Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Heidi Kaastrup Müller is active.

Publication


Featured researches published by Heidi Kaastrup Müller.


The International Journal of Neuropsychopharmacology | 2010

Inverse correlation of brain and blood BDNF levels in a genetic rat model of depression

Pia Høgh Plougmann; Heidi Kaastrup Müller; Aleksander A. Mathé; Raben Rosenberg; Gregers Wegener

There is accumulating evidence that brain-derived neurotrophic factor (BDNF) plays a critical role in the pathophysiology of depression. Decreased serum levels have been reported in major depression, and a correlation between BDNF reduction and the severity of the disease was found. Moreover, in post-mortem hippocampal tissue, increased levels of BDNF immunoreactivity have been reported in subjects treated with antidepressants compared to untreated subjects. These findings indicate parallel changes in brain and serum BDNF levels during depression. BDNF has been measured in selected brain areas in several animal models. In investigations between Flinders Sensitive Line (FSL) and Flinders Resistant Line (FRL) rats, a genetic rat model of depression, no differences were found in BDNF levels in the frontal cortex and hippocampus, areas believed to be core brain regions in depression. However, to our knowledge brain and serum BDNF levels have never been reported in parallel for any psychiatric disease model. Therefore, we examined the levels of BDNF in whole blood, serum, cerebrospinal fluid (CSF), hippocampus, and frontal cortex in male FSL and FRL rats. BDNF levels in serum and whole blood of FSL rats were significantly increased compared to FRL rats. In contrast, in the hippocampus the BDNF level was significantly decreased in FSL compared to FRL rats while no differences were found in the frontal cortex and CSF. The differential regulation of the BDNF levels in hippocampus, serum, and whole blood in FSL/FRL rats adds to the hypothesis that neurotrophic factors are related to the pathophysiology of depression.


The International Journal of Neuropsychopharmacology | 2010

Increased stress-evoked nitric oxide signalling in the Flinders sensitive line (FSL) rat: a genetic animal model of depression

Gregers Wegener; Brian H. Harvey; Birgit Egeskov Bonefeld; Heidi Kaastrup Müller; Vallo Volke; David H. Overstreet

Stress engenders the precipitation and progression of affective disorders, while stress-related release of excitatory mediators is implicated in the degenerative pathology observed especially in the hippocampus of patients with severe depression. Nitric oxide (NO) release following stress-evoked N-methyl-d-aspartate (NMDA) receptor activation modulates neurotransmission, cellular memory and neuronal toxicity. We have investigated the Flinders rat (FSL/FRL), a genetic animal model of depression, regarding the response of the hippocampal nitrergic system following exposure to an escapable stress/inescapable stress (ES-IS) paradigm. Hippocampal tissue from naive FSL/FRL rats and those exposed to ES-IS were studied with respect to constitutive nitric oxide synthase (cNOS) activity and neuronal nitric oxide synthase (nNOS) protein levels, as well as transcript expression of upstream regulatory proteins in the NMDA-NO signalling pathway, including NMDAR1, nNOS, CAPON, PIN and PSD95. Within stress-naive animals, no differences in hippocampal cNOS activity and nNOS expression or PIN were evident in FSL and FRL rats, although transcripts for NMDAR1 and CAPON were increased in FSL rats. Within the group of ES-IS animals, we found an increase in total hippocampal cNOS activity, nNOS protein levels and mRNA expression in FSL vs. FRL rats, together with an increase in PSD95 transcripts, and a reduction in PIN. In conclusion, ES-IS enhanced hippocampal cNOS activity in FSL rats, but not FRL rats, confirming the NMDA-NO cascade as an important vulnerability factor in the depressive phenotype of the FSL rat.


