Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 62
1.
Article En | MEDLINE | ID: mdl-38636702

BACKGROUND: Reserpine (RES), a Vesicular Monoamine Transporter 2 (VMAT2) inhibitor agent, has been used in preclinical research for many years to create animal models for depression and to test experimental antidepressant strategies. Nevertheless, evidence of the potential use and validity of RES as a chronic pharmacological model for depression is lacking, and there are no comprehensive studies of the behavioral effects in conjunction with molecular outcomes. METHODS: Experiment 1. Following baseline behavior testing sensitive to depression-like phenotype and locomotion (Phase 1), 27 Sprague-Dawley (SD) rats received i.p. either vehicle solution (0.0 mg/kg), low (0.2 mg/kg) or high (0.8 mg/kg) RES dose for 20 days using a pre-determined schedule and reassessed for behavioral phenotypes (Phase 2). After 10 days washout period, and a final behavioral assessment (Phase 3), the brains were collected 16 days after the last injection for mRNA-expression assessment. Experiment 2. In a similar timetable as in Experiment 1 but without the behavioral testing, 12 SD rats underwent repetitive dopamine D2/3 receptor PET scanning with [18F]DMFP following each Phase. The binding potential (BPND) of [18F]DMFP was quantified by kinetic analysis as a marker of striatal D2/3R availability. Weight and welfare were monitored throughout the study. RESULTS: Significant, dose-dependent weight loss and behavioral deficits including both motor (hypo-locomotion) and non-motor behavior (anhedonia, mild anxiety and reduced exploration) were found for both the low and high dose groups with significant decrease in D2R mRNA expression in the accumbal region for the low RES group after Phase 3. Both RES treated groups showed substantial increase in [18F]DMFP BPND (in line with dopamine depletion) during Phase 2 and 3 compared to baseline and Controls. CONCLUSIONS: The longitudinal design of the study demonstrated that chronic RES administration induced striatal dopamine depletion that persisted even after the wash-out period. However, the behavior phenotype observed were transient. The data suggest that RES administration can induce a rodent model for depression with mild face validity.


Depression , Disease Models, Animal , Positron-Emission Tomography , Rats, Sprague-Dawley , Reserpine , Animals , Reserpine/pharmacology , Male , Rats , Depression/chemically induced , Depression/metabolism , Behavior, Animal/drug effects , Receptors, Dopamine/metabolism , Dose-Response Relationship, Drug , Brain/metabolism , Brain/drug effects , Brain/diagnostic imaging , Vesicular Monoamine Transport Proteins/metabolism , Motor Activity/drug effects
2.
Neurol Int ; 15(3): 896-907, 2023 Jul 24.
Article En | MEDLINE | ID: mdl-37489363

High-resolution imaging in small animal models of neurologic disease is a technical challenge. In a pilot project, we have explored a non-destructive synchrotron imaging technique for the 3D visualization of intracerebral tissue transplants in a well-established small animal model of Huntington's disease. Four adult female Sprague Dawley rats each received injections of 0.12 M quinolinic acid (QA) into two target positions in the left striatum, thus creating unilateral left-sided striatal lesions similar to those frequently seen in patients suffering from Huntington's disease. One week after lesioning, the animals received transplants prepared from whole ganglionic eminences (wGEs) obtained from 13- to 14-day-old rat embryos. Of the four lesioned animals, three received transplants of GNP-loaded cells and one animal received a transplant of naïve cells, serving as control. Post-mortem synchrotron-based microCT was used to obtain images of the neurotransplants. The images obtained of GNP-loaded tissue transplants at the synchrotron corresponded in size and shape to the histological images of transplants developed from naïve cells. Thus, we conclude that non-destructive synchrotron imaging techniques such as phase-contrast imaging are suitable to obtain high-resolution images of GNP-loaded tissue transplants.

3.
Brain Stimul ; 16(2): 670-681, 2023.
Article En | MEDLINE | ID: mdl-37028755

BACKGROUND: Understanding prefrontal cortex projections to diencephalic-mesencephalic junction (DMJ), especially to subthalamic nucleus (STN) and ventral mesencephalic tegmentum (VMT) helps our comprehension of Deep Brain Stimulation (DBS) in major depression (MD) and obsessive-compulsive disorder (OCD). Fiber routes are complex and tract tracing studies in non-human primate species (NHP) have yielded conflicting results. The superolateral medial forebrain bundle (slMFB) is a promising target for DBS in MD and OCD. It has become a focus of criticism owing to its name and its diffusion weighted-imaging based primary description. OBJECTIVE: To investigate DMJ connectivity in NHP with a special focus on slMFB and the limbic hyperdirect pathway utilizing three-dimensional and data driven techniques. METHODS: We performed left prefrontal adeno-associated virus - tracer based injections in the common marmoset monkey (n = 52). Histology and two-photon microscopy were integrated into a common space. Manual and data driven cluster analyses of DMJ, subthalamic nucleus and VMT together, followed by anterior tract tracing streamline (ATTS) tractography were deployed. RESULTS: Typical pre- and supplementary motor hyperdirect connectivity was confirmed. The advanced tract tracing unraveled the complex connectivity to the DMJ. Limbic prefrontal territories directly projected to the VMT but not STN. DISCUSSION: Intricate results of tract tracing studies warrant the application of advanced three-dimensional analyses to understand complex fiber-anatomical routes. The applied three-dimensional techniques can enhance anatomical understanding also in other regions with complex fiber anatomy. CONCLUSION: Our work confirms slMFB anatomy and enfeebles previous misconceptions. The rigorous NHP approach strengthens the role of the slMFB as a target structure for DBS predominantly in psychiatric indications like MD and OCD.


Deep Brain Stimulation , Subthalamic Nucleus , Animals , Callithrix , Deep Brain Stimulation/methods , Medial Forebrain Bundle , Mesencephalon
4.
Brain Sci ; 12(8)2022 Aug 19.
Article En | MEDLINE | ID: mdl-36009169

New technologies, such as fiber photometry, can overcome long-standing methodological limitations and promote a better understanding of neuronal mechanisms. This study, for the first time, aimed at employing the newly available dopamine indicator (GRABDA2m) in combination with this novel imaging technique. Here, we present a detailed methodological roadmap leading to longitudinal repetitive transmitter release monitoring in in vivo freely moving animals and provide proof-of-concept data. This novel approach enables a fresh look at dopamine release patterns in the nucleus accumbens, following the medial forebrain bundle (mfb) DBS in a rodent model. Our results suggest reliable readouts of dopamine levels over at least 14 days of DBS-induced photometric measurements. We show that mfb-DBS can elicit an increased dopamine response during stimulation (5 s and 20 s DBS) compared to its baseline dopamine activity state, reaching its maximum peak amplitude in about 1 s and then recovering back after stimulation. The effect of different DBS pulse widths (PWs) also suggests a potential differential effect on this neurotransmitter response, but future studies would need to verify this. Using the described approach, we aim to gain insights into the differences between pathological and healthy models and to elucidate more exhaustively the mechanisms under which DBS exerts its therapeutic action.

5.
Neuroscience ; 498: 31-49, 2022 08 21.
Article En | MEDLINE | ID: mdl-35750113

Major Depressive Disorder (MDD) is an affective disorder typically accompanied by sleep disturbances. Deep brain stimulation (DBS) of the medial forebrain bundle (MFB) is an emerging intervention for treatment-resistant depression, but its effect on sleep has not been closely examined. Here we aimed to characterise sleep deficits in the Flinders sensitive line, an established rodent model of depression, and investigate the consequences of MFB stimulation on sleep-related phenotypes. Rats were implanted with bilateral stimulation electrodes in the MFB, surface electrodes to record electrocorticography and electromyography for sleep scoring and electrodes within the prelimbic cortex, nucleus accumbens (NAc) and dorsal hippocampus. Recordings of sleep and oscillatory activity were conducted prior to and following twenty-four hours of MFB stimulation. Behavioural anti-depressant effects were monitored using the forced swim test. Previously unreported abnormalities in the Flinders sensitive line rats were observed during slow wave sleep, including decreased circadian amplitude of its rhythm, a reduction in slow wave activity and elevated gamma band oscillations. Previously established rapid eye movement sleep deficits were replicated. MFB stimulation had anti-depressant effects on behavioural phenotype, but did not significantly impact sleep architecture; it suppressed elevated gamma activity during slow wave sleep in the electrocorticogram and prelimbic cortex signals. Diverse abnormalities in Flinders sensitive line rats emphasise slow wave sleep as a state of dysfunction in affective disorders. MFB stimulation is able to affect behaviour and sleep physiology without influencing sleep architecture. Gamma modulation may represent a component of antidepressant mechanism.


Deep Brain Stimulation , Depressive Disorder, Major , Sleep, Slow-Wave , Animals , Depression , Medial Forebrain Bundle , Nucleus Accumbens , Rats , Rodentia
6.
Brain Sci ; 12(4)2022 Mar 25.
Article En | MEDLINE | ID: mdl-35447971

More than a decade ago, deep brain stimulation (DBS) of the superolateral medial forebrain bundle (slMFB), as part of the greater MFB system, had been proposed as a putative yet experimental treatment strategy for therapy refractory depression (TRD) and later for obsessive-compulsive disorders (OCD). Antidepressant and anti-OCD efficacy have been shown in open case series and smaller trials and were independently replicated. The MFB is anato-physiologically confluent with the SEEKING system promoting euphoric drive, reward anticipation and reward; functions realized through the mesocorticolimbic dopaminergic system. Growing clinical experience concerning surgical and stimulation aspects from a larger number of patients shows an MFB functionality beyond SEEKING and now re-informs the scientific rationale concerning the MFB's (patho-) physiology. In this white paper, we combine observations from more than 75 cases of slMFB DBS. We integrate these observations with a selected literature review to provide a new neuroethological view on the MFB. We here formulate a re-interpretation of the MFB as the main structure of an integrated SEEKING/MAINTENANCE circuitry, allowing for individual homeostasis and well-being through emotional arousal, basic and higher affect valence, bodily reactions, motor programing, vigor and flexible behavior, as the basis for the antidepressant and anti-OCD efficacy.

