Open Access

Neurodegeneration and convergent factors contributing to the deterioration of the cytoskeleton in Alzheimer's disease, cerebral ischemia and multiple sclerosis (Review)

  • Authors:
    • Johanna Andrea Gutiérrez‑Vargas
    • John Fredy Castro‑Álvarez
    • Jose Fernando Zapata‑Berruecos
    • Komal Abdul‑Rahim
    • Anibal Arteaga‑Noriega
  • View Affiliations

  • Published online on: February 18, 2022     https://doi.org/10.3892/br.2022.1510
  • Article Number: 27
  • Copyright: © Gutiérrez‑Vargas et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The cytoskeleton is the main intracellular structure that determines the morphology of neurons and maintains their integrity. Therefore, disruption of its structure and function may underlie several neurodegenerative diseases. This review summarizes the current literature on the tau protein, microtubule‑associated protein 2 (MAP2) and neurofilaments as common denominators in pathological conditions such as Alzheimer's disease (AD), cerebral ischemia, and multiple sclerosis (MS). Insights obtained from experimental models using biochemical and immunocytochemical techniques highlight that changes in these proteins may be potentially used as protein targets in clinical settings, which provides novel opportunities for the detection, monitoring and treatment of patients with these neurodegenerative diseases.

1. Introduction

The cytoskeleton of neurons is the primary structure that regulates cell shape, protein localization and transport from the soma to dendrites and axons or vice versa. This structure consists of microfilaments (MFs) (diameter of ~8 nm), intermediate filaments (IFs) (diameter ranging from 7-11 nm) and microtubules (MTs) (diameter of ~25 nm) (1).

MFs are primarily composed of actin, enriched in cortical regions near the cell membrane and are particularly concentrated in presynaptic terminals, dendritic spines and growth cones. MFs are also called actin filaments; they are formed during proliferation, are considered to be versatile due to their ability to create and destroy themselves, used as construction materials and bind to accessory proteins, regulating the creation and destruction of filaments (2,3).

IFs are components of the cytoskeleton that extend from the nucleus to the cell periphery, and their main function is to provide mechanical resistance. IFs show an unusual degree of cell specificity and are often used as markers of cell differentiation. These filaments are subdivided into multiple types, among which neurofilaments (neurons) and glial filaments (glial cells) are noteworthy structures in the current discussion (4). Neurofilaments (NFs) support axons and dendrites (5) and play an important role in determining the axonal caliber of myelinated fibers (1). These filaments have been a focus of research, as an increase in their presence is potentially related to axonal damage and the severity of neurological diseases (6).

MTs consist of dimers of α and β-tubulin, and these dimers subsequently bind from one end to the other to form a ‘protofilament’ of tubulin. Additionally, 13 protofilaments are aligned in parallel in a circular manner to form an empty tube that is nm-µm long. The tube is polarized: it has head (+) and tail (-) ends. Polymerization occurs by adding new subunits at the (+) end. In this process, the units are removed from the (-) end and added to the (+) end. This process requires various accessory factors; two of the most important accessory factors are microtubule accessory proteins (MAPs) and tau protein. These accessory proteins appear to be involved both in the polymerization process and in the stabilization of tubulin once it is polymerized (7).

Due to the significant role of the cytoskeleton in cell transport, in addition to being essential for the normal functioning of neurons, cytoskeletal anomalies result in neuronal damage and cell death, which are the common denominators of neurodegenerative diseases (8,9). Although the causes of transport abnormalities may vary among the various pathological conditions, in many circumstances, deficiencies in transport are caused by a decrease in the stability of its components, such as alterations in neurofilaments, microtubule stability, loss of actin dynamics (9), and an increase in the phosphorylation rates of some proteins that comprise this network (10), as is the case for the tau protein (11). Based on these observations, cytoskeletal organization defects may be a common feature that contributes to neurodegeneration in pathologies such as Alzheimer's disease (AD), cerebral ischemia and multiple sclerosis (MS). Therefore, the potential benefits of cytoskeletal stabilizing agents in improving axonal transport and nerve function in patients with these diseases has been highlighted.

2. Cytoskeletal damage and neurodegeneration

The process of destabilization and aggregation of cytoskeletal proteins causes neurons to become unstable, and interferes with the antegrade and retrograde transport of biomolecules along their axons and dendrites, and, in some cases, generates protein aggregates that lead to neurodegeneration (12). For example, in AD and cerebral ischemia, the hyperphosphorylation of the tau protein leads to the formation of neurofibrillary tangles, axonal damage, dendritic damage and subsequent cell death (13,14). Axonal damage associated with the tau protein has also been reported in other pathologies, such as MS (15), although in the latter, neurofilaments are the central filaments contributing to deterioration due to demyelination (16,17). These proteins have become potential biomarkers, since they are related to the pathophysiology of the disease, and therefore have a significant association with diagnosis, treatment and prognosis, facilitating the diagnosis of individuals at earlier stages of the disease with less severe symptoms and long-term repercussions.

In this review, we discuss the current body of literature with regard to the abnormalities of the cytoskeleton and related proteins that are considered to play critical roles in the pathology of AD, cerebral ischemia and MS, such as microtubule-associated protein 2 (MAP2), tau and neurofilaments.

3. Alzheimer's disease

AD is considered the most prevalent cause of dementia worldwide (18). AD is characterized by two proteinopathies that result in a loss of cell and tissue homeostasis, leading to progressive neuronal degeneration and loss of cognitive functions in patients (19). Amyloidopathy is a product of the abnormal cleavage of amyloid precursor protein (APP), which leads to the accumulation of the Aβ peptide in senile or amyloid plaques, and tauopathy, which is due to hyperphosphorylation and aggregation of the tau protein (14). This protein is normally associated with MTs, but when it is abnormally phosphorylated, it dissociates and aggregates, leading to the formation of paired helical filaments (PHFs) and resulting structures called neurofibrillary tangles (NFTs) (20) (Fig. 1).

Tau and MAPs in Alzheimer's disease

Tauopathy in AD is responsible for a set of abnormalities that lead to neuronal degeneration, such as defects in axonal transport and mitochondrial and lysosomal function, among other functions associated with MTs (21,22). These abnormalities are due to the overactivation or loss of inhibition of various kinases and phosphatases that regulate the binding of MAPs and NFs to the neuronal cytoskeleton and their functions (23). Under pathological conditions, the tau protein itself leads to a loss of function, preventing the binding or shedding of MTs and accumulating in the cytoplasm along with other MAPs, forming PHFs and subsequently resulting in extracellular accumulation in NFTs, which leads to brain deterioration (14,24,25).

In mice deficient in the tau protein, decreases in the numbers of MTs and small caliber axons, muscle spasms and behavioral deficits have been identified, although MAP2 partially compensates for the tau deficit (26). In neurons, the tau protein regulates the establishment of MT dynamics in axons and has been linked to the formation of cytoplasmic extensions, axonal transport and protection against compounds that are deleterious to the cell (13). In vitro models related to AD, such as glutamate excitotoxicity, generate a cellular environment that resembles the conditions in the neurons of a patient (27). This model shows the disassembly of MTs, the retraction of dendritic and axonal processes and the hyperphosphorylation and aggregation of PHFs in the cell soma in the short term. Increased intracellular calcium levels, abnormal activation of signaling cascades, and loss of cellular functions trigger cell death signals that represent the neurodegenerative process of the disease (28).

