Next Article in Journal
High-Density Lipoproteins at the Interface between the NLRP3 Inflammasome and Myocardial Infarction
Next Article in Special Issue
Targeting Mitochondrial Dynamics during Lower-Limb Ischemia Reperfusion in Young and Old Mice: Effect of Mitochondrial Fission Inhibitor-1 (mDivi-1)
Previous Article in Journal
Genotype Combinations Drive Variability in the Microbiome Configuration of the Rhizosphere of Maize/Bean Intercropping System
Previous Article in Special Issue
Sulforaphane Is Protective against Warm Ischemia/Reperfusion Injury and Partial Hepatectomy in Rats
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Perspective

Ischemia-Reperfusion Programming of Alzheimer’s Disease-Related Genes—A New Perspective on Brain Neurodegeneration after Cardiac Arrest

by
Ryszard Pluta
* and
Stanisław J. Czuczwar
Department of Pathophysiology, Medical University of Lublin, 20-090 Lublin, Poland
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2024, 25(2), 1291; https://doi.org/10.3390/ijms25021291
Submission received: 29 December 2023 / Revised: 17 January 2024 / Accepted: 18 January 2024 / Published: 20 January 2024

Abstract

:
The article presents the latest data on pathological changes after cerebral ischemia caused by cardiac arrest. The data include amyloid accumulation, tau protein modification, neurodegenerative and cognitive changes, and gene and protein changes associated with Alzheimer’s disease. We present the latest data on the dysregulation of genes related to the metabolism of the amyloid protein precursor, tau protein, autophagy, mitophagy, apoptosis, and amyloid and tau protein transport genes. We report that neuronal death after cerebral ischemia due to cardiac arrest may be dependent and independent of caspase. Moreover, neuronal death dependent on amyloid and modified tau protein has been demonstrated. Finally, the results clearly indicate that changes in the expression of the presented genes play an important role in acute and secondary brain damage and the development of post-ischemic brain neurodegeneration with the Alzheimer’s disease phenotype. The data indicate that the above genes may be a potential therapeutic target for brain therapy after ischemia due to cardiac arrest. Overall, the studies show that the genes studied represent attractive targets for the development of new therapies to minimize ischemic brain injury and neurological dysfunction. Additionally, amyloid-related genes expression and tau protein gene modification after cerebral ischemia due to cardiac arrest are useful in identifying ischemic mechanisms associated with Alzheimer’s disease. Cardiac arrest illustrates the progressive, time- and area-specific development of neuropathology in the brain with the expression of genes responsible for the processing of amyloid protein precursor and the occurrence of tau protein and symptoms of dementia such as those occurring in patients with Alzheimer’s disease. By carefully examining the common genetic processes involved in these two diseases, these data may help unravel phenomena associated with the development of Alzheimer’s disease and neurodegeneration after cerebral ischemia and may lead future research on Alzheimer’s disease or cerebral ischemia in new directions.

1. Cardiac Arrest

Cardiac arrest occurs as a result of a sudden stop of the heartbeat and its mechanical activity, which causes cessation of systemic circulation and blood flow in the brain, which triggers global brain ischemia [1,2]. A comprehensive analysis of studies conducted in Europe, China, and the USA showed that the average age of patients who suffered cardiac arrest was 60–66 years, and 58–81% of them were men [3,4,5]. However, rates of mortality, depression, anxiety, and cognitive impairment were higher in women than in men [6,7]. The incidence of cardiac arrest worldwide exceeds 3.7 million annually [8,9]. Among others, in Europe there are 275,000, in the USA 450,000, in Taiwan 10,000, of whom about 10–15% survive until discharge from hospital [10,11,12,13]. The annual incidence of in-hospital cardiac arrest is estimated at 35–55 per 100,000 in Europe and the USA and 28–43 per 100,000 in Asia [9,14,15,16]. In Europe, based on data from the EuReCa ONE and EuReCa TWO studies, the annual incidence of out-of-hospital cardiac arrest was estimated at 84 per 100,000 patients and 89 per 100,000 patients, respectively [17,18], with a moderate average survival rate of 8% [18,19]. In European countries, it is estimated that 56–62 people per 100,000 are treated by emergency services for out-of-hospital cardiac arrest each year [15,18]. In Latin America and Colombia, the epidemiological characteristics and treatment outcomes of patients with out-of-hospital cardiac arrest are similar to those described in the world literature [20]. Similar statistics apply to the United States, Australia, and New Zealand [21,22]. Generally, the return of spontaneous circulation after cardiac arrest ranges from 33.7 to 35.5% [5,23]. Male gender and advanced age have been shown to be independent factors for failure to return spontaneous circulation after cardiac arrest [3,4,5].
Patients who are successfully resuscitated suffer from cerebral hypoxia and ischemia, which is the main cause of side-effects and death after admission to the intensive care unit [24]. Brain damage after cardiac arrest is the leading cause of death in patients resuscitated after cardiac arrest and the leading cause of long-term disability in those who survive the acute phase [25,26]. Therefore, cardiopulmonary resuscitation is the most important therapeutic activity that restores life to patients after cardiac arrest [1,2]. Despite progress in the use of cardiopulmonary resuscitation and targeted brain temperature control by lowering the head temperature to 32–34 °C [12,27,28,29], only 10–15% of patients after cardiac arrest survive until discharge from the hospital [12,13]. Therefore, cardiac arrest and cardiopulmonary resuscitation constitute a serious challenge for emergency physicians around the world and their actions are crucial for patients’ survival. Two key prognostic indicators that are extremely important in post-cardiac arrest are the return of spontaneous systemic circulation and the patient’s neurological status after resuscitation. Unfortunately, cardiac arrest is characterized by a high incidence, limited return of spontaneous systemic circulation, poor neurological outcomes, and poor survival to discharge. Despite significant progress in pre-hospital and in-hospital care, sudden cardiac arrest is still characterized by high morbidity and mortality. Therefore, the impact of cardiac arrest on the quality of life is of great importance worldwide. We believe that the presented article on similar brain neurodegeneration following cardiac arrest and Alzheimer’s disease will both inform and stimulate further interest in the mechanisms of changes and the development of causal treatments for dementia after cerebral ischemia from cardiac arrest and in Alzheimer’s disease. Therefore, there is an urgent need to search for new molecular mechanisms and therapeutic strategies for patients after cardiac arrest. While the effect of cardiac arrest on the brain is obvious, the mechanisms underlying this process remain unclear [30]. The paper presents the neurodegeneration of the brain after cardiac arrest, with particular emphasis on the occurrence of cognitive disorders and dementia, and with attention to new proteomic and genomic data.

2. Brain Neurodegeneration after Cardiac Arrest

Brain neuropathology after cardiac arrest includes primary ischemic injury and secondary reperfusion injury, which occur sequentially, acutely during cardiac arrest and resuscitation, and chronically in the post-resuscitation stage [24]. Transient global brain ischemia resulting from cardiac arrest in humans and animals causes neurodegeneration of neurons in the hippocampus, brain cortex, amygdala, basal ganglia, thalamus, dorsal and lateral septum, olfactory tubercle, primary olfactory cortex, entorhinal cortex, and brainstem [2,13,30,31,32,33,34,35,36]. The hippocampus is one of the brain regions most susceptible to ischemia after cardiac arrest in humans [37,38,39] and in animals [40,41,42,43]. Transient global brain ischemia causes selective neurodegeneration in the CA1 area of the hippocampus in humans and animals within 2–7 days after reperfusion [30,42]. Two years after ischemia in animals, in addition to neuronal loss in ischemia-sensitive areas, various stages of neuronal pathology were also observed in other areas [42]. Acute and chronic neuronal changes have been demonstrated in brain areas unrelated to primary ischemic pathology, i.e., hippocampal areas CA2, CA3, and CA4 [42]. In the hippocampus, activation of glial cells precedes neuronal loss and continues for a long time after an ischemic event in animals [44,45] and humans [46]. In the brains of humans and animals after ischemia, changes in the white matter combined with the proliferation of glial cells have been documented [42,44,45]. Autopsy of brains after experimental ischemia with a survival time of up to 2 years and in humans after ischemia showed severe hippocampal atrophy [42,47,48]. These neuropathological alterations have been clearly associated with progressive cognitive decline in humans [48,49,50] as well as in animals [51,52].
Brain ischemia caused by cardiac arrest increases the permeability of the blood–brain barrier in humans [53] as well as animals to cellular and non-cellular blood elements, i.e., platelets [54] and amyloids [55]. In the case of post-ischemic blood–brain barrier leakage, two facts deserve attention: the first is related to the passage of amyloid into the brain, and the second is the penetration of, among others, platelets containing huge amounts of soluble amyloid, which causes additional neurotoxic damage to the brain parenchyma [42,56]. Additionally, the permeability of the blood–brain barrier was influenced by oxidative stress, neuroinflammation [45,46,57], and the LRP1 and RAGE genes related to amyloid transport, described in detail later in the work. Soluble amyloid is delivered to the brain after cardiac arrest from the circulatory system [4,58] and additionally contributes to brain vessel vasoconstriction, amyloidosis, and cerebral amyloid angiopathy [49,50].
After brain ischemia, hippocampal atrophy was demonstrated with simultaneous neurodegenerative damage to the cortex of the temporal lobe [42,49,50]. Neuronal death after cerebral ischemia due to cardiac arrest may be dependent and independent of caspase. Moreover, neuronal death dependent on amyloid and modified tau protein has been demonstrated. Current preclinical and clinical evidence indicates that cardiac arrest causes chronic neuroinflammatory responses in the brain, which further exacerbate neurodegenerative changes [44,45,46]. Particularly extensive microglial activation and neurodegeneration in the CA1 area of the hippocampus in humans and animals are evident after cardiac arrest and lead to serious neurological sequelae [45,46,57].