7.
J Neurosci Res ; 100(3): 897-911, 2022 03.
Article En | MEDLINE | ID: mdl-35088434

Major depressive disorder is one of the most common mental disorders, and more than 300 million of people suffer from depression worldwide. Recent clinical trials indicate that deep brain stimulation of the superolateral medial forebrain bundle (mfb) can have rapid and long-term antidepressant effects in patients with treatment-resistant depression. However, the mechanisms of action are elusive. In this study, using female rats, we demonstrate the antidepressant effects of selective optogenetic stimulation of the ventral tegmental area's dopaminergic (DA) neurons passing through the mfb and compare different stimulation patterns. Chronic mild unpredictable stress (CMUS) induced depressive-like, but not anxiety-like phenotype. Short-term and long-term stimulation demonstrated antidepressant effect (OSST) and improved anxiolytic effect (EPM), while long-term stimulation during CMUS induction prevented depressive-like behavior (OSST and USV) and improved anxiolytic effect (EPM). The results highlight that long-term accumulative stimulation on DA pathways is required for antidepressant and anxiolytic effect.


Deep Brain Stimulation , Depressive Disorder, Major , Animals , Deep Brain Stimulation/methods , Depression/therapy , Depressive Disorder, Major/metabolism , Dopamine/metabolism , Dopaminergic Neurons/metabolism , Female , Humans , Optogenetics , Rats , Rodentia/metabolism , Ventral Tegmental Area/physiology
8.
Brain Struct Funct ; 227(1): 23-47, 2022 Jan.
Article En | MEDLINE | ID: mdl-34482443

Uncertainties concerning anatomy and function of cortico-subcortical projections have arisen during the recent years. A clear distinction between cortico-subthalamic (hyperdirect) and cortico-tegmental projections (superolateral medial forebrain bundle, slMFB) so far is elusive. Deep Brain Stimulation (DBS) of the slMFB (for major depression, MD and obsessive compulsive disorders, OCD) has on the one hand been interpreted as actually involving limbic (prefrontal) hyperdirect pathways. On the other hand slMFB's stimulation region in the mesencephalic ventral tegmentum is said to impact on other structures too, going beyond the antidepressant (or anti OCD) efficacy of sole modulation of the cortico-tegmental reward-associated pathways. We have here used a normative diffusion MRT template (HCP, n = 80) for long-range tractography and augmented this dataset with ex-vivo high resolution data (n = 1) in a stochastic brain space. We compared this data with histological information and used the high resolution ex-vivo data set to scrutinize the mesencephalic tegmentum for small fiber pathways present. Our work resolves an existing ambiguity between slMFB and prefrontal hyperdirect pathways which-for the first time-are described as co-existent. DBS of the slMFB does not appear to modulate prefrontal hyperdirect cortico-subthalamic but rather cortico-tegmental projections. Smaller fiber structures in the target region-as far as they can be discerned-appear not to be involved in slMFB DBS. Our work enfeebles previous anatomical criticism and strengthens the position of the slMFB DBS target for its use in MD and OCD.


Prefrontal Cortex , Subthalamic Nucleus , Deep Brain Stimulation , Medial Forebrain Bundle , Tegmentum Mesencephali
9.
Eur J Neurosci ; 53(1): 89-113, 2021 01.
Article En | MEDLINE | ID: mdl-32931064

Deep brain stimulation (DBS) in psychiatric illnesses has been clinically tested over the past 20 years. The clinical application of DBS to the superolateral branch of the medial forebrain bundle in treatment-resistant depressed patients-one of several targets under investigation-has shown to be promising in a number of uncontrolled open label trials. However, there are remain numerous questions that need to be investigated to understand and optimize the clinical use of DBS in depression, including, for example, the relationship between the symptoms, the biological substrates/projections and the stimulation itself. In the context of precision and customized medicine, the current paper focuses on clinical and experimental research of medial forebrain bundle DBS in depression or in animal models of depression, demonstrating how clinical and scientific progress can work in tandem to test the therapeutic value and investigate the mechanisms of this experimental treatment. As one of the hypotheses is that depression engenders changes in the reward and motivational networks, the review looks at how stimulation of the medial forebrain bundle impacts the dopaminergic system.


