Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 1.042
1.
Tidsskr Nor Laegeforen ; 144(7)2024 06 04.
Article En, Nor | MEDLINE | ID: mdl-38832603

Spinal cord stimulation is an important modality of treatment for some patients with chronic pain. Patient satisfaction following this treatment is comparable to outcomes from spine surgery in Norway.


Chronic Pain , Spinal Cord Stimulation , Humans , Chronic Pain/therapy , Spinal Cord Stimulation/methods , Pain Management/methods
2.
BMC Neurol ; 24(1): 167, 2024 May 21.
Article En | MEDLINE | ID: mdl-38773417

BACKGROUND: Postural abnormalities (PA) are common in the advanced stages of Parkinson's disease (PD), but effective therapies are lacking. A few studies suggested that spinal cord stimulation (SCS) could be a potential therapy whereas its effect is still uncertain. We aimed to investigate whether SCS had potential for benefiting PD patients with PA. METHODS: T8-12 SCS was operated on six PD patients with PA and all patients were followed for one year. Evaluations were made before and after SCS. Moreover, three patients were tested separately with SCS on-state and off-state to confirm the efficacy of SCS. RESULTS: Improvements in lateral trunk flexion degree, anterior thoracolumbar flexion degree and motor function were found after SCS. The improvements diminished while SCS was turned off. CONCLUSIONS: Lower thoracic SCS may be effective for improving PA in PD patients, but further studies are needed to confirm this conclusion. TRIAL REGISTRATION: Chinese Clinical Trial Registry, ChiCTR1900024326, Registered on 6th July 2019; https://www.chictr.org.cn/showproj.aspx?proj=40835 .


Parkinson Disease , Postural Balance , Spinal Cord Stimulation , Humans , Spinal Cord Stimulation/methods , Parkinson Disease/therapy , Parkinson Disease/complications , Parkinson Disease/physiopathology , Pilot Projects , Male , Female , Middle Aged , Aged , Prospective Studies , Postural Balance/physiology , Treatment Outcome
3.
Nat Med ; 30(5): 1276-1283, 2024 May.
Article En | MEDLINE | ID: mdl-38769431

Cervical spinal cord injury (SCI) leads to permanent impairment of arm and hand functions. Here we conducted a prospective, single-arm, multicenter, open-label, non-significant risk trial that evaluated the safety and efficacy of ARCEX Therapy to improve arm and hand functions in people with chronic SCI. ARCEX Therapy involves the delivery of externally applied electrical stimulation over the cervical spinal cord during structured rehabilitation. The primary endpoints were safety and efficacy as measured by whether the majority of participants exhibited significant improvement in both strength and functional performance in response to ARCEX Therapy compared to the end of an equivalent period of rehabilitation alone. Sixty participants completed the protocol. No serious adverse events related to ARCEX Therapy were reported, and the primary effectiveness endpoint was met. Seventy-two percent of participants demonstrated improvements greater than the minimally important difference criteria for both strength and functional domains. Secondary endpoint analysis revealed significant improvements in fingertip pinch force, hand prehension and strength, upper extremity motor and sensory abilities and self-reported increases in quality of life. These results demonstrate the safety and efficacy of ARCEX Therapy to improve hand and arm functions in people living with cervical SCI. ClinicalTrials.gov identifier: NCT04697472 .


Arm , Hand , Quadriplegia , Spinal Cord Injuries , Humans , Quadriplegia/therapy , Quadriplegia/physiopathology , Male , Hand/physiopathology , Female , Middle Aged , Adult , Arm/physiopathology , Spinal Cord Injuries/therapy , Spinal Cord Injuries/physiopathology , Spinal Cord Injuries/rehabilitation , Spinal Cord Stimulation/methods , Treatment Outcome , Quality of Life , Prospective Studies , Chronic Disease , Aged , Electric Stimulation Therapy/methods , Electric Stimulation Therapy/adverse effects
4.
Physiol Rep ; 12(9): e16039, 2024 May.
Article En | MEDLINE | ID: mdl-38740563

