Abstract
Extracellular vesicles (EV) are nano-sized vesicles that have been garnering a lot of attention for their valuable role as potential diagnostic markers and therapeutic vehicles for a plethora of pathologies. Whilst EV markers from biofluids such as plasma, serum, urine, cerebrospinal fluid and in vitro cell culture based platforms have been extensively studied, a significant knowledge gap that remains is the characterization of specific organ derived EVs (ODE). Here, we present a standardized protocol for isolation and characterization of purified EV isolated from brain, heart, lung, kidney and liver from rat and postmortem human tissue. Next, using quantitative mass spectrometry based proteomics, we characterized the respective tissue EV proteomes that identified synaptophysin (SYP), caveolin-3 (CAV3), solute carrier family 22 member 2 (SLC22A2), surfactant protein B (SP-B), and fatty acid-binding protein 1 (FABP1) as potential markers for the brain, heart, kidney, lung, and liver-EV, respectively. These respective tissue specific markers were further validated using both immunoblotting and a nanoplasmonic platform- single EV imaging analysis in the two species. To summarize, our study for the first time using traditional biochemical and high precision technology platforms provide a valuable proof of concept approach in defining specific ODE markers which further could be developed as potential therapeutic candidates for respective end-organ associated pathologies.
Keywords: Extracellular Vesicles, Nanovesicles, Organ specific markers, Organ derived extracellular vesicles, nanoplasmonic, Quantitative proteomics, Single EV imaging
Graphical Abstract
INTRODUCTION
Extracellular vesicles (EV) are phospholipid nanovesicles that include (based on their origin or size), apoptotic bodies, microvesicles (MVs) and exosomes, and are garnering significant attention as molecules involved in cell-cell communication between cells and regulating the pathophysiology of several diseases.1-3 EV carry molecular cargo such as proteins, lipids, and RNA 4-6, and the molecular composition of the EV-cargo generally depends on their cells/tissue of origin. Furthermore, recent works demonstrating the role of EV as next-generation biomarkers based on their feasibility as therapeutic drug-delivery nanocarriers and in immunotherapy 7, 8 have made them exciting candidates for diagnostics and therapeutics in an array of diseases. 9-11 A recent study revealed the use of a quantum dot-based fluorescent immunoassay capable of distinguishing pancreatic cancer patients from healthy controls based on EV as biomarkers.12 Similarly, assays such as nanoscale Fluorescence Analysis and Cytometric Sorting (nanoFACS) techniques have been developed to detect tumor-specific antigens on EV.13 Nanoengineered ExoProfile chip had enabled simultaneous detection of up to eight biomarkers from one EV sample to enhance the distinction between early and late-stage cancer diagnosis.14 Furthermore, plasma-derived exosomes have been analyzed to provide insights into the microRNA cargo profiles in the diagnosis of disease such as Type-1 diabetes mellitus.15
Recently, several studies were performed to detect neuronal EV in plasma in an effort to identify and establish biomarkers for cognitive impairment in human immunodeficiency virus (HIV) infection, and neurodegenerative diseases.16, 17 In addition to plasma 18, 19, serum and cerebrospinal fluid-derived EV have also been studied to reveal potential diagnostic and prognostic biomarkers in glioma patients.20-23 Since body fluids (e.g., blood plasma, urine, cerebrospinal fluid) can be serially obtained at different disease stages, isolation of EV from them lend a distinct advantage of providing real-time monitoring of the progression of the illness 24 and thus making them attractive targets for diagnostic applications.25-27
Despite the many advantages of body fluids associated EV as potential biomarkers, a significant limitation is the identification of organ specific EV markers which can offer more depth in understanding the respective end-organs and associated pathologies. Most studies have been conducted on cell-lines or bio-fluids with little understanding of how organ-specific EV are expressed in situ. Although we and others have reported the feasibility of EV isolation from brain28-33 and liver tissues29, till date there is no existing literature on comparing EV from different organs including identification of definitive organ-specific EV markers. To address these challenges, the current study performed a comprehensive analysis of the respective tissue EV proteomes from the brain, heart, lung, kidney, and liver from both preclinical (rat) and clinical (postmortem human tissues). We identified synaptophysin (SYP), caveolin-3 (CAV3), solute carrier family 22 member 2 (SLC22A2), surfactant protein B (SP-B), and fatty acid-binding protein 1 (FABP1) as potential markers for the brain, heart, kidney, lung, and liver-EV, respectively. These respective tissue specific markers were further validated using both immunoblotting and a nanoplasmonic platform- single EV imaging analysis in the two species. To summarize, our study for the first time using traditional biochemical and high precision technology platforms provide a valuable proof of concept approach in defining specific ODE markers which further could be developed as potential therapeutic candidates for respective end-organ associated pathologies.
RESULTS AND DISCUSSION
Isolation and characterization of EV from body organs
One main goal of this study was to establish a standard protocol for isolation and characterization of EV isolated from different organ tissues. Figure 1 shows the scheme for isolating organ-derived EV (ODE), with minor modifications from previously published protocols.31, 32 Both transmission electron microscopy (TEM) and nanoparticle tracking analysis (NTA) revealed that EV purified from non-perfused rat organs displayed sizes between ~70-250 nm (Figure 2A). Interestingly, lung derived EV displayed characteristic membrane protrusions that resemble plasma membrane budding (Figure 2A). Next, we characterized four EV protein markers (Hsp-70, Flotillin-1, CD63, and CD81), and a negative marker (calnexin) by western blot on rat organ derived EV as well as in whole tissue lysates. Figure 3A shows the expression profile of all the EV makers validated in the different ODE and tissue lysates. Expression of Hsp-70 was observed in the brain, heart, lung, and liver but not in the kidney derived EV while Flotillin-1 (Flot-1) was detected in all except the lung derived EV. Flot-1 has been shown to be ubiquitously present in all EV types, however, it is less enriched in the small EV (sEV).34 Its absence from lung derived EV indicates that there may be a different biogenesis mechanism in lungs and could also explain its unique morphological characteristic. These markers were, however, detected in all the tissue lysates from the different organs. The tetraspanins CD63 and CD81 were detected in all ODE. Lastly, the negative maker calnexin was expressed only in the tissue lysates of the different organs. In summary, our western blot analysis showed positive protein bands for at least three classical EV markers for all organs (brain, heart, kidney, lung, and liver). Further, we ascertained if perfusion of animals with saline prior to harvesting tissues affects ODE isolations. Interestingly, we did not observe any differences in total EV protein yield when comparing perfused and non-perfused organs (see Table S1). TEM and western blot analysis revealed no major differences in size and EV marker expression between non-perfused and perfused ODE (Figures S1 & S2). Furthermore, western blot results also revealed similar EV marker profiles as seen with ODE isolated from non-perfused animals. This data corroborates with a recent work conducted on brain derived EV where the authors failed to see significant differences in total EV number from perfused and non-perfused brains.35 Presence of heterogenous EV sizes could reveal the presence of various EV subtypes in organs. Generally, the tetraspanin markers CD63 and CD81 are predominantly present in smaller sized EV subtype, exosomes, whereas Flot-1 and Hsp-70 has been shown to be present in other larger sized EV populations. 34, 36 The absence of Flot-1 and Hsp-70 markers from lung and kidney, respectively, indicates the absence of a subset of larger EV population. Overall, the data demonstrates the feasibility, rigor, and reproductivity of isolating ODE from different rat organs in non-perfused and perfused conditions.