Studies have described how the inhibition of various cytoskeletal proteins is directly and indirectly involved in the disassembly of MTs, the accumulation of the tau protein and neuronal death (27,29). The tau protein kinases described here are grouped into three classes: proline-directed protein kinases (PDPKs), protein kinases (non-PDPKs) and tyrosine protein kinases (TPKs). Each kinase was assessed based on its structure, roles, regulation, involvement in tau phosphorylation and neurodegeneration linked to AD (30,31). Cyclin Dependent Kinase 5 (CDK5), a kinase related to the tau protein and its activators p35/p25, cytoskeletal modifying proteins such as p120 catenin, and enzymes related to the processing of β amyloid such as BACE1, have been linked to hyperphosphorylation of tau and neuronal death (Fig. 1). Silencing of the CDK5 protein, and the spatial and functional regulation of the activator p35, as well as its cleaved form p25 occupy central nodes in the mechanism regulating cellular signaling associated with the hyperphosphorylation of tau (23,28,32). CDK5 regulates NFs and MAPs either by forming protein complexes or by phosphorylating them directly, as in the case of NF or MAP2, MAP1b and the tau protein, which, when phosphorylated, induce the formation and stabilization of MTs (33,34). In turn, proteins associated with the processing of β amyloid, such as BACE1, participate in tau hyperphosphorylation as part of the cellular and molecular machinery that exerts a synergistic effect on the development of histopathological markers of the disease (35,36).

One of the main pathways for degradation of hyperphosphorylated tau is autophagy, which is a selective cellular catabolic process by which cytoplasmic material is transported from the cytoskeleton to lysosomes for degradation (37). This mechanism functions at basal levels in all cells and is required to maintain homeostasis (38). This process is particularly important in neurons, since they undergo cell division only at a low rate, and therefore must survive throughout the lifespan of the organism through the exchange of proteins and organelles (39). In neurodegenerative diseases such as AD, the cytoskeleton plays a key role in the degradation of PHFs, and when cytoskeletal integrity is compromised, PHFs are more likely to aggregate and accumulate (40). The current body of literature show an increase in autophagy in the early stages of AD in the 3xTg-AD model, which loses its effect as the pathological process progresses (41). The regulation of CDK5 and BACE1 and the effects on the cytoskeleton restore the degradation of the transgenically expressed tau protein and promote the functional recovery of the brain (23,35).

The contribution of other MT-associated proteins, such as MAP2, MAP1B and MAP1A, in the pathogenesis and progression of AD may vary as they generate different MT dynamics dependent on their location in the models described, despite sharing similar sequences and functions in stabilizing MTs with the tau protein (42). Unlike the tau protein, which is abundantly distributed in the axonal compartment, MAP2 is located exclusively in the somatodendritic compartment, whereas MAP1B and MAP1A are located in both compartments (43). Likewise, MAP2, MAP1B and MAP1A undergo hyperphosphorylation but fail to form filaments, as does the tau protein (44). In turn, a decrease in MAP2 levels or an increase in the levels of the soluble protein due to abnormal phosphorylation have been suggested to trigger neurotoxic processes (22). These changes are related to the loss of neuronal connectivity induced by amyloid β, tauopathy and the excitotoxic microenvironment that compromises the dynamics of dendritic spines and postsynaptic compartments (22,45).

Neurofilaments in Alzheimer's disease

NFs are the other type of essential filament involved in the neurodegeneration process; they are the largest component of the neuronal cytoskeleton, and together with the tau protein, they predominate in the nervous system (40). These filaments are composed of a family of 5 intermediate filaments termed NF heavy (NFH), medium (NFM) and light (NFL) chains, and α-internexin and peripherin (46). The assembly of NFs is essential for the growth and stability of axons in both the central and peripheral nervous systems (47). Its functions are broad and mainly mediate the stabilization of the microtubule content and interactions with organelles such as the mitochondria (48). In AD and other neurodegenerative processes, NFs have been described as a common biomarker that reflects the changes in the neuronal cytoskeleton and the progression of the neurodegenerative process that underlies the pathology (48).

NFs have been detected in both cerebrospinal fluid (CSF) and peripheral blood (SP) (49). In a cohort of 1,070 PSEN1 E280A mutation carriers and 1,074 noncarriers with baseline assessments and 242 mutation carriers and 262 noncarriers with longitudinal (6±3 years) measures, ranging in age from 8 to 75 years, plasma NFL levels increased with age in both groups and began to differentiate carriers from noncarriers at age 22 (22 years before the estimated median age of mild cognitive impairment) (50,51). This biomarker is released in to the CSF and SP as the inflammatory process and gliosis progress in the white matter, and has been correlated with cortical and hippocampal atrophy, widening of the ventricles, memory impairment and mild cognitive impairment (52,53). Although NFs have proline-directed phosphorylation sites and are identified in NFTs as one of the proteins that accumulate with PHFs, their contribution to aggregate formation has not been clarified (24). Axonal damage associated with MTs and synaptic compromise are common pathologies such as AD, Down syndrome, frontotemporal dementia, amyotrophic lateral sclerosis, dementia with Lewy bodies, progressive supra-nuclear palsy, and cortico-basal syndrome; along with the aging process, they convert NFs into biomarkers of CNS and peripheral involvement (54-56).

4. Cerebral ischemia

Cerebral ischemia is a disorder that affects the brain tissue and is characterized mainly by the disrupted supply of oxygen and other nutrients, which leads to tissue death (57). This pathology triggers cognitive impairment or dementia, conditions that include alterations in learning, memory and functions needed to perform basic activities of daily life (58). Within the pathophysiology of the disease, changes in the cytoskeleton that reflect neuronal damage have been identified (59).

MAP2 and tau in cerebral ischemia

Numerous antibodies have been used in experimental models to identify the damage and chronological changes in cytoskeletal proteins, including antibodies against microtubule proteins such as MAP2, a marker of cytoskeletal disruption; in fact, studies have documented a correlation of MAP2 expression with prolonged postischemic periods (60-62). In animal models, immunohistochemistry and immunofluorescence techniques revealed the loss or discontinuity of MAP2 immunoreactivity in lesions. The discontinuity reflects the fragmentation of dendrites. The translocation of this protein to neuronal cells has also been reported (63-65) (Fig. 1). Taken together, these data reveal the changes in dendrites that lead to cell death in areas of both the ischemic focus and in regions of the ischemic penumbra, as well as the loss of synaptic plasticity in regions further from the ischemic focus (65). The loss of MAP2 may contribute to the initial phase of neuronal dysfunction, and dendritic degradation may be the first sign of neurodegeneration 1 h after cerebral ischemia (66). In patients who have suffered cerebral ischemia, a decrease in MAP2 immunoreactivity has been observed in the motor (area 4), temporal (area 21), frontal (area 10) and visual (area 17) cortices in the left hemisphere; additionally, in all the areas studied, the most significant decrease in MAP2 expression was detected in cortical layers II-III compared to cortical layers V-VII. The maximum reduction in MAP2-positive pyramidal neurons was observed in cortical layers II-III of the motor cortex after 1 year of survival after cerebral infarction (67). Based on this finding, markers of the cytoskeleton are sensitive to the deterioration that occurs long after cerebral infarction.

Another protein involved in microtubule disruption is the tau protein. The hyperphosphorylation of the tau protein is associated with the development of dementia in the late postischemia phase. Its roles in initiating synaptic and cognitive dysfunction, in addition to neuronal toxicity and neurodegeneration, have been described (68). Experimental animals subjected to cerebral ischemia present disruptions in memory and learning processes accompanied by tau hyperphosphorylation, the formation of PHFs and changes in the immunoreactivity of the MAP2 protein (69).

