- Reviews
- Open access
- Published:
Translational approach to assess brain injury after cardiac arrest in preclinical models: a narrative review
Intensive Care Medicine Experimental volume 13, Article number: 3 (2025)
Introduction
Post-cardiac arrest brain injury (PCABI) is the main cause of death and disability in cardiac arrest (CA) patients [1]. Our understanding of the processes linking the cellular and molecular mechanisms that mediate brain structural and functional damage after CA is still incomplete, partly because of the paucity of relevant preclinical studies.
Increasing evidence suggests that, despite the many positive results from experimental studies, several neuroprotective drugs have failed to demonstrate clinical benefits in humans after CA [2]. This discrepancy may be attributed to biases or misinterpretation of preclinical results, such as the use of cerebral ischemia models without CA, unclear reporting of random assignment, lack of blinding and power analysis, poor reproducibility and insufficiently detailed study protocols [3]. In addition, there is often a failure to rigorously adhere to ARRIVE guidelines [4]. To improve the translatability of experimental results, preclinical study designs should more closely align with clinical settings. Indeed, many preclinical findings are difficult to translate into clinical practice, partly due to differences in injury models and functional assessment methods [3].
In PCABI animal studies, neurological damage and outcome are typically assessed using mainly neuropathology and animal-specific neurological deficit scores [3], with the addition of blood biomarkers of brain injury and neuroimaging techniques in some cases. In clinical practice, neuroprognostication after cardiac arrest relies on a multimodal approach incorporating clinical examination, circulating biomarkers, neurophysiology, and imaging to enhance diagnostic accuracy [5]. However, experimental studies rarely employ a multimodal approach or focus on evaluating whether individual methods correlate strongly with functional outcomes, contributing to the frequent failure in translating preclinical findings to clinical settings. To address this gap, it is crucial that preclinical research adopts methodologies similar to those used clinically. Drawing from clinical practice, preclinical studies should emphasize combining multiple techniques to provide a comprehensive assessment of brain injury evolution and recovery.
This narrative review aims to provide a comprehensive overview of the translational methods employed in preclinical research for evaluating brain injury and neurological recovery after CA. It seeks to analyze how different methods such as histological analysis, blood biomarkers, neurophysiology and neuroimaging techniques are employed in preclinical studies and will highlight the strengths and limitations of these methods in correlating with functional outcomes. The primary objective is to identify methodological approaches that more accurately reflect functional outcomes in the animal models. In addition, the review seeks to propose a multimodal approach that combines different techniques in animal’s studies, with the goal of bridging the gap between preclinical findings and clinical outcomes.
Materials and methods
Search strategy
This narrative review was conducted in accordance with the Preferred Reporting Items for Systematic Reviews and Meta-analysis (PRISMA, Fig. 1) [6, 7]. The PubMed database was selected as the primary source for data retrieval. We identified studies focusing on CA, brain injury and neurological outcomes in animal models published between 2021 and 2024. The search combined both keywords, Title/Abstract (Tiab) and MeSH (Medical Subject Headings) terms to ensure comprehensive coverage of relevant literature.
((cardiac arrest [Tiab]) OR (cardiopulmonary resuscitation [Tiab]) OR (cardiopolmunary resuscitation [MeSH]) OR (heart arrest [MeSH])) AND ((brain injury [Tiab]) OR (neurological outcome [Tiab]) OR (neurological recovery [Tiab]) OR (neurological dysfunction [Tiab]) OR (brain dysfunction [Tiab]) OR (neuroprotection [MeSH]) OR (brain injuries [MeSH]) OR (recovery of function [MeSH])) AND animal [Filter] AND (2021:2024[pdat]).
Articles eligible for inclusion in this narrative review were independently assessed for quality by two authors (FM and MC) and differences in scoring were resolved through discussion with the corresponding author (FF). The selection criteria further refine the search 141 results and after application of selection criteria, we identified 59 eligible studies.
Data collection
From all eligible articles, the following information were recorded: PMID number, DOI reference, name of the first author, publication year, title, aim, CA model, animal model, description of neurological function evaluation, histopathology, imaging, electrophysiology, circulating biomarkers, and main findings.
Selection criteria
The selection of studies was guided by specific criteria to ensure a comprehensive evaluation of brain injury and recovery following CA in animal models. The primary inclusion criteria was the presence of functional outcome tests. Only studies that included assessments of functional outcomes, tests that measure behavioral, cognitive, or neurological recovery, were considered. Another inclusion criterion was the evaluation of brain injury. Studies needed to include at least one of the following methods for assessing brain injury:
-
Histological evaluation: this involves the microscopic examination of brain tissue to assess structural damage at the cellular level.
-
In vivo neuro imaging techniques: methods such as magnetic resonance imaging (MRI) or computed tomography (CT) scans are used to visualize brain injury or recovery, offering a non-invasive way to track changes in brain structure and function over time.
-
Electroencephalography (EEG): monitoring of brain electrical activity to assess the severity of brain dysfunction and track neurological recovery over time.
-
Circulating biomarkers: measurement of specific protein biomarkers, such as S100 calcium‐binding protein B (S100B), neuro-specific enolase (NSE), neurofilament light (Nfl) and lactate, in the bloodstream. These biomarkers reflect multi-organ failure and neuronal damage and are valuable for assessing the severity and progression of brain injury after CA.
By adhering to these inclusion criteria, this review aims to highlight studies that not only examine the extent of brain damage but also provide methodological approaches that more accurately reflect functional outcomes.
