Review

Molecular Biomarkers for Diagnosis & Therapies of Alzheimer’s Disease

  • Received: 26 August 2016 Accepted: 26 October 2016 Published: 02 November 2016
  • Alzheimer’s disease (AD) has been discovered before the century but scientists still have not found the way to cure the disease. The basic requirement for successful cure requires the early diagnosis of the disease. Presence of Aβ42, total tau protein and phosphorylated tau have been used in the earlier days but these diagnostic markers fail the detection in the initial stages. Hence the need of the hour is to identify the various biomarkers which can be detected in the earlier stages of AD. Impaired cellular signaling is common in all the diseases and identification of particular signaling pathway helps in the identification of biomarker. Important signaling pathways such as Akt, FAS/NO, MAPK, Ca2+ are found to be altered in the AD brain. Protein molecules upstream or downstream to these signaling molecules can be potential molecular markers in the diagnosis of AD. This review comprises molecular markers which are crucial in the AD and play a significant role by altering the signaling pathways. These biomarkers will not only help in the understanding of pathobiology of AD but also provide an insight for the researchers working in the direction of AD biomarker discovery and drug discovery.

    Citation: Pravin D Potdar, Aashutosh U Shetti. Molecular Biomarkers for Diagnosis & Therapies of Alzheimer’s Disease[J]. AIMS Neuroscience, 2016, 3(4): 433-453. doi: 10.3934/Neuroscience.2016.4.433

    Related Papers:

    [1] Danton H. O’Day . Alzheimer’s Disease: A short introduction to the calmodulin hypothesis. AIMS Neuroscience, 2019, 6(4): 231-239. doi: 10.3934/Neuroscience.2019.4.231
    [2] Craig T. Vollert, Jason L. Eriksen . Microglia in the Alzheimers brain: a help or a hindrance?. AIMS Neuroscience, 2014, 1(3): 210-224. doi: 10.3934/Neuroscience.2014.3.210
    [3] Kiran Kumar Siddappaji, Shubha Gopal . Molecular mechanisms in Alzheimer's disease and the impact of physical exercise with advancements in therapeutic approaches. AIMS Neuroscience, 2021, 8(3): 357-389. doi: 10.3934/Neuroscience.2021020
    [4] Marco Calabrò, Carmela Rinaldi, Giuseppe Santoro, Concetta Crisafulli . The biological pathways of Alzheimer disease: a review. AIMS Neuroscience, 2021, 8(1): 86-132. doi: 10.3934/Neuroscience.2021005
    [5] Lourdes A. Vega Rasgado, Arantxa Tabernero Urbieta, José María Medina Jiménez . Affected albumin endocytosis as a new neurotoxicity mechanism of amyloid beta. AIMS Neuroscience, 2020, 7(3): 344-359. doi: 10.3934/Neuroscience.2020021
    [6] Jean A. Rondal . Down syndrome: A curative prospect?. AIMS Neuroscience, 2020, 7(2): 168-193. doi: 10.3934/Neuroscience.2020012
    [7] Frank O. Bastian . Is Alzheimers Disease Infectious?
    Relative to the CJD Bacterial Infection Model of Neurodegeneration. AIMS Neuroscience, 2015, 2(4): 240-258. doi: 10.3934/Neuroscience.2015.4.240
    [8] Khue Vu Nguyen . Special Issue: Alzheimer’s disease. AIMS Neuroscience, 2018, 5(1): 74-80. doi: 10.3934/Neuroscience.2018.1.74
    [9] Arosh S. Perera Molligoda Arachchige, Anton Kristoffer Garner . Seven Tesla MRI in Alzheimer's disease research: State of the art and future directions: A narrative review. AIMS Neuroscience, 2023, 10(4): 401-422. doi: 10.3934/Neuroscience.2023030
    [10] Khue Vu Nguyen . β-Amyloid precursor protein (APP) and the human diseases. AIMS Neuroscience, 2019, 6(4): 273-281. doi: 10.3934/Neuroscience.2019.4.273
  • Alzheimer’s disease (AD) has been discovered before the century but scientists still have not found the way to cure the disease. The basic requirement for successful cure requires the early diagnosis of the disease. Presence of Aβ42, total tau protein and phosphorylated tau have been used in the earlier days but these diagnostic markers fail the detection in the initial stages. Hence the need of the hour is to identify the various biomarkers which can be detected in the earlier stages of AD. Impaired cellular signaling is common in all the diseases and identification of particular signaling pathway helps in the identification of biomarker. Important signaling pathways such as Akt, FAS/NO, MAPK, Ca2+ are found to be altered in the AD brain. Protein molecules upstream or downstream to these signaling molecules can be potential molecular markers in the diagnosis of AD. This review comprises molecular markers which are crucial in the AD and play a significant role by altering the signaling pathways. These biomarkers will not only help in the understanding of pathobiology of AD but also provide an insight for the researchers working in the direction of AD biomarker discovery and drug discovery.


    1. Introduction

    Among many other neurological diseases Alzheimer’s disease is the most wide spread and deadliest disorder. The symptoms of this disease start with frequent memory loss but ends with severe immobility, total loss of verbal skill and confusion about past and present. In the current scenario by the time the individual is diagnosed with AD, he is almost into the 2nd or 3rd stage of the disease. According to Alzheimer’s Association, AD is the 6th leading cause of death in USA and every 66 seconds someone in the USA develops AD. According to Alzheimer’s disease International UK, approximately worldwide 46.8 million people were affected with AD in 2015 and the number will rise by 68% by 2050. The numbers suggest why it is important to study the pathobiology of Alzheimer’s disease and come up with molecular marker for early detection and treatment. In order to develop a biomarker it is essential to understand the mechanism behind the development and progression of disease.


    2. Mechanism behind the Alzheimer’s Disease

    Memory loss in an AD is a result of constant neuronal damage caused by development of amyloid plaque and neurofibrillary tangles (NFTs) [1,2]. Amyloid plaques mainly comprises of insoluble Aβ fragments of varying amino acid length [3]. These fragments are generally formed due to the cleavage of amyloid precursor protein abbreviated as APP. APP is a substrate of variety of proteases including α-secretase, β-secretase and γ-secretase which are involved in the pathogenesis of AD [4]. APP is processed by one of the two competing pathways. In a healthy person the pathway which is predominated involves, cleavage of APP by α-secretase and γ-secretase resulting in the formation of soluble fragment [4]. This soluble fragment is generally known as sAPPα. The generated sAPPα fragment has shown to have various neuro-protective effective [5]. The other pathway involves β and γ-secretase cleavage of APP. This pathway results in the release of insoluble Aβ fragments which are capable of forming toxic amyloid plaques [4]. Cathespins, an another class of proteases have also shown β-secretase like activity [6]. APP cleaving by β and γ-secretase also releases amyloid precursor protein intracellular domain (AICD) [7]. This AICD generated due to amyloidgenic pathway can only perform nuclear signaling which may result in the progression of Alzheimer’s disease [8]. AICD act as a gene expression regulator, controlling expression of many genes related to AD pathology including NFT [8].

    NFT is another important pathological condition related to AD [9]. The most crucial component of these NFTs is hyper-phosphorylated tau (pTau) protein [9]. The hyper-phosphorylation is carried out by variety of protein kinases, some of which are GSK3β, CDK-5, mitogen activated protein kinase (MAPK1) and casein kinase 1 [10]. Tau proteins are usually associated within the neurons and causes many neurological disorders which involve Alzheimer’s disease, Pick’s disease, progressive supranuclear palsy are some of the taupathies extensively studied [11]. Figure 1 shows the generalized processing of APP and Tau aggregation in normal vs AD brain.

    Figure 1. Schematic differentiation between Non-amyloidogenic and Amyloidogenic Pathway Genetic mutations behind Alzheimer’s disease.