The relationship between the CA1 region of the hippocampus and the expression of the tau gene has been established after transient global cerebral ischemia in rats with a survival period of 2, 7 and 30 days (70). In the hippocampal CA1 region, the expression of the tau gene increased to a maximum 3.3-fold change on the second day after cerebral ischemia. A total of 7 days after ischemic episodes, the expression ranged from 0.2 to 0.5 times the base value (70). On the 30th day of survival after ischemic injury, the expression of the tau protein gene decreased to 0.4 times that of the base value (70). Studies have shown that tau protein phosphorylation patterns differ depending on the models of cerebral ischemia. After ischemia and global brain recirculation, the tau protein is phosphorylated and slowly accumulates (71). Transient focal cerebral ischemia with 1 day of reperfusion induces local hyperphosphorylation (regions of injury) of the tau protein (72,73). Current research indicates that after ischemia, hyperphosphorylated tau protein accumulates in cortical neuronal cells and is accompanied by apoptosis (72,74). Additionally, hyperphosphorylation of tau leads to the formation of NFTs 24 h postinfarction in regions such as the motor, sensory and hippocampal cortex. These tangles persist in these same regions for up to 1 month after infarction, and lead to learning and memory deficits in the affected animals (69).

In clinical studies, an increase in total tau levels in human CSF and blood has been reported after brain injury, including ischemic stroke (75-78). Measurable tau has been detected in serum within 6 h after the onset of ischemic symptoms (78). The concentration may peak after 3-5 days (78) or later (79). Furthermore, a significant correlation between serum tau levels and the severity of the clinical deficit or disability evaluated using the Barthel index (BI) was not observed. However, serum tau levels correlate with infarct volumes (7-48 ml) and functional results at 90 days postischemia (78). These findings are consistent with other studies indicating that the absence of serum tau during the acute phase (<24 h) of ischemia might predict good clinical outcomes 90 days after stroke (80). Patients in whom tau was detected in serum had more severe neurological deficits and poorer functional outcomes than patients without tau (81). However, other researchers found that tau protein levels are correlated with neurological deficit (BI) scores after 48 h. Additionally, serum tau levels did not have a significant correlation with the etiology of stroke, as represented by the TOAST criteria (82). A prospective study revealed that tau levels in both plasma and CSF were closely related not only to stroke severity assessed using the National Institutes of Health Stroke Scale but also to long-term outcomes (83). In particular, the study of autopsy specimens of the brains from patients with cerebral infarction showed an increase in tau immunoreactivity and tau deposition in the ischemic area (73,84). However, the tau protein has been detected in the serum of ~40% of patients with stroke (78,79). Some researchers propose that tau appears in the blood due to disruption of the blood-brain barrier (BBB). Some factors, such as matrix metalloproteinase 9 (MMP9), may play a key role in the release of tau into the circulation (79) (Fig. 1).

In addition to BBB damage after stroke, another major cause of persistent disability after stroke is neuroaxonal damage, which is crucial for the functional outcomes and long-term survival of patients with stroke. Predicting functional outcomes after ischemic stroke is very important for patients and clinicians in terms of allocating healthcare resources and optimizing patient care. Furthermore, the amount of acute neuroaxonal damage reflected by the infarct nucleus guides patient selection for stroke therapies, e.g., endovascular thrombectomy beyond the 6-h time window. To date, only diffusion magnetic resonance imaging (MRI) and computed tomography (CT) perfusion approaches serve to assess the core of the infarct; therefore, blood biomarkers are urgently needed to guide individualized treatments of patients. NFs may be a suitable candidate for this purpose as they are part of the neuronal cytoskeleton, are exclusively expressed in neurons (85), and show encouraging results, as described further below.

Neurofilaments in cerebral ischemia

After the occlusion of a cerebral artery and subsequent neuroaxonal damage, the NF protein is released into the CSF and to a lesser extent in SP. Ultrasensitive assays with the single molecule matrix method facilitate the highly sensitive quantification of NF levels in blood (86). Due to the ease and wide applicability, research on NFs in various diseases, such as MS and other neurodegenerative diseases, including cerebral infarction, is emerging (16). In a study conducted by Peters et al (87), where they evaluated 503 patients with small vessel disease (SVD), NFs were associated with the presence of lacunae and microbleeds and with MRI imaging markers related to SVD. In addition, NF was associated with gap incidents during patient follow-up, as well as with future cognitive decline after adjustment for age, sex, education and depression. The risk of dementia increased with higher NF levels. Therefore, NFs were proposed as an emerging blood biomarker for neuroaxonal damage in various neurological diseases affecting the elderly, including small vessel neurodegenerative and cerebral disease (88,89). Thus, cerebrovascular diseases appear to be a major vascular contributor to dementia (88-90).

In 2019, Timo Uphaus and colleagues were the first to show that NF is a valuable biomarker for functional independence at 90 days after ischemic stroke and predicts long-term cardiovascular outcomes (85). Accordingly, NF may be useful in selecting patients at high risk of future cardiovascular events. The superiority of NF over other existing biomarkers was also shown with respect to its predictive value for functional outcomes and cardiovascular survival after ischemic stroke (85). However, studies examining larger patient cohorts are required to confirm the results and to change current clinical practice, since NF may be used in clinical practice as a screening biomarker to select patients at high risk of accident occurrence, recurrent cerebrovascular disease and those more susceptible to death following cerebral ischemia.

5. Multiple sclerosis

MS is a chronic demyelinating disease of the central nervous system (CNS). Although its etiology remains unknown, immune cells are generally accepted to invade the CNS, where demyelination occurs. This process is attributed to the migration of autoreactive T lymphocytes from the periphery toward the CNS with the capacity to cross the BBB and progressively compromise brain function (91) (Fig. 1). The clinical manifestation of MS represents the final stage of a process that involves inflammation, demyelination, remyelination and the depletion of oligodendrocytes, astrocytes and neurons along with axonal degeneration (91).

Tau and MAP2 in multiple sclerosis

In the affected part of the CNS, constitutively expressed proteins in the axon, such as tau protein and NFs, can be detected using antibodies (Fig. 1). CSF tau protein levels are associated with axonal injury, are elevated in patients with MS and correlate with the progression of disability (75,92). In serum samples from patients with MS collected before and after autologous hematopoietic stem cell transplantation, tau protein levels were increased 3 months after transplantation; this observation may reflect brain atrophy induced by chemotherapy-mediated toxicity (88). In a case-controlled study of 30 healthy women and 30 women with MS, the levels of the total tau protein in serum and saliva were analyzed. Total tau protein was level in the serum of patients with MS compared with the control group; therefore, tau represents a potential biomarker for MS. However, the levels of tau protein in saliva was not a suitable biomarker for the detection of MS (93). Some MS studies have sought to investigate the mechanisms of remyelination by analyzing the MAP2 protein levels in brain lesions affected by MS using immunocytochemistry and a series of specific monoclonal antibodies. MAP2 expression was increased in the brains of patients with MS (94). MAP2 was expressed in the regenerating oligodendrocytes associated with demyelinated lesions, with the highest levels detected in regions with extensive remyelination. Using electron microscopy, MAP2 was shown to be located in oligodendrocytes involved in remyelination, as evidenced by the extension of its process and association with finely myelinated (remyelinated) axons (94).

Neurofilaments in multiple sclerosis

NF, a cytoskeletal protein that has been most closely associated with axonal deterioration in MS, has been used as a biomarker for axonal degeneration and may be used to predict the neurological outcomes of patients (48,86). Following axonal damage in the CNS, NF proteins released into the CSF reflect the degree of axonal damage and neuronal death. Since neurofilaments are located in the cytoplasm of neurons, all diseases that lead to neuronal and axonal damage potentially increase the levels of these proteins in the CSF (86). In fact, NFs have been evaluated both in clinical studies and in in vivo models that simulate the degenerative processes of MS, where the most commonly used model is the experimental autoimmune encephalitis (EAE) model. In this model, the mean concentration of NFL is higher in the supernatant and brain pellet of rats with all EAE subtypes compared to samples collected from controls (95). Furthermore, NFM and NFH levels change in the later phases of the disease. Therefore, the NFL biomarker may reflect acute axonal damage mediated by inflammatory mechanisms, and may have prognostic value for the evaluation of MS, while NFM and NFH may be better indicators of nerve regeneration after acute inflammation (95).

