Skip to main content
medRxiv
  • Home
  • About
  • Submit
  • ALERTS / RSS
Advanced Search

Calpain-mediated sodium channel fragments in the monocentric SpasT-SCI-T trial: Biomarkers for central nervous system injuries and spasticity prediction

Guillaume Baucher, Sylvie Liabeuf, Cécile Brocard, Aurélie Ponz, View ORCID ProfileKarine Baumstarck, Lucas Troude, View ORCID ProfileMarc Leone, Pierre-Hugues Roche, View ORCID ProfileFrédéric Brocard
doi: https://doi.org/10.1101/2025.02.07.25321849
Guillaume Baucher
1Institut de Neurosciences de la Timone, Aix-Marseille Université, CNRS, Marseille, France
2AP-HM, Hôpital Universitaire Nord, Neurochirurgie adulte, Marseille, France
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • For correspondence: guillaume.baucher{at}ap-hm.fr frederic.brocard{at}univ-amu.fr
Sylvie Liabeuf
1Institut de Neurosciences de la Timone, Aix-Marseille Université, CNRS, Marseille, France
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Cécile Brocard
1Institut de Neurosciences de la Timone, Aix-Marseille Université, CNRS, Marseille, France
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Aurélie Ponz
3AP-HM, Direction de la Recherche Santé et Maladies Rares, Marseille, France
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Karine Baumstarck
3AP-HM, Direction de la Recherche Santé et Maladies Rares, Marseille, France
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • ORCID record for Karine Baumstarck
Lucas Troude
2AP-HM, Hôpital Universitaire Nord, Neurochirurgie adulte, Marseille, France
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Marc Leone
4AP-HM, Hôpital Universitaire Nord, Service d’Anesthésie et de Réanimation, Marseille, France
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • ORCID record for Marc Leone
Pierre-Hugues Roche
1Institut de Neurosciences de la Timone, Aix-Marseille Université, CNRS, Marseille, France
2AP-HM, Hôpital Universitaire Nord, Neurochirurgie adulte, Marseille, France
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Frédéric Brocard
1Institut de Neurosciences de la Timone, Aix-Marseille Université, CNRS, Marseille, France
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • ORCID record for Frédéric Brocard
  • For correspondence: guillaume.baucher{at}ap-hm.fr frederic.brocard{at}univ-amu.fr
  • Abstract
  • Full Text
  • Info/History
  • Metrics
  • Data/Code
  • Preview PDF
Loading

ABSTRACT

Spinal cord injury and traumatic brain injury are major causes of long-term disability and are often complicated by spasticity, a motor disorder characterized by increased muscle tone and exaggerated reflexes that significantly impair quality of life. Current diagnostic methods lack the sensitivity needed to accurately predict the severity of injury or the onset and progression of spasticity. Trauma-induced calcium dysregulation activates calpains, a family of proteases that cleave sodium channels, disrupting their inactivation and increasing persistent sodium currents. This cascade drives the overexcitability of motoneurons, contributing to the development of spasticity. Consequently, sodium channel fragments have emerged as promising biomarkers that link injury mechanisms to clinical outcomes.

The present SpasT-SCI-T clinical trial protocol aims to evaluate sodium channel fragments as blood biomarkers for assessing the severity of spinal cord and traumatic brain injuries, as well as their potential to predict clinical outcomes, including the development of spasticity. This prospective, multicenter, case-control and cohort study involves 40 participants: 20 individuals with spinal cord injury, 10 individuals with traumatic brain injury, and 10 healthy controls. Blood samples are collected within six hours of injury and at follow-up points over six months. Clinical outcomes, including spasticity (assessed using the Modified Ashworth Scale), neurological recovery (measured by the American Spinal Injury Association Impairment Scale and Glasgow Coma Scale), and quality of life (evaluated using the Short Form-36 Health Survey), are analyzed in correlation with biomarker levels.

We anticipate that calpain-mediated sodium channel fragments will transform the management of central nervous system injuries by enabling early diagnosis, improving prognostic accuracy, and guiding personalized therapeutic strategies. The clinical trial is registered on ClinicalTrials.gov (NCT06532760, January 10, 2024), with Assistance Publique–Hôpitaux de Marseille as the sponsor.

INTRODUCTION

Spinal cord injury (SCI) is a tragic condition with lifelong physical, psychological, and economic consequences, affecting millions of individuals worldwide [1–3]. Accurate and timely assessment of its severity is paramount for predicting outcomes and guiding therapeutic interventions. However, current diagnostic methods, which rely primarily on clinical examinations and imaging, lack the sensitivity required for precise evaluation, especially during the critical acute phase [4–6]. This diagnostic gap underscores the urgent need for novel, reliable biomarkers to improve clinical decision-making.

One of the most prevalent and disabling complications of SCI is spasticity, which affects up to 75% of patients within the first year post-injury [7,8]. Spasticity, classically defined as hypertonia with a velocity-dependent increase in stretch reflexes [9], encompasses a spectrum of clinical manifestations, including clonus, exaggerated reflexes, muscle spasms, and involuntary leg crossing [10,11]. These symptoms severely disrupt daily activities, quality of life, and rehabilitation efforts. Without early and effective management, spasticity progresses to complications such as muscle shortening, joint contractures, and painful pressure sores [12,13]. The unpredictable onset and variable severity of spasticity, ranging from mild stiffness to severe, uncontrollable spasms, pose significant challenges for clinicians, emphasizing the urgent need for reliable predictive biomarkers [14,15].

Advances in our understanding of spasticity pathophysiology have highlighted promising targets for biomarker discovery [16,17]. Secondary injury processes in SCI trigger molecular cascades, including excessive glutamate release, intracellular calcium overload, and calpain activation [18,19]. Calpains, calcium-activated proteases, cleaves Nav1.6 voltage-gated sodium channels [20–22], which are predominantly expressed in spinal motoneurons [23–25]. This proteolysis disrupts the normal inactivation of Nav1.6 channels, leading to an upregulationg of persistent sodium currents (INaP) [20–22]. The pathological INaP drives motoneuron hyperexcitability and sustained depolarizations (plateau potentials), directly contributing to spasticity in both animals [26–31] and humans [32–34].

The identification of sodium channel fragments as products of calpain-mediated cleavage offers a novel biomarker opportunity, linking the molecular pathology of SCI to clinical outcomes. These fragments hold potential not only for assessing SCI severity but also for predicting the onset and magnitude of spasticity [21]. Notably, similar sodium channel cleavage mechanisms have been observed in traumatic brain injury (TBI), suggesting broader applicability of these biomarkers across central nervous system (CNS) injuries [35–37].

The present SpasT-SCI-T clinical trial protocol aims to evaluate calpain-mediated sodium channel fragments as diagnostic biomarkers for SCI and TBI and as prognostic markers for spasticity. By bridging molecular mechanisms and clinical outcomes, this study seeks to advance the management of CNS injuries and spasticity, paving the way for more accurate diagnoses, enhanced prognostic precision, and personalized therapeutic strategies.

METHODS AND ANALYSIS

STUDY AIMS

The primary objective of the SpasT-SCI-T study is to detect and quantify sodium channel fragments in blood samples from patients with SCI or TBI. These fragments will be measured at multiple post-trauma time points, ranging from 6 hours to 6 months after injury.

The secondary objectives are threefold:

  1. To characterize in SCI and TBI patients the temporal kinetics of sodium channel fragments, providing insight into their dynamic expression and persistence over time.

  2. To evaluate the relation between plasma fragment levels and the onset, progression, and severity of spasticity in SCI patients.

  3. To compare plasma fragment levels in SCI and TBI patients with those of healthy controls, establishing baseline values and identifying injury-specific signatures.

STUDY DESIGN

The SpasT-SCI-T study is a prospective, multicenter observational trial performed in France. The study employs a combination of case-control and cohort designs to address its primary and secondary objectives. A total of 40 participants are recruited, divided into three groups:

  1. Group 1 (Healthy Controls): Ten healthy volunteers provide baseline blood samples for comparison.

  2. Group 2 (SCI Patients): Twenty patients with acute traumatic SCI confirmed by imaging and no associated TBI.

  3. Group 3 (TBI Patients): Ten patients with acute TBI confirmed by imaging and no associated SCI.

The trial consists of three phases:

  1. Inclusion Phase: Participants are enrolled into one of the three study groups based on predefined eligibility criteria.