Journal of Biological Chemistry | 2006

Subcellular Redistribution of the Serotonin Transporter by Secretory Carrier Membrane Protein 2

Heidi Kaastrup Müller; Ove Wiborg; Jana Haase

The serotonin transporter (SERT) belongs to the SLC6 family of sodium- and chloride-dependent neurotransmitter transporters responsible for uptake of amino acids and biogenic amines from extracellular spaces. Their activities and subcellular distributions are regulated by various cellular mechanisms, including interactions with other proteins. Using the yeast two-hybrid approach we screened a human brain cDNA library and identified secretory carrier membrane protein 2 (SCAMP2) as a novel SERT-interacting protein. GST-pulldown assays confirmed the physical interaction between SCAMP2 and the N-terminal domain of SERT. In addition, SERT was found to form a complex with SCAMP2 as demonstrated by co-immunoprecipitation from a heterologous expression system and from rat brain homogenate. Co-expression of SERT and SCAMP2 in mammalian cells results in the subcellular redistribution of SERT with a decrease in cell surface SERT and a concomitant reduction in 5-HT uptake activity. Using confocal microscopy we show that in neuronal cells endogenous SERT co-localizes with SCAMP2 in discrete structures also containing the lipid raft marker flotillin-1 and the SNARE protein syntaxin 1A. In contrast, SERT immunoreactivity is clearly segregated from transferrin receptor-containing endosomes. A single amino acid mutation, cysteine-201 to alanine, within the conserved cytoplasmic E peptide of SCAMP2, abolished SCAMP2-mediated down-regulation of SERT, although this mutation had no effect on the physical interaction between SERT and SCAMP2. Taken together, our results suggest that SCAMP2 plays an important role in the regulation of the subcellular distribution of SERT.


Brain Research | 2011

Differential expression of synaptic proteins after chronic restraint stress in rat prefrontal cortex and hippocampus

Heidi Kaastrup Müller; Gregers Wegener; Maurizio Popoli

Prolonged stress has been associated with altered synaptic plasticity but little is known about the molecular components and mechanisms involved in the stress response. In this study, we examined the effect of chronic restraint stress (CRS) on the expression of genes associated with synaptic vesicle exocytosis in rat prefrontal cortex and hippocampus. Rats were stressed daily using a 21day restraint stress paradigm, with durations of half an hour or 6h. RNA and protein were extracted from the same tissue sample and used for real-time quantitative polymerase chain reaction (real-time qPCR) and immunoblotting, respectively. Focusing on the SNARE complex, we investigated the expression of the SNARE core components syntaxin 1A, SNAP-25, and VAMP2 at both transcriptional and protein levels. In addition, the expression of 10 SNARE regulatory proteins was investigated at the transcriptional level. Overall, the prefrontal cortex was more sensitive to CRS compared to the hippocampus. In prefrontal cortex, CRS induced increased mRNA levels of VAMP2, VAMP1, syntaxin 1A, snapin, synaptotagmins I and III, and synapsins I and II, whereas SNAP-25 was down-regulated after CRS. Immunoblotting demonstrated equivalent changes in protein levels of VAMP2, syntaxin 1A, and SNAP-25. In hippocampus, we found increased mRNA levels of VAMP2 and SNAP-29 and a decrease in VAMP1 levels. Immunoblotting revealed decreased VAMP2 protein levels despite increased mRNA levels. Changes in the expression of synaptic proteins may accompany or contribute to the morphological, functional, and behavioral changes observed in experimental models of stress and may have relevance to the pathophysiology of stress-related disorders.


Biochemical and Biophysical Research Communications | 2002

The fatty acid translocase (FAT)/CD36 and the glucose transporter GLUT4 are localized in different cellular compartments in rat cardiac muscle.