    AD is broadly divided into two types, sporadic and familial AD. Sporadic is further classified into two categories, early onset and late onset AD. Early-onset AD (EOAD) is rarely observed where the people are affected and diagnosed with AD before the age of 65. Late onset AD (LOAD) is very frequent form of dementia affecting in people of age more than 65. Familial type of AD (FAD) is a form where the disease is observed within the family due to mutation and is inherited in the generations. Point mutation at SMT2 gene on chromosome 1 is responsible for AD [12]. Chromosome10 have various loci which were observed to increase the Aβ accumulation in AD patients. Many of the loci were found near Insulin degrading enzyme (IDE) [13]. IDE is capable of degrading Aβ fragments. This suggests that mutation at gene coding for IDE may result in abnormal increase in Aβ accumulation and amyloid formation. A risk locus has been found on chromosome 12 [14]. The SNP is observed near the gene coding for Vitamin D receptor (VDR). Vitamin D deficiency is associated with learning and memory dysfunction in adults [15]. Hence it is possible that alteration in the expression of VDR may lead to memory impairment in AD patients. Evidence exist for locus near to α-1-antichymotrypsin gene present on chromosome 14 can also results in FAD [16,17]. The well-known mutation in the AD is mutation in the gene coding for presenilins (PSEN). Presenilins 1/2 are important component of APP cleaving enzyme γ-secretase. Gene for presenilin 1 is present on the chromosome 14 whereas gene for presenilin 2 is observed on chromosome 1. Several studies show that mutation in presenilin 2 is a relatively rare cause of AD when compared to presenilin 1. Chromosome 19 is also associated with both sporadic and familial AD due to apolipoprotein E gene allele ε4 located at 19q13.2 [18,19]. Apoliprotein E combines with the fats to form lipoprotein, this lipoprotein carries out transfer of cholesterol along the bloodstream. Individual inheriting one or more than one copy of apoliprotein ε4 allele are said to be more susceptible for AD [20]. Merely exhibiting apo ε4 allele does not necessitates AD, since study exist where apo ε2 and apo ε3 can lead to taupathies observed in AD [21]. Last but not the least, mutation of an APP coding gene on chromosome 21 is considered important for an EOAD [22]. All these mutations are useful for the diagnosis of familial type of AD i.e. early onset Alzheimer’s disease. The diagnosis of LOAD is carried out by neuropsychological testing and brain imaging. Brain imaging studies involve techniques using Magnetic resonance imaging (MRI), computerized tomography and positron emission tomography.


    3. Molecular Markers Involved in AD

    In the early stages of AD, symptoms of the disease coincides with the other neurological dementia. It is important to distinguish the AD from the other diseases in the early stages. Established biomarkers involved in AD are based on the analysis of cerebrospinal fluid (CSF) content for lowered Aβ1-42, increased phosphorylated tau (pTau) and total tau (tTau) [23]. These biomarkers can only be seen in the CAF in later stages of AD. Hence it is crucial to identify early diagnostic markers for AD, since early diagnosis helps in starting the treatment in the earlier stages. These biomarkers can be either CSF based or blood based biomarkers. AD is a complex disease and the actual cause of the disease is unknown. In the initial stage Aβ1-42 and tau proteins act as signaling molecule in AD and they cause the disease progression by impairing cellular signaling. But what causes the production of Aβ and tau is a mystery and faulty cellular signaling can be a potential reason. In this review, we have tried to summarize about these potential biomarkers for the diagnosis of AD. These biomarkers are important since they are related to the production of Aβ fragments, tau filaments and the aggregation of these proteins. There are some molecular markers which are also associated with other neurological disease which can be used along with other AD markers in chip based or micro-array based method for the diagnosis of AD. These molecular markers can be classified under 3 types which are Aβ aggregation related markers, NFT Formation related markers and Ca2+ signaling pathway related molecular markers.


    4. Aβ1-42 Aggregation Related Molecular Markers


    4.1. Human Kallikrein 6

    Kallikreins belong to the subfamily of serine proteases which can cleave a vasoactive peptide known as kinin from the kininogen. Human kallikrein 6 is one of the various isoforms of kallikrein and it has several names such as zyme, protease M and neurosin. It is highly expressed in the neurons along with many other cell types [24]. The important function of this enzyme is to degrade the α-synuclein (α-syn) which is released by the neuronal cells and is harmful outside the cell. α-syn forms the protein aggregates and spread in similar fashion as that of amyloid plaque and the disease condition is known as synucleinopathies [25]. Some of the examples of synucleinopathies are Parkinson’s disease (PD), pure autonomic failure, multiple system atrophy and dementia with lewy bodies [26]. Comparison of kallikrein 6 immunolabeling performed on AD, PD and control patient revealed that the enzyme was rarely present in the damaged area of brain and RT-PCR showed decreased m-RNA indicating low gene expression of kallikrain 6 [27] which shows non specificity of kallikrain as a biomarker for AD. Since kallikrain is a serine protease it may possess the Aβ clearance activity and decrease in its expression may be associated with the Aβ accumulation as shown in figure 2. Study conducted by Diamandis et al. 2000 reported first time the increased concentration of kallikrain in CSF of AD patients and proposed the idea of using this enzyme as a biomarker in AD. This suggest that protein and m-RNA level of kallikrein 6 can be done in the brain as well as CSF. The study was conducted on the small group of people and required further confirmation in large sample size [28].

    Figure 2. Signaling cascade showing potential biomarkers involved in AD

    4.2. Hypocretin-1/Orexin A

    Hypocretin name was given based on their secretion from hypothalamus. Hypocretins are involved in the formation of neurotransmitter system [29]. These are the important peptide which have the ability to regulate neuroexcitory activity, circadian rhythm and sleep [30]. Hypocretin-1 is widely studied for their role in the neurological disorders which include AD, PD and Huntington’s disease (HD). In PD and HD patients there is no significant alteration in CSF levels of hypocretins-1 [31,32]. This may state that hypocretin can be a specific biomarker for AD. In AD patient’s CSF increased hypocretin-1 levels in the initial staged were observed by Dauviellers et al. Its expression in CSF is increased along with the tTau and pTau content which causes sleep impairment mainly insomnia. In vivo study has shown the effect of sleep impairment on the aggregation of the Aβ. Sleep restriction caused increase in the rate of Aβ accumulation. Sleep enhancement via blockage of orexin receptor inhibited the Aβ accumulation. The protein level were determined in the CSF of the sample [33]. The contradicting results have been shown by Friedman et al. 2007 stating decreased hypocretin-1 level in the early stages of AD. This study was supported by another study conducted by Slats et al. 2012 which showed decreased hypocretin-1 and Aβ42 in CSF of AD patient [34,35]. One of the studies has revealed gender specificity for immunoreactivity of hypocretin producing neurons in control group. Female AD patients have shown less orexin producing neurons when compared with the male AD patient [36]. Polymorphism in the gene HCRTR2 expressing hypocretin receptor 2 is also associated with the risk of AD [37]. Overall hypocretin levels in CSF can be used as a potential biomarker, but the various contradicting results along with the gender specificity discourage its exploration in biomarker related aspects.


    4.3. Neuroserpins

    Some of the proteases have the ability to degrade the Aβ filaments and oligomers, for example plasmin. Plasmin is serine protease which are produced as a non-active form called plasminogen [38]. The conversion of inactive plasminogen to active plasmin is carried out by the tissue plasminogen activator (tPA). Formation of plasmin is inhibited by inactivation of tPA which is carried out by neuroserpins. Neuroserpins are exclusively found in brain tissue and it is involved in morphological maturation of neurons [39]. In AD brain plasmin and tPA both have shown the neuro-protective nature. Plasmin and tPA can both increase APP processing by α-secretase [40]. Increased expression of neuroserpins has been observed in CSF of AD patients [41]. The neuroserpins and tPA both are co-localized with amyloid plaque in AD patient brain [42]. Knocking out of neuroserpins reduced the Aβ aggregation and showed almost normal memory function [43]. Interestingly study performed by Kinghorn et al. 2006 showed that the complex formed by the neuroserpins and Aβ is neuroprotective in nature [39]. Up-regulated neuroserpins expression observed by western blotting is co-related with the increased expression of Thyroid hormone rerceptor-1-β (THR-1β) and RNA-binding protein HuD. Binding of thyroid hormone to THR-1β induces expression of many genes one of which is HuD. The HuD increases the expression of neuroserpins by stabilizing the mRNA. This suggest that thyroid hormone response system is playing an important role in the development and progression of AD [44]. Overall, neuroserpins protein level quantification can be done in CSF of AD patient but further studies have to be done in order to validate the biomarker potential of neuroserpin. Also there is no evidence for the up-regulated expression of neuroserpins in PD and HD in order to understand its specificity as an AD Biomarker.


    4.4. Spingosine-1-phosphate (S1P)

    S1P is phosphorylated form of spingosine, a membrane lipid. The reaction is catalyzed by enzymes spingosine kinase 1 (SphK1) and spingosine kinase 2 (SphK2) [45]. The expression of SphK1 is decreased by the Aβ42 whereas, SphK2 expression is not altered [46]. SphK1 play an important role in the neuroprotection by activation of Insulin-like growth factor (IGF-I) as shown in figure 2. IGF-I is an important factor in AD since it is one of the enzyme having capability to cleave the Aβ peptides [47]. The phosphorylation of spingosine carried out by SphK1 results in the product S1P. S1P is an important signaling and cytoprotective molecule acting via G-protein coupled receptor [45]. It is also important in neuronal development since it is involved in the signaling pathways of neurogenesis and angiogenesis[48]. The link between AD and spingosine starts from the ApoE. Where the secretion of S1P is controlled by the ApoE. The variation in the ApoE gene and reduced expression of SphK1 can result in the loss of S1P [49]. This study was done by performing western blotting on the brain tissue sample of AD patient. It showed region specific expression of S1P. It was expressed in regions of brains which are affected early in the AD this involves hippocampus and temporal lobe. The loss in the S1P and SphK1 is found in the early AD event which states its importance in its use as a biomarker in AD. However down-regulated S1P is observed in case of PD also [50]. S1P is degraded by the enzyme sphingosine-1-phosphate lyase (SPL). Immunohistochemistry have revealed that in AD brain SPL expression is increased whereas, expression of SphK1 is reduced [49]. Interestingly experiment carried out by Takasugi et al. 2012 reported increased expression of SphK2 in AD patients. Importantly S1P was found to be associated with the activity of β-secretase [51]. Hagen et al. 2011 found that whenever the S1P is produced by the SphK2 it induces the neurodegeneration [52].