Results
The search identified 59 articles (Tables 1, 2, 3, 4, 5 and 6). Of these, 39 were models of asphyxia-induced CA (Tables 1 and 2), 6 were model of asystolia-induced CA (Table 3) and 15 were models of ventricular fibrillation-induced CA (Tables 4, 5 and 6). The search was not limited to a single species, but it encompasses a diverse range of species: seven articles used mice model, one used a rabbit model, 43 employed rats and eight works pigs.
This analysis showed that histology was the most commonly used technique, with 82% of studies employing histological or immunohistochemical evaluations to assess brain injury. In contrast, circulating biomarkers, imaging, and EEG were used less frequently, appearing in 33%, 10%, and 7%, of animal studies, respectively.
Survival rates were tracked in most studies at various timepoints, including 24 h and 72 h post-CA. A subset of studies extended the monitoring period to 14 days, which is critical for understanding long-term outcomes.
Neurological functional outcome was measured by general neurological behavior test using clinical deficit scores with specific grids for each species. In other 23 studies, animals underwent behavioral testing, to assess distinct cognitive behavior, such as Open Field, Y-Maze, Novel Object recognition, Morris Water Maze, Rotarod and Tape Removal tests.
Forty-nine articles employed histological examination to measure the severity of PCABI. Common markers included Nissl, Iba1, GFAP, and Tunel, which were used to analyze neuronal damage, microglial and astrocytes activation and apoptosis (Fig. 2A).
Six studies employed advanced neuroimaging techniques. MRI–diffusion-weighted imaging (MRI–DWI) and MRI–diffusion tensor imaging (MRI–DTI) were used to evaluate structural brain damage in five studies. In two studies [8, 9], MRI was used in combination with other assessments, such as neurological function or biomarker measurements. This multimodal approach offers a more comprehensive understanding of both functional and structural changes in the brain. One study employed combined CT scans as a reference for precise anatomical localization of glucose uptake in different brain regions [10]. In general, this analysis indicates a moderate reliance on advanced imaging techniques, primarily MRI, for detecting structural damage (Fig. 2A).
Only 4 studies used EEG approach to evaluate neurophysiology [11,12,13,14], indeed real-time functional assessment of brain injury remains underexplored in these preclinical studies (Fig. 2A).
Our analysis revealed that in 20 papers circulating biomarkers were evaluated, with NSE and S100B, being the most used. Lactate was analyzed in 8 different studies, as marker of metabolic distress and brain hypoxia (Fig. 2A). Meanwhile, NfL was measured in only two studies.
To determine whether a multimodal approach was employed in evaluating PCABI we also examined how many experimental studies utilized more than one method. Among the 59 articles reviewed, none fully adopted a multimodal approach. Only two studies used three methods to evaluate PCABI in mouse and rat models, specifically combining histological analysis, MRI and NfL levels [8, 9]. One study, investigated PCABI employing histological analysis, EEG evaluation and lactate levels after CA [11]. In contrast, the majority of the studies employed only one or two techniques, with 42 using a single approach and 14 using two (Fig. 2B).
Discussion
Our analysis reveals significant discrepancies between preclinical study designs and clinical practice, underscoring the urgent need for more clinically relevant and multimodal approaches to enhance the translational potential of experimental findings. Specifically, our study shows that most of preclinical studies on CA were not designed based on a clinically derived approach, thus limiting the translatability of results.
The need for neuroprotective treatments
There is an urgent need to identify novel neuroprotective treatments after CA. Over the past few decades, many treatments have been tested in the preclinical setting. Remarkably, the majority of them showed promising results in animal studies but failed to replicate the same success in large clinical trials. One reason for this translational failure is the discrepancy between the methods used in preclinical research and those applied in clinical practice.
In clinical neuroprognostication—the prediction of neurological recovery—it is recommended to use a multimodal approach as no single test has sufficient specificity to avoid false positives[2]. Embracing a multimodal methodology in animal research, could increase the translatability of findings, mirroring clinical practice, where combining multiple assessment tools improves the prediction of neurological outcomes.
Post-resuscitation care and neuroprognostication
The post-resuscitation care in years has gained increasing importance. In 2010 guidelines, the post-resuscitation care was a paragraph incorporated in Advance Life Support section [15]. In 2015, the ERC and ESICM published guidelines specifically focused on the post-resuscitation care section, emphasizing the importance of high-quality post-resuscitation care and highlighting how this is a crucial factor in the chain of survival. Moreover, in 2015, it has been introduced for the first time the concept of the multimodal approach for neuroprognostication [16].
Two-thirds of deaths occurring in OHCA patients are due to PCABI, with the majority resulting from the active withdrawal of life-sustaining treatment (WLST) based on neuroprognostication. Indeed, it was important to minimize the risk of falsely poor prediction [16]. ERC guidelines 2015, suggested for the first time a multimodal neuroprognostication strategy, so a combination of distinct parameters that can increase the sensitivity to predict a poor outcome in patients. Specifically, this prognostication strategy algorithm was composed by one or both of no pupillary and corneal reflexes and bilaterally absent N20 SSEP wave [16]. In addition, they recommended that in none of those signs were present, it can be evaluated also a group of less accurate predictors: the presence of early status myoclonus, high values of NSE at 48–72 h after Return of Spontaneous Circulation (ROSC), an unreactive malignant EEG pattern and the presence of anoxic injury evaluated by brain CT or brain MRI scans [16]. ERC and ESICM proposed this algorithm based on evidence that none of these predictors singularly predicted poor outcome with 100% of accuracy, but combined together, then, they can increase safety and sensitivity to avoid falsely pessimistic prediction.