3. Cognitive Deficits after Cardiac Arrest

The most sensitive brain regions to ischemic damage from cardiac arrest are the hippocampus, brain cortex, basal ganglia, thalamus, and amygdala [13,32,47]. These areas are closely related to cognitive domains (Figure 1) [13,51,52]. Cognitive functions depend on complex interactions between cortical and subcortical areas through various brain networks (Figure 1) [13]. Cognitive problems develop in 42–50% of patients who survive cardiac arrest up to several years after discharge from hospital [13,59]. Other studies have reported that in people who survived cardiac arrest, 50–100% of them experienced cognitive, mood, and functioning disorders [6,7,60]. Even among patients who returned to good neurological condition after discharge from the hospital, 29% experienced memory problems (short-term memory and spatial or contextual memory) and 43% experienced cognitive impairment [28,59].
The post-ischemic hippocampus is believed to be the main structure underlying episodic memory impairment, which is the earliest and most visible clinical symptom preceding the development of dementia (Figure 1) [42,48,49,50,51]. Ischemia causes damage to the temporal cortex, which is the target area of the main axon output network from the hippocampus, so these areas are structurally and functionally related to each other and are important for learning and memory processes [13,49,50]. Thus, existing evidence indicates that cognitive and functional impairment after cardiac arrest is one of the important areas of concern for physicians [61,62,63,64,65,66,67]. The primary goal of research in the past was to improve survival after cardiac arrest. Currently, in clinical conditions, there is a search for methods that can improve neurological outcomes. Therefore, it is not surprising that research has moved beyond prognostication in the acute period following successful cardiopulmonary resuscitation to identifying mechanisms of long-term brain damage. Another currently promising area of research is the implementation of therapies that can reduce neuronal damage resulting from cerebral hypoxia and ischemia. It should be added that it is unlikely that survivors could be considered fully recovered immediately after being discharged from the hospital. Typically, sooner or later during long-term follow-up, they develop cognitive deficits, mainly related to memory and executive functions, or mild to severe impairment, including memory loss, deterioration of psychomotor, executive, and visuospatial functions, and emotional problems, including anxiety and depression [28,68,69,70]. The above-mentioned disorders affect the vast majority of patients after cardiac arrest—estimated to be as many as 88% [71]. The most frequently stated cognitive deficit is memory loss in patients who regain consciousness after cardiac arrest. It should be noted that memory with delayed recall and recognition is usually most affected, while isolated memory loss is rare. It can be summarized that the typical pattern of impairment after cardiac arrest includes deficits in memory, fine motor skills, and in executive functions [72]. According to the latest recommendations, before leaving the hospital, patients should be thoroughly assessed for short- and long-term memory deficits, cognitive and executive dysfunction, depression, and possible progression to dementia [62]. Overall, the prognosis for recovery of cognitive function to pre-arrest levels is uncertain. Despite uncertain prognoses, long-term rehabilitation has been shown to improve overall performance across a wide range of activities and should be pursued [73]. In most cases, patients are unable to return to daily activities within the first year after injury, and deficiencies are detected up to 8 years after cardiac arrest [74,75]. A study from Sweden, with an extremely long-term follow-up of 17 years after cardiac arrest, found a trend towards lower scores on cognitive tests and lower self-reported quality of life [71]. The authors concluded that survivors of cardiac arrest may experience permanent cognitive impairment, progressing to Alzheimer’s disease-type dementia [71,76]. It is suggested that the above-mentioned population is at increased risk of dementia due to the hypoxia occurring during cardiac arrest. It has been noted that the progression of cognitive decline following cardiac arrest in humans is characterized by greater memory impairment than executive dysfunction, making it similar to the prodromal form of Alzheimer’s disease dementia [76].
Cardiac arrest in humans and animals has been shown to significantly increase the risk of mild cognitive impairment, vascular cognitive impairment, and dementia, and dementia of Alzheimer’s disease type [46,51,52,77,78]. More than a quarter of patients experienced cognitive impairment four years after cardiac arrest [46,79]. Cohort studies have shown a high prevalence (54.4%) of long-term cognitive deficits and functional limitations in cardiac arrest survivors [46,80], even in those with apparently favorable neurological outcomes [46,81].

4. Alzheimer’s Disease Related Genes after Cardiac Arrest

It has been shown that in the CA1 and CA3 regions of the hippocampus, as well as in the temporal cortex, genes associated with Alzheimer’s disease, such as these encoding α-secretase, amyloid precursor protein, β-secretase, presenilin 1 and 2, and tau protein are dysregulated as a result of ischemia caused by cardiac arrest (Table 1) [82]. Changes in gene expression in the CA1 area of the hippocampus include all genes examined at all time observations after cardiac arrest (Table 1) [82]. However, changes in gene expression in the CA3 area are less severe, do not occur at all times after ischemia, and are delayed in time in relation to the CA1 region (Table 1) [82]. Specifically, changes in gene expression in the temporal cortex occurred immediately after cardiac arrest but did not occur at all times after ischemia and did not affect all genes (Table 1) [82]. The results indicate that a brain ischemic episode caused by cardiac arrest is a trigger of amyloidogenic processes in brain parenchyma [42,47,83,84,85]. After cardiac arrest, amyloidogenic processing of amyloid protein precursor was observed with varying intensity depending on the structure. The consequences are additional changes in ischemic neurons depending on the toxic properties of amyloid. This creates conditions for the accumulation of amyloid plaques in the brain as in Alzheimer’s disease.

5. Amyloid and Tau Protein in Humans after Cardiac Arrest

Studies of human brains after cardiac arrest have shown intense accumulation of amyloid in the brain parenchyma [86]. The presence of both diffuse and senile amyloid plaques has been demonstrated in areas of the brain susceptible to ischemia, in the border zones of arteries, and in the brain cortex [86]. Intense amyloid accumulation was noted in the middle layers of the brain cortex, which are sensitive to ischemia [86]. In the brains of patients with survival up to 1 month after cardiac arrest, strong amyloid staining was found in neurons and perivascular areas [86]. The neurons of the brain cortex and hippocampus stained most intensively for amyloid. Ependymal and epithelial cells also stained for amyloid. The brain’s gray and white matter vessels were surrounded by numerous amyloid deposits that looked like cuffs. In some brains, the walls of meningeal and cortical vessels were stained for amyloid. The accumulation of amyloid in the perivascular spaces of the blood–brain barrier vessels suggests that the amyloid originated from the blood. Evidence supporting this hypothesis comes from clinical studies indicating the presence of significantly elevated blood amyloid levels in patients after cardiac arrest [4,58]. The level of amyloid increase correlated negatively with clinical outcome after cerebral ischemia, and this, in turn, likely indicates the degree of brain damage [4,58]. These data confirm that brain ischemia resulting from cardiac arrest may play a key role in the amyloidogenic metabolism of the amyloid precursor protein.
Tau protein was detected in the blood of patients after complete brain ischemia due to cardiac arrest with two peaks on the second and fourth day, which probably indicates the degree of neuronal injury [4,87]. The detected two-phase changes in the level of tau protein in the blood correlate with two types of neuronal death after ischemia, as a result of necrosis and delayed neuronal death [88]. The presented studies show that the concentration of tau protein in the serum may be a biomarker for assessing brain damage after cardiac arrest [87,88,89,90].

6. Amyloid and Tau Protein in Animals after Cardiac Arrest

After experimental brain ischemia resulting from cardiac arrest with survival of 1 year, various fragments of amyloid precursor protein were found in the extra- and intracellular spaces [42,47,83]. In rats that survived up to 0.5 years after brain ischemia, N- and C-terminal deposits of amyloid precursor protein and amyloid were visible in the extracellular space of the hippocampus, brain cortex, white matter, and around the lateral ventricles [42,83]. Various parts of the amyloid precursor protein have also been found to accumulate in neuronal cells, neuroglia, endothelium, pericytes, and ependymal cells [42,47,85]. In particular, astrocytes around the microvessels showed intense staining of very long, thin processes that adhered to or surrounded the capillaries. However, after 0.5 years of post-ischemic survival, only staining of the C-terminal of the amyloid precursor protein and amyloid was noted [42]. Amyloid accumulation in response to reversible brain ischemia was not a transient phenomenon, as diffuse amyloid deposits converted to amyloid plaques in animals with 1-year survival [84].
Evidence shows that after ischemia, neuronal cells are dominated by amyloid accumulation and hyperphosphorylated tau protein, which accompany apoptosis [42,91,92,93]. It is also evident that paired helical filaments [94], neurofibrillary tangles-like [95], and neurofibrillary tangles were observed in neurons after ischemia [96,97,98]. Data indicate that brain ischemia additionally activates neuronal death in an amyloid- and tau-protein-dependent manner [96,97,98].