Evaluating reciprocal inhibition of the thigh muscles is important to investigate the neural circuits of locomotor behaviors. However, measurements of reciprocal inhibition of thigh muscles using spinal reflex, such as H-reflex, have never been systematically established owing to methodological limitations. The present study aimed to clarify the existence of reciprocal inhibition in the thigh muscles using transcutaneous spinal cord stimulation (tSCS). Twenty able-bodied male individuals were enrolled. We evoked spinal reflex from the biceps femoris muscle (BF) by tSCS on the lumber posterior root. We examined whether the tSCS-evoked BF reflex was reciprocally inhibited by the following conditionings: (1) single-pulse electrical stimulation on the femoral nerve innervating the rectus femoris muscle (RF) at various inter-stimulus intervals in the resting condition; (2) voluntary contraction of the RF; and (3) vibration stimulus on the RF. The BF reflex was significantly inhibited when the conditioning electrical stimulation was delivered at 10 and 20 ms prior to tSCS, during voluntary contraction of the RF, and during vibration on the RF. These data suggested a piece of evidence of the existence of reciprocal inhibition from the RF to the BF muscle in humans and highlighted the utility of methods for evaluating reciprocal inhibition of the thigh muscles using tSCS.


Spinal Cord Stimulation , Thigh , Humans , Male , Spinal Cord Stimulation/methods , Adult , Thigh/physiology , Thigh/innervation , Muscle, Skeletal/physiology , Muscle, Skeletal/innervation , Muscle Contraction/physiology , Transcutaneous Electric Nerve Stimulation/methods , Young Adult , H-Reflex/physiology , Femoral Nerve/physiology , Neural Inhibition/physiology , Quadriceps Muscle/physiology , Quadriceps Muscle/innervation , Hamstring Muscles/physiology , Electromyography
5.
J Neural Eng ; 21(3)2024 Jun 07.
Article En | MEDLINE | ID: mdl-38772354

Objective. Spinal cord stimulation (SCS) is a well-established treatment for managing certain chronic pain conditions. More recently, it has also garnered attention as a means of modulating neural activity to restore lost autonomic or sensory-motor function. Personalized modeling and treatment planning are critical aspects of safe and effective SCS (Rowald and Amft 2022 Front. Neurorobotics 16 983072, Wagneret al2018 Nature 563 65-71). However, the generation of spine models at the required level of detail and accuracy requires time and labor intensive manual image segmentation by human experts. This study aims to develop a maximally automated segmentation routine capable of producing high-quality anatomical models, even with limited data, to facilitate safe and effective personalized SCS treatment planning.Approach. We developed an automated image segmentation and model generation pipeline based on a novel convolutional neural network (CNN) architecture trained on feline spinal cord magnetic resonance imaging data. The pipeline includes steps for image preprocessing, data augmentation, transfer learning, and cleanup. To assess the relative importance of each step in the pipeline and our choice of CNN architecture, we systematically dropped steps or substituted architectures, quantifying the downstream effects in terms of tissue segmentation quality (Jaccard index and Hausdorff distance) and predicted nerve recruitment (estimated axonal depolarization).Main results. The leave-one-out analysis demonstrated that each pipeline step contributed a small but measurable increment to mean segmentation quality. Surprisingly, minor differences in segmentation accuracy translated to significant deviations (ranging between 4% and 13% for each pipeline step) in predicted nerve recruitment, highlighting the importance of careful workflow design. Additionally, transfer learning techniques enhanced segmentation metric consistency and allowed generalization to a completely different spine region with minimal additional training data.Significance. To our knowledge, this work is the first to assess the downstream impacts of segmentation quality differences on neurostimulation predictions. It highlights the role of each step in the pipeline and paves the way towards fully automated, personalized SCS treatment planning in clinical settings.


Neural Networks, Computer , Spinal Cord Stimulation , Spinal Cord , Animals , Cats , Spinal Cord Stimulation/methods , Spinal Cord/physiology , Spinal Cord/diagnostic imaging , Image Processing, Computer-Assisted/methods , Magnetic Resonance Imaging/methods
7.
Sci Rep ; 14(1): 9654, 2024 04 26.
Article En | MEDLINE | ID: mdl-38670988