In the latest 2021 guidelines the concept of neuroprognostication has been revisited and improved: since 2015 there has been many studies regarding prognostication, which validated and confirmed the reliability of the algorithm presented in the last guidelines of 2015. Therefore, based on these data, they simplified the two-stage strategy algorithm so that poor outcome is considered likely when two or more listed predictors are present: no pupillary or corneal reflex at ≥ 72 h, bilaterally absent N20 SSEP wave at ≥ 24 h, NSE > 60 μg/L at 48 h and/or 72 h, presence of status myoclonus ≤ 72 h and a diffuse and extensive anoxic brain injury on CT or MRI scans [2].
The role of histological analysis
Histological analysis is an essential component of preclinical research, providing critical insights into the biological mechanisms underlying injury evolution following CA. Although the findings from histological studies are not always translatable to clinical settings, they are vital for understanding the pathophysiological changes that occur after brain injury. Histological and immunohistochemical methods remain essential tools for evaluating the severity of PCABI. Techniques such as hematoxylin and eosin (H&E), Nissl and Fluoro-Jade staining allow researchers to examine neuronal death and neurodegeneration. [17]. Immunohistochemistry is particularly valuable for investigating the inflammatory response following CA [17], focusing on key cellular populations like microglia and astrocytes, which mediate the brain's immune response and hold a pivot role in contributing to long term outcome. [1, 5]. These glial cells are highly active in the injured brain and play a pivotal role as immune cells that mediate inflammation. Their behavior and activation states, as revealed through immunohistochemistry, provide important clues about the extent and nature of the brain's response to injury after CA [1, 5]. Susceptibility to ischemia reperfusion injury due to CA, varies significantly depending on the neuronal subtype and region, with area more susceptible than others: the neocortex, the hippocampus, basal ganglia, cerebellum and thalamus [5].
However, it is essential to recognize that histological analysis cannot be obtained at the bedside and not always directly correlate with functional outcomes, which can limit its applicability in clinical contexts. Therefore, integrating histological analysis with other assessments, such as neuroimaging, biomarkers, and functional outcome measures, is crucial for a more comprehensive understanding of brain injury and recovery in conjunction with behavioral assessments.
Multimodal approaches in preclinical studies
Inspired by the multimodal approach used in clinical practice for neuroprognostication, we propose applying a similar strategy in preclinical research to improve the assessment of brain injury and functional recovery after CA.
Functional outcome
Given the poor prognosis of cardiopulmonary resuscitation (CPR) with regard to both survival and neurological outcome, functional evaluation constitute one of the primary measures of outcome [18].
In preclinical research on CA, neurological deficit tests play a critical role in assessing the extent of brain injury and functional recovery in animal models. These tests can be broadly categorized into two groups: those evaluating general neurological behavior and those focused on cognitive and behavioral assessments.
General neurological behavior
General neurological behavior tests provide a basic overview of the animal’s neurological status post-CA. These tests are essential for identifying the immediate and overt effects of brain injury. Specifically, distinct aspects of animal’s behavior can be assessed using clinical deficit scores with specific grids for each species.
They are based on the clinical evaluation of consciousness, behavior, breathing, reflexes, coordination, motor and sensory activity and seizure. The consciousness and general behavior, is asses by observing any changes in movement or responsiveness of the animal. The brainstem performance, is the evaluation of reflexes controlled by brainstem, i.e., pupillary, responses and breathing. The coordination, is assessed by testing the motor coordination through balance observations. Furthermore, the corneal reflex is evaluated by blink response to corneal stimulation and it indicates the cranial nerve function. The motor and sensory activity is the assessment of animal’s voluntary movements and response to sensory stimuli. Finally, is often present the study of seizure activity, so monitoring the occurrence of seizure or convulsions post-CA.
These analyses provide insight into the severity of neurological damage and recovery following CA. However, they primarily focus on broad neurological outcomes and may not capture the full spectrum of cognitive or behavioral impairments.
Cognitive and behavioral assessments
The second group offers a more specific evaluation of specific brain functions. These tests explore aspects like memory, anxiety, exploratory behavior, and learning, critical in understanding the deeper impact of CA on brain function. The key tests in this category include:
-
Open field test: used to assess anxiety levels and exploratory behavior by measuring the animal's movement and interaction with a novel environment [8, 19,20,21,22];
-
Novel object recognition test: evaluates recognition memory, based on the animal's ability to differentiate between familiar and new objects [20, 23];
-
Morris water maze: a well-established test for assessing spatial learning and memory, where animals must navigate to a hidden platform in a pool of water [19, 21, 24,25,26,27,28,29];
-
Tape removal test: evaluating sensorimotor function by placing adhesive tape on the forepaw and recording the time it takes for the animal to detect and remove it, reflecting sensory and motor coordination [8, 30, 31];
-
Y-maze: Used to assess working memory by analyzing spontaneous alternation behavior as the animal explores different arms of the maze [32, 33];
-
Motor activity test: assesses general locomotor activity by tracking the animal’s movement, measuring distinct parameters like distance traveled, speed, time spent moving versus resting and spontaneous motor activity during light and dark phase of day [8, 9, 19, 21, 22, 34].
These cognitive and behavioral tests are particularly valuable for examining long-term brain function, providing a more detailed understanding of how CA impacts learning, memory, and emotional regulation.