Heidi Kaastrup Müller; Karin Deckers; Jiirgen Eckel

The fatty acid translocase (FAT)/CD36 plays an important role in the acute regulation of fatty acid uptake in muscle tissue. We studied the subcellular distribution of FAT/CD36 in rat cardiac muscle after in vivo insulin stimulation by membrane fractionation and immunoisolation of GLUT4- and FAT/CD36-vesicles. FAT/CD36 was equally present in both plasma and microsomal membranes with no effect of insulin on the cellular distribution, whereas GLUT4 increased 2- to 3-fold in the plasma membrane. FAT/CD36 resides in one intracellular pool, whereas GLUT4 is present in two distinct pools. Immunoadsorption of GLUT4-vesicles indicated that FAT/CD36 is undetectable in these vesicles. Likewise, no GLUT4 could be detected in FAT/CD36-vesicles. These vesicles contain a high amount of Rab11 that remained unaffected after insulin stimulation, whereas Rab11 increased about 3-fold in the GLUT4-vesicles in response to insulin. These data show that GLUT4 and FAT/CD36 do not co-localize in cardiac muscle and that FAT/CD36 is not redistributed in response to insulin in the heart. Rab11 may be involved in endosomal recycling of FAT/CD36, however, insulin-associated Rab11 functions appear to be limited to GLUT4-vesicles.


Journal of Psychiatric Research | 2013

Ketamine regulates the presynaptic release machinery in the hippocampus.

Heidi Kaastrup Müller; Gregers Wegener; Nico Liebenberg; Carlos A. Zarate; Maurizio Popoli

In the search for new drug targets, that may help point the way to develop fast-acting treatments for mood disorders, we have explored molecular pathways regulated by ketamine, an NMDA receptor antagonist, which has consistently shown antidepressant response within a few hours of administration. Using Sprague-Dawley rats we investigated the effects of ketamine on the presynaptic release machinery responsible for neurotransmitter release at 1, 2 and 4 h as well as 7 days after administration of a single subanesthetic dose of ketamine (15 mg/kg). A large reduction in the accumulation of SNARE complexes was observed in hippocampal synaptic membranes after 1, 2 and 4 h of ketamine administration. In parallel, we found a selective reduction in the expression of the synaptic vesicle protein synaptotagmin I and an increase in the levels of synapsin I in hippocampal synaptosomes suggesting a mechanism by which ketamine reduces SNARE complex formation, in part, by regulating the number of synaptic vesicles in the nerve terminals. Moreover, ketamine reduced Thr(286)-phosphorylated αCaMKII and its interaction with syntaxin 1A, which identifies CaMKII as a potential target for second messenger-mediated actions of ketamine. In addition, despite previous reports of ketamine-induced inhibition of GSK-3, we were unable to detect regulation of its activity after ketamine administration. Our findings demonstrate that ketamine rapidly induces changes in the hippocampal presynaptic machinery similar to those that are obtained only with chronic treatments with traditional antidepressants. This suggests that reduction of neurotransmitter release in the hippocampus has possible relevance for the rapid antidepressant effect of ketamine.


Journal of Molecular Neuroscience | 2009

Membrane Glycoprotein M6B Interacts with the Human Serotonin Transporter

Anja W. Fjorback; Heidi Kaastrup Müller; Ove Wiborg

The serotonin transporter (SERT) belongs to a family of sodium- and chloride-dependent neurotransmitter transporters that are responsible for the active re-uptake of the neurotransmitter serotonin from the synapse. In the present study, using the yeast two-hybrid system, we identified the membrane glycoprotein M6B as a binding partner of SERT. This interaction was further verified by co-immunoprecipitation and glutathione-S-transferase pull-down assays. M6B belongs to a proteolipid protein family, which is expressed in neurons and in oligodendrocytes in the brain. The knowledge of the biological function of this protein family is sparse, but their expression in most brain regions have led to the hypothesis that they are involved in cellular housekeeping functions such as membrane trafficking and cell-to-cell communication. The co-expression of SERT with M6B results in a significant decrease in SERT-mediated serotonin uptake caused by a down-regulation of SERT surface expression. Furthermore, we find, using confocal microscopy, that M6B co-localizes with SERT when transiently expressed in HEK-MSR-293 cells and when endogenously expressed in RN46A cells. Taken together, our data suggest that M6B regulates the serotonin uptake by affecting cellular trafficking of the serotonin transporter.