Several neurologic diseases including spinal cord injury, Parkinson's disease or multiple sclerosis are accompanied by disturbances of the lower urinary tract functions. Clinical data indicates that chronic spinal cord stimulation can improve not only motor function but also ability to store urine and control micturition. Decoding the spinal mechanisms that regulate the functioning of detrusor (Detr) and external urethral sphincter (EUS) muscles is essential for effective neuromodulation therapy in patients with disturbances of micturition. In the present work we performed a mapping of Detr and EUS activity by applying epidural electrical stimulation (EES) at different levels of the spinal cord in decerebrated cat model. The study was performed in 5 adult male cats, evoked potentials were generated by EES aiming to recruit various spinal pathways responsible for LUT and hindlimbs control. Recruitment of Detr occurred mainly with stimulation of the lower thoracic and upper lumbar spinal cord (T13-L1 spinal segments). Responses in the EUS, in general, occurred with stimulation of all the studied sites of the spinal cord, however, a pronounced specificity was noted for the lower lumbar/upper sacral sections (L7-S1 spinal segments). These features were confirmed by comparing the normalized values of the slope angles used to approximate the recruitment curve data by the linear regression method. Thus, these findings are in accordance with our previous data obtained in rats and could be used for development of novel site-specific neuromodulation therapeutic approaches.


Spinal Cord , Animals , Cats , Male , Spinal Cord/physiopathology , Electric Stimulation/methods , Spinal Cord Stimulation/methods , Urinary Bladder/physiopathology , Decerebrate State/physiopathology , Urinary Tract/physiopathology , Urethra/physiopathology , Urination/physiology , Epidural Space
8.
Int J Mol Sci ; 25(8)2024 Apr 19.
Article En | MEDLINE | ID: mdl-38674065

Transcutaneous multisegmental spinal cord stimulation (tSCS) has shown superior efficacy in modulating spinal locomotor circuits compared to single-site stimulation in individuals with spinal cord injury (SCI). Building on these findings, we hypothesized that administering a single session of tSCS at multiple spinal segments may yield greater enhancements in muscle strength and gait function during stimulation compared to tSCS at only one or two segments. In our study, tSCS was applied at single segments (C5, L1, and Coc1), two segments (C5-L1, C5-Coc1, and L1-Coc1), or multisegments (C5-L1-Coc1) in a randomized order. We evaluated the 6-m walking test (6MWT) and maximum voluntary contraction (MVC) and assessed the Hmax/Mmax ratio during stimulation in ten individuals with incomplete motor SCI. Our findings indicate that multisegmental tSCS improved walking time and reduced spinal cord excitability, as measured by the Hmax/Mmax ratio, similar to some single or two-site tSCS interventions. However, only multisegmental tSCS resulted in increased tibialis anterior (TA) muscle strength. These results suggest that multisegmental tSCS holds promise for enhancing walking capacity, increasing muscle strength, and altering spinal cord excitability in individuals with incomplete SCI.


Spinal Cord Injuries , Spinal Cord Stimulation , Walking , Humans , Spinal Cord Injuries/therapy , Spinal Cord Injuries/physiopathology , Spinal Cord Injuries/rehabilitation , Walking/physiology , Male , Female , Adult , Middle Aged , Spinal Cord Stimulation/methods , Muscle Strength , Spinal Cord/physiopathology , Muscle, Skeletal/physiopathology , Gait/physiology
9.
Presse Med ; 53(2): 104231, 2024 Jun.
Article En | MEDLINE | ID: mdl-38636785

Neuropathic pain occurs in people experiencing lesion or disease affecting the somatosensorial system. It is present in 7 % of the general population and may not fully respond to first- and second-line treatments in up to 40 % of cases. Neuromodulation approaches are often proposed for those not tolerating or not responding to usual pharmacological management. These approaches can be delivered surgically (invasively) or non-invasively. Invasive neuromodulation techniques were the first to be employed in neuropathic pain. Among them is spinal cord stimulation (SCS), which consists of the implantation of epidural electrodes over the spinal cord. It is recommended in some guidelines for peripheral neuropathic pain. While recent studies have called into question its efficacy, others have provided promising data, driven by advances in techniques, battery capabilities, programming algorithms and software developments. Deep brain stimulation (DBS) is another well-stablished neuromodulation therapy routinely used for movement disorders; however, its role in pain management remains limited to specific research centers. This is not only due to variable results in the literature contesting its efficacy, but also because several different brain targets have been explored in small trials, compromising comparisons between these studies. Structures such as the periaqueductal grey, posterior thalamus, anterior cingulate cortex, ventral striatum/anterior limb of the internal capsule and the insula are the main targets described to date in literature. SCS and DBS present diverse rationales for use, mechanistic backgrounds, and varying levels of support from experimental studies. The present review aims to present their methodological details, main mechanisms of action for analgesia and their place in the current body of evidence in the management of patients with neuropathic pain, as well their particularities, effectiveness, safety and limitations.


