Generic placeholder image

Current Alzheimer Research

Editor-in-Chief

ISSN (Print): 1567-2050
ISSN (Online): 1875-5828

Research Article

Metabolic Alterations in the Outer Membrane Vesicles of Patients with Alzheimer’s Disease: An LC-MS/MS-based Metabolomics Analysis

Author(s): Shou-Chao Wei, Wei Wei, Wan-Juan Peng, Zhou Liu*, Zhi-You Cai* and Bin Zhao*

Volume 16, Issue 13, 2019

Page: [1183 - 1195] Pages: 13

DOI: 10.2174/1567205016666191121141352

Price: $65

Abstract

Objective: To characterize the specific metabolomics profiles in the outer membrane vesicles (OMVs) of patients with Alzheimer’s Disease (AD) and to explore potential metabolic biomarkers and their diagnostic roles.

Methods: Nine AD patients and age- and sex-matched healthy controls were enrolled, and feces were collected. OMVs were extracted, purified, and then analyzed using liquid chromatography-tandem mass chromatography (LC-MS/MS) method coupled with a series of multivariate statistical analyses.

Results: Remarkable differences were found between the OMVs from AD patients and those from healthy controls. A number of differential metabolites and several top-altered metabolic pathways were identified. The levels of aspartate, L-aspartate, imidazole-4-acetate and L-glutamate were confirmed to be highly upregulated in AD-OMVs. Other differential metabolites, such as arachidic acid, prostaglandin G2, and leukotriene B4, were also identified. Furthermore, the differential metabolites possessed higher areas under the ROC curve (AUCs).

Conclusion: Metabolic activity is significantly altered in the OMVs from AD patients. This data might be helpful for identifying novel biomarkers and their diagnostic roles in AD.

Furthermore, OMVs metabolomics analysis combined with GWAS could enrich our understanding of the genetic spectrum of AD and lead to early predictions and diagnosis and clinical applications of better AD treatments.

Keywords: Alzheimer’s disease, outer membrane vesicles, metabolomics, dementia, LC-MS/MS, genome-wide association studies.

[1]
Chang C-H, Lane H-Y, Lin C-H. Brain Stimulation in Alzheimer’s Disease. Front Psychiatry 9: 201. (2018)
[2]
2016 Alzheimer’s disease facts and figures. Alzheimer’s Dementia: J Alzheimer’s Assoc 12: 459-509. (2016)
[3]
Li K, Wei S, Liu Z, Hu L, Lin J, Tan S, et al. The prevalence of alzheimer’s disease in china: a systematic review and meta-analysis. Iran J Public Health 47: 1615-26. (2018)
[4]
Codagnone MG, Spichak S, O’Mahony SM, O’Leary OF, Clarke G, Stanton C, et al. Programming bugs: microbiota and the developmental origins of brain health and disease. Biol Psychiatry 85: 150-63. (2019)
[5]
Wang HX, Wang YP. Gut Microbiota-brain Axis. Chin Med J 129: 2373-80. (2016)
[6]
Mancuso C, Santangelo R. Alzheimer’s disease and gut microbiota modifications: the long way between preclinical studies and clinical evidence. Pharmacol Res 129: 329-36. (2018)
[7]
Dinan TG, Cryan JF. Brain-gut-microbiota axis and mental health. Psych Med 79: 920-6. (2017)
[8]
Liu P, et al. Altered microbiomes distinguish Alzheimer’s disease from amnestic mild cognitive impairment and health in a Chinese cohort. Brain, behavior, and immunity 80: 633-43. (2019)
[http://dx.doi.org/10.1016/j.bbi.2019.05.008]
[9]
Harach T, Marungruang N, Duthilleul N, Cheatham V, Mc Coy KD, Frisoni G, et al. Reduction of Abeta amyloid pathology in APPPS1 transgenic mice in the absence of gut microbiota. Sci Rep 7: 41802. (2017)
[10]
Chutkan H, Macdonald I, Manning A, Kuehn MJ. Quantitative and qualitative preparations of bacterial outer membrane vesicles. Methods in molecular biology (Clifton, NJ) 966: 259-72. (2013)
[11]
O’Donoghue EJ, Sirisaengtaksin N. Lipopolysaccharide structure impacts the entry kinetics of bacterial outer membrane vesicles into host cells. Aging 13e1006760 (2017)
[12]
Koeppen K, Hampton TH, Jarek M, Scharfe M, Gerber SA, Mielcarz DW, et al. A novel mechanism of host-pathogen interaction through sRNA in bacterial outer membrane vesicles. PLoS Pathog 12e1005672 (2016)
[13]
Fiehn O. Combining genomics, metabolome analysis, and biochemical modelling to understand metabolic networks. Compar Fun Genom 2: 155-68. (2001)
[14]
Rae CD, Williams SR. Glutathione in the human brain: Review of its roles and measurement by magnetic resonance spectroscopy. Anal Biochem 529: 127-43. (2017)
[15]
Király M, Dalmadiné Kiss B, Vékey K, Antal I, Ludányi K. Mass spectrometry: past and present. Acta pharmaceutica Hungarica 86: 3-11. (2016)
[16]
Zakharzhevskaya NB, Vanyushkina AA, Altukhov IA, Shavarda AL, Butenko IO, Rakitina DV, et al. Outer membrane vesicles secreted by pathogenic and nonpathogenic Bacteroides fragilis represent different metabolic activities. Scientifi Rep 7: 5008. (2017)
[17]
Bryant WA, Stentz R, Le Gall G, Sternberg MJE, Carding SR, Wilhelm T. Analysis of the small molecule content of outer membrane vesicles produced by indicates an extensive metabolic link between microbe and host. Front Microbiol 8: 2440. (2017)
[18]
McKhann G, Drachman D, Folstein M, Katzman R, Price D, Stadlan EM. 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: 939-44. (1984)
[19]
Boulesteix A-L, Strimmer K. Partial least squares: a versatile tool for the analysis of high-dimensional genomic data. Brief Bioinform 8: 32-44. (2007)
[20]
Wang J-B, Pu SB, Sun Y, Li ZF, Niu M, Yan XZ, et al. Metabolomic profiling of autoimmune hepatitis: the diagnostic utility of nuclear magnetic resonance spectroscopy. J Proteome Res 13: 3792-801. (2014)
[21]
Goentoro L, Kirschner MW. Evidence that fold-change, and not absolute level, of beta-catenin dictates Wnt signaling. Mol Cell 36: 872-84. (2009)
[22]
Michael H, Tian L, Ghebremichael M. The ROC curve for regularly measured longitudinal biomarkers. Biostatistics 20: 433-51. (2019)
[23]
Baldacci F, Lista S, Cavedo E, Bonuccelli U, Hampel H. Diagnostic function of the neuroinflammatory biomarker YKL-40 in Alzheimer’s disease and other neurodegenerative diseases. Expert Rev Proteomics 14: 285-99. (2017)
[24]
Hassan M, Abbas Q, Seo SY, Shahzadi S, Al Ashwal H, Zaki N, et al. Computational modeling and biomarker studies of pharmacological treatment of Alzheimer’s disease.(Review). Mol Med Rep 18: 639-55. (2018)
[25]
Diaz Heijtz R, Wang S, Anuar F, Qian Y, Björkholm B, Samuelsson A, et al. Normal gut microbiota modulates brain development and behavior. Proc Natl Acad Sci USA 108: 3047-52. (2011)
[26]
Collins SM, Surette M, Bercik P. The interplay between the intestinal microbiota and the brain. Nat Rev Microbiol 10: 735-42. (2012)
[27]
Medzhitov R. Recognition of microorganisms and activation of the immune response. Nature 449: 819-26. (2007)
[28]
Banks WA. The blood-brain barrier in psychoneuroimmunology. Immunol Allergy Clin North Am 29: 223-8. (2009)
[29]
Vizcarra JA, Wilson-Perez HE, Espay AJ. The power in numbers: gut microbiota in Parkinson’s disease. Mov Disord 30: 296-8. (2015)
[30]
Moco S, Bino RJ, De Vos RCH, Vervoort J. Metabolomics technologies and metabolite identification. Trends Analyt Chem 26: 855-66. (2007)
[31]
Kim YH, Shim HS, Kim KH, et al. Metabolomic analysis identifies alterations of amino acid metabolome signatures in the postmortem brain of alzheimer’s disease. Exp Neurobiol 28: 376-89. (2019)
[32]
Tsuruoka M, Hara J, Hirayama A, Sugimoto M, Soga T, Shankle WR, et al. Capillary electrophoresis-mass spectrometry-based metabolome analysis of serum and saliva from neurodegenerative dementia patients. Electrophoresis 34: 2865-72. (2013)
[33]
Sanchez-Mejia RO, Mucke L. Phospholipase A2 and arachidonic acid in Alzheimer’s disease. Biochimica et Biophysica Acta 1801: 784-90. (2010)
[34]
Choi S-H, Bosetti F. Cyclooxygenase-1 null mice show reduced neuroinflammation in response to beta-amyloid. Aging (Albany NY) 1: 234-44. (2009)
[35]
Rinne JO, Rinne JO, Laakso K, Lönnberg P, Mölsä P, Paljärvi L, et al. Brain muscarinic receptors in senile dementia. Brain Res 336: 19-25. (1985)
[36]
Sanchez-Mejia RO, Newman JW, Toh S, Yu GQ, Zhou Y, Halabisky B, et al. Phospholipase A2 reduction ameliorates cognitive deficits in a mouse model of Alzheimers disease. Nat Neurosci 11: 1311. (2008)
[37]
Esposito G, Giovacchini G, Liow JS, Bhattacharjee AK, Greenstein D, Schapiro M, et al. Imaging neuroinflammation in Alzheimer’s disease with radiolabeled arachidonic acid and PET. J Nucl Med 49: 1414-21. (2008)
[38]
Allsop D, Mayes J. Amyloid β-peptide and Alzheimer’s disease. Essays Biochem 56: 99-110. (2014)
[39]
Amtul Z, Uhrig M, Wang L, Rozmahel RF, Beyreuther K. Detrimental effects of arachidonic acid and its metabolites in cellular and mouse models of Alzheimer’s disease: structural insight. Neurobiol Aging 33(4): 831.e21-31. (2012)
[40]
Smirnov A, Henkel AW, Trupp A, Bloch E, Wiltfang J. Stimulation of platelet-secreted amyloid peptides by Ca2+ and prostaglandin 2. Alzheimers Dement 5: 174. (2009)
[41]
Sugaya K, Uz T, Kumar V, Manev H. New anti-inflammatory treatment strategy in Alzheimer’s disease. J Pharmacol 82: 85-94. (2000)
[42]
Joshi YB, Di Meco A, Praticó D. Modulation of amyloid-β production by leukotriene B4 via the γ-secretase pathway. J Alzheimers Dis 38: 503-6. (2014)
[43]
Maass A, Berron D, Harrison TM, Adams JN, La Joie R, Baker S, et al. Alzheimer’s pathology targets distinct memory networks in the ageing brain. Brain 142: 2492-09. (2019)
[44]
Campbell MA, Prusakiewicz JJ, Caouette D, Nolan CE, Cadellina GW, Lu Y, et al. The effect of 5-lipoxygenase inhibition on Aβ and inflammation in models of Alzheimer-like pathology. Alzheimers Dement 6: S567-8. (2010)
[45]
Benarroch EE, Schmeichel AM, Parisi JE, Low PA. Histaminergic tuberomammillary neuron loss in multiple system atrophy and dementia with Lewy bodies. Mov Disord 30: 1133-9. (2015)
[46]
Summers KL, Schilling KM, Roseman G, Markham KA, Dolgova NV, Kroll T, et al. X-ray absorption spectroscopy investigations of Copper(ii) coordination in the human amyloid β peptide. Inorg Chem 58: 6294-11. (2019)
[47]
Liu J, Chang L, Song Y, Li H, Wu Y. The role of NMDA receptors in Alzheimer’s disease. Front Neurosci 13: 43. (2019)
[48]
Cacabelos R, Takeda M, Winblad B. The glutamatergic system and neurodegeneration in dementia: preventive strategies in Alzheimer’s disease. International journal of geriatric psychiatry 14: 3-47. (1999)
[49]
Areosa SA, Sherriff F, McShane R. Memantine for dementia. Cochrane Database Syst Rev 20(3)CD003154 (2005)
[50]
Forsythe P, Sudo N, Dinan T, Taylor VH, Bienenstock J. Mood and gut feelings. Brain Behav Immun 24: 9-16. (2010)
[51]
Lyte M. Microbial endocrinology in the microbiome-gut-brain axis: how bacterial production and utilization of neurochemicals influence behavior. PLoS Pathogens 9: e1003726 (2013).
[52]
Gibb AJ. Choline and acetylcholine: what a difference an acetate makes. J Physiol 595: 1021-2. (2017)
[53]
Cui Y, Liu X, Wang M, Liu L, Sun X, Ma L, et al. Lysophosphatidylcholine and amide as metabolites for detecting Alzheimer disease using ultrahigh-performance liquid chromatography-quadrupole time-of-flight mass spectrometry-based metabonomics. J Neuropathol Exp Neurol 73: 954-63. (2014)
[54]
Cryan JF, O’Riordan KJ, Cowan CSM, Sandhu KV, Bastiaanssen TFS, Boehme M, et al. The microbiota-gut-brain axis. Physiol Rev 99: 1877-2013. (2019)
[55]
Chen X, D’Souza R, Hong ST. The role of gut microbiota in the gut-brain axis: current challenges and perspectives. Protein Cell 4: 403-14. (2013)
[56]
Muraca M, Putignani L, Fierabracci A, Teti A, & Perilongo G. Gut microbiota-derived outer membrane vesicles: under-recognized major players in health and disease? Discovery medicine 19: 343-8. (2015)
[57]
Ratajczak W, Rył A, Mizerski A, Walczakiewicz K, Sipak O, Laszczyńska M. Immunomodulatory potential of gut microbiome-derived short-chain fatty acids (SCFAs). Acta Biochimica Pol 66: 1-12. (2019)
[58]
Shen L, Jia J. An overview of genome-wide association studies in Alzheimer’s disease. Neurosci Bulletin 3: 183-90. (2016)
[59]
Martinelli-Boneschi F, Giacalone G, Magnani G, Biella G, Coppi E, Santangelo R, et al. Pharmacogenomics in Alzheimer's disease: a genome-wide association study of response to cholinesterase inhibitors Neurobiol Aging 34: 1711.e1717-1713(2013)
[60]
Davenport ER, Cusanovich DA, Michelini K, Barreiro LB, Ober C, Gilad Y. Genome-wide association studies of the human gut microbiota. PLoS One 10e0140301 (2015)

Rights & Permissions Print Cite
© 2024 Bentham Science Publishers | Privacy Policy