Biomarkers
Multi-organ failure and neuronal damage can be measured in the serum or plasma after CA as biomarkers. Major advantages of blood biomarkers are that they are easy to obtain and offer a quantitative and easily interpreted measure of the extent of brain injury [1].
Neuron-specific enolase
NSE is a neuronal glycolytic enzyme that is abundant in the neurons of brain gray matter and involved in axonal transport [35]. In healthy individuals, serum levels of NSE remain low. In contrast, upon damage to neuronal tissue, such as anoxic brain injury, NSE serum concentration increases and consequently acts as a biomarker for brain damage. European Resuscitation Council (ERC) guidelines 2021 indicates that increasing values between 24 and 48 h or 72 h in combination with high values at 48 and 72 h indicate a poor prognosis [2].
S100 calcium‐binding protein B
S100B is abundant in glial cells expressed in astrocytes surrounding the blood vessels in the brain and in the Schwann cells of the peripheral nervous system, where they stimulate cellular processes such as proliferation, differentiation, and regulation of intracellular Ca2+ homeostasis. At least 80–90% of the total S100B pool is found within the brain, the remainder being located in other non-neuronal tissues. S100B is released from damaged astrocytes into the bloodstream after ischemic–reperfusion injury that occur after CA. Is considered an early biomarker, as the level usually peaks at 24 h and elevated levels are associated with poor outcome [35].
Neurofilament light
Neurofilament light chain (NfL) is a subunit of neurofilaments, which are cylindrical proteins exclusively located in the neuronal cytoplasm, predominantly within large, myelinated axons within the cerebral white matter. Their function is largely unknown but hypothesized to be essential for radial growth and enabling rapid nerve conduction [35]. Under physiological conditions, low levels of NfL are constantly released from axons, probably in an age-dependent manner, however, in response to CNS axonal damage because of pathological problem, such as an ischemic insult as occurs during CA, the release of NfL sharply increases. High levels in CA patients are index of poor outcomes [35].
Lactate
Lactate, a product of pyruvate reduction during glycolysis, has been suggested to be an indicator of multi-organ failure hypoxia resulting from reduced cardiac output and in CA patients elevated arterial blood lactate levels are associated with poor neurologic outcome [36]. Lactatemia during the first hours after CA is associated with hypoperfusion after the cessation of blood flow and the inflammatory reaction due to ischemia–reperfusion injury [36]. Hyperlactatemia few h after ROSC may indicate complication in patients, such as poor neurological function. Hence, lactate levels in CA are a critical marker for assessing the severity of ischemic–reperfusion injury, as well as predicting neurological outcomes.
Electroencephalography
Electroencephalography (EEG) is a highly sensitive tool for detecting the severity of PCABI since assesses cortical synaptic activity [5]. Moreover, EEG is able to evaluate the occurrence of seizures as well as the appearance of spikes/sharp waves and epileptiform discharges. These, together with malignant EEG patterns (persistent iso-electricity, low voltage activity, or low burst-suppression patterns), are used to prognosticate outcome after CA. ERC guidelines 2021 suggest that highly malignant EEG after 24 h is an indicator of poor neurological outcome [2].
Imaging
Brain CT is extensively used shortly after CA to rule out neurological causes of arrest, especially an intracranial hemorrhage that would contraindicate percutaneous coronary interventions. However, CT also allows assessing the severity of PCABI by detecting brain oedema [1].
The use of MRI for prognostication is more recent, but has rapidly gained interest during the last decade. One of the main advantages of MRI is the ability to assess the anatomical distribution of diffusion restrictions [37]. MRI allows the detection of cytotoxic edema, which occurs within hours after CA. Restricted diffusion by cytotoxic edema can be quantified by the Apparent Diffusion Coefficient (ADC) value of each voxel. Low ADC values, thus restricted water diffusion (DWI lesions) are associated with poor outcome after CA [2]. Moreover, an ADC values < 650X10−6 mm2/s in > 10% of the brain at 7 days after CA is highly specific for poor outcome.
DTI is an extension of DWI that allows the evaluation of microstructural integrity of brain white matter by directional assessment of water diffusion. Although DTI is not a criterion in the strategy algorithm for neuroprognostication, it was found that changes in DTI parameters can predict poor outcome in CA patients with 85% sensitivity [37].
Bridging the gap between preclinical and clinical studies
Our review highlights several gaps between preclinical models and clinical practices. Preclinical studies often rely heavily on brain histopathology as a primary endpoint, neglecting correlations with functional outcomes or biomarkers. Furthermore, animal models typically involve young, healthy subjects, which do not adequately reflect the comorbidities present in clinical patients.
Significant correlations are seen between imaging, neurological deficits, and biomarkers, reflecting the severity of brain injury in CA patients, suggesting similar correlations should be explored in preclinical models. Indeed, in our analysis we did not find studies that explored possible correlations between outcomes. Interestingly, our review highlights a significant gap in the literature, as very few studies have explored potential correlations between histology, neuroimaging, electrophysiology, blood biomarkers, and neurobehavioral outcomes. Notably, there are only two studies addressing these correlations that demonstrated strong, positive correlations between apparent diffusion coefficient (ADC) values and the neurological deficit score, showing that the severity of brain cytotoxic edema is closely associated with worsened neurological function in the early phase after CA [8, 9]. In addition, one of these studies found a strong correlation between diffusion tensor imaging (DTI) metrics and the brain injury biomarker NfL [8]. These findings provide compelling evidence that such correlation can be identified underscoring a critical parallel between preclinical models and the clinical scenarios, emphasizing the translational relevance of incorporating multimodal assessments in preclinical research.