Deep Brain Stimulation , Neuralgia , Spinal Cord Stimulation , Humans , Neuralgia/therapy , Deep Brain Stimulation/methods , Spinal Cord Stimulation/methods , Pain Management/methods
10.
J Neurophysiol ; 131(6): 1101-1111, 2024 Jun 01.
Article En | MEDLINE | ID: mdl-38656134

Transspinal (or transcutaneous spinal cord) stimulation is a noninvasive, cost-effective, easily applied method with great potential as a therapeutic modality for recovering somatic and nonsomatic functions in upper motor neuron disorders. However, how transspinal stimulation affects motor neuron depolarization is poorly understood, limiting the development of effective transspinal stimulation protocols for rehabilitation. In this study, we characterized the responses of soleus α motor neurons to single-pulse transspinal stimulation using single-motor unit (SMU) discharges as a proxy given the 1:1 discharge activation between the motor neuron and the motor unit. Peristimulus time histogram, peristimulus frequencygram, and surface electromyography (sEMG) were used to characterize the postsynaptic potentials of soleus motor neurons. Transspinal stimulation produced short-latency excitatory postsynaptic potentials (EPSPs) followed by two distinct phases of inhibitory postsynaptic potentials (IPSPs) in most soleus motor neurons and only IPSPs in others. Transspinal stimulation generated double discharges at short interspike intervals in a few motor units. The short-latency EPSPs were likely mediated by muscle spindle group Ia and II afferents, and the IPSPs via excitation of group Ib afferents and recurrent collaterals of motor neurons leading to activation of diverse spinal inhibitory interneuronal circuits. Further studies are warranted to understand better how transspinal stimulation affects depolarization of α motor neurons over multiple spinal segments. This knowledge will be seminal for developing effective transspinal stimulation protocols in upper motor neuron lesions.NEW & NOTEWORTHY Transspinal stimulation produces distinct actions on soleus motor neurons: an early short-latency excitation followed by two inhibitions or only inhibition and doublets. These results show how transspinal stimulation affects depolarization of soleus α motor neurons in healthy humans.


Motor Neurons , Muscle, Skeletal , Humans , Motor Neurons/physiology , Male , Adult , Muscle, Skeletal/physiology , Female , Excitatory Postsynaptic Potentials/physiology , Spinal Cord Stimulation/methods , Inhibitory Postsynaptic Potentials/physiology , Electromyography , Young Adult , Spinal Cord/physiology
12.
A A Pract ; 18(3): e01766, 2024 Mar 01.
Article En | MEDLINE | ID: mdl-38502524

Dorsal root ganglion stimulation (DRG-S) is a relatively new neuromodulation technique that has shown promising results in the treatment of chronic pain conditions. We present a case of a difficult lead extraction during the explantation of a DRG-S device. The lead was unable to be removed despite multiple attempts until a sheath and stylet were used to facilitate extraction. As DRG-S utilization becomes more widespread, DRG-S device explantation will inevitably become more common. The technique described in this report may be beneficial in certain cases of difficult DRG-S lead extraction.


Chronic Pain , Neuralgia , Spinal Cord Stimulation , Humans , Ganglia, Spinal/physiology , Spinal Cord Stimulation/methods , Chronic Pain/therapy , Neuralgia/therapy , Pain Management/methods
13.
Trials ; 25(1): 223, 2024 Mar 28.
Article En | MEDLINE | ID: mdl-38549128