  2. Acute Phase (0–14 days): This phase focuses on the immediate response to trauma, capturing rapid changes in sodium channel fragment levels during the critical early period. Blood samples are collected within 6 hours post-trauma and at multiple time points (Days 1, 3, 5, 7, and 14) to monitor early biomarker kinetics.

  3. Late Follow-Up Phase (3 and 6 months): The late follow-up phase assesses long-term changes in sodium channel fragment levels and their association with recovery trajectories and clinical outcomes, including the onset and severity of spasticity. Blood samples are collected during follow-up consultations at 3 and 6 months post-trauma.

The CONSORT diagram in Fig 1 provides details on the timeline for enrollment, interventions, and assessments across the participant groups, while the overall study design is summarized in Fig 2.

Fig 1
  • Download figure
  • Open in new tab
Fig 1 CONSORT flow diagram of the SpasT-SCI-T study.

(SCI: spinal cord injury; TBI: traumatic brain injury; MAS: modified Ashworth scale; ASIA: American Spinal Injury Association; GOS: Glasgow Outcome Scale; H: hour; D: Day; M: Month).

Fig 2
  • Download figure
  • Open in new tab
Fig 2 Graphical overview of SpasT-SCI-T study design.

Overview of the three study groups and their respective schedules for blood sample collections and clinical assessments. Group 1 (10 healthy subjects) provides a single baseline blood sample. Group 2 (20 SCI patients) and Group 3 (10 TBI patients) undergo sampling within 6 hours post-trauma, during the early follow-up phase (days 1, 3, 5, 7, and 14), and at late follow-ups (3 and 6 months). Clinical assessments include the American Spinal Injury Association (ASIA) impairment scale, Glasgow Coma Scale (GCS), Glasgow Outcome Scale (GOS), and Modified Ashworth Scale (MAS), depending on the group. These outcome measures are defined and detailed in the “Outcomes” section.

SITES AND RESSOURCES

The trial spans 30 months and is performed within the Assistance Publique– Hôpitaux de Marseille (AP-HM) network, leveraging multiple specialized facilities for recruitment, data collection, and biological sample processing:

  1. Neurosurgery Department, Hôpital Nord: Led by Professor Pierre-Hugues Roche, the Neurosurgery Department serves as the primary site for recruiting SCI and TBI patients. It specializes in acute neurological trauma management and advanced imaging (CT and MRI) to confirm eligibility. Follow-up visits at 3 and 6 months, including Modified Ashworth Scale (MAS) evaluations for spasticity in SCI patients, are coordinated here.

  2. Intensive Care Unit, Hôpital Nord: Headed by Professor Marc Leone, the center manages sever trauma patients during the acute injury phase and ensures the inclusion of patients unable to provide immediate consent. Baseline blood samples are collected within 6 hours post-trauma, with additional samples taken during early follow-up on Days 1, 3, 5, 7 and 14.

  3. Clinical Investigation Center, Hôpital Nord: Healthy controls are recruited and assessed at this Center. It provides a controlled environment for obtaining informed consent and baseline blood samples. Standardized protocols ensure consistency across participant groups.

  4. BioMINT Platform, INT UMR7289, Aix-Marseille University la Timone: Biological sample processing is centralized at the BioMINT platform, part of the Institute of Neurosciences (INT UMR7289). Plasma samples are analyzed using semi-automated Western blot techniques with Pan-Nav antibodies, ensuring high-quality, standardized biomarker data. Samples are stored at - 80°C until analysis and transported monthly from clinical sites.

  5. Rehabilitation Centers for Long-Term Follow-Up: Patients transitioning to affiliated rehabilitation centers within the AP-HM network participate in long-term follow-up. These centers collaborate with the Neurosurgery Department to ensure the collection of clinical data, including MAS evaluations, at 3 and 6 months post-trauma.

PARTICIPANTS

The selection of participants is based on strict inclusion and exclusion criteria (Table 1).

View this table:
  • View inline
  • View popup
  • Download powerpoint
Table 1 SCI - spinal cord injury; GCS: Glasgow Coma Scale; TBI: traumatic brain injury; ASIA: American Spinal Injury Association.

Inclusion Criteria

  1. Group 1 (Healthy Controls):

    • Adults (male or female) aged 18–75 years.

    • No history of central or peripheral nervous system trauma.

    • Absence of any chronic neurological, psychiatric, or systemic diseases.

    • Ability to provide informed consent prior to inclusion.

    • Affiliation with a social security system.

  2. Group 2 (SCI Patients):

    • Adults (male or female) aged 18–75 years

    • Confirmed acute SCI (C4-T12) based on imaging (CT and MRI).

    • Neurological assessment using the American Spinal Injury Association Impairment Scale (ASIA): grades A to D.

    • Inclusion must occur within 6 hours post-trauma.

    • No concurrent TBI.

    • Affiliation with a social security system.

  3. Group 3 (TBI Patients):

    • Adults (male or female) aged 18–75 years

    • Confirmed acute TBI based on CT imaging (Marshall Grade II or higher).

    • Neurological assessment using the Glasgow Coma Scale (GCS) ≤13.

    • Inclusion must occur within 6 hours post-trauma.

    • No concurrent SCI.

    • Affiliation with a social security system.

Exclusion Criteria (All Groups)

  • Pregnancy or breastfeeding.

  • Pre-existing neurological disorders, such as multiple sclerosis or Parkinson’s disease.

  • History of psychiatric illness or neurodegenerative conditions.

  • Concurrent severe trauma affecting study participation (e.g., multi-organ failure).

  • Inability to provide consent, and absence of a legally authorized representative.

  • Pre-existing cardiovascular or pulmonary diseases.

Informed consent (S3 Supplemental material) is obtained from all participants after verifying their eligibility according to the inclusion and exclusion criteria. Recognizing the urgency of inclusion and the acute medical care context, the consent process is tailored to accommodate the specific needs of SCI and TBI patients. The following procedures ensure compliance with ethical and legal requirements while prioritizing participant autonomy and rights:

  1. For conscious patients capable of decision-making, informed consent is obtained directly within the 6-hour inclusion window. The investigator or clinical team provides the patient with a detailed information sheet outlining the study’s objectives, procedures, potential risks and benefits. Patients are given the opportunity to ask questions and are encouraged to make an informed decision. Once all concerns are addressed, they sign the consent form, confirming their voluntary participation.

  2. For unconscious or incapacitated patients, a legally authorized representative (e.g., a close relative) is approached to provide proxy consent. The representative is provided with the same detailed information about the study, and their consent is carefully documented. Once the patient regains the capacity to decide, they are fully informed about their participation, following the same process as for conscious patients. This includes providing them with the study information sheet, explaining the objectives, procedures, risks, and potential benefits, and addressing any questions they may have. Written continuation consent is then requested to confirm their voluntary agreement to remain in the study. These procedures comply with Article L1122-1-3 of the French Public Health Code, ensuring adherence to legal and ethical standards for proxy and emergency consent.

  3. Emergency Consent: In cases where the patient is unconscious and no proxy is available within the inclusion timeframe (6 hours post-trauma), a physician not involved in the study may authorize emergency consent. This process allows for the patient’s inclusion while ensuring that: 1) Consent is obtained as soon as possible from the patient or their representative once circumstances allow. Once the patient regains capacity or a relative is available, they are fully informed of their inclusion and written consent is requested, in compliance with Article L1122-1-3 of the French Public Health Code.

  4. For healthy volunteers, written informed consent is obtained prior to any study-related procedures. The consent process ensures they understand the study’s objectives, any potential risks, and their right to withdraw at any time without consequences.

All participants, regardless of group, continue to receive standard medical care, including routine imaging, rehabilitation, and clinical follow-ups, without interference from study procedures. Withdrawal from the study, whether initiated by the participant or deemed necessary for medical reasons, does not impact their access to or quality of standard medical care.

INTERVENTION PROCEDURES

The SpasT-SCI-T trial outlines specific intervention procedures for each participant group. These interventions encompass blood sampling, imaging, and clinical assessments aimed at investigating sodium channel fragments as potential biomarkers and correlating their dynamics with clinical outcomes. All procedures are rigorously standardized to ensure reproducibility and protocol adherence.

