Dersleri yüzünden oldukça stresli bir ruh haline sikiş hikayeleri bürünüp özel matematik dersinden önce rahatlayabilmek için amatör pornolar kendisini yatak odasına kapatan genç adam telefonundan porno resimleri açtığı porno filmini keyifle seyir ederek yatağını mobil porno okşar ruh dinlendirici olduğunu iddia ettikleri özel sex resim bir masaj salonunda çalışan genç masör hem sağlık hem de huzur sikiş için gelip masaj yaptıracak olan kadını gördüğünde porn nutku tutulur tüm gün boyu seksi lezbiyenleri sikiş dikizleyerek onları en savunmasız anlarında fotoğraflayan azılı erkek lavaboya geçerek fotoğraflara bakıp koca yarağını keyifle okşamaya başlar

GET THE APP

Proteomic Biomarkers in Alzheimer's disease
Make the best use of Scientific Research and information from our 700+ peer reviewed, Open Access Journals that operates with the help of 50,000+ Editorial Board Members and esteemed reviewers and 1000+ Scientific associations in Medical, Clinical, Pharmaceutical, Engineering, Technology and Management Fields.
  • Review Article   
  • Diagn Pathol Open 2016, Vol 1(1): 103
  • DOI: 10.4172/2476-2024.1000103

Proteomic Biomarkers in Alzheimer's disease

Ramón Cacabelos1,2*
1Chair of Genomic Medicine, Camilo José Cela University, Madrid, Spain
2Institute of Medical Science and Genomic Medicine, EuroEspes Biomedical Research Center, Corunna, Spain
*Corresponding Author: Ramón Cacabelos, EuroEspes Biomedical Research Center, Institute of Medical Science and Genomic Medicine, Camilo José Cela University, 15165-Bergondo, Corunna, Spain, Tel: +34981780505, Fax: +34981780511, Email: rcacabelos@euroespes.com

Received: 02-Dec-2015 / Accepted Date: 24-Dec-2015 / Published Date: 02-Jan-2016 DOI: 10.4172/2476-2024.1000103

Alzheimer’s Disease: Proteomic Biomarkers

Alzheimer’s disease (AD) is a major problem of health in developed countries. From a pathogenic perspective, this prevalent form of dementia is a polygenic/complex disorder in which over 600 defective genes distributed across the human genome may be involved, in conjunction with epigenetic aberrations and diverse environmental risk factors [1]. The abnormal expression of genes and subsequent post-transcriptional modifications may lead to changes in proteins, either conformational, mutant forms or quantitative variants. Pathognomonic hallmarks are represented by the accumulation of extracellular β-amyloid protein (Aβ) in senile plaques and intracellular hyperphosphorylated tau protein in neurofibrillary tangles, which have been used as potential biomarkers for AD in brain tissue and body fluids (plasma, cerebrospinal fluid) [2-4]. These and other still undefined neuropathological hallmarks may be expressed in brain tissue and body fluids 2-3 decades prior to the onset of the disease. Consequently, the identification of predictive biomarkers is of paramount importance for the implementation of preventive programs in the population at risk [1,5,6]. Furthermore, at present, the clinical diagnosis of AD is not an easy task, and there is a great interest in developing specific, sensitive, and practical tools to differentially diagnose and discriminate the different types of dementia [7].

During the past decade, a great effort has been made to identify proteomic biomarkers for AD with still poor results. Over 1,000 proteins have been identified to be differentially expressed in proteomic analyses of AD cases; however, interpretation of results is difficult, and a direct connection between specific proteomic profiles and AD pathogenesis is still undefined [8]. Similarly, several epigenetic signatures (DNA methylation, histone modifications, miRNA dysregulation) have been reported as diagnostic aids with still unclear consequences [9,10]. Therefore, AD biomarkers are urgently needed for both early and accurate diagnosis and prediction of disease progression [11], as well as for monitoring treatment [1,12].

