TABLE 3 - uploaded by Christoph Kleinschnitz
Content may be subject to copyright.
Comparison of Gait Parameters Using the CatWalk System: Before Photothrombosis and Thereafter, When Mesencephalic Locomotor Region High-Frequency Stimulation Was Applied

Comparison of Gait Parameters Using the CatWalk System: Before Photothrombosis and Thereafter, When Mesencephalic Locomotor Region High-Frequency Stimulation Was Applied

Source publication
Article
Full-text available
Objective: One-third of all stroke survivors are unable to walk, even after intensive physiotherapy. Thus, other concepts to restore walking are needed. Since electrical stimulation of the mesencephalic locomotor region (MLR) is known to elicit gait movements, this area might be a promising target for restorative neurostimulation in stroke patient...

Context in source publication

Context 1
... we compared gait parameters before photo- thrombosis and thereafter when MLR-HFS was applied (Table 3). There was no significant difference with respect to step cycle and stride length between the base- line and stimulated stroke condition and duty cycle and swing speed of the forepaws. ...

Similar publications

Article
Full-text available
Recovery of upper limb (UL) impairment after stroke is limited in stroke survivors. Since stroke can be considered as a network disorder, neuromodulation may be an approach to improve UL motor dysfunction. Here, we evaluated the effect of high-frequency stimulation (HFS) of the subthalamic nucleus (STN) in rats on forelimb grasping using the single...
Article
Full-text available
Deep brain stimulation of the mesencephalic locomotor region (MLR) improves the motor symptoms in Parkinson’s disease and experimental stroke by intervening in the motor cerebral network. Whether high-frequency stimulation (HFS) of the MLR is involved in non-motor processes, such as neuroprotection and inflammation in the area surrounding the photo...

Citations

... In our data, we observed that stereotactic hematoma puncture and drainage not only significantly increased the survival rate of PPH patients but also led to a notable increase in the proportion of patients with moderate to severe disability, thereby improving functional outcomes to some extent. Furthermore, with the development of various rehabilitation techniques, such as neurostimulation therapy showing tremendous potential in enhancing functional outcomes for stroke patients, surviving PPH patients, even those with moderate to severe or profound disability, may benefit from rehabilitation therapy, thereby improving the ultimate clinical outcome [21][22][23]. However, the selection of surgical treatment for PPH remains subjective, lacking an objective scale to predict the outcome of patients after stereotactic hematoma puncture and drainage [24].To address this, we formulated a nomogram to predict the probability of death Moderately severe disability, unable to walk without assistance and unable to attend to own bodily needs without assistance. ...
... There is also evidence for the proposed use of new technologies and techniques, such as deep brain stimulation of midbrain motor centers (mesencephalic motor cortex), as therapeutic strategies to restore motor function after SCIs or stroke provoking plastic changes. Very promising results have been obtained in rodent models with a >80% spinal cord transection, and this technique resulted in acutely functional hindlimb walking and swimming movements [130,131]. The use of stem cellbased regenerative therapy is an innovative approach to inducing a new type of plasticity, which can be produced through SCIs. ...
Article
Full-text available
A spinal cord injury (SCI) causes changes in brain structure and brain function due to the direct effects of nerve damage, secondary mechanisms, and long-term effects of the injury, such as paralysis and neuropathic pain (NP). Recovery takes place over weeks to months, which is a time frame well beyond the duration of spinal shock and is the phase in which the spinal cord remains unstimulated below the level of injury and is associated with adaptations occurring throughout the nervous system, often referred to as neuronal plasticity. Such changes occur at different anatomical sites and also at different physiological and molecular biological levels. This review aims to investigate brain plasticity in patients with SCIs and its influence on the rehabilitation process. Studies were identified from an online search of the PubMed, Web of Science, and Scopus databases. Studies published between 2013 and 2023 were selected. This review has been registered on OSF under (n) 9QP45. We found that neuroplasticity can affect the sensory-motor network, and different protocols or rehabilitation interventions can activate this process in different ways. Exercise rehabilitation training in humans with SCIs can elicit white matter plasticity in the form of increased myelin water content. This review has demonstrated that SCI patients may experience plastic changes either spontaneously or as a result of specific neurorehabilitation training, which may lead to positive outcomes in functional recovery. Clinical and experimental evidence convincingly displays that plasticity occurs in the adult CNS through a variety of events following traumatic or non-traumatic SCI. Furthermore, efficacy-based, pharmacological, and genetic approaches, alone or in combination, are increasingly effective in promoting plasticity.
... In healthy subjects, skilled hand movements are related to coordinated neural networks, which are disturbed after stroke [14]. A promising tool for retuning brain networks is electrical stimulation of distinct brain areas which has been shown to enhance recovery in animal stroke models [60][61][62]. Here we sought to determine the impact of STN-HFS on skilled forelimb reaching and its modulating effect on brain networks in rats undergoing photothrombotic stroke. ...
... Induction of photothrombotic stroke was carried out in rats as described elsewhere [60]. In brief, under deep anesthesia (isoflurane 2.5%), the head of the rat was fixed in a stereotactic frame. ...
... Lesion size was visualized using T2-weighted (T2w) magnetic resonance imaging (MRI) on a 3.0T scanner (MAGNETOM Trio; Siemens, Erlangen Germany) in rats undergoing a 7-day STN-HFS or sham stimulation as reported elsewhere [60]. T2w scans were acquired with turbo spin-echo sequences (echo time, 105 ms, repetition time, 2100 ms) and infarct volume was determined using ImageJ Analysis Software 1.45s (National Institutes of Health, Bethesda, MD, USA. ...
Article
Full-text available
Recovery of upper limb (UL) impairment after stroke is limited in stroke survivors. Since stroke can be considered as a network disorder, neuromodulation may be an approach to improve UL motor dysfunction. Here, we evaluated the effect of high-frequency stimulation (HFS) of the subthalamic nucleus (STN) in rats on forelimb grasping using the single-pellet reaching (SPR) test after stroke and determined costimulated brain regions during STN-HFS using 2-[18F]Fluoro-2-deoxyglucose-([18F]FDG)-positron emission tomography (PET). After a 4-week training of SPR, photothrombotic stroke was induced in the sensorimotor cortex of the dominant hemisphere. Thereafter, an electrode was implanted in the STN ipsilateral to the infarction, followed by a continuous STN-HFS or sham stimulation for 7 days. On postinterventional day 2 and 7, an SPR test was performed during STN-HFS. Success rate of grasping was compared between these two time points. [18F]FDG-PET was conducted on day 2 and 3 after stroke, without and with STN-HFS, respectively. STN-HFS resulted in a significant improvement of SPR compared to sham stimulation. During STN-HFS, a significantly higher [18F]FDG-uptake was observed in the corticosubthalamic/pallidosubthalamic circuit, particularly ipsilateral to the stimulated side. Additionally, STN-HFS led to an increased glucose metabolism within the brainstem. These data demonstrate that STN-HFS supports rehabilitation of skilled forelimb movements, probably by retuning dysfunctional motor centers within the cerebral network.
... Äquivalent zu Versuch A war die Präparation des Tier-Modells kein Bestandteil dieser Doktorarbeit. Für tiefergehende Informationen hinsichtlich der Erzeugung des Schlaganfalls sowie der Implantation der Elektrode wird auf die Literaturangaben[54,58] verwiesen. ...
