Loading

Commentary Open Access
Volume 1 | Issue 3 | DOI: https://doi.org/10.33696/Neurol.1.016

Effect of Exosomes on Alzheimer’s Disease

  • 1Department of Neurology, People’s Hospital of Zhengzhou University, Zhengzhou, Henan, 450003, China
+ Affiliations - Affiliations

*Corresponding Author

Jiewen Zhang, zhangjiewen9900@126.com

Received Date: June 29, 2020

Accepted Date: July 31, 2020

Abstract

Alzheimer’s disease (AD) is a neurodegenerative disease characterized by latent and progressive onset. At present, there is a lack of effective diagnosis and treatment of AD, and AD constitutes an exceptional burden on the lives of patients and their families. The study of exosomes has uncovered promising new possibilities for the diagnosis and treatment of AD. This commentary describes the role of exosomes in Alzheimer’s disease.

 

Keywords

Exosomes; Alzheimer’s disease; Urinary

Commentary

AD is a neurodegenerative disease characterized by progressive cognitive impairment, behavioral changes, memory loss and executive dysfunction, all of which present serious threats to the health of older people [1]. It has been proposed that the pathology of AD spreads between functionally and anatomically connected brain regions through prion-like mechanisms [2-6], which promote the progression of neurological diseases. Methods for early diagnosis and treatment of AD are urgently needed in order to delay the progress of the disease and preserve the quality of life of patients.

After the fusion of multi-vesicle bodies (MVBs) and the plasma membrane, exosomes are released into the extracellular space [7]. When exosomes are absorbed by receptor cells, they transfer their contents [8], which can promote the transmission of pathological proteins between nerve cells and encourage the development of neurodegenerative diseases. Exocrine bodies are abundant in blood, saliva, urine, semen, amniotic fluid, ascites, cerebrospinal fluid (CSF) and breast milk [9]. There is growing evidence that exosomes freely cross the bloodbrain barrier (BBB) [10-12], and it follows that exosomes in CSF can cross the blood-brain barrier to reach the blood and then pass through the kidneys into the urine. Given consistent control of other factors , such as age, region, education level, and no other related diseases, changes in the contents of plasma and urine exosomes can reflect changes in the brain to a certain extent.

Exosomes vary in size and shape depending on their origin and function [13], and they can be distinguished in clinical studies by transmission electron microscopy (TEM). Negative staining is considered to be the best method for TEM imaging of exocrine bodies [14], because it can detect most exosomes. Gradual technical advances in this method have made it possible to observe the detailed structure of exosomes [15].

Nanoparticle tracking analysis (NTA) is a method that was developed in recent years allowing quantitative measurement of the size of particles with diameters ranging from 30 nm to 1 μm. It can also be used to measure the diameter and number of exosomes. The NTA technology depends on the NanoSight instrument, which is equipped with a highly sensitive camera and a vertical microscope with a laser light source. The particle size distribution and particle number in the liquid suspension are obtained by using the characteristics of light scattering and Brownian motion. The laser beam passes through the sample chamber and follows the path of scattered light through the particles in the suspension, so that the particles can be observed through a 20x magnification microscope with a camera. Einstein’s equation is used to calculate the hydrodynamic diameter of the particles.

In the manuscript titled “A Pilot Study of Urinary Exosomes in Alzheimer’s Disease”, enzyme-linked immunosorbent assay was used to detect the levels of Aβ1–42 and P-S396-tau (normalized by CD63) in urinary exosomes derived from AD patients and matched healthy subjects. TEM and NTA were used to characterize the exosomes. Urinary exosomes from AD patients were found to contain higher levels of Aβ1–42 and P-S396-tau compared to those of matched healthy controls, and exosomes taken from AD patients were also more numerous. The difference in levels of Aβ1–42 and P-S396-tau and the disparate quantity of urinary exosomes between AD patients and healthy controls may provide a basis for early diagnosis of AD.

However, the sample size in this study is relatively small, and the sample may not be representative. Studies of pathological protein in urinary exosomes of patients with AD are relatively few, and the difficulty of operation and experimental uncertainty are higher than in detection of plasma exosomes. More samples are needed to verify the conclusion. In addition, the vesicles tested in the study included exosomes as well as other extracellular vesicles. If further purification is done using technology such as neuron-derived markers (NCAM or L1CAM antibodies), which only exist on the surface of neuron-derived exosomes, but not on other vesicles, the results will be more reliable [16].

Intracellular Aβ accumulation is the result of amyloid precursor protein (APP-transport imbalance where in extra proteins that exceed the degradation capacity of glial cells and lysosomes are released into the extracellular space among brain cells by exosomes [17-19]. Phosphorylated tau protein accumulation leads to the formation of doublestranded microfilaments that in turn accumulate to form neurofibrillary tangles and then destroy the normal function of neurons, eventually leading to the onset of AD [20]. In AD, the level of tau protein is 300% higher than in normal older adults, and hyperphosphorylated tau protein plays a major role in the progression of AD [21]. Upregulation and then failure of neuronal autophagic-lysosomal systems in patients with AD may cause lysosomal proteins to be added to exosome shipments, inappropriately facilitating their removal from neurons [22,23], so the level of lysosomal proteins in exosomes in patients with AD may be aberrant. The direct neuropathological consequences of insulin resistance include increased formation and accumulation of Aβ1-42 oligomer, increased level of tau phosphorylation, and higher levels of insulin in the brain, which can competitively inhibit the degradation of Aβ1-42 protease [24-26]. It can be concluded then that insulin resistance plays a key role in the disease process of AD. A decrease in the level of synaptic protein SNAP- 25 is considered to be a biomarker indicating the degree of synaptic degeneration [27,28]. Loss of synaptic density and connectivity, accompanied by decreased expression of synaptic protein SNAP-25, has been observed in multiple brain regions of patients with AD.

Regarding treatment, only highly fat soluble substances with molecular weights below 400Da can reach the brain through blood circulation due to the blood-brain barrier. However, over 98% of drug molecules fail to meet this standard, so the effect of most drugs is limited [29]. In recent years, in order to improve intracranial drug delivery, researchers have used a variety of methods, such as surgery, infusion of hypertonic fluid, and drug chemical modification, to increase the permeability of the bloodbrain barrier and consequently increase the total amount of drugs reaching the brain. However, this method also inevitably increases the risk of intracranial infection and may have more adverse effects on patients. Because its lipid bimolecular structure can effectively load hydrophobic and hydrophilic drugs, and can freely cross the blood-brain barrier [30], exosomes are also considered ideal carriers for drug delivery [31]. Exosomes’ lipid bimolecular structures can effectively load both hydrophobic and hydrophilic drugs [32]. In the process of ligand receptor-mediated drug delivery, exosomes facilitate targeting [33], laying a foundation for highly specific therapies for nervous system diseases such as AD.

AD is a progressive neurodegenerative disease. The vast majority of AD patients do not show any symptoms even in the few years immediately preceding the onset of dementia, although memory impairment may sometimes reach the threshold for diagnosis [34]. At this time, the effect of drug treatment is very limited, seriously affecting the quality of life of patients as well as their families. Therefore, the development of methods allowing early diagnosis before the clinical stage is imperative, because preventive treatment may be more effective. The study of exosomes has opened up a new path for the diagnosis and treatment of AD. It is believed that early diagnosis and treatment of AD will be finally possible with further study.

 

Conflicts of Interest

The authors have no relevant disclosures or conflicts of interest to declare.

Acknowledgements

We would like to thank all subjects for their participation, as well as all further contributors for their work and support for this study. This research was supported by the National Natural Science Foundation of China (Grants 81671068, 81873727) and Key Science and Technology Program of Henan Province, China (201701020).

 

References

1. Murakami K, Watanabe T, Koike T, Kamata M, Igari T, Kondo S. Pharmacological properties of a novel and potent γ-secretase modulator as a therapeutic option for the treatment of Alzheimer’s disease. Brain Research. 2016 Feb 15;1633:73-86.

2. Eisele YS, Bolmont T, Heikenwalder M, Langer F, Jacobson LH, Yan ZX, et al. Induction of cerebral ß-amyloidosis: intracerebral versus systemic Aß inoculation. Proceedings of the National Academy of Sciences. 2009 Aug 4;106(31):12926-31.

3. Frost B, Diamond MI. Prion-like mechanisms in neurodegenerative diseases. Nature Reviews Neuroscience. 2010 Mar;11(3):155-9.

4. Acquatella-Tran Van Ba I, Imberdis T, Perrier V. From prion diseases to prion-like propagation mechanisms of neurodegenerative diseases. International Journal of Cell Biology. 2013 Oct 10;2013.

5. Braak H, Braak E. Neuropathological stageing of Alzheimer-related changes. Acta Neuropathologica. 1991 Sep 1;82(4):239-59.

6. Braak H, Rüb U, Schultz C, Tredici KD. Vulnerability of cortical neurons to Alzheimer’s and Parkinson’s diseases. Journal of Alzheimer’s Disease. 2006 Jan 1;9(s3):35-44.

7. Kowal J, Arras G, Colombo M, Jouve M, Morath JP, Primdal-Bengtson B, et al. Proteomic comparison defines novel markers to characterize heterogeneous populations of extracellular vesicle subtypes. Proceedings of the National Academy of Sciences. 2016 Feb 23;113(8):E968-77.

8. Février B, Raposo G. Exosomes: endosomal-derived vesicles shipping extracellular messages. Current Opinion in Cell Biology. 2004 Aug 1;16(4):415-21.

9. Urbanelli L, Magini A, Buratta S, Brozzi A, Sagini K, Polchi A, et al. Signaling pathways in exosomes biogenesis, secretion and fate. Genes. 2013 Jun;4(2):152-70.

10. Wiklander OP, Nordin JZ, O’Loughlin A, Gustafsson Y, Corso G, Mäger I, et al. Extracellular vesicle in vivo biodistribution is determined by cell source, route of administration and targeting. Journal of Extracellular Vesicles. 2015 Jan 1;4(1):26316.

11. Bala S, Csak T, Momen-Heravi F, Lippai D, Kodys K, Catalano D, et al. Biodistribution and function of extracellular miRNA-155 in mice. Scientific Reports. 2015 May 29;5(1):1-2.

12. Chen CC, Liu L, Ma F, Wong CW, Guo XE, Chacko JV, et al. Elucidation of exosome migration across the blood–brain barrier model in vitro. Cellular and Molecular Bioengineering. 2016 Dec 1;9(4):509-29.

13. Lötvall J, Hill AF, Hochberg F, Buzás EI, Di Vizio D, Gardiner C, et al. Minimal experimental requirements for definition of extracellular vesicles and their functions: a position statement from the International Society for Extracellular Vesicles.

14. Théry C, Amigorena S, Raposo G, Clayton A. Isolation and characterization of exosomes from cell culture supernatants and biological fluids. Current Protocols in Cell Biology. 2006 Mar;30(1):3-22.

15. Liu X, Wang HW. Single particle electron microscopy reconstruction of the exosome complex using the random conical tilt method. JoVE (Journal of Visualized Experiments). 2011 Mar 28(49):e2574.

16. Goetzl EJ, Boxer A, Schwartz JB, Abner EL, Petersen RC, Miller BL, et al. Low neural exosomal levels of cellular survival factors in Alzheimer’s disease. Annals of Clinical and Translational Neurology. 2015 Jul;2(7):769-73.

17. Eitan E, Hutchison ER, Marosi K, Comotto J, Mustapic M, Nigam SM, et al. Extracellular vesicle-associated Aß mediates trans-neuronal bioenergetic and Ca 2+-handling deficits in Alzheimer’s disease models. NPJ Aging and Mechanisms of Disease. 2016 Sep 22;2(1):1-1.

18. Xiao T, Zhang W, Jiao B, Pan CZ, Liu X, Shen L. The role of exosomes in the pathogenesis of Alzheimer’disease. Translational Neurodegeneration. 2017 Dec 1;6(1):3.

19. Rajendran L, Honsho M, Zahn TR, Keller P, Geiger KD, Verkade P, et al. Alzheimer’s disease ß-amyloid peptides are released in association with exosomes. Proceedings of the National Academy of Sciences. 2006 Jul 25;103(30):11172-7.

20. Metaxas A, Kempf SJ. Neurofibrillary tangles in Alzheimer’s disease: elucidation of the molecular mechanism by immunohistochemistry and tau protein phospho-proteomics. Neural Regeneration Research. 2016 Oct;11(10):1579.

21. Banzhaf-Strathmann J, Benito E, May S, Arzberger T, Tahirovic S, Kretzschmar H, et al. Micro RNA-125b induces tau hyperphosphorylation and cognitive deficits in Alzheimer’s disease. The EMBO Journal. 2014 Aug 1;33(15):1667-80.

22. Inouye H, Gleason KA, Zhang D, Decatur SM, Kirschner DA. Differential effects of phe19 and phe20 on fibril formation by amyloidogenic peptide Aß16–22 (Ac-KLVFFAE-NH2). Proteins: Structure, Function, and Bioinformatics. 2010 Aug 1;78(10):2306-21.

23. Ihara Y, Morishima-Kawashima M, Nixon R. The ubiquitin–proteasome system and the autophagic– lysosomal system in Alzheimer disease. Cold Spring Harbor perspectives in medicine. 2012 Aug 1;2(8):a006361.

24. Craft S, Watson GS. Insulin and neurodegenerative disease: shared and specific mechanisms. The Lancet Neurology. 2004 Mar 1;3(3):169-78.

25. Planel E, Tatebayashi Y, Miyasaka T, Liu L, Wang L, Herman M, et al. Insulin dysfunction induces in vivo tau hyperphosphorylation through distinct mechanisms. Journal of Neuroscience. 2007 Dec 12;27(50):13635-48.

26. Zhao WQ, Lacor PN, Chen H, Lambert MP, Quon MJ, Krafft GA, et al. Insulin receptor dysfunction impairs cellular clearance of neurotoxic oligomeric Aß. Journal of Biological Chemistry. 2009 Jul 10;284(28):18742-53.

27. Kvartsberg H, Duits FH, Ingelsson M, Andreasen N, Öhrfelt A, Andersson K, et al. Cerebrospinal fluid levels of the synaptic protein neurogranin correlates with cognitive decline in prodromal Alzheimer’s disease. Alzheimer’s & Dementia. 2015 Oct 1;11(10):1180-90.

28. Brinkmalm A, Brinkmalm G, Honer WG, Frölich L, Hausner L, Minthon L, et al. SNAP-25 is a promising novel cerebrospinal fluid biomarker for synapse degeneration in Alzheimer’s disease. Molecular Neurodegeneration. 2014 Dec 1;9(1):53.

29. Maussang D, Rip J, van Kregten J, van den Heuvel A, van der Pol S, van der Boom B, et al. Glutathione conjugation dose-dependently increases brain-specific liposomal drug delivery in vitro and in vivo. Drug Discovery Today: Technologies. 2016 Jun 1;20:59-69.

30. Qu M, Lin Q, Huang L, Fu Y, Wang L, He S, et al. Dopamine-loaded blood exosomes targeted to brain for better treatment of Parkinson’s disease. Journal of Controlled Release. 2018 Oct 10;287:156-66.

31. Li H, Yang C, Shi Y, Zhao L. Exosomes derived from siRNA against GRP78 modified bone-marrow-derived mesenchymal stem cells suppress Sorafenib resistance in hepatocellular carcinoma. Journal of Nanobiotechnology. 2018 Dec;16(1):1-3.

32. Qu M, Lin Q, Huang L, Fu Y, Wang L, He S, et al. Dopamine-loaded blood exosomes targeted to brain for better treatment of Parkinson’s disease. Journal of Controlled Release. 2018 Oct 10;287:156-66.

33. Van Niel G, d’Angelo G, Raposo G. Shedding light on the cell biology of extracellular vesicles. Nature reviews Molecular Cell Biology. 2018 Apr;19(4):213.

34. Alzheimer’s Association. 2015 Alzheimer’s disease facts and figures. Alzheimer’s & Dementia. 2015 Mar;11(3):332-84.

Author Information X