7. Transport Genes of Amyloid and Tau Protein in the Brain after Cardiac Arrest

The expression of the receptor for the advanced glycation end products (RAGE) gene increases significantly in CA3 area during 7–30 days after ischemia, and at 12–24 months its expression gradually and significantly decreases (Table 2) [99]. Regarding the expression of the low-density lipoprotein receptor-related protein 1 (LRP1) gene, the opposite is true (Table 2). In the early phase post-ischemia (2–7 days), a significant decrease in its expression was observed [99]. Then, 12–24 months after cardiac arrest, its expression gradually increased and was statistically significant [99]. In other words, in the early phase after ischemia, 7–30 days, the expression of the gene RAGE transporting amyloid and tau protein to the brain increased with increase in the neurotoxic activity of amyloid and tau protein. Conversely, in the late phase after ischemia, i.e., 12–24 months, the activity of the amyloid clearing gene LRP1 increased spontaneously in the CA3 area, reducing and/or preventing further neuronal injury or facilitating healing (Table 2).
In the brains of patients after ischemia caused by cardiac arrest, immunostaining for the receptor for advanced glycation end products was localized in the epithelial cells of the choroid plexus as well as in the cells of the lining of the brain ventricles [100]. The above cells form the cerebrospinal fluid–brain barrier and the blood–cerebrospinal fluid barrier. In these cases, amyloid staining was observed in the blood vessels of the choroid plexus and the basement membrane of the choroid plexus epithelium [100]. These data indicate that the choroid plexus epithelium and cells lining the ventricles are equipped with the advanced glycation end-product receptor and play a significant role in the transport and accumulation of amyloid in the brain parenchyma.

8. Genes Involved in Neuronal Death in Cardiac Arrest and Alzheimer’s Disease

Autophagy (BECN1) gene expression in the CA1 area of the hippocampus after cardiac arrest with survival at 2, 7, and 30 days was within control limits (Table 3) [101]. However, mitophagy (BNIP3) gene expression 2 days after ischemia in CA1 was above the control value, and on days 7 and 30 after cardiac arrest, it was within the control range (Table 3). As for the expression of the apoptosis (caspase 3) gene in the CA1 area, its overexpression was found after 2 and 7 days of survival, while on the 30th day after cardiac arrest, the expression was below the control value (Table 3) [101].
BECN1 gene expression in CA3 increased significantly on day 30 after cardiac arrest (Table 3) [102]. However, BNIP3 gene expression after cardiac arrest was below control values for 2–30 days of observation (Table 3) [102]. Studies of the caspase 3 gene showed increased activity on days 7 and 30 after cardiac arrest (Table 3) [102].
BECN1 gene expression in the temporal cortex 2 days after cardiac arrest was above the control value, while after 7 and 30 days it oscillated around control values (Table 3) [103]. However, 2 days after cardiac arrest, the expression of the BNIP3 gene was below the control value, and after 7 and 30 days its expression increased significantly above the control value (Table 3) [103]. As for the expression of the caspase 3 gene in this structure, 2 days after cardiac arrest, it was below the control value, and on days 7 and 30, its expression was above the control value (Table 3) [103].

9. Discussion

Harvested evidence suggests that there is a remarkable parallelism between the neuropathogenesis of Alzheimer’s disease and brain ischemia in animals and humans resulting from cardiac arrest [42,104,105,106,107,108,109,110,111]. Both the Alzheimer’s disease and post-cardiac arrest brain are characterized by neuronal death, blood–brain barrier permeability, neuroinflammation, amyloid plaques, neurofibrillary tangles, cerebral amyloid angiopathy, and hippocampal and brain atrophy [42,44,45,49,50,84,85,93,96,97]. Collectively, these types of evidence point to common proteomic and genomic mechanisms of cerebral ischemia caused by cardiac arrest and Alzheimer’s disease [42,44,45,82,84,85,96,97,98].
After cardiac arrest, neuronal death is observed in the hippocampus, cerebral cortex, basal ganglia, thalamus, and amygdala, structures responsible for memory, as well as general brain atrophy and, ultimately, progressive dementia of Alzheimer’s disease phenotype (Figure 1) [51,52,59,77]. One proposal explaining the above changes suggests that long non-coding RNA increases the expression of the adapter protein ShcA in the hippocampus, which causes cognitive impairment after cardiac arrest as a result of neuroinflammation and apoptosis of pyramidal neurons [112,113,114]. Another hypothesis links hypoxia to molecular abnormalities of the cerebral cortex resembling Alzheimer’s disease, which may facilitate the development of this dementia phenotype. It has been shown that transient hypoxia is associated with hyperphosphorylation of cytoskeletal proteins and abnormalities in the functioning of neuronal mitochondria, which may lead to Alzheimer’s disease-like changes and may also cause an increase in the level of amyloid, which is an important element in the pathology of Alzheimer’s disease [115]. Yet another possible pathway leading to the pathogenesis of a post-ischemic Alzheimer’s disease phenotype is dysregulation of the expression of genes that metabolize the amyloid precursor protein to amyloid and modification of the tau protein, observed after global cerebral ischemia in an experimental model of cardiac arrest [116,117,118,119,120]. Brain ischemia is the second naturally occurring brain pathology after Alzheimer’s disease, causing the death of pyramidal neurons in the hippocampus [42]. An increase in amyloid levels in the brain and blood indicates activation of the amyloidogenic pathway in response to acute cerebral ischemia caused by cardiac arrest. The above observations allow us to suggest that brain ischemia may be the cause of Alzheimer’s disease [42,45,93,104,105,107,108,109,110,111].
Currently, a small group of scientists specifically proposes the involvement of ischemia in the development of Alzheimer’s disease at three levels: cellular, brain, and patient [42,104,105,106,107,108,109,110,111]. They discuss the involvement of ischemia in Alzheimer’s disease as dysregulation of neurovascular coupling, reduced cerebral blood flow, blood–brain barrier permeability, and reduced amyloid clearance from the brain. Finally, they present a two-step etiology of Alzheimer’s disease that involves a two-step positive feedback loop in which cerebral microvascular damage(s) precedes the production and accumulation of amyloid and tau protein abnormalities in the brain [42,104,105,106,107,108,109,110,111].

10. Conclusions

In previous studies, the main goal was to increase survival after cardiac arrest. Unfortunately, the current high incidence of cognitive impairment observed in cardiac arrest survivors remains a significant challenge for both the research and clinical communities. It seems certain that future research should focus on the prevention of neurological damage and effective therapeutic approaches that could improve neurological outcomes after cardiac arrest. Future research efforts should focus on finding pharmacological and nonpharmacological interventions to improve cognitive function, mental health, and quality of life in cardiac arrest survivors. The conclusions drawn from the study of Alzheimer’s disease-related proteins after cardiac arrest and their genes in the hippocampus and temporal cortex, which contribute to neuronal death, amyloid production, and the formation of neurofibrillary tangles, are important for the development of therapeutic targets also in the treatment of Alzheimer’s disease.
Finally, the results clearly indicate that changes in the expression of the presented genes play an important role in acute and secondary brain damage and the development of post-ischemic brain neurodegeneration with the Alzheimer’s disease phonotype. The data indicate that the above genes may be a potential therapeutic target for brain therapy after ischemia due to cardiac arrest. Overall, the studies show that the genes studied represent attractive targets for the development of new therapies to minimize ischemic brain injury and neurological dysfunction. Additionally, amyloid-related genes expression and tau protein gene modification after cerebral ischemia due to cardiac arrest are useful in identifying ischemic mechanisms associated with Alzheimer’s disease. Cardiac arrest illustrates the progressive, time- and area-specific development of neuropathology in the brain with the expression of genes responsible for the processing of amyloid protein precursor and the occurrence of tau protein and symptoms of dementia, such as those occurring in patients with Alzheimer’s disease. By carefully examining the common genetic processes involved in these two diseases, these data may help unravel phenomena associated with the development of Alzheimer’s disease and neurodegeneration after cerebral ischemia and may lead future research on Alzheimer’s disease or cerebral ischemia in new directions.

Author Contributions

Conceptualization, R.P.; methodology, S.J.C.; software, S.J.C.; validation, R.P. and S.J.C.; formal analysis, R.P.; investigation, R.P. and S.J.C.; resources, R.P. and S.J.C.; data curation, R.P. and S.J.C.; writing—original draft preparation, R.P.; writing—review and editing, R.P. and S.J.C.; visualization, R.P.; supervision, R.P.; project administration, R.P.; funding acquisition, S.J.C. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Acknowledgments

The authors acknowledge the support of the Medical University of Lublin, Poland (DS 721/22-SJC).