Group 1: Healthy Subjects

Healthy participants are selected from a pre-screened volunteer registry. Upon enrollment, they are invited to the Clinical Investigation Center at Hôpital Nord at their convenience. During the inclusion visit a single venous blood sample of 4 mL is collected in BP100 tubes containing protease inhibitors to prevent biomarker degradation. The sample is processed within 2 hours of collection by centrifugation at 1500g to extract plasma, which is then stored at -80°C until analysis at the BioMINT platform. No follow-up visits or additional interventions are required for this group, ensuring minimal participant burden.

Group 2: Spinal Cord Injury (SCI) Patients

SCI patients are prospectively recruited from the Intensive Care Unit or Neurosurgical Department at Hôpital Nord within 6 hours post-trauma. Upon admission, interventions for these patients begin with clinical and imaging assessments. A computed tomography (CT) scan is performed on admission to confirm vertebral fractures or spinal column instability. Magnetic resonance imaging (MRI) provides detailed characterization of the SCI, documenting patterns such as contusion, compression, hemorrhage, or edema. Injury severity is further classified using the AOSpine fracture classification [38] and BASIC scoring system [39,40].

Baseline neurological status is documented using the American Spinal Injury Association (ASIA) Impairment Scale, which evaluates the severity and level of the SCI [38,41]. Additional documentation includes respiratory and hemodynamic stability, renal clearance, liver function, and hemoglobin levels.

Blood sampling begins with a 4 mL venous sample collected within the 6 hours of trauma. Additional samples are collected during the acute phase on days 1, 3, 5, 7 and 14 to monitor early biomarker kinetics. Further samples are obtained at 3 months (M3) and 6 months (M6) post-trauma, offering insights into long-term biomarker dynamics. As part of the follow-up interventions, at M3 and M6, spasticity is assessed using the Modified Ashworth Scale (MAS, table 2), alongside detailed evaluations of neurological recovery and residual deficits. These assessments are complemented by documentation of neurological recovery and residual deficits, enabling correlations between clinical outcomes and sodium channel fragment levels.

View this table:
  • View inline
  • View popup
  • Download powerpoint
Table 2 Modified Ashworth scale for spasticity assessment

[43].

Group 3: Traumatic Brain Injury (TBI) Patients

Interventions for TBI patients are tailored to intracranial trauma and are similar in structure to those for SCI patients, with modifications to address brain injury specifics. A whole-body CT scan is performed upon admission to confirm intracranial hemorrhage and exclude SCI, ensuring group specificity. Neurological status is assessed using the Glasgow Coma Scale (GCS) to document the severity of brain injury [42]. Trauma-related data, including respiratory and hemodynamic stability, renal clearance, liver function, and hemoglobin levels, are recorded. Blood sampling begins with a 4 mL venous sample collected within the 6 hours of trauma during the routine biological workup. Additional samples are obtained during the acute phase at Days 1, 3, 5, 7 and 14 to capture early biomarker dynamics. Further samples are collected during late follow-up visits at M3 and M6 to evaluate the longer-term biomarker behavior. General neurological assessments are performed at M3 and M6 to evaluate recovery. These outcomes are correlated with sodium channel fragment levels to explore potential biomarker-based prognostication.

For SCI and TBI patients (Groups 2 and 3), detailed records of all medical treatments administered during hospitalization will be maintained. This includes, but is not limited to: anticoagulation therapy, antibiotic regimens, anti-spastic medications, and pain management protocols. This documentation allows for the evaluation of potential confounding factors and ensures that biomarker changes can be interpreted in the context of treatment.

OUTCOMES

The primary outcome of the study is the detection and quantification of sodium channel fragments in blood samples. These fragments are quantified using a semi-automated Western blot technique employing the commercial Pan-Nav antibody, which is validated for its high sensitivity in detecting α-subunit cleavage products generated by calpain-mediated proteolysis. Sodium channel fragment levels are compared across SCI, TBI and healthy participants to identify injury-specific biomarker signatures. These data aim to provide valuable insights into the molecular mechanisms underlying SCI and TBI.

The secondary outcomes include:

  1. Temporal Dynamics of Sodium Channel Fragments: The temporal kinetics of sodium channel fragments are analyzed to characterize their dynamic expression and persistence during the acute and chronic phases of injury. Longitudinal changes in fragment levels are assessed from the baseline (acute phase) through follow-up visits at 3 months (M3) and 6 months (M6). These data are related with key clinical and functional outcomes to evaluate the role of sodium channel fragments as potential prognostic biomarkers. For SCI patients, changes in neurological function are monitored using the American Spinal Injury Association (ASIA) Impairment Scale, which provides detailed assessments of sensory and motor function recovery. For TBI patients, functional recovery is evaluated using the Glasgow Outcome Scale (GOS) and the more granular Glasgow Outcome Scale-Extended (GOSE), which capture subtle variations in recovery trajectories. For both groups, quality of life is assessed using the Short Form Health Survey (SF-36), a comprehensive tool that evaluates health-related quality of life across physical, mental, and social dimensions. These correlations aim to establish the predictive value of sodium channel fragment levels for neurological recovery, functional outcomes, and overall quality of life, providing a deeper understanding of the molecular mechanisms underlying injury progression and recovery.

  2. Spasticity and Biomarker Levels in SCI Patients: The relationship between sodium channel fragment levels and spasticity is specifically investigated in SCI patients. Spasticity is assessed during follow-up visits at M3 and M6 using the Modified Ashworth Scale (MAS), a widely used tool for grading muscle tone. The relationship between MAS scores and fragment levels is evaluated using correlation analyses. This correlation is expected to provide evidence of the prognostic utility of sodium channel fragments in predicting the onset and severity of spasticity. These analyses aim to establish the prognostic utility of sodium channel fragments in predicting the onset, progression, and severity of spasticity, providing a molecular link between injury mechanisms and clinical outcomes. Such insights may inform personalized therapeutic strategies for managing spasticity in SCI patients.

  3. Injury Severity and Biomarker Associations: The study investigates the association between sodium channel fragment levels and injury severity in both SCI and TBI patients. For SCI patients, neurological severity is assessed using the ASIA Impairment Scale, which evaluates the level and completeness of spinal cord injury through sensory and motor function tests. For TBI patients, the severity of brain injury is determined using the GCS, which provides a standardized assessment of consciousness based on eye, verbal, and motor responses. By correlating sodium channel fragment levels with these established severity metrics, the study aims to elucidate the relationship between molecular biomarkers and clinical injury severity, enhancing the understanding of the pathophysiology of CNS trauma and informing future diagnostic approaches.

Biomarker Levels

Sodium channel fragment levels are quantified using the Jess system (ProteinSimple, Bio-techne), a capillary-based automated Western blot platform. Plasma samples are resuspended in 0.1X Sample buffer (SimpleWestern, Bio-techne). Protein concentrations are determined using the DC Protein Assay (Biorad) to ensure consistent loading across all samples. Samples are prepared without reducing agents or heat denaturation to preserve protein integrity. Prepared plasma samples (3 µL) are loaded into the Jess assay plate along with controls, standards, and molecular weight ladders. Proteins are loaded into the capillary automatically (12–230 kDa range), electrophoresed and separated by size. Antibodies against the α-subunit of sodium channels, and chemiluminescence are used for signal detection. Quantitative data are captured by automated software, and protein band intensities are normalized to either a housekeeping protein or total protein for variability control. The native sodium channel (∼250 kDa) and its lower molecular weight fragments are identified and quantified based on band intensity, facilitating the comparison of fragment expression across groups.

Spasticity Evaluations

Spasticity is assessed at M3 and M6 follow-up visits using the Modified Ashworth Scale (MAS), which grades muscle tone on a scale from 0 (no increase in muscle tone) to 4 (rigid muscle tone) [43]. This score measures the resistance perceived by the rater when passively moving a joint through its full range of movement. All evaluations are performed by a single, trained physiotherapist to minimize inter-observer variability. Associated phenomena, such as clonus and spasms, are also recorded to provide a comprehensive spasticity profile. Factors influencing variability (e.g., patient positioning, medication, and pain) are meticulously documented.