Proteomic analysis of organ derived EV
It is well known that EV protein cargo can be used as biomarkers for detection of various diseases such as cancer 37, neurological 17, and cardiovascular diseases.38 To determine the composition of the EV proteome from the different organs, we performed comprehensive profiling of ODE using quantitative mass spectrometry based proteomics. Since we did not find major differences in size or markers between the perfused and non-perfused organ isolations, non-perfused rat organs were used for this proteomics study. We analyzed three biological replicates of ODE isolated from our rat organs. A protein was identified as differentially expressed if the FDR (Benjamini and Hochberg’s method) corrected P-value was ≤0.05, and log2 fold change was ≥1.5. As seen in Figure 3B, principal component analysis (PCA) revealed a distinguishable organ-specific clustering of ODE proteins. Overall, PCA analysis shows that the EV protein contents of various organs are significantly different from each other.
Next, we assessed the differential expression of proteins expressed in ODE isolated from different organs. Proteins identified with at least two unique peptides were included in the downstream differential analysis. Figure 4A (i) shows the number of upregulated proteins in the brain when compared with heart, kidney, lung, and liver EV proteins. We identified a total of 399 EV proteins that were upregulated in brain EV when compared to the rest of the organs analyzed. Of these, 147 had significantly higher expression across all comparisons (Figure 4A and Table S2). A similar analysis of heart, kidney, lung, and liver also identified several EV proteins that were highly abundant in one organ compared the rest analyzed in this study (Figure 4A (ii-v) and Table S2). Functional annotation using the bioinformatic tool, ClueGo, revealed that among the 147 differentially upregulated brain-EV proteins, a majority of these were involved in synaptic vesicle recycling, protein-amino acid transport, and neuronal synaptic plasticity (Figure 4B (i)). The majority of heart-EV proteins were involved with proton transport and oxidative phosphorylation pathways (Figure 4B (ii)). EV proteins from the kidney (7 groups), lung (8 groups), and liver (5 groups) were enriched with transmembrane transporter activity, regulation of actin filament polymerization, and PPAR signaling pathway, respectively (Figure 4B (iii-v)). Bar charts of the data are represented in Figure S3. It is widely accepted in the EV field that most of the cargo content expressed represents proteins or RNA carried in the host cells. It is interesting to note that protein cargos expressed in ODE from organs represent the major proteins (membrane proteins, receptors and transporters) expressed in that particular organ. For example, brain derived EV express largely synaptic membrane proteins and receptors (Figure 4B (i)). Similarly, kidney derived EV express mostly transporters which are abundantly expressed in kidneys (Figure 4B (iii)). In summary, the proteomic analysis of ODE revealed distinct organ specific protein signatures.
Identification and characterization of organ specific markers
Based on the bioinformatic analysis and normalized spectral count, we characterized the top 25 most abundant ODE proteins (Figure 5 and Table S3). Since high throughout approaches such as proteomics yield a lot of data, it is imperative to validate the identified potential hits. Accordingly, we performed post-validation of the respective ODE markers by western blot. In our quest to narrow our search for a good organ specific marker, we first compared the proteins abundant in different organs with the publicly available tissue enrichment dataset (https://v15.proteinatlas.org/tissue). Using this approach, we selected the following ODE markers for the respective organs. Brain- Synaptophysin (SYP), neural cell adhesion molecule 1 (NCAM1), excitatory amino acid transporter 1 (EAA1), and myelin oligodendrocyte glycoprotein (MOG)]; Heart-Caveolin-3 (CAV3) and cytochrome c oxidase subunit 5B (COX5B); Kidney: solute carrier family 22 member 2 (SLC22A2 and solute carrier family 12 member 1 (S12A1); Lung: surfactant protein B (SP-B), Intercellular adhesion molecule 1 (ICAM1), annexin A5 (ANXA5), and acidic mammalian chitinase (CHIA); and Liver: fatty acid-binding protein 1 (FABP1), glutathione S-transferase Mu 1 (GSTM1), microsomal glutathione S-transferase 1 (MGST1), and 3 beta-hydroxysteroid dehydrogenase type 7 (HSD3B7)] (Figures 6A(i-v) & S3 (A-E)). Following immunoblotting, we confirmed the validation of SYP, CAV3, SLC22A2, SP-B, and FABP1 as a potential brain, heart, kidney, lung, and liver-EV markers respectively from non-perfused, perfused ODE and whole tissue lysates (Figure 6B). The other markers identified showed either non-specificity or significant expression in more than one primary organ of interest and, therefore, could not meet the criteria for an organ-specific marker (Figure S4).
Although our results confirmed the presence of one predominant marker, we also found more than one significantly expressed EV protein on ODE. For example, brain given its complexity in structure and function is one of the most studied organs in EV biology. EV can readily cross the blood-brain barrier (BBB), making them attractive biomarkers for prognostic and diagnostic purposes for mental disorders. 39 Recent studies have shown some success in identifying neuron-specific EV marker, L1CAM, in plasma.40-44 In one recent study, it was shown that cleaved forms of L1CAM could be immunoprecipitated by biotin-labeled antibody from the brain and sera of HIV-1 transgenic (Tg) rats.42 They also show that the cleaved forms are enriched in the HIV-1 Tg rats when compared to wild type (WT). Whether the cleaved forms of L1CAM are found on the surface of EV or inside is not clear from these studies. In our proteomics screen under physiological conditions, we found that in brain L1CAM is significantly less expressed than SYP, an enriched neuronal marker (Figure S5). It is important to note that both L1CAM and SYP are expressed in other organs as well but highly enriched in the brain. SYP is an integral membrane protein localized to synaptic vesicles and the presynaptic membrane.45 SYP has been previously shown to be differentially regulated in neuronal derived exosomes (NDE) isolated from plasma of individuals with frontotemporal dementia, Alzheimer’s disease (AD) and HIV neurological disease.44, 46 Interestingly, another synaptic protein, synaptosomal-associated protein, 25kDa (SNAP-25), has also been shown to increase in NDE isolated from serum of AD patients 47. Further, we detected excitatory Amino Acid Transporter 1 (EAA1) or GLAST in brain-EV. GLAST has been recently reported as a marker for identifying astrocyte-derived EV (ADEV) in plasma 48. Another glial specific marker, S100β was also detected in our proteomics screen.
Similarly, for heart-EV, Caveolin-3 (CAV3) was validated to be heart specific EV marker. CAV3 is a cholesterol-binding protein and is one of the main protein components of caveolae and is involved in many cardiovascular processes such as myocardial hypertrophy, myocardial ischemia, and arrhythmogenesis.49 One recent study showed that CAV3 is present in microvesicles released from cardiomyocytes.50 Due to its highest expression in heart-EV when compared to rest of the ODE, CAV3 could be potentially used to isolate heart-EV from peripheral biofluids to determine cardiovascular dysfunction in individuals with heart disease. Intriguingly, organ or tissue-specific EV could serve as non-invasive biomarkers to monitor immunologic rejections of transplanted tissue. 51 A recent heart transplant study in mice showed that EV released from the donor's heart could predict early acute rejection in the host. 52 Alterations in the expression of CAV3 could potentially serve as a biomarker to detect heart specific alterations. Kidney specific marker, SLC22A2 is an organic cation transporter 2 (OCT2), plays an important role in the renal secretion of various cationic compounds. 53-56 Further, EV isolated from human urine could potentially serve as a non-invasive biomarker for renal dysfunction.57, 58 . Lung-derived EV and its associated cargo can be used as a non-invasive biomarker for diagnostic, prognostic, and therapeutic treatment of lung disorders, especially cancer.59-62 SP-B is a lipophilic protein that is required to facilitate lung inflation and minimize the work of respiration by reducing surface tension in the lung, thus maintaining alveolar stability. Mature SP-B forms are seen in multi-vesicular bodies (MVB), indicating their likely presence on EV.63 Circulating immature forms of SP-B (pro-SP-B) were identified as lung-specific biomarkers for alveolar-capillary membrane (ACM) dysfunction, increased risk of lung cancer, and chronic obstructive pulmonary disease (COPD).64-66 Finally, for the liver, we found FABP1 to be a specific marker. FABP1 is critical for fatty acid metabolism and plays a protective role during oxidative stress, starvation, and infections.67 Additionally, FABP1 has previously been used as molecular biomarkers in drug-induced liver injury (DILI).68 A recent finding indicated that in patients with type 2 diabetes mellitus (T2DM), FABP1 is linked to the pathogenesis of the nonalcoholic fatty liver disease (NAFLD).69 FABP1 plasma levels have shown a positive correlation with body mass index (BMI), creatinine levels, waist circumference, and many hepatic diseases.69 Interestingly, one study reported the presence of FABP1 in exosomes derived from hepatocytes.70
Intriguingly, many of the organ specific EV markers that were identified through our proteomic screening are also invariably enriched in the host tissue and subcellular organelles. For example, liver specific FABP1 and brain specific SYP are also found in organelles such as peroxisomes and synaptic vesicles respectively. However, the isolation of peroxisomes for instance uses entirely different protocol than EV which includes freezing of the density step-gradient, a prerequisite for a successful separation71. Similarly, synaptic vesicle preparations include breaking of the synaptosome membranes before the high centrifugation step to release synaptic vesicles72-76. Hence, the variances in the isolations method largely prevent any organelle contaminations. To summarize, our results conclude that it is imperative to determine more than one organ specific EV marker in biofluids to discern the EV origin and therefore its relevance to pathogenesis.