In humans, NFs were used for the first time as markers of neuronal damage in a study of 12 patients with amyotrophic lateral sclerosis and 11 patients with AD (96). Subsequently, CSF NF levels were higher in 60 patients with relapsing-remitting multiple sclerosis than in the control subjects (97), suggesting that these proteins may serve as a biomarker of the disease activity of MS.

NFs have been associated with the progression of disability in patients with MS (74), and according to studies comparing the serum and CSF levels of NFs, they are strongly correlated (83). An ultrasensitive technique called single molecule arrays (SIMOA) has been developed recently to detect NF levels in blood, enabling, for the first time, the detection of NF levels in serum. Compared to detection using ELISA or ECL-based assays, SIMOA has >25-fold higher analytical sensitivity (SIMOA: 0.62 pg/ml, ECL: 15.6 pg/ml, ELISA: 78, 0 pg/ml) (98).

NFL is associated with axonal damage, while NFH is associated with the progression of disability (87,99). In some of the population based studies, NFL levels were shown to be associated with an increased risk of progressive phenotypes (16), and they have also been useful in diagnosing the pathology; when NFL levels were compared between patients with MS and controls, where the values between the two groups were different, they were higher in patients with MS (100).

A recent meta-analysis found that patients with MS present with axonal injury (101), which may explain the neurodegeneration caused by the disease and its effect on NF levels, especially in the early stages of the disease. The cause of axonal loss is not yet known, but a destructive process directed against components of the axonal cytoskeleton appears to contribute to the progression of the disability (99). Serum NF levels also correlate with MRI activity, degree of disability and rate of brain atrophy (102,103). Furthermore, NFs are also suitable as a prognostic biomarker for the conversion of clinically isolated syndrome (CIS) to MS (104,105). A recent study showed the prognostic importance of serum NF levels in the conversion of isolated radiological syndrome to CIS (106).

The serum concentration of NFL, which does not require a lumbar puncture, appears to correlate with several clinical and magnetic tomographic features of MS (107,108). Therefore, it may conceivably be established as a prognostic biomarker in clinical practice in the future.

6. Conclusions

Cytoskeletal damage, including disruptions to microtubule stability, NFs and axonal transport, have been characterized in several unrelated neurodegenerative conditions. Although no direct link has been established between the different pathologies addressed in this review, common cytoskeletal players have been shown to contribute to deterioration, suggesting that defects in the organization of the cytoskeleton are a common feature that contributes to neurodegeneration. Findings from the in vitro and in vivo experimental models discussed in this review show that the tau protein, MAP2 and NFs are essential for maintaining the stability of the cytoskeleton, and that their dysregulation triggered by anoxia (cerebral ischemia), the Aβ peptide (AD) and demyelination (MS) leads to the disassembly of MAP2 and NFs, hyperphosphorylation and tau accumulation. These proteins are subsequently released into the SP after the disruption of the BBB through the actions of metalloproteases such as MMP9 or the post-traumatic inflammatory response (Fig. 1). The latter suggests that cytoskeletal components detected in the SP are promising protein targets that may facilitate the diagnosis and progression of cerebral ischemia, AD and MS in a practical and safe manner, as reported in several clinical studies discussed in the review. However, more studies are required to correlate the clinical manifestations presented by patients with these pathologies.

Acknowledgements

Not applicable.

Funding

Funding: We would like to thank Remington University Corporation for the financial support.

Availability of data and materials

Not applicable.

Authors' contributions

JAGV, JFCA, JFZB, KAR and AAN wrote and revised the current review. All authors have read and approved the final manuscript. Data authentication is not applicable.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Fletcher DA and Mullins RD: Cell mechanics and the cytoskeleton. Nature. 463:485–492. 2010.PubMed/NCBI View Article : Google Scholar

2 

Rottner K, Faix J, Bogdan S, Linder S and Kerkhoff E: Actin assembly mechanisms at a glance. J Cell Sci. 130:3427–3435. 2017.PubMed/NCBI View Article : Google Scholar

3 

Jay D, García EJ, Lara JE, Medina MA and de la Luz Ibarra M: Determination of a cAMP-dependent protein kinase phosphorylation site in the C-terminal region of human endothelial actin-binding protein. Arch Biochem Biophys. 377:80–84. 2000.PubMed/NCBI View Article : Google Scholar

4 

Liem RKH and Messing A: Dysfunctions of neuronal and glial intermediate filaments in disease. J Clin Invest. 119:1814–1824. 2009.PubMed/NCBI View Article : Google Scholar

5 

Da Silva JS and Dotti CG: Breaking the neuronal sphere: Regulation of the actin cytoskeleton in neuritogenesis. Nat Rev Neurosci. 3:694–704. 2002.PubMed/NCBI View Article : Google Scholar

6 

Medana IM and Esiri MM: Axonal damage: A key predictor of outcome in human CNS diseases. Brain. 126:515–530. 2003.PubMed/NCBI View Article : Google Scholar

7 

Goodson HV and Jonasson EM: Microtubules and microtubule-associated proteins. Cold Spring Harb Perspect Biol. 10(a022608)2018.PubMed/NCBI View Article : Google Scholar

8 

Jellinger KA: Cell death mechanisms in neurodegeneration. J Cell Mol Med. 5:1–17. 2001.PubMed/NCBI View Article : Google Scholar

9 

Muñoz-Lasso DC, Romá-Mateo C, Pallardó FV and Gonzalez-Cabo P: Much more than a scaffold: Cytoskeletal proteins in neurological disorders. Cells. 9(E358)2020.PubMed/NCBI View Article : Google Scholar

10 

Henriques AG, Müller T, Oliveira JM, Cova M, da Cruz e Silva CB and da Cruz E Silva OA: Altered protein phosphorylation as a resource for potential AD biomarkers. Sci Rep. 6(30319)2016.PubMed/NCBI View Article : Google Scholar

11 

Mietelska-Porowska A, Wasik U, Goras M, Filipek A and Niewiadomska G: Tau protein modifications and interactions: Their role in function and dysfunction. Int J Mol Sci. 15:4671–4713. 2014.PubMed/NCBI View Article : Google Scholar

12 

McMurray CT: Neurodegeneration: Diseases of the cytoskeleton? Cell Death Differ. 7:861–865. 2000.PubMed/NCBI View Article : Google Scholar

13 

Guo T, Noble W and Hanger DP: Roles of tau protein in health and disease. Acta Neuropathol. 133:665–704. 2017.PubMed/NCBI View Article : Google Scholar

14 

Kosik KS, Joachim CL and Selkoe DJ: Microtubule-associated protein tau (tau) is a major antigenic component of paired helical filaments in Alzheimer disease. Acta Neuropathol. 83:4044–4048. 1986.PubMed/NCBI View Article : Google Scholar

15 

Islas-Hernandez A, Aguilar-Talamantes HS, Bertado-Cortes B, Mejia-delCastillo GJ, Carrera-Pineda R, Cuevas-Garcia CF and Garcia-delaTorre P: BDNF and Tau as biomarkers of severity in multiple sclerosis. Biomark Med. 12:717–726. 2018.PubMed/NCBI View Article : Google Scholar

16 

Siller N, Kuhle J, Muthuraman M, Barro C, Uphaus T, Groppa S, Kappos L, Zipp F and Bittner S: Serum neurofilament light chain is a biomarker of acute and chronic neuronal damage in early multiple sclerosis. Mult Scler. 25:678–686. 2019.PubMed/NCBI View Article : Google Scholar