Deep Brain Stimulation , Mental Disorders , Animals , Humans , Medial Forebrain Bundle , Mental Disorders/therapy , Motivation , Reward
10.
Exp Neurol ; 327: 113224, 2020 05.
Article En | MEDLINE | ID: mdl-32035070

BACKGROUND: Medial forebrain bundle (MFB) deep brain stimulation (DBS) has anti-depressant effects clinically and in depression models. Currently, therapeutic mechanisms of MFB DBS or how stimulation parameters acutely impact neurotransmitter release, particularly dopamine, are unknown. Experimentally, MFB DBS has been shown to evoke dopamine response in healthy controls, but not yet in a rodent model of depression. OBJECTIVE: The study investigated the impact of clinically used stimulation parameters on the dopamine induced response in a validated rodent depression model and in healthy controls. METHOD: The stimulation-induced dopamine response in Flinders Sensitive Line (FSL, n = 6) rat model of depression was compared with Sprague Dawley (SD, n = 6) rats following MFB DSB, using Fast Scan Cyclic Voltammetry to assess the induced response in the nucleus accumbens. Stimulation parameters were 130 Hz ("clinically" relevant) with pulse widths between 100 and 350 µs. RESULTS: Linear mixed model analysis showed significant impact in both models following MFB DBS both at 130 and 60 Hz with 100 µs pulse width in inducing dopamine response. Furthermore, at 130 Hz the evoked dopamine responses were different across the groups at the different pulse widths. CONCLUSION: The differential impact of MFB DBS on the induced dopamine response, including different response patterns at given pulse widths, is suggestive of physiological and anatomical divergence in the MFB in the pathological and healthy state. Studying how varying stimulation parameters affect the physiological outcome will promote a better understanding of the biological substrate of the disease and the possible anti-depressant mechanisms at play in clinical MFB DBS.


Deep Brain Stimulation/methods , Depression/metabolism , Dopamine/metabolism , Medial Forebrain Bundle/metabolism , Nucleus Accumbens/metabolism , Animals , Depression/physiopathology , Disease Models, Animal , Female , Medial Forebrain Bundle/physiopathology , Nucleus Accumbens/physiopathology , Rats , Rats, Sprague-Dawley
11.
Stereotact Funct Neurosurg ; 98(1): 8-20, 2020.
Article En | MEDLINE | ID: mdl-31982883

BACKGROUND: Deep brain stimulation (DBS) of the medial forebrain bundle (MFB) can reverse depressive-like symptoms clinically and in experimental models of depression, but the mechanisms of action are unknown. OBJECTIVES: This study investigated the role of dopaminergic mechanisms in MFB stimulation-mediated behavior changes, in conjunction with raclopride administration and micropositron emission tomography (micro-PET). METHODS: Flinders Sensitive Line (FSL) rats were allocated into 4 groups: FSL (no treatment), FSL+ (DBS), FSL.R (FSL with raclopride), and FSL.R+ (FSL with raclopride and DBS). Animals were implanted with bilateral electrodes targeting the MFB and given 11 days access to raclopride in the drinking water with or without concurrent continuous bilateral DBS over the last 10 days. Behavioral testing was conducted after stimulation. A PET scan using [18F]desmethoxyfallypride was performed to determine D2 receptor availability before and after raclopride treatment. Changes in gene expression in the nucleus accumbens and the hippocampus were assessed using quantitative polymerase chain reaction. RESULTS: Micro-PET imaging showed that raclopride administration blocked 36% of the D2 receptor in the striatum, but the relative level of blockade was reduced/modulated by stimulation. Raclopride treatment enhanced depressive-like symptoms in several tasks, and the MFB DBS partially reversed the depressive-like phenotype. The raclopride-treated MFB DBS animals had increased levels of mRNA coding for dopamine receptor D1 and D2 suggestive of a stimulation-mediated increase in dopamine receptors. CONCLUSION: Data suggest that chronic and continuous MFB DBS could act via the modulation of the midbrain dopaminergic transmission, including impacting on the postsynaptic dopamine receptor profile.


Deep Brain Stimulation/methods , Depression/metabolism , Dopamine/metabolism , Medial Forebrain Bundle/metabolism , Positron-Emission Tomography/methods , Raclopride/metabolism , Animals , Depression/diagnostic imaging , Depression/therapy , Dopamine Antagonists/metabolism , Dopamine Antagonists/pharmacology , Dopamine Antagonists/therapeutic use , Male , Medial Forebrain Bundle/diagnostic imaging , Medial Forebrain Bundle/drug effects , Raclopride/pharmacology , Raclopride/therapeutic use , Rats , Rodentia/metabolism , X-Ray Microtomography/methods
12.
Neuroimage Clin ; 25: 102165, 2020.
Article En | MEDLINE | ID: mdl-31954987