BACKGROUND: The prevalence of sacroiliac joint pain (SIJP) is estimated to be 10-30% in patients with chronic low back pain. Numerous conservative and surgical treatment modalities for SIJP have been described with limited evidence regarding long-term pain relief. Spinal cord stimulation (SCS) is a well-established technique to treat patients with chronic low back pain. However, the effect on patients with SIJP is not consistent. Therefore, peripheral nerve stimulation (PNS) for chronic SIJP was implemented in experimental trials. Clinical data on PNS for SIJP is still lacking. The authors present a case series and a protocol for a prospective, multicenter study to determine the effect of PNS in patients with chronic intractable SIJP. METHOD: A multicenter, prospective randomized controlled trial was designed. Patients with chronic intractable SIJP will be recruited and randomized in a 4:3 ratio to either the peripheral nerve stimulation group or to the best medical treatment group. A total of 90 patients are planned to be enrolled (52 in the PNS group and 38 in the BMT group). Patients in the intervention group receive a percutaneous implantation of a unilateral or bilateral lead which is externalized for a trial phase for 3-14 days. After trial phase only patients with at least 50% reduction of pain receive an impulse generator for permanent stimulation. Regular visits for participants are planned on day 0, after 3 months (± 30 days), 6 months (± 30 days), and 12 months (± 60 days). The primary outcome measurements is the difference in Numeric Pain Rating Scale (NRS) between baseline and after 6 months. Secondary outcomes is improvement of pain associated disability (ODI) and improvement of health-related quality of life after 6 and 12 months. DISCUSSION: We have described the protocol for a prospective, multicenter, randomized trial evaluating the influence of PNS on patients with chronic sacroiliac joint syndrome. We believe that PNS on patients with chronic sacroiliac joint syndrome will show promising results regarding pain relief and quality of life in comparison to BMT after 12 months. The design of this trial promises high evidence in comparison to the data to date. TRIAL REGISTRATION: ClinicalTrials.gov, NCT05357300. Registered on April 26, 2022.


Chronic Pain , Low Back Pain , Spinal Cord Stimulation , Humans , Low Back Pain/diagnosis , Low Back Pain/therapy , Sacroiliac Joint , Quality of Life , Prospective Studies , Chronic Pain/diagnosis , Chronic Pain/therapy , Spinal Cord Stimulation/methods , Peripheral Nerves , Treatment Outcome , Randomized Controlled Trials as Topic , Multicenter Studies as Topic
14.
Exp Neurol ; 376: 114754, 2024 Jun.
Article En | MEDLINE | ID: mdl-38493983

Spasticity is a complex and multidimensional disorder that impacts nearly 75% of individuals with spinal cord injury (SCI) and currently lacks adequate treatment options. This sensorimotor condition is burdensome as hyperexcitability of reflex pathways result in exacerbated reflex responses, co-contractions of antagonistic muscles, and involuntary movements. Transcutaneous spinal cord stimulation (tSCS) has become a popular tool in the human SCI research field. The likeliness for this intervention to be successful as a noninvasive anti-spastic therapy after SCI is suggested by a mild and transitory improvement in spastic symptoms following a single stimulation session, but it remains to be determined if repeated tSCS over the course of weeks can produce more profound effects. Despite its popularity, the neuroplasticity induced by tSCS also remains widely unexplored, particularly due to the lack of suitable animal models to investigate this intervention. Thus, the basis of this work was to use tSCS over multiple sessions (multi-session tSCS) in a rat model to target spasticity after SCI and identify the long-term physiological improvements and anatomical neuroplasticity occurring in the spinal cord. Here, we show that multi-session tSCS in rats with an incomplete (severe T9 contusion) SCI (1) decreases hyperreflexia, (2) increases the low frequency-dependent modulation of the H-reflex, (3) prevents potassium-chloride cotransporter isoform 2 (KCC2) membrane downregulation in lumbar motoneurons, and (4) generally augments motor output, i.e., EMG amplitude in response to single pulses of tSCS, particularly in extensor muscles. Together, this work displays that multi-session tSCS can target and diminish spasticity after SCI as an alternative to pharmacological interventions and begins to highlight the underlying neuroplasticity contributing to its success in improving functional recovery.


Homeostasis , Rats, Sprague-Dawley , Reflex, Abnormal , Spinal Cord Injuries , Spinal Cord Stimulation , Animals , Spinal Cord Injuries/complications , Spinal Cord Injuries/physiopathology , Spinal Cord Injuries/therapy , Rats , Homeostasis/physiology , Reflex, Abnormal/physiology , Spinal Cord Stimulation/methods , Female , Chlorides/metabolism , Muscle Spasticity/etiology , Muscle Spasticity/therapy , Neuronal Plasticity/physiology
15.
Ann Neurol ; 95(5): 966-983, 2024 May.
Article En | MEDLINE | ID: mdl-38450773