Neurological Evaluations (GCS, ASIA)

The Glasgow Coma Scale (GCS) is a widely used, standardized tool for assessing consciousness in patients with TBI. It evaluates three key components: eye-opening, verbal, and motor responses, providing a rapid and reliable measure of neurological status. The Eye-Opening Response is scored from 4 to 1: 4 for spontaneous eye opening, 3 for a response to verbal stimuli, 2 for a response to painful stimuli, and 1 if there is no response. The Verbal Response evaluates the orientation and ability to communicate, scoring 5 for coherent and oriented speech, 4 for confusion, 3 for inappropriate words, 2 for incomprehensible sounds, and 1 for no vocalization. The Motor Response measures movement in response to commands or stimuli, scoring 6 for obeying commands, 5 for localizing pain, 4 for withdrawing from pain, 3 for abnormal flexion (decorticate posturing), 2 for abnormal extension (decerebrate posturing), and 1 for no response. The total GCS score ranges from 3 (deep coma) to 15 (full consciousness), allowing clinicians to classify the severity of brain injury and monitor changes in neurological status over time.

The American Spinal Injury Association (ASIA) Impairment Scale is a standardized framework for evaluating and classifying the severity of SCI [38,41]. It assesses sensory and motor functions across key dermatomes and myotomes, as well as sacral sparing, to determine the neurological level and completeness of the injury. Sensory evaluation tests light touch and pinprick sensations across 28 dermatomes bilaterally, from C2 to S4/S5. A cotton swab is used for light touch, and a safety pin for pinprick testing, with scores assigned as 2 for normal sensation, 1 for impaired sensation, and 0 for absent sensation. Special attention is given to the S4/S5 dermatome, as sacral sparing is critical for determining whether the injury is complete or incomplete. Motor function is evaluated by testing strength in 10 muscle groups corresponding to specific myotomes, ranging from elbow flexion (C5) to ankle plantar flexion (S1), scored from 5 (normal strength) to 0 (no contraction). Sacral motor function is further assessed by voluntary contraction of the external anal sphincter. The ASIA Impairment Scale assigns grades based on the completeness of the injury: Grade A indicates complete injury with no sensory or motor function preserved in sacral segments, Grades B-D reflect varying degrees of incomplete injuries with some preserved sensory or motor function below the injury level, and Grade E signifies normal sensory and motor function. The neurological level of injury is defined as the lowest spinal segment with intact sensory and motor function bilaterally.

Functional Scales (GOS, GOSE, SF-36)

The Glasgow Outcome Scale (GOS) is a widely recognized tool for evaluating functional recovery following TBI [44]. It categorizes outcomes into five levels: death, vegetative state, severe disability, moderate disability, and good recovery. The GOS provides a high-level summary of a patient’s ability to reintegrate into daily life post-injury. To enhance its sensitivity, the Glasgow Outcome Scale-Extended (GOSE) subdivides each GOS category into upper and lower levels, offering a more nuanced evaluation of recovery trajectories. This refined scale is particularly beneficial for detecting subtle differences in functional outcomes, making it a valuable tool in clinical trials. Both the GOS and GOSE are exclusively applied to TBI patients in this study to monitor recovery.

The Short Form Health Survey (SF-36) is a generic quality-of-life questionnaire used for both SCI and TBI patients. It evaluates health-related quality of life (HRQoL) across eight domains, including physical functioning, pain, general health, vitality, social functioning, and mental health [45]. The SF-36 captures the broader impact of injuries and treatments on physical, emotional, and social well-being. Administered during follow-up visits, it complements the GOS and GOSE for TBI patients by offering additional insights into their quality of life. For SCI patients, it serves as a critical tool for assessing long-term functional impairments and evaluating the effectiveness of rehabilitation strategies.

Pain Assessments (VAS, DN4)

Pain assessments are critical for evaluating quality of life and rehabilitation in SCI and TBI patients. The Visual Analogue Scale (VAS) is a simple yet effective tool for quantifying pain intensity. Patients rate their pain on a 10-centimeter line, with anchors at “no pain” (0 cm) and “worst imaginable pain” (10 cm). This approach enables longitudinal tracking of pain severity, offering valuable insights into the effectiveness of therapeutic interventions over time. The DN4 questionnaire (Douleur Neuropathique 4 Questions) is specifically designed to identify neuropathic pain, which is prevalent among SCI patients [46]. It comprises 10 items: 7 symptom-related questions (e.g., burning, tingling, electric shocks) and 3 clinical examination items (e.g., hypoesthesia, allodynia). A score of ≥4 indicates the presence of neuropathic pain. Together, the VAS and DN4 provide a comprehensive evaluation of pain intensity and type, enabling targeted therapeutic strategies and effective monitoring of patient progress.

Sphincter Functions

Sphincter function is a crucial outcome in SCI patients, as SCI often compromise bladder and bowel control, significantly affecting quality of life. Assessments include bladder management (permanent or intermittent catheterization, normal urination) and bowel function (fecal incontinence, constipation), focusing on their impact on quality of life. The Table 3 summarizes the main data collected from the patients in the case report forms, from their inclusion in the study to the 6-month follow-up consultation.

View this table:
  • View inline
  • View popup
Table 3

Main data collected from the patients in the case report forms, from their inclusion in the study to the 6-month follow-up consultation (ASIA: American Spinal Injury Association; GCS: Glasgow Coma Scale; BASIC: Brain and Spinal Injury Center[40]; AOSpine[38]; AIS: Abbreviated Injury Score; ISS: Injury Severity Score[47]; SAPS II: Simplified Acute Physiology Score II[48]; SOFA: Sepsis related Organ Faillure Assessment[49]; Kdigo[50]; modified Ashworth scale[43]; DN4[46]; GOS: Glasgow Outcome Scale; GOSE: Glasgow Outcome Scale Extended[44]; SF36: Short Form 36 Health Survey[45]).

JUSTIFICATION OF SAMPLE SIZE

The study includes a total of 40 participants distributed as follows: 20 patients with spinal cord injury (SCI), 10 patients with traumatic brain injury (TBI), and 10 healthy controls. The selection of 20 SCI patients is based on the annual admission rates for SCI at the study institution, ensuring a representative sample of the population served. The inclusion of 10 TBI patients is justified to account for potential confounding factors arising from the overlapping but distinct injury mechanisms of SCI and TBI. Furthermore, 10 healthy controls are included to provide a robust baseline for sodium channel fragment levels, facilitating a balanced comparison across the three groups.

This sample size is sufficient to detect meaningful trends and associations between sodium channel fragments and clinical outcomes, including spasticity, while maintaining feasibility within the scope of a pilot study. The findings will generate preliminary evidence regarding the diagnostic and prognostic utility of sodium channel fragments in neurological injuries and will guide the design of larger-scale clinical trials.

Given the exploratory nature of this study, no formal interim analyses are planned. As the trial involves minimal-risk procedures, such as blood sampling, no pre-specified stopping rules have been established. In the event of significant deviations from the protocol or unforeseen safety concerns, the trial steering committee will consult the Ethics Committee to determine whether adjustments or study termination are necessary. All such decisions will be transparently documented and communicated to relevant stakeholders, including regulatory authorities, to ensure compliance with ethical standards.

RANDOMISATION, BLINDING

The SpasT-SCI-T trial is an observational study employing a case-control and cohort design; therefore, randomization is not applicable. Participants are allocated to groups based on predefined eligibility criteria rather than random assignment. Consequently, sequence generation, allocation concealment, and implementation of randomization do not apply to this trial. To mitigate bias and maintain data integrity, laboratory personnel conducting biochemical analyses remain blinded to group allocation. This blinding ensures that sodium channel fragment quantification is performed without preconceived assumptions about the samples, enhancing the reliability and validity of the results.

STATISTICAL METHODS

The primary objective of this study is to quantify sodium channel fragments in blood samples and compare their levels across the three participant groups: healthy controls (Group 1), SCI patients (Group 2) and TBI patients (Group 3). To address this objective, statistical analyses will begin with group comparisons.

  1. Group Comparisons: For normally distributed data, one-way analysis of variance (ANOVA) will be employed to identify differences in sodium channel fragment levels among the three groups. If data are not normally distributed, the Kruskal-Wallis test will be applied as a non-parametric alternative. Post-hoc analyses will follow these tests to assess pairwise comparisons, using Tukey’s test for ANOVA or the Mann-Whitney U test for non-parametric data.

  2. Effect Size Calculation: Effect sizes (e.g. Cohen’s d for t-tests or η² for ANOVA) will be calculated to quantify the magnitude of differences between groups.

For secondary outcomes, analyses will focus on the temporal kinetics of sodium channel fragments and their correlation with clinical outcomes.

  1. Temporal Kinetics: Changes in fragment levels over time, measured at baseline (6 hours), during the acute phase (Days 1, 3, 5, 7), and at follow-up (Months 3 and 6), will be analyzed using repeated-measures ANOVA for normally distributed data. For non-normally distributed data, the Friedman test will be used. Bonferroni corrections will be applied during multiple comparisons.

  2. Spasticity Correlation: The relationship between sodium channel fragment levels and spasticity severity in SCI patients will be assessed using Spearman’s rank correlation coefficients, with spasticity evaluated at Months 3 and 6 using the Modified Ashworth Scale (MAS).

  3. Subgroup Analysis (SCI Severity): Subgroup analyses will investigate whether sodium channel fragment levels differ by injury severity. In SCI patients, severity will be categorized based on the American Spinal Injury Association (ASIA) Impairment Scale grades (A–D). Similarly, spasticity onset will be analyzed as a binary outcome (presence vs. absence). For TBI patients, the Glasgow Coma Scale (GCS) will be used to classify injury severity, enabling comparisons of sodium channel fragment levels across defined severity subgroups.

Additional statistical methods will include multivariable linear regression models to adjust for potential confounders, such as age, sex, and comorbidities, ensuring that observed associations are robust and not influenced by baseline characteristics. Receiver operating characteristic (ROC) curves will be generated to identify threshold levels of sodium channel fragments that differentiate SCI from TBI or predict spasticity onset. To address missing data, multiple imputation methods will be applied, minimizing bias and preserving the validity of the analyses.

Continuous variables will be expressed as means with standard deviations for normally distributed data or medians with interquartile ranges for non-normally distributed data. Categorical variables will be summarized as frequencies and percentages. For all primary and secondary analyses, a two-tailed p-value < 0.05 will be considered statistically significant. Patients who die before the 3-month follow-up will be excluded from spasticity-related analyses due to insufficient time for spasticity development, but all data collected up to the time of death will be included in secondary analyses, such as demographic characterization and early biomarker levels.

ETHICAL APPROVAL AND DISSEMINATION POLICY

RESEARCH ETHICS APPROVAL

The SpasT-SCI-T clinical trial adheres to the ethical principles outlined in the Declaration of Helsinki [63] and complies with the French Public Health Code. The study received the approval from the local ethical committee (CPP number 2024-A00265-42) on October 7, 2024, and the authorization of the National Agency for Medicines and Health Products in France on July 10, 2017. The trial is registered in the French National Clinical Portal and listed on Clinicaltrials.gov under registration n°NCT06532760. All research procedures have been designed to safeguard participant safety, rights, and confidentiality. Under French law, the trial is classified as a category 2 interventional study, denoting minimal risk and constraints for participants. Reporting will adhere to the Standard Protocol Items Recommendations for Interventional Trials (SPIRIT) guidelines [64], and both SPIRIT and CONSORT checklists are available as supplementary materials (see S1 and S2 Supplemental Materials, respectively).

PROTOCOL AMENDMENTS

Any modifications to the trial protocol, including changes to eligibility criteria, outcome measures, or analytical methods, will be promptly communicated to all relevant stakeholders. These include the investigators, the Ethics Committee (CPP), regulatory authorities such as the ANSM (Agence Nationale de Sécurité du Médicament), and trial participants, when applicable. Updates will also be reflected in trial registries (e.g., ClinicalTrials.gov) and disseminated through publications and conference presentations as needed. All amendments will undergo prior review and approval by the CPP to ensure continued ethical compliance and transparency.

DATA CONFIDENTIALITY AND MANAGEMENT

Participant confidentiality is prioritized throughout the trial. All data are pseudonymized, and unique patient identifiers are used for sample tracking and clinical data management. Personal identifying information will not be associated with biological samples or research databases. Data will be securely stored in a password-protected electronic Case Report Form (eCRF) developed using REDCap, hosted on encrypted servers at Assistance Publique–Hôpitaux de Marseille (AP-HM). Access to the eCRF is restricted to authorized personnel, including the principal investigators and the designated data management team, through encrypted login credentials. Data integrity is ensured through daily backups, and all procedures comply with the European General Data Protection Regulation (GDPR). At the conclusion of the trial, all biological samples will be destroyed, and the pseudonymized data will be archived securely for 15 years, as mandated by French regulations.

DECLARATION OF INTERESTS

The trial sponsor, Assistance Publique–Hôpitaux de Marseille (AP-HM), has no financial or commercial interests in this study. All investigators declare the absence of conflicts of interest that could influence the conduct or reporting of the trial. The study is funded through institutional grants and public research funding bodies, with no involvement from private-sector entities.

ANCILLARY AND POST-TRIAL CARE

As the trial involves minimal-risk procedures, participants are not expected to require ancillary or post-trial care related to their involvement. In the event of harm directly resulting from study participation, liability insurance provided by the sponsor ensures coverage. Participants will retain access to their treating physicians for medical concerns during and after the trial.

DISSEMINATION PLAN

The results of the SpasT-SCI-T clinical trial will be disseminated through peer-reviewed journals, conference presentations, and publicly accessible databases. The findings will adhere to CONSORT guidelines to ensure transparency and rigor in reporting. Participants who express interest in the study’s results will be provided with an aggregated summary by the investigators, in compliance with French regulatory requirements. To maintain confidentiality, no individual results will be disclosed. Authorship of publications will follow the guidelines of the International Committee of Medical Journal Editors (ICMJE), ensuring that all contributors to the study design, conduct, analysis, and reporting are appropriately credited.

BASELINE DATA

Baseline data were systematically collected to characterize the study population and ensure comparability across the three participant groups (healthy controls, SCI patients, and TBI patients). Key baseline variables are presented in a detailed summary table to promote transparency and support balanced comparisons between groups.

DISCUSSION

The CNS injury initiates a cascade of molecular events, culminating in the proteolysis of Nav1.6 sodium channels, generating fragments that emerge as accessible biomarkers in blood vessels (Fig 3). The clinical trial builds upon prior evidence that calpain activity is positively correlated with the severity of spasticity [21], leading us to hypothesize that breakdown products of calpain substrates may serve as reliable biomarkers for predicting functional outcomes in SCI and TBI. This aligns with the growing recognition of biomarkers as invaluable tools in CNS injury management, enhancing our understanding of disease progression, clinical decision-making, treatment strategies, and overall patient care [14,15,51].

Fig 3
  • Download figure
  • Open in new tab
Fig 3 Calpain activation following spinal cord injury.

Traumatic injury to the spinal cord triggers glutamate release into the extracellular space, causing a sustained increase in intracellular calcium concentrations in motoneurons. This calcium influx activates calpain, a calcium-dependent protease, which cleaves Nav1.6 voltage-gated sodium channels. The cleavage results in an upregulation of persistent sodium currents (INaP), contributing to motoneuron hyperexcitability and the development of spasticity. Additionally, fragments of cleaved sodium channels accumulate in the sublesional spinal cord and can be detected in the bloodstream. These fragments represent a promising biomarker for assessing SCI severity and predicting spasticity outcomes.

Previous studies have identified fragments of calpain-mediated α-II spectrin cleavage as potential indicators of calpain activity following SCI [51]. However, several limitations diminish their utility. First, α-II spectrin is also a substrate for caspase-3, activated during apoptosis after CNS trauma, confounding its specificity for calpain activity [52]. Second, spectrin breakdown products are transiently expressed, limiting their utility to the acute phase and precluding their role in monitoring spasticity progression or therapeutic efficacy during the chronic phase [15]. Third, the high expression of spectrin in red blood cells raises concerns about its diagnostic accuracy in polytraumatized patients [51]. These challenges highlight the critical need for novel biomarkers that are both detectable shortly after injury and persist for months, facilitating long-term monitoring. Our preliminary data, combined with evidence that Nav channels are exclusively expressed in the nervous system and are resistant to caspase-3 cleavage [37], suggest that calpain-mediated breakdown products from Nav channels may fulfill this need. These fragments represent a promising biomarker for both CNS trauma and spasticity, providing specificity, sensitivity, and temporal versatility.

The implications of this research for SCI and TBI management are significant. Current diagnostic methods, which rely heavily on clinical assessments and imaging, often lack the sensitivity to detect molecular changes or predict long-term outcomes and secondary complications. Sodium channel fragments could provide real-time insights into injury dynamics, allowing for the precise monitoring of therapeutic efficacy and the progression of secondary complications like spasticity. This is particularly critical for spasticity, which affects up to 75% of SCI patients and significantly impairs quality of life and rehabilitation outcomes [7,8]. By identifying patients at risk of developing spasticity early, clinicians could implement targeted interventions during the acute phase, potentially reducing its severity and improving recovery trajectories. The inclusion of TBI patients in this study underscores the potential broad applicability of these biomarkers across various CNS injuries. The detection of sodium channel fragments in both SCI and TBI highlights a shared pathological mechanism, underscoring their relevance as universal biomarkers for neural trauma.

Despite their promise, several challenges must be addressed before sodium channel fragments can be fully integrated into clinical practice. Factors such as patient demographics and comorbidities may influence fragment levels, necessitating extensive studies to establish normative data and clinical thresholds. Additionally, blood sample processing protocols must be standardized to ensure reproducibility and accuracy. Lastly, integrating biomarker analysis into existing clinical workflows requires careful planning, infrastructure adaptation, and validation. Future research should focus on integrating these biomarkers with advanced imaging techniques to enhance diagnostic precision and prognostic modeling. A multimodal approach combining molecular biomarkers and imaging could offer a more comprehensive assessment of injury severity, therapeutic response, and recovery trajectories.

CONCLUSIONS

The SpasT-SCI-T trial marks a pivotal step toward bridging molecular mechanisms and clinical outcomes in SCI and TBI. By validating sodium channel fragments as reliable biomarkers, this study has the potential to modernize CNS trauma management, enabling personalized therapeutic strategies and improving long-term recovery prospects. Although challenges remain, the findings from this trial could herald a new era of biomarker-driven approaches in neurology, advancing precision medicine in the management of CNS disorders.

Data Availability

All relevant data are within the manuscript and its Supporting Information files.

SUPPORTING INFORMATION

S1 Checklist. The SPIRIT 2013 checklist.

S2 Checklist. CONSORT 2010 checklist of information to include when reporting a randomized trial.

S3 Model consent form and other related documents given to participants.

S4 Trial study protocol (English version)

S5 Trial study protocol (French version).

OTHERS INFORMATION

Registration

The SpasT-SCI-T clinical trial has been registered on the ClinicalTrials.gov database under the identifier NCT06532760.

Protocol Availability

The full trial protocol for the SpasT-SCI-T study is publicly available on ClinicalTrials.gov under the identifier NCT06532760. The full trial protocol, including all details regarding the study design, objectives, methodology, and ethical considerations are described in the present paper.

Funding and Support

This study was supported by the French National Research Agency (ANR) under grant number ANR-21-CE17-0060, awarded to FB.

AUTHOR CONTRIBUTIONS

Conceptualization

Frédéric Brocard, Pierre-Hugues Roche, Marc Léone

Funding acquisition

Frédéric Brocard

Investigation

Frédéric Brocard, Cécile Brocard, Sylvie Liabeuf , Pierre-Hugues Roche, Marc Léone, Guillaume Baucher, Lucas Troude

Methodology

Frédéric Brocard, Cécile Brocard, Sylvie Liabeuf, Aurélie Ponz, Karine Baumstarck, Pierre-Hugues Roche, Guillaume Baucher

Project administration

Aurélie Ponz

Validation

Frédéric Brocard, Pierre-Hugues Roche, Marc Léone, Aurélie Ponz, Karine Baumstarck

Writing – original draft

Guillaume Baucher, Frédéric Brocard

Writing – reviewing & editing

Frédéric Brocard, Pierre-Hugues Roche, Marc Léone, Karine Baumstarck

ACKNOWLEDGMENTS

We sincerely thank Tiphaine Villaume for her invaluable help in reviewing, correcting, and providing insightful advice on the drafting of the Ethics Committee submission.

REFERENCES

  1. 1.↵
    Svensson J, Borg S, Nilsson P. Costs and quality of life in multiple sclerosis patients with spasticity. Acta Neurol Scand. 2014;129: 13–20. doi:10.1111/ane.12139
    OpenUrlCrossRefPubMed
  2. 2.
    Ding W, Hu S, Wang P, Kang H, Peng R, Dong Y, et al. Spinal Cord Injury: The Global Incidence, Prevalence, and Disability From the Global Burden of Disease Study 2019. Spine. 2022;47: 1532–1540. doi:10.1097/BRS.0000000000004417
    OpenUrlCrossRef
  3. 3.↵
    Khazaeipour Z, Taheri-Otaghsara S-M, Naghdi M. Depression Following Spinal Cord Injury: Its Relationship to Demographic and Socioeconomic Indicators. Topics in Spinal Cord Injury Rehabilitation. 2015;21: 149–155. doi:10.1310/sci2102-149
    OpenUrlCrossRefPubMed
  4. 4.↵
    Ellingson BM, Salamon N, Holly LT. Imaging Techniques in Spinal Cord Injury. World Neurosurgery. 2014;82: 1351–1358. doi:10.1016/j.wneu.2012.12.004
    OpenUrlCrossRefPubMed
  5. 5.
    van Middendorp JJ, Goss B, Urquhart S, Atresh S, Williams RP, Schuetz M. Diagnosis and prognosis of traumatic spinal cord injury. Global Spine J. 2011;1: 1–8. doi:10.1055/s-0031-1296049
    OpenUrlCrossRefPubMed
  6. 6.↵
    Krishna V, Andrews H, Varma A, Mintzer J, Kindy MS, Guest J. Spinal cord injury: how can we improve the classification and quantification of its severity and prognosis? J Neurotrauma. 2014;31: 215–227. doi:10.1089/neu.2013.2982
    OpenUrlCrossRefPubMed
  7. 7.↵
    Adams MM, Hicks AL. Spasticity after spinal cord injury. Spinal Cord. 2005;43: 577–586. doi:10.1038/sj.sc.3101757
    OpenUrlCrossRefPubMedWeb of Science
  8. 8.↵
    Biering-Sørensen F, Nielsen JB, Klinge K. Spasticity-assessment: a review. Spinal Cord. 2006;44: 708–722. doi:10.1038/sj.sc.3101928
    OpenUrlCrossRefPubMedWeb of Science
  9. 9.↵
    Lance JW. The control of muscle tone, reflexes, and movement: Robert Wartenbeg Lecture. Neurology. 1980;30: 1303–1303. doi:10.1212/WNL.30.12.1303
    OpenUrlCrossRefPubMed
  10. 10.↵
    Sheean G, McGuire JR. Spastic Hypertonia and Movement Disorders: Pathophysiology, Clinical Presentation, and Quantification. PM&R. 2009;1: 827–833. doi:10.1016/j.pmrj.2009.08.002
    OpenUrlCrossRefPubMedWeb of Science
  11. 11.↵
    Pandyan A, Gregoric M, Barnes M, Wood D, Wijck FV, Burridge J, et al. Spasticity: Clinical perceptions, neurological realities and meaningful measurement. Disability and Rehabilitation. 2005;27: 2–6. doi:10.1080/09638280400014576
    OpenUrlCrossRefPubMedWeb of Science
  12. 12.↵
    Vural M, Yalcinkaya EY, Celik EC, Gunduz B, Bozan A, Erhan B. Assessment of quality of life in relation to spasticity severity and socio-demographic and clinical factors among patients with spinal cord injury. The Journal of Spinal Cord Medicine. 2020;43: 193–200. doi:10.1080/10790268.2018.1543093
    OpenUrlCrossRefPubMed
  13. 13.↵
    Field-Fote EC, Furbish CL, Tripp NE, Zanca JM, Dyson-Hudson T, Kirshblum S, et al. Characterizing the Experience of Spasticity after Spinal Cord Injury: A National Survey Project of the Spinal Cord Injury Model Systems Centers. Archives of Physical Medicine and Rehabilitation. 2022;103: 764–772.e2. doi:10.1016/j.apmr.2021.03.040
    OpenUrlCrossRefPubMed
  14. 14.↵
    De Mello Rieder M, Oses JP, Kutchak FM, Sartor M, Cecchini A, Rodolphi MS, et al. Serum Biomarkers and Clinical Outcomes in Traumatic Spinal Cord Injury: Prospective Cohort Study. World Neurosurgery. 2019;122: e1028–e1036. doi:10.1016/j.wneu.2018.10.206
    OpenUrlCrossRefPubMed
  15. 15.↵
    Rodrigues LF, Moura-Neto V, E Spohr TCLDS. Biomarkers in Spinal Cord Injury: from Prognosis to Treatment. Mol Neurobiol. 2018;55: 6436–6448. doi:10.1007/s12035-017-0858-y
    OpenUrlCrossRefPubMed
  16. 16.↵
    Nielsen JB, Crone C, Hultborn H. The spinal pathophysiology of spasticity – from a basic science point of view. Acta Physiologica. 2007;189: 171–180. doi:10.1111/j.1748-1716.2006.01652.x
    OpenUrlCrossRefPubMedWeb of Science
  17. 17.↵
    Sheean G. The pathophysiology of spasticity. Euro J of Neurology. 2002;9: 3–9. doi:10.1046/j.1468-1331.2002.0090s1003.x
    OpenUrlCrossRefPubMedWeb of Science
  18. 18.↵
    Dingu N, Bras H, Brocard F. Calpain role in the pathophysiology of spasticity after spinal cord injury. Cellular, Molecular, Physiological, and Behavioral Aspects of Spinal Cord Injury. Elsevier; 2022. pp. 249–261. doi:10.1016/B978-0-12-822427-4.00021-6
    OpenUrlCrossRef
  19. 19.↵
    Alizadeh A, Dyck SM, Karimi-Abdolrezaee S. Traumatic Spinal Cord Injury: An Overview of Pathophysiology, Models and Acute Injury Mechanisms. Front Neurol. 2019;10: 282. doi:10.3389/fneur.2019.00282
    OpenUrlCrossRefPubMed
  20. 20.↵
    Plantier V, Sanchez-Brualla I, Dingu N, Brocard C, Liabeuf S, Gackière F, et al. Calpain fosters the hyperexcitability of motoneurons after spinal cord injury and leads to spasticity. eLife. 2019;8: e51404. doi:10.7554/eLife.51404
    OpenUrlCrossRef
  21. 21.↵
    Brocard C, Plantier V, Boulenguez P, Liabeuf S, Bouhadfane M, Viallat-Lieutaud A, et al. Cleavage of Na+ channels by calpain increases persistent Na+ current and promotes spasticity after spinal cord injury. Nat Med. 2016;22: 404–411. doi:10.1038/nm.4061
    OpenUrlCrossRefPubMed
  22. 22.↵
    Plantier V, Brocard F. Une nouvelle cible thérapeutique dans le traitement de la spasticité après une lésion de la moelle épinière : la calpaïne. Med Sci (Paris). 2017;33: 629–636. doi:10.1051/medsci/20173306020
    OpenUrlCrossRefPubMed
  23. 23.↵
    Harris-Warrick RM, Pecchi E, Drouillas B, Brocard F, Bos R. Effect of size on expression of bistability in mouse spinal motoneurons. Journal of Neurophysiology. 2024;131: 577–588. doi:10.1152/jn.00320.2023
    OpenUrlCrossRefPubMed
  24. 24.
    Duflocq A, Le Bras B, Bullier E, Couraud F, Davenne M. Nav1.1 is predominantly expressed in nodes of Ranvier and axon initial segments. Molecular and Cellular Neuroscience. 2008;39: 180–192. doi:10.1016/j.mcn.2008.06.008
    OpenUrlCrossRefPubMedWeb of Science
  25. 25.↵
    Drouillas B, Brocard C, Zanella S, Bos R, Brocard F. Persistent Nav1.1 and Nav1.6 currents drive spinal locomotor functions through nonlinear dynamics. Cell Reports. 2023;42: 113085. doi:10.1016/j.celrep.2023.113085
    OpenUrlCrossRefPubMed
  26. 26.↵
    Li Y, Gorassini MA, Bennett DJ. Role of Persistent Sodium and Calcium Currents in Motoneuron Firing and Spasticity in Chronic Spinal Rats. Journal of Neurophysiology. 2004;91: 767–783. doi:10.1152/jn.00788.2003
    OpenUrlCrossRefPubMedWeb of Science
  27. 27.
    Bennett DJ, Li Y, Siu M. Plateau Potentials in Sacrocaudal Motoneurons of Chronic Spinal Rats, Recorded In Vitro. Journal of Neurophysiology. 2001;86: 1955–1971. doi:10.1152/jn.2001.86.4.1955
    OpenUrlCrossRefPubMedWeb of Science
  28. 28.
    Bennett DJ, Li Y, Harvey PJ, Gorassini M. Evidence for Plateau Potentials in Tail Motoneurons of Awake Chronic Spinal Rats With Spasticity. Journal of Neurophysiology. 2001;86: 1972–1982. doi:10.1152/jn.2001.86.4.1972
    OpenUrlCrossRefPubMedWeb of Science
  29. 29.
    Harvey PJ, Li X, Li Y, Bennett DJ. 5-HT2 Receptor Activation Facilitates a Persistent Sodium Current and Repetitive Firing in Spinal Motoneurons of Rats With and Without Chronic Spinal Cord Injury. Journal of Neurophysiology. 2006;96: 1158–1170. doi:10.1152/jn.01088.2005
    OpenUrlCrossRefPubMedWeb of Science
  30. 30.
    Harvey PJ, Li Y, Li X, Bennett DJ. Persistent Sodium Currents and Repetitive Firing in Motoneurons of the Sacrocaudal Spinal Cord of Adult Rats. Journal of Neurophysiology. 2006;96: 1141–1157. doi:10.1152/jn.00335.2005
    OpenUrlCrossRefPubMedWeb of Science
  31. 31.↵
    Li Y, Bennett DJ. Persistent Sodium and Calcium Currents Cause Plateau Potentials in Motoneurons of Chronic Spinal Rats. Journal of Neurophysiology. 2003;90: 857–869. doi:10.1152/jn.00236.2003
    OpenUrlCrossRefPubMedWeb of Science
  32. 32.↵
    Gorassini MA, Knash ME, Harvey PJ, Bennett DJ, Yang JF. Role of motoneurons in the generation of muscle spasms after spinal cord injury. Brain. 2004;127: 2247–2258. doi:10.1093/brain/awh243
    OpenUrlCrossRefPubMedWeb of Science
  33. 33.
    Hornby TG, Rymer WZ, Benz EN, Schmit BD. Windup of Flexion Reflexes in Chronic Human Spinal Cord Injury: A Marker for Neuronal Plateau Potentials? Journal of Neurophysiology. 2003;89: 416–426. doi:10.1152/jn.00979.2001
    OpenUrlCrossRefPubMedWeb of Science
  34. 34.↵
    Nickolls P, Collins DF, Gorman RB, Burke D, Gandevia SC. Forces consistent with plateau-like behaviour of spinal neurons evoked in patients with spinal cord injuries. Brain. 2004;127: 660–670. doi:10.1093/brain/awh073
    OpenUrlCrossRefPubMedWeb of Science
  35. 35.↵
    Schoch KM, von Reyn CR, Bian J, Telling GC, Meaney DF, Saatman KE. Brain injury-induced proteolysis is reduced in a novel calpastatin-overexpressing transgenic mouse. J Neurochem. 2013;125: 909–920. doi:10.1111/jnc.12144
    OpenUrlCrossRefPubMedWeb of Science
  36. 36.
    Von Reyn CR, Mott RE, Siman R, Smith DH, Meaney DF. Mechanisms of calpain mediated proteolysis of voltage gated sodium channel α-subunits following in vitro dynamic stretch injury. Journal of Neurochemistry. 2012;121: 793–805. doi:10.1111/j.1471-4159.2012.07735.x
    OpenUrlCrossRefPubMed
  37. 37.↵
    Von Reyn CR, Spaethling JM, Mesfin MN, Ma M, Neumar RW, Smith DH, et al. Calpain Mediates Proteolysis of the Voltage-Gated Sodium Channel α-Subunit. J Neurosci. 2009;29: 10350–10356. doi:10.1523/JNEUROSCI.2339-09.2009
    OpenUrlAbstract/FREE Full Text
  38. 38.↵
    Schnake KJ, Schroeder GD, Vaccaro AR, Oner C. AOSpine Classification Systems (Subaxial, Thoracolumbar). Journal of Orthopaedic Trauma. 2017;31: S14–S23. doi:10.1097/BOT.0000000000000947
    OpenUrlCrossRefPubMed
  39. 39.↵
    Guest J, Datta N, Jimsheleishvili G, Gater DR. Pathophysiology, Classification and Comorbidities after Traumatic Spinal Cord Injury. JPM. 2022;12: 1126. doi:10.3390/jpm12071126
    OpenUrlCrossRefPubMed
  40. 40.↵
    Talbott JF, Whetstone WD, Readdy WJ, Ferguson AR, Bresnahan JC, Saigal R, et al. The Brain and Spinal Injury Center score: a novel, simple, and reproducible method for assessing the severity of acute cervical spinal cord injury with axial T2-weighted MRI findings. SPI. 2015;23: 495–504. doi:10.3171/2015.1.SPINE141033
    OpenUrlCrossRefPubMed
  41. 41.↵
    El Masry WS, Tsubo M, Katoh S, El Miligui YHS, Khan A. Validation of the American Spinal Injury Association (ASIA) Motor Score and the National Acute Spinal Cord Injury Study (NASCIS) Motor Score: Spine. 1996;21: 614–619. doi:10.1097/00007632-199603010-00015
    OpenUrlCrossRefPubMedWeb of Science
  42. 42.↵
    Gabbe BJ, Cameron PA, Finch CF. The status of the Glasgow Coma Scale. Emergency Medicine. 2003;15: 353–360. doi:10.1046/j.1442-2026.2003.00474.x
    OpenUrlCrossRef
  43. 43.↵
    Harb A, Kishner S. Modified Ashworth Scale. StatPearls. Treasure Island (FL): StatPearls Publishing; 2024. Available: http://www.ncbi.nlm.nih.gov/books/NBK554572/
  44. 44.↵
    McMillan T, Wilson L, Ponsford J, Levin H, Teasdale G, Bond M. The Glasgow Outcome Scale — 40 years of application and refinement. Nat Rev Neurol. 2016;12: 477–485. doi:10.1038/nrneurol.2016.89
    OpenUrlCrossRefPubMed
  45. 45.↵
    Jenkinson C, Coulter A, Wright L. Short form 36 (SF36) health survey questionnaire: normative data for adults of working age. BMJ. 1993;306: 1437–1440. doi:10.1136/bmj.306.6890.1437
    OpenUrlAbstract/FREE Full Text
  46. 46.↵
    Bouhassira D, Attal N, Alchaar H, Boureau F, Brochet B, Bruxelle J, et al. Comparison of pain syndromes associated with nervous or somatic lesions and development of a new neuropathic pain diagnostic questionnaire (DN4). Pain. 2005;114: 29–36. doi:10.1016/j.pain.2004.12.010
    OpenUrlCrossRefPubMedWeb of Science
  47. 47.↵
    Copes WS, Champion HR, Sacco WJ, Lawnick MM, Keast SL, Bain LW. The Injury Severity Score Revisited: The Journal of Trauma: Injury, Infection, and Critical Care. 1988;28: 69–77. doi:10.1097/00005373-198801000-00010
    OpenUrlCrossRefPubMedWeb of Science
  48. 48.↵
    Le Gall J-R. A New Simplified Acute Physiology Score (SAPS II) Based on a European/North American Multicenter Study. JAMA. 1993;270: 2957. doi:10.1001/jama.1993.03510240069035
    OpenUrlCrossRefPubMedWeb of Science
  49. 49.↵
    Moreno R, Vincent J-L, Matos R, Mendonça A, Cantraine F, Thijs L, et al. The use of maximum SOFA score to quantify organ dysfunction/failure in intensive care. Results of a prospective, multicentre study. Intensive Care Medicine. 1999;25: 686–696. doi:10.1007/s001340050931
    OpenUrlCrossRefPubMedWeb of Science
  50. 50.↵
    Khwaja A. KDIGO Clinical Practice Guidelines for Acute Kidney Injury. Nephron Clin Pract. 2012;120: c179–c184. doi:10.1159/000339789
    OpenUrlCrossRefPubMed
  51. 51.↵
    Yokobori S, Zhang Z, Moghieb A, Mondello S, Gajavelli S, Dietrich WD, et al. Acute Diagnostic Biomarkers for Spinal Cord Injury: Review of the Literature and Preliminary Research Report. World Neurosurgery. 2015;83: 867–878. doi:10.1016/j.wneu.2013.03.012
    OpenUrlCrossRefPubMed
  52. 52.↵
    Pineda JA, Lewis SB, Valadka AB, Papa L, Hannay HJ, Heaton SC, et al. Clinical Significance of α II-Spectrin Breakdown Products in Cerebrospinal Fluid after Severe Traumatic Brain Injury. Journal of Neurotrauma. 2007;24: 354–366. doi:10.1089/neu.2006.003789
    OpenUrlCrossRefPubMedWeb of Science
Back to top
PreviousNext
Posted February 08, 2025.
Download PDF
Data/Code
Email

Thank you for your interest in spreading the word about medRxiv.

NOTE: Your email address is requested solely to identify you as the sender of this article.

Enter multiple addresses on separate lines or separate them with commas.
Calpain-mediated sodium channel fragments in the monocentric SpasT-SCI-T trial: Biomarkers for central nervous system injuries and spasticity prediction
(Your Name) has forwarded a page to you from medRxiv
(Your Name) thought you would like to see this page from the medRxiv website.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
Share
Calpain-mediated sodium channel fragments in the monocentric SpasT-SCI-T trial: Biomarkers for central nervous system injuries and spasticity prediction
Guillaume Baucher, Sylvie Liabeuf, Cécile Brocard, Aurélie Ponz, Karine Baumstarck, Lucas Troude, Marc Leone, Pierre-Hugues Roche, Frédéric Brocard
medRxiv 2025.02.07.25321849; doi: https://doi.org/10.1101/2025.02.07.25321849
Twitter logo Facebook logo LinkedIn logo Mendeley logo
Citation Tools
Calpain-mediated sodium channel fragments in the monocentric SpasT-SCI-T trial: Biomarkers for central nervous system injuries and spasticity prediction
Guillaume Baucher, Sylvie Liabeuf, Cécile Brocard, Aurélie Ponz, Karine Baumstarck, Lucas Troude, Marc Leone, Pierre-Hugues Roche, Frédéric Brocard
medRxiv 2025.02.07.25321849; doi: https://doi.org/10.1101/2025.02.07.25321849

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
  • Tweet Widget
  • Facebook Like
  • Google Plus One

Subject Area

  • Intensive Care and Critical Care Medicine
Subject Areas
All Articles
  • Addiction Medicine (434)
  • Allergy and Immunology (758)
  • Anesthesia (222)
  • Cardiovascular Medicine (3310)
  • Dentistry and Oral Medicine (366)
  • Dermatology (282)
  • Emergency Medicine (479)
  • Endocrinology (including Diabetes Mellitus and Metabolic Disease) (1175)
  • Epidemiology (13396)
  • Forensic Medicine (19)
  • Gastroenterology (900)
  • Genetic and Genomic Medicine (5171)
  • Geriatric Medicine (482)
  • Health Economics (785)
  • Health Informatics (3282)
  • Health Policy (1144)
  • Health Systems and Quality Improvement (1198)
  • Hematology (432)
  • HIV/AIDS (1022)
  • Infectious Diseases (except HIV/AIDS) (14649)
  • Intensive Care and Critical Care Medicine (914)
  • Medical Education (478)
  • Medical Ethics (128)
  • Nephrology (525)
  • Neurology (4945)
  • Nursing (262)
  • Nutrition (734)
  • Obstetrics and Gynecology (888)
  • Occupational and Environmental Health (796)
  • Oncology (2528)
  • Ophthalmology (730)
  • Orthopedics (284)
  • Otolaryngology (347)
  • Pain Medicine (323)
  • Palliative Medicine (90)
  • Pathology (546)
  • Pediatrics (1304)
  • Pharmacology and Therapeutics (551)
  • Primary Care Research (558)
  • Psychiatry and Clinical Psychology (4223)
  • Public and Global Health (7524)
  • Radiology and Imaging (1713)
  • Rehabilitation Medicine and Physical Therapy (1017)
  • Respiratory Medicine (981)
  • Rheumatology (480)
  • Sexual and Reproductive Health (500)
  • Sports Medicine (425)
  • Surgery (551)
  • Toxicology (72)
  • Transplantation (237)
  • Urology (206)