BACKGROUND: Major depression (MD) and obsessive-compulsive disorder (OCD) are psychiatric diseases with a huge impact on individual well-being. Despite optimal treatment regiments a subgroup of patients remains treatment resistant and stereotactic surgery (stereotactic lesion surgery, SLS or Deep Brain Stimulation, DBS) might be an option. Recent research has described four networks related to MD and OCD (affect, reward, cognitive control, default network) but only on a cortical and the adjacent sub-cortical level. Despite the enormous impact of comparative neuroanatomy, animal science and stereotactic approaches a holistic theory of subcortical and cortical network interactions is elusive. Because of the dominant hierarchical rank of the neocortex, corticofugal approaches have been used to identify connections in subcortical anatomy without anatomical priors and in part confusing results. We here propose a different corticopetal approach by identifying subcortical networks and search for neocortical convergences thereby following the principle of phylogenetic and ontogenetic network development. MATERIAL AND METHODS: This work used a diffusion tensor imaging data from a normative cohort (Human Connectome Project, HCP; n = 200) to describe eight subcortical fiber projection pathways (PPs) from subthalamic nucleus (STN), substantia nigra (SNR), red nucleus (RN), ventral tegmental area (VTA), ventrolateral thalamus (VLT) and mediodorsal thalamus (MDT) in a normative space (MNI). Subcortical and cortical convergences were described including an assignment of the specific pathways to MD/OCD-related networks. Volumes of activated tissue for different stereotactic stimulation sites and procedures were simulated to understand the role of the distinct networks, with respect to symptoms and treatment of OCD and MD. RESULTS: The detailed course of eight subcortical PPs (stnPP, snrPP, rnPP, vlATR, vlATRc, mdATR, mdATRc, vtaPP/slMFB) were described together with their subcortical and cortical convergences. The anterior limb of the internal capsule can be subdivided with respect to network occurrences in ventral-dorsal and medio-lateral gradients. Simulation of stereotactic procedures for OCD and MD showed dominant involvement of mdATR/mdATRc (affect network) and vtaPP/slMFB (reward network). DISCUSSION: Corticofugal search strategies for the evaluation of stereotactic approaches without anatomical priors often lead to confusing results which do not allow for a clear assignment of a procedure to an involved network. According to our simulation of stereotactic procedures in the treatment of OCD and MD, most of the target regions directly involve the reward (and affect) networks, while side-effects can in part be explained with a co-modulation of the control network. CONCLUSION: The here proposed corticopetal approach of a hierarchical description of 8 subcortical PPs with subcortical and cortical convergences represents a new systematics of networks found in all different evolutionary and distinct parts of the human brain.


Depressive Disorder, Major/pathology , Diffusion Tensor Imaging/methods , Internal Capsule/pathology , Mesencephalon/pathology , Neocortex/pathology , Nerve Net/pathology , Obsessive-Compulsive Disorder/pathology , Adult , Cohort Studies , Connectome , Depressive Disorder, Major/diagnostic imaging , Humans , Internal Capsule/diagnostic imaging , Mesencephalon/diagnostic imaging , Neocortex/diagnostic imaging , Nerve Net/diagnostic imaging , Neural Pathways/diagnostic imaging , Neural Pathways/pathology , Obsessive-Compulsive Disorder/diagnostic imaging
13.
Neuropharmacology ; 162: 107834, 2020 01 01.
Article En | MEDLINE | ID: mdl-31682853

Resilience to stress is critical for the development of depression. Enhanced adenosine A1 receptor (A1R) signaling mediates the antidepressant effects of acute sleep deprivation (SD). However, chronic SD causes long-lasting upregulation of brain A1R and increases the risk of depression. To investigate the effects of A1R on mood, we utilized two transgenic mouse lines with inducible A1R overexpression in forebrain neurons. These two lines have identical levels of A1R increase in the cortex, but differ in the transgenic A1R expression in the hippocampus. Switching on the transgene promotes robust antidepressant and anxiolytic effects in both lines. The mice of the line without transgenic A1R overexpression in the hippocampus (A1Hipp-) show very strong resistance towards development of stress-induced chronic depression-like behavior. In contrast, the mice of the line in which A1R upregulation extends to the hippocampus (A1Hipp+), exhibit decreased resilience to depression as compared to A1Hipp-. Similarly, automatic analysis of reward behavior of the two lines reveals that depression resistant A1Hipp-transgenic mice exhibit high sucrose preference, while mice of the vulnerable A1Hipp + line developed stress-induced anhedonic phenotype. The A1Hipp + mice have increased Homer1a expression in hippocampus, correlating with impaired long-term potentiation in the CA1 region, mimicking the stressed mice. Furthermore, virus-mediated overexpression of Homer1a in the hippocampus decreases stress resilience. Taken together our data indicate for first time that increased expression of A1R and Homer1a in the hippocampus modulates the resilience to stress-induced depression and thus might potentially mediate the detrimental effects of chronic sleep restriction on mood.


Cerebral Cortex/metabolism , Depression/genetics , Hippocampus/metabolism , Homer Scaffolding Proteins/genetics , Receptor, Adenosine A1/genetics , Resilience, Psychological , Sleep Deprivation/metabolism , Stress, Psychological/genetics , Animals , Behavior, Animal , CA1 Region, Hippocampal/metabolism , Depression/metabolism , Depression/psychology , Elevated Plus Maze Test , Excitatory Postsynaptic Potentials , Hindlimb Suspension , Homer Scaffolding Proteins/metabolism , Long-Term Potentiation/genetics , Mice , Mice, Transgenic , Neurons/metabolism , Open Field Test , Prosencephalon , Receptor, Adenosine A1/metabolism , Reward , Sleep Deprivation/psychology
14.
Neuron ; 104(2): 338-352.e7, 2019 10 23.
Article En | MEDLINE | ID: mdl-31420117