Thesis
Full-text available
Die präklinische Forschung stellt den ersten wichtigen Meilenstein in der Klärung und Untersuchung klinisch-relevanter Erkrankungen dar. Darüber hinaus unterstützt die präklinische Forschung erheblich die Entwicklung von Therapien. Die Kleintier-Positronenemissionstomographie (µ-PET) spielt dabei eine wichtige Rolle, da sie in der Lage ist, funktionelle, physiologische und biochemische Prozesse in vivo darzustellen und zu quantifizieren. Trotz diverser etablierter PET-Datenauswertungs-Programme bleibt die Analyse von in vivo akquirierten Bilddaten aufgrund der Vielzahl an medizinischen Fragestellungen, der Komplexität der Krankheitsbilder, sowie der Etablierung neuer Radiotracer weiterhin eine große Herausforderung in der Medizin. Ziel dieser Doktorarbeit ist es daher, ein geeignetes, brauchbares Auswertungstool für eine einfache und effiziente Analyse von akquirierten µ-PET-Daten zu entwickeln und zu etablieren, welches das Spektrum bereits vorhandener Programme erweitert. Das entwickelte nuklearmedizinische Datenverarbeitungs-Analyseprogramm (engl. nuclear medicine data processing analysis tool, NU_DPA) wurde in Matlab implementiert und anhand dreier präklinischer Versuchs- bzw. Testreihen erprobt und etabliert. Bei den Datenreihen handelt es sich um µ-PET-Datensätze verschiedener Schlaganfall-Rattenhirnmodelle unter Verwendung folgender Radiotracer. Zum einen die im Gehirn homogen akkumulierende 2-[18F]Fluor-2-desoxy-glukose ([18F]FDG) zum anderen das spezifisch an P-Selektin anreichernde [68Ga]Fucoidan. Das NU_DPA umfasst die automatische Selektion des Zielvolumens (volume-of-interest, VOI) aus dem vollständigen PET-Bild und die anschließende Ausrichtung des VOI mit Hilfe eines PET-Templates (gemittelter PET-Datensatz). Dieses PET Template wird aus den eigenen akquirierten PET-Daten erstellt. Durch das Einbinden eines geeigneten anatomischen MRT-Atlas‘ (anpassbar) können die ausgerichteten PET-Daten einzelnen, Atlas-spezifischen Teilregionen zugeordnet werden. Eine solche Subklassifikation des VOI erlaubt eine genauere Betrachtung und Auswertung der Radiotracer-Akkumulation. Des Weiteren bietet NU_DPA die Möglichkeit einer semiquantitativen Auswertung der PET-Bilddaten anhand von drei unterschiedlichen Parametern, der normalisierten Aktivität, dem Standardized Uptake Value und der Uptake Ratio. Durch die Matlab-integrierten Statistik-Algorithmen ist zusätzlich eine Möglichkeit der statistischen Auswertung der zuvor berechneten Parameter gegeben. Das NU_DPA-Programm stellt somit ein semi-automatisiertes Datenauswertungs-Programm dar, das sowohl die Registrierung als auch die semiquantitative Auswertung von PET-Bilddaten innerhalb einer Versuchsreihe ermöglicht und bereits erfolgreich für die Radiotracer [18F]FDG und [68Ga]Fucoidan in Tiermodellen getestet wurde. Nach derzeitigem Kenntnisstand ist kein Datenauswertungs-Programm bekannt, das PET-Bilddaten unter Verwendung des hinzugefügten Atlas‘ semi-automatisiert analysieren kann und potenziell für homogene und Target-spezifisch akkumulierende Radiotracer geeignet ist.
... [24][25][26][27] On the other hand, the CNF is known to be a main control region for locomotion initiation, maintenance and speed regulation. 23 28 29 Recently, the MLR has gained scientific and clinical interest as target for DBS to improve deficient gait after SCI 16 and stroke 30 with the CNF being proposed as main therapeutic target in recent rodent studies. 23 28 29 Acute electrical activation of the rat MLR has been shown to enable close to physiological hindlimb movements during walking and swimming in a rodent model of chronic incomplete SCI resembling an American Spinal Injury Association (ASIA) Impairment Scale (AIS) D score in humans. ...
... In an acute rodent stroke model, MLR-DBS was able to improve walking speed and limb coordination. 30 DBS in humans is considered safe, reversible and minimally invasive, and is being routinely and successfully applied in the treatment of various movement disorders [31][32][33][34][35][36] with great technical progress in recent years. [37][38][39] While DBS of the PPN in Parkinson's disease has not only yielded clearly positive therapeutic effects, 40 the CNF might be a promising target for locomotion initiation. ...
Article
Full-text available
Introduction Spinal cord injury (SCI) is a devastating condition with immediate impact on the individual’s health and quality of life. Major functional recovery reaches a plateau 3–4 months after injury despite intensive rehabilitative training. To enhance training efficacy and improve long-term outcomes, the combination of rehabilitation with electrical modulation of the spinal cord and brain has recently aroused scientific interest with encouraging results. The mesencephalic locomotor region (MLR), an evolutionarily conserved brainstem locomotor command and control centre, is considered a promising target for deep brain stimulation (DBS) in patients with SCI. Experiments showed that MLR-DBS can induce locomotion in rats with spinal white matter destructions of >85%. Methods and analysis In this prospective one-armed multi-centre study, we investigate the safety, feasibility, and therapeutic efficacy of MLR-DBS to enable and enhance locomotor training in severely affected, subchronic and chronic American Spinal Injury Association Impairment Scale C patients in order to improve functional recovery. Patients undergo an intensive training programme with MLR-DBS while being regularly followed up until 6 months post-implantation. The acquired data of each timepoint are compared with baseline while the primary endpoint is performance in the 6-minute walking test. The clinical trial protocol was written in accordance with the Standard Protocol Items: Recommendations for Interventional Trials checklist. Ethics and dissemination This first in-man study investigates the therapeutic potential of MLR-DBS in SCI patients. One patient has already been implanted with electrodes and underwent MLR stimulation during locomotion. Based on the preliminary results which promise safety and feasibility, recruitment of further patients is currently ongoing. Ethical approval has been obtained from the Ethical Committee of the Canton of Zurich (case number BASEC 2016-01104) and Swissmedic (10000316). Results will be published in peer-reviewed journals and presented at conferences. Trial registration number NCT03053791 .
... We used deep learning to detect locomotor movements in a linear corridor and in an open-field arena. It is relevant to determine whether MLR-evoked locomotion can be dynamically adapted to the environment, as MLR stimulation is explored to improve locomotor function in Parkinson's disease (Plaha and Gill, 2005;Hamani et al., 2016a,b;Goetz et al., 2019) and in animal models of spinal cord injury (Bachmann et al., 2013;Richardson, 2014;Roussel et al., 2019;for review Chari et al., 2017) and stroke (Fluri et al., 2017). We focused on the CnF, which is increasingly considered as the optimal subregion to target within the MLR (Chang et al., 2020). ...
... This supports the idea that distinct brainstem circuits control speed (Lee et al., 2014;Roseberry et al., 2016;Capelli et al., 2017;Caggiano et al., 2018;Josset et al., 2018) and braking/turning movements in mammals (Bouvier et al., 2015;Lemieux and Bretzner, 2019;Cregg et al., 2020;Usseglio et al., 2020; Figure 7). This also suggests that MLR glutamatergic neurons (and especially CnF glutamatergic neurons, Chang et al., 2020) are a relevant target to improve navigation adaptable to the environment in conditions where locomotion is impaired such as Parkinson's disease (Plaha and Gill, 2005;Hamani et al., 2016a,b;Goetz et al., 2019), spinal cord injury (Bachmann et al., 2013;Richardson, 2014;Roussel et al., 2019;for review Chari et al., 2017) and stroke (Fluri et al., 2017). ...
Article
Full-text available
A key function of the mesencephalic locomotor region (MLR) is to control the speed of forward symmetrical locomotor movements. However, the ability of freely moving mammals to integrate environmental cues to brake and turn during MLR stimulation is poorly documented. Here, we investigated whether freely behaving mice could brake or turn, based on environmental cues during MLR stimulation. We photostimulated the cuneiform nucleus (part of the MLR) in mice expressing channelrhodopsin in Vglut2-positive neurons in a Cre-dependent manner (Vglut2-ChR2-EYFP) using optogenetics. We detected locomotor movements using deep learning. We used patch-clamp recordings to validate the functional expression of channelrhodopsin and neuroanatomy to visualize the stimulation sites. In the linear corridor, gait diagram and limb kinematics were similar during spontaneous and optogenetic-evoked locomotion. In the open-field arena, optogenetic stimulation of the MLR evoked locomotion, and increasing laser power increased locomotor speed. Mice could brake and make sharp turns (~90°) when approaching a corner during MLR stimulation in the open-field arena. The speed during the turn was scaled with the speed before the turn, and with the turn angle. Patch-clamp recordings in Vglut2-ChR2-EYFP mice show that blue light evoked short-latency spiking in MLR neurons. Our results strengthen the idea that different brainstem neurons convey braking/turning and MLR speed commands in mammals. Our study also shows that Vglut2-positive neurons of the cuneiform nucleus are a relevant target to increase locomotor activity without impeding the ability to brake and turn when approaching obstacles, thus ensuring smooth and adaptable navigation. Our observations may have clinical relevance since cuneiform nucleus stimulation is increasingly considered to improve locomotion function in pathological states such as Parkinson’s disease, spinal cord injury, or stroke.
... Gait parameters were labeled right forepaw (RF), right hind paw (RH), left forepaw (LF), and left hind paw (LH). Data were analyzed using CatWalk TM XT 10 software (Fluri et al., 2017). ...
Article
Full-text available
Cerebral ischemia is one of the leading causes of death. Reperfusion is a critical stage after thrombolysis or thrombectomy, accompanied by oxidative stress, excitotoxicity, neuroinflammation, and defects in synapse structure. The process is closely related to the dephosphorylation of actin-binding proteins (e.g., cofilin-1) by specific phosphatases. Although studies of the molecular mechanisms of the actin cytoskeleton have been ongoing for decades, limited studies have directly investigated reperfusion-induced reorganization of actin-binding protein, and little is known about the gene expression of actin-binding proteins. The exact mechanism is still uncertain. The motor cortex is very important to save nerve function; therefore, we chose the penumbra to study the relationship between cerebral ischemia-reperfusion and actin-binding protein. After transient middle cerebral artery occlusion (MCAO) and reperfusion, we confirmed reperfusion and motor function deficit by cerebral blood flow and gait analysis. PCR was used to screen the high expression mRNAs in penumbra of the motor cortex. The high expression of cofilin in this region was confirmed by immunohistochemistry (IHC) and Western blot (WB). The change in cofilin-1 expression appears at the same time as gait imbalance, especially maximum variation and left front swing. It is suggested that cofilin-1 may partially affect motor cortex function. This result provides a potential mechanism for understanding cerebral ischemia-reperfusion.
... The MLR is a physiologically defined area of the midbrain, where low threshold electrical stimulation can evoke locomotion in both decerebrate and alert animals [118][119][120]. It has been identified as a key, phylogenetically preserved, regulatory node within the supraspinal locomotor circuit, believed to integrate numerous sensorimotor, cognitive, and limbic inputs into a final descending "locomotor command" through reticulospinal and monoaminergic neurons projecting to spinal central pattern generators [121][122][123][124]. Interestingly, several studies have shown that electrical stimulation of the MLR is capable of ameliorating gait and postural deficits in rodent models of SCI, stroke, and parkinsonism [125][126][127]. ...
Article
Full-text available
Background: Deep brain stimulation (DBS) of the mesencephalic locomotor region (MLR) has been studied as a therapeutic target in rodent models of stroke, parkinsonism, and spinal cord injury. Clinical DBS trials have targeted the closely related pedunculopontine nucleus in patients with Parkinson’s disease as a therapy for gait dysfunction, with mixed reported outcomes. Recent studies suggest that optimizing the MLR target could improve its effectiveness. Objective: We sought to determine if stereotaxic targeting and DBS in the midbrain of the pig, in a region anatomically similar to that previously identified as the MLR in other species, could initiate and modulate ongoing locomotion, as a step towards generating a large animal neuromodulation model of gait. Methods: We implanted Medtronic 3389 electrodes into putative MLR structures in Yucatan micropigs to characterize the locomotor effects of acute DBS in this region, using EMG recordings, joint kinematics, and speed measurements on a manual treadmill. Results: MLR DBS initiated and augmented locomotion in freely moving micropigs. Effective locomotor sites centered around the cuneiform nucleus and stimulation frequency controlled locomotor speed and stepping frequency. Off-target stimulation evoked defensive and aversive behaviors that precluded locomotion in the animals. Conclusion: Pigs appear to have an MLR and can be used to model neuromodulation of this gait-promoting center. These results indicate that the pig is a useful model to guide future clinical studies for optimizing MLR DBS in cases of gait deficiencies associated with such conditions as Parkinson’s disease, spinal cord injury, or stroke.
... A photothrombotic lesion was inflicted within the right cerebral cortex of all animals as described recently [44]. Briefly, after induction of anesthesia (isoflurane 2.5%), rats were fixed in a stereotactic frame and the skull was exposed. ...
... After inducing a photothrombotic stroke, a stimulating monopolar microelectrode was inserted in the right MLR (coordinates: 7.8 mm posterior, 2.0 mm lateral, and 5.8 mm ventral to the bregma) as described elsewhere [44]. Briefly, a microelectrode (FHC Inc., Bowdoin, ME, USA) was implanted into the MLR using a micromanipulator. ...
... The stimulating amplitude was determined, as follows: beginning with 20 µA, the intensity was increased in steps of 10 µA until maximal locomotion was observed. The lowest amplitude evoking locomotor behavior was chosen for the 24 h MLR-HFS (for details see [44]). At the same time, control animals were connected with a stimulus generator, which was turned off. ...
Article
Full-text available
Inflammation is crucial in the pathophysiology of stroke and thus a promising therapeutic target. High-frequency stimulation (HFS) of the mesencephalic locomotor region (MLR) reduces perilesional inflammation after photothrombotic stroke (PTS). However, the underlying mechanism is not completely understood. Since distinct neural and immune cells respond to electrical stimulation by releasing acetylcholine, we hypothesize that HFS might trigger the cholinergic anti-inflammatory pathway via activation of the α7 nicotinic acetylcholine receptor (α7nAchR). To test this hypothesis, rats underwent PTS and implantation of a microelectrode into the MLR. Three hours after intervention, either HFS or sham-stimulation of the MLR was applied for 24 h. IFN-γ, TNF-α, and IL-1α were quantified by cytometric bead array. Choline acetyltransferase (ChAT)+ CD4+-cells and α7nAchR+-cells were quantified visually using immunohistochemistry. Phosphorylation of NFĸB, ERK1/2, Akt, and Stat3 was determined by Western blot analyses. IFN-γ, TNF-α, and IL-1α were decreased in the perilesional area of stimulated rats compared to controls. The number of ChAT+ CD4+-cells increased after MLR-HFS, whereas the amount of α7nAchR+-cells was similar in both groups. Phospho-ERK1/2 was reduced significantly in stimulated rats. The present study suggests that MLR-HFS may trigger anti-inflammatory processes within the perilesional area by modulating the cholinergic system, probably via activation of the α7nAchR.
... Wang, Bontempi, et al., 2008;Y. Liu et al., 2013;Fluri et al., 2017). Thus, we used gait analysis as one of the parameters to compare functional deficits in stVAChT-KO mice and littermate controls 7 days after stroke. ...
Article
Full-text available
Acetylcholine (ACh) has been suggested to facilitate plasticity and improve functional recovery after different types of brain lesions. Interestingly, numerous studies have shown that striatal cholinergic interneurons are relatively resistant to acute ischemic insults, but whether ACh released by these neurons enhances functional recovery after stroke is unknown. We investigated the role of endogenous striatal ACh in stroke lesion volume and functional outcomes following middle cerebral artery occlusion to induce focal ischemia in striatum-selective vesicular acetylcholine transporter-deficient mice (stVAChT-KO). As transporter expression is almost completely eliminated in the striatum of stVAChT-KO mice, ACh release is nearly abolished in this area. Conversely, in other brain areas, VAChT expression and ACh release are preserved. Our results demonstrate a larger infarct size after ischemic insult in stVAChT-KO mice, with more pronounced functional impairments and increased mortality than in littermate controls. These changes are associated with increased activation of GSK-3, decreased levels of β-catenin, and a higher permeability of the blood-brain barrier in mice with loss of VAChT in striatum neurons. These results support a framework in which endogenous ACh secretion originating from cholinergic interneurons in the striatum helps to protect brain tissue against ischemia-induced damage and facilitates brain recovery by supporting blood-brain barrier function.