17 

Zetterberg H: Plasma Neurofilament light in progressive multiple sclerosis. Acta Neurol Scand. 141:14–15. 2020.PubMed/NCBI View Article : Google Scholar

18 

GBD 2017 Disease and Injury Incidence and Prevalence Collaborators. Global, Regional, and national incidence, prevalence, and years lived with disability for 354 Diseases and Injuries for 195 countries and territories, 1990-2017: A systematic analysis for the Global Burden of Disease Study 2017. Lancet. 392:1789–1858. 2018.PubMed/NCBI View Article : Google Scholar

19 

Sanchez JS, Hanseeuw BJ, Lopera F, Sperling RA, Baena A, Bocanegra Y, Aguillon D, Guzmán-Vélez E, Pardilla-Delgado E, Ramirez-Gomez L, et al: Longitudinal amyloid and tau accumulation in autosomal dominant Alzheimer's disease: Findings from the Colombia-Boston (COLBOS) biomarker study. Alzheimers Res Ther. 13(27)2021.PubMed/NCBI View Article : Google Scholar

20 

Busche MA and Hyman BT: Synergy between amyloid-β and tau in Alzheimer's disease. Nat Neurosci. 23:1183–1193. 2020.PubMed/NCBI View Article : Google Scholar

21 

Drummond E, Pires G, MacMurray C, Askenazi M, Nayak S, Bourdon M, Safar J, Ueberheide B and Wisniewski T: Phosphorylated tau interactome in the human Alzheimer's disease brain. Brain. 143:2803–2817. 2020.PubMed/NCBI View Article : Google Scholar

22 

Kandimalla R, Manczak M, Yin X, Wang R and Reddy PH: Hippocampal phosphorylated tau induced cognitive decline, dendritic spine loss and mitochondrial abnormalities in a mouse model of Alzheimer's disease. Hum Mol Genet. 27:30–40. 2018.PubMed/NCBI View Article : Google Scholar

23 

Castro-Alvarez JF, Uribe-Arias A, Raigoza DM and Cardona-Gómez GP: Cyclin-dependent kinase 5, a node protein in diminished tauopathy: A systems biology approach. Front Aging Neurosci. 6(232)2014.PubMed/NCBI View Article : Google Scholar

24 

Rudrabhatla P, Jaffe H and Pant HC: Direct evidence of phosphorylated neuronal intermediate filament proteins in neurofibrillary tangles (NFTs): Phosphoproteomics of Alzheimer's NFTs. FASEB J. 25:3896–3905. 2011.PubMed/NCBI View Article : Google Scholar

25 

Richetin K, Steullet P, Pachoud M, Perbet R, Parietti E, Maheswaran M, Eddarkaoui S, Bégard S, Pythoud C, Rey M, et al: Tau accumulation in astrocytes of the dentate gyrus induces neuronal dysfunction and memory deficits in Alzheimer's disease. Nat Neurosci. 23:1567–1579. 2020.PubMed/NCBI View Article : Google Scholar

26 

Ma QL, Zuo X, Yang F, Ubeda OJ, Gant DJ, Alaverdyan M, Kiosea NC, Nazari S, Chen PP, Nothias F, et al: Loss of MAP function leads to hippocampal synapse loss and deficits in the Morris water maze with aging. J Neurosci. 34:7124–7136. 2014.PubMed/NCBI View Article : Google Scholar

27 

Lopez-Tobón A, Cepeda-Prado E and Cardona-Gómez GP: Decrease of tau hyperphosphorylation by 17β estradiol requires sphingosine kinase in a glutamate toxicity model. Neurochem Res. 34:2206–2214. 2009.PubMed/NCBI View Article : Google Scholar

28 

Posada-Duque RA, Ramirez O, Härtel S, Inestrosa NC, Bodaleo F, González-Billault C, Kirkwood A and Cardona-Gómez GP: CDK5 downregulation enhances synaptic plasticity. Cell Mol Life Sci. 74:153–172. 2017.PubMed/NCBI View Article : Google Scholar

29 

Uribe-Arias A, Posada-Duque RA, González-Billault C, Villegas A, Lopera F and Cardona-Gómez GP: p120-catenin is necessary for neuroprotection induced by CDK5 silencing in models of Alzheimer's disease. J Neurochem. 138:624–639. 2016.PubMed/NCBI View Article : Google Scholar

30 

Martin L, Latypova X, Wilson CM, Magnaudeix A, Perrin ML, Yardin C and Terro F: Tau protein kinases: Involvement in Alzheimer's disease. Ageing Res Rev. 12:289–309. 2013.PubMed/NCBI View Article : Google Scholar

31 

Reimer L, Betzer C, Kofoed RH, Volbracht C, Fog K, Kurhade C, Nilsson E, Överby AK and Jensen PH: PKR kinase directly regulates tau expression and Alzheimer's disease-related tau phosphorylation. Brain Pathol. 31:103–119. 2021.PubMed/NCBI View Article : Google Scholar

32 

Posada-Duque RA, López-Tobón A, Piedrahita D, González-Billault C and Cardona-Gómez GP: p35 and Rac1 underlie the neuroprotection and cognitive improvement induced by CDK5 silencing. J Neurochem. 134:354–370. 2015.PubMed/NCBI View Article : Google Scholar

33 

Zheng YL, Li BS, Kanungo J, Kesavapany S, Amin N, Grant P and Pant HC: Cdk5 modulation of mitogen-activated protein kinase signaling regulates neuronal survival. Mol Biol Cell. 18:404–413. 2007.PubMed/NCBI View Article : Google Scholar

34 

Cicero S and Herrup K: Cyclin-dependent kinase 5 is essential for neuronal cell cycle arrest and differentiation. J Neurosci. 25:9658–9668. 2005.PubMed/NCBI View Article : Google Scholar

35 

Piedrahita D, Castro-Alvarez JF, Boudreau RL, Villegas-Lanau A, Kosik KS, Gallego-Gomez JC and Cardona-Gómez GP: β-Secretase 1's targeting reduces hyperphosphorilated tau, implying autophagy actors in 3xTg-AD mice. Front Cell Neurosci. 9(498)2016.PubMed/NCBI View Article : Google Scholar

36 

Choi SH, Kim YH, Hebisch M, Sliwinski C, Lee S, D'Avanzo C, Chen H, Hooli B, Asselin C, Muffat J, et al: A three-dimensional human neural cell culture model of Alzheimer's disease. Nature. 515:274–278. 2014.PubMed/NCBI View Article : Google Scholar

37 

Choi AMK, Ryter SW and Levine B: Autophagy in human health and disease. N Engl J Med. 368:651–662. 2013.PubMed/NCBI View Article : Google Scholar

38 

Komatsu M, Qing JW, Holstein GR, Friedrich VL Jr, Iwata J, Kominami E, Chait BT, Tanaka K and Yue Z: Essential role for autophagy protein Atg7 in the maintenance of axonal homeostasis and the prevention of axonal degeneration. Proc Natl Acad Sci USA. 104:14489–14494. 2007.PubMed/NCBI View Article : Google Scholar

39 

Komatsu M, Waguri S, Chiba T, Murata S, Iwata J, Tanida I, Ueno T, Koike M, Uchiyama Y, Kominami E and Tanaka K: Loss of autophagy in the central nervous system causes neurodegeneration in mice. Nature. 441:880–884. 2006.PubMed/NCBI View Article : Google Scholar

40 

Kast DJ and Dominguez R: The cytoskeleton-autophagy connection. Curr Biol. 27:R318–R326. 2017.PubMed/NCBI View Article : Google Scholar

41 

Villamil Ortiz JG and Cardona Gomez GP: Comparative analysis of autophagy and tauopathy related markers in cerebral ischemia and Alzheimer's disease animal models. Front Aging Neurosci. 7(84)2015.PubMed/NCBI View Article : Google Scholar