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Stub, D.; Bernard, S.; Duffy, S.J.; Kaye, D.M. Post cardiac arrest syndrome: A review of therapeutic strategies. Circulation 2011, 123, 1428–1435. [Google Scholar] [CrossRef] [PubMed]
  2. Sandroni, C.; Cronberg, T.; Sekhon, M. Brain injury after cardiac arrest: Pathophysiology, treatment, and prognosis. Intensiv. Care Med. 2021, 47, 1393–1414. [Google Scholar] [CrossRef] [PubMed]
  3. Andersen, L.W.; Holmberg, M.J.; Berg, K.M.; Donnino, M.W.; Granfeldt, A. In-hospital cardiac arrest: A review. JAMA 2019, 321, 1200–1210. [Google Scholar] [CrossRef] [PubMed]
  4. Ashton, N.J.; Moseby-Knappe, M.; Benedet, A.L.; Grötschel, L.; Lantero-Rodriguez, J.; Karikari, T.K.; Hassager, C.; Wise, M.P.; Stammet, P.; Kjaergaard, J.; et al. Alzheimer Disease Blood Biomarkers in Patients with Out-of-Hospital Cardiac Arrest. JAMA Neurol. 2023, 80, 388–396. [Google Scholar] [CrossRef] [PubMed]
  5. Li, Z.; Xing, J. A model for predicting return of spontaneous circulation and neurological outcomes in adults after in-hospital cardiac arrest: Development and evaluation. Front. Neurol. 2023, 14, 1323721. [Google Scholar] [CrossRef] [PubMed]
  6. Green, C.R.; Botha, J.A.; Tiruvoipati, R. Cognitive function, quality of life and mental health in survivors of our-of-hospital cardiac arrest: A review. Anaesth. Intensive Care 2015, 43, 568–576. [Google Scholar] [CrossRef]
  7. Glimmerveen, A.; Verhulst, M.; Verbunt, J.; Van Heugten, C.; Hofmeijer, J. Predicting Long-Term Cognitive Impairments in Survivors after Cardiac Arrest: A Systematic Review. J. Rehabil. Med. 2023, 55, jrm00368. [Google Scholar] [CrossRef]
  8. Mehra, R. Global public health problem of sudden cardiac death. J. Electrocardiol. 2007, 40, S118–S122. [Google Scholar] [CrossRef]
  9. Jung, E.; Park, J.H.; Ro, Y.S.; Ryu, H.H.; Cha, K.C.; Do Shin, S.; Hwang, S.O. Cardiac Arrest Pursuit Trial with Unique Registration, Epidemiologic Surveillance (CAPTURES) project investigators. Family history, socioeconomic factors, comorbidities, health behaviors, and the risk of sudden cardiac arrest. Sci. Rep. 2023, 13, 21341. [Google Scholar] [CrossRef]
  10. Wissenberg, M.; Lippert, F.K.; Folke, F.; Weeke, P.; Hansen, C.M.; Christensen, E.F. Association of national initiatives to improve cardiac arrest management with rates of bystander intervention and patient survival after out-of-hospital cardiac arrest. JAMA 2013, 310, 1377–1384. [Google Scholar] [CrossRef]
  11. Wang, C.Y.; Wang, J.Y.; Teng, N.C.; Chao, T.T.; Tsai, S.L.; Chen, C.L.; Hsu, J.Y.; Wu, C.P.; Lai, C.C.; Chen, L. The secular trends in the incidence rate and outcomes of out-of-hospital cardiac arrest in Taiwan-A nationwide population-based study. PLoS ONE 2015, 10, e0122675. [Google Scholar] [CrossRef] [PubMed]
  12. Hosseini, M.; Wilson, R.H.; Crouzet, C.; Amirhekmat, A.; Wei, K.S.; Akbari, Y. Resuscitating the Globally Ischemic Brain: TTM and Beyond. Neurotherapeutics 2020, 17, 539–562. [Google Scholar] [CrossRef] [PubMed]
  13. Hagberg, G.; Ihle-Hansen, H.; Sandset, E.C.; Jacobsen, D.; Wimmer Hand Ihle-Hansen, H. Long Term Cognitive Function after Cardiac Arrest: A Mini-Review. Front. Aging Neurosci. 2022, 14, 885226. [Google Scholar] [CrossRef] [PubMed]
  14. de Vreede-Swagemakers, J.J.; Gorgels, A.P.; Dubois-Arbouw, W.I.; van Ree, J.W.; Daemen, M.J.; Houben, L.G.; Wellens, H.J. Out-of-hospital cardiac arrest in the 1990’s: A population-based study in the Maastricht area on incidence, characteristics and survival. J. Am. Coll. Cardiol. 1997, 30, 1500–1505. [Google Scholar] [CrossRef] [PubMed]
  15. Berdowski, J.; Berg, R.A.; Tijssen, J.G.; Koster, R.W. Global incidences of out-of-hospital cardiac arrest and survival rates: Systematic review of 67 prospective studies. Resuscitation 2010, 81, 1479–1487. [Google Scholar] [CrossRef] [PubMed]
  16. Hwang, S.O.; Cha, K.C.; Jung, W.J.; Roh, Y.I.; Kim, T.Y.; Chung, S.P.; Kim, Y.M.; Park, J.D.; Kim, H.S.; Lee, M.J.; et al. Steering Committee of the 2020 Korean Guidelines for Cardiopulmonary Resuscitation and Emergency Cardiovascular Care. 2020 Korean Guidelines for Cardiopulmonary Resuscitation. Part 2. Environment for cardiac arrest survival and the chain of survival. Clin. Exp. Emerg. Med. 2021, 8, S8–S14. [Google Scholar] [CrossRef]
  17. Gräsner, J.T.; Lefering, R.; Koster, R.W.; Masterson, S.; Böttiger, B.W.; Herlitz, J.; Wnent, J.; Tjelmeland, I.B.; Ortiz, F.R.; Maurer, H.; et al. EuReCa ONE-27 Nations, ONE Europe, ONE Registry: A prospective one month analysis of out-of-hospital cardiac arrest outcomes in 27 countries in Europe. Resuscitation 2016, 105, 188–195. [Google Scholar] [CrossRef]
  18. Gräsner, J.T.; Wnent, J.; Herlitz, J.; Perkins, G.D.; Lefering, R.; Tjelmeland, I.; Koster, R.W.; Masterson, S.; Rossell-Ortiz, F.; Maurer, H.; et al. Survival after out-of-hospital cardiac arrest in Europe –Results of the EuReCa TWO study. Resuscitation 2020, 148, 218–226. [Google Scholar] [CrossRef]
  19. Nichol, G.; Thomas, E.; Callaway, C.W.; Hedges, J.; Powell, J.L.; Aufderheide, T.P.; Rea, T.; Lowe, R.; Brown, T.; Dreyer, J.; et al. Resuscitation Outcomes Consortium Investigators. Regional variation in out-of-hospital cardiac arrest incidence and outcome. JAMA 2008, 300, 1423–1431. [Google Scholar] [CrossRef]
  20. Medina, J.A.; Quintero, J.A.; de Paz, D.A.; Scarpetta, D.F.; Castro, C.A.; Paker, N.A.; Carvajal, S.M. Cardiac arrest in an emergency department in Colombia during 2011–2020: A descriptive study. Int. J. Crit. Illn. Inj. Sci. 2023, 13, 132–137. [Google Scholar]
  21. Beck, B.; Bray, J.; Cameron, P.; Smith, K.; Walker, T.; Grantham, H.; Hein, C.; Thorrowgood, M.; Smith, A.; Inoue, M.; et al. Aus-ROC Steering Committee. Regional variation in the characteristics, incidence and outcomes of out-of-hospital cardiac arrest in Australia and New Zealand: Results from the Aus-ROC Epistry. Resuscitation 2018, 126, 49–57. [Google Scholar] [CrossRef] [PubMed]
  22. Benjamin, E.J.; Muntner, P.; Alonso, A.; Bittencourt, M.S.; Callaway, C.W.; Carson, A.P.; Chamberlain, A.M.; Chang, A.R.; Cheng, S.; Das, S.R.; et al. Heart disease and stroke statistics—2019 update: A report from the American Heart Association. Circulation 2019, 139, e56–e528. [Google Scholar] [CrossRef] [PubMed]
  23. Shao, F.; Li, C.S.; Liang, L.R.; Qin, J.; Ding, N.; Fu, Y.; Yang, K.; Zhang, G.Q.; Zhao, L.; Zhao, B.; et al. Incidence and outcome of adult in-hospital cardiac arrest in Beijing, China. Resuscitation 2016, 102, 51–56. [Google Scholar] [CrossRef] [PubMed]
  24. Sekhon, M.S.; Ainslie, P.N.; Griesdale, D.E. Clinical pathophysiology of hypoxic ischemic brain injury after cardiac arrest: A “two-hit” model. Crit. Care 2017, 21, 90. [Google Scholar] [CrossRef] [PubMed]
  25. Lemiale, V.; Dumas, F.; Mongardon, N.; Giovanetti, O.; Charpentier, J.; Chiche, J.D.; Carli, P.; Mira, J.P.; Nolan, J.; Cariou, A. Intensive care unit mortality after cardiac arrest: The relative contribution of shock and brain injury in a large cohort. Intensive Care Med. 2013, 39, 1972–1980. [Google Scholar] [CrossRef]
  26. Geocadin, R.G.; Callaway, C.W.; Fink, E.L.; Golan, E.; Greer, D.M.; Ko, N.U.; Lang, E.; Licht, D.J.; Marino, B.S.; McNair, N.D.; et al. Standards for studies of neurological prognostication in comatose survivors of cardiac arrest: A scientific statement from the American Heart Association. Circulation 2019, 140, e517–e542. [Google Scholar] [CrossRef] [PubMed]