Conventional antidepressants have limited efficacy and many side effects, highlighting the need for fast-acting and specific medications. Induction of the synaptic protein Homer1a mediates the effects of different antidepressant treatments, including the rapid action of ketamine and sleep deprivation (SD). We show here that mimicking Homer1a upregulation via intravenous injection of cell-membrane-permeable TAT-Homer1a elicits rapid antidepressant effects in various tests. Similar to ketamine and SD, in vitro and in vivo application of TAT-Homer1a enhances mGlu5 signaling, resulting in increased mTOR pathway phosphorylation, and upregulates synaptic AMPA receptor expression and activity. The antidepressant action of SD and Homer1a induction depends on mGlu5 activation specifically in excitatory CaMK2a neurons and requires enhanced AMPA receptor activity, translation, and trafficking. Moreover, our data demonstrate a pronounced therapeutic potential of different TAT-fused peptides that directly modulate mGlu5 and AMPA receptor activity and thus might provide a novel strategy for rapid and effective antidepressant treatment.


Behavior, Animal/drug effects , Brain/metabolism , Depressive Disorder, Major/metabolism , Homer Scaffolding Proteins/pharmacology , Receptor, Metabotropic Glutamate 5/drug effects , Receptors, AMPA/drug effects , Synapses/drug effects , Animals , Depressive Disorder, Major/genetics , Disease Models, Animal , Gene Products, tat , Homer Scaffolding Proteins/genetics , Homer Scaffolding Proteins/metabolism , Mice , Mice, Knockout , Peptide Fragments , Receptor, Metabotropic Glutamate 5/genetics , Receptor, Metabotropic Glutamate 5/metabolism , Receptors, AMPA/metabolism , Signal Transduction/drug effects , Sleep Deprivation/metabolism , Synapses/metabolism , TOR Serine-Threonine Kinases/drug effects , Up-Regulation
15.
PLoS One ; 14(6): e0218130, 2019.
Article En | MEDLINE | ID: mdl-31181111

BACKGROUND: Unresponsiveness to dopaminergic therapies is a key feature in the diagnosis of multiple system atrophy (MSA) and a major unmet need in the treatment of MSA patients caused by combined striatonigral degeneration (SND). Transgenic, alpha-synuclein animal models do not recapitulate this lack of levodopa responsiveness. In order to preclinically study interventions including striatal cell grafts, models that feature SND are required. Most of the previous studies focused on extensive nigral and striatal lesions corresponding to advanced MSA-P/SND. The aim of the current study was to replicate mild stage MSA-P/SND with L-dopa failure. METHODS AND RESULTS: Two different striatal quinolinic acid (QA) lesions following a striatal 6-OHDA lesion replicating mild and severe MSA-P/SND, respectively, were investigated and compared to 6-OHDA lesioned animals. After the initial 6-OHDA lesion there was a significant improvement of motor performance after dopaminergic stimulation in the cylinder and stepping test (p<0.001). Response to L-dopa treatment declined in both MSA-P/SND groups reflecting striatal damage of lateral motor areas in contrast to the 6-OHDA only lesioned animals (p<0.01). The remaining striatal volume correlated strongly with contralateral apomorphine induced rotation behaviour and contralateral paw use during L-dopa treatment in cylinder and stepping test (p<0.001). CONCLUSION: Our novel L-dopa response data suggest that L-dopa failure can be induced by restricted lateral striatal lesions combined with dopaminergic denervation. We propose that this sequential striatal double-lesion model replicates a mild stage of MSA-P/SND and is suitable to address neuro-regenerative therapies aimed at restoring dopaminergic responsiveness.


Levodopa/pharmacology , Multiple System Atrophy/etiology , Striatonigral Degeneration/drug therapy , Animals , Behavior, Animal/drug effects , Corpus Striatum/pathology , Denervation , Disease Models, Animal , Motor Activity/drug effects , Quinolinic Acid/adverse effects , Rats
16.
Exp Neurol ; 318: 135-144, 2019 08.
Article En | MEDLINE | ID: mdl-31028828

Inhibitors of cell cycle proteins are known to reduce glial activation and to be neuroprotective in a number of settings. In the context of intracerebral grafting, glial activation is documented to correlate with graft rejection. However, the effects of modification of glial reactivity following grafting in the CNS are poorly understood. Moreover, it is not completely clear if the glial cells themselves trigger the rejection process, or are they secondarily activated. The present study investigated the effect of microglial inhibition by the cyclin-dependant kinase 5 (CDK5) inhibitor roscovitine following intracerebral transplantation in the rodent model of Parkinson's disease. Single cell suspension of rat E14 ventral mesencephalic tissue was transplanted to the dopamine-depleted striatum of unilaterally 6-hydroxydopamine (6-OHDA) lesioned male Sprague-Dawley rats. Experimental animals received injections of roscovitine (20 mg/kg) or a vehicle solution three times following the procedure. Immunohistochemistry was carried out on Day 7 and Day 28 to quantitatively describe the glial reaction adjacent to grafts. The data confirm that systemic roscovitine treatment significantly reduced microglial recruitment adjacent to the grafts on Day 28, without exhibiting significant effects on astroglia. However, this was not found to correlate with elevated numbers of neurons in the grafts. Moreover, microglial reaction surrounding grafts was less pronounced compared to control animals, subjected to the mechanical influence only, even without roscovitine treatment. Our results are the first to show the effect of cell cycle inhibition in the context of neuronal transplantation. The findings suggest that microglial activation around intracerebral grafts can be modified pharmacologically. However, the results do not confirm direct neuroprotective effects of cell cycle inhibition after intracerebral transplantation. Reducing microglial recruitment around grafts could be beneficial by reducing inflammation-related degenerative processes. Sparing astrocytes in the same time provides transplanted cells with essential trophics and support. We consider microglial inhibition to be a possible approach for reducing later graft-related complications.


Astrocytes/drug effects , Brain Tissue Transplantation/methods , Microglia/drug effects , Neuroprotective Agents/pharmacology , Roscovitine/pharmacology , Animals , Brain/drug effects , Brain/pathology , Cyclin-Dependent Kinase 5/antagonists & inhibitors , Male , Parkinsonian Disorders , Protein Kinase Inhibitors/pharmacology , Rats , Rats, Sprague-Dawley
17.
Exp Neurol ; 303: 153-161, 2018 05.
Article En | MEDLINE | ID: mdl-29428214

BACKGROUND: Clinical trials of supra-lateral medial forebrain bundle (MFB) Deep Brain Stimulation (DBS) in treatment resistant major depressive patients have shown rapid and long-term benefits. OBJECTIVE/HYPOTHESIS: The study used Flinders Sensitive Line (FSL) rats with previously identified depressive-like phenotype to assess the range of behavior modification achieved by MFB DBS. METHODS: Male FSL and wild-type Sprague-Dawley rats as Controls were tested on mood/anxiety/exploration, cognitive and motor behaviors. The animals were implanted with bipolar stimulation electrodes in the MFB, and recovery was followed by 10 days of bilateral, chronic and continuous stimulation. RESULTS: Weight dynamics was assessed continuously and indicated similar growth rates although the FSL rats weighed approximately 20-25% less. MFB DBS had no impact on ultrasound calls emitted and the FSL rats continued to vocalize significantly less in the positive affect frequency compared to controls. Similarly, stimulation did not influence the FSL's exploration level (Elevated Plus Maze), nor locomotion (Open Field), although it reduced their freezing behavior (Open Field). Importantly, MFB DBS improved cognitive performance (Double-H) compared to Controls by reducing the time required and the number of errors committed to complete a spatial task. CONCLUSION: MFB DBS in the FSL animals selectively affected certain types of behaviors. Exploration and vocalization remained unaltered, but cognitive performance such as speed and precision of memory recall improved compared to unstimulated and stimulated controls. Future studies should focus on the mechanisms of action of MFB DBS, and in particular on the role of dopamine in the stimulation-dependent phenotype changes.


Deep Brain Stimulation/methods , Depression/therapy , Disease Models, Animal , Medial Forebrain Bundle/physiology , Animals , Body Weight/physiology , Depression/genetics , Electrodes, Implanted , Exploratory Behavior , Male , Maze Learning , Rats , Rats, Mutant Strains , Rats, Sprague-Dawley , Swimming , Time Factors , Tyrosine 3-Monooxygenase/metabolism , Vocalization, Animal/physiology
18.
Balkan Med J ; 35(2): 141-147, 2018 03 15.
Article En | MEDLINE | ID: mdl-29039346

BACKGROUND: Transplantation of fetal mesencephalic tissue is a well-established concept for functional reinnervation of the dopamine-depleted rat striatum. However, there is no extensive description of the glial response of the host brain following this procedure. AIMS: The present study aimed to quantitatively and qualitatively analyse astrogliosis surrounding intrastriatal grafts and compare it to the reaction to mechanical injury with the transplantation instrument only. STUDY DESIGN: Animal experimentation. METHODS: The standard 6-hydroxydopamine-induced unilateral model of Parkinson's disease was used. The experimental animals received transplantation of a single-cell suspension of E14 ventral mesencephalic tissue. Control animals (sham-transplanted) were subjected to injury by the transplantation cannula, without injection of a cell suspension. Histological analyses were carried out 7 and 28 days following the procedure by immunohistochemistry assays for tyrosine hydroxylase and glial fibrillary acidic protein. To evaluate astrogliosis, the cell density and immunopositive area were measured in distinct zones within and surrounding the grafts or the cannula tract. RESULTS: Statistical analysis revealed that astrogliosis in the grafted striatum increased from day 7 to day 28, as shown by a significant change in both cell density and the immunopositive area. The cell density increased from 816.7±370.6 to 1403±272.1 cells/mm2 (p<0.0001) аnd from 523±245.9 to 1164±304.8 cells/mm2 (p<0.0001) in the two zones in the graft core, and from 1151±218.6 to 1485±210.6 cells/mm2 (p<0.05) for the zone in the striatum immediately adjacent to the graft. The glial fibrillary acidic protein-expressing area increased from 0.3109±0.1843 to 0.7949±0.1910 (p<0.0001) and from 0.1449±0.1240 to 0.702±0.2558 (p<0.0001) for the same zones in the graft core, and from 0.5277±0.1502 to 0.6969±0.1223 (p<0.0001) for the same area adjacent to the graft zone. However, astrogliosis caused by mechanical impact only (control) did not display such dynamics. This finding suggests an influence of the grafted cells on the host's glia, possibly through cross-talk between astrocytes and transplanted neurons. CONCLUSION: This bidirectional relationship is affected by multiple factors beyond the mechanical trauma. Elucidation of these factors might help achieve better functional outcomes after intracerebral transplantation.


