253

Journal of Alzheimer’s Disease 39 (2014) 253–259 DOI 10.3233/JAD-130932 IOS Press

MicroRNAs in Plasma and Cerebrospinal Fluid as Potential Markers for Alzheimer’s Disease Takehiro Kikoa , Kiyotaka Nakagawaa,∗ , Tsuyoshi Tsudukib , Katsutoshi Furukawac , Hiroyuki Araic and Teruo Miyazawaa a Food

& Biodynamic Chemistry Laboratory, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan b Laboratory of Food and Biomolecular Science, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan c Department of Geriatrics and Gerontology, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan

Accepted 6 September 2013

Abstract. The development of Alzheimer’s disease (AD) biomarkers remains an unmet challenge, and new approaches that can improve current AD biomarker strategies are needed. Recent reports suggested that microRNA (miRNA) profiling of biological fluids has emerged as a diagnostic tool for several pathologic conditions. In this study, we measured six candidate miRNAs (miR-9, miR-29a, miR-29b, miR-34a, miR-125b, and miR-146a) in plasma and cerebrospinal fluid (CSF) of AD and normal subjects by using quantitative reverse transcriptase-polymerase chain reaction (qRT-PCR) to evaluate their potential usability as AD biomarkers. The qRT-PCR results showed that plasma miR-34a and miR-146a levels, and CSF miR-34a, miR-125b, and miR-146a levels in AD patients were significantly lower than in control subjects. On the other hand, CSF miR-29a and miR-29b levels were significantly higher than in control subjects. Our results provide a possibility that miRNAs detected in plasma and CSF can serve as biomarkers for AD. Keywords: Alzheimer’s disease, cerebrospinal fluid, miRNA, plasma

INTRODUCTION Alzheimer’s disease (AD) is the most common form of dementia and a chronic neurodegenerative disease of the brain [1, 2]. For detecting and monitoring AD, in addition to minimally invasive tests, there are strategies for developing AD biomarkers by using metabolomics and proteomics techniques. However, identification of such biomarkers remains an unmet challenge [3–6], ∗ Correspondence

to: Kiyotaka Nakagawa, PhD, Food & Biodynamic Chemistry Laboratory, Graduate School of Agricultural Science, Tohoku University, 1-1 Tsutsumidori-Amamiyamachi, Aobaku, Sendai 981-8555, Japan. Tel.: +81 22 717 8904; Fax: +81 22 717 8905; E-mail: [email protected].

and new approaches that can improve current AD biomarker strategies are needed. An ideal biomarker should be specific, predictive, and easy to measure in an accessible fluid (e.g., blood and cerebrospinal fluid (CSF)). MicroRNAs (miRNAs) are a family of short noncoding RNAs whose final product is a 22-nucleotide functional RNA molecule that regulates the expression of target genes by binding to complementary regions of transcripts to repress their translation or cause mRNA degradation [7]. Aberrant expression of miRNAs has been implicated in diseases including cancer, viral hepatitis, and heart disease [8–11]. In addition to these diseases, since some miRNAs (e.g., miR-9, miR-29a,