ODE Profiling Through Single EV Imaging
In additional to the traditional western blotting, we also validated the specificity of ODE markers by using a nanoplasmonic technology platform -single EV analysis (SEA). SEA technology is capable of robust, multiplexed protein biomarker measurement in individual vesicles. EV are immobilized inside a microfluidic chamber and then on-chip immuno-staining and imaging is performed.77 Here, marker sets that showed the highest selectivity among organs were used: SYP (brain), CAV3 (heart), SLC22A (kidney), SP-B (lung), and FABP1 (liver). We first matched the concentration of ODEs harvested from different organs (1.0×1010 EV/mL). These samples were then immuno-labeled for fluorescent imaging. As a representative result, Figure 7A shows ODE images with SYP labeling. Overall, the marker expression was dominant only in brain-derived EV, corroborating the validation from our immunoblotting platform. We further used marker-positive EV numbers as a metric for quantitative comparison (Figure 7B). SYP-labeling imparted a significant contrast between brain-derived EV and other ODEs. For a given marker, we defined its selectivity as the ratio between marker-positive and negative EV counts. For SYP, these values were > 65. Similarly, we observed high selectivity for other markers (Figure 7C; see Figure S6 for all images); the average selectivity was 30.
Currently available diagnostics are less sensitive and require 105–106 EV per biomarker to measure protein (e.g., Western, ELISA) or 102– 103 EV for the more sensitive methods μNMR, nPLEX).77 This newly developed method could be invariably used to isolate and quantitate organ specific EV changes from biofluids to follow the progression of disease pathology during therapy.
Validation of ODE from human postmortem tissues
To further extend our preclinical findings from a rat model to a clinical setting, we validated the identified ODE markers in postmortem human tissues. We isolated EV from postmortem human brain, heart, kidney, lung, and liver tissues, and performed TEM, NTA, and SEA as performed previously in our rat studies. TEM analysis confirmed that we collected nanoscale vesicles from human organs (Figure 8A and Figure S7). Intriguingly, we obtained less total EV protein when compared to EV protein isolated from rat organs even when using similar weights of the starting material (Table S5). This discrepancy could be attributed to the time elapsed between death and collection of tissues (PMI) that could invariably result in loss of protein integrity that could lead to denaturation of tissue proteins. Interestingly, EV derived from human lung showed no bud membrane protrusions as seen with rat lung derived EV (Figure S6). The size range of these vesicles ranged between 50-300 nm. Next, we performed molecular analyses on select protein markers. Immunoblotting showed that ODE samples were positive with EV-specific protein markers (Hsp-70, CD63, CD81), whereas the negative marker (calnexin) was present only in the whole tissue lysates (Figure S8). Both ODE and tissue samples also showed the presence of tissue markers identified from our rat study (Figure 8B): SYP (brain), CAV3 (heart), SLC22A2 (kidney), SP-B (lung), and FABP1 (liver). Single EV imaging further confirmed the tissue-specific nature of these markers (Figure 8C); the number of marker-positive vesicles was the highest only when tissue-type was matched. Because total EV concentrations varied among ODE sources, we normalized each column to the maximum EV numbers detected. The number of marker-positive EVs were counted and heatmap was generated as shown in Supplementary Figure S9. In summary, both rat and human ODE show similar EV marker expression profiles, thus denoting their respective use as potential markers in both pre-clinical and clinical applications.
CONCLUSION
Using both biochemical and single-EV imaging methods, we show a proof of concept for identifying specific EV markers for brain, lung, heart, kidney, and liver using both preclinical (rat) and clinical (human) model systems. ODE can cross tissue barriers and circulate in blood, thereby serving as a promising non-invasive biomarker to detect organ dysfunctions. Our proteomic analysis of ODE revealed that the protein signatures identified from different organs are distinct from one another. Post-validations by western blot and single-EV imaging on both rat and human ODE, we confirmed SYP, CAV3, SLC22A2, SP-B, and FABP1 as specific markers for brain, heart, kidney, lung, and liver, respectively.
Although we validated one marker for each organ, there is a strong possibility of more potential organ specific-markers in ODE. Therefore, more stringent strategies such as tandem or sequential immunoprecipitation might be required to pull down organ-specific EV. A more thorough analysis of the data needs to be performed to ascertain the cellular origin of EV. Therefore, we recommend using more than one EV protein markers for organ-specific screening. In summary our robust approach provides a valuable proof of concept approach in defining specific ODE markers which further could be developed as potential therapeutic candidates for respective end-organ associated pathologies.
MATERIALS AND METHODS
Animals.
Male and female Sprague Dawley rats were obtained from Charles River Laboratories Inc. (Wilmington, MA, USA) and group housed in a 12-h light-dark cycle and fed ad libitum. All procedures and protocols were approved by the Institutional Animal Care and Use Committee of the University of Nebraska Medical Center and conducted in accordance with the National Institutes of Health Guide for the Care and Use of Laboratory Animals.
Isolation of Organs.
We employed both perfusion and non-perfusion methods to isolate brain, heart, kidney, lung, and liver from the animals. For isolation employing perfusion, animals were deeply anesthetized using isoflurane followed by infusion with 1X phosphate-buffered saline (PBS; 70013-032, Gibco-Thermo Fisher Scientific, Waltham, MA, USA) using a peristaltic pump (Fisherbrand Variable-Flow Peristaltic Pump, Waltham, MA, USA) to clear the blood. Animals that did not undergo perfusions were sacrificed via overdose with isoflurane followed by decapitation and retrieval of organs. The different organs were harvested and immediately flash-frozen on dry ice, followed by storage at −80 °C until further use.
Human Organs.
Human postmortem tissues (Heart, Lung, Liver and Kidney) from two donors were obtained from the National Disease Research Interchange (NDRI) (Philadelphia, PA, USA), and human postmortem Brain samples from two additional donors were obtained from the UCLA National Neurological AIDS Bank of the National NeuroAIDS Tissue Consortium (NNTC). All samples were stored at −80° C until further use. See Table S3 for additional information on donors.
Extracellular Vesicles (EV) Isolations.