Astrocytes/transplantation , Brain Tissue Transplantation , Cell Transplantation , Disease Models, Animal , Fetal Tissue Transplantation , Parkinson Disease/therapy , Animals , Male , Rats , Rats, Sprague-Dawley , Rodentia
19.
eNeuro ; 4(5)2017.
Article En | MEDLINE | ID: mdl-28966974

Restorative therapy concepts, such as cell based therapies aim to restitute impaired neurotransmission in neurodegenerative diseases. New strategies to enhance grafted cell survival and integration are still needed to improve functional recovery. Anodal direct current stimulation (DCS) promotes neuronal activity and secretion of the trophic factor BDNF in the motor cortex. Transcranial DCS applied to the motor cortex transiently improves motor symptoms in Parkinson's disease (PD) patients. In this proof-of-concept study, we combine cell based therapy and noninvasive neuromodulation to assess whether neurotrophic support via transcranial DCS would enhance the restitution of striatal neurotransmission by fetal dopaminergic transplants in a rat Parkinson model. Transcranial DCS was applied daily for 20 min on 14 consecutive days following striatal transplantation of fetal ventral mesencephalic (fVM) cells derived from transgenic rat embryos ubiquitously expressing GFP. Anodal but not cathodal transcranial DCS significantly enhanced graft survival and dopaminergic reinnervation of the surrounding striatal tissue relative to sham stimulation. Behavioral recovery was more pronounced following anodal transcranial DCS, and behavioral effects correlated with the degree of striatal innervation. Our results suggest anodal transcranial DCS may help advance cell-based restorative therapies in neurodegenerative diseases. In particular, such an assistive approach may be beneficial for the already established cell transplantation therapy in PD.


Cell Transplantation/methods , Dopaminergic Neurons/transplantation , Parkinson Disease/therapy , Transcranial Direct Current Stimulation/methods , Adrenergic Agents/toxicity , Animals , Cell Survival , Disease Models, Animal , Dopaminergic Neurons/physiology , Female , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Motor Activity , Oxidopamine/toxicity , Parkinson Disease/etiology , Rats , Rats, Sprague-Dawley , Recovery of Function , Tyrosine 3-Monooxygenase/metabolism
20.
Prog Brain Res ; 230: 305-329, 2017.
Article En | MEDLINE | ID: mdl-28552234

Over the last decade, neural transplantation has emerged as one of the more promising, albeit highly experimental, potential therapeutics in neurodegenerative disease. Preclinical studies in rat lesion models of Huntington's disease (HD) and Parkinson's disease (PD) have shown that transplanted precursor neuronal tissue from a fetus into the lesioned striatum can survive, integrate, and reconnect circuitry. Importantly, specific training on behavioral tasks that target striatal function is required to encourage functional integration of the graft to the host tissue. Indeed, "learning to use the graft" is a concept recently adopted in preclinical studies to account for unpredicted profiles of recovery posttransplantation and is an emerging strategy for improving graft functionality. Clinical transplant studies in HD and PD have resulted in mixed outcomes. Small sample sizes and nonstandardized experimental procedures from trial to trial may explain some of this variability. However, it is becoming increasingly apparent that simply replacing the lost neurons may not be sufficient to ensure the optimal graft effects. The knowledge gained from preclinical grafting and training studies suggests that lifestyle factors, including physical activity and specific cognitive and/or motor training, may be required to drive the functional integration of grafted cells and to facilitate the development of compensatory neural networks. The clear implications of preclinical studies are that physical activity and cognitive training strategies are likely to be crucial components of clinical cell replacement therapies in the future. In this chapter, we evaluate the role of general activity in mediating the physical ability of cells to survive, sprout, and extend processes following transplantation in the adult mammalian brain, and we consider the impact of general and specific activity at the behavioral level on functional integration at the cellular and physiological level. We then highlight specific research questions related to timing, intensity, and specificity of training in preclinical models and synthesize the current state of knowledge in clinical populations to inform the development of a strategy for neural transplantation rehabilitation training.


Brain Tissue Transplantation/rehabilitation , Corpus Striatum , Huntington Disease/therapy , Neurons/transplantation , Parkinson Disease/therapy , Animals , Rats
...