  27. Varon, J.; Marik, P.E.; Einav, S. Therapeutic hypothermia: A state-of-the-art emergency medicine perspective. Am. J. Emerg. Med. 2012, 30, 800–810. [Google Scholar] [CrossRef]
  28. Lilja, G.; Nielsen, N.; Friberg, H.; Horn, J.; Kjaergaard, J.; Nilsson, F.; Pellis, T.; Wetterslev, J.; Wise, M.P.; Bosch, F.; et al. Cognitive function in survivors of out-of-hospital cardiac arrest after target temperature management at 33 °C versus 36 °C. Circulation 2015, 131, 1340–1349. [Google Scholar] [CrossRef]
  29. Ahn, J.H.; Lee, T.K.; Tae, H.J.; Kim, B.; Sim, H.; Lee, J.C.; Kim, D.W.; Kim, Y.S.; Shin, M.C.; Park, Y.; et al. Neuronal Death in the CNS Autonomic Control Center Comes Very Early after Cardiac Arrest and Is Not Significantly Attenuated by Prompt Hypothermic Treatment in Rats. Cells 2021, 10, 60. [Google Scholar] [CrossRef]
  30. Weng, Y.C.; Huang, Y.T.; Chiang, I.C.; Chuang, H.C.; Lee, T.H.; Tan, T.H.; Chou, W.H. DUSP6 deficiency attenuates neurodegeneration after global cerebral ischemia. Int. J. Mol. Sci. 2023, 24, 7690. [Google Scholar] [CrossRef]
  31. Kagstrom, E.; Smith, M.L.; Siesjo, B.K. Local cerebral blood flow in the recovery period following complete cerebral ischemia in the rat. J. Cereb. Blood Flow Metab. 1983, 3, 170–182. [Google Scholar] [CrossRef] [PubMed]
  32. Blomqvist, P.; Wieloch, T. Ischemic brain damage in rats following cardiac arrest using a long-term recovery model. J. Cereb. Blood Flow Metab. 1985, 5, 420–423. [Google Scholar] [CrossRef] [PubMed]
  33. Kawai, K.; Nitecka, L.; Ruetzler, C.A.; Nagashima, G.; Joó, F.; Mies, G.; Nowak TSJr Saito, N.; Lohr, J.M.; Klatzo, I. Global cerebral ischemia associated with cardiac arrest in the rat: I. Dynamics of early neuronal changes. J. Cereb. Blood Flow Metab. 1992, 12, 238–249. [Google Scholar] [CrossRef] [PubMed]
  34. Krajewski, S.; Mai, J.K.; Krajewska, M.; Sikorska, M.; Mossakowski, M.J.; Reed, J.C. Upregulation of bax protein levels in neurons following cerebral ischemia. J. Neurosci. 1995, 15, 6364–6376. [Google Scholar] [CrossRef] [PubMed]
  35. Kawai, K.; Nowak, T.S., Jr.; Klatzo, I. Loss of parvalbumin immunoreactivity defines selectively vulnerable thalamic reticular nucleus neurons following cardiac arrest in the rat. Acta Neuropathol. 1995, 89, 262–269. [Google Scholar] [CrossRef] [PubMed]
  36. Böttiger, B.W.; Schmitz, B.; Wiessner, C.; Vogel, P.; Hossmann, K.A. Neuronal stress response and neuronal cell damage after cardiocirculatory arrest in rats. J. Cereb. Blood Flow Metab. 1998, 18, 1077–1087. [Google Scholar] [CrossRef] [PubMed]
  37. Petito, C.K.; Feldmann, E.; Pulsinelli, W.A.; Plum, F. Delayed hippocampal damage in humans following cardiorespiratory arrest. Neurology 1987, 37, 1281–1286. [Google Scholar] [CrossRef] [PubMed]
  38. Hu, B.R.; Liu, C.L.; Park, D.J. Alteration of MAP kinase pathways after transient forebrain ischemia. J. Cereb. Blood Flow Metab. 2000, 20, 1089–1095. [Google Scholar] [CrossRef]
  39. Sawe, N.; Steinberg, G.; Zhao, H. Dual roles of the MAPK/ERK1/2 cell signaling pathway after stroke. J. Neurosci. Res. 2008, 86, 1659–1669. [Google Scholar] [CrossRef]
  40. Sadowski, M.; Wisniewski, H.M.; Jakubowska-Sadowska, K.; Tarnawski, M.; Lazarewicz, J.W.; Mossakowski, M.J. Pattern of neuronal loss in the rat hippocampus following experimental cardiac arrest-induced ischemia. J. Neurol. Sci. 1999, 168, 13–20. [Google Scholar] [CrossRef]
  41. Sadowski, M.; Lazarewicz, J.W.; Jakubowska-Sadowska, K.; Wisniewski, H.M.; Mossakowski, M.J.; Brown, W.T. Long-term changes in calbindin D(28K) immunoreactivity in the rat hippocampus after cardiac arrest. Neurosci. Lett. 2002, 321, 90–94. [Google Scholar] [CrossRef] [PubMed]
  42. Pluta, R.; Ułamek, M.; Jabłoński, M. Alzheimer’s mechanisms in ischemic brain degeneration. Anat. Rec. 2009, 292, 1863–1881. [Google Scholar] [CrossRef] [PubMed]
  43. Warenits, A.M.; Hatami, J.; Müllebner, A.; Ettl, F.; Teubenbacher, U.; Magnet, I.A.M.; Bauder, B.; Janata, A.; Miller, I.; Moldzio, R.; et al. Motor Cortex and Hippocampus Display Decreased Heme Oxygenase Activity 2 Weeks after Ventricular Fibrillation Cardiac Arrest in Rats. Front. Med. 2020, 7, 513. [Google Scholar] [CrossRef] [PubMed]
  44. Sekeljic, V.; Bataveljic, D.; Stamenkovic, S.; Ułamek, M.; Jabłoński, M.; Radenovic, L.; Pluta, R.; Andjus, P.R. Cellular markers of neuroinflammation and neurogenesis after ischemic brain injury in the long-term survival rat model. Brain Struct. Funct. 2012, 217, 411–420. [Google Scholar] [CrossRef] [PubMed]
  45. Radenovic, L.; Nenadic, M.; Ułamek-Kozioł, M.; Januszewski, S.; Czuczwar, S.J.; Andjus, P.R.; Pluta, R. Heterogeneity in brain distribution of activated microglia and astrocytes in a rat ischemic model of Alzheimer’s disease after 2 years of survival. Aging 2020, 12, 12251–12267. [Google Scholar] [CrossRef] [PubMed]
  46. Traub, J.; Frey, A.; Störk, S. Chronic Neuroinflammation and Cognitive Decline in Patients with Cardiac Disease: Evidence, Relevance, and Therapeutic Implications. Life 2023, 13, 329. [Google Scholar] [CrossRef] [PubMed]
  47. Pluta, R. The role of apolipoprotein E in the deposition of β-amyloid peptide during ischemia–reperfusion brain injury. A model of early Alzheimer’s disease. Ann. N. Y. Acad. Sci. 2000, 903, 324–334. [Google Scholar] [CrossRef] [PubMed]
  48. Gemmell, E.; Tam, E.; Allan, L.; Hall, R.; Khundakar, A.; Oakley, A.E.; Thomas, A.; Deramecourt, V.; Kalaria, R.N. Neuron volumes in hippocampal subfields in delayed poststroke and aging-related dementias. J. Neuropathol. Exp. Neurol. 2014, 73, 305–311. [Google Scholar] [CrossRef]
  49. Goulay, R.; Mena Romo, L.; Hol, E.M.; Dijkhuizen, R.M. From stroke to dementia: A Comprehensive review exposing tight interactions between stroke and amyloid-β formation. Transl. Stroke Res. 2020, 11, 601–614. [Google Scholar] [CrossRef]
  50. Rost, N.S.; Brodtmann, A.; Pase, M.P.; van Veluw, S.J.; Biffi, A.; Duering, M.; Hinman, J.D.; Dichgans, M. Post-stroke cognitive impairment and dementia. Circ. Res. 2022, 130, 1252–1271. [Google Scholar] [CrossRef]
  51. Kiryk, A.; Pluta, R.; Figiel, I.; Mikosz, M.; Ulamek, M.; Niewiadomska, G.; Jablonski, M.; Kaczmarek, L. Transient brain ische-mia due to cardiac arrest causes irreversible long-lasting cognitive injury. Behav. Brain Res. 2011, 219, 1–7. [Google Scholar] [CrossRef] [PubMed]
  52. Cohan, C.H.; Neumann, J.T.; Dave, K.R.; Alekseyenko, A.; Binkert, M.; Stransky, K.; Lin, H.W.; Barnes, C.A.; Wright, C.B.; Perez Pinzon, M.A. Effect of cardiac arrest on cognitive impairment and hippocampal plasticity in middle-aged rats. PLoS ONE 2015, 10, e0124918. [Google Scholar] [CrossRef] [PubMed]
  53. Park, J.S.; You, Y.; Min, J.H.; Yoo, I.; Jeong, W.; Cho, Y.; Ryu, S.; Lee, J.; Kim, S.W.; Cho, S.U.; et al. Study on the timing of severe blood-brain barrier disruption using cerebrospinal fluid-serum albumin quotient in post cardiac arrest patients treated with targeted temperature management. Resuscitation 2019, 135, 118–123. [Google Scholar] [CrossRef] [PubMed]
  54. Pluta, R.; Lossinsky, A.S.; Walski, M.; Wisniewski, H.M.; Mossakowski, M.J. Platelet occlusion phenomenon after short- and long-term survival following complete cerebral ischemia in rats produced by cardiac arrest. J. Hirnforsch. 1994, 35, 463–471. [Google Scholar] [PubMed]