OBJECTIVE: Neuropathic pain poses a persistent challenge in clinical management. Neuromodulation has emerged as a last-resort therapy. Conventional spinal cord stimulation (Con SCS) often causes abnormal sensations and provides short analgesia, whereas high-frequency spinal cord stimulation (HF SCS) is a newer therapy that effectively alleviates pain without paresthesia. However, the modes of action of 10kHz HF SCS (HF10 SCS) in pain relief remain unclear. To bridge this knowledge gap, we employed preclinical models that mimic certain features of clinical SCS to explore the underlying mechanisms of HF10 SCS. Addressing these issues would provide the scientific basis for improving and evaluating the effectiveness, reliability, and practicality of different frequency SCS in clinical settings. METHODS: We established a preclinical SCS model to examine its effects in a neuropathic pain rat model. We conducted bulk and single-cell RNA sequencing in the spinal dorsal horn (SDH) to examine cellular and molecular changes under different treatments. We employed genetic manipulations through intrathecal injection of a lentiviral system to explore the SCS-mediated signaling axis in pain. Various behavioral tests were performed to evaluate pain conditions under different treatments. RESULTS: We found that HF10 SCS significantly reduces immune responses in the SDH by inactivating the Kaiso-P2X7R pathological axis in microglia, promoting long-lasting pain relief. Targeting Kaiso-P2X7R in microglia dramatically improved efficacy of Con SCS treatment, leading to reduced neuroinflammation and long-lasting pain relief. INTERPRETATION: HF10 SCS could improve the immunopathologic state in the SDH, extending its benefits beyond symptom relief. Targeting the Kaiso-P2X7R axis may enhance Con SCS therapy and offer a new strategy for pain management. ANN NEUROL 2024;95:966-983.


Inflammation , Microglia , Neuralgia , Rats, Sprague-Dawley , Receptors, Purinergic P2X7 , Spinal Cord Stimulation , Animals , Neuralgia/therapy , Neuralgia/metabolism , Rats , Microglia/metabolism , Spinal Cord Stimulation/methods , Male , Receptors, Purinergic P2X7/metabolism , Receptors, Purinergic P2X7/genetics , Inflammation/therapy , Disease Models, Animal
16.
World Neurosurg ; 185: e820-e826, 2024 May.
Article En | MEDLINE | ID: mdl-38432508

OBJECTIVE: To examine if the use of an antibacterial envelope (TYRX) decreases the rate of postoperative infection in chronic pain patients undergoing treatment with spinal cord stimulation (SCS) involving device implantation. METHODS: Single-center retrospective cohort study comparing postoperative infections rates in non-TYRX recipients from 2018 to 2020 with recipients of a TYRX antibacterial envelope from 2020 to 2021. Infection was registered if a patient received any form of antibiotic treatment after hospital discharge within a follow-up period of 100 days. RESULTS: A total of 198 patients were included: 100 in the TYRX group and 98 in the non-TYRX group. There were no significant differences between the 2 groups regarding age, body mass index (BMI), smoking, diabetes, and use of immunosuppression. The overall infection rate was 5.6%. The infection rate was 4% in the TYRX group and 7.1% in the non-TYRX group (P = 0.6). However, the 4 cases of postoperative infection in the TYRX group could be effectively managed with oral antibiotics alone, whereas 6 out of the 7 patients in the non-TYRX group required intravenous antibiotics. Moreover, device explantation was necessary in 3 of these patients suggesting the event of more severe infections in the non-TYRX group (P = 0.014). CONCLUSIONS: The TYRX antibacterial envelope displayed infection rates reducing capabilities, along with a clear tendency to reduce revision surgeries and system removals due to infections.


Anti-Bacterial Agents , Spinal Cord Stimulation , Humans , Spinal Cord Stimulation/methods , Female , Middle Aged , Male , Anti-Bacterial Agents/therapeutic use , Retrospective Studies , Aged , Adult , Surgical Wound Infection/prevention & control , Surgical Wound Infection/epidemiology , Chronic Pain/therapy , Cohort Studies
17.
Exp Brain Res ; 242(4): 959-970, 2024 Apr.
Article En | MEDLINE | ID: mdl-38416179

Transcutaneous spinal stimulation (TSS) studies rely on the depolarization of afferent fibers to provide input to the spinal cord; however, this has not been routinely ascertained. Thus, we aimed to characterize the types of responses evoked by TSS and establish paired-pulse ratio cutoffs that distinguish posterior root reflexes, evoked by stimulation of afferent nerve fibers, from motor responses, evoked by stimulation of efferent nerve fibers. Twelve neurologically intact participants (six women) underwent unipolar TSS (cathode over T11-12 spinal processes, anode paraumbilically) while resting supine. In six participants, unipolar TSS was repeated 2-3 months later and also compared to a bipolar TSS configuration (cathode 2.5 cm below T11-12, anode 5 cm above cathode). EMG signals were recorded from 16 leg muscles. A paired-pulse paradigm was applied at interstimulus intervals (ISIs) of 25, 50, 100, 200, and 400 ms. Responses were categorized by three assessors into reflexes, motor responses, or their combination (mixed responses) based on the visual presence/absence of paired-pulse suppression across ISIs. The paired-pulse ratio that best discriminated between response types was derived for each ISI. These cutoffs were validated by repeating unipolar TSS 2-3 months later and with bipolar TSS. Unipolar TSS evoked only reflexes (90%) and mixed responses (10%), which were mainly recorded in the quadriceps muscles (25-42%). Paired-pulse ratios of 0.51 (25-ms ISI) and 0.47 (50-ms ISI) best distinguished reflexes from mixed responses (100% sensitivity, > 99.2% specificity). These cutoffs performed well in the repeated unipolar TSS session (100% sensitivity, > 89% specificity). Bipolar TSS exclusively elicited reflexes which were all correctly classified. These results can be utilized in future studies to ensure that the input to the spinal cord originates from the depolarization of large afferents. This knowledge can be applied to improve the design of future neurophysiological studies and increase the fidelity of neuromodulation interventions.


Spinal Cord Stimulation , Spinal Cord , Humans , Female , Spinal Cord/physiology , Reflex/physiology , Muscle, Skeletal/physiology , Leg/physiology , Spinal Cord Stimulation/methods , Electric Stimulation/methods
18.
Neurotherapeutics ; 21(3): e00330, 2024 Apr.
Article En | MEDLINE | ID: mdl-38340524

Over the past 30 years, the field of neuromodulation has witnessed remarkable advancements. These developments encompass a spectrum of techniques, both non-invasive and invasive, that possess the ability to both probe and influence the central nervous system. In many cases neuromodulation therapies have been adopted into standard care treatments. Transcranial magnetic stimulation (TMS), transcranial direct current stimulation (tDCS), and transcranial ultrasound stimulation (TUS) are the most common non-invasive methods in use today. Deep brain stimulation (DBS), spinal cord stimulation (SCS), and vagus nerve stimulation (VNS), are leading surgical methods for neuromodulation. Ongoing active clinical trials using are uncovering novel applications and paradigms for these interventions.


Deep Brain Stimulation , Transcranial Direct Current Stimulation , Transcranial Magnetic Stimulation , Humans , Deep Brain Stimulation/methods , Transcranial Magnetic Stimulation/methods , Transcranial Direct Current Stimulation/methods , Spinal Cord Stimulation/methods , Vagus Nerve Stimulation/methods , Vagus Nerve Stimulation/trends
19.
Neurophysiol Clin ; 54(3): 102945, 2024 May.
Article En | MEDLINE | ID: mdl-38422720

Neuromodulation therapy, like spinal cord stimulation (SCS), benefits individuals with chronic diseases, improving outcomes of patients with heart failure (HF). This systematic review aims to investigate the efficacy of SCS when used as an adjunctive therapy in HF. A systematic analysis of all studies that included SCS therapy in human participants with HF was conducted. After excluding studies not meeting specific criteria, 4 studies involving a total of 125 participants were selected. All participants had heart failure with the New York Heart Association (NYHA) classification ranging from 2.2 ± 0.4 to 3. The primary endpoints for assessment included the impact of SCS in HF-related symptoms, Left ventricular function, VO2 max, and NT-proBNP. All the studies could demonstrate safety and feasibility of SCS therapy, although the outcomes varied. Two studies reported improvement in NYHA classification, MLHFQ and QoL parameters after SCS. Concerning LVEF and VO2 max, only one study indicated positive changes. None of the studies found a significant change of NT-proBNP following SCS therapy. Given methodological variation, discrepancies in the results could be attributed to the diversity of the induction technique. Further studies are needed to develop a solid approach for employing SCS in human patients with HF.


Heart Failure , Spinal Cord Stimulation , Humans , Spinal Cord Stimulation/methods , Heart Failure/therapy , Heart Failure/physiopathology , Treatment Outcome
...