To improve translational value, preclinical models should integrate multimodal assessments and better mimic clinical conditions providing a more comprehensive assessment of the brain during CA.
Conclusion
Our review advocates for the adoption of a multimodal approach in preclinical studies, integrating neuroimaging, biomarkers, and EEG, to better reflect the severity of brain injury, similar to the strategies used in clinical neuroprognostication. These approaches must always be integrated with histology, which plays a pivotal role in preclinical studies in understanding the pathophysiological mechanisms underlying PCABI. This comprehensive approach aligns with clinical methodologies, where the combination of multiple assessment tools provides a more reliable evaluation of neurological outcomes in patients. By implementing such a multimodal strategy in preclinical research, we can improve the translational relevance of experimental findings, helping to bridge the gap between laboratory models and clinical practice.
Availability of data and materials
The data sets generated and/or analyzed during the current study are available at: https://zenodo.org/records/13928458
References
Sandroni C, Cronberg T, Sekhon M (2021) Brain injury after cardiac arrest: pathophysiology, treatment, and prognosis. Intensive Care Med 47:1393–1414. https://doi.org/10.1007/s00134-021-06548-2
Nolan JP, Sandroni C, Böttiger BW et al (2021) European resuscitation council and European society of intensive care medicine guidelines 2021: post-resuscitation care. Intensive Care Med 47:369–421. https://doi.org/10.1007/s00134-021-06368-4
Perkins GD, Neumar R, Hsu CH et al (2024) Improving outcomes after post-cardiac arrest brain injury: a scientific statement from the international liaison committee on resuscitation. Circulation. https://doi.org/10.1161/CIR.0000000000001219
Idris AH, Becker LB, Ornato JP et al (1996) Utstein-style guidelines for uniform reporting of laboratory CPR research: a statement for healthcare professionals from a task force of the American heart association, the American college of emergency physicians, the American college of cardiology, the European resuscitation council, the heart and stroke foundation of Canada, the institute of critical care medicine, the Safar Center for resuscitation research, and the society for academic emergency medicine. Circulation 94:2324–2336. https://doi.org/10.1161/01.CIR.94.9.2324
Carroll E, Lewis A (2021) Neuroprognostication after cardiac arrest: who recovers? who progresses to brain death? Semin Neurol 41:606–618. https://doi.org/10.1055/s-0041-1733789
Page MJ, Moher D, Bossuyt PM et al (2021) PRISMA 2020 explanation and elaboration: updated guidance and exemplars for reporting systematic reviews. BMJ. https://doi.org/10.1136/bmj.n160
Page MJ, McKenzie JE, Bossuyt PM et al (2021) The PRISMA 2020 statement: an updated guideline for reporting systematic reviews. BMJ. https://doi.org/10.1136/bmj.n71
Perego C, Fumagalli F, Motta F et al (2024) Evolution of brain injury and neurological dysfunction after cardiac arrest in the rat – A multimodal and comprehensive model. J Cereb Blood Flow Metab. https://doi.org/10.1177/0271678X241255599
Magliocca A, Perego C, Motta F et al (2023) Indoleamine 2,3-dioxygenase deletion to modulate kynurenine pathway and to prevent brain injury after cardiac arrest in mice. Anesthesiology 139:628–645. https://doi.org/10.1097/ALN.0000000000004713
Zhang HJ, Mitchell S, Fang Y-H et al (2021) Assessment of brain glucose metabolism following cardiac arrest by [18F]FDG positron emission tomography. Neurocrit Care 34:64–72. https://doi.org/10.1007/s12028-020-00984-6
Shoaib M, Choudhary RC, Chillale RK et al (2022) Metformin-mediated mitochondrial protection post-cardiac arrest improves EEG activity and confers neuroprotection and survival benefit. FASEB J. https://doi.org/10.1096/fj.202200121R
Wang Z, Du J, Lachance BB et al (2021) Intracerebroventricular administration of hNSCs improves neurological recovery after cardiac arrest in rats. Stem Cell Rev and Rep 17:923–937. https://doi.org/10.1007/s12015-020-10067-w
Dai C, Wang J, Li J et al (2021) Repetitive anodal transcranial direct current stimulation improves neurological recovery by preserving the neuroplasticity in an asphyxial rat model of cardiac arrest. Brain Stimul 14:407–416. https://doi.org/10.1016/j.brs.2021.02.008
Wang J, Li J, Chen B et al (2021) Interaction between gender and post resuscitation interventions on neurological outcome in an asphyxial rat model of cardiac arrest. BMC Cardiovasc Disord 21:441. https://doi.org/10.1186/s12872-021-02262-5
Berg RA, Hemphill R, Abella BS et al (2010) Part 5: adult basic life support: 2010 American heart association guidelines for cardiopulmonary resuscitation and emergency cardiovascular care. Circulation. https://doi.org/10.1161/CIRCULATIONAHA.110.970939