  55. Pluta, R.; Barcikowska, M.; Januszewski, S.; Misicka, A.; Lipkowski, A.W. Evidence of blood-brain barrier permeability/leakage for circulating human Alzheimer’s beta-amyloid-(1-42)-peptide. Neuroreport 1996, 7, 1261–1265. [Google Scholar] [CrossRef] [PubMed]
  56. Pluta, R.; Miziak, B.; Czuczwar, S.J. Post-Ischemic Permeability of the Blood-Brain Barrier to Amyloid and Platelets as a Factor in the Maturation of Alzheimer’s Disease-Type Brain Neurodegeneration. Int. J. Mol. Sci. 2023, 24, 10739. [Google Scholar] [CrossRef] [PubMed]
  57. Ousta, A.; Piao, L.; Fang, Y.H.; Vera, A.; Nallamothu, T.; Garcia, A.J., 3rd; Sharp, W.W. Microglial Activation and Neurological Outcomes in a Murine Model of Cardiac Arrest. Neurocrit. Care 2022, 36, 61–70. [Google Scholar] [CrossRef] [PubMed]
  58. Zetterberg, H.; Mörtberg, E.; Song, L.; Chang, L.; Provuncher, G.K.; Patel, P.P.; Ferrell, E.; Fournier, D.R.; Kan, C.W.; Campbell, T.G.; et al. Hypoxia due to cardiac arrest induces a time-dependent increase in serum amyloid β levels in humans. PLoS ONE 2011, 6, e28263. [Google Scholar] [CrossRef]
  59. Peeters-Scholte, C.; Meilin, S.; Berckovich, Y.; Westers, P. 2-iminobiotin, a selective inhibitor of nitric oxide synthase, improves memory and learning in a rat model after four vessel occlusion, mimicking cardiac arrest. PLoS ONE 2023, 18, e0291915. [Google Scholar] [CrossRef]
  60. Moulaert, V.R.; Verbunt, J.A.; van Heugten, C.M.; Wade, D.T. Cognitive impairments in survivors of out-of-hospital cardiac arrest: A systematic review. Resuscitation 2009, 80, 297–305. [Google Scholar] [CrossRef]
  61. Sabedra, A.R.; Kristan, J.; Raina, K.; Holm, M.B.; Callaway, C.W.; Guyette, F.X.; Dezfulian, C.; Doshi, A.A.; Rittenberger, J.C.; Post Cardiac Arrest Service. Neurocognitive outcomes following successful resuscitation from cardiac arrest. Resuscitation 2015, 90, 67–72. [Google Scholar] [CrossRef] [PubMed]
  62. Perez, C.A.; Samudra, N.; Aiyagari, V. Cognitive and Functional Consequence of Cardiac Arrest. Curr. Neurol. Neurosci. Rep. 2016, 16, 70. [Google Scholar] [CrossRef]
  63. Steinbusch, C.V.M.; van Heugten, C.M.; Rasquin, S.M.C.; Verbunt, J.A.; Moulaert, V.R.M. Cognitive impairments and subjective cognitive complaints after survival of cardiac arrest: A prospective longitudinal cohort study. Resuscitation 2017, 120, 132–137. [Google Scholar] [CrossRef] [PubMed]
  64. Wagner, M.K.; Berg, S.K.; Hassager, C.; Armand, S.; Møller, J.E.; Ekholm, O.; Rasmussen, T.B.; Fisher, P.M.; Knudsen, G.M.; Stenbæk, D.S. Cognitive impairment and psychopathology in out-of-hospital cardiac arrest survivors in Denmark: The REVIVAL cohort study protocol. BMJ Open 2020, 10, e038633. [Google Scholar] [CrossRef] [PubMed]
  65. Joshi, V.L.; Tang, L.H.; Borregaard, B.; Zinckernagel, L.; Mikkelsen, T.B.; Taylor, R.S.; Christiansen, S.R.; Nielsen, J.F.; Zwisler, A.D. Long-term physical and psychological outcomes after out-of-hospital cardiac arrest-protocol for a national cross-sectional survey of survivors and their relatives (the DANCAS survey). BMJ Open 2021, 11, e045668. [Google Scholar] [CrossRef] [PubMed]
  66. Amacher, S.A.; Bohren, C.; Blatter, R.; Becker, C.; Beck, K.; Mueller, J.; Loretz, N.; Gross, S.; Tisljar, K.; Sutter, R.; et al. Long-term survival after out-of-hospital cardiac arrest: A systematic review and meta-analysis. JAMA Cardiol. 2022, 7, 633–643. [Google Scholar] [CrossRef] [PubMed]
  67. Huebschmann, N.A.; Cook, N.E.; Murphy, S.; Iverson, G.L. Cognitive and Psychological Outcomes Following Pediatric Cardiac Arrest. Front. Pediatr. 2022, 10, 780251. [Google Scholar] [CrossRef] [PubMed]
  68. Lilja, G.; Nilsson, G.; Nielsen, N.; Friberg, H.; Hassager, C.; Koopmans, M.; Kuiper, M.; Martini, A.; Mellinghoff, J.; Pelosi, P.; et al. Anxiety and depression among out-of-hospital cardiac arrest survivors. Resuscitation 2015, 97, 68–75. [Google Scholar] [CrossRef]
  69. Mion, M.; Simpson, R.; Johnson, T.; Oriolo, V.; Gudde, E.; Rees, P.; Quinn, T.; Vopelius-Feldt, V.J.; Gallagher, S.; Mozid, A.; et al. British cardiovascular intervention society consensus position statement on out-of-hospital cardiac arrest 2: Post-discharge rehabilitation. Interv. Cardiol. 2022, 17, e19. [Google Scholar] [CrossRef]
  70. Kim, J.S.; Kim, Y.J.; Ryoo, S.M.; Ahn, S.; Kim, W.Y. Telephone-based evaluation of cognitive impairment and mood disorders in cardiac arrest survivors with good neurologic outcomes: A retrospective cohort study. Sci. Rep. 2023, 13, 18065. [Google Scholar] [CrossRef]
  71. Andersson, A.E.; Rosén, H.; Sunnerhagen, K.S. Life after cardiac arrest: A very long-term follow-up. Resuscitation 2015, 91, 99–103. [Google Scholar] [CrossRef] [PubMed]
  72. Lim, C.; Verfaellie, M.; Schnyer, D.; Lafleche, G.; Alexander, M.P. Recovery, long-term cognitive outcome and quality of life following out-of-hospital cardiac arrest. J. Rehabil. Med. 2014, 46, 691–697. [Google Scholar] [CrossRef] [PubMed]
  73. Fertl, E.; Vass, K.; Sterz, F.; Gabriel, H.; Auff, E. Neurological rehabilitation of severely disabled cardiac arrest survivors. Part I. Course of post-acute inpatient treatment. Resuscitation 2000, 47, 231–239. [Google Scholar] [CrossRef] [PubMed]
  74. Lundgren-Nilsson, Å.; Rosén, H.; Hofgren, C.; Sunnerhagen, K.S. The first year after successful cardiac resuscitation: Function, activity, participation and quality of life. Resuscitation 2005, 66, 285–289. [Google Scholar] [CrossRef] [PubMed]
  75. Mateen, F.J.; Josephs, K.A.; Trenerry, M.R.; Felmlee-Devine, M.D.; Weaver, A.L.; Carone, M.; White, R.D. Long-term cognitive outcomes following out-of-hospital cardiac arrest. Neurology 2011, 77, 1438–1445. [Google Scholar] [CrossRef] [PubMed]
  76. Czuczwar, M. Cognitive impairment and dementia following ischemic stroke and cardiac arrest in humans. In Brain Ischemia: Alzheimer’s Disease Mechanisms; Pluta, R., Ed.; Nova Science Publishers, Inc.: New York, NY, USA, 2019; pp. 1–10. [Google Scholar]
  77. Wolters, F.J.; Segufa, R.A.; Darweesh, S.K.L.; Bos, D.; Ikram, M.A.; Sabayan, B.; Hofman, A.; Sedaghat, S. Coronary heart disease, heart failure, and the risk of dementia: A systematic review and meta-analysis. Alzheimers Dement. 2018, 14, 1493–1504. [Google Scholar] [CrossRef] [PubMed]
  78. Zhang, W.; Luo, P. Myocardial Infarction Predisposes Neurodegenerative Diseases. J. Alzheimers Dis. 2020, 74, 579–587. [Google Scholar] [CrossRef] [PubMed]
  79. Buanes, E.A.; Gramstad, A.; Sovig, K.K.; Hufthammer, K.O.; Flaatten, H.; Husby, T.; Langorgen, J.; Heltne, J.K. Cognitive function and health-related quality of life four years after cardiac arrest. Resuscitation 2015, 89, 13–18. [Google Scholar] [CrossRef]
  80. Caro-Codon, J.; Rey, J.R.; Lopez-de-Sa, E.; Gonzalez Fernandez, O.; Rosillo, S.O.; Armada, E.; Iniesta, A.M.; Fernandez de Bobadilla, J.; Ruiz Cantador, J.; Rodriguez Sotelo, L.; et al. Long-term neurological outcomes in out-of-hospital cardiac arrest patients treated with targeted-temperature management. Resuscitation 2018, 133, 33–39. [Google Scholar] [CrossRef]
  81. Byron-Alhassan, A.; Collins, B.; Bedard, M.; Quinlan, B.; Le May, M.; Duchesne, L.; Osborne, C.; Wells, G.; Smith, A.M.; Tulloch, H.E. Cognitive dysfunction after out-of-hospital cardiac arrest: Rate of impairment and clinical predictors. Resuscitation 2021, 165, 154–160. [Google Scholar] [CrossRef]
  82. Pluta, R. Alzheimer’s disease connected genes in the post-ischemic hippocampus and temporal cortex. Genes 2022, 13, 1059. [Google Scholar] [CrossRef] [PubMed]