42 

Mohan R and John A: Microtubule-associated proteins as direct crosslinkers of actin filaments and microtubules. IUBMB Life. 67:395–403. 2015.PubMed/NCBI View Article : Google Scholar

43 

Xie C, Soeda Y, Shinzaki Y, In Y, Tomoo K, Ihara Y and Miyasaka T: Identification of key amino acids responsible for the distinct aggregation properties of microtubule-associated protein 2 and tau. J Neurochem. 135:19–26. 2015.PubMed/NCBI View Article : Google Scholar

44 

Xie C, Miyasaka T, Yoshimura S, Hatsuta H, Yoshina S, Kage-Nakadai E, Mitani S, Murayama S and Ihara Y: The homologous carboxyl-terminal domains of microtubule-associated protein 2 and Tau induce neuronal dysfunction and have differential fates in the evolution of neurofibrillary tangles. PLoS One. 9(e89796)2014.PubMed/NCBI View Article : Google Scholar

45 

Takahashi RH, Capetillo-Zarate E, Lin MT, Milner TA and Gouras GK: Accumulation of Intraneuronal β-Amyloid 42 peptides is associated with early changes in microtubule-associated protein 2 in neurites and synapses. PLoS One. 8(e51965)2013.PubMed/NCBI View Article : Google Scholar

46 

Yuan A, Sasaki T, Kumar A, Peterhoff CM, Rao MV, Liem RK, Julien JP and Nixon RA: Peripherin is a subunit of peripheral nerve neurofilaments: Implications for differential vulnerability of CNS and peripheral nervous system axons. J Neurosci. 32:8501–8508. 2012.PubMed/NCBI View Article : Google Scholar

47 

Yuan A, Sershen H, Veeranna Basavarajappa BS, Kumar A, Hashim A, Berg M, Lee JH, Sato Y, Rao MV, et al: Neurofilament subunits are integral components of synapses and modulate neurotransmission and behavior in vivo. Mol Psychiatry. 20:986–994. 2015.PubMed/NCBI View Article : Google Scholar

48 

Gafson AR, Barthélemy NR, Bomont P, Carare RO, Durham HD, Julien JP, Kuhle J, Leppert D, Nixon RA, Weller RO, et al: Neurofilaments: Neurobiological foundations for biomarker applications. Brain. 143:1975–1998. 2020.PubMed/NCBI View Article : Google Scholar

49 

Barry DM, Stevenson W, Bober BG, Wiese PJ, Dale JM, Barry GS, Byers NS, Strope JD, Chang R, Schulz DJ, et al: Expansion of Neurofilament Medium C terminus increases axonal diameter independent of increases in conduction velocity or myelin thickness. J Neurosci. 32:6209–6219. 2012.PubMed/NCBI View Article : Google Scholar

50 

Guzmán-Vélez E, Zetterberg H, Fox-Fuller JT, Vila-Castelar C, Sanchez JS, Baena A, Garcia-Ospina G, Aguillon D, Pardilla-Delgado E, Gatchel JR, et al: Associations between plasma neurofilament light, in vivo brain pathology, and cognition in non-demented individuals with autosomal-dominant Alzheimer's disease. Alzheimers Dement. 17:813–821. 2021.PubMed/NCBI View Article : Google Scholar

51 

Quiroz YT, Zetterberg H, Reiman EM, Chen Y, Su Y, Fox-Fuller JT, Garcia G, Villegas A, Sepulveda-Falla D, Villada M, et al: Plasma neurofilament light chain in the presenilin 1 E280A autosomal dominant Alzheimer's disease kindred: A cross-sectional and longitudinal cohort study. Lancet Neurol. 19:513–521. 2020.PubMed/NCBI View Article : Google Scholar

52 

Rajan KB, Aggarwal NT, McAninch EA, Weuve J, Barnes LL, Wilson RS, DeCarli C and Evans DA: Remote blood biomarkers of longitudinal cognitive outcomes in a population study. Ann Neurol. 88:1065–1076. 2020.PubMed/NCBI View Article : Google Scholar

53 

Walsh P, Sudre CH, Fiford CM, Ryan NS, Lashley T, Frost C and Barnes J: ADNI Investigators. The age-dependent associations of white matter hyperintensities and neurofilament light in early- and late-stage Alzheimer's disease. Neurobiol Aging. 97:10–17. 2021.PubMed/NCBI View Article : Google Scholar

54 

Delaby C, Alcolea D, Carmona-Iragui M, Illán-Gala I, Morenas-Rodríguez E, Barroeta I, Altuna M, Estellés T, Santos-Santos M, Turon-Sans J, et al: Differential levels of Neurofilament Light protein in cerebrospinal fluid in patients with a wide range of neurodegenerative disorders. Sci Rep. 10(9161)2020.PubMed/NCBI View Article : Google Scholar

55 

Idland AV, Sala-Llonch R, Borza T, Watne LO, Wyller TB, Brækhus A, Zetterberg H, Blennow K, Walhovd KB and Fjell AM: CSF neurofilament light levels predict hippocampal atrophy in cognitively healthy older adults. Neurobiol Aging. 49:138–144. 2017.PubMed/NCBI View Article : Google Scholar

56 

Henson RL, Doran E, Christian BT, Handen BL, Klunk WE, Lai F, Lee JH, Rosas HD, Schupf N, Zaman SH, et al: Cerebrospinal fluid biomarkers of Alzheimer's disease in a cohort of adults with Down syndrome. Alzheimers Dement (Amst). 12(e12057)2020.PubMed/NCBI View Article : Google Scholar

57 

Sveinsson OA, Kjartansson O and Valdimarsson EM: Cerebral ischemia/infarction-epidemiology, causes and symptoms. Laeknabladid. 100:271–279. 2014.PubMed/NCBI View Article : Google Scholar : (In Icelandic).

58 

Cao L, Tan L, Wang HF, Jiang T, Zhu XC and Yu JT: Cerebral Microinfarcts and dementia: A systematic review and metaanalysis. Curr Alzheimer Res. 14:802–808. 2017.PubMed/NCBI View Article : Google Scholar

59 

Pluta R, Januszewski S and Czuczwar SJ: Brain ischemia as a prelude to Alzheimer's disease. Front Aging Neurosci. 13(636653)2021.PubMed/NCBI View Article : Google Scholar

60 

Yoshimi K, Takeda M, Nishimura T, Kudo T, Nakamura Y, Tada K and Iwata N: An immunohistochemical study of MAP2 and clathrin in gerbil hippocampus after cerebral ischemia. Brain Res. 560:149–158. 1991.PubMed/NCBI View Article : Google Scholar

61 

Vanicky I, Balchen T and Diemer NH: Alterations in MAP2 immunostainability after prolonged complete brain ischaemia in the rat. Neuroreport. 7:161–164. 1995.PubMed/NCBI

62 

Mages B, Fuhs T, Aleithe S, Blietz A, Hobusch C, Härtig W, Schob S, Krueger M and Michalski D: The Cytoskeletal Elements MAP2 and NF-L show substantial alterations in different stroke models while elevated serum levels highlight especially MAP2 as a sensitive biomarker in stroke patients. Mol Neurobiol. 58:4051–4069. 2021.PubMed/NCBI View Article : Google Scholar

63 

Johanna GV, Fredy CA, David VC, Natalia MV, Angel CR and Patricia CG: Rac1 activity changes are associated with neuronal pathology and spatial memory long-term recovery after global cerebral ischemia. Neurochem Int. 57:762–773. 2010.PubMed/NCBI View Article : Google Scholar

64 

Gutiérrez-Vargas JA, Moreno H and Cardona-Gómez GP: Targeting CDK5 post-stroke provides long-term neuroprotection and rescues synaptic plasticity. J Cereb Blood Flow Metab. 37:2208–2223. 2017.PubMed/NCBI View Article : Google Scholar

65 

Pérez-Corredor PA, Gutiérrez-Vargas JA, Ciro-Ramírez L, Balcazar N and Cardona-Gómez GP: High fructose diet-induced obesity worsens post-ischemic brain injury in the hippocampus of female rats. Nutr Neurosci: Mar 2, 2020 (Epub ahead of print).