    4.5. Cystatin C (Cys C)

    Cys C also called as γ-trace was first identified in CSF. The Cys C have many biological activities which includes tumor metastasis, cell proliferation, growth and inflammatory response modulation [53]. The decrease in the Cys C is observed in various neurological disease which include ALS and AD [54,55]. Whereas in case of PD, protein is found to be elevated in the serum of a patient [56]. The Cys C levels in the CSF and plasma were found to be lowered in AD patients which was measured by ELISA [57]. The polymorphism within the CST3 gene coding for Cys C have shown increased susceptibility to cause AD. The polymorphism induced AD condition was seen in the LOAD than the EOAD [58]. Earlier in this article we stated presenilin 1 mutation causing AD when compared to presenilin 2. But the recent studies have shown mutation in presenilin 2 caused reduced expression of Cys C [59]. Taking into consideration the neuroprotective activity of Cys C it can be said that, though presenilin 2 mutation is less likely involved in the AD pathogenesis but it may be the crucial component in progression of AD. One of the important function of Cys C is inhibition of cathespin B as shown in figure 2 [60]. Hence decreased Cys C leads to the increased amyloid plaque formation. Binding of Cys C to Aβ also inhibit the amyloid plaque formation [61]. Contradicting to neuroprotection of Cys C by binding to Aβ another study stated that aggregation of Cys C and Aβ in cerebral vessels will lead to the cerebral hemorrhage in the AD patient [62]. Also strong Cys C neuronal immunostaining was seen in the AD patient brain whereas, the staining was absent in the control. Also the neurons showing the immunostaining were most susceptible to the cellular death when compared to the neurons without staining [63]. The role of Cys C in the AD development is controversial also taking into an account it is deregulated in various neurological disease it is important to consider it with multiple AD related markers.


    4.6. Calsyntenin (Clstn)

    Calsyntenin are transmembrane protein belonging to the cadherins superfamily. These are synaptic proteins expressed in 3 forms Clstn1, 2 and 3. Clstn1 is an adaptor protein which is involved in trafficking and processing of APP [64]. Clstn1 is reduced in the AD brain and it plays crucial role in the increased production of Aβ and hence it makes Clstn1 a potential biomarker for the AD. The quantification of protein was done by immunoblot assay on the frozen tissue of AD brain. APP within the neurons is transported through motor protein kinesin-1. Clstn1 is protein which bind to this kinesin and it is colocalized with APP in the vesicles [65]. Whether the presence of Clstn1 for the transport of APP within transporting vesicles is important or not is yet to be evaluated. Clstn1 is found to be down-regulated in CSF of both AD and PD [66]. As Clstn1 is down-regulated in AD and PD, for further diagnostics, AD specific marker need to be analyzed simultaneously, most probably by chip based assay. Studies have demonstrated that Clstn1 protects the APP cleavage by BACE1 within the vesicles. Decreased expression of Clstn1 mediated via si-RNA increased the APP processing by BACE1. This suggest the association between the APP and Clstn1 which protects from the toxic Aβ generation [67]. Clearly further study needs to be done in order to establish Clstn1 as a biomarker for early diagnosis of AD. The other calsyntetin, Clstn 3 is also found to be related with the AD. Clstn3 is important neurodevelopmental molecule correlated with the cognition. Clstn 3 is up-regulated in the AD brain and the up-regulation is mainly induced by the Aβ. Quantification of protein was done by immunofluorescence assay on transformed embryonic rat cells. The increased expression of Clstn-3 leads to the death of the neurons. The Clstn 3 surround the amyloid plaque generated [68]. Further studies showed Clstn 3 is substrate for α-secretase and γ-secretase. The C terminal fragment of this proteolysis accumulates surrounding Aβ in mouse brain. This shows the pathological role of the Clstn-3 in the AD [69]. Clstn 3 have not been explored as a biomarker in the AD diagnostics but future studies will help in proving it as an emerging biomarker.


    5. NFT Formation Related Molecular Markers


    5.1. Reelin

    Reelin is a glycoprotein with the serine protease activity with the molecular weight of 388kD. The gene for this protein is present on the chromosome 7 [70]. The protein is an extracellular secreted matrix protein. It is an important signaling molecule for its crucial role in the neural development, maintenance and plasticity of neural networks [71]. Any alteration in the function of this protein or its relevant receptors, results in the altered memory and its cognition [71]. Because of its neurological importance the protein is widely studied in many neurological disorders and have found to play important role in schizophrenia, autism and in AD [72,73,74]. One of the signaling pathway of reelin results in the inhibition of the glycogen synthase kinase 3β (GSK-3β) and cyclin dependent kinase 5 (CDK5), both of which have found to play an important role in hyper-phosphorylation of tau. Depletion in the reelin gene expression is noted during early stages of AD murine model and patients without affecting downstream signaling molecules [75]. Another study was conducted on frozen brain tissue sample and change in the concentration of reelin was observed at protein level by western blotting and at m-RNA level by RT-PCR [76]. These molecules involve apolyprotein E receptor 2, very low density lipoprotein receptor and phosphorylated disabled-1 (Dab1). Various studies have reported the neuro-protective effect of reelin against the toxicity caused by the Aβ42. In-vivo and in-vitro studies have showed the potency of reelin to alter the kinetics of Aβ42 aggregation [77]. This effect is mainly due to binding of reelin to soluble Aβ42 fragment and also to the fibrils formed due to aggregation. The same study also reported that overexpression of reelin in mice models sufficed the recovery of functional and behavioral conditions [77]. Reelin when it is bound to the Aβ peptide cannot activate the Dab-1. Dab-1 is an important component of Akt signaling pathway which leads to the inhibition of GSK-3β [78] as shown in figure 2. Unavailability of reelin due to its co-localization with Aβ renders Dab-1 inactive for GSK-3β inhibition [79]. This might be the potential cause for the increase tau hyper-phosphorylation. Though having been studied extensively for its role in Alzheimer’s disease reelin is not yet explored in biomarker aspect. Studies involving change in concentration of reelin in CSF and blood of AD patients need to be carried out. Association of reelin with the Aβ fragment may provide information depleted reelin protein concentration in the CSF or blood in AD patient.


    5.2. Bridging integrator 1 (BIN1)

    BIN 1 also known as amphiphysin 2 or AMPH2. There are around 15 isoforms of BIN1, some of which can be categorized based on their size. It was identified to be one of the Myc interacting protein. Myc gene plays an important role in the progression of cell cycle and tumor development. This lead to the hypothesis of tumor suppressor ability of BIN1 [80]. BIN1 expressions were found to be altered in the AD brain [81]. The large isoform of BIN1 was decreased and smaller isoforms were increased in the AD patient when compared with the age matched control [82]. One of the study revealed that increased BIN1 can lead to AD. BIN1 knockdown have successfully suppressed the neurotoxicity caused by the NFTs as shown in figure 2. The altered DNA methylation pattern was observed at the locus of BIN1 in the AD patients. This DNA methylation has been predicted to have potential in the early onset of AD. The potential use of plasma BIN1 level as a biomarker have been carried out in AD patient [83]. The study revealed that m-RNA and protein expression levels of BIN1 in the plasma are increased significantly compared to control. Study conducted by Glennon et al. 2013 proposed that BIN1 levels are decreased in sporadic AD and not in the familial AD [84]. Changes in the locus of BIN1 are found to be associated with the PD but change in its expression in case of PD has to be evaluated [85]. Hence convincing evidence about BIN1 as a potential biomarker in AD is still to be found.


    5.3. Transactive Response DNA Binding Protein 43 (TDP-43)

    As stated in the name TDP 43 is DNA and also RNA binding protein with the approximate molecular mass 43kDa. TDP 43 as it binds to DNA/RNA can bring about transcription repression of genes and also the gene splicing [86]. TDP 43 in its phosphorylated form is associated with many neurological disorders to name a few frontotemporal lobar degeneration (FTLD), ALS and AD [87]. TDP is a nuclear protein but in these diseases it is deposited in cytoplasm of the neuronal cells [88]. Immunofluorescence assay have revealed the presence of these deposits in the brain region similar to the NFTs of AD. It is also found in the association with the NFTs and α-synuclein as shown in figure 2. TDP-43 is observed within diseased neurons in an ubiquitinated form. This ubiquitinated TDP-43 further undergoes proteolytic cleavage. The fragments of this proteolytic cleavage are abundant in the deposits found in the AD [89]. C-terminal fragment generated due to aberrant cleavage of TDP-43 is more abundant in the aggregation rather than complete TDP-43 peptide [90]. TDP-43 is found to be reduced in other neurological diseases including HD and spinocerebellar ataxia 3 [91]. Study performed by Josephs et al have already revealed its importance as a biomarker and therapeutic target in AD [92]. But whether its change in expression can be observed in CSF and blood plasma has to be elucidated.