ISSN 1387-2877/14/$27.50 © 2014 – IOS Press and the authors. All rights reserved

254

T. Kiko et al. / miRNAs Determination in Plasma and CSF of AD Patients

miR-29b, miR-34a, miR-125b, and miR-146a) have been reported to be expressed in mammalian brain [12], their importance in central nervous system (CNS) function and disease is now being recognized [13–15]. For example, miR-29a and miR-29b may facilitate increases in both amyloid-␤ protein precursor (A␤PP) and ␤-site A␤PP-cleaving enzyme (BACE1) in AD brain [16]. Other studies reported that oxidative stress and DNA damage, generated in part by elevations in amyloid-␤ peptide (A␤), iron, and reactive oxygen species, would induce expression of miR-34a and miR146a [17, 18]. Based on these studies and together with recent reports about the occurrence of miRNAs in body fluids [19–23], the above candidate miRNAs in plasma and CSF of AD and normal subjects were analyzed by using quantitative reverse transcriptase-polymerase chain reaction (qRT-PCR) in order to evaluate their potential usability as AD biomarkers. To the best of our knowledge, there are only two reports of miRNA analysis in body fluids (CSF and white blood cells) of AD patients [24, 25], and the utility of plasma and CSF miRNAs as AD biomarkers for developing diagnostic tools and/or for monitoring pathology remains unclear. MATERIALS AND METHODS Ethics statement AD patient volunteers seen at the Tohoku University Hospital and healthy volunteer control subjects participated in this study (Table 1). Diagnosis of AD was set according to the NINCDS-ADRDA criteria [26]. Magnetic resonance imaging was taken in all the subjects. All the AD cases showed significant cerebral atrophy and no cerebrovascular lesions. Disease stage was rated by the Mini-Mental State Examination (MMSE), which is a brief cognitive test used widely in clinical practice and epidemiologic studies. This test was administered in order to grade the subjects’ global cognitive impairment. The study protocol was in accordance with the Declaration of Helsinki and was approved by the Ethical Committee of the Graduate School of Medicine at Tohoku University. All subjects gave written informed consent to the experimental protocol. Plasma and CSF samples from AD and healthy subjects Blood was collected into a tube containing EDTA2Na as an anticoagulant and was subjected to

Table 1 Physical characteristics of AD patients and control subjects1 Total number of subjects Males Females Age (years) MMSE CSF-A␤ (1-42) (pg/ml) CSF-tau (pg/ml) 1 Means ± SD;

AD patients

Control subjects

10 3 7 80.7 ± 5.8 21.1 ± 3.5 378.4 ± 112.9 480.7 ± 218.9

10 4 6 73.0 ± 5.2 29.5 ± 0.7 560.4 ± 90.5 209.8 ± 64.2

n = 10.

centrifugation at 1,000 g for 10 min at 4◦ C. CSF samples were centrifuged at 500 g for 10 min at room temperature. Plasma and CSF were stored at −80◦ C until further analyzed. RNA isolation from plasma and CSF Total RNA, including miRNAs, was isolated from plasma or CSF with the miRNeasy kit (Qiagen). In brief, 2800 ␮L of QIAzol reagent was added to 800 ␮L of plasma or CSF samples. The sample was mixed with 560 ␮L of chloroform in a tube. After mixing vigorously for 15 s, the sample was centrifuged at 12,000 g for 15 min. The upper aqueous phase was carefully transferred to a new collection tube, and 1.5 volume of ethanol was added. The sample was then applied directly to a silica membrane containing column and the RNA was bound and cleaned by using buffers provided by the manufacturer to remove impurities. The immobilized RNA was collected from the membrane with a low salt elution buffer. The RNA concentration was determined by a NanoDrop ND-1000 Spectrophotometers (Thermo Fisher Scientific Inc.). RNA quality was evaluated by an Agilent 2100 Bioanalyzer (Agilent Technologies) using an Agilent Small RNA kit. Real-time quantitative RT-PCR analysis Using High-Capacity cDNA Reverse Transcription Kits (Applied Biosystems), RNA was reverse transcribed to cDNA in the presence of 50 units of MultiScribe Reverse Transcriptase in a total volume of 15 ␮L with the following conditions: 30 min at 16◦ C, 30 min at 42◦ C, and 5 min at 85◦ C. For qRT-PCR, TaqMan MicroRNA Assays (Applied Biosystems), including undiluted cDNA, 20×TaqMan MicroRNA Assay mix, TaqMan 2 × Universal PCR Master Mix, and nuclease-free water, were used. Each PCR reaction was performed in triplicate with MicroAmp™ optical 384-well reaction plates using a 7900HT Fast

T. Kiko et al. / miRNAs Determination in Plasma and CSF of AD Patients

255

Fig. 1. miRNA levels in plasma of healthy human volunteers and AD patients. Values are mean ± SD (n = 10). Difference is considered significant from control subjects (p < 0.05).

Real-Time PCR System (Applied Biosystems), with reactions incubated at 95◦ C for 10 min, followed by 40 cycles of 95◦ C for 15 s and 60◦ C for 1 min. Fluorescence readings were taken during the 60◦ C step. The relative amounts of miRNAs were calculated by using the comparative cycle threshold (Ct) method. Human U6 and miR-24 served as an internal control and were used to normalize the plasma and CSF samples, respectively. CSF tau analysis CSF total tau was measured in duplicate using a double-sandwich enzyme-linked immunosorbent assay method (Innogenetics, Gent, Belgium) according to the manufacturer’s instructions. CSF Aβ analysis CSF A␤ [1-42] was measured in duplicate using a double-sandwich enzyme-linked immunosorbent assay method (WAKO, Osaka, Japan) according to the manufacturer’s instructions.

RESULTS Plasma miRNAs in AD patients and healthy subjects The 6 miRNAs (miR-9, miR-29a, miR-29b, miR34a, miR-125b, and miR-146a) could be detected in plasma by qRT-PCR (Fig. 1). Plasma miR-34a and miR-146a levels in AD patients were significantly lower than in control subjects. The same tendency, although not significant, was observed for plasma miR-125b. No significant differences in plasma miR9, miR-29a, and miR-29b were found between AD patients and control subjects. CSF miRNA in AD patients and healthy subjects Similar to plasma, the six miRNAs could be detected in CSF by qRT-PCR (Fig. 2), and CSF miR-34a, miR125b, and miR-146a in AD patients were significantly lower than in control subjects. On the other hand, CSF miR-29a and miR-29b were significantly higher than in control subjects. No significant difference in CSF miR9 was found between AD patients and control subjects.

Statistical analyses Data are presented as mean ± SD. Differences in miRNA concentrations between control and AD subjects were compared using Student’s t-test or Welch’s t-test for equal or unequal variances; the MannWhitney U test was used when the distribution was skewed. A difference was considered significant at p < 0.05 or p < 0.01.

Relationship between miRNA and other parameters The levels of A␤ and tau in the CSF of AD cases were lower and higher, respectively, than the mean of controls (Table 1). However, probably, the number of volunteers was small, and therefore, no significant correlation between the miRNAs levels in plasma or CSF

256

T. Kiko et al. / miRNAs Determination in Plasma and CSF of AD Patients

Fig. 2. miRNA levels in CSF of healthy human volunteers and AD patients. Values are mean ± SD (n = 10). Difference is considered significant from control subjects (p < 0.05).

and other parameters (such as age, MMSE, CSF A␤, or CSF tau) of AD patients and control subjects was found (Supplementary Figs. 1–8). DISCUSSION miRNAs are recognized as key molecules in intracellular regulatory networks for gene expression, and the spectra and levels of some miRNAs are emerging as biomarkers for various pathological conditions [27]. Recent findings suggest that circulating miRNAs in blood plasma may be useful as biomarkers for the diagnosis of cancers [19, 21, 28, 29] and tissue injuries [30, 31]. In the present study, we selected miR-9, miR-29a, miR-29b, miR-34a, miR-125b, and miR-146a, because these 6 miRNAs were expressed in human brain and there were many reports about association of these miRNAs to AD mechanisms [32–34]. We measured the candidate 6 miRNAs (miR-9, miR-29a, miR-29b, miR-34a, miR-125b, and miR-146a) in plasma as well as CSF by qRT-PCR assays, and showed changes in circulating miRNA levels as a result of AD (Figs. 1, 2). It has been reported that in brains of AD patients, miR-29a and miR-29b are decreased, which leads to increased brain BACE1 expression [16]. BACE1 is the rate-limiting enzyme responsible for A␤ production in the brain, and increased BACE1 expression is thought to be a risk for sporadic AD [35–37]. Additionally, the miR-29 family is reported to regulate DNA methyltransferase 3A and 3B [38]. Abnormal DNA methylation is usually found in AD brains and this can cause cell death [39]. Thus, decreasing miR-29a and miR-29b in brain would lead to increased A␤ genera-

tion and DNA methylation, thereby becoming a burden for aged neurons [40]. In contrast to AD brain, in this study, we found that miR-29a and miR-29b were increased in CSF of AD patients (Fig. 2). Therefore, we might expect that miR-29a and miR-29b found in CSF originated from the brain, and these miRNAs were secreted from the AD brain to CSF. On the other hand, plasma miR-29a and miR-29b levels were unchanged (Fig. 1). Thus, it is hypothesized that miR-29a and miR-29b may be actively released from brain to CSF, but not to blood plasma. We need to clarify the mechanism of how these miR-29a and miR-29b levels were changed in brain and CSF, but not in plasma. miR-34a levels were reportedly increased in AD brain [41]. It has been shown that miR-34a overexpression can induce apoptosis in SH-SY5Y cells [42] utilizing the mechanism of interaction of miR-34a with the 3 -UTR of bcl-2 [42]. Bcl-2 is an antiapoptotic protein that prevents caspase-9 activation through an interaction with Apaf-1. Various studies have shown that bcl-2 overexpression is neuroprotective against apoptotic cell death or reduces the pathological changes in AD by inhibiting the activation of caspases [43, 44]. These effects of bcl-2 expression could be diminished by ectopic expression of miR-34a or knockdown of miR-34a [42]. In the present study, miR-34a was found to be decreased in both CSF and plasma of AD patients (Figs. 1, 2). Therefore, it was thought that miR-34a could not be secreted from AD brain into CSF and plasma. It is tempting to speculate that this may be one reason for AD development. However these are our hypotheses and further study is needed.

T. Kiko et al. / miRNAs Determination in Plasma and CSF of AD Patients

Transcription of miR-146a is induced by pathophysiological stress factors, such as the pro-inflammatory cytokine IL-1␤ [45]. Relatively high levels of miR146a in brains of AD animal models as well as AD superior temporal lobe neocortex and hippocampus underscore the potential importance of miR-146a in neurodegenerative disease [45]. Upregulation of miR146a contributes to mediate innate immune response, inflammatory and synaptic pathobiology, and the manipulation of physiological stressors and environmental factors [45], which drive the AD process. In this study, miR-146a was found to be decreased in plasma and CSF of AD patients (Figs. 1, 2). Therefore, like miR-34a, miR-146a might accumulate in AD brain and not be released from brain to CSF and plasma. This may be one of the reasons for AD; however, research is needed to further understand the roles that these miRNAs play in AD. miR-9 and miR-125b are brain-enriched miRNAs [46, 47]. miR-9 has been associated with neurogenesis, morphogenesis, developmental patterning, brain cell proliferation, and glioblastoma [48, 49]. miR-9 and miR-125b increases in abundance in AD neocortex, suggesting that these brain-enriched miRNAs play some regulatory role in the degenerative process [47]. These miRNAs abundances are significantly changed during development and aging and exhibit a specifically altered expression profile in different regions of AD brain [50, 51]. In this study, no significant differences in plasma and CSF miR-9 were found between AD patients and control subjects (Figs. 1, 2). On the other hand, CSF miR-125b in AD patients was significantly lower than in control subjects, and the same tendency was observed for plasma miR-125b (Figs. 1, 2). As mentioned above, since both miR-9 and miR-125b are enriched in brain, the present findings may imply that these miRNAs are released from the brain in different ways. Further research is needed to evaluate the release of these miRNAs into the body fluids. Extracellular miRNAs circulating in the peripheral blood are included in cell membrane-derived particles, such as apoptotic bodies, microvesicles, and exosomes [52]. There is evidence that packaging of miRNAs into these particles results in protection from endogenous ribonucleases in plasma/serum [19]. For miRNAs in CSF, they showed a remarkable stability against exposure to exogenous RNase [53]. However, the role of extracellular miRNAs remains largely unknown, and additional studies exploring a potential biological function of miRNAs circulating in body fluids such as peripheral blood and CSF are required.

257

Sample size of our study was rather small, and this may cause restriction to develop AD biomarkers. However, degree of data variation was satisfactory, and no outlying observations were found (Figs. 1 and 2), suggesting reliability of our data. This means that our present results could provide the rationale for future investigations of miRNAs in the plasma and CSF for diagnostic and prognostic purposes in a variety of CNS disease, including AD. To further clarify the usability for the biomarker of miRNAs, we need to show the results from large sample size. Such study will be presented in the near future as a different investigation. In conclusion, our results provide a possibility that miRNA detected in the CSF and plasma can serve as markers for AD. This finding is a preliminary indication of the abundant possibilities of using miRNAs to detect specific pathological conditions. In the present study of AD group, since we did not include other neurodegenerative disorders such as dementia with Lewy bodies and frontotemporal lobar degeneration, we consider that these miRNAs would be changed as a result of AD. For the possibility whether these miRNA change or not in other neurodegenerative diseases, this needs to examine in future experiments. There are no significant correlation between miRNAs and MMSE, CSF A␤, or CSF tau in both AD and control subjects. The reason may relate to plasma and CSF miRNA properties (incorporation into recipient cells followed by degradation, or urinary excretion), however these were still unclear and further study is needed. A more comprehensive study in humans is needed to determine the specificity and sensitivity of selected miRNA species. By means of qRT-PCR as well as miRNA array technologies, plasma and CSF-based miRNA markers that are specific for particular CNS disorders may be discovered in the future. Although the number of patients and controls studied here is small, our study provides the rationale for future investigations of miRNAs in the plasma and CSF for diagnostic and prognostic purposes in a variety of CNS disease, including AD. DISCLOSURE STATEMENT Authors’ disclosures available online (http://www.jalz.com/disclosures/view.php?id=1944). SUPPLEMENTARY MATERIAL Supplementary tables are available in the electronic version of this article: http://dx.doi.org/10.3233/JAD130932.

258

T. Kiko et al. / miRNAs Determination in Plasma and CSF of AD Patients

REFERENCES [1]

[2] [3]

[4]

[5]

[6]

[7] [8] [9]

[10]

[11]

[12]

[13] [14] [15]

[16]

de la Torre JC (2004) Is Alzheimer’s disease a neurodegenerative or a vascular disorder? Data, dogma, and dialectics. Lancet Neurol 3, 184-190. Younkin SG (1995) Evidence that A␤42 is the real culprit in Alzheimer’s disease. Ann Neurol 37, 287-288. Oreˇsiˇc M, Hy¨otyl¨ainen T, Herukka SK, Sysi-Aho M, Mattila I, Sepp¨anan-Laakso T, Julkunen V, Gopalacharyulu PV, Hallikainen M, Koikkalainen J, Kivipelto M, Helisalmi S, L¨otj¨onen J, Soininen H (2011) Metabolome in progression to Alzheimer’s disease. Transl Psychiatry 1, e57. Han X, Rozen S, Boyle SH, Hellegers C, Cheng H, Burke JR, Welsh-Bohmer KA, Doraiswamy PM, Kaddurah-Daouk R (2011) Metabolomics in early Alzheimer’s disease: Identification of altered plasma sphingolipidome using shotgun lipidomics. PLoS One 6, e21643. Thambisetty M, Tripaldi R, Riddoch-Contreras J, Hye A, An Y, Campbell J, Sojkova J, Kinsey A, Lynham S, Zhou Y, Ferrucci L, Wong DF, Lovestone S, Resnick SM (2010) Proteome-based plasma markers of brain amyloid-␤ deposition in non-demented older individuals. J Alzheimers Dis 22, 1099-1109. Hye A, Lynham S, Thambisetty M, Causevic M, Campbell J, Byers HL, Hooper C, Rijsdijk F, Tabrizi SJ, Banner S, Shaw CE, Foy C, Poppe M, Archer N, Hamilton G, Powell J, Brown RG, Sham P, Ward M, Lovestone S (2006) Proteome-based plasma biomarkers for Alzheimer’s disease. Brain 129, 30423050. Bartel DP (2004) MicroRNAs: Genomics, biogenesis, mechanism, and function. Cell 116, 281-297. Dillhoff M, Wojcik SE, Bloomston M (2009) MicroRNAs in solid tumors. J Surg Res 154, 349-354. Mitchell PS, Parkin RK, Kroh EM, Fritz BR, Wyman SK, Pogosova-Agadjanyan EL, Peterson A, Noteboom J, O’Briant KC, Allen A, Lin DW, Urban N, Drescher CW, Knudsen BS, Stirewalt DL, Gentleman R, Vessella RL, Nelson PS, Martin DB, Tewari M (2008) Circulating microRNAs as stable bloodbased markers for cancer detection. Proc Natl Acad Sci U S A 105, 10513-10518. Ura S, Honda M, Yamashita T, Ueda T, Takatori H, Nishino R, Sunakozaka H, Sakai Y, Horimoto K, Kaneko S (2009) Differential microRNA expression between hepatitis B and hepatitis C leading disease progression to hepatocellular carcinoma. Hepatology 49, 1098-1112. Ikeda S, Kong SW, Lu J, Bisping E, Zhang H, Allen PD, Golub TR, Pieske B, Pu WT (2007) Altered microRNA expression in human heart disease. Physiol Genomics 31, 367-373. Sempere LF, Freemantle S, Pitha-Rowe I, Moss E, Dmitrovsky E, Ambros V (2004) Expression profiling of mammalian microRNAs uncovers a subset of brain-expressed microRNAs with possible roles in murine and human neuronal differentiation. Genome Biol 5, R13. Kosik KS, Krichevsky AM (2005) The elegance of the microRNAs: A neuronal perspective. Neuron 47, 779-782. Kosik KS (2006) The neuronal microRNA system. Nat Rev Neurosci 7, 911-920. Maes OC, Chertkow HM, Wang E, Schipper HM (2009) MicroRNA: Implications for Alzheimer Disease and other human CNS disorders. Curr Genomics 10, 154-168. Hebert SS, Horre K, Nicolai L, Papadopoulou AS, Mandemakers W, Silahtaroglu AN, Kauppinen S, Delacourte A, De Strooper B (2008) Loss of microRNA cluster miR-29a/b1 in sporadic Alzheimer’s disease correlates with increased

[17]

[18]

[19]

[20]

[21]

[22]

[23]

[24]

[25]

[26]

[27] [28]

[29]

BACE1/betasecretase expression. Proc Natl Acad Sci U S A 105, 6415-6420. Ohyagi Y, Asahara H, Chui DH, Tsuruta Y, Sakae N, Miyoshi K, Yamada T, Kikuchi H, Taniwaki T, Murai H, Ikezoe K, Furuya H, Kawarabayashi T, Shoji M, Checler F, Iwaki T, Makifuchi T, Takeda K, Kira J, Tabira T (2005) Intracellular Abeta42 activates p53 promoter: A pathway to neurodegeneration in Alzheimer’s disease. FASEB J 19, 255-257. He L, He X, Lim LP, de Stanchina E, Xuan Z, Liang Y, Xue W, Zender L, Magnus J, Ridzon D, Jackson AL, Linsley PS, Chen C, Lowe SW, Cleary MA, Hannon GJ (2007) A microRNA component of the p53 tumour suppressor network. Nature 447, 1130-1134. Mitchell PS, Parkin RK, Kroh EM, Fritz BR, Wyman SK, Pogosova-Agadjanyan EL, Peterson A, Noteboom J, O’Briant KC, Allen A, Lin DW, Urban N, Drescher CW, Knudsen BS, Stirewalt DL, Gentleman R, Vessella RL, Nelson PS, Martin DB, Tewari M (2008) Circulating microRNAs as stable bloodbased markers for cancer detection. Proc Natl Acad Sci U S A 105, 10513-10518. Baraniskin A, Kuhnhenn J, Schlegel U, Chan A, Deckert M, Gold R, Maghnouj A, Z¨ollner H, Reinacher-Schick A, Schmiegel W, Hahn SA, Schroers R (2011) Identification of microRNAs in the cerebrospinal fluid as marker for primary diffuse large B-cell lymphoma of the central nervous system. Blood 117, 3140-3146. Hanke M, Hoefig K, Merz H, Feller AC, Kausch I, Jocham D, Warnecke JM, Sczakiel G (2010) A robust methodology to study urine microRNA as tumor marker: MicroRNA-126 and microRNA-182 are related to urinary bladder cancer. Urol Oncol 28, 655-661. Park NJ, Zhou H, Elashoff D, Henson BS, Kastratovic DA, Abemayor E, Wong DT (2009) Salivary microRNA: Discovery, characterization, and clinical utility for oral cancer detection. Clin Cancer Res 15, 5473-5477. Michael A, Bajracharya SD, Yuen PS, Zhou H, Star RA, Illei GG, Alevizos I (2010) Exosomes from human saliva as a source of microRNA biomarkers. Oral Dis 16, 34-38. Cogswell JP, Ward J, Taylor IA, Waters M, Shi Y, Cannon B, Kelnar K, Kemppainen J, Brown D, Chen C, Prinjha RK, Richardson JC, Saunders AM, Roses AD, Richards CA (2008) Identification of miRNA changes in Alzheimer’s disease brain and CSF yields putative biomarkers and insights into disease pathways. J Alzheimers Dis 14, 27-41. Schipper HM, Maes OC, Chertkow HM, Wang E (2007) MicroRNA expression in Alzheimer blood mononuclear cells. Gene Regul Syst Bio 1, 263-274. McKhann G, Drachman D, Folstein M, Katzman R, Price D, Stadlan EM (1984) Clinical diagnosis of Alzheimer’s disease: Report of the NINCDS-ADRDA Work Group under the auspices of Department of Health and Human Services Task Force on Alzheimer’s Disease. Neurology 34, 939-944. Waldman SA, Terzic A (2008) MicroRNA signatures as diagnostic and therapeutic targets. Clin Chem 54, 943-944. Chen X, Ba Y, Ma L, Cai X, Yin Y, Wang K, Guo J, Zhang Y, Chen J, Guo X, Li Q, Li X, Wang W, Zhang Y, Wang J, Jiang X, Xiang Y, Xu C, Zheng P, Zhang J, Li R, Zhang H, Shang X, Gong T, Ning G, Wang J, Zen K, Zhang J, Zhang CY (2008) Characterization of microRNAs in serum: A novel class of biomarkers for diagnosis of cancer and other diseases. Cell Res 18, 997-1006. Huang Z, Huang D, Ni S, Peng Z, Sheng W, Du X (2010) Plasma microRNAs are promising novel biomarkers for early detection of colorectal cancer. Int J Cancer 127, 118-126.