Nolan JP, Soar J, Cariou A et al (2015) European resuscitation council and European society of intensive care medicine 2015 guidelines for post-resuscitation care. Intensive Care Med 41:2039–2056. https://doi.org/10.1007/s00134-015-4051-3
Rahaman P, Del Bigio MR (2018) Histology of brain trauma and hypoxia-ischemia. Acad Forensic Pathol 8:539–554. https://doi.org/10.1177/1925362118797728
Albertsmeier M, Teschendorf P, Popp E et al (2007) Evaluation of a tape removal test to assess neurological deficit after cardiac arrest in rats. Resuscitation 74:552–558. https://doi.org/10.1016/j.resuscitation.2007.01.040
Zhang H, Wang R (2023) TWEAK knockdown alleviates post-cardiac arrest brain injury via the p38 MAPK/NF-κB pathway. Disc Med 35:503. https://doi.org/10.24976/Discov.Med.202335177.51
Peng F, Zhang Y-H, Zhang L et al (2022) Ketogenic diet attenuates post-cardiac arrest brain injury by upregulation of pentose phosphate pathway–mediated antioxidant defense in a mouse model of cardiac arrest. Nutrition 103–104:111814. https://doi.org/10.1016/j.nut.2022.111814
Chen J, Chang Y, Zhu J et al (2022) Flufenamic acid improves survival and neurologic outcome after successful cardiopulmonary resuscitation in mice. J Neuroinflammation 19:214. https://doi.org/10.1186/s12974-022-02571-2
Duan W, Sun Q, Wu X et al (2022) Cervical vagus nerve stimulation improves neurologic outcome after cardiac arrest in mice by attenuating oxidative stress and excessive autophagy. Neuromod Technol Neural Int 25:414–423. https://doi.org/10.1016/j.neurom.2021.12.014
Sun Y, Cong T, Li L et al (2022) Baclofen attenuates cognitive deficits in post-cardiac arrest brain injury. Biochem Biophys Res Commun 602:135–141. https://doi.org/10.1016/j.bbrc.2022.02.106
Chen C, Xia Z, Zhang M et al (2024) Molecular mechanism of HDAC6-mediated pyroptosis in neurological function recovery after cardiopulmonary resuscitation in rats. Brain Res 1843:149121. https://doi.org/10.1016/j.brainres.2024.149121
Xu M, Zhu J, Wang Z et al (2024) Neuroprotective effect of autologous mitochondrial transplantation against global ischemia/reperfusion injury in a rat model of cardiac arrest. Mitochondrion 78:101924. https://doi.org/10.1016/j.mito.2024.101924
Oghifobibi OA, Toader AE, Nicholas MA et al (2022) Resuscitation with epinephrine worsens cerebral capillary no-reflow after experimental pediatric cardiac arrest: An in vivo multiphoton microscopy evaluation. J Cereb Blood Flow Metab 42:2255–2269. https://doi.org/10.1177/0271678X221113022
Xia P, Marjan M, Liu Z et al (2022) Chrysophanol postconditioning attenuated cerebral ischemia-reperfusion injury induced NLRP3-related pyroptosis in a TRAF6-dependent manner. Exp Neurol 357:114197. https://doi.org/10.1016/j.expneurol.2022.114197
He Y, Chang Y, Peng Y et al (2022) Glibenclamide directly prevents neuroinflammation by targeting SUR1-TRPM4-mediated NLRP3 inflammasome activation in microglia. Mol Neurobiol 59:6590–6607. https://doi.org/10.1007/s12035-022-02998-x
Liu W, Ye Q, Xi W et al (2021) The ERK/CREB/PTN/syndecan-3 pathway involves in heparin-mediated neuro-protection and neuro-regeneration against cerebral ischemia-reperfusion injury following cardiac arrest. Int Immunopharmacol 98:107689. https://doi.org/10.1016/j.intimp.2021.107689
Cai S, Li Q, Fan J et al (2023) Therapeutic hypothermia combined with hydrogen sulfide treatment attenuated early blood-brain barrier disruption and brain edema induced by cardiac arrest and resuscitation in rat model. Neurochem Res 48:967–979. https://doi.org/10.1007/s11064-022-03824-5
Ousta A, Piao L, Fang YH et al (2022) Microglial activation and neurological outcomes in a murine model of cardiac arrest. Neurocrit Care 36:61–70. https://doi.org/10.1007/s12028-021-01253-w
Tan Y, Zhang J, Ge Q et al (2022) Ketone body improves neurological outcomes after cardiac arrest by inhibiting mitochondrial fission in rats. Oxid Med Cell Longev 2022:1–13. https://doi.org/10.1155/2022/7736416
Lee RH-C, Wu CY-C, Citadin CT et al (2022) Activation of neuropeptide Y2 receptor can inhibit global cerebral ischemia-induced brain injury. Neuromol Med 24:97–112. https://doi.org/10.1007/s12017-021-08665-z
Nishikimi M, Shoaib M, Choudhary RC et al (2022) Preserving brain LPC-DHA by plasma supplementation attenuates brain injury after cardiac arrest. Ann Neurol 91:389–403. https://doi.org/10.1002/ana.26296
Humaloja J, Ashton NJ, Skrifvars MB (2022) Brain injury biomarkers for predicting outcome after cardiac arrest. Crit Care 26:81. https://doi.org/10.1186/s13054-022-03913-5
Laurikkala J, Skrifvars MB, Bäcklund M et al (2019) Early lactate values after out-of-hospital cardiac arrest: associations with one-year outcome. Shock 51:168–173. https://doi.org/10.1097/SHK.0000000000001145
Keijzer HM, Duering M, Pasternak O et al (2023) Free water corrected diffusion tensor imaging discriminates between good and poor outcomes of comatose patients after cardiac arrest. Eur Radiol 33:2139–2148. https://doi.org/10.1007/s00330-022-09245-w
Xia Y, Zou C, Kang W et al (2024) Invasive metastatic tumor-camouflaged ROS responsive nanosystem for targeting therapeutic brain injury after cardiac arrest. Biomaterials 311:122678. https://doi.org/10.1016/j.biomaterials.2024.122678
Chang R, Hsu M, Lee T et al (2024) Selective brain perfusion improves the neurological outcomes after extracorporeal cardiopulmonary resuscitation in a rat model. Artif Organs 48:743–752. https://doi.org/10.1111/aor.14732
Li J, Shen Y, Wang J et al (2024) Combination of hyperoxygenation and targeted temperature management improves functional outcomes of post cardiac arrest syndrome irrespective of causes of arrest in rats. Shock 61:934–941. https://doi.org/10.1097/SHK.0000000000002338
Dai Z, Zhang S, Wang H et al (2024) Comparison between active abdominal compression-decompression cardiopulmonary resuscitation and standard cardiopulmonary resuscitation in Asphyctic cardiac arrest rats with multiple rib fractures. Shock 61:266–273. https://doi.org/10.1097/SHK.0000000000002283
Hu Y, Zhao X, Jiang G et al (2023) Prophylactic supplement with melatonin prevented the brain injury after cardiac arrest in rats. Sci Rep 13:20100. https://doi.org/10.1038/s41598-023-47424-x
Zhan H, Zhang Q, Zhang C et al (2023) Targeted activation of HNF4α by AMPK inhibits apoptosis and ameliorates neurological injury caused by cardiac arrest in rats. Neurochem Res 48:3129–3145. https://doi.org/10.1007/s11064-023-03957-1
Zhou Y, Zhang X, Yang H et al (2023) Mechanism of cAMP response element-binding protein 1/death-associated protein kinase 1 axis-mediated hippocampal neuron apoptosis in rat brain injury after cardiopulmonary resuscitation. Neuroscience 526:175–184. https://doi.org/10.1016/j.neuroscience.2023.06.024
Tang X, Ke J, Chen F et al (2023) Hypoxic preconditioned mesenchymal stem cells ameliorate rat brain injury after cardiopulmonary resuscitation by suppressing neuronal pyroptosis. J Cellular Molecular Medi 27:1836–1858. https://doi.org/10.1111/jcmm.17782
Zhang Q, Zhan H, Liu C et al (2023) Neuroprotective effect of miR-483-5p against cardiac arrest-induced mitochondrial dysfunction mediated through the TNFSF8/AMPK/JNK signaling pathway. Cell Mol Neurobiol 43:2179–2202. https://doi.org/10.1007/s10571-022-01296-3
Choudhary RC, Shoaib M, Hayashida K et al (2023) Multi-drug cocktail therapy improves survival and neurological function after asphyxial cardiac arrest in rodents. Cells 12:1548. https://doi.org/10.3390/cells12111548
Hayashida K, Takegawa R, Endo Y et al (2023) Exogenous mitochondrial transplantation improves survival and neurological outcomes after resuscitation from cardiac arrest. BMC Med 21:56. https://doi.org/10.1186/s12916-023-02759-0
Wang C, Huang C, Tsai M et al (2022) Inhaled carbon dioxide improves neurological outcomes by downregulating hippocampal autophagy and apoptosis in an asphyxia-induced cardiac arrest and resuscitation rat model. JAHA 11:e027685. https://doi.org/10.1161/JAHA.122.027685
Jackson TC, Dezfulian C, Vagni VA et al (2022) PHLPP inhibitor NSC74429 is neuroprotective in rodent models of cardiac arrest and traumatic brain injury. Biomolecules 12:1352. https://doi.org/10.3390/biom12101352
Guo Y, Cho S-M, Wei Z et al (2022) Early thalamocortical reperfusion leads to neurologic recovery in a rodent cardiac arrest model. Neurocrit Care 37:60–72. https://doi.org/10.1007/s12028-021-01432-9
Liu Y, Zhang L, Han R et al (2022) Electroacupuncture attenuates brain injury through α7 nicotinic acetylcholine receptor-mediated suppression of neuroinflammation in a rat model of asphyxial cardiac arrest. J Neuroimmunol 367:577873. https://doi.org/10.1016/j.jneuroim.2022.577873
Zhang L, Liang W, Li Y et al (2021) Mild therapeutic hypothermia improves neurological outcomes in a rat model of cardiac arrest. Brain Res Bull 173:97–107. https://doi.org/10.1016/j.brainresbull.2021.05.014
Wu S-N, Tsai M-S, Huang C-H, Chen W-J (2022) Omecamtiv mecarbil treatment improves post-resuscitation cardiac function and neurological outcome in a rat model. PLoS ONE 17:e0264165. https://doi.org/10.1371/journal.pone.0264165
Liu Z, Liu T, Cai J et al (2022) Quantitative magnetic resonance imaging assessment of brain injury after successful cardiopulmonary resuscitation in a rat model of asphyxia cardiac arrest. Brain Imaging Behav 16:270–280. https://doi.org/10.1007/s11682-021-00500-0
Shao R, Wang X, Xu T et al (2021) The balance between AIM2-associated inflammation and autophagy: the role of CHMP2A in brain injury after cardiac arrest. J Neuroinflammation 18:257. https://doi.org/10.1186/s12974-021-02307-8
Yang S, Yu C, Yang Z et al (2021) DL-3-n-butylphthalide-induced neuroprotection in rat models of asphyxia-induced cardiac arrest followed by cardiopulmonary resuscitation. J Cell Physiol 236:7464–7472. https://doi.org/10.1002/jcp.30442
Zhang R, Bryson TD, Fogo GM et al (2022) Rapid treatment with intramuscular magnesium sulfate during cardiopulmonary resuscitation does not provide neuroprotection following cardiac arrest. Mol Neurobiol 59:1872–1881. https://doi.org/10.1007/s12035-021-02645-x