66 

Dawson DA and Hallenbeck JM: Acute focal ischemia-induced alterations in MAP2 immunostaining: Description of temporal changes and utilization as a marker for volumetric assessment of acute brain injury. J Cereb Blood Flow Metab. 16:170–174. 1996.PubMed/NCBI View Article : Google Scholar

67 

Akulinin VA and Dahlstrom A: Quantitative analysis of MAP2 immunoreactivity in human neocortex of three patients surviving after brain ischemia. Neurochem Res. 28:373–378. 2003.PubMed/NCBI View Article : Google Scholar

68 

Pluta R, Ułamek-Kozioł M, Januszewski S and Czuczwar SJ: Tau protein dysfunction after brain ischemia. J Alzheimers Dis. 66:429–437. 2018.PubMed/NCBI View Article : Google Scholar

69 

Gutiérrez-Vargas JA, Múnera A and Cardona-Gómez GP: CDK5 knockdown prevents hippocampal degeneration and cognitive dysfunction produced by cerebral ischemia. J Cereb Blood Flow Metab. 35:1937–1949. 2015.PubMed/NCBI View Article : Google Scholar

70 

Pluta R, Bogucka-Kocka A, Ułamek-Kozioł M, Bogucki J, Januszewski S, Kocki J and Czuczwar SJ: 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. 70:881–884. 2018.PubMed/NCBI View Article : Google Scholar

71 

Mailliot C, Podevin-Dimster V, Rosenthal RE, Sergeant N, Delacourte A, Fiskum G and Buée L: Rapid tau protein dephosphorylation and differential rephosphorylation during cardiac arrest-induced cerebral ischemia and reperfusion. J Cereb Blood Flow Metab. 20:543–549. 2000.PubMed/NCBI View Article : Google Scholar

72 

Wen Y, Yang S, Liu R and Simpkins JW: Transient cerebral ischemia induces site-specific hyperphosphorylation of tau protein. Brain Res. 1022:30–38. 2004.PubMed/NCBI View Article : Google Scholar

73 

Uchihara T, Nakamura A, Arai T, Ikeda K and Tsuchiya K: Microglial tau undergoes phosphorylation-independent modification after ischemia. Glia. 45:180–187. 2004.PubMed/NCBI View Article : Google Scholar

74 

Fujii H, Takahashi T, Mukai T, Tanaka S, Hosomi N, Maruyama H, Sakai N and Matsumoto M: Modifications of tau protein after cerebral ischemia and reperfusion in rats are similar to those occurring in Alzheimer's disease-Hyperphosphorylation and cleavage of 4- and 3-repeat tau. J Cereb Blood Flow Metab. 37:2441–2457. 2017.PubMed/NCBI View Article : Google Scholar

75 

Shiiya N, Kunihara T, Miyatake T, Matsuzaki K and Yasuda K: Tau protein in the cerebrospinal fluid is a marker of brain injury after aortic surgery. Ann Thorac Surg. 77:2034–2038. 2004.PubMed/NCBI View Article : Google Scholar

76 

Hesse C, Rosengren L, Andreasen N, Davidsson P, Vanderstichele H, Vanmechelen E and Blennow K: Transient increase in total tau but not phospho-tau in human cerebrospinal fluid after acute stroke. Neurosci Lett. 297:187–190. 2001.PubMed/NCBI View Article : Google Scholar

77 

Onatsu J, Vanninen R, JÄkÄlÄ P, Mustonen P, Pulkki K, Korhonen M, Hedman M, HÖglund K, Blennow K, Zetterberg H, et al: Tau, S100B and NSE as blood biomarkers in acute cerebrovascular events. In Vivo. 34:2577–2586. 2020.PubMed/NCBI View Article : Google Scholar

78 

Bitsch A, Horn C, Kemmling Y, Seipelt M, Hellenbrand U, Stiefel M, Ciesielczyk B, Cepek L, Bahn E, Ratzka P, et al: Serum tau protein level as a marker of axonal damage in acute ischemic stroke. Eur Neurol. 47:45–51. 2002.PubMed/NCBI View Article : Google Scholar

79 

Kurzepa J, Bielewicz J, Grabarska A, Stelmasiak Z, Stryjecka-Zimmer M and Bartosik-Psujek H: Matrix metalloproteinase-9 contributes to the increase of tau protein in serum during acute ischemic stroke. J Clin Neurosci. 17:997–999. 2010.PubMed/NCBI View Article : Google Scholar

80 

Lasek-Bal A, Jedrzejowska-Szypulka H, Rozycka J, Bal W, Kowalczyk A, Holecki M, Dulawa J and Lewin-Kowalik J: The presence of Tau protein in blood as a potential prognostic factor in stroke patients. J Physiol Pharmacol. 67:691–696. 2016.PubMed/NCBI

81 

Bielewicz J, Kurzepa J, Czekajska-Chehab E, Stelmasiak Z and Bartosik-Psujek H: Does serum Tau protein predict the outcome of patients with ischemic stroke? J Mol Neurosci. 43:241–245. 2011.PubMed/NCBI View Article : Google Scholar

82 

Wunderlich MT, Lins H, Skalej M, Wallesch CW and Goertler M: Neuron-specific enolase and tau protein as neurobiochemical markers of neuronal damage are related to early clinical course and long-term outcome in acute ischemic stroke. Clin Neurol Neurosurg. 108:558–563. 2006.PubMed/NCBI View Article : Google Scholar

83 

De Vos A, Bjerke M, Brouns R, De Roeck N, Jacobs D, Van den Abbeele L, Guldolf K, Zetterberg H, Blennow K, Engelborghs S and Vanmechelen E: Neurogranin and tau in cerebrospinal fluid and plasma of patients with acute ischemic stroke. BMC Neurol. 17(170)2017.PubMed/NCBI View Article : Google Scholar

84 

Irving EA, Nicoll J, Graham DI and Dewar D: Increased tau immunoreactivity in oligodendrocytes following human stroke and head injury. Neurosci Lett. 213:189–192. 1996.PubMed/NCBI View Article : Google Scholar

85 

Uphaus T, Bittner S, Gröschel S, Steffen F, Muthuraman M, Wasser K, Weber-Krüger M, Zipp F, Wachter R and Gröschel K: NfL (Neurofilament Light Chain) levels as a predictive marker for long-term outcome after ischemic stroke. Stroke. 50:3077–3084. 2019.PubMed/NCBI View Article : Google Scholar

86 

Khalil M, Teunissen CE, Otto M, Piehl F, Sormani MP, Gattringer T, Barro C, Kappos L, Comabella M, Fazekas F, et al: Neurofilaments as biomarkers in neurological disorders. Nat Rev Neurol. 14:577–589. 2018.PubMed/NCBI View Article : Google Scholar

87 

Peters N, van Leijsen E, Tuladhar AM, Barro C, Konieczny MJ, Ewers M, Lyrer P, Engelter ST, Kuhle J, Duering M and de Leeuw FE: Serum Neurofilament light Chain is associated with incident Lacunes in progressive cerebral small vessel disease. J Stroke. 22:369–376. 2020.PubMed/NCBI View Article : Google Scholar