The abnormal expression of proteins in AD brains is a frequent finding. Some of these proteins might represent novel markers for AD neuropathology; however, in most studies, similar anomalies in protein expression are currently seen in different neurodegenerative disorders [13,14]. In AD, Aβ interacts with a variety of Aβ-associated proteins, some of which can form complexes with Aβ and influence its clearance, aggregation or toxicity. The secreted Wnt pathway protein Dickkopf-related protein 3 (Dkk-3) is a potential Aβ-associated protein. Dkk-3 co-localizes with Aβ in the brain, is expressed in neurons and in blood vessel walls, is secreted by leptomeningeal smooth muscle cells in vitro, and is abundantly present in both cerebrospinal fluid and serum, but its levels are similar in nondemented controls and patients with AD, Lewy body dementia, and frontotemporal dementia [15]. In a study of the human brain-insoluble proteome in AD by mass spectrometry, 4,216 proteins have been identified, among which 36 proteins accumulate in the disease, including U1-70K and other U1 small nuclear ribonucleoprotein (U1 snRNP) spliceosome components. Similar accumulations in mild cognitive impairment cases indicate that spliceosome changes occur in early stages of AD. A dysregulated RNA processing with accumulation of unspliced RNA species, including myc box-dependent-interacting protein 1, clusterin, and presenilin-1, occurs in AD. U1-70K knockdown or antisense oligonucleotide inhibition of U1 snRNP increases the protein level of amyloid precursor protein (APP), indicating U1 snRNP pathology and abnormal RNA splicing in AD pathogenesis [16]. Brain tissue from diabetic patients with cerebrovascular dementia or AD contains significant deposits of oligomerized amylin [17]. Visinin-like protein 1, RUFY3 protein, and Copine 6 are abnormally expressed in the olfactory bulb of autopsy cases with AD, Lewy body disease, frontotemporal lobar degeneration, and mixed dementia; and only dipeptidyl aminopeptidase-like protein 6 shows a specific down-regulation in AD [18].

Changes in phosphorylation levels were found in 19 proteins involved in energy metabolism, neuronal plasticity, signal transduction, and oxidative stress response in the parietal cortex of AD patients at different stages of dementia [19]. Histone post-translational modifications have been found in the frontal cortex of AD patients, affecting methylation of H2B K108, methylation of H4 R55, and ubiquitination of H2B K120 [20]. Novel techniques have been developed for proteomic bioanalysis of microdissected cells; 400 proteins have been identified in microdissected neurons, 50% of which were associated with AD [21].

Materials And Methods

The expression levels of DCC-interacting protein 13-beta, serum albumin, creatine kinase B-type, heat shock 70 kDa protein 1A, Tcomplex protein 1 subunit beta, adenylate kinase isoenzyme 1, pyruvate dehydrogenase E1 component subunit beta mitochondrial, and V-type proton ATPase catalytic subunit A have been found to be 1.5 times higher in transgenic AβPPswe/PS1dE9 mice which express a chimeric mouse/human amyloid-β protein precursor (Mo/ HuAβPP695swe) and mutant human presenilin 1 (PS1-dE9) associated with early-onset AD; in contrast, the expression levels of dihydropyrimidinase-related protein 2, actin cytoplasmic 2, isoform 1 of V-type proton ATPase catalytic subunit, tubulin alpha-1C chain, Factin- capping protein subunit alpha-2, ubiquitin carboxyl-terminal hydrolase isozyme L1, and actin cytoplasmic 1 were lower in the transgenic model. These proteins are involved in cytoskeletal structure, energy metabolism, synaptic components, and protein degradation [22]. Using a redox-proteomic approach, 12 proteins involved in energy metabolism, protein folding, cell structure, signal transduction and excitotoxicity were found to be significantly altered in the levels of protein carbonyls in the hippocampus of 3xTg-AD mice before the appearance of Aβ plaques and neurofibrillary tangles. Alpha-enolase and glutamine synthetase were identified as the common targets of oxidation in the brains of 3 × Tg-AD mice, mild cognitive impairment (MCI) sufferers and AD patients. The oxidation of t-complex protein 1 subunit epsilon and protein disulfide-isomerase A3 was reported to be associated with AD [23].