Huang Y, Gao X, Zhou X et al (2021) Remote ischemic postconditioning inhibited mitophagy to achieve neuroprotective effects in the rat model of cardiac arrest. Neurochem Res 46:573–583. https://doi.org/10.1007/s11064-020-03193-x
Li Y, Zhu H, Cheng D, Zhao Z (2022) Inhibition of Γδ T cells alleviates brain ischemic injury in cardiopulmonary-cerebral resuscitation mice. Transplant Proc 54:1984–1991. https://doi.org/10.1016/j.transproceed.2022.05.033
Ye Z, Zhang F, Wang P et al (2023) Baicalein relieves brain injury via inhibiting ferroptosis and endoplasmic reticulum stress in a rat model of cardiac arrest. Shock 59:434–441. https://doi.org/10.1097/SHK.0000000000002058
Yuan Z-L, Zhang Z-X, Mo Y-Z et al (2022) Inhibition of extracellular signal-regulated kinase downregulates endoplasmic reticulum stress-induced apoptosis and decreases brain injury in a cardiac arrest rat model. Physiol Res. https://doi.org/10.33549/physiolres.934882
Wang W, Xie L, Zou X et al (2021) Pomelo peel oil suppresses TNF-α-induced necroptosis and cerebral ischaemia–reperfusion injury in a rat model of cardiac arrest. Pharm Biol 59:399–407. https://doi.org/10.1080/13880209.2021.1903046
Tsai M, Huang C, Wang C et al (2021) Post-cardiac arrest hydrocortisone use ameliorates cardiac mitochondrial injury in a male rat model of ventricular fibrillation cardiac arrest. JAHA 10:e019837. https://doi.org/10.1161/JAHA.120.019837
Motta F, De Giorgio D, Cerrato M et al (2024) Postresuscitation ventilation with a mixture of argon and hydrogen reduces brain injury after cardiac arrest in a pig model. JAHA 13:e033367. https://doi.org/10.1161/JAHA.123.033367
Shen R, Liu Z, Fei L et al (2024) Remimazolam improves the markers of postresuscitation cerebral injury in a swine model of cardiac arrest. Shock 61:783–790. https://doi.org/10.1097/SHK.0000000000002331
Wider JM, Gruley E, Morse PT et al (2023) Modulation of mitochondrial function with near-infrared light reduces brain injury in a translational model of cardiac arrest. Crit Care 27:491. https://doi.org/10.1186/s13054-023-04745-7
Vammen L, Johannsen CM, Baltsen CD et al (2023) Thiamine for the treatment of cardiac arrest-induced neurological injury: a randomized, blinded. Placebo-Controlled Experimental Study JAHA 12:e028558. https://doi.org/10.1161/JAHA.122.028558
Pooth J-S, Brixius SJ, Scherer C et al (2023) Limiting calcium overload after cardiac arrest: the role of human albumin in controlled automated reperfusion of the whole body. Perfusion 38:622–630. https://doi.org/10.1177/02676591211073779
Wang L, Sun Y, Kong F et al (2023) Mild hypothermia alleviates complement C5a-induced neuronal autophagy during brain ischemia-reperfusion injury after cardiac arrest. Cell Mol Neurobiol 43:1957–1974. https://doi.org/10.1007/s10571-022-01275-8
Diao M, Xu J, Wang J et al (2022) Alda-1, an activator of ALDH2, improves postresuscitation cardiac and neurological outcomes by inhibiting pyroptosis in swine. Neurochem Res 47:1097–1109. https://doi.org/10.1007/s11064-021-03511-x
Shen R, Pan D, Wang Z et al (2021) The effects of dexmedetomidine post-conditioning on cardiac and neurological outcomes after cardiac arrest and resuscitation in swine. Shock 55:388–395. https://doi.org/10.1097/SHK.0000000000001637
Boissady E, Abi Zeid Daou Y, Faucher E et al (2023) High-mobility group box 1–signaling inhibition with glycyrrhizin prevents cerebral t-cell infiltration after cardiac arrest. JAHA 12:e027749. https://doi.org/10.1161/JAHA.122.027749
Acknowledgements
We would like to thank Irene Possenti for her valuable assistance in developing the search terms and drafting the PubMed search string.
Funding
Not applicable.
Author information
Authors and Affiliations
Contributions
F.M.: conceptualisation, investigation, data validation, visualisation, and writing—original draft; F.F. and C.P.: conceptualisation, visualisation, and writing—review and editing; M.C.: visualisation, data validation, and review and editing; D.D.G., A.M., G.M., and A.S: visualisation, and review and editing; G.R. and E.R.Z.: conceptualisation, supervision, and review and editing. All authors read and approved the final version of the manuscript.
Corresponding author
Ethics declarations
Ethics approval and consent to participate
Not applicable.
Consent for publication
Not applicable.
Competing interests
Not applicable.
Additional information
Publisher's Note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Rights and permissions
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/.
About this article
Cite this article
Motta, F., Cerrato, M., De Giorgio, D. et al. Translational approach to assess brain injury after cardiac arrest in preclinical models: a narrative review. ICMx 13, 3 (2025). https://doi.org/10.1186/s40635-024-00710-y
Received:
Accepted:
Published:
DOI: https://doi.org/10.1186/s40635-024-00710-y