  83. Pluta, R.; Kida, E.; Lossinsky, A.S.; Golabek, A.A.; Mossakowski, M.J.; Wisniewski, H.M. Complete cerebral ischemia with short-term survival in rats induced by cardiac arrest: I. Extracellular accumulation of Alzheimer’s β-amyloid protein precursor in the brain. Brain Res. 1994, 649, 323–328. [Google Scholar] [CrossRef] [PubMed]
  84. van Groen, T.; Puurunen, K.; Mäki, H.M.; Sivenius, J.; Jolkkonen, J. Transformation of diffuse β-amyloid precursor protein and β-amyloid deposits to plaques in the thalamus after transient occlusion of the middle cerebral artery in rats. Stroke 2005, 36, 1551–1556. [Google Scholar] [CrossRef] [PubMed]
  85. Qi, J.; Wu, H.; Yang, Y.; Wang, D.D.; Chen, Y.X.; Gu, Y.H.; Liu, T. Cerebral ischemia and Alzheimer’s disease: The expression of amyloid and apolipoprotein E in human hippocampus. J. Alzheimer’s Dis. 2007, 12, 335–341. [Google Scholar] [CrossRef] [PubMed]
  86. Wiśniewski, H.M.; Maślińska, D. Beta-protein immunoreactivity in the human brain after cardiac arrest. Folia Neuropathol. 1996, 34, 65–71. [Google Scholar] [PubMed]
  87. Mörtberg, E.; Zetterberg, H.; Nordmark, J.; Blennow, K.; Catry, C.; Decraemer, H.; Vanmechelen, E.; Rubertsson, S. Plasma tau protein in comatose patients after cardiac arrest treated with therapeutic hypothermia. Acta Anaesthesiol. Scand. 2011, 55, 1132–1138. [Google Scholar] [CrossRef]
  88. Randall, J.; Mörtberg, E.; Provuncher, G.K.; Fournier, D.R.; Duffy, D.C.; Rubertsson, S.; Blennow, K.; Zetterberg, H.; Wilson, D.H. Tau proteins in serum predict neurological outcome after hypoxic brain injury from cardiac arrest: Results of a pilot study. Resuscitation 2013, 84, 351–356. [Google Scholar] [CrossRef]
  89. Mattsson, N.; Zetterberg, H.; Nielsen, N.; Blennow, K.; Dankiewicz, J.; Friberg, H.; Lilja, G.; Insel, P.S.; Rylander, C.; Stammet, P.; et al. Serum tau and neurological outcome in cardiac arrest. Ann. Neurol. 2017, 82, 665–675. [Google Scholar] [CrossRef]
  90. Zhang, Q.; Zhang, C.; Liu, C.; Zhan, H.; Li, B.; Lu, Y.; Wei, H.; Cheng, J.; Li, S.; Wang, C.; et al. Identification and Validation of Novel Potential Pathogenesis and Biomarkers to Predict the Neurological Outcome after Cardiac Arrest. Brain Sci. 2022, 12, 928. [Google Scholar] [CrossRef]
  91. Majd, S.; Power, J.H.; Koblar, S.A.; Grantham, H.J. Early glycogen synthase kinase-3 and protein phosphatase 2A independent tau dephosphorylation during global brain ischaemia and reperfusion following cardiac arrest and the role of the adenosine monophosphate kinase pathway. Eur. J. Neurosci. 2016, 44, 1987–1997. [Google Scholar] [CrossRef]
  92. Majd, S.; Power, J.H.; Koblar, S.A.; Grantham, H.J.M. Introducing a developed model of reversible cardiac arrest to produce global brain ischemia and its impact on microtubule-associated protein tau phosphorylation at Ser396. Int. J. Neurol. Neurother. 2016, 3, 040. [Google Scholar] [CrossRef]
  93. Pluta, R.; Januszewski, S.; Czuczwar, S.J. Brain ischemia as a prelude to Alzheimer’s disease. Front. Aging Neurosci. 2021, 13, 636653. [Google Scholar] [CrossRef] [PubMed]
  94. Khan, S.; Yuldasheva, N.Y.; Batten, T.F.C.; Pickles, A.R.; Kellett, K.A.B.; Saha, S. Tau pathology and neurochemical changes associated with memory dysfunction in an optimized murine model of global cerebral ischaemia—A potential model for vascular dementia? Neurochem. Int. 2018, 118, 134–144. [Google Scholar] [CrossRef] [PubMed]
  95. Wen, Y.; Yang, S.H.; Liu, R.; Perez, E.J.; Brun-Ziukemagel, A.M.; Koulen, P.; Simpkins, J.W. Cdk5 is involved in NFT-like tauopathy induced by transient cerebral ischemia in female rats. Biochim. Biophys. Acta 2007, 1772, 473–483. [Google Scholar] [CrossRef] [PubMed]
  96. Kato, T.; Hirano, A.; Katagiri, T.; Sasaki, H.; Yamada, S. Neurofibrillary tangle formation in the nucleus basalis of Meynert ipsilateral to a massive cerebral infarct. Ann. Neurol. 1988, 23, 620–623. [Google Scholar] [CrossRef]
  97. Hatsuta, H.; Takao, M.; Nogami, A.; Uchino, A.; Sumikura, H.; Takata, T.; Morimoto, S.; Kanemaru, K.; Adachi, T.; Arai, T.; et al. Tau and TDP-43 accumulation of the basal nucleus of Meynert in individuals with cerebral lobar infarcts or hemorrhage. Acta Neuropathol. Commun. 2019, 7, 49. [Google Scholar] [CrossRef]
  98. Pluta, R.; Czuczwar, S.J.; Januszewski, S.; Jabłoński, M. The many faces of post-ischemic tau protein in brain neurodegeneration of the Alzheimer’s disease type. Cells 2021, 10, 2213. [Google Scholar] [CrossRef]
  99. Pluta, R.; Kocki, J.; Bogucki, J.; Bogucka-Kocka, A.; Czuczwar, S.J. LRP1 and RAGE Genes Transporting Amyloid and Tau Protein in the Hippocampal CA3 Area in an Ischemic Model of Alzheimer’s Disease with 2-Year Survival. Cells 2023, 12, 2763. [Google Scholar] [CrossRef]
  100. Maślińska, D.; Laure-Kamionowska, M.; Taraszewska, A.; Deręgowski, K.; Maśliński, S. Immunodistribution of amyloid beta protein (Aβ) and advanced glycation end-product receptors (RAGE) in choroid plexus and ependyma of resuscitated patients. Folia Neuropathol. 2011, 49, 295–300. [Google Scholar]
  101. Ułamek-Kozioł, M.; Kocki, J.; Bogucka-Kocka, A.; Januszewski, S.; Bogucki, J.; Czuczwar, S.J.; Pluta, R. Autophagy, mitophagy and apoptotic gene changes in the hippocampal CA1 area in a rat ischemic model of Alzheimer’s disease. Pharmacol. Rep. 2017, 69, 1289–1294. [Google Scholar] [CrossRef]
  102. Ułamek-Kozioł, M.; Czuczwar, S.J.; Kocki, J.; Januszewski, S.; Bogucki, J.; Bogucka-Kocka, A.; Pluta, R. Dysregulation of autophagy, mitophagy, and apoptosis genes in the CA3 region of the hippocampus in the ischemic model of Alzheimer’s dis-ease in the rat. J. Alzheimers Dis. 2019, 72, 1279–1286. [Google Scholar] [CrossRef] [PubMed]
  103. Ułamek-Kozioł, M.; Kocki, J.; Bogucka-Kocka, A.; Petniak, A.; Gil-Kulik, P.; Januszewski, S.; Bogucki, J.; Jabłoński, M.; Furmaga-Jabłońska, W.; Brzozowska, J.; et al. Dysregulation of Autophagy, Mitophagy, and Apoptotic Genes in the Medial Temporal Lobe Cortex in an Ischemic Model of Alzheimer’s Disease. J. Alzheimers Dis. 2016, 54, 113–121. [Google Scholar] [CrossRef] [PubMed]
  104. Salminen, A.; Kauppinen, A.; Kaarniranta, K. Hypoxia/ischemia activate processing of amyloid precursor protein: Impact of vascular dysfunction in the pathogenesis of Alzheimer’s disease. J. Neurochem. 2017, 140, 536–549. [Google Scholar] [CrossRef] [PubMed]
  105. Sweeney, M.D.; Montagne, A.; Sagare, A.P.; Nation, D.A.; Schneider, L.S.; Chui, H.C.; Harrington, M.G.; Pa, J.; Law, M.; Wang, D.J.J.; et al. Vascular dysfunction the disregarded partner of Alzheimer’s disease. Alzheimers Dement. 2019, 15, 158–167. [Google Scholar] [CrossRef] [PubMed]
  106. Kriska, J.; Hermanova, Z.; Knotek, T.; Tureckova, J.; Anderova, M. On the Common Journey of Neural Cells through Ischemic Brain Injury and Alzheimer’s Disease. Int. J. Mol. Sci. 2021, 22, 9689. [Google Scholar] [CrossRef] [PubMed]
  107. Elman-Shina, K.; Efrati, S. Ischemia as a common trigger for Alzheimer’s disease. Front. Aging Neurosci. 2022, 14, 1012779. [Google Scholar] [CrossRef] [PubMed]
  108. Fisher, R.A.; Miners, J.S.; Love, S. Pathological changes within the cerebral vasculature in Alzheimer’s disease: New perspectives. Brain Pathol. 2022, 32, e13061. [Google Scholar] [CrossRef]
  109. Lecordier, S.; Pons, V.; Rivest, S.; ElAli, A. Multifocal Cerebral Microinfarcts Modulate Early Alzheimer’s Disease Pathology in a Sex-Dependent Manner. Front. Immunol. 2022, 12, 813536. [Google Scholar] [CrossRef]