    6. Ca2+ Signaling Pathway Related Molecular Markers


    6.1. Visinin like protein 1 (VLP 1)

    VLP 1 is a protein which belongs to the family of neuronal calcium sensing protein. The protein is known to play an important role in intracellular neuronal signaling. This signaling pathway is mainly driven by the change in concentration of Ca2+ ions [93]. The protein is expressed only in the brain tissue and is absent in other peripheral tissue [94]. VLP1 have been associated with the AD. Ca2+ levels are altered in the AD patient brain due to the damage caused by accumulation of Aβ42. AB42 is capable of inducing Ca2+ pores on the neurons which leads to the rapid influx of Ca2+ ions within neurons. Ca2+ may then lead to activation of many mitochondrial as well as endoplasmic reticulum pathway which causes increased oxidative stress [85,86]. One of the hypothesis suggest that the increased concentration of Ca2+ may lead to the death of neurons which express VLPs [97]. VLP-1 has been found to be associated with the hyper-phosphorylation of tau protein which further causes NFTs formation as shown in figure 2. Same study also showed that not only increased expression of VLP-1 is associated with the AD but also the decreased level of calcium buffering protein known as calbinding-D28K is affected [98]. Lee et al. 2008 carried out evaluation of VLP-1 as biomarker in AD. Their result suggest that increased VLP-1 values directly co-relate with the neuronal death as more dead neurons will release more amount of VLP-1. Even though the results of this study sounds positive the sample size of AD patient was less [99]. Also VLP-1 expression was higher in homozygous APOε4 than the homozygous APOε3 and heterozygous APOε3/4 [99]. This increases the complexity in using VLP-1 since genomic aspects interplay with the biomarker level in the blood. Also the VLP-1 is a brain injury biomarker and is implicated in various neurological disorders such as amyotrophic lateral sclerosis (ALS) stroke and schizophrenia [90,91,92]. One of the study demonstrated VLP-1 and a Chitinase-3-like protein 1 levels increased as the time progressed in Memory cognitive impairment (MCI) patients [103]. VLP-1 protein levels has been significantly detected in the CSF and plasma of AD patient and its level are found to be significantly different from the normal patient [104]. This implies the significant role of VLP-1 in the AD and its potential as a biomarker in its diagnosis.


    6.2. Calreticulin (CRT)

    Discussing about Ca2+ signaling pathway, one important endoplasmic reticulum protein which sequester and store Ca2+ in endoplasmic reticulum (ER)is calreticulin. CRT plays an important role in signaling pathways [105]. Because of its important cellular signaling function it is found in various neurological disease which include HD and PD[106]. In ALS mice model the study was performed in order to identify the cause of rapid neuro-degeneration. The outcome of the study stated that CRT is one of the downstream signaling molecule in Fas signaling [107]. Activation of Fas/NO pathway leads to the decreased expression of the CRT followed by rapid neuronal degeneration. Decreased expression of CRT within neurons makes them sensitive to ER related oxidative stress [108]. As discussed earlier excess Ca2+ levels may be the reason for the oxidative stress observed in this condition. One important role of CRT is its ability to bind to misfolded proteins and stop their transportation to golgi complex [109]. Importantly in vitro studies have shown the ability of CRT to bind Aβ42 peptide. The binding was stimulated in the presence of Ca2+ ions. This suggests the dual role and altered signaling of Ca2+ in AD. Ca2+ can increase the binding of Aβ42 to CRT but in its increased concentration it results in the oxidative stress and neuronal death via its signaling pathways as shown in figure 2 [109,110]. CRT is a receptor for various other protein molecules one of which is C1q. C1q has been found to increase the reactive oxygen species (ROS) generation [111]. Evaluation of CRT as a biomarker in AD have mentioned its lowered protein and m-RNA expression in AD patient serum [112]. CRT can bind to the Aβ42 and C1q and its lower expression suggest that these bindings may be altered in the AD patients which result in the amyloid deposit and also ROS mediated neuronal death [113]. Hence it can be stated that CRT is associated with the early development of AD and the inflammatory cascade is also altered in the AD brain.


    6.3. Neurogranin (Ng)

    Neurogranin is a small protein first discovered first identified in rat brain and is also named as RC3, BICKS [114]. It is an important protein regulating the synaptic plasticity and signaling within the neurons. Ng regulates the synaptic signaling by controlling the availability of calmodulin an important signalling molecule in Ca2+-Calmodulin pathway [114]. Ng levels have been up-regulated in the AD brain and they have been explored for the early detection of AD and progression of the disease. The study evaluated concentration of the Ng in CSF of AD patient by immunoassay [115]. Vos et al. (2005)showed the presence of C-terminal Ng is increased in CSF but not in the plasma of AD patient. In human Ng is present as peptide of various lengths. Out of these 7 are distinct for the plasma and are not observed in the CSF [116]. Ng peptide 48 to 76 which was absent in plasma was found to be increased in CSF of AD brain when compared with the normal patient [117]. Ng was used in combination with the YKL40 which is also known as chitinase-3 like protein 1 for the evaluation of there accuracy in AD biomarker discovery. The comparison between YLK40/Ng and established biomarker Aβ42/pTau provided the evidence for more accurate and sensitive detection by Aβ42/pTau than YLK40/Ng [118].


    7. Conclusion & Future Direction

    Alzheimer’s disease is a complex disease involving the alteration of variety of the key components. It is important to identify the early disease specific biomarker so as to better treatment of the disease. Many of the biomarkers have been identify in AD but none of them seems to be efficient in the early diagnostics. The mutations observed within the FAD can be useful for the early diagnosis but the same does not serve the purpose in the LOAD. Hence for LOAD many neuropsychological and brain imaging testing have been adopted. A deep study of AD is required in order to identify the potential biomarkers which will also help in therapeutic targeting of the disease. Overall, Human Kalikrain 6 m-RNA and protein level is found to be altered in the brain tissue as well as in CSF. Hypocretin, Neuroserpins, VLP-1 and Ng showed increase protein concentration in the CSF of the AD patient. Spingosine and reelin showed elevated level of protein in the brain tissue sample of patient suffering from AD, for reelin, even m-RNA level were elevated indicating increased gene expression. Clstn1 protein level was found to be reduced in the brain tissue of patient whereas Clstn3 protein level is increased in AD rat model. Cystacin C protein level was reduced in the CSF and plasma of AD patient. Increased m-RNA and protein level of Bin1 within the plasma is observed by the studies done. TDP43 at protein level is reduced in the AD brain due to its co-localization with the Amyloid. Decreased expression of calreticulin in AD is observed by m-RNA in the serum.

    In conclusion these molecular markers can form the basis for studies involving development of biomarker in AD. Use of Chip-based technology or Micro-array technology will be useful in measuring the gene level expression of these markers in the plasma or CSF, this may even help to distinguish AD from other neurological diseases. Even though many of the markers involved are also markers for other neurological diseases these markers can be used to study therapeutic efficacy of a drug developed against AD.


    Acknowledgements

    Authors wish to thank Management of Jaslok Hospital & Research Centre, Mumbai for providing the facilities to carry out research work in the field of Neurobiology.


    Conflict of Interest

    All authors declare no conflicts of interest in this paper.