88 

Duering M, Konieczny MJ, Tiedt S, Baykara E, Tuladhar AM, Leijsen EV, Lyrer P, Engelter ST, Gesierich B, Achmüller M, et al: Serum Neurofilament Light Chain levels are related to small vessel disease burden. J Stroke. 20:228–238. 2018.PubMed/NCBI View Article : Google Scholar

89 

Paolini Paoletti F, Simoni S, Parnetti L and Gaetani L: The contribution of small vessel disease to neurodegeneration: Focus on Alzheimer's disease, Parkinson's disease and multiple sclerosis. Int J Mol Sci. 22(4958)2021.PubMed/NCBI View Article : Google Scholar

90 

Knopman DS: Cerebrovascular disease and dementia. Br J Radiol. 80 (Suppl 2):S121–S127. 2007.PubMed/NCBI View Article : Google Scholar

91 

Dendrou CA, Fugger L and Friese MA: Immunopathology of multiple sclerosis. Nat Rev Immunol. 15:545–558. 2015.PubMed/NCBI View Article : Google Scholar

92 

Virgilio E, Vecchio D, Crespi I, Serino R, Cantello R, Dianzani U and Comi C: Cerebrospinal Tau levels as a predictor of early disability in multiple sclerosis. Mult Scler Relat Disord. 56(103231)2021.PubMed/NCBI View Article : Google Scholar

93 

Mirzaii-Dizgah MH, Mirzaii-Dizgah MR and Mirzaii-Dizgah I: Serum and saliva total tau protein as a marker for relapsing-remitting multiple sclerosis. Med Hypotheses. 135(109476)2020.PubMed/NCBI View Article : Google Scholar

94 

Shafit-Zagardo B, Kress Y, Zhao ML and Lee SC: A novel microtubule-associated protein-2 expressed in oligodendrocytes in multiple sclerosis lesions. J Neurochem. 73:2531–2537. 1999.PubMed/NCBI View Article : Google Scholar

95 

Wang P, Jiang LL, Wang C, Zhu Z and Lai C: Neurofilament protein as a potential biomarker of axonal degeneration in experimental autoimmune encephalomyelitis. Neurol India. 68:364–367. 2020.PubMed/NCBI View Article : Google Scholar

96 

Rosengren LE, Karlsson JE, Karlsson JO, Persson LI and Wikkelsø C: Patients with amyotrophic lateral sclerosis and other neurodegenerative diseases have increased levels of neurofilament protein in CSF. J Neurochem. 67:2013–2018. 1996.PubMed/NCBI View Article : Google Scholar

97 

Lycke JN, Karlsson JE, Andersen O and Rosengren LE: Neurofilament protein in cerebrospinal fluid: A potential marker of activity in multiple sclerosis. J Neurol Neurosurg Psychiatry. 64:402–404. 1998.PubMed/NCBI View Article : Google Scholar

98 

Kuhle J, Barro C, Andreasson U, Derfuss T, Lindberg R, Sandelius Å, Liman V, Norgren N, Blennow K and Zetterberg H: Comparison of three analytical platforms for quantification of the neurofilament light chain in blood samples: ELISA, electrochemiluminescence immunoassay and Simoa. Clin Chem Lab Med. 54:1655–1661. 2016.PubMed/NCBI View Article : Google Scholar

99 

Varhaug KN, Torkildsen Ø, Myhr KM and Vedeler CA: Neurofilament light Chain as a biomarker in multiple sclerosis. Front Neurol. 10(338)2019.PubMed/NCBI View Article : Google Scholar

100 

Norgren N, Rosengren L and Stigbrand T: Elevated neurofilament levels in neurological diseases. Brain Res. 987:25–31. 2003.PubMed/NCBI View Article : Google Scholar

101 

Cai L and Huang J: Neurofilament light chain as a biological marker for multiple sclerosis: A meta-analysis study. Neuropsychiatr Dis Treat. 14:2241–2254. 2018.PubMed/NCBI View Article : Google Scholar

102 

Disanto G, Barro C, Benkert P, Naegelin Y, Schädelin S, Giardiello A, Zecca C, Blennow K, Zetterberg H, Leppert D, et al: Serum Neurofilament light: A biomarker of neuronal damage in multiple sclerosis. Ann Neurol. 81:857–870. 2017.PubMed/NCBI View Article : Google Scholar

103 

Barro C, Benkert P, Disanto G, Tsagkas C, Amann M, Naegelin Y, Leppert D, Gobbi C, Granziera C, Yaldizli Ö, et al: Serum neurofilament as a predictor of disease worsening and brain and spinal cord atrophy in multiple sclerosis. Brain. 141:2382–2391. 2018.PubMed/NCBI View Article : Google Scholar

104 

Arrambide G, Espejo C, Eixarch H, Villar LM, Alvarez-Cermeño JC, Picón C, Kuhle J, Disanto G, Kappos L, Sastre-Garriga J, et al: Neurofilament light chain level is a weak risk factor for the development of MS. Neurology. 87:1076–1084. 2016.PubMed/NCBI View Article : Google Scholar

105 

Comabella M and Montalban X: Body fluid biomarkers in multiple sclerosis. Lancet Neurol. 13:113–126. 2014.PubMed/NCBI View Article : Google Scholar

106 

Matute-Blanch C, Villar LM, Álvarez-Cermeño JC, Rejdak K, Evdoshenko E, Makshakov G, Nazarov V, Lapin S, Midaglia L, Vidal-Jordana A, et al: Neurofilament light chain and oligoclonal bands are prognostic biomarkers in radiologically isolated syndrome. Brain. 141:1085–1093. 2018.PubMed/NCBI View Article : Google Scholar

107 

Khalil M: Are neurofilaments valuable biomarkers for long-term disease prognostication in MS? Mult Scler. 24:1270–1271. 2018.PubMed/NCBI View Article : Google Scholar

108 

Giovannoni G: Peripheral blood neurofilament light chain levels: The neurologist's C-reactive protein? Brain. 141:2235–2237. 2018.PubMed/NCBI View Article : Google Scholar

Related Articles

Journal Cover

April-2022
Volume 16 Issue 4

Print ISSN: 2049-9434
Online ISSN:2049-9442

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Gutiérrez‑Vargas JA, Castro‑Álvarez JF, Zapata‑Berruecos JF, Abdul‑Rahim K and Arteaga‑Noriega A: Neurodegeneration and convergent factors contributing to the deterioration of the cytoskeleton in Alzheimer's disease, cerebral ischemia and multiple sclerosis (Review). Biomed Rep 16: 27, 2022
APA
Gutiérrez‑Vargas, J.A., Castro‑Álvarez, J.F., Zapata‑Berruecos, J.F., Abdul‑Rahim, K., & Arteaga‑Noriega, A. (2022). Neurodegeneration and convergent factors contributing to the deterioration of the cytoskeleton in Alzheimer's disease, cerebral ischemia and multiple sclerosis (Review). Biomedical Reports, 16, 27. https://doi.org/10.3892/br.2022.1510
MLA
Gutiérrez‑Vargas, J. A., Castro‑Álvarez, J. F., Zapata‑Berruecos, J. F., Abdul‑Rahim, K., Arteaga‑Noriega, A."Neurodegeneration and convergent factors contributing to the deterioration of the cytoskeleton in Alzheimer's disease, cerebral ischemia and multiple sclerosis (Review)". Biomedical Reports 16.4 (2022): 27.
Chicago
Gutiérrez‑Vargas, J. A., Castro‑Álvarez, J. F., Zapata‑Berruecos, J. F., Abdul‑Rahim, K., Arteaga‑Noriega, A."Neurodegeneration and convergent factors contributing to the deterioration of the cytoskeleton in Alzheimer's disease, cerebral ischemia and multiple sclerosis (Review)". Biomedical Reports 16, no. 4 (2022): 27. https://doi.org/10.3892/br.2022.1510