striatal tyrosine hydroxylase-positive (TH+) neurons, which are capable of producing either L-DOPA or dopamine (DA) in target areas of ventral midbrain DAergic neurons. To address this issue, a daily L-DOPA dose was administered over the course of 15 days to mice with unilateral 6-hydroxydopamine-induced lesions of the medial forebrain bundle and LIDs were evaluated. Remarkably, the number of striatal TH+ neurons strongly correlated with both induction and severity of LID as well as ΔFosB expression as an established molecular marker for LID. Furthermore, dyskinetic mice showed a marked augmentation of serotonergic fiber innervation in the striatum, enabling the decarboxylation of L-DOPA to DA. Axial, limb and orolingual dyskinesias were predominantly associated with TH+ neurons in the lateral striatum, whereas medially located TH+ neurons triggered locomotive rotations. In contrast, identified accumbal and cortical TH+ cells did not contribute to the generation of LID. Thus, striatal TH+ cells and serotonergic terminals may cooperatively synthesize DA and subsequently contribute to supraphysiological synaptic DA concentrations, an accepted cause in LID pathogenesis., (Copyright © 2015 IBRO. Published by Elsevier Ltd. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
26. Early subcellular Ca2+ remodelling and increased propensity for Ca2+ alternans in left atrial myocytes from hypertensive rats.
- Author
-
Pluteanu F, Heß J, Plackic J, Nikonova Y, Preisenberger J, Bukowska A, Schotten U, Rinne A, Kienitz MC, Schäfer MK, Weihe E, Goette A, and Kockskämper J
- Subjects
- Animals, Arrhythmias, Cardiac physiopathology, Calcium Channels, L-Type metabolism, Disease Models, Animal, Heart Atria pathology, Hypertension pathology, Male, Myocytes, Cardiac pathology, Patch-Clamp Techniques, Rats, Rats, Inbred SHR, Rats, Inbred WKY, Risk Factors, Sarcoplasmic Reticulum metabolism, Sodium metabolism, Sodium-Calcium Exchanger metabolism, Tachycardia epidemiology, Tachycardia metabolism, Tachycardia physiopathology, Arrhythmias, Cardiac epidemiology, Arrhythmias, Cardiac metabolism, Atrial Remodeling physiology, Calcium metabolism, Heart Atria metabolism, Hypertension metabolism, Myocytes, Cardiac metabolism
- Abstract
Aims: Hypertension is a major risk factor for atrial fibrillation. We hypothesized that arterial hypertension would alter atrial myocyte calcium (Ca2+) handling and that these alterations would serve to trigger atrial tachyarrhythmias., Methods and Results: Left atria or left atrial (LA) myocytes were isolated from spontaneously hypertensive rats (SHR) or normotensive Wistar-Kyoto (WKY) controls. Early after the onset of hypertension, at 3 months of age, there were no differences in Ca2+ transients (CaTs) or expression and phosphorylation of Ca2+ handling proteins between SHR and WKY. At 7 months of age, when left ventricular (LV) hypertrophy had progressed and markers of fibrosis were increased in left atrium, CaTs (at 1 Hz stimulation) were still unchanged. Subcellular alterations in Ca2+ handling were observed, however, in SHR atrial myocytes including (i) reduced expression of the α1C subunit of and reduced Ca2+ influx through L-type Ca2+ channels, (ii) reduced expression of ryanodine receptors with increased phosphorylation at Ser2808, (iii) decreased activity of the Na+ / Ca2+ exchanger (at unaltered intracellular Na+ concentration), and (iv) increased SR Ca2+ load with reduced fractional release. These changes were associated with an increased propensity of SHR atrial myocytes to develop frequency-dependent, arrhythmogenic Ca2+ alternans., Conclusions: In SHR, hypertension induces early subcellular LA myocyte Ca2+ remodelling during compensated LV hypertrophy. In basal conditions, atrial myocyte CaTs are not changed. At increased stimulation frequency, however, SHR atrial myocytes become more prone to arrhythmogenic Ca2+ alternans, suggesting a link between hypertension, atrial Ca2+ homeostasis, and development of atrial tachyarrhythmias., (Published on behalf of the European Society of Cardiology. All rights reserved. © The Author 2015. For permissions please email: journals.permissions@oup.com.)
- Published
- 2015
- Full Text
- View/download PDF
27. Satb2-independent acquisition of the cholinergic sudomotor phenotype in rodents.
- Author
-
Schütz B, Schäfer MK, Gördes M, Eiden LE, and Weihe E
- Subjects
- Animals, Animals, Newborn, Antibody Specificity, Biomarkers metabolism, Extremities innervation, Female, Gene Expression Regulation, Male, Matrix Attachment Region Binding Proteins deficiency, Matrix Attachment Region Binding Proteins genetics, Mice, Inbred BALB C, Peptides metabolism, Phenotype, RNA, Messenger genetics, RNA, Messenger metabolism, Rats, Wistar, Stellate Ganglion metabolism, Sweat Glands innervation, Sympathetic Nervous System metabolism, Time Factors, Transcription Factors deficiency, Transcription Factors genetics, Vesicular Acetylcholine Transport Proteins, Cholinergic Neurons metabolism, Matrix Attachment Region Binding Proteins metabolism, Motor Activity, Transcription Factors metabolism
- Abstract
Expression of Satb2 (Special AT-rich sequence-binding protein-2) elicits expression of the vesicular acetylcholine transporter (VAChT) and choline acetyltransferase (ChAT) in cultured rat sympathetic neurons exposed to soluble differentiation factors. Here, we determined whether or not Satb2 plays a similar role in cholinergic differentiation in vivo, by comparing the postnatal profile of Satb2 expression in the rodent stellate ganglion to that of VAChT and ChAT. Throughout postnatal development, VAChT and ChAT were found to be co-expressed in a numerically stable subpopulation of rat stellate ganglion neurons. Nerve fibers innervating rat forepaw sweat glands on P1 were VAChT immunoreactive, while ChAT was detectable at this target only after P5. The postnatal abundance of VAChT transcripts in the stellate ganglion was at maximum already on P1, whereas ChAT mRNA levels increased from low levels on P1 to reach maximum levels between P5 and P21. Satb2 mRNA was detected in cholinergic neurons in the stellate ganglion beginning with P8, thus coincident with the onset of unequivocal detection of ChAT immunoreactivity in forepaw sweat gland endings. Satb2 knockout mice exhibited no change in the P1 cholinergic VAChT/ChAT co-phenotype in stellate ganglion neurons. Thus, cholinergic phenotype maturation involves first, early target (sweat-gland)-independent expression and trafficking of VAChT, and later, potentially target- and Satb2-dependent elevation of ChAT mRNA and protein transport into sweat gland endings. In rat sudomotor neurons that, unlike mouse sudomotor neurons, co-express calcitonin gene-related peptide (CGRP), Satb2 may also be related to the establishment of species-specific neuropeptide co-phenotypes during postnatal development.