    [1] Butterfield D, Castegna A, Lauderback C, et al. (2002) Evidence that amyloid beta-peptide-induced lipid peroxidation and its sequelae in Alzheimer’s disease brain contribute to neuronal death. Neurobiol Aging 23: 655-664. doi:10.1016/S0197-4580(01)00340-2. doi: 10.1016/S0197-4580(01)00340-2
    [2] Ramsden M, Kotilinek L, Forster C, et al. (2005) Age-dependent neurofibrillary tangle formation, neuron loss, and memory impairment in a mouse model of human tauopathy (P301L). J Neurosci 25: 10637-10647. doi:10.1523/JNEUROSCI.3279-05.2005. doi: 10.1523/JNEUROSCI.3279-05.2005
    [3] Serpell LC (2000) Alzheimer’s amyloid fibrils: structure and assembly. Biochim Biophys Acta- Mol Basis Dis 1502: 16-30. doi:10.1016/S0925-4439(00)00029-6. doi: 10.1016/S0925-4439(00)00029-6
    [4] Nunan J, Small DH (2000) Regulation of APP cleavage by α-, β- and γ-secretases. FEBS Lett 483: 6-10. doi:10.1016/S0014-5793(00)02076-7. doi: 10.1016/S0014-5793(00)02076-7
    [5] Chasseigneaux S, Allinquant B (2012) Functions of Aβ, sAPPα and sAPPβ : similarities and differences. J Neurochem 120 Suppl: 99-108. doi:10.1111/j.1471-4159.2011.07584.x.
    [6] Sadik G, Kaji H, Takeda K, et al. (1999) In vitro processing of amyloid precursor protein by cathepsin D. Int J Biochem Cell Biol 31: 1327-1337. http://www.ncbi.nlm.nih.gov/pubmed/10605825. Accessed January 3, 2016. doi: 10.1016/S1357-2725(99)00053-9
    [7] Kume H, Maruyama K, Kametani F (2004) Intracellular domain generation of amyloid precursor protein by epsilon-cleavage depends on C-terminal fragment by alpha-secretase cleavage. Int J Mol Med 13: 121-125. http://www.ncbi.nlm.nih.gov/pubmed/14654982.
    [8] Konietzko U (2012) AICD nuclear signaling and its possible contribution to Alzheimer’s disease. Curr Alzheimer Res 9: 200-216. http://www.ncbi.nlm.nih.gov/pubmed/21605035. doi: 10.2174/156720512799361673
    [9] Brion JP, Couck AM, Passareiro E, et al. (1985) Neurofibrillary tangles of Alzheimer’s disease: an immunohistochemical study. J Submicrosc Cytol 17: 89-96. http://europepmc.org/abstract/med/3973960.
    [10] Ferrer I, Gomez-Isla T, Puig B, et al. (2005) Current advances on different kinases involved in tau phosphorylation, and implications in Alzheimer’s disease and tauopathies. Curr Alzheimer Res 2: 3-18. http://www.ncbi.nlm.nih.gov/pubmed/15977985. doi: 10.2174/1567205052772713
    [11] Williams DR (2006) Tauopathies: classification and clinical update on neurodegenerative diseases associated with microtubule-associated protein tau. Intern Med J 36: 652-660. doi:10.1111/j.1445-5994.2006.01153.x. doi: 10.1111/j.1445-5994.2006.01153.x
    [12] Levy-Lahad E, Wasco W, Poorkaj P, et al. (1995) Candidate gene for the chromosome 1 familial Alzheimer’s disease locus. Science 269: 973-977. doi:10.1126/science.7638622. doi: 10.1126/science.7638622
    [13] Bertram L, Blacker D, Mullin K, et al. (2000) Evidence for genetic linkage of Alzheimer’s disease to chromosome 10q. Science 290: 2302-2303. doi:10.1126/science.290.5500.2302. doi: 10.1126/science.290.5500.2302
    [14] Beecham GW, Martin ER, Li Y-J, et al. (2008) Genome-wide association study implicates a chromosome 12 risk locus for late-onset Alzheimer disease. Am J Hum Genet 84: 35-43. doi:10.1016/j.ajhg.2008.12.008.
    [15] Wilkins CH, Sheline YI, Roe CM, et al. (2006) Vitamin D deficiency is associated with low mood and worse cognitive performance in older adults. Am J Geriatr Psychiatry 14: 1032-1040. doi:10.1097/01.JGP.0000240986.74642.7c. doi: 10.1097/01.JGP.0000240986.74642.7c
    [16] Mullan M, Houlden H, Windelspecht M, et al. (1992) A locus for familial early-onset Alzheimer’s disease on the long arm of chromosome 14, proximal to the alpha 1-antichymotrypsin gene. Nat Genet 2: 340-342. doi:10.1038/ng1292-340. doi: 10.1038/ng1292-340
    [17] St George-Hyslop P, Haines J, Rogaev E, et al. (1992) Genetic evidence for a novel familial Alzheimer’s disease locus on chromosome 14. Nat Genet 2: 330-334. doi:10.1038/ng1292-330. doi: 10.1038/ng1292-330
    [18] Pericak-Vance M a, Bebout JL, Gaskell PC, et al. (1991) Linkage studies in familial Alzheimer disease: evidence for chromosome 19 linkage. Am J Hum Genet 48: 1034-1050. http://www.ncbi.nlm.nih.gov/pmc/articles/PMC1683100/pdf/ajhg00090-0019.pdf.
    [19] Chartier-Harlin MC, Parfitt M, Legrain S, et al. (1994) Apolipoprotein E, epsilon 4 allele as a major risk factor for sporadic early and late-onset forms of Alzheimer’s disease: analysis of the 19q13.2 chromosomal region. Hum Mol Genet 3: 569-574. doi:10.1093/hmg/3.4.569. doi: 10.1093/hmg/3.4.569
    [20] Yu J-T, Tan L, Hardy J (2014) Apolipoprotein E in Alzheimer’s disease: an update. Annu Rev Neurosci 37: 79-100. doi:10.1146/annurev-neuro-071013-014300. doi: 10.1146/annurev-neuro-071013-014300
    [21] Strittmatter WJ, Weisgraber KH, Goedert M, et al. (1994) Hypothesis: microtubule instability and paired helical filament formation in the Alzheimer disease brain are related to apolipoprotein E genotype. Exp Neurol 125: 163-171. doi:S0014488684710193. doi: 10.1006/exnr.1994.1019
    [22] Wu L, Rosa-Neto P, Hsiung G-YR, et al. (2012) Early-onset familial Alzheimer’s disease (EOFAD). Can J Neurol Sci 39: 436-445. doi:W4438L6488727555. doi: 10.1017/S0317167100013949
    [23] Olsson A, Vanderstichele H, Andreasen N, et al. (2005) Simultaneous measurement of β-amyloid (1-42), total Tau, and phosphorylated Tau (Thr181) in cerebrospinal fluid by the xMAP technology. Clin Chem 51: 336-345. doi:10.1373/clinchem.2004.039347. doi: 10.1373/clinchem.2004.039347
    [24] Petraki CD, Karavana VN, Skoufogiannis PT, et al. (2001) The spectrum of human kallikrein 6 (zyme/protease M/neurosin) expression in human tissues as assessed by immunohistochemistry. J Histochem Cytochem 49: 1431-1441. http://www.ncbi.nlm.nih.gov/pubmed/11668196. doi: 10.1177/002215540104901111
    [25] Rockenstein E, Nuber S, Overk CR, et al. (2014) Accumulation of oligomer-prone α-synuclein exacerbates synaptic and neuronal degeneration in vivo. Brain 137: 1496-1513. doi:10.1093/brain/awu057. doi: 10.1093/brain/awu057
    [26] Spencer B, Valera E, Rockenstein E, et al. (2015) A brain-targeted, modified neurosin (kallikrein-6) reduces α-synuclein accumulation in a mouse model of multiple system atrophy. Mol Neurodegener 10: 48. doi:10.1186/s13024-015-0043-6. doi: 10.1186/s13024-015-0043-6
    [27] Ogawa K, Yamada T, Tsujioka Y, et al. (2000) Localization of a novel type trypsin-like serine protease, neurosin, in brain tissues of Alzheimer’s disease and Parkinson's disease. Psychiatry Clin Neurosci 54: 419-426. doi:10.1046/j.1440-1819.2000.00731.x. doi: 10.1046/j.1440-1819.2000.00731.x
    [28] Diamandis EP, Yousef GM, Petraki C, et al. (2000). Human kallikrein 6 as a biomarker of alzheimer’s disease. Clin Biochem 33:663-667. http://www.ncbi.nlm.nih.gov/pubmed/11166014. doi: 10.1016/S0009-9120(00)00185-5
    [29] Saper CB, Scammell TE, Lu J (2005) Hypothalamic regulation of sleep and circadian rhythms. Nature 437: 1257-1263. doi:10.1038/nature04284. doi: 10.1038/nature04284
    [30] de Lecea L, Kilduff TS, Peyron C, et al. (1998) The hypocretins: Hypothalamus-specific peptides with neuroexcitatory activity. Proc Natl Acad Sci 95: 322-327. doi:10.1073/pnas.95.1.322. doi: 10.1073/pnas.95.1.322
    [31] Liguori C, Romigi A, Nuccetelli M, et al. (2014) Orexinergic System Dysregulation, Sleep Impairment, and Cognitive Decline in Alzheimer Disease. JAMA Neurol 71: 1498-1505. doi:10.1001/jamaneurol.2014.2510. doi: 10.1001/jamaneurol.2014.2510
    [32] Baumann CR, Hersberger M, Bassetti CL (2006) Hypocretin-1 (orexin A) levels are normal in Huntington’s disease. J Neurol 253: 1232-1233. doi:10.1007/s00415-006-0146-7. doi: 10.1007/s00415-006-0146-7
    [33] Dauvilliers YA, Lehmann S, Jaussent I, et al. (2014). Hypocretin and brain β-amyloid peptide interactions in cognitive disorders and narcolepsy. Front Aging Neurosci 6. doi:10.3389/fnagi.2014.00119.
    [34] Friedman LF, Zeitzer JM, Lin L, et al. et al. (2007) In Alzheimer disease, increased wake fragmentation found in those with lower hypocretin-1. Neurology 68: 793-794. doi:10.1212/01.wnl.0000256731.57544.f9. doi: 10.1212/01.wnl.0000256731.57544.f9
    [35] Slats D, Claassen JAHR, Lammers GJ, et al. (2012) Association between hypocretin-1 and amyloid-β42 cerebrospinal fluid levels in Alzheimer’s disease and healthy controls. Curr Alzheimer Res 9: 1119-1125. http://www.ncbi.nlm.nih.gov/pubmed/22742854. doi: 10.2174/156720512804142840
    [36] Fronczek R, van Geest S, Frölich M, et al. (2012) Hypocretin (orexin) loss in Alzheimer’s disease. Neurobiol Aging 33: 1642-1650. doi:10.1016/j.neurobiolaging.2011.03.014. doi: 10.1016/j.neurobiolaging.2011.03.014
    [37] Gallone S, Boschi S, Rubino E, et al. et al. (2014) Is HCRTR2 a genetic risk factor for Alzheimer’s disease? Dement Geriatr Cogn Disord 38: 245-253. doi:10.1159/000359964. doi: 10.1159/000359964
    [38] Tucker HM, Kihiko M, Caldwell JN, et al. (2000) The plasmin system is induced by and degrades amyloid-beta aggregates. J Neurosci 20: 3937-3946. doi:20/11/3937.
    [39] Man H-Y, Ma X-M (2012) A role for neuroserpin in neuron morphological development. J Neurochem 121: 495-496. doi:10.1111/j.1471-4159.2012.07655.x. doi: 10.1111/j.1471-4159.2012.07655.x
    [40] Ledesma MD, Da Silva JS, Crassaerts K, et al. (2000) Brain plasmin enhances APP alpha-cleavage and Abeta degradation and is reduced in Alzheimer’s disease brains. EMBO Rep 1: 530-535. doi:10.1093/embo-reports/kvd107. doi: 10.1093/embo-reports/kvd107
    [41] Hanzel CE, Iulita MF, Eyjolfsdottir H, et al. (2014) Analysis of matrix metallo-proteases and the plasminogen system in mild cognitive impairment and Alzheimer’s disease cerebrospinal fluid. J Alzheimers Dis 40: 667-678. doi:10.3233/JAD-132282.
    [42] Kinghorn KJ, Crowther DC, Sharp LK, et al. (2006) Neuroserpin binds Abeta and is a neuroprotective component of amyloid plaques in Alzheimer disease. J Biol Chem 281: 29268-29277. doi:10.1074/jbc.M600690200. doi: 10.1074/jbc.M600690200
    [43] Fabbro S, Schaller K, Seeds NW (2011) Amyloid-beta levels are significantly reduced and spatial memory defects are rescued in a novel neuroserpin-deficient Alzheimer’s disease transgenic mouse model. J Neurochem 118: 928-938. doi:10.1111/j.1471-4159.2011.07359.x. doi: 10.1111/j.1471-4159.2011.07359.x
    [44] Subhadra B, Schaller K, Seeds NW (2013) Neuroserpin up-regulation in the Alzheimer’s disease brain is associated with elevated thyroid hormone receptor-β1 and HuD expression. Neurochem Int 63: 476-481. doi:10.1016/j.neuint.2013.08.010. doi: 10.1016/j.neuint.2013.08.010
    [45] Okada T, Kajimoto T, Jahangeer S, et al. (2009) Sphingosine kinase/sphingosine 1-phosphate signalling in central nervous system. Cell Signal 21: 7-13. doi:10.1016/j.cellsig.2008.07.011. doi: 10.1016/j.cellsig.2008.07.011
    [46] Cieślik M, Czapski GA, Strosznajder JB (2015) The Molecular Mechanism of Amyloid β42 Peptide Toxicity: The Role of Sphingosine Kinase-1 and Mitochondrial Sirtuins. PLoS One 10: e0137193. doi:10.1371/journal.pone.0137193. doi: 10.1371/journal.pone.0137193
    [47] Carro E, Trejo JL, Gomez-Isla T, et al. (2002) Serum insulin-like growth factor I regulates brain amyloid-beta levels. Nat Med 8: 1390-1397. doi:10.1038/nm793. doi: 10.1038/nm1202-793
    [48] Mizugishi K, Yamashita T, Olivera A, et al. (2005) Essential role for sphingosine kinases in neural and vascular development. Mol Cell Biol 2511113-11121. doi:10.1128/MCB.25.24.11113-11121.2005.
    [49] Couttas T a, Kain N, Daniels B, et al. (2014) Loss of the neuroprotective factor Sphingosine 1-phosphate early in Alzheimer’s disease pathogenesis. Acta Neuropathol Commun 2: 9. doi:10.1186/2051-5960-2-9. doi: 10.1186/2051-5960-2-9
    [50] Sivasubramanian M, Kanagaraj N, Dheen ST, et al. (2015) Sphingosine kinase 2 and sphingosine-1-phosphate promotes mitochondrial function in dopaminergic neurons of mouse model of Parkinson’s disease and in MPP+-treated MN9D cells in vitro. Neuroscience 290: 636-648. doi:10.1016/j.neuroscience.2015.01.032. doi: 10.1016/j.neuroscience.2015.01.032
    [51] Takasugi N, Sasaki T, Suzuki K, et al. (2011) BACE1 activity is modulated by cell-associated sphingosine-1-phosphate. J Neurosci 31: 6850-6857. doi:10.1523/JNEUROSCI.6467-10. doi: 10.1523/JNEUROSCI.6467-10.2011
    [52] Hagen N, Hans M, Hartmann D, et al. (2011) Sphingosine-1-phosphate links glycosphingolipid metabolism to neurodegeneration via a calpain-mediated mechanism. Cell Death Differ 18: 1356-1365. doi:10.1038/cdd.2011.7. doi: 10.1038/cdd.2011.7
    [53] Huh CG, Håkansson K, Nathanson CM, et al. (1999) Decreased metastatic spread in mice homozygous for a null allele of the cystatin C protease inhibitor gene. Mol Pathol 52: 332-340. http://www.pubmedcentral.nih.gov/articlerender.fcgi?artid=395718&tool=pmcentrez&rendertype=abstract. doi: 10.1136/mp.52.6.332
    [54] Wilson ME, Boumaza I, Lacomis D, et al. (2010) Cystatin C: a candidate biomarker for amyotrophic lateral sclerosis. PLoS One 5: e15133. doi:10.1371/journal.pone.0015133. doi: 10.1371/journal.pone.0015133
    [55] Simonsen AH, McGuire J, Podust VN, et al. (2007) A novel panel of cerebrospinal fluid biomarkers for the differential diagnosis of Alzheimer’s disease versus normal aging and frontotemporal dementia. Dement Geriatr Cogn Disord 24: 434-440. doi:10.1159/000110576. doi: 10.1159/000110576
    [56] Chen J, Liu C (2014) Association Of Serum Cystatin C Levels On The Progression And Cognition In Parkinson’s Disease (P4.045). Neurology 82: 45.
    [57] Chuo L-J, Sheu WHH, Pai M-C, et al. (2007) Genotype and plasma concentration of cystatin C in patients with late-onset Alzheimer disease. Dement Geriatr Cogn Disord 23: 251-257. doi:10.1159/000100021. doi: 10.1159/000100021
    [58] Crawford FC, Freeman MJ, Schinka JA, et al. (2000) A polymorphism in the cystatin C gene is a novel risk factor for late-onset Alzheimer’s disease. Neurology 55: 763-768. http://www.ncbi.nlm.nih.gov/pubmed/10993992. doi: 10.1212/WNL.55.6.763
    [59] Ghidoni R, Paterlini A, Albertini V, et al. (2011) Cystatin C is released in association with exosomes: a new tool of neuronal communication which is unbalanced in Alzheimer’s disease. Neurobiol Aging 32: 1435-1442. doi:10.1016/j.neurobiolaging.2009.08.013. doi: 10.1016/j.neurobiolaging.2009.08.013
    [60] Afonso S, Romagnano L, Babiarz B (2016) The expression and function of cystatin C and cathepsin B and cathepsin L during mouse embryo implantation and placentation. Development 124: 3415-3425. http://www.ncbi.nlm.nih.gov/pubmed/9310336.
    [61] Sastre M, Calero M, Pawlik M, et al. (2004) Binding of cystatin C to Alzheimer’s amyloid beta inhibits in vitro amyloid fibril formation. Neurobiol Aging 25: 1033-1043. doi:10.1016/j.neurobiolaging.2003.11.006. doi: 10.1016/j.neurobiolaging.2003.11.006
    [62] Maruyama K, Ikeda S, Ishihara T, et al. (1990) Immunohistochemical characterization of cerebrovascular amyloid in 46 autopsied cases using antibodies to beta protein and cystatin C. Stroke 21: 397-403. doi:10.1161/01.STR.21.3.397. doi: 10.1161/01.STR.21.3.397
    [63] Deng A, Irizarry MC, Nitsch RM, et al. (2001). Elevation of cystatin C in susceptible neurons in Alzheimer’s disease. Am J Pathol 159: 1061-1068. doi:10.1016/S0002-9440(10)61781-6. doi: 10.1016/S0002-9440(10)61781-6
    [64] Ponomareva OY, Holmen IC, Sperry AJ, et al. (2014) Calsyntenin-1 regulates axon branching and endosomal trafficking during sensory neuron development in vivo. J Neurosci 34: 9235-9248. doi:10.1523/JNEUROSCI.0561-14.2014. doi: 10.1523/JNEUROSCI.0561-14.2014
    [65] Vagnoni A, Perkinton MS, Gray EH, et al. (2012) Calsyntenin-1 mediates axonal transport of the amyloid precursor protein and regulates Aβ production. Hum Mol Genet 21: 2845-2854. doi:10.1093/hmg/dds109. doi: 10.1093/hmg/dds109
    [66] Yin GN, Lee HW, Cho J-Y, et al. (2009) Neuronal pentraxin receptor in cerebrospinal fluid as a potential biomarker for neurodegenerative diseases. Brain Res 1265: 158-170. doi:10.1016/j.brainres.2009.01.058. doi: 10.1016/j.brainres.2009.01.058
    [67] Ludwig A, Blume J, Diep T-M, et al. (2009) Calsyntenins mediate TGN exit of APP in a kinesin-1-dependent manner. Traffic 10: 572-589. doi:10.1111/j.1600-0854.2009.00886.x. doi: 10.1111/j.1600-0854.2009.00886.x
    [68] Pettem KL, Yokomaku D, Luo L, et al. (2013) The specific α-neurexin interactor calsyntenin-3 promotes excitatory and inhibitory synapse development. Neuron 80: 113-128. doi:10.1016/j.neuron.2013.07.016. doi: 10.1016/j.neuron.2013.07.016
    [69] Uchida Y, Gomi F, Murayama S, et al. (2013) Calsyntenin-3 C-terminal fragment accumulates in dystrophic neurites surrounding aβ plaques in tg2576 mouse and Alzheimer disease brains: its neurotoxic role in mediating dystrophic neurite formation. Am J Pathol 182: 1718-1726. doi:10.1016/j.ajpath.2013.01.014. doi: 10.1016/j.ajpath.2013.01.014
    [70] DeSilva U, D’Arcangelo G, Braden VV, et al. (1997) The human reelin gene: Isolation, sequencing, and mapping on chromosome 7. Genome Res 7: 157-164. doi:10.1101/gr.7.2.157. doi: 10.1101/gr.7.2.157
    [71] Stranahan AM, Erion JR, Wosiski-Kuhn M (2013) Reelin signaling in development, maintenance, and plasticity of neural networks. Ageing Res Rev 12: 815-822. doi:10.1016/j.arr.2013.01.005. doi: 10.1016/j.arr.2013.01.005
    [72] Eastwood SL, Harrison PJ (2003) Interstitial white matter neurons express less reelin and are abnormally distributed in schizophrenia: towards an integration of molecular and morphologic aspects of the neurodevelopmental hypothesis. Mol Psychiatry 8: 821-831. doi:10.1038/sj.mp.4001399. doi: 10.1038/sj.mp.4001371
    [73] Fatemi SH, Stary JM, Egan EA (2002) Reduced blood levels of reelin as a vulnerability factor in pathophysiology of autistic disorder. Cell Mol Neurobiol 22: 139-152. doi:http://dx.doi.org/10.1023/A:1019857620251. doi: 10.1023/A:1019857620251
    [74] Botella-López A, Burgaya F, Gavín R, et al. (2006) Reelin expression and glycosylation patterns are altered in Alzheimer’s disease. Proc Natl Acad Sci U S A 103: 5573-5578. doi:10.1073/pnas.0601279103. doi: 10.1073/pnas.0601279103
    [75] Herring A, Donath A, Steiner KM, et al. (2012) Reelin depletion is an early phenomenon of alzheimer’s pathology. J Alzheimer’s Dis 30: 963-979. doi:10.3233/JAD-2012-112069.
    [76] Chin J, Massaro CM, Palop JJ, et al. (2007) Reelin Depletion in the Entorhinal Cortex of Human Amyloid Precursor Protein Transgenic Mice and Humans with Alzheimer’s Disease. J Neurosci 27.
    [77] Pujadas L, Rossi D, Andrés R, et al. (2014) Reelin delays amyloid-beta fibril formation and rescues cognitive deficits in a model of Alzheimer’s disease. Nat Commun 5: 3443. doi:10.1038/ncomms4443.
    [78] Cuchillo-Ibáñez I, Balmaceda V, Botella-López A, et al. (2013) Beta-Amyloid Impairs Reelin Signaling. PLoS One 8: 1-10. doi:10.1371/journal.pone.0072297.
    [79] Patrick GN, Zukerberg L, Nikolic M, et al. (1999) Conversion of p35 to p25 deregulates Cdk5 activity and promotes neurodegeneration. Nature 402: 615-622. doi:10.1038/45159. doi: 10.1038/45159
    [80] Sakamuro D, Elliott KJ, Wechsler-Reya R, et al. (1996) BIN1 is a novel MYC-interacting protein with features of a tumour suppressor. Nat Genet 14: 69-77. doi:10.1038/ng0996-69. doi: 10.1038/ng0996-69
    [81] Chapuis J, Hansmannel F, Gistelinck M, et al. (2013) Increased expression of BIN1 mediates Alzheimer genetic risk by modulating tau pathology. Mol Psychiatry 18: 1225-1234. doi:10.1038/mp.2013.1. doi: 10.1038/mp.2013.1
    [82] Holler CJ, Davis PR, Beckett TL, et al. (2014) Bridging integrator 1 (BIN1) protein expression increases in the Alzheimer’s disease brain and correlates with neurofibrillary tangle pathology. J Alzheimers Dis 42: 1221-1227. doi:10.3233/JAD-132450.
    [83] Sun L, Tan M-S, Hu N, et al. (2013) Exploring the value of plasma BIN1 as a potential biomarker for alzheimer’s disease. J Alzheimers Dis 37: 291-295. doi:10.3233/JAD-130392.
    [84] Glennon EBC, Whitehouse IJ, Miners JS, et al. (2013) BIN1 Is Decreased in Sporadic but Not Familial Alzheimer’s Disease or in Aging. PLoS One 8. doi:10.1371/journal.pone.0078806.
    [85] Gan-Or Z, Amshalom I, Bar-Shira A, et al. (2015) The Alzheimer disease BIN1 locus as a modifier of GBA-associated Parkinson disease. J Neurol 262: 2443-2447. doi:10.1007/s00415-015-7868-3. doi: 10.1007/s00415-015-7868-3
    [86] Ou SH, Wu F, Harrich D, et al. (1995) Cloning and characterization of a novel cellular protein, TDP-43, that binds to human immunodeficiency virus type 1 TAR DNA sequence motifs. J Virol 69: 3584-3596. http://www.pubmedcentral.nih.gov/articlerender.fcgi?artid=189073&tool=pmcentrez&rendertype=abstract.
    [87] Arai T, Mackenzie IRA, Hasegawa M, et al. (2009) Phosphorylated TDP-43 in Alzheimer’s disease and dementia with Lewy bodies. Acta Neuropathol 117: 125-136. doi:10.1007/s00401-008-0480-1. doi: 10.1007/s00401-008-0480-1
    [88] Foulds P, McAuley E, Gibbons L, et al. (2008) TDP-43 protein in plasma may index TDP-43 brain pathology in Alzheimer’s disease and frontotemporal lobar degeneration. Acta Neuropathol 116: 141-146. doi:10.1007/s00401-008-0389-8. doi: 10.1007/s00401-008-0389-8
    [89] Higashi S, Iseki E, Yamamoto R, et al. (2007) Concurrence of TDP-43, tau and alpha-synuclein pathology in brains of Alzheimer’s disease and dementia with Lewy bodies. Brain Res 1184: 284-294. doi:10.1016/j.brainres.2007.09.048. doi: 10.1016/j.brainres.2007.09.048
    [90] Zhang Y-J, Xu Y-F, Cook C, et al. (2009) Aberrant cleavage of TDP-43 enhances aggregation and cellular toxicity. Proc Natl Acad Sci U S A 106: 7607-7612. doi:10.1073/pnas.0900688106. doi: 10.1073/pnas.0900688106
    [91] Tauffenberger A, Chitramuthu BP, Bateman A, et al. (2013) Reduction of polyglutamine toxicity by TDP-43, FUS and progranulin in Huntington’s disease models. Hum Mol Genet 22: 782-794. doi:10.1093/hmg/dds485. doi: 10.1093/hmg/dds485
    [92] Josephs KA, Whitwell JL, Weigand SD, et al. (2014) TDP-43 is a key player in the clinical features associated with Alzheimer’s disease. Acta Neuropathol 127: 811-824. doi:10.1007/s00401-014-1269-z. doi: 10.1007/s00401-014-1269-z
    [93] Spilker C, Braunewell K-H (2003) Calcium-myristoyl switch, subcellular localization, and calcium-dependent translocation of the neuronal calcium sensor protein VILIP-3, and comparison with VILIP-1 in hippocampal neurons. Mol Cell Neurosci 24: 766-778. http://www.ncbi.nlm.nih.gov/pubmed/14664824. Accessed February 15, 2016. doi: 10.1016/S1044-7431(03)00242-2
    [94] Laterza OF, Modur VR, Crimmins DL, et al. (2006) Identification of novel brain biomarkers. Clin Chem 52: 1713-1721. doi:10.1373/clinchem.2006.070912. doi: 10.1373/clinchem.2006.070912
    [95] Braunewell KH (2012) The visinin-like proteins VILIP-1 and VILIP-3 in Alzheimer’s disease-old wine in new bottles. Front Mol Neurosci 5: 20. doi:10.3389/fnmol.2012.00020.
    [96] Chakroborty S, Stutzmann GE (2011) Early calcium dysregulation in Alzheimer’s disease: Setting the stage for synaptic dysfunction. Sci China Life Sci 54: 752-762. doi:10.1007/s11427-011-4205-7. doi: 10.1007/s11427-011-4205-7
    [97] Bezprozvanny I, Mattson MP (2008) Neuronal calcium mishandling and the pathogenesis of Alzheimer’s disease. Trends Neurosci 31: 454-463. doi:10.1016/j.tins.2008.06.005. doi: 10.1016/j.tins.2008.06.005
    [98] Schnurra I, Bernstein HG, Riederer P, et al. (2001) The neuronal calcium sensor protein VILIP-1 is associated with amyloid plaques and extracellular tangles in Alzheimer’s disease and promotes cell death and tau phosphorylation in vitro: a link between calcium sensors and Alzheimer's disease? Neurobiol Dis 8: 900-909. doi:10.1006/nbdi.2001.0432. doi: 10.1006/nbdi.2001.0432
    [99] Lee JM, Blennow K, Andreasen N, et al. (2008) The brain injury biomarker VLP-1 is increased in the cerebrospinal fluid of Alzheimer disease patients. Clin Chem 54: 1617-1623. doi:10.1373/clinchem.2008.104497. doi: 10.1373/clinchem.2008.104497
    [100] Liebl MP, Kaya AM, Tenzer S, et al. (2014) Dimerization of visinin-like protein 1 is regulated by oxidative stress and calcium and is a pathological hallmark of amyotrophic lateral sclerosis. Free Radic Biol Med 72: 41-54. doi:10.1016/j.freeradbiomed.2014.04.008. doi: 10.1016/j.freeradbiomed.2014.04.008
    [101] Stejskal D, Sporova L, Svestak M, et al. (2011) Determination of serum visinin like protein-1 and its potential for the diagnosis of brain injury due to the stroke: a pilot study. Biomed Pap Med Fac Univ Palacký Olomouc Czechoslov 155: 263-268. doi:10.5507/bp.2011.049. doi: 10.5507/bp.2011.049
    [102] Bernstein H-G, Braunewell K-H, Spilker C, et al. (2002) Hippocampal expression of the calcium sensor protein visinin-like protein-1 in schizophrenia. Neuroreport 13: 393-396. http://www.ncbi.nlm.nih.gov/pubmed/11930147. doi: 10.1097/00001756-200203250-00006
    [103] Kester MI, Teunissen CE, Sutphen C, et al. Cerebrospinal fluid VILIP-1 and YKL-40, candidate biomarkers to diagnose, predict and monitor Alzheimer’s disease in a memory clinic cohort. Alzheimers Res Ther 7: 59. doi:10.1186/s13195-015-0142-1.
    [104] Tarawneh R, D’Angelo G, Macy E, et al. (2011) Visinin-like protein-1: diagnostic and prognostic biomarker in Alzheimer disease. Ann Neurol 70: 274-285. doi:10.1002/ana.22448. doi: 10.1002/ana.22448
    [105] Lin Q, Cao Y, Gao J (2014) Serum calreticulin is a negative biomarker in patients with Alzheimer’s disease. Int J Mol Sci 15: 21740-21753. doi:10.3390/ijms151221740. doi: 10.3390/ijms151221740
    [106] Wu J-C, Liang Z-Q, Qin Z-H (2006) Quality control system of the endoplasmic reticulum and related diseases. Acta Biochim Biophys Sin 38: 219-226. doi:10.1111/j.1745-7270.2006.00156.x.
    [107] Bernard-Marissal N, Moumen a., Sunyach C, et al. (2012) Reduced Calreticulin Levels Link Endoplasmic Reticulum Stress and Fas-Triggered Cell Death in Motoneurons Vulnerable to ALS. J Neurosci 32: 4901-4912. doi:10.1523/JNEUROSCI.5431-11. doi: 10.1523/JNEUROSCI.5431-11.2012
    [108] Gelebart P, Opas M, Michalak M (2004) Calreticulin, a Ca2+-binding chaperone of the endoplasmic reticulum. Int J Biochem Cell Biol 37: 260-266. doi:10.1016/j.biocel.2004.02.030.
    [109] Peterson JR, Ora A, Van PN, et al. (1995) Transient, lectin-like association of calreticulin with folding intermediates of cellular and viral glycoproteins. Mol Biol Cell 6: 1173-1184. doi:10.1091/mbc.6.9.1173. doi: 10.1091/mbc.6.9.1173
    [110] Duus K, Hansen PR, Houen G (2008) Interaction of calreticulin with amyloid beta peptide 1–42. Protein Pept Lett 15: 103-107. http://www.ncbi.nlm.nih.gov/pubmed/18221019.. doi: 10.2174/092986608783330459
    [111] Erickson RR, Dunning LM, Olson DA, et al. (2005) In cerebrospinal fluid ER chaperones ERp57 and calreticulin bind beta-amyloid. Biochem Biophys Res Commun 332: 50-57. doi:10.1016/j.bbrc.2005.04.090. doi: 10.1016/j.bbrc.2005.04.090
    [112] Taguchi J, Fujii A, Fujino Y, et al. (2000) Different expression of calreticulin and immunoglobulin binding protein in Alzheimer’s disease brain. Acta Neuropathol 100: 153-160. doi:10.1007/s004019900165. doi: 10.1007/s004019900165
    [113] Luo X, Weber GA, Zheng J, et al. (2003) C1q-calreticulin induced oxidative neurotoxicity: Relevance for the neuropathogenesis of Alzheimer’s disease. J Neuroimmunol 135: 62-71. doi:10.1016/S0165-5728(02)00444-7. doi: 10.1016/S0165-5728(02)00444-7
    [114] Díez-Guerra FJ (2010) Neurogranin, a link between calcium/calmodulin and protein kinase C signaling in synaptic plasticity. IUBMB Life 62: 597-606. doi:10.1002/iub.357. doi: 10.1002/iub.357
    [115] Kester MI, Teunissen CE, Crimmins DL, et al. (2015) Neurogranin as a Cerebrospinal Fluid Biomarker for Synaptic Loss in Symptomatic Alzheimer Disease. JAMA Neurol 72: 1275-1280. doi:10.1001/jamaneurol.2015.1867. doi: 10.1001/jamaneurol.2015.1867
    [116] Fyfe I (2015) Alzheimer disease: neurogranin in the CSF signals early Alzheimer disease and predicts disease progression. Nat Rev Neurol 11: 609. doi:10.1038/nrneurol.2015.178. doi: 10.1038/nrneurol.2015.178
    [117] Portelius E, Zetterberg H, Skillbäck T, et al. (2015) Cerebrospinal fluid neurogranin: relation to cognition and neurodegeneration in Alzheimer’s disease. Brain 138: 3373-3385. doi:10.1093/brain/awv267. doi: 10.1093/brain/awv267
    [118] De Vos A, Jacobs D, Struyfs H, et al. (2015) C-terminal neurogranin is increased in cerebrospinal fluid but unchanged in plasma in Alzheimer’s disease. Alzheimers Dement 11: 1461-1469. doi:10.1016/j.jalz.2015.05.012. doi: 10.1016/j.jalz.2015.05.012
  • Reader Comments
  • © 2016 the Author(s), licensee AIMS Press. This is an open access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/4.0)
通讯作者: 陈斌, bchen63@163.com
  • 1. 

    沈阳化工大学材料科学与工程学院 沈阳 110142

  1. 本站搜索
  2. 百度学术搜索
  3. 万方数据库搜索
  4. CNKI搜索

Metrics

Article views(7492) PDF downloads(1224) Cited by(0)

Article outline

Figures and Tables

Figures(2)

Other Articles By Authors

/

DownLoad:  Full-Size Img  PowerPoint
Return
Return

Catalog