EV isolations were carried out as described in our previous studies with minor modifications.31, 32, 78 This protocol is well verified and we have consistently got similar results in all the runs that were performed for this study. Briefly, tissue weighing ~450 mg (Table S4) each were minced and treated with 20 unit/ml papain (P4762, Sigma-Aldrich, MO, USA) in 3.5 ml of Hibernate A (NC0176976, Fisher Scientific, Hampton, NH, USA) solution and rocked for 15-25 min (brain, 15 min; kidney and liver, 20 min; heart and lung, 25 min) at 37 °C. The reaction was then quenched with 6.5 ml cold Hibernate A solution and gently homogenized using a glass pipette. Tissue homogenate was sequentially centrifuged at 300 × g for 10 min, 2000 × g for 10 min, and at 10,000 × g for 30 min respectively at 4°C to remove dead cells, cell debris, and larger vesicles, respectively. The supernatant was then filtered through a 0.20 μm syringe filter (09-754-13, Corning Fisher Scientific, Hampton, NH, USA) and centrifuged at 100,000 × g for 70 min at 4 °C to pellet down the EV. The EV pellet was then resuspended in 37 ml particle-free cold 1X PBS and centrifuged again at 100,000 × g for 70 min at 4 °C. A sucrose gradient was prepared using 2 ml steps starting from 2.0 M sucrose at the bottom with a 0.25 M sucrose solution at the top. The washed EV pellet was resuspended in 2 ml of 0.95 M sucrose solution and layered between 0.60 M and 1.30 M sucrose layers. The gradients were then centrifuged at 200,000 × g for 16 h at 4 °C. To make sure layers should not mix, we used lower acceleration (7) and lower deceleration (7) during our overnight run. After run is complete we immediately take out respective layers. For collecting the purified EV, a 2 ml fraction was discarded from the top of the gradient, and the next 8 ml of the gradient were collected, diluted to 37 ml with cold particle-free PBS, and centrifuged at 100,000 × g at 4 °C for 70 min. EV pellets were suspended in 50 - 100 μl of particle-free PBS and further used for downstream analysis.
Nanoparticle Tracking Analysis (NTA).
EV size distribution curves and concentration measurements were carried out by NTA using a Nanosight NS300TM (Malvern Instruments, UK). For NTA analysis, EV pellets were resuspended in 100 μl PBS, following which 10 μl of the sample was diluted to 1:100 – 1:1000 in PBS prior to measurements. All samples were loaded with the laser module outside the instrument. As the sample was loaded, care was taken to avoid air pockets. The machine was equipped with a 488 nm laser and a syringe pump system, with a pump infusion speed of 20. The standard measurement option was selected for the scripted workflow to capture videos. The number and duration of captures were set to 5 and 60s, respectively. The base filename and location were selected prior to starting the run. The camera level was set at 11. Background measurements were performed with filtered PBS, which revealed the absence of any kind of particles. Five video recordings were carried out for each EV preparation with a duration of the 60s with frame rates of 25 frames/sec. Once the videos were recorded, the NTA 3.1 software version was used to analyze the sample videos. For analysis, the screen gain was set to 1.0, and the detection threshold was adjusted to set the minimum brightness of pixels to be considered. At the end of the analyses, the dilution factor for the samples was updated before data export.
Transmission Electron Microscopy (TEM).
For TEM, a 10 μl EV sample was mixed with 90 μL of TEM fix buffer (2% glutaraldehyde, 2% paraformaldehyde, and 0.1 M phosphate buffer). A 10 μl drop of EV-buffer mix sample was spotted onto formvar/silicon monoxide coated 200 mesh copper grids (Ted Pella Inc. Redding, CA) and allowed to sit for 3 min. Grids were glow discharged for 60 seconds at 20 μA with a GloQube glow discharge unit (Quorum Technologies, East Sussex, UK) prior to use. The excess solution was blotted off by using a clean piece of Whatman #50 filter paper held by forceps at 45° angle to touch only the edge of the grid, leaving behind a thin film of the sample that was allowed to dry for 1 min. Samples were negatively stained with NanoVan (Nanoprobes, New York, NY) and examined on a Tecnai G2 Spirit TWIN (FEI, Hillsboro, OR) operating at an accelerating voltage of 80 kV. Images were acquired digitally with an AMT (Woburn, MA) digital imaging system.
Western Blotting.
EV pellets were homogenized in RIPA buffer containing 1% SDS, and protein estimated using the BCA protein assay kit (Thermo Fisher Scientific, Rockford, IL, USA). Western blotting was performed on the various tissue-specific EV as described previously.31, 78, 79 Briefly, 20-40 μg protein was loaded onto NuPAGE 4–12% Bis-Tris gels (Invitrogen) and run either under non-reducing (CD63 and CD81) or reducing (Hsp-70 and flotillin-1) conditions followed by their transfer onto nitrocellulose membranes using the iBlot 2 Gel Transfer Device (Invitrogen). Membranes were blocked in TBS SuperBlock buffer (Thermo Fisher Scientific) for 30 min, and immunoblotting carried out overnight at 4 °C with primary antibodies (see Table 1 for details). The next day, membranes were incubated with respective HRP conjugated secondary antibodies for 1.5 hr at room temperature on a rocker. Blots were developed with 1:1 solution of Radiance Chemiluminescent Substrate and Luminol/Enhancer (Azure Biosystems) and visualized using a c300 imaging system (Azure Biosystems). Images were quantified using the ImageJ software.
Table 1.
Marker | Running Condition |
Antibody (Catalog #) |
Sample species |
1° Ab dilution |
2° Ab dilution |
---|---|---|---|---|---|
Hsp-70 | Reducing | SAB4200714 | Rat/Human | 1:1000 | 1:2000 |
Flotillin-1 | Reducing | abcam41927 | Rat/Human | 1:1000 | 1:2000 |
Calnexin | Reducing | ab22595 | Rat/Human | 1:1000 | 1:2000 |
Synaptophysin | Reducing | PA1-1043 | Rat/Human | 1:1000 | 1:2000 |
Caveolin-3 | Reducing | ab2912 | Rat | 1:1000 | 1:2000 |
Caveolin-3 | Reducing | MAB6706 | Human | 1:1000 | 1:2000 |
SLC22A2 | Reducing | PA5-37290 | Rat | 1:500 | 1:1000 |
SLC22A2 | Reducing | MAB6547 | Human | 1:500 | 1:1000 |
ANXA5 | Reducing | ab14196 | Rat | 1:500 | 1:2000 |
FABP1 | Reducing | Invitrogen 720242 |
Rat/Human | 1:500 | 1:1000 |
COX5B | Reducing | ab180136 | Rat | 1:1000 0 |
1:2000 0 |
ICAM-1 | Reducing | sc-107 | Rat | 1:500 | 1:1000 |
MOG | Reducing | MA5-24645 | Rat | 1:500 | 1:1000 |
SLC12A1 | Reducing | ab171747 | Rat | 1:1000 | 1:2000 |
NCAM1 | Reducing | ab232781 | Rat | 1:500 | 1:1000 |
MGST1 | Reducing | ab129175 | Rat | 1:500 | 1:1000 |
GSTM1 | Reducing | ab108524 | Rat | 1:1000 | 1:2000 |
EAAT1 | Reducing | sc-515839 | Rat | 1:1000 | 1:2000 |
HSD3B7 | Reducing | ab190223 | Rat | 1:500 | 1:1000 |
CHIA | Reducing | PA5-03057 | Rat | 1:1000 | 1:2000 |
SP-B | Non- reducing |
WRAB-48604 | Rat/Human | 1:1000 | 1:2000 |
CD81 | Non- reducing |
MCA1846 | Rat | 1:500 | 1:1000 |
CD81 | Non- reducing |
MAB4615 | Human | 1:500 | 1:1000 |
CD63 | Non- reducing |
BD551458 | Rat/Human | 1:500 | 1:1000 |
Proteomics.