- Published
- 2015
- Full Text
- View/download PDF
28. Perivascular microglia promote blood vessel disintegration in the ischemic penumbra.
- Author
-
Jolivel V, Bicker F, Binamé F, Ploen R, Keller S, Gollan R, Jurek B, Birkenstock J, Poisa-Beiro L, Bruttger J, Opitz V, Thal SC, Waisman A, Bäuerle T, Schäfer MK, Zipp F, and Schmidt MHH
- Subjects
- Animals, Blood-Brain Barrier pathology, Blood-Brain Barrier physiopathology, Brain pathology, Brain Ischemia pathology, CX3C Chemokine Receptor 1, Cell Line, Disease Models, Animal, Green Fluorescent Proteins genetics, Green Fluorescent Proteins metabolism, Mice, 129 Strain, Mice, Inbred C57BL, Mice, Transgenic, Microglia pathology, Phagocytosis physiology, Receptors, Chemokine genetics, Receptors, Chemokine metabolism, Stroke pathology, Brain blood supply, Brain physiopathology, Brain Ischemia physiopathology, Microglia physiology, Stroke physiopathology
- Abstract
The contribution of microglia to ischemic cortical stroke is of particular therapeutic interest because of the impact on the survival of brain tissue in the ischemic penumbra, a region that is potentially salvable upon a brain infarct. Whether or not tissue in the penumbra survives critically depends on blood flow and vessel perfusion. To study the role of microglia in cortical stroke and blood vessel stability, CX3CR1(+/GFP) mice were subjected to transient middle cerebral artery occlusion and then microglia were investigated using time-lapse two-photon microscopy in vivo. Soon after reperfusion, microglia became activated in the stroke penumbra and started to expand cellular protrusions towards adjacent blood vessels. All microglia in the penumbra were found associated with blood vessels within 24 h post reperfusion and partially fully engulfed them. In the same time frame blood vessels became permissive for blood serum components. Migration assays in vitro showed that blood serum proteins leaking into the tissue provided molecular cues leading to the recruitment of microglia to blood vessels and to their activation. Subsequently, these perivascular microglia started to eat up endothelial cells by phagocytosis, which caused an activation of the local endothelium and contributed to the disintegration of blood vessels with an eventual break down of the blood brain barrier. Loss-of-microglia-function studies using CX3CR1(GFP/GFP) mice displayed a decrease in stroke size and a reduction in the extravasation of contrast agent into the brain penumbra as measured by MRI. Potentially, medication directed at inhibiting microglia activation within the first day after stroke could stabilize blood vessels in the penumbra, increase blood flow, and serve as a valuable treatment for patients suffering from ischemic stroke.
- Published
- 2015
- Full Text
- View/download PDF
29. Golgi fragmentation in pmn mice is due to a defective ARF1/TBCE cross-talk that coordinates COPI vesicle formation and tubulin polymerization.
- Author
-
Bellouze S, Schäfer MK, Buttigieg D, Baillat G, Rabouille C, and Haase G
- Subjects
- ADP-Ribosylation Factor 1 genetics, Amyotrophic Lateral Sclerosis genetics, Animals, COP-Coated Vesicles genetics, Coat Protein Complex I metabolism, Disease Models, Animal, Golgi Apparatus chemistry, Humans, Mice, Mice, Inbred C57BL, Molecular Chaperones genetics, Motor Neurons chemistry, Motor Neurons metabolism, Muscular Atrophy, Spinal genetics, Polymerization, Signal Transduction, Tubulin chemistry, ADP-Ribosylation Factor 1 metabolism, Amyotrophic Lateral Sclerosis metabolism, COP-Coated Vesicles metabolism, Golgi Apparatus metabolism, Molecular Chaperones metabolism, Muscular Atrophy, Spinal metabolism, Tubulin metabolism
- Abstract
Golgi fragmentation is an early hallmark of many neurodegenerative diseases but its pathophysiological relevance and molecular mechanisms are unclear. We here demonstrate severe and progressive Golgi fragmentation in motor neurons of progressive motor neuronopathy (pmn) mice due to loss of the Golgi-localized tubulin-binding cofactor E (TBCE). Loss of TBCE in mutant pmn and TBCE-depleted motor neuron cultures causes defects in Golgi-derived microtubules, as expected, but surprisingly also reduced levels of COPI subunits, decreased recruitment of tethering factors p115/GM130 and impaired Golgi SNARE-mediated vesicle fusion. Conversely, ARF1, which stimulates COPI vesicle formation, enhances the recruitment of TBCE to the Golgi, increases polymerization of Golgi-derived microtubules and rescues TBCE-linked Golgi fragmentation. These data indicate an ARF1/TBCE-mediated cross-talk that coordinates COPI formation and tubulin polymerization at the Golgi. We conclude that interruption of this cross-talk causes Golgi fragmentation in pmn mice and hypothesize that similar mechanisms operate in human amyotrophic lateral sclerosis and spinal muscular atrophy., (© The Author 2014. Published by Oxford University Press. All rights reserved. For Permissions, please email: journals.permissions@oup.com.)
- Published
- 2014
- Full Text
- View/download PDF
30. Transcriptional and structural plasticity of tyrosine hydroxylase expressing neurons in both striatum and nucleus accumbens following dopaminergic denervation.
- Author
-
Depboylu C, Klietz M, Maurer L, Oertel WH, Kobayashi K, Weihe E, Höglinger GU, and Schäfer MK
- Subjects
- Animals, Axotomy, Corpus Striatum cytology, Immunohistochemistry, Medial Forebrain Bundle surgery, Mice, Mice, Inbred C57BL, Mice, Transgenic, Nucleus Accumbens cytology, Nucleus Accumbens metabolism, Transcription, Genetic, Transfection, Corpus Striatum metabolism, Neurons metabolism, Tyrosine 3-Monooxygenase biosynthesis
- Abstract
Mice that express green fluorescent protein (GFP) under the control of tyrosine hydroxylase (TH) gene promoter were used to visualize transcriptional as well as structural regulation of TH cells following prolonged dopaminergic denervation. A unilateral lesion of the medial forebrain bundle was induced by 6-hydroxydopamine. In the unlesioned contralateral striatum and nucleus accumbens surprisingly high numbers of resident GFP-positive neurons (about 2653 and 422 per striatum and accumbens, respectively) were observed while only much lower TH-positive neurons (about 214 and 102 per striatum and accumbens, respectively) were detectable. In the lesioned hemisphere the number of GFP neurons was slightly increased already at day 4 by 16% and more at day 40 by 47% while the number of TH-immunoreactive neurons was dramatically increased by 848% at day 4 and by 1139% at day 40 over the control side. Additionally and particularly pronounced in the nucleus accumbens, GFP-positive neurons demonstrated increased sprouting of their projections over time, stronger than observed by TH-immunostaining. The load in TH protein may be essentially determined by post-transcriptional suppression/degradation while GFP may rather reflect the gross transcriptional activity. Thus, permanent dopaminergic pathway injury induces both transcriptional as well as structural plasticity of TH expressing neurons in striatal and accumbal target areas of ventral midbrain dopaminergic neurons., (Copyright © 2014 Elsevier B.V. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
31. Regulators of mitochondrial Ca(2+) homeostasis in cerebral ischemia.
- Author
-
Schäfer MK, Pfeiffer A, Jaeckel M, Pouya A, Dolga AM, and Methner A
- Subjects
- Animals, Cations, Divalent metabolism, Endoplasmic Reticulum metabolism, Humans, Inositol 1,4,5-Trisphosphate Receptors metabolism, Mitochondrial Membrane Transport Proteins metabolism, Mitochondrial Permeability Transition Pore, Neurons metabolism, Sodium-Calcium Exchanger metabolism, Voltage-Dependent Anion Channels metabolism, Brain Ischemia metabolism, Homeostasis, Mitochondria metabolism
- Abstract
Cerebral ischemia is a key pathophysiological feature of various brain insults. Inadequate oxygen supply can manifest regionally in stroke or as a result of traumatic brain injury or globally following cardiac arrest, all leading to irreversible brain damage. Mitochondrial function is essential for neuronal survival, since neurons critically depend on ATP synthesis generated by mitochondrial oxidative phosphorylation. Mitochondrial activity depends on Ca(2+) and is fueled either by Ca(2+) from the extracellular space when triggered by neuronal activity or by Ca(2+) released from the endoplasmic reticulum (ER) and taken up through specialized contact sites between the ER and mitochondria known as mitochondrial-associated ER membranes. The coordination of these Ca(2+) pools is required to synchronize mitochondrial respiration rates and ATP synthesis to physiological demands. In this review, we discuss the role of the proteins involved in mitochondrial Ca(2+) homeostasis in models of ischemia. The proteins include those important for the Ca(2+)-dependent motility of mitochondria and for Ca(2+) transfer from the ER to mitochondria, the tethering proteins that bring the two organelles together, inositol 1,4,5-triphosphate receptors that enable Ca(2+) release from the ER, voltage-dependent anion channels that allow Ca(2+) entry through the highly permeable outer mitochondrial membrane and the mitochondrial Ca(2+) uniporter together with its regulatory proteins that permit Ca(2+) entry into the mitochondrial matrix. Finally, we address those proteins important for the extrusion of Ca(2+) from the mitochondria such as the mitochondrial Na(+)/Ca(2+) exchanger or, if the mitochondrial Ca(2+) concentration exceeds a certain threshold, the mitochondrial permeability transition pore.
- Published
- 2014
- Full Text
- View/download PDF
32. 2-Methoxyestradiol confers neuroprotection and inhibits a maladaptive HIF-1α response after traumatic brain injury in mice.
- Author
-
Schaible EV, Windschügl J, Bobkiewicz W, Kaburov Y, Dangel L, Krämer T, Huang C, Sebastiani A, Luh C, Werner C, Engelhard K, Thal SC, and Schäfer MK
- Subjects
- Alternative Splicing, Animals, Blotting, Western, Brain Ischemia drug therapy, Brain Ischemia metabolism, Brain Ischemia pathology, Cell Nucleus metabolism, Estradiol pharmacology, Exons genetics, Gene Expression Regulation drug effects, Immunohistochemistry, Injections, Intraperitoneal, Male, Membrane Proteins biosynthesis, Mice, Mice, Inbred C57BL, Mitochondrial Proteins biosynthesis, Neurons metabolism, Plasminogen Activator Inhibitor 1 metabolism, Protein Transport, Subcellular Fractions metabolism, Tumor Necrosis Factor-alpha metabolism, Up-Regulation genetics, Up-Regulation physiology, Brain Injuries drug therapy, Brain Injuries metabolism, Estradiol analogs & derivatives, Hypoxia-Inducible Factor 1, alpha Subunit antagonists & inhibitors, Neuroprotective Agents
- Abstract
HIF-1α is pivotal for cellular homeostasis in response to cerebral ischemia. Pharmacological inhibition of HIF-1α may reduce secondary brain damage by targeting post-translational mechanisms associated with its proteasomal degradation and nuclear translocation. This study examined the neuroprotective effects of 2-methoxyestradiol (2ME2), the involved HIF-1α-dependent response, and alternative splicing in exon 14 of HIF-1α (HIF-1α∆Ex14) after traumatic brain injury (TBI) in mice. Intraperitoneal 2ME2 administration 30 min after TBI caused a dose-dependent reduction in secondary brain damage after 24 h. 2ME2 was physiologically tolerated, showed no effects on immune cell brain migration, and mitigated trauma-induced brain expression of neuropathologically relevant HIF-1α target genes encoding for Plasminogen activator inhibitor 1 and tumor necrosis factor alpha. Moreover, TBI-induced expression of pro-apoptotic BNIP3 was attenuated by 2ME2 treatment. Alternatively, spliced HIF-1α∆Ex14 was substantially up-regulated from 6 to 48 h after TBI. In vitro, nuclear location and gene transcription activity of HIF-1α∆Ex14 were impaired compared to full-length HIF-1α, but no effects on nuclear translocation of the transcriptional complex partner HIF-1β were observed. This study demonstrates that 2ME2 confers neuroprotection after TBI. While the role of alternatively spliced HIF-1α∆Ex14 remains elusive, the in vivo data provide evidence that inhibition of a maladaptive HIF-1α-dependent response contributes to the neuroprotective effects of 2ME2. We examined neuroprotective effects of 2-methoxyestradiol (2ME2) and the hypoxia-inducible factor 1-α (HIF-1α) response following traumatic brain injury in mice. Early 2ME2 administration reduced the secondary brain damage and neuronal HIF-1α probably involving ubiquitin proteasome system-mediated degradation. The up-regulation of neuropathological HIF-1α target genes and pro-apoptotic BNIP3 protein was attenuated. We propose that the inhibition of a maladaptive HIF-1α response may contribute to 2ME2-mediated neuroprotection., (© 2014 International Society for Neurochemistry.)
- Published
- 2014
- Full Text
- View/download PDF
33. The sympathetic nervous system modulates CD4(+)Foxp3(+) regulatory T cells via noradrenaline-dependent apoptosis in a murine model of lymphoproliferative disease.
- Author
-
Wirth T, Westendorf AM, Bloemker D, Wildmann J, Engler H, Mollerus S, Wadwa M, Schäfer MK, Schedlowski M, and del Rey A
- Subjects
- Animals, Disease Models, Animal, Female, Lymphoproliferative Disorders metabolism, Male, Mice, Mice, Inbred C57BL, Spleen innervation, Spleen metabolism, Apoptosis, Forkhead Transcription Factors metabolism, Lymphoproliferative Disorders immunology, Norepinephrine metabolism, Sympathetic Nervous System metabolism, T-Lymphocytes, Regulatory metabolism
- Abstract
The sympathetic nervous system (SNS) plays a crucial role in the course and development of autoimmune disease in Fas-deficient lpr/lpr mice. As regulatory T cells (Tregs) are considered important modulators of autoimmune processes, we analyzed the interaction between the SNS and Tregs in this murine model of lymphoproliferative disease. We found that the percentage of Tregs among CD4(+) T cells is increased in the spleen, lymph nodes, and thymus of lpr/lpr mice as compared to age-matched C57Bl/6J (B6) mice. Furthermore, noradrenaline (NA), the main sympathetic neurotransmitter, induced apoptosis in B6- and lpr/lpr-derived Tregs. NA also reduced the frequency of Foxp3(+) cells and Foxp3 mRNA expression via β2-adrenoceptor (β2-AR)-mediated mechanisms in a concentration and time-dependent manner. Destruction of peripheral sympathetic nerves by 6-hydroxydopamine significantly increased the percentage of Tregs in B6 control mice to an extent comparable to aged-matched lpr/lpr mice. The concentration of splenic NA negatively correlated with the frequency of CD4(+)Foxp3(+) Tregs. Additionally, 60days after sympathectomy, a partial recovery of NA concentrations led to Treg percentages comparable to those of intact, vehicle-treated controls. Immunohistochemical analysis of the spleen revealed localization of single Foxp3(+) Tregs in proximity to NA-producing nerve fibers, providing an interface between Tregs and the SNS. Taken together, our data suggest a relation between the degree of splenic sympathetic innervation and the size of the Treg compartment. While there are few examples of endogenous substances capable of affecting Tregs, our results provide a possible explanation of how the magnitude of the Treg compartment in the spleen can be regulated by the SNS., (Copyright © 2014 Elsevier Inc. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
34. Mitochondrial function and energy metabolism in neuronal HT22 cells resistant to oxidative stress.
- Author
-
Pfeiffer A, Jaeckel M, Lewerenz J, Noack R, Pouya A, Schacht T, Hoffmann C, Winter J, Schweiger S, Schäfer MK, and Methner A
- Subjects
- Animals, Calcium metabolism, Cell Count, Cell Death drug effects, Cell Death physiology, Cell Respiration drug effects, Cell Respiration physiology, Deoxyglucose pharmacology, Drug Resistance physiology, Energy Metabolism drug effects, Glucosephosphate Dehydrogenase metabolism, Glutamic Acid pharmacology, Glutathione metabolism, Hexokinase metabolism, Hippocampus drug effects, Lactic Acid metabolism, Mechanistic Target of Rapamycin Complex 1, Mice, Mitochondria drug effects, Mitochondria metabolism, Multiprotein Complexes metabolism, Neurons drug effects, Neurons enzymology, Neurons metabolism, Oligomycins pharmacology, Oxygen Consumption drug effects, Superoxides metabolism, TOR Serine-Threonine Kinases metabolism, Energy Metabolism physiology, Hippocampus physiopathology, Mitochondria physiology, Neurons physiology, Oxidative Stress physiology
- Abstract
Background and Purpose: The hippocampal cell line HT22 is an excellent model for studying the consequences of endogenous oxidative stress. Extracellular glutamate depletes cellular glutathione by blocking the glutamate/cystine antiporter system xc-. Glutathione depletion induces a well-defined programme of cell death characterized by an increase in reactive oxygen species and mitochondrial dysfunction., Experimental Approach: We compared the mitochondrial shape, the abundance of mitochondrial complexes and the mitochondrial respiration of HT22 cells, selected based on their resistance to glutamate, with those of the glutamate-sensitive parental cell line., Key Results: Glutamate-resistant mitochondria were less fragmented and displayed seemingly contradictory features: mitochondrial calcium and superoxide were increased while high-resolution respirometry suggested a reduction in mitochondrial respiration. This was interpreted as a reverse activity of the ATP synthase under oxidative stress, leading to hydrolysis of ATP to maintain or even elevate the mitochondrial membrane potential, suggesting these cells endure ineffective energy metabolism to protect their membrane potential. Glutamate-resistant cells were also resistant to oligomycin, an inhibitor of the ATP synthase, but sensitive to deoxyglucose, an inhibitor of hexokinases. Exchanging glucose with galactose rendered resistant cells 1000-fold more sensitive to oligomycin. These results, together with a strong increase in cytosolic hexokinase 1 and 2, a reduced lactate production and an increased activity of glucose-6-phosphate dehydrogenase, suggest that glutamate-resistant HT22 cells shuttle most available glucose towards the hexose monophosphate shunt to increase glutathione recovery., Conclusions and Implications: These results indicate that mitochondrial and metabolic adaptations play an important role in the resistance of cells to oxidative stress., (© 2013 The British Pharmacological Society.)
- Published
- 2014
- Full Text
- View/download PDF
35. Single administration of tripeptide α-MSH(11-13) attenuates brain damage by reduced inflammation and apoptosis after experimental traumatic brain injury in mice.
- Author
-
Schaible EV, Steinsträßer A, Jahn-Eimermacher A, Luh C, Sebastiani A, Kornes F, Pieter D, Schäfer MK, Engelhard K, and Thal SC
- Subjects
- Animals, Brain drug effects, Brain immunology, Brain metabolism, Brain Injuries immunology, Brain Injuries metabolism, Calcium-Binding Proteins metabolism, Gene Expression, Inflammation Mediators metabolism, Male, Mice, Mice, Inbred C57BL, Microfilament Proteins metabolism, Microglia drug effects, Microglia metabolism, Pro-Opiomelanocortin genetics, Pro-Opiomelanocortin metabolism, Receptor, Melanocortin, Type 1 genetics, Receptor, Melanocortin, Type 1 metabolism, Receptor, Melanocortin, Type 4 genetics, Receptor, Melanocortin, Type 4 metabolism, Anti-Inflammatory Agents administration & dosage, Apoptosis drug effects, Brain Injuries drug therapy, Melanocyte-Stimulating Hormones administration & dosage, Peptide Fragments administration & dosage
- Abstract
Following traumatic brain injury (TBI) neuroinflammatory processes promote neuronal cell loss. Alpha-melanocyte-stimulating hormone (α-MSH) is a neuropeptide with immunomodulatory properties, which may offer neuroprotection. Due to short half-life and pigmentary side-effects of α-MSH, the C-terminal tripeptide α-MSH(11-13) may be an anti-inflammatory alternative. The present study investigated the mRNA concentrations of the precursor hormone proopiomelanocortin (POMC) and of melanocortin receptors 1 and 4 (MC1R/MC4R) in naive mice and 15 min, 6, 12, 24, and 48 h after controlled cortical impact (CCI). Regulation of POMC and MC4R expression did not change after trauma, while MC1R levels increased over time with a 3-fold maximum at 12 h compared to naive brain tissue. The effect of α-MSH(11-13) on secondary lesion volume determined in cresyl violet stained sections (intraperitoneal injection 30 min after insult of 1 mg/kg α-MSH(11-13) or 0.9% NaCl) showed a considerable smaller trauma in α-MSH(11-13) injected mice. The expression of the inflammatory markers TNF-α and IL-1β as well as the total amount of Iba-1 positive cells were not reduced. However, cell branch counting of Iba-1 positive cells revealed a reduced activation of microglia. Furthermore, tripeptide injection reduced neuronal apoptosis analyzed by cleaved caspase-3 and NeuN staining. Based on the results single α-MSH(11-13) administration offers a promising neuroprotective property by modulation of inflammation and prevention of apoptosis after traumatic brain injury.
- Published
- 2013
- Full Text
- View/download PDF
36. PACAP signaling exerts opposing effects on neuroprotection and neuroinflammation during disease progression in the SOD1(G93A) mouse model of amyotrophic lateral sclerosis.
- Author
-
Ringer C, Büning LS, Schäfer MK, Eiden LE, Weihe E, and Schütz B
- Subjects
- Amyotrophic Lateral Sclerosis pathology, Animals, Disease Models, Animal, Disease Progression, Humans, Immunohistochemistry, In Situ Hybridization, Inflammation metabolism, Inflammation pathology, Mice, Mice, Inbred C57BL, Mice, Mutant Strains, Mice, Transgenic, Motor Neurons metabolism, Motor Neurons pathology, Nerve Degeneration pathology, Superoxide Dismutase genetics, Superoxide Dismutase-1, Amyotrophic Lateral Sclerosis metabolism, Nerve Degeneration metabolism, Pituitary Adenylate Cyclase-Activating Polypeptide metabolism, Signal Transduction physiology
- Abstract
Pituitary adenylate cyclase-activating polypeptide (PACAP) is a pleiotropic peptide with autocrine neuroprotective and paracrine anti-inflammatory properties in various models of acute neuronal damage and neurodegenerative diseases. Therefore, we examined a possible beneficial role of endogenous PACAP in the superoxide dismutase 1, SOD1(G93A), mouse model of amyotrophic lateral sclerosis (ALS), a lethal neurodegenerative disease particularly affecting somatomotor neurons. In wild-type mice, somatomotor and visceromotor neurons in brain stem and spinal cord were found to express the PACAP specific receptor PAC1, but only visceromotor neurons expressed PACAP as a potential autocrine source of regulation of these receptors. In SOD1(G93A) mice, only a small subset of the surviving somatomotor neurons showed induction of PACAP mRNA, and somatomotor neuron degeneration was unchanged in PACAP-deficient SOD1(G93A) mice. Pre-ganglionic sympathetic visceromotor neurons were found to be resistant in SOD1(G93A) mice, while pre-ganglionic parasympathetic neurons degenerated during ALS disease progression in this mouse model. PACAP-deficient SOD1(G93A) mice showed even greater pre-ganglionic parasympathetic neuron loss compared to SOD1(G93A) mice, and additional degeneration of pre-ganglionic sympathetic neurons. Thus, constitutive expression of PACAP and PAC1 may confer neuroprotection to central visceromotor neurons in SOD1(G93A) mice via autocrine pathways. Regarding the progression of neuroinflammation, the switch from amoeboid to hypertrophic microglial phenotype observed in SOD1(G93A) mice was absent in PACAP-deficient SOD1(G93A) mice. Thus, endogenous PACAP may promote microglial cytodestructive functions thought to drive ALS disease progression. This hypothesis was consistent with prolongation of life expectancy and preserved tongue motor function in PACAP-deficient SOD1(G93A) mice, compared to SOD1(G93A) mice. Given the protective role of PACAP expression in visceromotor neurons and the opposing effect on microglial function in SOD1(G93A) mice, both PACAP agonism and antagonism may be promising therapeutic tools for ALS treatment, if stage of disease progression and targeting the specific auto- and paracrine signaling pathways are carefully considered., (Copyright © 2013 Elsevier Inc. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
37. Species-specific vesicular monoamine transporter 2 (VMAT2) expression in mammalian pancreatic beta cells: implications for optimising radioligand-based human beta cell mass (BCM) imaging in animal models.
- Author
-
Schäfer MK, Hartwig NR, Kalmbach N, Klietz M, Anlauf M, Eiden LE, and Weihe E
- Subjects
- Aged, Animals, Female, Gene Expression Regulation, Humans, Insulin-Secreting Cells cytology, Insulin-Secreting Cells immunology, Ligands, Male, Mast Cells cytology, Mast Cells immunology, Mast Cells metabolism, Mice, Middle Aged, Nerve Endings metabolism, Pancreas cytology, Pancreas immunology, Pancreas innervation, Radioligand Assay, Rats, Species Specificity, Sus scrofa, Sympathetic Nervous System cytology, Sympathetic Nervous System metabolism, Tetrabenazine analogs & derivatives, Tetrabenazine metabolism, Vesicular Monoamine Transport Proteins genetics, Insulin-Secreting Cells metabolism, Pancreas metabolism, Vesicular Monoamine Transport Proteins metabolism
- Abstract
Aims/hypothesis: Imaging of beta cell mass (BCM) is a major challenge in diabetes research. The vesicular monoamine transporter 2 (VMAT2) is abundantly expressed in human beta cells. Radiolabelled analogues of tetrabenazine (TBZ; a low-molecular-weight, cell-permeant VMAT2-selective ligand) have been employed for pancreatic islet imaging in humans. Since reports on TBZ-based VMAT2 imaging in rodent pancreas have been fraught with confusion, we compared VMAT2 gene expression patterns in the mouse, rat, pig and human pancreas, to identify appropriate animal models with which to further validate and optimise TBZ imaging in humans., Methods: We used a panel of highly sensitive VMAT2 antibodies developed against equivalently antigenic regions of the transporter from each species in combination with immunostaining for insulin and species-specific in situ hybridisation probes. Individual pancreatic islets were obtained by laser-capture microdissection and subjected to analysis of mRNA expression of VMAT2., Results: The VMAT2 protein was not expressed in beta cells in the adult pancreas of common mouse or rat laboratory strains, in contrast to its expression in beta cells (but not other pancreatic endocrine cell types) in the pancreas of pigs and humans. VMAT2- and tyrosine hydroxylase co-positive (catecholaminergic) innervation was less abundant in humans than in rodents. VMAT2-positive mast cells were identified in the pancreas of all species., Conclusions/interpretation: Primates and pigs are suitable models for TBZ imaging of beta cells. Rodents, because of a complete lack of VMAT2 expression in the endocrine pancreas, are a 'null' model for assessing interference with BCM measurements by VMAT2-positive mast cells and sympathetic innervation in the pancreas.
- Published
- 2013
- Full Text
- View/download PDF
38. EGFL7 ligates αvβ3 integrin to enhance vessel formation.
- Author
-
Nikolic I, Stankovic ND, Bicker F, Meister J, Braun H, Awwad K, Baumgart J, Simon K, Thal SC, Patra C, Harter PN, Plate KH, Engel FB, Dimmeler S, Eble JA, Mittelbronn M, Schäfer MK, Jungblut B, Chavakis E, Fleming I, and Schmidt MHH
- Subjects
- Amino Acid Motifs genetics, Animals, Calcium-Binding Proteins, Cell Adhesion genetics, Cell Movement genetics, EGF Family of Proteins, Embryo, Nonmammalian blood supply, Embryo, Nonmammalian metabolism, Endothelial Growth Factors genetics, Endothelial Growth Factors pharmacology, Extracellular Matrix metabolism, Gene Expression, HEK293 Cells, Humans, Immunohistochemistry, Immunoprecipitation, Infarction, Middle Cerebral Artery genetics, Infarction, Middle Cerebral Artery metabolism, Integrin alphaVbeta3 genetics, Mice, Mice, Nude, Phosphorylation drug effects, Protein Binding, RNA Interference, Reverse Transcriptase Polymerase Chain Reaction, Zebrafish, Blood Vessels metabolism, Endothelial Growth Factors metabolism, Human Umbilical Vein Endothelial Cells metabolism, Integrin alphaVbeta3 metabolism
- Abstract
Angiogenesis, defined as blood vessel formation from a preexisting vasculature, is governed by multiple signal cascades including integrin receptors, in particular integrin αVβ3. Here we identify the endothelial cell (EC)-secreted factor epidermal growth factor-like protein 7 (EGFL7) as a novel specific ligand of integrin αVβ3, thus providing mechanistic insight into its proangiogenic actions in vitro and in vivo. Specifically, EGFL7 attaches to the extracellular matrix and by its interaction with integrin αVβ3 increases the motility of EC, which allows EC to move on a sticky underground during vessel remodeling. We provide evidence that the deregulation of EGFL7 in zebrafish embryos leads to a severe integrin-dependent malformation of the caudal venous plexus, pointing toward the significance of EGFL7 in vessel development. In biopsy specimens of patients with neurologic diseases, vascular EGFL7 expression rose with increasing EC proliferation. Further, EGFL7 became upregulated in vessels of the stroke penumbra using a mouse model of reversible middle cerebral artery occlusion. Our data suggest that EGFL7 expression depends on the remodeling state of the existing vasculature rather than on the phenotype of neurologic disease analyzed. In sum, our work sheds a novel light on the molecular mechanism EGFL7 engages to govern physiological and pathological angiogenesis.
- Published
- 2013
- Full Text
- View/download PDF
39. Role of L1CAM for axon sprouting and branching.
- Author
-
Schäfer MK and Frotscher M
- Subjects
- Animals, Ankyrins metabolism, Axons pathology, Cytoskeletal Proteins metabolism, Humans, Models, Biological, Neural Cell Adhesion Molecule L1 chemistry, Axons metabolism, Neural Cell Adhesion Molecule L1 metabolism
- Abstract
The central nervous system (CNS) has been traditionally considered as an organ that fails to regenerate in response to injury. Indeed, the lesioned CNS faces a number of obstacles during regeneration, including an overall non-permissive environment for axonal regeneration. However, research during the last few decades has identified axon sprouting as an anatomical correlate for the regenerative capability of the CNS to establish new connections. The immunoglobulin superfamily member L1CAM has been shown to promote the capability of neurons for regenerative axon sprouting and to improve behavioral outcomes after CNS injury. Here, we discuss the cell-autonomous role of L1CAM for axon sprouting in experimental rodent injury models and highlight the molecular interactions of L1CAM with ankyrins, ezrin-radixin-moesin proteins and the Sema3A/Neuropilin ligand-receptor complex in the context of axonal branching.
- Published
- 2012
- Full Text
- View/download PDF
40. Pathomechanistic characterization of two exonic L1CAM variants located in trans in an obligate carrier of X-linked hydrocephalus.
- Author
-
Marx M, Diestel S, Bozon M, Keglowich L, Drouot N, Bouché E, Frebourg T, Minz M, Saugier-Veber P, Castellani V, and Schäfer MK
- Subjects
- Adult, Cell Line, Cerebral Aqueduct abnormalities, Cerebral Aqueduct metabolism, Cerebral Aqueduct pathology, DNA Mutational Analysis, Female, Genetic Diseases, X-Linked metabolism, Genetic Diseases, X-Linked pathology, Humans, Hydrocephalus metabolism, Hydrocephalus pathology, Male, Middle Aged, Mutation, Neurons cytology, Neurons physiology, Pedigree, Exons, Genetic Diseases, X-Linked genetics, Genetic Variation, Hydrocephalus genetics, Neural Cell Adhesion Molecule L1 genetics
- Abstract
Mutations in the gene encoding the neural cell adhesion molecule L1CAM cause several neurological disorders collectively referred to as L1 syndrome. We report here a family case of X-linked hydrocephalus in which an obligate female carrier has two exonic L1CAM missense mutations in trans substituting amino acids in the first (p.W635C) or second (p.V768I) fibronectin-type III domains. We performed various biochemical and cell biological in vitro assays to evaluate the pathogenicity of these variants. Mutant L1-W635C protein accumulates in the endoplasmic reticulum (ER), is not transported into axons, and fails to promote L1CAM-mediated cell-cell adhesion as well as neurite growth. Immunoprecipitation experiments show that L1-W635C associates with the molecular ER chaperone calnexin and is modified by poly-ubiquitination. The mutant L1-V768I protein localizes at the cell surface, is not retained in the ER, and promotes neurite growth similar to wild-type L1CAM. However, the p.V768I mutation impairs L1CAM-mediated cell-cell adhesion albeit less severe than L1-W635C. These data indicate that p.W635C is a novel loss-of-function L1 syndrome mutation. The p.V768I mutation may represent a non-pathogenic variant or a variant associated with low penetrance. The poly-ubiquitination of L1-W635C and its association with the ER chaperone calnexin provide further insights into the molecular mechanisms underlying defective cell surface trafficking of L1CAM in L1 syndrome.
- Published
- 2012
- Full Text
- View/download PDF
41. Influence of age on brain edema formation, secondary brain damage and inflammatory response after brain trauma in mice.
- Author
-
Timaru-Kast R, Luh C, Gotthardt P, Huang C, Schäfer MK, Engelhard K, and Thal SC
- Subjects
- Aging, Animals, Brain Injuries metabolism, Brain Injuries pathology, Cell Movement, Cerebrum pathology, Cyclooxygenase 2 genetics, Gene Expression Regulation, Hematologic Tests, Inflammation complications, Interleukin-1beta genetics, Interleukin-6 genetics, Lung metabolism, Male, Mice, Mice, Inbred C57BL, Nitric Oxide Synthase Type II genetics, Organ Size, RNA, Messenger genetics, RNA, Messenger metabolism, T-Lymphocytes cytology, Tumor Necrosis Factor-alpha genetics, Water metabolism, Brain Injuries complications, Brain Injuries physiopathology, Edema complications
- Abstract
After traumatic brain injury (TBI) elderly patients suffer from higher mortality rate and worse functional outcome compared to young patients. However, experimental TBI research is primarily performed in young animals. Aim of the present study was to clarify whether age affects functional outcome, neuroinflammation and secondary brain damage after brain trauma in mice. Young (2 months) and old (21 months) male C57Bl6N mice were anesthetized and subjected to a controlled cortical impact injury (CCI) on the right parietal cortex. Animals of both ages were randomly assigned to 15 min, 24 h, and 72 h survival. At the end of the observation periods, contusion volume, brain water content, neurologic function, cerebral and systemic inflammation (CD3+ T cell migration, inflammatory cytokine expression in brain and lung, blood differential cell count) were determined. Old animals showed worse neurological function 72 h after CCI and a high mortality rate (19.2%) compared to young (0%). This did not correlate with histopathological damage, as contusion volumes were equal in both age groups. Although a more pronounced brain edema formation was detected in old mice 24 hours after TBI, lack of correlation between brain water content and neurological deficit indicated that brain edema formation is not solely responsible for age-dependent differences in neurological outcome. Brains of old naïve mice were about 8% smaller compared to young naïve brains, suggesting age-related brain atrophy with possible decline in plasticity. Onset of cerebral inflammation started earlier and primarily ipsilateral to damage in old mice, whereas in young mice inflammation was delayed and present in both hemispheres with a characteristic T cell migration pattern. Pulmonary interleukin 1β expression was up-regulated after cerebral injury only in young, not aged mice. The results therefore indicate that old animals are prone to functional deficits and strong ipsilateral cerebral inflammation without major differences in morphological brain damage compared to young.
- Published
- 2012
- Full Text
- View/download PDF
42. Functional inactivation of the genome-wide association study obesity gene neuronal growth regulator 1 in mice causes a body mass phenotype.
- Author
-
Lee AW, Hengstler H, Schwald K, Berriel-Diaz M, Loreth D, Kirsch M, Kretz O, Haas CA, de Angelis MH, Herzig S, Brümmendorf T, Klingenspor M, Rathjen FG, Rozman J, Nicholson G, Cox RD, and Schäfer MK
- Subjects
- Alleles, Animals, Body Height genetics, Cell Adhesion, Cell Line, Diet, High-Fat adverse effects, Eating genetics, Endoplasmic Reticulum metabolism, Energy Metabolism genetics, Female, Gene Knockout Techniques, Genotype, Humans, Hypothalamus cytology, Hypothalamus metabolism, Male, Membrane Proteins metabolism, Mice, Motor Activity genetics, Nerve Tissue Proteins metabolism, Neurites metabolism, Obesity metabolism, Obesity pathology, Obesity physiopathology, Phenotype, Body Weight genetics, Gene Silencing, Genome-Wide Association Study, Membrane Proteins deficiency, Membrane Proteins genetics, Nerve Tissue Proteins deficiency, Nerve Tissue Proteins genetics, Obesity genetics
- Abstract
To date, genome-wide association studies (GWAS) have identified at least 32 novel loci for obesity and body mass-related traits. However, the causal genetic variant and molecular mechanisms of specific susceptibility genes in relation to obesity are yet to be fully confirmed and characterised. Here, we examined whether the candidate gene NEGR1 encoding the neuronal growth regulator 1, also termed neurotractin or Kilon, accounts for the obesity association. To characterise the function of NEGR1 for body weight control in vivo, we generated two novel mutant mouse lines, including a constitutive NEGR1-deficient mouse line as well as an ENU-mutagenised line carrying a loss-of-function mutation (Negr1-I87N) and performed metabolic phenotypic analyses. Ablation of NEGR1 results in a small but steady reduction of body mass in both mutant lines, accompanied with a small reduction in body length in the Negr1-I87N mutants. Magnetic resonance scanning reveals that the reduction of body mass in Negr1-I87N mice is due to a reduced proportion of lean mass. Negr1-I87N mutants display reduced food intake and physical activity while normalised energy expenditure remains unchanged. Expression analyses confirmed the brain-specific distribution of NEGR1 including strong expression in the hypothalamus. In vitro assays show that NEGR1 promotes cell-cell adhesion and neurite growth of hypothalamic neurons. Our results indicate a role of NEGR1 in the control of body weight and food intake. This study provides evidence that supports the link of the GWAS candidate gene NEGR1 with body weight control.
- Published
- 2012
- Full Text
- View/download PDF
43. Gene expression of enzymes required for the de novo synthesis and degradation of pyrimidines in rat peripheral tissues and brain.
- Author
-
Gerlach J, Löffler M, and Schäfer MK
- Subjects
- Animals, Dihydrouracil Dehydrogenase (NADP) genetics, Dihydrouracil Dehydrogenase (NADP) metabolism, Immunohistochemistry, Kidney cytology, Kidney enzymology, Male, Rats, Rats, Wistar, Brain enzymology, Gene Expression Regulation, Enzymologic, Organ Specificity, Pyrimidines biosynthesis
- Abstract
Tissue-specific expression of the genes coding for the six enzymes of the de novo pyrimidine synthesis and for the first enzyme of the degradation pathway, dihydropyrimidine dehydrogenase (DPD), was analyzed in the rat using the in situ hybridization technique. Transcripts of the biosynthetic enzymes were detected in liver, kidney, and spleen with the highest expression in the white pulp. DPD was also transcribed in these organs with a striking layer-specific localization of DPD mRNA and protein in the kidney. All enzyme mRNAs were present in brain at low levels, but with region- and cell-specific differences. The relatively high expression in cortical regions including cerebellum and hippocampus points to a fundamental role of pyrimidine metabolism in brain function.
- Published
- 2011
- Full Text
- View/download PDF
44. Upregulation of microglial C1q expression has no effects on nigrostriatal dopaminergic injury in the MPTP mouse model of Parkinson disease.
- Author
-
Depboylu C, Schorlemmer K, Klietz M, Oertel WH, Weihe E, Höglinger GU, and Schäfer MK
- Subjects
- Animals, Corpus Striatum pathology, Disease Models, Animal, MPTP Poisoning pathology, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Microglia pathology, Parkinson Disease pathology, Substantia Nigra pathology, Complement C1q biosynthesis, Corpus Striatum metabolism, Dopamine physiology, MPTP Poisoning metabolism, Microglia metabolism, Parkinson Disease metabolism, Substantia Nigra metabolism, Up-Regulation physiology
- Abstract
Here we analyzed C1q, the initial recognition subcomponent of classical complement activation cascade, in an experimental model of Parkinson disease (PD). Nigrostriatal dopaminergic pathway injury was induced by treatment of wildtype mice subchronically with 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). Constitutive expression of C1q was restricted to microglia throughout the brain, and microglial C1q expression was early and transiently upregulated after MPTP in the substantia nigra (SN) and striatum, as analyzed by immunohistochemistry and in situ hybridization. C1q-positive microglia exhibited morphological characteristics of activated macrophage-type of cells, co-stained for MHCII, proliferated and were in close contact with degenerating dopaminergic neurons and fibers in the MPTP-lesioned SN. However, mice deficient in functional C1q protein were not significantly different in MPTP-induced loss of nigral dopaminergic neurons, striatal dopaminergic fibers and dopamine levels than their control littermates. In conclusion, C1q is upregulated and considered to be a marker of microglial activation in the nigrostriatal system after subchronic MPTP, but nigrostriatal dopaminergic injury may be not affected by C1q in this model., (Copyright © 2011 Elsevier B.V. All rights reserved.)
- Published
- 2011
- Full Text
- View/download PDF
45. Full-length L1CAM and not its Δ2Δ27 splice variant promotes metastasis through induction of gelatinase expression.
- Author
-
Hauser S, Bickel L, Weinspach D, Gerg M, Schäfer MK, Pfeifer M, Hazin J, Schelter F, Weidle UH, Ramser J, Volkmann J, Meindl A, Schmitt M, Schrötzlmair F, Altevogt P, and Krüger A
- Subjects
- Animals, Cell Line, Tumor, Colorectal Neoplasms genetics, Colorectal Neoplasms pathology, Disease Progression, Endosomes metabolism, Enzyme Induction, Female, Fibrosarcoma genetics, Fibrosarcoma pathology, Humans, Lymphoma, T-Cell genetics, Lymphoma, T-Cell pathology, Mice, Neoplasms genetics, Ovarian Neoplasms genetics, Ovarian Neoplasms pathology, Protein Transport, Alternative Splicing genetics, Matrix Metalloproteinase 2 biosynthesis, Matrix Metalloproteinase 9 biosynthesis, Neoplasm Metastasis genetics, Neoplasms enzymology, Neoplasms pathology, Neural Cell Adhesion Molecule L1 genetics
- Abstract
Tumour-specific splicing is known to contribute to cancer progression. In the case of the L1 cell adhesion molecule (L1CAM), which is expressed in many human tumours and often linked to bad prognosis, alternative splicing results in a full-length form (FL-L1CAM) and a splice variant lacking exons 2 and 27 (SV-L1CAM). It has not been elucidated so far whether SV-L1CAM, classically considered as tumour-associated, or whether FL-L1CAM is the metastasis-promoting isoform. Here, we show that both variants were expressed in human ovarian carcinoma and that exposure of tumour cells to pro-metastatic factors led to an exclusive increase of FL-L1CAM expression. Selective overexpression of one isoform in different tumour cells revealed that only FL-L1CAM promoted experimental lung and/or liver metastasis in mice. In addition, metastasis formation upon up-regulation of FL-L1CAM correlated with increased invasive potential and elevated Matrix metalloproteinase (MMP)-2 and -9 expression and activity in vitro as well as enhanced gelatinolytic activity in vivo. In conclusion, we identified FL-L1CAM as the metastasis-promoting isoform, thereby exemplifying that high expression of a so-called tumour-associated variant, here SV-L1CAM, is not per se equivalent to a decisive role of this isoform in tumour progression.
- Published
- 2011
- Full Text
- View/download PDF
46. Epileptiform activity interferes with proteolytic processing of Reelin required for dentate granule cell positioning.
- Author
-
Tinnes S, Schäfer MK, Flubacher A, Münzner G, Frotscher M, and Haas CA
- Subjects
- Animals, Cell Adhesion Molecules, Neuronal genetics, Endoplasmic Reticulum metabolism, Excitatory Amino Acid Agonists pharmacology, Extracellular Matrix metabolism, Extracellular Matrix pathology, Extracellular Matrix Proteins genetics, Gelatinases metabolism, Gene Expression physiology, Golgi Apparatus metabolism, Interneurons pathology, Interneurons physiology, Kainic Acid pharmacology, Nerve Tissue Proteins genetics, Organ Culture Techniques, Peptides, Potassium Chloride pharmacology, Rats, Reelin Protein, Serine Endopeptidases genetics, Cell Adhesion Molecules, Neuronal metabolism, Dentate Gyrus metabolism, Dentate Gyrus pathology, Dentate Gyrus physiopathology, Epilepsy metabolism, Epilepsy pathology, Epilepsy physiopathology, Extracellular Matrix Proteins metabolism, Nerve Tissue Proteins metabolism, Peptide Hydrolases metabolism, Serine Endopeptidases metabolism
- Abstract
The extracellular matrix protein Reelin is an essential regulator of neuronal migration and lamination in the developing and mature brain. Lack of Reelin causes severe disturbances in cerebral layering, such as the reeler phenotype and granule cell dispersion in temporal lobe epilepsy. Reelin is synthesized and secreted by Cajal-Retzius cells and GABAergic interneurons, and its function depends on proteolytic cleavage after secretion. The mechanisms regulating these processes are largely unknown. Here, we used rat hippocampal slice cultures to investigate the effect of neuronal activation and hyperexcitation on Reelin synthesis, secretion, and proteolytic processing. We show that enhanced neuronal activity does not modulate Reelin synthesis or secretion. Moreover, we found that intracellular Reelin resides predominantly in the endoplasmic reticulum before it is constitutively secreted via the early secretory pathway. Epileptiform activity, however, impairs the proteolytic processing of Reelin and leads to accumulation of Reelin in the extracellular matrix. We found that both conditions, epileptiform activity and impaired proteolytic cleavage of Reelin, cause granule cell dispersion via inhibition of metalloproteinases. Taken together, our results strongly suggest that secretion of Reelin is activity-independent and that proteolytic processing of Reelin is required for the maintenance of granule cell lamination in the dentate gyrus.
- Published
- 2011
- Full Text
- View/download PDF
47. Possible involvement of complement factor C1q in the clearance of extracellular neuromelanin from the substantia nigra in Parkinson disease.
- Author
-
Depboylu C, Schäfer MK, Arias-Carrión O, Oertel WH, Weihe E, and Höglinger GU
- Subjects
- Aged, Autopsy, Digoxigenin, Extracellular Space metabolism, Female, Fluorescent Antibody Technique, Humans, Image Processing, Computer-Assisted, Immunity, Innate physiology, Immunohistochemistry, In Situ Hybridization, Male, Phagocytosis, Tissue Fixation, Up-Regulation, Uridine Triphosphate, Complement C1q physiology, Melanins metabolism, Parkinson Disease metabolism, Substantia Nigra metabolism
- Abstract
Activation of the complement system promotes the removal of pathogens and tissue damage products from the brain and may also be involved in neuronal cell death in neurodegenerative diseases. Here, we analyzed the expression of C1q, the initial recognition subcomponent of the classic complement cascade, in the substantia nigra pars compacta (SNc) in Parkinson disease (PD) and control cases using immunohistochemistry and in situ hybridization. Microglia were determined to be the only cells that expressed C1q in the SNc and other brain areas. In the SNc of PD cases, there was increased deposition of extracellular neuromelanin in the parenchyma, resulting from degeneration of dopaminergic neurons. Neuromelanin granules and blebs of degenerated neurons seemed to be opsonized by C1q and phagocytosed by C1q-positive microglia and macrophages in the parenchyma and in the perivascular spaces. Neuromelanin-laden C1q-positive cells were also attached to the luminal surfaces of blood vessels in the SNc in PD. Thus, we present evidence suggesting that microglia are capable of phagocytosing and clearing cellular debris of degenerating neurons from the SNc through a C1q-mediated pathway in PD.
- Published
- 2011
- Full Text
- View/download PDF
48. L1CAM ubiquitination facilitates its lysosomal degradation.
- Author
-
Schäfer MK, Schmitz B, and Diestel S
- Subjects
- Animals, Cell Line, Tumor, Cell Movement, Growth Cones metabolism, Humans, Intracellular Space metabolism, Mice, Protein Transport, Lysosomes metabolism, Neural Cell Adhesion Molecule L1 metabolism, Ubiquitination
- Abstract
The cell adhesion molecule L1 is implicated in several processes in the developing and adult nervous system. Intracellular trafficking of L1 is important for cell migration, neurite growth and adhesion. We demonstrate here that L1 is ubiquitinated at the plasma membrane and in early endosomes. Mono-ubiquitination regulates L1 intracellular trafficking by enhancing its lysosomal degradation. We propose that L1's ubiquitination might be an additional mechanism to control its re-appearance at the cell surface thereby influencing processes like neurite growth and cell adhesion., (Copyright © 2010 Federation of European Biochemical Societies. Published by Elsevier B.V. All rights reserved.)
- Published
- 2010
- Full Text
- View/download PDF
49. L1 syndrome mutations impair neuronal L1 function at different levels by divergent mechanisms.
- Author
-
Schäfer MK, Nam YC, Moumen A, Keglowich L, Bouché E, Küffner M, Bock HH, Rathjen FG, Raoul C, and Frotscher M
- Subjects
- Animals, Cell Line, Cell Membrane metabolism, Cell Membrane pathology, Cell Membrane ultrastructure, Cell Polarity genetics, Endoplasmic Reticulum metabolism, Endoplasmic Reticulum pathology, Endoplasmic Reticulum ultrastructure, Humans, Neural Cell Adhesion Molecule L1 physiology, Neurogenesis genetics, Neurons ultrastructure, Organ Culture Techniques, Protein Transport genetics, Rats, Rats, Wistar, Syndrome, CA3 Region, Hippocampal metabolism, CA3 Region, Hippocampal pathology, Mutation genetics, Neural Cell Adhesion Molecule L1 genetics, Neurons metabolism, Neurons pathology
- Abstract
Mutations in the human L1CAM gene cause neurodevelopmental disorders collectively referred to as L1 syndrome. Here, we investigated cellular pathomechanisms underlying two L1 syndrome mutations, R184Q and W1036L. We demonstrate that these mutations cause partial endoplasmic reticulum (ER) retention of L1, reduce L1 cell surface expression, but do not induce ER stress in neuronal NSC-34 cells. We provide evidence that surface trafficking of mutated L1 is affected by defective sorting to ER exit sites and attenuated ER export. However, in differentiated neuronal cultures and long-term cultured hippocampal slices, the L1-R184Q protein is restricted to cell bodies, whereas L1-W1036L also aberrantly localizes to dendrites. These trafficking defects preclude axonal targeting of L1, thereby affecting L1-mediated axon growth and arborization. Our results indicate that L1 syndrome mutations impair neuronal L1 function at different levels, firstly by attenuating ER export and secondly by interfering with polarized neuronal trafficking., ((c) 2010 Elsevier Inc. All rights reserved.)
- Published
- 2010
- Full Text
- View/download PDF
50. L1CAM malfunction in the nervous system and human carcinomas.
- Author
-
Schäfer MK and Altevogt P
- Subjects
- Female, Fetal Alcohol Spectrum Disorders genetics, Fetal Alcohol Spectrum Disorders metabolism, Gene Expression Regulation, Genetic Diseases, X-Linked genetics, Hirschsprung Disease genetics, Hirschsprung Disease metabolism, Humans, Kidney abnormalities, Neoplasms genetics, Nervous System Diseases genetics, Neural Cell Adhesion Molecule L1 chemistry, Neural Cell Adhesion Molecule L1 genetics, Neurons metabolism, Paraplegia genetics, Paraplegia metabolism, Phenotype, Pregnancy, Protein Processing, Post-Translational, Syndrome, Transcription, Genetic, Genetic Diseases, X-Linked metabolism, Neoplasms metabolism, Nervous System metabolism, Nervous System Diseases metabolism, Neural Cell Adhesion Molecule L1 metabolism
- Abstract
Research over the last 25 years on the cell adhesion molecule L1 has revealed its pivotal role in nervous system function. Mutations of the human L1CAM gene have been shown to cause neurodevelopmental disorders such as X-linked hydrocephalus, spastic paraplegia and mental retardation. Impaired L1 function has been also implicated in the aetiology of fetal alcohol spectrum disorders, defective enteric nervous system development and malformations of the renal system. Importantly, aberrant expression of L1 has emerged as a critical factor in the development of human carcinomas, where it enhances cell proliferation, motility and chemoresistance. This discovery promoted collaborative work between tumour biologists and neurobiologists, which has led to a substantial expansion of the basic knowledge about L1 function and regulation. Here we provide an overview of the pathological conditions caused by L1 malfunction. We further discuss how the available data on gene regulation, molecular interactions and posttranslational processing of L1 may contribute to a better understanding of associated neurological and cancerous diseases.
- Published
- 2010
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.