T. Kiko et al. / miRNAs Determination in Plasma and CSF of AD Patients [30]

[31]

[32]

[33] [34]

[35]

[36]

[37]

[38]

[39]

[40]

[41]

Laterza OF, Lim L, Garrett-Engele PW, Vlasakova K, Muniappa N, Tanaka WK, Johnson JM, Sina JF, Fare TL, Sistare FD, Glaab WE (2009) Plasma microRNAs as sensitive and specific biomarkers of tissue injury. Clinl Chem 55, 1977-1983. Wang K, Zhang S, Marzolf B, Troisch P, Brightman A, Hu Z, Hood LE, Galas DJ (2009) Circulating microRNAs, potential biomarkers for drug-induced liver injury. Proc Natl Acad Sci U S A 106, 4402-4407. Schonrock N, Matamales M, Ittner LM, G¨otz J (2012) MicroRNA networks surrounding APP and amyloid-␤ metabolism-implications for Alzheimer’s disease. Exp Neurol 235, 447-454. Tan L, Yu JT, Hu N, Tan L (2013) Non-coding RNAs in Alzheimer’s disease. Mol Neurobiol 47, 382-393. Alexandrov PN, Dua P, Hill JM, Bhattacharjee S, Zhao Y, Lukiw WJ (2012) microRNA (miRNA) speciation in Alzheimer’s disease (AD) cerebrospinal fluid (CSF) and extracellular fluid (ECF). Int J Biochem Mol Biol 3, 365-373. Fukumoto H, Cheung BS, Hyman BT, Irizarry MC (2002) Beta-secretase protein and activity are increased in the neocortex in Alzheimer disease. Arch Neurol 59, 1381-1389. Holsinger RM, McLean CA, Beyreuther K, Masters CL, Evin G (2002) Increased expression of the amyloid precursor betasecretase in Alzheimer’s disease. Ann Neurol 51, 783-786. Achard P, Herr A, Baulcombe DC, Harberd NP (2004) Modulation of floral development by a gibberellin-regulated microRNA. Development 131, 3357-3365. Calin GA, Liu CG, Ferracin M, Hyslop T, Spizzo R, Sevignani C, Fabbri M, Cimmino A, Lee EJ, Wojcik SE, Shimizu M, Tili E, Rossi S, Taccioli C, Pichiorri F, Liu X, Zupo S, Herlea V, Gramantieri L, Lanza G, Alder H, Rassenti L, Volinia S, Schmittgen TD, Kipps TJ, Negrini M, Croce CM (2007) Ultraconserved regions encoding ncRNAs are altered in human leukemias and carcinomas. Cancer Cell 12, 215-229. Mattson MP, Shea TB (2003) Folate and homocysteine metabolism in neural plasticity and neurodegenerative disorders. Trends Neurosci 26, 137-146. Shioya M, Obayashi S, Tabunoki H, Arima K, Saito Y, Ishida T, Satoh J (2010) Aberrant microRNA expression in the brains of neurodegenerative diseases: miR-29a decreased in Alzheimer disease brains targets neurone navigator 3. Neuropathol Appl Neurobiol 36, 320-330. Li X, Khanna A, Li N, Wang E (2011) Circulatory miR34a as an RNAbased, noninvasive biomarker for brain aging. Aging (Albany NY) 3, 985-1002.

[42]

[43]

[44]

[45]

[46] [47]

[48]

[49]

[50]

[51] [52]

[53]

259

Wang X, Liu P, Zhu H, Xu Y, Ma C, Dai X, Huang L, Liu Y, Zhang L, Qin C (2010) miR-34a, a microRNA up-regulated in a double transgenic mouse model of Alzheimer’s disease, inhibits bcl2 translation. Brain Res Bull 80, 268-273. Ferreiro E, Eufr´asio A, Pereira C, Oliveira CR, Rego AC (2007) Bcl-2 overexpression protects against amyloid-beta and prion toxicity in GT1-7 neural cells. J Alzheimers Dis 12, 223-228. Rohn TT, Vyas V, Hernandez-Estrada T, Nichol KE, Christie LA, Head E (2008) Lack of pathology in a triple transgenic mouse model of Alzheimer’s disease after overexpression of the anti-apoptotic protein Bcl-2. J Neurosci 19, 3051-3059. Li YY, Cui JG, Hill JM, Bhattacharjee S, Zhao Y, Lukiw WJ (2011) Increased expression of miRNA-146a in Alzheimer’s disease transgenic mouse models. Neurosci Lett 487, 94-98. Mattick JS, Makunin IV (2005) Small regulatory RNAs in mammals. Hum Mol Genet 14, 121-132. Sethi P, Lukiw WJ (2009) Micro-RNA abundance and stability in human brain: Specific alterations in Alzheimer’s disease temporal lobe neocortex. Neurosci Lett 459, 100-104. Ferretti E, De Smaele E, Po A, Di Marcotullio L, Tosi E, Espinola MS, Di Rocco C, Riccardi R, Giangaspero F, Farcomeni A, Nofroni I, Laneve P, Gioia U, Caffarelli E, Bozzoni I, Screpanti I, Gulino A (2009) MicroRNA profiling in human medulloblastoma. Int J Cancer 124, 568-577. Papadia S, Soriano FX, L´eveill´e F, Martel MA, Dakin KA, Hansen HH, Kaindl A, Sifringer M, Fowler J, Stefovska V, McKenzie G, Craigon M, Corriveau R, Ghazal P, Horsburgh K, Yankner BA, Wyllie DJ, Ikonomidou C, Hardingham GE (2008) Synaptic NMDA receptor activity boosts intrinsic antioxidant defenses. Nat Neurosci 11, 476-487. Mehler MF, Mattick JS (2007) Noncoding RNAs and RNA editing in brain development, functional diversification, and neurological disease. Physiol Rev 87, 799-823. Lukiw WJ (2007) Micro-RNA speciation in fetal, adult and Alzheimer’s disease hippocampus. Neuroreport 18, 297-300. Kosaka N, Iguchi H, Ochiya T (2010) Circulating microRNA in body fluid: A new potential biomarker for cancer diagnosis and prognosis. Cancer Sci 101, 2087-2092. Baraniskin A, Kuhnhenn J, Schlegel U, Chan A, Deckert M, Gold R, Maghnouj A, Z¨ollner H, Reinacher-Schick A, Schmiegel W, Hahn SA, Schroers R (2011) Identification of microRNAs in the cerebrospinal fluid as marker for primary diffuse large B-cell lymphoma of the central nervous system. Blood 117, 3140-3146.

MicroRNAs in plasma and cerebrospinal fluid as potential markers for Alzheimer's disease.

The development of Alzheimer's disease (AD) biomarkers remains an unmet challenge, and new approaches that can improve current AD biomarker strategies...
155KB Sizes 0 Downloads 0 Views