Abnormal processing of tau protein is central to the etiology of tauopathies (AD, frontotemporal dementia, progressive supranuclear palsy, post-traumatic dementia) [13]. Tau protein associates with the ribonucleoproteome, including major protein complexes involved in RNA processing and translation, and binds to several heat shock proteins, the proteasome- and microtubule- associated proteins. Expression of P301L mutant tau disrupts interactions of the Cterminal half of tau with heat shock proteins and the proteasome. The higher propensity of P301L mutant tau to aggregate may reflect a perturbation of its chaperone-assisted stabilization and proteasomedependent degradation [24]. MAP2c prevents arachidonic acidinduced in vitro aggregation of tau. MAP2c possesses chaperone-like activity while tau does not. Phosphorylation impairs the chaperone activity of MAP2c, implying a crucial role of chaperone in preventing tau fibrillation. MAP2c/MAP2 might be one of the regulators maintaining tau homeostasis in the cell [25]. The tau N-terminal domain may play a direct role in the regulation of microtubule stabilization, since the Gln124-tau fragment displays a strong ability to bind and stabilize microtubules [26].

Novel markers associated with major AD neuropathological hallmarks have been reported, including CSF oxidative stress signatures [27,28], heat-shock proteins [29], markers of the ubiquitinproteasome system (UPS) [30], and neuroimmune biomarkers [31].

The measurement of plasma Aβ levels has been used by researchers for years, with contradictory results. This failure might be attributed to the heterogeneity of the disease, deficient patient recruitment, nonspecificity of peripheral Aβ levels as an AD biomarker, and the nonrecognition that Aβ levels are genotype-dependent [32,33]. For the past 5 years some new biomarkers have been postulated as potential diagnostic and/or prognostic candidates for AD. Among peripheral candidate proteins, most AD-related proteins reported in the literature were not specific and were found to be affected by other brain disorders [34,35]. Several proteins associated with brain atrophy and cognitive decline have been identified in plasma, including the following: A1AT, alpha 1 antitrypsin; A1M, α-1-microglobulin; ApoA1, apolipoprotein A1; ApoA2, apolipoprotein A-2; ApoB, apolipoprotein B; ApoC3, apolipoprotein C3; ApoC4, apolipoprotein C4; ApoE, apolipoprotein E; ApoH, apolipoprotein-H; Aβ40, amyloid beta 1-40; BDNF, brain-derived neurotrophic factor; C3, complement C3; C3a, complement C3a; C4a, complement component C4a; C8, complement C8; CF1, complement factor-I; CgA, chromogranin A; Chk2, serine/threonine-protein kinase Chk2; FGG, γ-fibrinogen; GCSF, granulocyte-colonystimulating factor; ICAM, intercellular adhesion molecule; IFN-γ, interferon-gamma; IGFBP2, insulin-like growth factor binding protein 2; IL-10, interleukin-10; IL-13, interleukin-13; IL-1ra, interleukin 1 receptor antagonist; IL-2, interleukin-2; IL-4, interleukin-4; IL-6, interleukin-6; MIP1α, macrophage inhibitory protein 1α; NAP2, nucleosome assembly protein 2; NCAM, neural cell adhesion molecule; NSE, neuron-specific enolase; PDGF, platelet-derived growth factor; PPY, pancreatic polypeptide; PSA-ACT, prostate-specific antigen complexed to α1- antichymotrypsin; RANTES, regulated on activation normal T cell expressed and secreted; SAP, serum amyloid-P; sRAGE, soluble receptor for advanced glycation end products; TNF-α, tumor necrosis factor alpha; and TTR, transthyretin [35].

A novel serum proteomic approach to interrogate the lowmolecular weight proteome for serum AD found 59 novel potential AD biomarkers, 4 of which showed diagnostic replicability. These biomarkers were characterized as 602.3 (amino acid sequence L/ IAENR), 804.55 (a phosphatidylcholine with two fatty-acid side chains), 874.6 (a peroxidated phosphatidylcholine), and biomarker 804.53 (an oxidized glycerophosphatidylcholine (C42H78NO11P+H+) with 3 primary possible structures: 1-palmitoyl-2-(14,15-di-hydroxy9- keto-10, 12-octadecadienoyl) glycerophosphatidyl choline; 1- palmitoyl-2-(13,14-di-hydroxy-10-keto-8, 11-octadecadienoyl) glycerophosphatidyl choline; or 1-palmitoyl-2-(13-hydroperoxy-8- keto-9, 11octadecadienoyl)glycerophosphatidylcholine) [36].

Panel-based proteomics on plasma samples from Twins-UK subjects revealed that genetic factors explain ~26% of the variability in blood protein levels on average. The plasma level of the mitogen-activated protein kinase (MAPK) MAPKAPK5 protein was found to positively associate with the 10-year change, and MAP2K4 was found to associate negatively with the volume of the left entorhinal cortex [37].

Multiple markers were identified to be differentially expressed in the cerebrospinal fluid (CSF) of AD patients as compared with control subjects. The currently available cerebrospinal fluid (CSF) biomarkers for AD (Aβ, total Tau (t-Tau), phosphorylated Tau (p-Tau)) have a high sensitivity and specificity for AD, but there is still no test to effectively predict the development of AD in a pre-symptomatic stage [2,3,38,39]; however, measurement of Aβ1-42 and tau in CSF is a valuable marker in AD research, where low levels of Aβ and high levels of tau indicate AD [2,3,35]. In addition, CSF and plasma levels of Aβ are closely associated with APOE genotypes, with APOE-4/4 carriers exhibiting the lowest Aβ levels [1,12,32,33].

Neuronal secretory protein VGF and neuronal pentraxin receptor-1 (NPTXR) are decreased in AD (at baseline, 21% and 17%, respectively), with a decreased rate/year of 10.9% and 6.9%, respectively [40]. Thirty-eight proteins show abnormal expression in Lewy body dementia CSF by proteomic profiling; 4 proteins (ectonucleotide pyrophosphatase/phosphodiesterase 2, lysosomeassociated membrane protein 1, pro-orexin, transthyretin) are specific for the identification of AD from LBD; 9 proteins (4 novel: malate dehydrogenase 1, serum amyloid A4, GM2-activator protein, and prosaposin) are elevated in both AD and LBD versus controls [41]. Patients with mild cognitive impairment (MCI) who are converted into AD cases have an abnormal CSF glycosylation profile. CSF glycosylation changes may occur before the onset of the disease. Glycosyltransferase GnT-III might be involved in AD inducing specific sugar modifications in the BACE-1 glycoprotein [42].

From the vast majority of the studies reported so far, some conclusions can be drawn:

(i) Less than 30% of AD cases can be accurately diagnosed with genomic and/or proteomic biomarkers; (ii) a major problem with most proteomic analyses is the coexistence of similar findings in different forms of neurodegenerative disorders; (iii) most proteomic analyses with positive results as potential AD biomarkers need further validation and replicability; (iv) although the most accepted proteomic biomarker for AD is the analysis of CSF Aβ and tau, with decreased levels of Aβ and increased levels of tau reflecting AD, its clinical utility in elderly subjects is limited by the invasive nature of lumbar puncture; (v) attention should be paid to the standardization of measurement of AD biomarkers due to large inter-laboratory variation; (vi) CSF and/or plasma Aβ are APOE-dependent; (vii) from a practical perspective, blood-based biomarkers for AD represent a very attractive option; however, the complex composition of blood may pose technical challenges for an efficient identification of reliable biomarkers; (viii) the identification of a universal biomarker for AD is highly unlikely; and (ix) since protein expression depends on the genomic background of each patient, proteomic signatures should be correlated with the genomic profiles of the patients.

References

  1. Cacabelos R, Cacabelos P, Torrellas C, Tellado I, Carril JC (2014) Pharmacogenomics of Alzheimer's disease: novel therapeutic strategies for drug development. Methods MolBiol 1175: 323-556.
  2. Blennow K, Hampel H, Weiner M, Zetterberg H (2010) Cerebrospinal fluid and plasma biomarkers in Alzheimer disease. Nat Rev Neurol 6: 131-144.
  3. Blennow K, Zetterberg H (2015) The past and the future of Alzheimer's disease CSF biomarkers-a journey toward validated biochemical tests covering the whole spectrum of molecular events. Front Neurosci 9: 345.
  4. Viola KL, Klein WL (2015) Amyloid β oligomers in Alzheimer's disease pathogenesis, treatment, and diagnosis. ActaNeuropathol 129: 183-206.
  5. Lista S, O'Bryant SE, Blennow K, Dubois B (2015) Biomarkers in Sporadic and Familial Alzheimer's Disease. J Alzheimers Dis 47: 291-317.
  6. Nayak A, Salt G, Verma SK, Kishore U (2015) Proteomics Approach to Identify Biomarkers in Neurodegenerative Diseases. Int Rev Neurobiol 121: 59-86.
  7. Harbater O, Gannot I (2014) Fluorescent probes concentration estimation in vitro and ex vivo as a model for early detection of Alzheimer's disease. J Biomed Opt 19: 127007.
  8. Cacabelos R, Torrellas C, Carril JC, Aliev G, Teijido O (2015) Epigenomics and proteomics of brain disorders. Curr Genomics (in press).
  9. Cacabelos R, Torrellas C (2015) Epigenetics of Aging and Alzheimer's Disease: Implications for Pharmacogenomics and Drug Response. Int J MolSci 16: 30483-30543.
  10. Ren RJ, Zhang YF, Dammer EB, Zhou Y, Wang LL, et al. (2015) Peripheral Blood MicroRNA Expression Profiles in Alzheimer's Disease: Screening, Validation, Association with Clinical Phenotype and Implications for Molecular Mechanism. MolNeurobiol (Epub ahead of print).
  11. Moya-Alvarado G, Gershoni-Emek N, Perlson E, Bronfman FC (2015) Neurodegeneration and Alzheimer's disease. What can proteomics tell us about the Alzheimer's brain? Mol Cell Proteomics .
  12. Cacabelos R, Torrellas C, Carrera I (2015) Opportunities in Pharmacogenomics for the treatment of Alzheimer’s Disease. Future Neurology 10: 229-252.
  13. Kovacs GG, Ferrer I, Grinberg LT, Alafuzoff I (2015) Aging-related tau astrogliopathy (ARTAG): harmonized evaluation strategy. Acta Neuropathol.
  14. Shi Q, Chen LN, Zhang BY, Xiao K, Zhou W, et al. (2015) Proteomics analyses for the global proteins in the brain tissues of different human prion diseases. Mol Cell Proteomics 14: 854-69.
  15. Bruggink KA, Kuiperij HB, Gloerich J, Otte-Höller I, Rozemuller AJ, et al. (2015) Dickkopf-related protein 3 is a potential Aβ-associated protein in Alzheimer's Disease. J Neurochem 134: 1152-1162.
  16. Bai B, Hales CM, Chen PC, Gozal Y, Dammer EB, et al. (2013) U small nuclear ribonucleoprotein complex and RNA splicing alterations in Alzheimer's disease. ProcNatlAcadSci U S A 110: 16562-16567.
  17. Ly H, Despa F (2015) Hyperamylinemia as a risk factor for accelerated cognitive decline in diabetes. Expert Rev Proteomics 12: 575-577.
  18. Zelaya MV, Pérez-Valderrama E, de Morentin XM, Tuñon T, Ferrer I, et al. (2015) Olfactory bulb proteome dynamics during the progression of sporadic Alzheimer's disease: identification of common and distinct olfactory targets across Alzheimer-related co-pathologies. Oncotarget 6: 39437-39456.
  19. Triplett JC, Swomley AM, Cai J, Klein JB, Butterfield DA, et al. (2015) Quantitative Phosphoproteomic Analyses of the Inferior Parietal Lobule from Three Different Pathological Stages of Alzheimer's Disease. J Alzheimers Dis 49: 45-62.
  20. Anderson KW, Turko IV (2015) Histone post-translational modifications in frontal cortex from human donors with Alzheimer's disease. Clin Proteomics 12: 26.
  21. Drummond ES, Nayak S, Ueberheide B, Wisniewski T (2015) Proteomic analysis of neurons microdissected from formalin-fixed, paraffin-embedded Alzheimer's disease brain tissue. Sci Rep 5: 15456.
  22. Fu Y, Zhao D, Pan B, Wang J, Cui Y, et al. (2015) Proteomic Analysis of Protein Expression Throughout Disease Progression in a Mouse Model of Alzheimer's Disease. J Alzheimers Dis 47: 915-926.
  23. Shen L, Chen C, Yang A, Chen Y, Liu Q2, et al. (2015) Redox proteomics identification of specifically carbonylated proteins in the hippocampi of triple transgenic Alzheimer's disease mice at its earliest pathological stage. J Proteomics 123: 101-113.
  24. Gunawardana CG, Mehrabian M, Wang X, Mueller I, Lubambo IB, et al. (2015) The Human Tau Interactome: Binding to the Ribonucleoproteome, and Impaired Binding of the Proline-to-Leucine Mutant at Position 301 (P301L) to Chaperones and the Proteasome. Mol Cell Proteomics 14: 3000-3014.
  25. Mitra G, Gupta S, Poddar A, Bhattacharyya B (2015) MAPc prevents arachidonic acid-induced fibril formation of tau: Role of chaperone activity and phosphorylation. BiophysChem 205: 16-23.
  26. Derisbourg M, Leghay C, Chiappetta G, Fernandez-Gomez FJ, Laurent C, et al. (2015) Role of the Tau N-terminal region in microtubule stabilization revealed by new endogenous truncated forms. Sci Rep 5: 9659.
  27. Di Domenico F, Pupo G, Giraldo E, Badìa MC, Monllor P, et al. (2015) Oxidative signature of cerebrospinal fluid from mild cognitive impairment and Alzheimer disease patients. Free RadicBiol Med 91: 1-9.
  28. Swomley AM, Butterfield DA (2015) Oxidative stress in Alzheimer disease and mild cognitive impairment: evidence from human data provided by redox proteomics. Arch Toxicol 89: 1669-1680.
  29. Dattilo S, Mancuso C, Koverech G, Di Mauro P, Ontario ML, et al. (2015) Heat shock proteins and hormesis in the diagnosis and treatment of neurodegenerative diseases. Immun Ageing 12: 20.
  30. Gentier RJ, Verheijen BM, Zamboni M, Stroeken MM, Hermes DJ, et al. (2015) Localization of mutant ubiquitin in the brain of a transgenic mouse line with proteasomal inhibition and its validation at specific sites in Alzheimer's disease. Front Neuroanat 9: 26.
  31. Cacabelos R, Torrellas C, Fernández-Novoa L, Aliev G (2015) Neuroimmune Crosstalk in CNS Disorders: The Histamine Connection. Curr Pharm Des .
  32. Cacabelos R (2003) The application of functional genomics to Alzheimer's disease. Pharmacogenomics 4: 597-621.
  33. Cacabelos R, Lombardi V, Fernández-Novoa L, Kubota Y, Corzo L, et al. (2004) A functional genomics approach to the analysis of biological markers in Alzheimer disease. In: Molecular Neurobiology of Alzheimer’s Disease and Related Disorders. Takeda M, Tanaka T, Cacabelos R. (Eds.). Karger, Basel, 236-285.
  34. Chiam JT, Dobson RJ, Kiddle SJ, Sattlecker M (2015) Are blood-based protein biomarkers for Alzheimer's disease also involved in other brain disorders? A systematic review. J Alzheimers Dis 43: 303-314.
  35. Baird AL, Westwood S, Lovestone S (2015) Blood-Based Proteomic Biomarkers of Alzheimer's Disease Pathology. Front Neurol 6: 236.
  36. Shah DJ, Rohlfing F, Anand S, Johnson WE, Alvarez MT, et al. (2015) Discovery and Subsequent Confirmation of Novel Serum Biomarkers Diagnosing Alzheimer's Disease. J Alzheimers Dis 49: 317-327.
  37. Kiddle SJ, Steves CJ, Mehta M, Simmons A, Xu X, et al. (2015) Plasma protein biomarkers of Alzheimer's disease endophenotypes in asymptomatic older twins: early cognitive decline and regional brain volumes. Transl Psychiatry 5: e584.
  38. Ahmed RM, Paterson RW, Warren JD, Zetterberg H, O'Brien JT, et al. (2014) Biomarkers in dementia: clinical utility and new directions. J NeurolNeurosurg Psychiatry 85: 1426-1434.
  39. Galozzi S, Marcus K, Barkovits K (2015) Amyloid-β as a biomarker for Alzheimer's disease: quantification methods in body fluids. Expert Rev Proteomics 12: 343-354.
  40. Hendrickson RC, Lee AY, Song Q, Liaw A, Wiener M, et al. (2015) High Resolution Discovery Proteomics Reveals Candidate Disease Progression Markers of Alzheimer's Disease in Human Cerebrospinal Fluid. PLoS One 10: e0135365.
  41. Heywood WE, Galimberti D, Bliss E, Sirka E (2015) Identification of novel CSF biomarkers for neurodegeneration and their validation by a high-throughput multiplexed targeted proteomic assay. MolNeurodegener 10: 64.
  42. Palmigiano A, Barone R, Sturiale L, Sanfilippo C, Bua RO, et al. (2016) CSF N-glycoproteomics for early diagnosis in Alzheimer's disease. J Proteomics 131: 29-37.

Citation: Cacabelos R (2016) Proteomic Biomarkers in Alzheimer’s disease. Diagn Pathol Open 1: 103. Doi: 10.4172/2476-2024.1000103

Copyright: © 2015 Cacabelos R. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

Review summary

  1. May Henry
    Posted on Aug 24 2016 at 2:24 pm
    The short communication provides a brief overview of diagnostic biomarkers for Alzheimer's Disease. The author discussed the biomarkers involved in Alzheimer’s disease. The author said that Aβ1-42 in CSF is a valuable marker in AD and also discussed the limitations its use as a diagnostic and prognostic parameter.

Review summary

  1. May Henry
    Posted on Aug 24 2016 at 2:24 pm
    The short communication provides a brief overview of diagnostic biomarkers for Alzheimer's Disease. The author discussed the biomarkers involved in Alzheimer’s disease. The author said that Aβ1-42 in CSF is a valuable marker in AD and also discussed the limitations its use as a diagnostic and prognostic parameter.

Post your comment

captcha   Reload  Can't read the image? click here to refresh
Peer Reviewed Journals

Make the best use of Scientific Research and information from our 700 + peer reviewed, Open Access Journals

Top