100 μg of EV protein prepared using RIPA buffer with 1% SDS per sample from three biological replicates and detergent removed by chloroform/methanol extraction. Samples were then subjected to quantitative mass spectrometry based proteomics. Briefly, the protein pellet was re-suspended in 100 mM ammonium bicarbonate and digested with MS-grade trypsin (Pierce) overnight at 37 °C. Peptides cleaned with PepClean C18 spin columns (Thermo Fisher Scientific) were resuspended in 2% acetonitrile (ACN) and 0.1% formic acid (FA). 500 ng from each sample was loaded onto trap column Acclaim PepMap 100 75μm x 2 cm C18 LC Columns (Thermo Fisher Scientific) at a flow rate of 4 μl/min. Subsequently, samples were separated with a Thermo RSLC Ultimate 3000 (Thermo Fisher Scientific) on a Thermo Easy-Spray PepMap RSLC C18 75μm x 50cm C-18 2 μm column (Thermo Fisher Scientific, Waltham, MA, USA) with a step gradient of 4–25% solvent B (0.1% FA in 80 % ACN) from 10-130 min and 25–45% solvent B for 130–145 min at 300 nL/min and 50 °C with a 180 min total run time. Thermo Orbitrap Fusion Lumos Tribrid (Thermo Fisher Scientific) mass spectrometer (data-dependent acquisition mode) was used to analyze the eluted peptides. By Orbitrap, with a resolution of 120,000, the full survey scan MS (from m/z 350–1800) was acquired. The ion filling time was set as 100 ms, while the AGC target for MS1 was set as 4 × 105. The precursor ions with charge state 2-6 were isolated in 3 s cycle and fragmented using HCD fragmentation with 35 % normalized collision energy and were detected at a mass resolution of 30,000 at 200 m/z. For MS/MS, the AGC target was set as 5 × 104, and ion filling time was set 60 ms while dynamic separation was set for 30 s with a 10 ppm mass window. The protein identification was performed by searching MS/MS data against the swiss-prot Rattus norvegicus protein database downloaded on Feb 13, 2019, using the in house mascot 2.6.2 (Matrix Science) search engine. With a maximum of two unexploited cleavage sites, the search was set up for full tryptic peptides. N-terminus acetylation and oxidized methionine were included as variable modifications, whereas cysteine carbamidomethylation was set as a fixed modification. Forerunner mass tolerance threshold was set at 10 ppm, and the maximum fragment mass error was 0.02 Da. The significance threshold score of the ion was calculated based on a false discovery rate (FDR, calculated using false positive matches/true positive matches+false positive matches) of ≤ 1% (http://www.matrixscience.com/help/decoy_help.html). Qualitative analysis was performed using progenesis QI proteomics 4.1 (Nonlinear Dynamics). Sample to sample experimental variation during sample runs was normalized using default settings (Normalize to all proteins) in progenesis QI proteomics.
Data Analysis and Bioinformatics.
Proteins identified with at least two unique peptides were included in the downstream differential analysis. Proteins were further filtered from downstream analysis if the spectral counts were missing in 80% of the samples, irrespective of the sample type. Spectral counts were analyzed using limma 80 to identify differentially expressed proteins between any two EV types. A total of ten pairwise differential expression analyses were performed across all sample groups based on the tissue of origin. A protein was identified as differentially expressed if the FDR (Benjamini and Hochberg’s method) corrected P-value was ≤0.05, and log2 fold change was ≥1.5. A heatmap of the top 25 proteins in each organ that exhibited significantly higher or lower expression with respect to other comparisons was plotted using the function heatmap.2 in the R (version 3.6.0) package gplots. Principal Component Analysis (PCA) was performed using ClustVis.81 Gene Ontology (GO) analysis of differentially expressed proteins was performed using the Cytoscape plugin ClueGO.82 Only proteins exhibiting significantly higher expression across all comparisons were included in this analysis. The biological process, molecular function, and KEGG pathways were included for GO enrichment analysis.
Single EV analysis (SEA) for organ specific protein markers.
EV from different organ tissues were prepared as described above. EV pellets were suspended in PBS and then filtered with a 0.22 μm syringe filter (SLGVR04NL, Millipore Sigma). Filtered EV were captured on a glass slide. Following 30 min incubation (20 °C), the slide was washed with PBS buffer containing 0.001% Tween 20 (PBST). After incubation with fixation buffer (4% formaldehyde), permiabilization buffer (BD perm/wash buffer, 554723, BD Biosciences) and blocking buffer (Superblock, 37515, Thermo Scientific), EV samples were incubated with primary antibodies (see Table 2 for antibody details). After washing with PBST, the fluorescence-conjugated secondary antibody was incubated for 30 min (20 °C) and washed again. Fluorescence images were taken with an upright fluorescent microscope (BX63, Olympus) using a 40× objective lense. Acquisition settings (i.e., magnification, exposure time, camera gains, illumination) were kept constant for all images.
Table 2.
Marker | Antibody (Catalog #) |
Sample species |
1° Ab dilution |
2° Ab dilution |
---|---|---|---|---|
Synaptophysin | PA1-1043 | Rat/Human | 1:100/1:50 | 1:500/1:200 |
Caveolin-3 | ab2912 | Rat | 1:100 | 1:500 |
Caveolin-3 | MAB6706 | Human | 1:50 | 1:400 |
SLC22A2 | PA5-37290 | Rat | 1:100 | 1:500 |
SLC22A2 | MAB6547 | Human | 1:50 | 1:400 |
ANXA5 | ab14196 | Rat | 1:100 | 1:500 |
SP-B | WRAB-48604 | Human | 1:50 | 1:100 |
FABP1 | CST 13368 | Rat/Human | 1:50/1:50 | 1:200/1:100 |
Supplementary Material
ACKNOWLEDGMENTS
We thank the University of Nebraska Medical Center Mass Spectrometry and Proteomics Core Facility for its expert assistance. The UNMC Mass Spectrometry and Proteomics Core Facility is administrated through the Office of the Vice Chancellor for Research and supported by state funds from the Nebraska Research Initiative (NRI). We also thank Tom Bargar and Nicholas Conoan for their expert technical assistance for TEM.
Funding Sources
This study is supported by NIDA grants R21DA046855 and R01DA042379 awarded to SVY, R21DA049577 to GP and HL and R01DA046852 to GP and SVY. The bioinformatics work has been partially supported by the National Institutes of Health grants to CG [5P20GM103427, 1P30GM127200].
NOTES
The authors declare the following competing financial interest(s): The authors have filed intellectual property for organ specific EV markers that were identified in this study.
The proteomics data have been deposited to the ProteomeXchange Consortium via the PRIDE partner repository with the data set identifier PXD020568.
Footnotes
Supplementary Information Statement
The Supporting Information is available free of charge on the ACS Publications website.
TEM analysis of EV isolated from perfused rat organs, Western blot analysis of EV purified from perfused rat organs, Bar Charts representing the enriched GO annotations, Characterization of organ-specific EV protein markers. Brain (A), heart (B), kidney (C), lung (D), and liver (E), Comparative expression of L1CAM and SYP in rat EV, Single EV imaging for ODEs with five organ-specific markers, TEM and NTA analysis of EV isolated from postmortem human organs, Western blot analysis of human EV and human tissue lysates, Signal contrast among human ODE types and organ-specific markers was compared, Total EV protein yield from non-perfused and perfused rat tissues, Differentially expressed proteins in brain, heart, kidney, lung, and liver, Spectral counts of abundant proteins from the EV of rat brain, heart, kidney, lung, and liver, Additional information on human donors, EV protein yields for different human organs.
REFERENCES
- 1.Porro C; Trotta T; Panaro MA, Microvesicles in the brain: Biomarker, messenger or mediator? J Neuroimmunol 2015, 288, 70–8. [DOI] [PubMed] [Google Scholar]
- 2.Mathieu M; Martin-Jaular L; Lavieu G; Thery C, Specificities of secretion and uptake of exosomes and other extracellular vesicles for cell-to-cell communication. Nat Cell Biol 2019, 21 (1), 9–17. [DOI] [PubMed] [Google Scholar]
- 3.Antonyak MA; Cerione RA, Emerging picture of the distinct traits and functions of microvesicles and exosomes. Proceedings of the National Academy of Sciences of the United States of America 2015, 112 (12), 3589–3590. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Maas SLN; Breakefield XO; Weaver AM, Extracellular Vesicles: Unique Intercellular Delivery Vehicles. Trends Cell Biol 2017, 27 (3), 172–188. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Raposo G; Stoorvogel W, Extracellular vesicles: exosomes, microvesicles, and friends. J Cell Biol 2013, 200 (4), 373–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Valadi H; Ekström K; Bossios A; Sjöstrand M; Lee JJ; Lötvall JO, Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nature Cell Biology 2007, 9, 654. [DOI] [PubMed] [Google Scholar]
- 7.Chen R; Xu X; Qian Z; Zhang C; Niu Y; Wang Z; Sun J; Zhang X; Yu Y, The biological functions and clinical applications of exosomes in lung cancer. Cell Mol Life Sci 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Shah R; Patel T; Freedman JE, Circulating Extracellular Vesicles in Human Disease. N Engl J Med 2018, 379 (10), 958–966. [DOI] [PubMed] [Google Scholar]
- 9.Jiang L; Gu Y; Du Y; Liu J, Exosomes: Diagnostic Biomarkers and Therapeutic Delivery Vehicles for Cancer. Mol Pharm 2019, 16 (8), 3333–3349. [DOI] [PubMed] [Google Scholar]
- 10.Wu M; Wang G; Hu W; Yao Y; Yu XF, Emerging roles and therapeutic value of exosomes in cancer metastasis. Mol Cancer 2019, 18 (1), 53. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Hosaka T; Yamashita T; Tamaoka A; Kwak S, Extracellular RNAs as Biomarkers of Sporadic Amyotrophic Lateral Sclerosis and Other Neurodegenerative Diseases. Int J Mol Sci 2019, 20 (13). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Rodrigues M; Richards N; Ning B; Lyon CJ; Hu TY, Rapid Lipid-Based Approach for Normalization of Quantum-Dot-Detected Biomarker Expression on Extracellular Vesicles in Complex Biological Samples. Nano Lett 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Morales-Kastresana A; Musich TA; Welsh JA; Telford W; Demberg T; Wood JCS; Bigos M; Ross CD; Kachynski A; Dean A; Felton EJ; Van Dyke J; Tigges J; Toxavidis V; Parks DR; Overton WR; Kesarwala AH; Freeman GJ; Rosner A; Perfetto SP; Pasquet L; Terabe M; McKinnon K; Kapoor V; Trepel JB; Puri A; Kobayashi H; Yung B; Chen X; Guion P; Choyke P; Knox SJ; Ghiran I; Robert-Guroff M; Berzofsky JA; Jones JC, High-fidelity detection and sorting of nanoscale vesicles in viral disease and cancer. J Extracell Vesicles 2019, 8 (1), 1597603. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Zhang P; Zhou X; Zeng Y, Multiplexed immunophenotyping of circulating exosomes on nano-engineered ExoProfile chip towards early diagnosis of cancer. Chem Sci 2019, 10 (21), 5495–5504. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Shahjin F; Chand S; Yelamanchili SV, Extracellular Vesicles as Drug Delivery Vehicles to the Central Nervous System. J Neuroimmune Pharmacol 2019. [DOI] [PubMed] [Google Scholar]
- 16.Kanninen KM; Bister N; Koistinaho J; Malm T, Exosomes as new diagnostic tools in CNS diseases. Biochim Biophys Acta 2016, 1862 (3), 403–10. [DOI] [PubMed] [Google Scholar]
- 17.Pulliam L; Sun B; Mustapic M; Chawla S; Kapogiannis D, Plasma neuronal exosomes serve as biomarkers of cognitive impairment in HIV infection and Alzheimer's disease. J Neurovirol 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Tang Y; Zhao S; Wang J; Li D; Ren Q; Tang Y, Plasma miR-122 as a potential diagnostic and prognostic indicator in human glioma. Neurol Sci 2017, 38 (6), 1087–1092. [DOI] [PubMed] [Google Scholar]
- 19.Shao N; Wang L; Xue L; Wang R; Lan Q, Plasma miR-454-3p as a potential prognostic indicator in human glioma. Neurol Sci 2015, 36 (2), 309–13. [DOI] [PubMed] [Google Scholar]
- 20.Lan F; Qing Q; Pan Q; Hu M; Yu H; Yue X, Serum exosomal miR-301a as a potential diagnostic and prognostic biomarker for human glioma. Cell Oncol (Dordr) 2018, 41 (1), 25–33. [DOI] [PubMed] [Google Scholar]
- 21.Yue X; Lan F; Hu M; Pan Q; Wang Q; Wang J, Downregulation of serum microRNA-205 as a potential diagnostic and prognostic biomarker for human glioma. J Neurosurg 2016, 124 (1), 122–8. [DOI] [PubMed] [Google Scholar]
- 22.Shi R; Wang PY; Li XY; Chen JX; Li Y; Zhang XZ; Zhang CG; Jiang T; Li WB; Ding W; Cheng SJ, Exosomal levels of miRNA-21 from cerebrospinal fluids associated with poor prognosis and tumor recurrence of glioma patients. Oncotarget 2015, 6 (29), 26971–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Li M; Zeringer E; Barta T; Schageman J; Cheng A; Vlassov AV, Analysis of the RNA content of the exosomes derived from blood serum and urine and its potential as biomarkers. Philos Trans R Soc Lond B Biol Sci 2014, 369 (1652). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Wong C-H; Chen Y-C, Clinical significance of exosomes as potential biomarkers in cancer. World journal of clinical cases 2019, 7 (2), 171–190. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Vlassov AV; Magdaleno S; Setterquist R; Conrad R, Exosomes: current knowledge of their composition, biological functions, and diagnostic and therapeutic potentials. Biochim Biophys Acta 2012, 1820 (7), 940–8. [DOI] [PubMed] [Google Scholar]
- 26.Revenfeld AL; Baek R; Nielsen MH; Stensballe A; Varming K; Jorgensen M, Diagnostic and prognostic potential of extracellular vesicles in peripheral blood. Clin Ther 2014, 36 (6), 830–46. [DOI] [PubMed] [Google Scholar]
- 27.Jansen F; Nickenig G; Werner N, Extracellular Vesicles in Cardiovascular Disease: Potential Applications in Diagnosis, Prognosis, and Epidemiology. Circ Res 2017, 120 (10), 1649–1657. [DOI] [PubMed] [Google Scholar]
- 28.Crescitelli R; Lasser C; Jang SC; Cvjetkovic A; Malmhall C; Karimi N; Hoog JL; Johansson I; Fuchs J; Thorsell A; Gho YS; Olofsson Bagge R; Lotvall J, Subpopulations of extracellular vesicles from human metastatic melanoma tissue identified by quantitative proteomics after optimized isolation. J Extracell Vesicles 2020, 9 (1), 1722433. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Ishiguro K; Yan IK; Patel T, Isolation of Tissue Extracellular Vesicles from the Liver. J Vis Exp 2019, (150). [DOI] [PubMed] [Google Scholar]
- 30.Vella LJ; Scicluna BJ; Cheng L; Bawden EG; Masters CL; Ang CS; Willamson N; McLean C; Barnham KJ; Hill AF, A rigorous method to enrich for exosomes from brain tissue. J Extracell Vesicles 2017, 6 (1), 1348885. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Yelamanchili SV; Lamberty BG; Rennard DA; Morsey BM; Hochfelder CG; Meays BM; Levy E; Fox HS, MiR-21 in Extracellular Vesicles Leads to Neurotoxicity via TLR7 Signaling in SIV Neurological Disease. PLoS Pathog 2015, 11 (7), e1005032. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Perez-Gonzalez R; Gauthier SA; Kumar A; Levy E, The exosome secretory pathway transports amyloid precursor protein carboxyl-terminal fragments from the cell into the brain extracellular space. J Biol Chem 2012, 287 (51), 43108–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Harrison EB; Hochfelder CG; Lamberty BG; Meays BM; Morsey BM; Kelso ML; Fox HS; Yelamanchili SV, Traumatic brain injury increases levels of miR-21 in extracellular vesicles: implications for neuroinflammation. FEBS Open Bio 2016, 6 (8), 835–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Kowal J; Arras G; Colombo M; Jouve M; Morath JP; Primdal-Bengtson B; Dingli F; Loew D; Tkach M; Thery C, Proteomic comparison defines novel markers to characterize heterogeneous populations of extracellular vesicle subtypes. Proc Natl Acad Sci U S A 2016, 113 (8), E968–77. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Huang Y; Cheng L; Turchinovich A; Mahairaki V; Troncoso JC; Pletniková O; Haughey NJ; Vella LJ; Hill AF; Zheng L; Witwer KW, Influence of species and processing parameters on recovery and content of brain tissue-derived extracellular vesicles. Journal of Extracellular Vesicles 2020, 9 (1), 1785746. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Vagner T; Chin A; Mariscal J; Bannykh S; Engman DM; Di Vizio D, Protein Composition Reflects Extracellular Vesicle Heterogeneity. Proteomics 2019, 19 (8), e1800167. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Jalalian SH; Ramezani M; Jalalian SA; Abnous K; Taghdisi SM, Exosomes, new biomarkers in early cancer detection. Anal Biochem 2019, 571, 1–13. [DOI] [PubMed] [Google Scholar]
- 38.Saenz-Pipaon G; San Martín P; Planell N; Maillo A; Ravassa S; Vilas-Zornoza A; Martinez-Aguilar E; Rodriguez JA; Alameda D; Lara-Astiaso D; Prosper F; Paramo JA; Orbe J; Gomez-Cabrero D; Roncal C, Functional and transcriptomic analysis of extracellular vesicles identifies calprotectin as a new prognostic marker in peripheral arterial disease (PAD). Journal of Extracellular Vesicles 2020, 9 (1), 1729646. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Saeedi S; Israel S; Nagy C; Turecki G, The emerging role of exosomes in mental disorders. Transl Psychiatry 2019, 9 (1), 122. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Fiandaca MS; Kapogiannis D; Mapstone M; Boxer A; Eitan E; Schwartz JB; Abner EL; Petersen RC; Federoff HJ; Miller BL; Goetzl EJ, Identification of preclinical Alzheimer's disease by a profile of pathogenic proteins in neurally derived blood exosomes: A case-control study. Alzheimers Dement 2015, 11 (6), 600–7.e1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Pulliam L; Sun B; Mustapic M; Chawla S; Kapogiannis D, Plasma neuronal exosomes serve as biomarkers of cognitive impairment in HIV infection and Alzheimer's disease. J Neurovirol 2019, 25 (5), 702–709. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Dagur RS; Liao K; Sil S; Niu F; Sun Z; Lyubchenko YL; Peeples ES; Hu G; Buch S, Neuronal-derived extracellular vesicles are enriched in the brain and serum of HIV-1 transgenic rats. J Extracell Vesicles 2020, 9 (1), 1703249. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Dagur RS; Liao K; Sil S; Niu F; Sun Z; Lyubchenko YL; Peeples ES; Hu G; Buch S, Neuronal-derived extracellular vesicles are enriched in the brain and serum of HIV-1 transgenic rats. Journal of Extracellular Vesicles 2020, 9 (1), 1703249. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Sun B; Dalvi P; Abadjian L; Tang N; Pulliam L, Blood neuron-derived exosomes as biomarkers of cognitive impairment in HIV. AIDS (London, England) 2017, 31 (14), F9–f17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Sudhof TC; Jahn R, Proteins of synaptic vesicles involved in exocytosis and membrane recycling. Neuron 1991, 6 (5), 665–77. [DOI] [PubMed] [Google Scholar]
- 46.Goetzl EJ; Kapogiannis D; Schwartz JB; Lobach IV; Goetzl L; Abner EL; Jicha GA; Karydas AM; Boxer A; Miller BL, Decreased synaptic proteins in neuronal exosomes of frontotemporal dementia and Alzheimer's disease. FASEB J 2016, 30 (12), 4141–4148. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Agliardi C; Guerini FR; Zanzottera M; Bianchi A; Nemni R; Clerici M, SNAP-25 in Serum Is Carried by Exosomes of Neuronal Origin and Is a Potential Biomarker of Alzheimer’s Disease. Molecular Neurobiology 2019, 56 (8), 5792–5798. [DOI] [PubMed] [Google Scholar]
- 48.Winston CN; Romero HK; Ellisman M; Nauss S; Julovich DA; Conger T; Hall JR; Campana W; O'Bryant SE; Nievergelt CM; Baker DG; Risbrough VB; Rissman RA, Assessing Neuronal and Astrocyte Derived Exosomes From Individuals With Mild Traumatic Brain Injury for Markers of Neurodegeneration and Cytotoxic Activity. Front Neurosci 2019, 13, 1005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Cohen AW; Hnasko R; Schubert W; Lisanti MP, Role of caveolae and caveolins in health and disease. Physiol Rev 2004, 84 (4), 1341–79. [DOI] [PubMed] [Google Scholar]
- 50.Waldenstrom A; Genneback N; Hellman U; Ronquist G, Cardiomyocyte microvesicles contain DNA/RNA and convey biological messages to target cells. PLoS One 2012, 7 (4), e34653. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Vallabhajosyula P; Korutla L; Habertheuer A; Yu M; Rostami S; Yuan CX; Reddy S; Liu C; Korutla V; Koeberlein B; Trofe-Clark J; Rickels MR; Naji A, Tissue-specific exosome biomarkers for noninvasively monitoring immunologic rejection of transplanted tissue. J Clin Invest 2017, 127 (4), 1375–1391. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Habertheuer A; Korutla L; Rostami S; Reddy S; Lal P; Naji A; Vallabhajosyula P, Donor tissue-specific exosome profiling enables noninvasive monitoring of acute rejection in mouse allogeneic heart transplantation. J Thorac Cardiovasc Surg 2018, 155 (6), 2479–2489. [DOI] [PubMed] [Google Scholar]
- 53.Nigam SK, The SLC22 Transporter Family: A Paradigm for the Impact of Drug Transporters on Metabolic Pathways, Signaling, and Disease. Annu Rev Pharmacol Toxicol 2018, 58, 663–687. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Urakami Y; Okuda M; Masuda S; Saito H; Inui KI, Functional characteristics and membrane localization of rat multispecific organic cation transporters, OCT1 and OCT2, mediating tubular secretion of cationic drugs. J Pharmacol Exp Ther 1998, 287 (2), 800–5. [PubMed] [Google Scholar]
- 55.Kimura N; Masuda S; Tanihara Y; Ueo H; Okuda M; Katsura T; Inui K, Metformin is a superior substrate for renal organic cation transporter OCT2 rather than hepatic OCT1. Drug Metab Pharmacokinet 2005, 20 (5), 379–86. [DOI] [PubMed] [Google Scholar]
- 56.Yonezawa A; Masuda S; Nishihara K; Yano I; Katsura T; Inui K, Association between tubular toxicity of cisplatin and expression of organic cation transporter rOCT2 (Slc22a2) in the rat. Biochem Pharmacol 2005, 70 (12), 1823–31. [DOI] [PubMed] [Google Scholar]
- 57.Gonzales PA; Pisitkun T; Hoffert JD; Tchapyjnikov D; Star RA; Kleta R; Wang NS; Knepper MA, Large-scale proteomics and phosphoproteomics of urinary exosomes. J Am Soc Nephrol 2009, 20 (2), 363–79. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Console L; Scalise M; Indiveri C, Exosomes in inflammation and role as biomarkers. Clin Chim Acta 2019, 488, 165–171. [DOI] [PubMed] [Google Scholar]
- 59.Amiri A; Pourhanifeh MH; Mirzaei HR; Nahand JS; Moghoofei M; Sahebnasagh R; Mirzaei H; Hamblin MR, Exosomes and Lung cancer: Roles in pathophysiology, diagnosis and therapeutic applications. Curr Med Chem 2020. [DOI] [PubMed] [Google Scholar]
- 60.Sun S; Chen H; Xu C; Zhang Y; Zhang Q; Chen L; Ding Q; Deng Z, Exosomal miR-106b serves as a novel marker for lung cancer and promotes cancer metastasis via targeting PTEN. Life Sci 2020, 244, 117297. [DOI] [PubMed] [Google Scholar]
- 61.Niu L; Song X; Wang N; Xue L; Song X; Xie L, Tumor-derived exosomal proteins as diagnostic biomarkers in non-small cell lung cancer. Cancer Sci 2019, 110 (1), 433–442. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Sandfeld-Paulsen B; Jakobsen KR; Baek R; Folkersen BH; Rasmussen TR; Meldgaard P; Varming K; Jorgensen MM; Sorensen BS, Exosomal Proteins as Diagnostic Biomarkers in Lung Cancer. J Thorac Oncol 2016, 11 (10), 1701–10. [DOI] [PubMed] [Google Scholar]
- 63.Jacob A; Morley M; Hawkins F; McCauley KB; Jean JC; Heins H; Na CL; Weaver TE; Vedaie M; Hurley K; Hinds A; Russo SJ; Kook S; Zacharias W; Ochs M; Traber K; Quinton LJ; Crane A; Davis BR; White FV; Wambach J; Whitsett JA; Cole FS; Morrisey EE; Guttentag SH; Beers MF; Kotton DN, Differentiation of Human Pluripotent Stem Cells into Functional Lung Alveolar Epithelial Cells. Cell Stem Cell 2017, 21 (4), 472–488 e10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Mannino DM, Biomarkers in COPD: the search continues! Eur Respir J 2015, 45 (4), 872–4. [DOI] [PubMed] [Google Scholar]
- 65.Sin DD; Tammemagi CM; Lam S; Barnett MJ; Duan X; Tam A; Auman H; Feng Z; Goodman GE; Hanash S; Taguchi A, Pro-surfactant protein B as a biomarker for lung cancer prediction. J Clin Oncol 2013, 31 (36), 4536–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Banfi C; Agostoni P, Surfactant protein B: From biochemistry to its potential role as diagnostic and prognostic marker in heart failure. Int J Cardiol 2016, 221, 456–62. [DOI] [PubMed] [Google Scholar]
- 67.Wang G; Bonkovsky HL; de Lemos A; Burczynski FJ, Recent insights into the biological functions of liver fatty acid binding protein 1. J Lipid Res 2015, 56 (12), 2238–47. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Fu S; Wu D; Jiang W; Li J; Long J; Jia C; Zhou T, Molecular Biomarkers in Drug-Induced Liver Injury: Challenges and Future Perspectives. Front Pharmacol 2019, 10, 1667. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Lu YC; Chang CC; Wang CP; Hung WC; Tsai IT; Tang WH; Wu CC; Wei CT; Chung FM; Lee YJ; Hsu CC, Circulating fatty acid-binding protein 1 (FABP1) and nonalcoholic fatty liver disease in patients with type 2 diabetes mellitus. Int J Med Sci 2020, 17 (2), 182–190. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Jia X; Chen J; Megger DA; Zhang X; Kozlowski M; Zhang L; Fang Z; Li J; Chu Q; Wu M; Li Y; Sitek B; Yuan Z, Label-free Proteomic Analysis of Exosomes Derived from Inducible Hepatitis B Virus-Replicating HepAD38 Cell Line. Mol Cell Proteomics 2017, 16 (4 suppl 1), S144–S160. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Manner A; Islinger M, Isolation of Peroxisomes from Rat Liver and Cultured Hepatoma Cells by Density Gradient Centrifugation. Methods Mol Biol 2017, 1595, 1–11. [DOI] [PubMed] [Google Scholar]
- 72.Darna M; Schmutz I; Richter K; Yelamanchili SV; Pendyala G; Holtje M; Albrecht U; Ahnert-Hilger G, Time of day-dependent sorting of the vesicular glutamate transporter to the plasma membrane. J Biol Chem 2009, 284 (7), 4300–7. [DOI] [PubMed] [Google Scholar]
- 73.Reisinger C; Yelamanchili SV; Hinz B; Mitter D; Becher A; Bigalke H; Ahnert-Hilger G, The synaptophysin/synaptobrevin complex dissociates independently of neuroexocytosis. J Neurochem 2004, 90 (1), 1–8. [DOI] [PubMed] [Google Scholar]
- 74.Mitter D; Reisinger C; Hinz B; Hollmann S; Yelamanchili SV; Treiber-Held S; Ohm TG; Herrmann A; Ahnert-Hilger G, The synaptophysin/synaptobrevin interaction critically depends on the cholesterol content. J Neurochem 2003, 84 (1), 35–42. [DOI] [PubMed] [Google Scholar]
- 75.Yelamanchili SV; Reisinger C; Becher A; Sikorra S; Bigalke H; Binz T; Ahnert-Hilger G, The C-terminal transmembrane region of synaptobrevin binds synaptophysin from adult synaptic vesicles. Eur J Cell Biol 2005, 84 (4), 467–75. [DOI] [PubMed] [Google Scholar]
- 76.Yelamanchili SV; Pendyala G; Brunk I; Darna M; Albrecht U; Ahnert-Hilger G, Differential sorting of the vesicular glutamate transporter 1 into a defined vesicular pool is regulated by light signaling involving the clock gene Period2. J Biol Chem 2006, 281 (23), 15671–9. [DOI] [PubMed] [Google Scholar]
- 77.Lee K; Fraser K; Ghaddar B; Yang K; Kim E; Balaj L; Chiocca EA; Breakefield XO; Lee H; Weissleder R, Multiplexed Profiling of Single Extracellular Vesicles. ACS Nano 2018, 12 (1), 494–503. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Moore D; Meays BM; Madduri LSV; Shahjin F; Chand S; Niu M; Albahrani A; Guda C; Pendyala G; Fox HS; Yelamanchili SV, Downregulation of an Evolutionary Young miR-1290 in an iPSC-Derived Neural Stem Cell Model of Autism Spectrum Disorder. Stem Cells Int 2019, 2019, 8710180. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Shahjin F; Guda RS; Schaal VL; Odegaard K; Clark A; Gowen A; Xiao P; Lisco SJ; Pendyala G; Yelamanchili SV, Brain-Derived Extracellular Vesicle microRNA Signatures Associated with In Utero and Postnatal Oxycodone Exposure. Cells 2019, 9 (1). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Ritchie ME; Phipson B; Wu D; Hu Y; Law CW; Shi W; Smyth GK, limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res 2015, 43 (7), e47. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Metsalu T; Vilo J, ClustVis: a web tool for visualizing clustering of multivariate data using Principal Component Analysis and heatmap. Nucleic Acids Res 2015, 43 (W1), W566–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Bindea G; Mlecnik B; Hackl H; Charoentong P; Tosolini M; Kirilovsky A; Fridman WH; Pages F; Trajanoski Z; Galon J, ClueGO: a Cytoscape plug-in to decipher functionally grouped gene ontology and pathway annotation networks. Bioinformatics 2009, 25 (8), 1091–3. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.