Stress | 2012

Wistar rats subjected to chronic restraint stress display increased hippocampal spine density paralleled by increased expression levels of synaptic scaffolding proteins

Dariusz Orlowski; Heidi Kaastrup Müller; Gregers Wegener; Carsten R. Bjarkam

The aim of this study was to investigate whether the previously reported effect of chronic restraint stress (CRS) on hippocampal neuron morphology and spine density is paralleled by a similar change in the expression levels of synaptic scaffolding proteins. Adult male Wistar rats were subjected either to CRS (6 h/day) for 21 days or to control conditions. The resulting brains were divided and one hemisphere was impregnated with Golgi-Cox before coronal sectioning and autometallographic development. Neurons from CA1, CA3b, CA3c, and dentate gyrus (DG) area were reconstructed and subjected to Sholl analysis and spine density estimation. The contralateral hippocampus was used for quantitative real-time polymerase chain reaction and protein analysis of genes associated with spine density and morphology (the synaptic scaffolding proteins: Spinophilin, Homer1-3, and Shank1-3). In the CA3c area, CRS decreased the number of apical dendrites and their total length, whereas CA1 and DG spine density were significantly increased. Analysis of the contralateral hippocampal homogenate displayed an increased gene expression of Spinophilin, Homer1, Shank1, and Shank2 and increased protein expression of Spinophilin and Homer1 in the CRS animals. In conclusion, CRS influences hippocampal neuroplasticity by modulation of dendrite branching pattern and spine density paralleled by increased expression levels of synaptic scaffolding proteins.


Journal of Neurochemistry | 2008

Characterisation of the zebrafish serotonin transporter functionally links TM10 to the ligand binding site

Kasper Severinsen; Steffen Sinning; Heidi Kaastrup Müller; Ove Wiborg

The selective serotonin reuptake inhibitors and tricyclic antidepressants act by inhibiting pre‐synaptic reuptake of serotonin (5‐HT) leading to elevated synaptic 5‐HT concentrations. However, despite extensive efforts little is known about the protein‐ligand interactions of serotonin transporter (SERT) and inhibitors. To identify domains and individual amino acids important for ligand binding, we cloned the serotonin transporter from zebrafish, Danio rerio, (drSERT) and compared its pharmacological profile to that of the human serotonin transporter (hSERT) with respect to inhibition of [3H]5‐HT uptake and [3H]‐escitalopram binding in transiently transfected human embryonic kidney cells; HEK293‐MSR. Residues responsible for altered affinities inhibitors were pinpointed by generating cross‐species chimeras and subsequent point mutations by site directed mutagenesis. drSERT has a higher affinity towards compounds of the imipramine class, desipramine in particular, exhibiting a 35‐fold increased affinity compared to hSERT. drSERT has a 15–30‐fold lower affinity towards cocaine and cocaine analogues. The differences in ligand recognition are shown to be primarily caused by interspecies differences in TM10 and were tracked down to three residues (Ala505, Leu506 and Ile507).


Biochemical and Biophysical Research Communications | 2009

Serotonin transporter oligomerization documented in RN46A cells and neurons by sensitized acceptor emission FRET and fluorescence lifetime imaging microscopy

Anja W. Fjorback; Patrick Pla; Heidi Kaastrup Müller; Ove Wiborg; Frédéric Saudou; Jens R. Nyengaard

The serotonin transporter is a member of the monoamine transporter family that also includes transporters of dopamine and norepinephrine. We have used sensitized acceptor emission fluorescence resonance energy transfer (FRET) and fluorescence lifetime imaging microscopy (FLIM) to study the oligomerization of SERT in HEK-MSR-239 cells, RN46A cells and in cultured hippocampal neurons. We were able to show identical FRET efficiencies in cell lines as well as in primary cultured hippocampal neurons, demonstrating that the oligomerization is cell type independent. The results obtained with both FRET approaches are very similar and furthermore, in agreement with previous results obtained by donor bleaching FRET microscopy.

Collaboration


Dive into the Heidi Kaastrup Müller's collaboration.

Top Co-Authors

Avatar

Gregers Wegener

Aarhus University Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jana Haase

University College Dublin

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge