Skip to main content

Employing nanoparticle tracking analysis of salivary neuronal exosomes for early detection of neurodegenerative diseases

Abstract

Neurodegenerative diseases are a set of progressive and currently incurable diseases that are primarily caused by neuron degeneration. Neurodegenerative diseases often lead to cognitive impairment and dyskinesias. It is now well recognized that molecular events precede the onset of clinical symptoms by years. Over the past decade, intensive research attempts have been aimed at the early diagnosis of these diseases. Recently, exosomes have been shown to play a pivotal role in the occurrence and progression of many diseases including cancer and neurodegenerative diseases. Additionally, because exosomes can cross the blood–brain barrier, they may serve as a diagnostic tool for neural dysfunction. In this review, we detail the mechanisms and current challenges of these diseases, briefly review the role of exosomes in the progression of neurodegenerative diseases, and propose a novel strategy based on salivary neuronal exosomes and nanoparticle tracking analysis that could be employed for screening the early onset of neurodegenerative diseases.

Background

Timely diagnosis of neurodegenerative diseases at an early stage provides an effective means to manage the ‘impending burden’ of the aging population globally. Alzheimer’s disease (AD) and Parkinson’s disease (PD) are the most common neurodegenerative diseases. In terms of prevalence, AD tops the list, followed by PD. The other spectrum of neurodegenerative diseases encompasses frontotemporal dementia (FTD), Huntington’s disease (HD), and amyotrophic lateral sclerosis (ALS) [1, 2]. Accumulation of amyloid fibrils in tissues is linked to many neurodegenerative diseases, although the associated protein varies among diseases, for instance, amyloid β (Aβ) in AD and α-synuclein (α-syn) in PD [3]. However, there is increasing evidence for significant overlap between misfolding proteins and various symptoms in these diseases [4]. AD is clinically characterized by decline in cognitive abilities and changes in behavior whereas pathologically, by extracellular and intracellular deposits of insoluble Aβ and neurofibrillary tangles (NFTs), respectively [5]. These senile amyloid plaques result from either overproduction of Aβ (~ 1% in familial forms) or dysfunction of Aβ clearance, which is hypothesized to be the main cause of Aβ accumulation in sporadic AD that accounts for 99% of total AD cases [6, 7]. On the contrary, NFTs consist of abnormally hyperphosphorylated insoluble forms of Tau (τ) protein. Both Aβ accumulation and NFTs can exert direct and indirect neurotoxic effects leading to inflammation that causes neuronal death. Hence, Aβ amyloid, total tau, and phosphorylated tau are considered “core” AD biomarkers [8].

On the other hand, PD is a chronic neurodegenerative disease characterized by motor impairments due to the death of dopaminergic neurons within the substantia nigra. Cognitive impairments can also follow during the course of the disease [9]. Generally, familial forms that are sporadic in origin but rare, are linked to mutations in SNCA, parkin, DJ-1, PINK-1 (PTEN-induced kinase 1), and Leucine-rich repeat kinase 2 (LRRK2) [10]. Although the detailed molecular pathogenesis is still unclear, it is widely accepted that α-syn plays a central role in causing PD pathology and is in turn responsible for neurodegeneration [11,12,13].

Both of these neurodegenerative diseases are currently incurable, and the pathological events start a decade before symptoms become severe enough to be diagnosed clinically using the current criteria. The development of PD has three stages, preclinical, prodromal, and clinical, which are characterized by the onset of neurodegeneration in the absence of symptoms, the presence of non-motor symptoms, and the occurrence of motor symptoms, respectively [14, 15]. The diagnosis of PD occurs at the clinical stage based on the presence of bradykinesia along with either tremor or rigidity. It is worth noting that by this time 70%–80% of dopamine has been lost in the striatum [16]. Similarly, to improve AD diagnosis, the progression of the disease is broken down majorly into three categories: preclinical, mild cognitive impairment (MCI), and dementia [17, 18]. The preclinical stage signifies no cognitive symptoms but some signs of pathology upon brain imaging, whereas MCI represents a transition zone between normal aging and AD. It is noteworthy that the annual conversion rate of MCI to AD is around 30% [19, 20]. Finally, accurate and early diagnosis is further complicated by overlaps in clinical features that hinder the discrimination between diseases. For example, mild PD associated with dementia has similar patterns of cognitive dysfunction as mild AD.

ALS is a neuromuscular disorder resulting from protein inclusions formed from TAR DNA-binding protein of 43 kDa (TDP-43), Cu/Zn superoxide dismutase (SOD-1) or fused in sarcoma (FUS) within motor neurons [21], which result in loss of motor functions, progressive degeneration and ultimately death from asphyxiation or inanition [22]. Sporadic ALS accounts for almost 90% of the cases and the remaining 10% falls under the familial form of the disease [23]. The diagnosis of ALS is difficult due to the variability in patient presentation and lack of a definitive biomarker for the disease. A primary pathological marker of ALS is the deposition of ubiquitinylated or hyperphosphorylated protein inclusions in motor neurons and glia of the spinal cord/brainstem and motor cortex. Most of the cases (~ 97%) show TDP-43 pathology rather than SOD-1 (~ 2%) or FUS (~ 1%) aggregates. It is also worth noting that aggregates of SOD-1 or FUS are only associated with mutations, whereas TDP-43 aggregates can result from the wild-type protein in a sporadic form and be associated with mutations in familial ALS [24].

HD is also an incurable neurodegenerative disease and the most common form of inherited neurodegenerative disease. HD is characterized by uncontrolled and excessive motor movements along with a significant cognitive loss. HD is associated with abnormal expansion of a CAG repeat in the IT15 gene that results in abnormal numbers of glutamine repeats (polyQ) in the huntingtin (Htt) protein. Six to 34 CAG repeats are normal but a person with over 40 repeats will develop HD and usually die 10–15 years after disease onset [25]. Therefore, a pathological hallmark of HD is the intracellular aggregates of mutant Htt called inclusion bodies. The most accepted causes for aggregation of mutant Htt are “proteasomal impairment” and various posttranslational modifications such as ubiquitination, sumoylation and acetylation [26, 27].

With a need for an efficient diagnostic methodology for neurodegenerative diseases, a variety of approaches have been tried ranging from tracking subtle functional changes in the brain using positron emission tomography (PET) [28,29,30] to identifying biomarkers in the skin [9] and body fluids like blood [31,32,33], urine [34, 35], cerebrospinal fluid [36,37,38], and whole saliva [39,40,41]. The utility of volatile organic compounds in breath samples has also been explored for early diagnosis and staging of PD [42]. Recently, exosomes from brain cells are considered as a fundamental mediator of intercellular communication that can dynamically reflect the pathological state of the donor cells. Despite their involvement in age-related neurodegeneration, research on exosomes and their cargos in neurodegenerative diseases is still in its infancy [43] (Fig. 1).

Fig. 1
figure 1

Schematic representation of general characteristics of exosomes and their characterization via NTA. Differing surface markers in the context of different neurodegenerative diseases are also presented

Exosomes and their role in neurodegenerative diseases

Exosomes are tiny extracellular vesicles ranging from 30 to 100 nm in diameter, and are released by all cell types. These vesicles are formed from late endosomes via inward budding of multivesicular bodies (MVBs). Invagination of endosomal membranes results in the formation of intraluminal vesicles (ILVs) within large MVBs (Fig. 2). During this process, certain proteins may be trapped into the invaginating membrane, and cytosolic materials are also engulfed and enclosed into the ILVs. The majority of ILVs that are released into extracellular space via fusion with the plasma membrane are termed “exosomes”. A small proportion of MVBs function as “garbage trucks” by guiding their contents to lysosomes for degradation and removal. RAB7 and RAB2 are primary mediators that coordinate traffic between late endosomes and lysosomes. RAB GTPases are a family of small GTPases belonging to the RAS superfamily [44]. Also, MVBs sometimes do not fuse directly to lysosomes but via autophagosome-dependent fusion to form auto-phagolysosomes, which degrade the content they encapsulate. However, the detailed mechanism that sorts MVB to plasma membrane and lysosome is unclear, but there exists a decision point between the two fates, i.e., inhibition of one path will increase the other [45, 46]. Alternatively, the formation of ILVs requires the function of endosomal sorting complex required for transport (ESCRT), a complex protein machinery consisting of four separate ESCRTs (ESCRT-0, -I, -II, and -III) that cooperate to facilitate MVB formation, protein cargo sorting and vesicle budding [47,48,49]. Initially, exosomes were considered as a mere waste-disposing system. It is now widely accepted that they play a central role in intercellular communication [50, 51]. Exosomes serve as a vehicle for not only proteins but also DNA, mRNA, miRNA, and other non-coding RNAs (ncRNAs) and thus contribute to the modulation of gene expression within target cells. In addition, specific markers on the membrane of exosomes reflect their origin as well as differentiating them from other extracellular vesicles. CD63, CD81, CD9, ALIX, and TSG101 are generally considered specific markers for exosomes [52, 53]. Neural-derived exosomes can largely be detected by the presence of L1-cell adhesion molecule (L1CAM), which is a central nervous system-specific marker [54, 55]. However, quite recently, there have been some contradicting reports questioning the use of L1CAM as a neuronal specific marker [56] as it is shown to be present in other cell types like T and B cells and at higher levels in several cancer types [57].

Extracellular vesicles including exosomes can be isolated by several techniques such as differential ultracentrifugation, size exclusion chromatography, ultrafiltration, immunoaffinity capture, and polyethylene glycol-based precipitation. Efficient isolation of exosomes from different biofluids has been largely studied and nicely reviewed elsewhere [58,59,60,61,62], thus will not be discussed in detail here. Each of these isolation methods has its pros and cons as discussed in the cited studies above, but it is worth noting that despite recent technological advances in this field, it is still difficult to obtain pure and homogeneous exosomal preparations in sufficient quantities by any of the currently available methods or technologies. In recent years, several studies have compared performances of different methods and their combinations in the isolation of exosomes from various biofluids [63,64,65,66], and shown that exosome isolation from the same biomaterial by different methods may vary significantly in the yield, purity, and biochemical compositions of exosomes. The type of biological fluid can also affect the parameters. Furthermore, the exosomal pool itself is quite heterogenous [67], consisting of subpopulations that differ in size, morphology, surface markers, and biochemical compositions [68].

Another important class of molecules contained within exosomes are miRNAs, which are usually small, non-coding, and about 22 nucleotides in length. A miRNA can repress the translation or regulate the degradation of over 100 mRNAs and one mRNA may be regulated by multiple miRNAs. Thus, they form a powerful gene regulation network and are involved in key biological processes including cell signaling, apoptosis, neuronal development, and plasticity [69, 70]. miRNAs are enriched in exosomes compared to cell-free serum or plasma because they are relatively more stable within exosomes [71, 72]. These exosomal miRNAs may be useful diagnostic and prognostic biomarkers of diseases. Besides being highly variable across neurodegenerative diseases, exosomal miRNAs can be used to discriminate disease subtypes [73]. For instance, the clinical phenotype of multiple sclerosis can be accurately distinguished via expression of different miRNAs at different stages. Ebrahim khani et al. identified 9 miRNAs that can differentiate relapsing-remitting multiple sclerosis from secondary/primary progressive multiple sclerosis [74].

Furthermore, exosomes have been strongly linked to the pathogenesis and progression of many neurodegenerative diseases [75, 76]. In the case of PD, the exosomal biogenesis machinery has been implicated in α-syn accumulation [77], and both in vitro and in vivo studies have noted pathological propagation of Tau aggregates by exosomes [78, 79]. It has also been reported that exosomes provide an ideal environment for α-syn to aggregate and this exosomal form of oligomeric α-syn is more easily taken up by recipient cells compared with the free form, resulting in potential propagation of the PD pathology [80, 81] (Fig. 2).

Fig. 2
figure 2

Exosome biogenesis in neuronal cells and their role in cell-to-cell transmission of various “infectious proteins”. These “cargo-loaded vesicles” are also released in the circulatory system. α-syn: alpha-synuclein; NFT: neurofibrillary tangle; Aβ: amyloid beta; MVB: multivesicular body

Some exosomal miRNAs have also been reported as PD biomarkers. For instance, miR-19b, miR-24 and miR-195 can be used for PD diagnosis, based on the target scan tool. Cao et al. [82] related these miRNAs to pathological process in PD, such as miR-19b related to Parkin RBR E3 ubiquitin protein ligase, miR-19b to LRRK2/PARK8, as well as miR-24 and miR-195 to ATP13A2/PARK9. Another study reported a panel of 5 exosomal miRNAs from CSF, comprising Let-7f-5p, miR-27a-3p, miR-125a-5p, miR-151a-3p and miR-423-5p, which shows 90% sensitivity and 80% specificity for differentiation of PD from controls [83].

Another set of misfolded proteins including superoxide dismutase 1 and TDP-43, which have shown associations with ALS, have been identified in exosomes [84, 85]. Also, in the case of AD, exosomes extracted from the brains of AD patients show significantly elevated levels of amyloid precursor protein, Aβ oligomers, Aβ1−42, and p-S396-tau [86,87,88]. Additionally, significant differences in miR-9-5p and miR-598 are detected in exosomes from CSF of AD  vs control participants [89]. Another study has shown that the serum exosomal miR-135a and miR-384 are upregulated in AD patients when compared with healthy cohorts, and miR-384 could be used for discriminating between AD, vascular dementia and Parkinson’s disease with dementia [90].

Several studies have pointed out that exosomes can also transport the expanded polyglutamine tract of both Htt RNA and protein as well as the mutant huntingtin protein (mHtt) aggregates, and thus trigger HD-related behavioral and pathological features [90,91,92]. Htt is a large protein of 350 kDa, which makes it hard to be packed into exosomes. Therefore, mHtt packaging and spreading through exosomes, which is supported by recent evidence [93], is a complex pathway that remains to be defined.

FTD is considered as a second most common cause of dementia with an age of onset < 65. In FTD, the most common mutations are from three genes: granulin (GRN) [94], C9orf72 (chromosome 9 open reading frame 72) [95] and MAPT (microtubule associated tau) [96]. It has been documented that many proteins involved in FTD pathogenesis are secreted by cells in association with exosomes. Furthermore, mutations in GRN result in the reduction of exosomes and the alteration of their composition [97]. Neurofilament light chain has been considered as a biomarker for axonal injury and is reportedly increased in sera of FTD patients [98, 99]. Another report showed higher levels of exosomal heat shock protein-70 (HSP70) than free HSP70 in plasma in FTD and AD, and the exosomal HSP70 level correlated with 18F-FDG-PET [100].

Recently, the biomarker potential of salivary neuronal exosomes has been explored in neurodegenerative diseases. Saliva is a type of easily accessible biofluid and isolation of salivary exosomes is a non-invasive, painless, and relatively simple procedure when compared with blood and CSF sampling [101]. Some recent studies have shown increased levels of α-syn oligomers within salivary exosomes form PD patinets when compared with the healthy cohorts [102, 103]. The only reported study connecting salivary exosomes concentration to cognitive impairment (CI) and AD showed an increased concentration of salivary exosomes in CI and AD than in healthy controls. The αβ oligomer and p-tau show high protein abundance in salivary exosomes in CI and AD in comparison to control subjects [104]. Despite the limited number of studies on salivary neuronal exosomes to date, the saliva-derived exosomes hold promise for various clinical applications, including use in a non-invasive biomarker panel and for disease progression tracking [105].

On the other hand, some research findings indicate that the production of exosomes may be involved in the improvement of pathological phenotype of diseases like AD [106, 107] and PD [108, 109]. In summary, exosomes can be considered as a “double-edged sword” suggesting their neuropathologic and neuroprotective roles. On the one hand, exosomes are involved in the dysregulation of communication between neurons or between neurons and glial cells, which triggers neurodegenerative diseases. On the other side, there is evidence for the involvement of exosomes in sequestering neurotoxic molecules from neural cells and the transfer of neuroprotective ones [110, 111]. However, the detailed mechanisms of the switch between the two sides remain unknown.

Nanoparticle tracking analysis (NTA): an emerging platform for screening neurodegenerative diseases

NTA has emerged as a state-of-the-art method for the characterization of exosomes [112, 113]. This method combines two different physical principles, light scattering and Brownian motion (Fig. 3). First, particles in liquid suspension are irradiated by a laser beam and the 2D trajectory of each particle is tracked to calculate the diffusion coefficient. Second, a video of the displacement of each particle under Brownian motion is captured by camera (Fig. 3). The hydrodynamic radius for individual particles is calculated via the Stokes–Einstein equations [114, 115] as follows:

$$\overline{{\left( {x,y} \right)^{2} }} = \frac{{2kTt}}{{3R_{h} \eta }}$$

where k = Boltzmann constant, T = temperature, and η = solvent viscosity. The particle size is reported as the hydrodynamic radius, Rh, determined by following the 2D trajectory of each particle over a tracking time t.

The advantage of NTA is that individual particles are tracked, hence one can obtain both the concentration and the size distribution, rather than just a mean size. One disadvantage with NTA is that smaller particles are not easily measurable, as the minimum size for detection depends on the refractive index increment. In addition, particles larger than 1 μm diffuse slowly and thus are also difficult to measure with NTA [116]. A seemingly similar instrument called differential light scattering (DLS) also works on the scattering principle and measures particle diffusion. However, the DLS obtains the total contribution from all scattering particles, which is slightly biased towards larger-sized particles and thereby swamps out contributions from smaller particles, while in NTA, more accurate measurements can be obtained by its particle-by-particle approach [117]. Many initial studies with NTA were focused on the validation of NTA measurements of mono- and multi-modal nanoparticles and compared its performance with DLS [114, 118]. When analyzing polydisperse samples such as proteins and vesicles, NTA provides a much better resolution than DLS [119, 120].

Fig. 3
figure 3

Schematic setup of a nanoparticle tracking analyzer. The particles in a sample are illuminated with a laser beam. The scattered light is recorded by a CMOS camera fitting at 90˚ to the illumination plane, in a built-in ultramicroscope system. A fluorescence filter is placed between the cell holder and the microscope. Light scattered by the particles is displayed in the “live-view” window of the software. The size of each particle is calculated by Brownian motion analysis of the individual tracks, allowing for simultaneous determination of size and concentration

The aggregation of proteins is central to many neurodegenerative disorders. The NTA system has been used for the characterization of fibrillar protein aggregates including Aβ and polyglutamine peptides [121], and analysis of aggregation kinetics of α-syn [122]. The NTA system can also be used to monitor bioconjugation. For instance, gold nanoparticle binding to protein A and subsequent interactions with immunoglobulin G can be measured with NTA, using hydrodynamic radius as a function of measurement [123, 124]. A more recent study has shown the utility of statistical mixture distribution combined with NTA to quantitate the amount and extent of particle binding in a mixture of nanomaterials [125]. Furthermore, antibody- and fluorescence-based NTA methods have been used to study specific populations of exosomes and provide better insights. Both antibody- and fluorescence-based methods allow accurate sizing and phenotyping of various exosomes based on their respective surface markers [126,127,128]. With the NTA system, many studies have reported an increase of neuronally derived exosomes in various biofluids in neurodegenerative disorders. For instance, in PD, brain-derived exosomes in plasma were found to be significantly increased when compared with the age-matched healthy controls [129]. In another supportive study, increased DJ-1 and α-syn were found in neural-derived exosomes from PD patients [130]. In the saliva samples from PD [131, 132] and AD patients [133], neuronal salivary exosomes were found to be increased in a similar pattern. This approach could be extended to other neurodegenerative diseases as well [62]. Hence, combining the specificity of fluorescence and antibody-based exosomal quantifications through the NTA system could serve as a promising screening methodology for a variety of neurodegenerative diseases, depending on their respective surface markers.

Conclusion

Despite significant advances in clinical imaging technologies, there exist several unaddressed challenges in the accurate diagnosis of neurodegenerative diseases. As the molecular changes in these diseases begin as early as 10–20 years prior to the clinical manifestations, tracking molecular events could aid in the early diagnosis. In this regard, exosomes are considered as new “hotspots” since their role in the progression of various diseases is turning out to be quite critical. Through this article, we put forward a screening technology, which could be employed along with other already established subjective assessments and imaging methods for early diagnosis of neurodegenerative disorders. Fluorescence NTA has the potential to quantitate neuronally derived exosomes from diseased and healthy cohorts, based on their specific surface markers.

As of now, there are several studies elucidating concentration differences of exosomal particles between diseased and healthy volunteers. But cautiously, more studies with increased sample sizes are needed to validate this methodology and  make it a “mass-screening technology” in the future. Another necessity of particular interest is the follow-up of participants in such studies to gain more confidence in such a screening methodology. In the foreseeable future, more efforts should be made in similar areas to tackle the increasing burden of neurodegenerative diseases, in addition to drug-based efforts for curing these debilitating diseases.

Data availability

Not applicable.

Abbreviations

NDs:

Neurodegerative diseases

AD:

Alzheimer’s disease

PD:

Parkinson’s disease

FTD:

Frontotemporal dementia

HD:

Huntington’s disease

ALS:

Amyotrophic lateral sclerosis

Aβ:

Amyloid β

α-syn:

α-Synuclein

NFT:

Neurofibrillary tangle

LRRK2:

Leucine-rich repeat kinase 2

MCI:

Mild cognitive impairment

PET:

Positron emission tomography

L1CAM:

L1-cell adhesion molecule

TDP-43:

Transactive response DNA binding protein 43 kDa

NTA:

Nanoparticle tracking analysis

DLS:

Differential light scattering

FUS:

Fused in sarcoma

ESCRT:

Endosomal sorting complex required for transport

ILV:

Intraluminal vesicle

References

  1. D’Anca M, Fenoglio C, Serpente M, Arosio B, Cesari M, Scarpini EA, et al. Exosome determinants of physiological aging and age-related neurodegenerative diseases. Front Aging Neurosci. 2019;11:232.

    Article  Google Scholar 

  2. Frost B, Diamond MI. Prion-like mechanisms in neurodegenerative diseases. Nat Rev Neurosci. 2010;11(3):155–9.

    Article  CAS  Google Scholar 

  3. Hoover BM, Murphy RM. Evaluation of nanoparticle tracking analysis for the detection of rod-shaped particles and protein aggregates. J Pharm Sci. 2020;109(1):452–63.

    Article  CAS  Google Scholar 

  4. Jellinger KA. Interaction between pathogenic proteins in neurodegenerative disorders. J Cell Mol Med. 2012;16(6):1166–83.

    Article  CAS  Google Scholar 

  5. Busciglio J, Gabuzda DH, Matsudaira P, Yankner BA. Generation of beta-amyloid in the secretory pathway in neuronal and nonneuronal cells. Proc Natl Acad Sci USA. 1993;90(5):2092–6.

    Article  CAS  Google Scholar 

  6. Yoon SS, Jo SA. Mechanisms of amyloid-β peptide clearance: potential therapeutic targets for Alzheimer’s disease. Biomol Ther (Seoul). 2012;20(3):245–55.

    Article  CAS  Google Scholar 

  7. Selkoe DJ, Hardy J. The amyloid hypothesis of Alzheimer’s disease at 25 years. EMBO Mol Med. 2016;8(6):595–608.

    Article  CAS  Google Scholar 

  8. Molinuevo JL, Ayton S, Batrla R, Bednar MM, Bittner T, Cummings J, et al. Current state of Alzheimer’s fluid biomarkers. Acta Neuropathol. 2018;136(6):821–53.

    Article  CAS  Google Scholar 

  9. Manne S, Kondru N, Jin H, Serrano GE, Anantharam V, Kanthasamy A, et al. Blinded RT-QuIC analysis of α-synuclein biomarker in skin tissue from Parkinson’s disease patients. Mov Disord. 2020;35(12):2230–9.

    Article  CAS  Google Scholar 

  10. Thomas B, Beal MF. Parkinson’s disease. Hum Mol Genet. 2007;16(R2):R183–94.

    Article  CAS  Google Scholar 

  11. Spillantini MG, Goedert M. Neurodegeneration and the ordered assembly of α-synuclein. Cell Tissue Res. 2018;373(1):137–48.

    Article  CAS  Google Scholar 

  12. Kramer ML, Schulz-Schaeffer WJ. Presynaptic alpha-synuclein aggregates, not Lewy bodies, cause neurodegeneration in dementia with Lewy bodies. J Neurosci. 2007;27(6):1405–10.

    Article  CAS  Google Scholar 

  13. Choudhury SP, Bano S, Sen S, Suchal K, Kumar S, Nikolajeff F, et al. Altered neural cell junctions and ion-channels leading to disrupted neuron communication in Parkinson’s disease. NPJ Parkinson Dis. 2022;8(1):66.

    Article  Google Scholar 

  14. Mehta SH, Adler CH. Advances in biomarker research in Parkinson’s disease. Curr Neurol Neurosci Rep. 2015;16(1):7.

    Article  Google Scholar 

  15. Berg D, Postuma RB, Adler CH, Bloem BR, Chan P, Dubois B, et al. MDS research criteria for prodromal Parkinson’s disease. Mov Disord. 2015;30(12):1600–11.

    Article  Google Scholar 

  16. Postuma RB, Berg D, Stern M, Poewe W, Olanow CW, Oertel W, et al. MDS clinical diagnostic criteria for Parkinson’s disease. Mov Disord. 2015;30(12):1591–601.

    Article  Google Scholar 

  17. Reisberg B, Ferris SH, de Leon MJ, Crook T. The global deterioration scale for assessment of primary degenerative dementia. Am J Psychiatry. 1982.

  18. Rasmussen J, Langerman H. Alzheimer’s disease–why we need early diagnosis. Degener Neurol Neuromuscul Dis. 2019;9:123.

    Google Scholar 

  19. Mueller SG, Weiner MW, Thal LJ, Petersen RC, Jack CR, Jagust W, et al. Ways toward an early diagnosis in Alzheimer’s disease: the Alzheimer’s disease neuroimaging initiative (ADNI). Alzheimers Dement. 2005;1(1):55–66.

    Article  Google Scholar 

  20. Ottoy J, Niemantsverdriet E, Verhaeghe J, De Roeck E, Struyfs H, Somers C, et al. Association of short-term cognitive decline and MCI-to-AD dementia conversion with CSF, MRI, amyloid- and 18F-FDG-PET imaging. NeuroImage. 2019;22:101771.

    Article  Google Scholar 

  21. McAlary L, Plotkin SS, Yerbury JJ, Cashman NR. Prion-like propagation of protein misfolding and aggregation in amyotrophic lateral sclerosis. Front Mol Neurosci. 2019;12:262.

    Article  CAS  Google Scholar 

  22. Hardiman O, Al-Chalabi A, Chio A, Corr EM, Logroscino G, Robberecht W, et al. Amyotrophic lateral sclerosis. Nat Rev Dis Primers. 2017;3(1):1–19.

    Google Scholar 

  23. Taylor JP, Brown RH, Cleveland DW. Decoding ALS: from genes to mechanism. Nature. 2016;539(7628):197–206.

    Article  Google Scholar 

  24. Ling S-C, Polymenidou M, Cleveland DW. Converging mechanisms in ALS and FTD: disrupted RNA and protein homeostasis. Neuron. 2013;79(3):416–38.

    Article  CAS  Google Scholar 

  25. Arrasate M, Finkbeiner S. Protein aggregates in Huntington’s disease. Exp Neurol. 2012;238(1):1–11.

    Article  CAS  Google Scholar 

  26. Kirkin V, McEwan DG, Novak I, Dikic I. A role for ubiquitin in selective autophagy. Mol Cell. 2009;34(3):259–69.

    Article  CAS  Google Scholar 

  27. Fukata Y, Fukata M. Protein palmitoylation in neuronal development and synaptic plasticity. Nat Rev Neurosci. 2010;11(3):161–75.

    Article  CAS  Google Scholar 

  28. Chételat G, Arbizu J, Barthel H, Garibotto V, Law I, Morbelli S, et al. Amyloid-PET and 18F-FDG-PET in the diagnostic investigation of Alzheimer’s disease and other dementias. Lancet Neurol. 2020;19(11):951–62.

    Article  Google Scholar 

  29. Smailagic N, Vacante M, Hyde C, Martin S, Ukoumunne O, Sachpekidis C. 18F-FDG PET for the early diagnosis of Alzheimer’s disease dementia and other dementias in people with mild cognitive impairment (MCI). Cochrane Database Syst Rev. 2015;1(1):Cd010632.

    Google Scholar 

  30. Walker Z, Gandolfo F, Orini S, Garibotto V, Agosta F, Arbizu J, et al. Clinical utility of FDG PET in Parkinson’s disease and atypical parkinsonism associated with dementia. Eur J Nucl Med Mol Image. 2018;45(9):1534–45.

    Article  CAS  Google Scholar 

  31. Gao A. Identification of blood-based biomarkers for early stage Parkinson’s disease. medRxiv. 2020:2020.10.22.20217893.

  32. Posavi M, Diaz-Ortiz M, Liu B, Swanson CR, Skrinak RT, Hernandez-Con P, et al. Characterization of Parkinson’s disease using blood-based biomarkers: a multicohort proteomic analysis. PLoS Med. 2019;16(10):e1002931.

    Article  CAS  Google Scholar 

  33. Chahine LM, Stern MB, Chen-Plotkin A. Blood-based biomarkers for Parkinson’s disease. Parkinsonism Relat Disord. 2014;20:99–S103.

    Article  Google Scholar 

  34. Yao F, Hong X, Li S, Zhang Y, Zhao Q, Du W, et al. Urine-based biomarkers for Alzheimer’s disease identified through coupling computational and experimental methods. J Alzheimers Dis. 2018;65(2):421–31.

    Article  CAS  Google Scholar 

  35. Seol W, Kim H, Son I. Urinary biomarkers for neurodegenerative diseases. Exp Neurobiol. 2020;29(5):325–33.

    Article  Google Scholar 

  36. Obrocki P, Khatun A, Ness D, Senkevich K, Hanrieder J, Capraro F, et al. Perspectives in fluid biomarkers in neurodegeneration from the 2019 biomarkers in neurodegenerative diseases course—a joint PhD student course at University College London and University of Gothenburg. Alzheimers Res Ther. 2020;12(1):20.

    Article  Google Scholar 

  37. Robey TT, Panegyres PK. Cerebrospinal fluid biomarkers in neurodegenerative disorders. Future Neurol. 2019;14(1):FNL6.

    Article  CAS  Google Scholar 

  38. Seino Y, Nakamura T, Kawarabayashi T, Hirohata M, Narita S, Wakasaya Y, et al. Cerebrospinal fluid and plasma biomarkers in neurodegenerative diseases. J Alzheimers Dis. 2019;68(1):395–404.

    Article  CAS  Google Scholar 

  39. Maciejczyk M, Zalewska A, Gerreth. Karolina. Salivary redox biomarkers in selected neurodegenerative diseases. J Clin Med. 2020;9(2):497.

    Article  CAS  Google Scholar 

  40. Pawlik P, Błochowiak K. The role of salivary biomarkers in the early diagnosis of Alzheimer’s disease and Parkinson’s disease. Diagnostics (Basel). 2021;11(2):371.

    Article  CAS  Google Scholar 

  41. Bermejo-Pareja F, del Ser T, Valentí M, de la Fuente M, Bartolome F, Carro E. Salivary lactoferrin as biomarker for Alzheimer’s disease: brain-immunity interactions. Alzheimers Dement. 2020;16(8):1196–204.

    Article  Google Scholar 

  42. Stott S, Broza YY, Gharra A, Wang Z, Barker RA, Haick H. The utility of breath analysis in the diagnosis and staging of Parkinson’s disease. J Parkinsons Dis. 2022;12(3):993–1002.

    Article  CAS  Google Scholar 

  43. D’Anca M, Fenoglio C, Serpente M, Arosio B, Cesari M, Scarpini EA, et al. Exosome determinants of physiological aging and age-related neurodegenerative diseases. Front Aging Neurosci. 2019;11:232.

    Article  Google Scholar 

  44. Xu M, Ji J, Jin D, Wu Y, Wu T, Lin R, et al. The biogenesis and secretion of exosomes and multivesicular bodies (MVBs): intercellular shuttles and implications in human diseases. Genes Dis. 2022. https://doi.org/10.1016/j.gendis.2022.03.021.

    Article  Google Scholar 

  45. Fader CM, Sánchez D, Furlán M, Colombo MI. Induction of autophagy promotes fusion of multivesicular bodies with autophagic vacuoles in k562 cells. Traffic. 2008;9(2):230–50.

    Article  CAS  Google Scholar 

  46. Eitan E, Suire C, Zhang S, Mattson MP. Impact of lysosome status on extracellular vesicle content and release. Ageing Res Rev. 2016;32:65–74.

    Article  CAS  Google Scholar 

  47. Sahu R, Kaushik S, Clement CC, Cannizzo ES, Scharf B, Follenzi A, et al. Microautophagy of cytosolic proteins by late endosomes. Dev Cell. 2011;20(1):131–9.

    Article  CAS  Google Scholar 

  48. Hurley JH. ESCRT s are everywhere. EMBO J. 2015;34(19):2398–407.

    Article  CAS  Google Scholar 

  49. Henne WM, Buchkovich NJ, Emr SD. The ESCRT pathway. Dev Cell. 2011;21(1):77–91.

    Article  CAS  Google Scholar 

  50. Zhang G, Yang P. A novel cell-cell communication mechanism in the nervous system: exosomes. J Neurosci Res. 2018;96(1):45–52.

    Article  CAS  Google Scholar 

  51. Rashed H, Bayraktar M, K Helal E, Abd-Ellah G, Amero MF, Chavez-Reyes P. Exosomes: from garbage bins to promising therapeutic targets. Int J Mol Sci. 2017;18(3):538.

    Article  Google Scholar 

  52. Deng F, Miller J. A review on protein markers of exosome from different bio-resources and the antibodies used for characterization. J Histotechnol. 2019;42(4):226–39.

    Article  CAS  Google Scholar 

  53. 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. Proc Natl Acad Sci USA. 2016;113(8):E968–77.

    Article  CAS  Google Scholar 

  54. Kenwrick S, Watkins A, Angelis ED. Neural cell recognition molecule L1: relating biological complexity to human disease mutations. Hum Mol Genet. 2000;9(6):879–86.

    Article  CAS  Google Scholar 

  55. Fauré J, Lachenal G, Court M, Hirrlinger J, Chatellard-Causse C, Blot B, et al. Exosomes are released by cultured cortical neurones. Mol Cell Neurosci. 2006;31(4):642–8.

    Article  Google Scholar 

  56. Norman M, Ter-Ovanesyan D, Trieu W, Lazarovits R, Kowal EJK, Lee JH, et al. L1CAM is not associated with extracellular vesicles in human cerebrospinal fluid or plasma. Nat Methods. 2021;18(6):631–4.

    Article  CAS  Google Scholar 

  57. Gomes DE, Witwer KW. L1CAM-associated extracellular vesicles: a systematic review of nomenclature, sources, separation, and characterization. J Extracell Biol. 2022;1(3):e35.

    Article  Google Scholar 

  58. Sidhom K, Obi PO, Saleem A. A review of exosomal isolation methods: is size exclusion chromatography the best option? Int J Mol Sci. 2020;21:18.

    Article  Google Scholar 

  59. Chen J, Li P, Zhang T, Xu Z, Huang X, Wang R, et al. Review on strategies and technologies for exosome isolation and purification. Front Bioeng Biotechnol. 2022;9:811971.

    Article  Google Scholar 

  60. Zhou M, Weber SR, Zhao Y, Chen H, Sundstrom JM. In: Edelstein L, Smythies J, Quesenberry P, Noble D, editors. Chapter 2-methods for exosome isolation and characterization. Exosomes: Academic Press; 2020. p. 23–38.

    Google Scholar 

  61. Mustafa K, Gülşah Erel A. Exosome isolation: is there an optimal method with regard to diagnosis or treatment? In: Jin W, editor. Novel implications of exosomes in diagnosis and treatment of cancer and infectious diseases. Rijeka: IntechOpen; 2017. Ch. 8.

    Google Scholar 

  62. Younas N, Fernandez Flores LC, Hopfner F, Höglinger GU, Zerr I. A new paradigm for diagnosis of neurodegenerative diseases: peripheral exosomes of brain origin. Transl Neurodegener. 2022;11(1):28.

    Article  Google Scholar 

  63. Brennan K, Martin K, FitzGerald S, O’sullivan J, Wu Y, Blanco A, et al. A comparison of methods for the isolation and separation of extracellular vesicles from protein and lipid particles in human serum. Sci Rep. 2020;10(1):1–13.

    Article  Google Scholar 

  64. Soares Martins T, Catita J, Martins Rosa I, Henriques AG. Exosome isolation from distinct biofluids using precipitation and column-based approaches. PLoS ONE. 2018;13:e0198820.

    Article  Google Scholar 

  65. Buschmann D, Kirchner B, Hermann S, Märte M, Wurmser C, Brandes F, et al. Evaluation of serum extracellular vesicle isolation methods for profiling miRNAs by next-generation sequencing. J Extracell Vesicles. 2018;7(1):1481321.

    Article  Google Scholar 

  66. Langevin SM, Kuhnell D, Biesiada J, Zhang X, Medvedovic M, Talaska GG, et al. Comparability of the small RNA secretome across human biofluids concomitantly collected from healthy adults. PLoS ONE. 2020;15(4):e0229976.

    Article  CAS  Google Scholar 

  67. Zhang H, Freitas D, Kim HS, Fabijanic K, Li Z, Chen H, et al. Identification of distinct nanoparticles and subsets of extracellular vesicles by asymmetric flow field-flow fractionation. Nat Cell Biol. 2018;20(3):332–43.

    Article  CAS  Google Scholar 

  68. Yakubovich EI, Polischouk AG, Evtushenko VI. Principles and problems of exosome isolation from biological fluids. Biochem Mosc Suppl Ser A Membr Cell Biol. 2022;16(2):115–26.

    Article  CAS  Google Scholar 

  69. Kim VN, Han J, Siomi MC. Biogenesis of small RNAs in animals. Nat Rev Mol Cell Biol. 2009;10(2):126–39.

    Article  CAS  Google Scholar 

  70. Ambros V. The functions of animal microRNAs. Nature. 2004;431(7006):350–5.

    Article  CAS  Google Scholar 

  71. Cheng L, Sharples RA, Scicluna BJ, Hill AF. Exosomes provide a protective and enriched source of miRNA for biomarker profiling compared to intracellular and cell-free blood. J Extracell Vesicles. 2014;3:23743.

    Article  Google Scholar 

  72. Hu G, Drescher KM, Chen XM. Exosomal miRNAs: biological properties and therapeutic potential. Front Genet. 2012;3:56.

    Article  CAS  Google Scholar 

  73. Sheinerman KS, Toledo JB, Tsivinsky VG, Irwin D, Grossman M, Weintraub D, et al. Circulating brain-enriched microRNAs as novel biomarkers for detection and differentiation of neurodegenerative diseases. Alzheimers Res Ther. 2017;9(1):89.

    Article  Google Scholar 

  74. Ebrahimkhani S, Vafaee F, Young PE, Hur SSJ, Hawke S, Devenney E, et al. Exosomal microRNA signatures in multiple sclerosis reflect disease status. Sci Rep. 2017;7(1):14293.

    Article  Google Scholar 

  75. Soria FN, Pampliega O, Bourdenx M, Meissner WG, Bezard E, Dehay B. Exosomes, an unmasked culprit in neurodegenerative diseases. Front Neurosci. 2017;11:26.

    Article  Google Scholar 

  76. Rastogi S, Sharma V, Bharti PS, Rani K, Modi GP, Nikolajeff F, et al. The evolving landscape of exosomes in neurodegenerative diseases: exosomes characteristics and a promising role in early diagnosis. Int J Mol Sci. 2021;22(1):440.

    Article  CAS  Google Scholar 

  77. Spencer B, Kim C, Gonzalez T, Bisquertt A, Patrick C, Rockenstein E, et al. α-Synuclein interferes with the ESCRT-III complex contributing to the pathogenesis of Lewy body disease. Hum Mol Genet. 2016;25(6):1100–15.

    Article  CAS  Google Scholar 

  78. Asai H, Ikezu S, Tsunoda S, Medalla M, Luebke J, Haydar T, et al. Depletion of microglia and inhibition of exosome synthesis halt tau propagation. Nat Neurosci. 2015;18(11):1584–93.

    Article  CAS  Google Scholar 

  79. Baker S, Polanco JC, Götz J. Extracellular vesicles containing P301L mutant tau accelerate pathological tau phosphorylation and oligomer formation but do not seed mature neurofibrillary tangles in ALZ17 mice. J Alzheimers Dis. 2016;54(3):1207–17.

    Article  CAS  Google Scholar 

  80. Grey M, Dunning CJ, Gaspar R, Grey C, Brundin P, Sparr E, et al. Acceleration of α-synuclein aggregation by exosomes. J Biol Chem. 2015;290(5):2969–82.

    Article  CAS  Google Scholar 

  81. Danzer KM, Kranich LR, Ruf WP, Cagsal-Getkin O, Winslow AR, Zhu L, et al. Exosomal cell-to-cell transmission of alpha synuclein oligomers. Mol Neurodegener. 2012;7(1):1–18.

    Article  Google Scholar 

  82. Leggio L, Vivarelli S, L’Episcopo F, Tirolo C, Caniglia S, Testa N, et al. microRNAs in Parkinson’s disease: from pathogenesis to novel diagnostic and therapeutic approaches. Int J Mol Sci. 2017;18(12):2698.

    Article  Google Scholar 

  83. Dos Santos MCT, Barreto-Sanz MA, Correia BRS, Bell R, Widnall C, Perez LT, et al. miRNA-based signatures in cerebrospinal fluid as potential diagnostic tools for early stage Parkinson’s disease. Oncotarget. 2018;9(25):17455–65.

    Article  Google Scholar 

  84. Grad LI, Yerbury JJ, Turner BJ, Guest WC, Pokrishevsky E, O’Neill MA, et al. Intercellular propagated misfolding of wild-type Cu/Zn superoxide dismutase occurs via exosome-dependent and-independent mechanisms. Proc Natl Acad Sci U S A. 2014;111(9):3620–5.

    Article  CAS  Google Scholar 

  85. Iguchi Y, Eid L, Parent M, Soucy G, Bareil C, Riku Y, et al. Exosome secretion is a key pathway for clearance of pathological TDP-43. Brain. 2016;139(12):3187–201.

    Article  Google Scholar 

  86. Gallart-Palau X, Guo X, Serra A, Sze SK. Alzheimer’s disease progression characterized by alterations in the molecular profiles and biogenesis of brain extracellular vesicles. Alzheimers Res Ther. 2020;12:1–15.

    Article  Google Scholar 

  87. Muraoka S, DeLeo AM, Sethi MK, Yukawa-Takamatsu K, Yang Z, Ko J, et al. Proteomic and biological profiling of extracellular vesicles from Alzheimer’s disease human brain tissues. Alzheimers Dement. 2020;16(6):896–907.

    Article  Google Scholar 

  88. Zhang T, Ma S, Lv J, Wang X, Afewerky HK, Li H, et al. The emerging role of exosomes in Alzheimer’s disease. Ageing Res Rev. 2021;68:101321.

    Article  CAS  Google Scholar 

  89. Riancho J, Vázquez-Higuera JL, Pozueta A, Lage C, Kazimierczak M, Bravo M, et al. MicroRNA profile in patients with Alzheimer’s disease: analysis of mir-9-5p and miR-598 in raw and exosome enriched cerebrospinal fluid samples. J Alzheimers Dis. 2017;57(2):483–91.

    Article  CAS  Google Scholar 

  90. Yang TT, Liu CG, Gao SC, Zhang Y, Wang PC. The serum exosome derived microRNA-135a, -193b, and -384 were potential Alzheimer’s disease biomarkers. Biomed Environ Sci. 2018;31(2):87–96.

    CAS  Google Scholar 

  91. Jeon I, Cicchetti F, Cisbani G, Lee S, Li E, Bae J, et al. Human-to-mouse prion-like propagation of mutant huntingtin protein. Acta Neuropathol. 2016;132(4):577–92.

    Article  CAS  Google Scholar 

  92. Zhang X, Abels ER, Redzic JS, Margulis J, Finkbeiner S, Breakefield XO. Potential transfer of polyglutamine and CAG-repeat RNA in extracellular vesicles in Huntington’s disease: background and evaluation in cell culture. Cell Mol Neurobiol. 2016;36(3):459–70.

    Article  Google Scholar 

  93. Diaz-Hidalgo L, Altuntas S, Rossin F, D’Eletto M, Marsella C, Farrace MG, et al. Transglutaminase type 2-dependent selective recruitment of proteins into exosomes under stressful cellular conditions. Biochim Biophys Acta. 2016;1863(8):2084–92.

    Article  CAS  Google Scholar 

  94. Baker M, Mackenzie IR, Pickering-Brown SM, Gass J, Rademakers R, Lindholm C, et al. Mutations in progranulin cause tau-negative frontotemporal dementia linked to chromosome 17. Nature. 2006;442(7105):916–9.

    Article  CAS  Google Scholar 

  95. DeJesus-Hernandez M, Mackenzie IR, Boeve BF, Boxer AL, Baker M, Rutherford NJ, et al. Expanded GGGGCC hexanucleotide repeat in noncoding region of C9ORF72 causes chromosome 9p-linked FTD and ALS. Neuron. 2011;72(2):245–56.

    Article  CAS  Google Scholar 

  96. Hutton M, Lendon CL, Rizzu P, Baker M, Froelich S, Houlden H, et al. Association of missense and 5′-splice-site mutations in tau with the inherited dementia FTDP-17. Nature. 1998;393(6686):702–5.

    Article  CAS  Google Scholar 

  97. Benussi L, Ciani M, Tonoli E, Morbin M, Palamara L, Albani D, et al. Loss of exosomes in progranulin-associated frontotemporal dementia. Neurobiol Aging. 2016;40:41–9.

    Article  CAS  Google Scholar 

  98. van der Ende EL, Meeter LH, Poos JM, Panman JL, Jiskoot LC, Dopper EG, et al. Serum neurofilament light chain in genetic frontotemporal dementia: a longitudinal, multicentre cohort study. Lancet Neurol. 2019;18(12):1103–11.

    Article  Google Scholar 

  99. Benussi A, Karikari TK, Ashton N, Gazzina S, Premi E, Benussi L, et al. Diagnostic and prognostic value of serum NfL and p-Tau181 in frontotemporal lobar degeneration. J Neurol Neurosurg Psychiatry. 2020;91(9):960–7.

    Article  Google Scholar 

  100. Chanteloup G, Cordonnier M, Moreno-Ramos T, Pytel V, Matías-Guiu J, Gobbo J, et al. Exosomal HSP70 for monitoring of frontotemporal dementia and Alzheimer’s disease: clinical and FDG-PET correlation. J Alzheimers Dis. 2019;71(4):1263–9.

    Article  CAS  Google Scholar 

  101. Zlotogorski-Hurvitz A, Dayan D, Chaushu G, Korvala J, Salo T, Sormunen R, et al. Human saliva-derived exosomes: comparing methods of isolation. J Histochem Cytochem. 2015;63(3):181–9.

    Article  CAS  Google Scholar 

  102. Cao Z, Wu Y, Liu G, Jiang Y, Wang X, Wang Z, et al. α-Synuclein in salivary extracellular vesicles as a potential biomarker of Parkinson’s disease. Neurosci Lett. 2019;696:114–20.

    Article  CAS  Google Scholar 

  103. Rani K, Mukherjee R, Singh E, Kumar S, Sharma V, Vishwakarma P, et al. Neuronal exosomes in saliva of Parkinson’s disease patients: a pilot study. Parkinsonism Relat Disord. 2019;67:21–3.

    Article  Google Scholar 

  104. Rani K, Rastogi S, Vishwakarma P, Bharti PS, Sharma V, Renu K, et al. A novel approach to correlate the salivary exosomes and their protein cargo in the progression of cognitive impairment into Alzheimer’s disease. J Neurosci Methods. 2021;347:108980.

    Article  CAS  Google Scholar 

  105. Younas N, Fernandez Flores LC, Hopfner F, Höglinger GU, Zerr I. A new paradigm for diagnosis of neurodegenerative diseases: peripheral exosomes of brain origin. Transl Neurodegen. 2022;11(1):28.

    Article  Google Scholar 

  106. Pacheco-Quinto J, Clausen D, Pérez-González R, Peng H, Meszaros A, Eckman CB, et al. Intracellular metalloprotease activity controls intraneuronal Aβ aggregation and limits secretion of Aβ via exosomes. FASEB J. 2019;33(3):3758–71.

    Article  CAS  Google Scholar 

  107. Glebov K, Walter J. Statins in unconventional secretion of insulin-degrading enzyme and degradation of the amyloid-β peptide. Neurodegener Dis. 2012;10(1–4):309–12.

    Article  CAS  Google Scholar 

  108. Sun T, Ding ZX, Luo X, Liu QS, Cheng Y. Blood exosomes have neuroprotective effects in a mouse model of Parkinson’s disease. Oxid Med Cell Longev. 2020;2020:3807476.

    Article  Google Scholar 

  109. Huo L, Du X, Li X, Liu S, Xu Y. The emerging role of neural cell-derived exosomes in intercellular communication in health and neurodegenerative diseases. Front Neurosci. 2021;15(1119):738442.

    Article  Google Scholar 

  110. Longoni B, Fasciani I, Kolachalam S, Pietrantoni I, Marampon F, Petragnano F, et al. Neurotoxic and neuroprotective role of exosomes in Parkinson’s disease. Curr Pharm Des. 2019;25(42):4510–22.

    Article  CAS  Google Scholar 

  111. Kalani A, Tyagi A, Tyagi N. Exosomes: mediators of neurodegeneration, neuroprotection and therapeutics. Mol Neurobiol. 2014;49(1):590–600.

    Article  CAS  Google Scholar 

  112. Bachurski D, Schuldner M, Nguyen PH, Malz A, Reiners KS, Grenzi PC, et al. Extracellular vesicle measurements with nanoparticle tracking analysis—an accuracy and repeatability comparison between NanoSight NS300 and ZetaView. J Extracell Vesicles. 2019;8(1):1596016.

    Article  CAS  Google Scholar 

  113. Comfort N, Cai K, Bloomquist TR, Strait MD, Ferrante AW Jr, Baccarelli AA. Nanoparticle tracking analysis for the quantification and size determination of extracellular vesicles. J Vis Exp. 2021;169:e62447.

    Google Scholar 

  114. Malloy A, Carr B. Nano particle tracking analysis–the halo™ system. Part Part Syst Charact. 2006;23(2):197–204.

    Article  Google Scholar 

  115. Gallego-Urrea JA, Tuoriniemi J, Hassellöv M. Applications of particle-tracking analysis to the determination of size distributions and concentrations of nanoparticles in environmental, biological and food samples. Trends Analyt Chem. 2011;30(3):473–83.

    Article  CAS  Google Scholar 

  116. Yang DT, Lu X, Fan Y, Murphy RM. Evaluation of nanoparticle tracking for characterization of fibrillar protein aggregates. AIChE J. 2014;60(4):1236–44.

    Article  CAS  Google Scholar 

  117. Lu X, Murphy RM. Nanoparticle tracking for protein aggregation research. Methods Mol Biol. 2018;1777:145–58.

    Article  CAS  Google Scholar 

  118. Bell NC, Minelli C, Tompkins J, Stevens MM, Shard AG. Emerging techniques for submicrometer particle sizing applied to Stober silica. Langmuir. 2012;28(29):10860–72.

    Article  CAS  Google Scholar 

  119. Gross J, Sayle S, Karow AR, Bakowsky U, Garidel P. Nanoparticle tracking analysis of particle size and concentration detection in suspensions of polymer and protein samples: influence of experimental and data evaluation parameters. Eur J Pharm Biopharm. 2016;104:30–41.

    Article  CAS  Google Scholar 

  120. Dragovic RA, Gardiner C, Brooks AS, Tannetta DS, Ferguson DJ, Hole P, et al. Sizing and phenotyping of cellular vesicles using nanoparticle tracking analysis. Nanomedicine. 2011;7(6):780–8.

    Article  CAS  Google Scholar 

  121. Yang DT, Lu X, Fan Y, Murphy RM. Evaluation of nanoparticle tracking for characterization of fibrillar protein aggregates. AIChE J. 2014;60(4):1236–44.

    Article  CAS  Google Scholar 

  122. Hoover BM, Murphy RM. Evaluation of nanoparticle tracking analysis for the detection of rod-shaped particles and protein aggregates. J Pharm Sci. 2020;109(1):452–63.

    Article  CAS  Google Scholar 

  123. James AE, Driskell JD. Monitoring gold nanoparticle conjugation and analysis of biomolecular binding with nanoparticle tracking analysis (NTA) and dynamic light scattering (DLS). Analyst. 2013;138(4):1212–8.

    Article  CAS  Google Scholar 

  124. Navarro Sanchez ME, Soulet D, Bonnet E, Guinchard F, Marco S, Vetter E, et al. Rabies vaccine characterization by nanoparticle tracking analysis. Sci Rep. 2020;10(1):8149.

    Article  CAS  Google Scholar 

  125. Foreman-Ortiz IU, Ma TF, Hoover BM, Wu M, Murphy CJ, Murphy RM, et al. Nanoparticle tracking analysis and statistical mixture distribution analysis to quantify nanoparticle–vesicle binding. J Colloid Interface Sci. 2022;615:50–8.

    Article  CAS  Google Scholar 

  126. Carnell-Morris P, Tannetta D, Siupa A, Hole P, Dragovic R. Analysis of extracellular vesicles using fluorescence nanoparticle tracking analysis. Methods Mol Biol. 2017;1660:153–73.

    Article  CAS  Google Scholar 

  127. Thane KE, Davis AM, Hoffman AM. Improved methods for fluorescent labeling and detection of single extracellular vesicles using nanoparticle tracking analysis. Sci Rep. 2019;9(1):12295.

    Article  Google Scholar 

  128. Andreu Z, Yáñez-Mó M. Tetraspanins in extracellular vesicle formation and function. Front Immunol. 2014;5:442.

    Article  Google Scholar 

  129. Ohmichi T, Mitsuhashi M, Tatebe H, Kasai T, Ali El-Agnaf OM, Tokuda T. Quantification of brain-derived extracellular vesicles in plasma as a biomarker to diagnose Parkinson’s and related diseases. Parkinsonism Relat Disord. 2019;61:82–7.

    Article  Google Scholar 

  130. Zhao ZH, Chen ZT, Zhou RL, Zhang X, Ye QY, Wang YZ. Increased DJ-1 and α-synuclein in plasma neural-derived exosomes as potential markers for Parkinson’s disease. Front Aging Neurosci. 2018;10:438.

    Article  CAS  Google Scholar 

  131. Rani K, Mukherjee R, Singh E, Kumar S, Sharma V, Vishwakarma P, et al. Neuronal exosomes in saliva of Parkinson’s disease patients: a pilot study. Parkinsonism Relat Disord. 2019;67:21–3.

    Article  Google Scholar 

  132. Kumar S, Rastogi S, Rani K, Rai S, Singh R, Sharma V, et al. Fluorescence tagged salivary exosomes as a nano tool in early diagnosis of Parkinson’s disease. 16 May 2022, PREPRINT (Version 1) available at Research Square https://doi.org/10.21203/rs.3.rs-1621589/v1.

  133. Rani K, Rastogi S, Vishwakarma P, Bharti PS, Sharma V, Renu K, et al. A novel approach to correlate the salivary exosomes and their protein cargo in the progression of cognitive impairment into Alzheimer’s disease. J Neurosci Methods. 2021;347:108980.

    Article  CAS  Google Scholar 

Download references

Acknowledgements

The authors extend their thanks to various funding agencies (ICMR, DHR, VINNOVA, Kempe foundation) for supporting the ongoing work at our laboratories at the Department of Biophysics, AIIMS, New Delhi, and Lulea University of Technology, Sweden. VS is thankful to Prahalad Bharti for his assistance in preparing figures. All authors are thankful to the reviewers for their insightful comments.

Funding

Open access funding provided by Lulea University of Technology.

Author information

Authors and Affiliations

Authors

Contributions

VS designed the review outline, performed literature search, wrote the manuscript, and designed the illustrations. SK and FN revised the manuscript. All authors made substantial contributions to the discussion of the content and reviewed and/or edited the article before submission. All authors read and approved the final manuscript.

Corresponding authors

Correspondence to Vaibhav Sharma or Saroj Kumar.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

All authors declare no competing interest.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Sharma, V., Nikolajeff, F. & Kumar, S. Employing nanoparticle tracking analysis of salivary neuronal exosomes for early detection of neurodegenerative diseases. Transl Neurodegener 12, 7 (2023). https://doi.org/10.1186/s40035-023-00339-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s40035-023-00339-z

Keywords