  110. Das, T.K.; Ganesh, B.P.; Fatima-Shad, K. Common signaling pathways involved in Alzheimer’s disease and stroke: Two Faces of the Same Coin. J. Alzheimers Dis. Rep. 2023, 7, 381–398. [Google Scholar] [CrossRef]
  111. Eisenmenger, L.B.; Peret, A.; Famakin, B.M.; Spahic, A.; Roberts, G.S.; Bockholt, J.H.; Johnson, K.M.; Paulsen, J.S. Vascular contributions to Alzheimer’s disease. Transl. Res. 2023, 254, 41–53. [Google Scholar] [CrossRef]
  112. Chen, C.; Liu, C.; Niu, Z.; Li, M.; Zhang, Y.; Gao, R.; Chen, H.; Wang, Q.; Zhang, S.; Zhou, R.; et al. RNA-seq analysis of the key long noncoding RNAs and mRNAs related to cognitive impairment after cardiac arrest and cardiopulmonary resuscitation. Aging 2020, 12, 14490–14505. [Google Scholar] [CrossRef] [PubMed]
  113. Jing, W.; Tuxiu, X.; Xiaobing, L.; Guijun, J.; Lulu, K.; Jie, J.; Lu, Y.; Liying, Z.; Xiaoxing, X.; Jingjun, L. LncRNA GAS5/miR-137 Is a Hypoxia-Responsive Axis Involved in Cardiac Arrest and Cardiopulmonary Cerebral Resuscitation. Front. Immunol. 2022, 12, 790750. [Google Scholar] [CrossRef] [PubMed]
  114. Zhang, Y.H.; Peng, F.; Zhang, L.; Kang, K.; Yang, M.; Chen, C.; Yu, H. Long noncoding RNA upregulates adapter ShcA protein expression to promote cognitive impairment after cardiac arrest and resuscitation. Shock 2022, 58, 169–178. [Google Scholar] [CrossRef] [PubMed]
  115. Chen, G.J.; Xu, J.; Lahousse, S.A.; Caggiano, N.L.; de la Monte, S.M. Transient hypoxia causes Alzheimer-type molecular and biochemical abnormalities in cortical neurons: Potential strategies for neuroprotection. J. Alzheimers Dis. 2003, 5, 209–228. [Google Scholar] [CrossRef]
  116. Kocki, J.; Ułamek-Kozioł, M.; Bogucka-Kocka, A.; Januszewski, S.; Jabłoński, M.; Gil-Kulik, P.; Brzozowska, J.; Petniak, A.; Furmaga-Jabłońska, W.; Bogucki, J.; et al. Dysregulation of Amyloid-β Protein Precursor, β-Secretase, Presenilin 1 and 2 Genes in the Rat Selectively Vulnerable CA1 Subfield of Hippocampus Following Transient Global Brain Ischemia. J. Alzheimers Dis. 2015, 47, 1047–1056. [Google Scholar] [CrossRef]
  117. Pluta, R.; Kocki, J.; Ułamek-Kozioł, M.; Petniak, A.; Gil-Kulik, P.; Januszewski, S.; Bogucki, J.; Jabłoński, M.; Brzozowska, J.; Furmaga-Jabłońska, W.; et al. Discrepancy in Expression of β-Secretase and Amyloid-β Protein Precursor in Alzheimer-Related Genes in the Rat Medial Temporal Lobe Cortex Following Transient Global Brain Ischemia. J. Alzheimers Dis. 2016, 51, 1023–1031. [Google Scholar] [CrossRef]
  118. Pluta, R.; Kocki, J.; Ułamek-Kozioł, M.; Bogucka-Kocka, A.; Gil-Kulik, P.; Januszewski, S.; Jabłoński, M.; Petniak, A.; Brzozowska, J.; Bogucki, J.; et al. Alzheimer-associated presenilin 2 gene is dysregulated in rat medial temporal lobe cortex after complete brain ischemia due to cardiac arrest. Pharmacol. Rep. 2016, 68, 155–161. [Google Scholar] [CrossRef]
  119. Pluta, R.; Bogucka-Kocka, A.; Ułamek-Kozioł, M.; Bogucki, J.; Januszewski, S.; Kocki, J.; Czuczwar, S.J. Ischemic tau protein gene induction as an additional key factor driving development of Alzheimer’s phenotype changes in CA1 area of hippocampus in an ischemic model of Alzheimer’s disease. Pharmacol. Rep. 2018, 70, 881–884. [Google Scholar] [CrossRef]
  120. Pluta, R.; Ułamek-Kozioł, M.; Kocki, J.; Bogucki, J.; Januszewski, S.; Bogucka-Kocka, A.; Czuczwar, S.J. Expression of the tau protein and amyloid protein precursor processing genes in the CA3 Area of the hippocampus in the ischemic model of Alzheimer’s Disease in the Rat. Mol. Neurobiol. 2020, 57, 1281–1290. [Google Scholar] [CrossRef]
Figure 1. Structures susceptible to ischemic brain injury due to cardiac arrest and responsible for cognition. The figure shows damage to structures affecting memory, behavior, and cognitive impairment after cardiac arrest in an identical manner to that seen in Alzheimer’s disease. Arrows indicate decreased activity.
Figure 1. Structures susceptible to ischemic brain injury due to cardiac arrest and responsible for cognition. The figure shows damage to structures affecting memory, behavior, and cognitive impairment after cardiac arrest in an identical manner to that seen in Alzheimer’s disease. Arrows indicate decreased activity.
Ijms 25 01291 g001
Table 1. Changes in the expression of the Alzheimer’s disease-linked genes in the hippocampus and temporal cortex at various times after ischemic brain injury due to cardiac arrest.
Table 1. Changes in the expression of the Alzheimer’s disease-linked genes in the hippocampus and temporal cortex at various times after ischemic brain injury due to cardiac arrest.
Genes
Survival
APPADAM10BACE1PSEN1PSEN2MAPT
CA1 subfield of hippocampus
2 daysN.A.↑↑↑↑↑↑
7 daysN.A.
30 daysN.A.
CA3 subfield of hippocampus
2 days
7 days
30 days
Medial temporal cortex
2 daysN.A.↑↑↑↑N.A.
7 daysN.A.N.A.
30 daysN.A.N.A.
Expression: ↑ increase; ↓ decrease; ↔ oscillation around control values; N.A., not available. Genes: APP, amyloid protein precursor; ADAM10, α-secretase; BACE1, β-secretase; PSEN1, presenilin 1; PSEN2, presenilin 2; MAPT, tau protein.
Table 2. Changes in the expression of amyloid and tau protein transporting genes in the hippocampal CA3 area at various times after ischemic brain injury due to cardiac arrest.
Table 2. Changes in the expression of amyloid and tau protein transporting genes in the hippocampal CA3 area at various times after ischemic brain injury due to cardiac arrest.
Survival
Genes
2 Days7 Days30 Days12 Months18 Months24 Months
LRP1↑↑
RAGE↓↓
Expression: ↑ increase; ↓ decrease; ↔ oscillation around control values. Genes: LRP1-low-density lipoprotein receptor-related protein 1; RAGE-receptor for advanced glycation end products.
Table 3. Changes in the expression of autophagy, mitophagy, and apoptosis genes linked to Alzheimer’s disease in the hippocampus and temporal cortex at various times after ischemic brain injury due to cardiac arrest.
Table 3. Changes in the expression of autophagy, mitophagy, and apoptosis genes linked to Alzheimer’s disease in the hippocampus and temporal cortex at various times after ischemic brain injury due to cardiac arrest.
Genes
Survival
BECN1BNIP3CASP3
CA1 subfield of hippocampus
2 days↑↑↑
7 days
30 days
CA3 subfield of hippocampus
2 days
7 days
30 days
Medial temporal cortex
2 days↓↓
7 days↑↑↑
30 days
Expression: ↑ increase; ↓ decrease; ↔ oscillation around control values. Genes: BECN1-autophagy; BINP3-mitophagy; CASP3-apoptosis.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Pluta, R.; Czuczwar, S.J. Ischemia-Reperfusion Programming of Alzheimer’s Disease-Related Genes—A New Perspective on Brain Neurodegeneration after Cardiac Arrest. Int. J. Mol. Sci. 2024, 25, 1291. https://doi.org/10.3390/ijms25021291

AMA Style

Pluta R, Czuczwar SJ. Ischemia-Reperfusion Programming of Alzheimer’s Disease-Related Genes—A New Perspective on Brain Neurodegeneration after Cardiac Arrest. International Journal of Molecular Sciences. 2024; 25(2):1291. https://doi.org/10.3390/ijms25021291

Chicago/Turabian Style

Pluta, Ryszard, and Stanisław J. Czuczwar. 2024. "Ischemia-Reperfusion Programming of Alzheimer’s Disease-Related Genes—A New Perspective on Brain Neurodegeneration after Cardiac Arrest" International Journal of Molecular Sciences 25, no. 2: 1291. https://doi.org/10.3390/ijms25021291

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop