Next Article in Journal
Heterogenic Origin of Micro RNAs in Atlantic Salmon (Salmo salar) Seminal Plasma
Next Article in Special Issue
MiR-519d-3p in Trophoblastic Cells: Effects, Targets and Transfer to Allogeneic Immune Cells via Extracellular Vesicles
Previous Article in Journal
In Vivo Targets of Pasteurella Multocida Toxin
Previous Article in Special Issue
Therapeutic Use of Extracellular Vesicles for Acute and Chronic Lung Disease
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

HIV-1 and Amyloid Beta Remodel Proteome of Brain Endothelial Extracellular Vesicles

Department of Biochemistry and Molecular Biology, University of Miami School of Medicine, Miami, FL 33136-1019, USA
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2020, 21(8), 2741; https://doi.org/10.3390/ijms21082741
Submission received: 11 March 2020 / Revised: 6 April 2020 / Accepted: 7 April 2020 / Published: 15 April 2020
(This article belongs to the Special Issue Extracellular Vesicles and Cell–Cell Communication)

Abstract

:
Amyloid beta (Aβ) depositions are more abundant in HIV-infected brains. The blood–brain barrier, with its backbone created by endothelial cells, is assumed to be a core player in Aβ homeostasis and may contribute to Aβ accumulation in the brain. Exposure to HIV increases shedding of extracellular vesicles (EVs) from human brain endothelial cells and alters EV-Aβ levels. EVs carrying various cargo molecules, including a complex set of proteins, can profoundly affect the biology of surrounding neurovascular unit cells. In the current study, we sought to examine how exposure to HIV, alone or together with Aβ, affects the surface and total proteomic landscape of brain endothelial EVs. By using this unbiased approach, we gained an unprecedented, high-resolution insight into these changes. Our data suggest that HIV and Aβ profoundly remodel the proteome of brain endothelial EVs, altering the pathway networks and functional interactions among proteins. These events may contribute to the EV-mediated amyloid pathology in the HIV-infected brain and may be relevant to HIV-1-associated neurocognitive disorders.

1. Introduction

HIV-infected brains tend to have enhanced amyloid beta (Aβ) deposition [1,2,3,4,5,6], mostly in the perivascular space [3,7,8,9]. Indeed, the blood–brain barrier (BBB) is thought to be a key player in the brain’s Aβ homeostasis [10]. It is now widely accepted that extracellular vesicles (EVs) may also be important in Aβ pathology [11,12,13,14,15,16,17]. Our earlier work has shown that HIV can increase the release of brain endothelial EVs and alter EV-Aβ levels. Moreover, brain endothelial cell-derived EVs can transfer Aβ to other cells of the neurovascular unit [18]. EVs carry specific cargo molecules, including a complex set of proteins, which can be transferred to the neighboring cells and affect their biology. Some of these proteins are on the EV surface. The surface proteins may allow for selective EV uptake by the recipient cells, like in the case of receptor-mediated endocytosis. Total proteomics can give detailed information on the EV protein cargo overall. Surface proteomics could indicate the “address” of a targeted delivery, while total proteomics would represent the delivered “package.”
In this work, we investigated how exposure to HIV, alone and together with Aβ, impacts the surface and total proteomic landscape of EVs from human brain microvascular endothelial cells (HBMEC-EVs). By using this unbiased strategy, we obtained a complex, high-resolution insight into these changes.

2. Results

2.1. Extracellular Vesicles from Human Brain Microvascular Endothelial Cells (HBMEC-EVs) Are Enriched with the Major EV Markers

At first, we examined whether proteins that are frequently identified in EVs/exosomes from various sources can be found in our isolated HBMEC-EVs. Based on the ExoCarta EV proteomics database from different human cell types that have been isolated using different approaches [19,20], we compiled the list of 100 marker proteins that are most often present on EVs (Table 1). The surface HBMEC-EV proteome, which contained a total of 283 identified proteins, included 62 of the top 100 ExoCarta EV markers (Figure 1A, Table 1). In addition, the total HBMEC-EV proteome, which contained 501 identified proteins, included 80 of such markers (Figure 1B, Table 1). These results demonstrate that our HBMEC-EV isolation was highly enriched with known EV markers.

2.2. Cellular Component Enrichment of the Identified Surface and Total EV Proteins

Using the Scaffold software, we next evaluated the HBMEC-EV proteins according to their known cellular localization. This approach may indicate the parent cellular compartment origin of the identified HBMEC-EV proteins. The majority of the HBMEC-EV surface proteins were extracellular region proteins, followed by cytoplasmic, intracellular organelle, membrane, nuclear, endoplasmic reticulum, cytoskeleton, Golgi, mitochondrial, endosomal, ribosomal proteins, and one unknown protein (Figure 1C). For the total HBMEC-EV proteome, the majority of proteins were cytoplasmic and extracellular region proteins (Figure 1D).

2.3. HIV and Aβ Exposure Results in Unique HBMEC-EV Proteome Signatures

We next focused on the unique proteins induced by the exposure to HIV and Aβ. Comparison of the control vs. HIV surface HBMEC-EV proteomes identified 112 unique proteins in the control and three unique proteins in the HIV group (Figure 2A). By contrast, a similar comparison for the total proteome identified only three unique proteins in the control and as many as 259 unique proteins in the HIV group (Figure 2B). Comparison of the surface proteome between the HIV vs. HIV+Aβ groups identified six unique proteins in the HIV group and 116 unique proteins in the HIV+Aβ group (Figure 2C). Finally, analysis of the total proteome revealed 28 unique proteins in the HIV group and 201 unique proteins in the HIV+Aβ group (Figure 2D). A list of these unique proteins is provided in Table 2 and Table 3 for the surface and total proteomes, respectively.

2.4. Functional Enrichment of the Unique HBMEC-EV Proteins

We next grouped these unique protein signatures into the biological process categories of the Scaffold software. Overall, 19 main categories were established, and the number of unique proteins mapping to these categories is illustrated in Figure 2, separately for the surface (A and C) and the total proteome (B and D). Note that individual proteins could map to more than one category; on the other hand, not all categories have been identified for all comparisons. This is consistent with the fact that selected group comparisons identified only a limited number of unique proteins that mapped to a limited number of categories. The number of unique proteins corresponding to the main biological process categories in the combined comparisons is illustrated on the bar graphs in Figure 2E for the surface proteome and Figure 2F for the total proteome. The majority of both surface and total unique proteins were mapped to “response to stimulus,” ”multicellular organismal process,” ”metabolic process,” and “localization” categories.
Next, we evaluated the unique proteins in the control vs. HIV and in the HIV vs. HIV+Aβ comparisons using STRING for functional enrichment in the biological processes and the Kyoto Encyclopedia of Genes and Genomes (KEGG) Pathways. In addition, we enriched these analyses for cellular components and PMID publications.
The results of these analyses for the EV surface proteome unique proteins in the control group in the control vs. HIV comparison are listed in Table 4 and Supplementary Table S1A. In addition, Supplementary Table S1B lists the enrichment for cellular components. The observed gene count (Obs), background gene count (Bgr), false discovery rate (FDR), and matched proteins are also included in these tables. The three unique proteins identified when comparing the surface proteome in the HIV group to the control group are dynein heavy chain 8, axonemal (DNAH8), titin (TTN), and immunoglobulin heavy constant gamma 2 (IGHG2). According to the description in the STRING or GeneCards database, DNAH8 is a force-generating protein of the respiratory cilia and is also involved in sperm motility. In addition, DNAH8 is highly expressed in prostate cancer [21]. Titin appears to be a key component of the vertebrate striated muscles [22]. IGHG2 may take part in antigen binding and the regulation of actin dynamics. It was linked to severe respiratory syncytial virus infection [23]. Overall, very limited or no data were found for the different enrichment analyses in STRING regarding these three proteins.
Next, we analyzed the EV surface proteome unique lists for the HIV vs. HIV+Aβ comparison in order to dissect the effect of exogenous EV-Aβ cargo in the context of HIV. In this analysis, six unique proteins were identified in the HIV group, namely, TTN, ninein (NIN), DNAH8, adenylyl cyclase-associated protein 1 (CAP1), actin-related protein 2/3 complex subunit 4 (ARPC4), and IGHG2. For these unique proteins, all enriched biological processes are shown in Table 5. No KEGG pathways were enriched; however, several PMID publications were found by textmining (Table 5). Cellular localization of these enriched proteins to only a few categories was found, namely, “cytoskeletal part” (ARPC4, CAP1, DNAH8, NIN, TTN), “actin cytoskeleton” (ARPC4, CAP1, TTN), “supramolecular fiber” (DNAH8, NIN, TTN), “microtubule” (DNAH8, NIN), “ciliary part” (DNAH8, NIN), and “cytoplasmic region” (CAP1, DNAH8).
For the unique proteins in the HIV+Aβ group in this comparison, the enriched biological processes, KEGG pathways, and PMID publications are presented in Table 6 and Supplementary Table S2A. The enrichment for cellular components is included in Supplementary Table S2B.
Next, we analyzed the EV total proteome unique lists for the control vs. HIV comparison. For the unique proteins in the control group, no gene ontology (GO) terms were found for biological processes. Similarly, no KEGG Pathways were enriched, likely because only three unique proteins were identified in this group and comparison. The cellular localization of these proteins is presented in Supplementary Table S3. In addition, the first 10 PMID publications enriched are shown in Table 7. The total proteome revealed 259 unique proteins in the HIV group that mapped to a variety of GO terms for biological processes (Table 8 and Supplementary Table S4A). They were also enriched in several KEGG pathways (Table 8) and assigned to diverse cellular components, as listed in Supplementary Table S4B. Textmining resulted in an unbiased PubMed search with the 10 most significant publications listed in Table 8.
Finally, we analyzed the list of the unique proteins present in the total HBMEC-EV proteome in the HIV and HIV+Aβ groups. The unique proteins in the HIV group in this comparison mapped to only one GO term for biological processes, namely, “cell envelope organization,” presented in Table 9. No KEGG pathways and no cellular components were enriched for this group. The first 10 textmined PMID citations are presented in Table 9. The unique proteins in the HIV+Aβ group were enriched to several biological processes, KEGG pathways, and PMID publications (Table 10 and Supplementary Table S5A). Supplementary Table S5B lists the enrichments for the cellular component in this group.

2.5. Analysis of Unique Protein Interactions

We also explored in STRING whether these unique proteins have functional interactions among each other. The statistical background assumed for this enrichment analysis was the whole human genome. We filtered our search for established interactions only for the input proteins, for the highest confidence (over 0.900), and for a static map without the protein structures. In the obtained interaction maps, different nodes are connected with colored lines depending on the functional association type. The results imply that the identified proteins have more interactions among themselves than what would be expected for a random set of proteins of similar size, drawn from the genome. Such enrichments indicate that the proteins are, at least partially, biologically connected as a group and may contribute jointly to shared functions.
The interactions of the 112 unique surface proteins in the control group as compared to the HIV group are illustrated in Figure 3A. The HIV group in this comparison had only three unique surface proteins (DNAH8, TTN, and IGHG2). Being present on the EV surface, these proteins may be prone to interact with their potential functional partners beyond the EV surface. Therefore, we examined their possible interactions not only with each other but with other proteins as well. The STRING program identified predicted functional partners for DNAH8 and TTN, and the top five candidates that were predicted with the highest confidence, as well as their interacting networks, are illustrated in Figure 3B.
Next, we evaluated the unique surface protein list in the HIV vs. HIV+Aβ group. No protein–protein interactions were found for the six proteins uniquely expressed in the HIV group. By contrast, the HIV+Aβ unique surface proteins had several complex interactions, as illustrated in Figure 3C.
Finally, we analyzed the interactions between the unique proteins present in the total HBMEC-EV proteome. No interactions were found in the control group as compared to the HIV group; however, the elaborate interaction map for the total unique proteins in the HIV group is presented in Figure 4A. For the HIV vs. HIV+Aβ comparison, the HIV group exhibited 28 unique proteins without any identified interactions. In contrast, the unique proteins in the HIV+Aβ group showed a complicated interaction network, as illustrated in Figure 4B.

3. Discussion

In the current study, we evaluated HBMEC-EV surface and total proteome changes evoked by HIV-1 alone and together with Aβ. We limited our analyses to the unique lists of proteins identified in the treatment groups; thus, we did not include the shared protein lists and the complex changes in the up- and down-regulated proteins. In addition, we specifically focused on the unique proteins in the control vs. HIV and in the HIV vs. HIV+Aβ group comparisons. The identified proteins were mapped to different gene ontology (GO) terms for biological processes, KEGG pathways, and Cell Components. We also explored the protein–protein interactions among the identified unique proteins.
Overall, the surface proteome control vs. HIV comparison indicated that the functions of the identified unique proteins ranged from diverse biological processes in the control (mainly “extracellular matrix organization,” “metabolic processes,” “vesicle-mediated transport,” “exocytosis”) and KEGG pathways (mainly “proteoglycans in cancer,” “focal adhesion,” “carbohydrate and cholesterol metabolism,” “HIF-1 signaling pathway”) to few or no distinct biological processes in the HIV group (Figure 2A and Table 4). The latter phenomenon was likely due to the limited number of proteins (namely, DNAH8, TTN, IGHG2) that were unique in the HIV-1 group when compared to the HBMEC-EV surface proteome of the controls. Nevertheless, we found several potential functional partners for DNAH8, such as platelet-activating factor acetylhydrolase IB subunit alpha (PAFAH1B1), dynactin subunit 1 (DCTN1), dynactin subunit 2 (DCTN2), CAP-Gly domain-containing linker protein 1 (CLIP1), and cytoplasmic dynein 1 light intermediate chain 1 (DYNC1LI1). Similarly, we identified several predicted functional partners for TTN, namely, nebulin (NEB), telethonin (TCAP), troponin C, skeletal muscle (TNNC2), myosin light chain 1/3, skeletal muscle isoform (MYL1), and alpha-actinin-2 (ACTN2) (Figure 3B). Thus, these few unique surface EV proteins in the HIV group may engage primarily with proteins of actin cytoskeleton/microtubule remodeling and vesicle-mediated transport.
The control EV proteome exhibited more than a hundred unique proteins; thus, it appears that after HIV-1 exposure of the parent cells, the EV surface proteome almost completely “blended” into the control proteome. This relative lack of surface HBMEC-EV protein signature in the HIV group is particularly striking in light of our previous findings where the exposure of HBMEC to HIV results in increased EV shedding [18] and the fact that EVs are involved in spreading HIV infection to the neighboring cells. However, it is possible that the localization of some proteins could alter from the EV surface to the vesicle lumen, resulting in a highly enriched total but not surface proteome. Indeed, comparison of the total proteome revealed a highly diverse number of 259 unique proteins in the HIV group as compared to the control that mapped to a variety of biological processes and KEGG pathways. The most prominent enrichment among the biological processes category was “vesicle-mediated transport,” followed by “extracellular structure organization.” In addition, mapping these unique proteins to “exocytosis” and “secretion by cell” categories points to processes that may be involved in EV release and EV transport (Figure 2B and Table 8). Likewise, the KEGG pathways were also diverse, from “focal adhesion” and “endothelial cell medium (ECM)-receptor interaction” to “proteoglycans in cancer,” different infections, “endocytosis,” “cholesterol metabolism,” and “glycolysis/gluconeogenesis” (Table 8). Thus, the total EV proteome in the HIV group, with a large number of unique proteins, may suggest that the rich, unique cargo is somewhat “hidden” within the EVs with a surface proteome that was barely altered. This notion is supported by the observations that the HIV group in the HIV versus HIV+Aβ group surface proteome comparison also exhibited only six unique proteins (Figure 2C). On the other hand, the relative lack of unique EV surface protein signatures may facilitate EV internalization and, thus, HIV transmission to other cells.
In addition to the effects of HIV-1, we explored the impact of Aβ on the HBMEC-EV proteome in the context of HIV-1. It was reported that increased brain Aβ induced profound proteome remodeling in multiple cell types, altering brain molecular pathways in an Alzheimer’s disease (AD) mouse model [24]. Another brain proteomic study using a different AD mouse model with amyloid and neurofibrillary tangle pathologies indicated age-dependent immune responses and synaptic dysfunctions. It was proposed that these changes were evoked by the advancing Aβ pathology in the brain [25], further demonstrating the importance of proteomic analyses in studies on the mechanisms of amyloid pathology.
Comparison of surface proteomes of EVs derived from HBMEC exposed to HIV alone vs. HIV+Aβ revealed profound changes, as demonstrated by 116 unique proteins in the HIV+Aβ group (Figure 2C). Aβ, acting on a HIV background, appeared to shift biological processes from mainly actin cytoskeleton organization (Table 5) to immune responses, extracellular matrix organization, and carbohydrate metabolic processes. In addition, enrichment of the “vesicle-mediated transport” and “exocytosis” also pointed to processes involved in EV release and EV transport (Figure 2C and Table 6). The KEGG pathways changed from a “blended” profile in the HIV group to a very diverse profile in the HIV+Aβ group, pointing mainly to the carbohydrate metabolic processes, “focal adhesion,” different infections, and signaling pathways as demonstrated by HIF-1, MAPK, and AGE-RAGE enrichment (Table 6). Regarding these signaling pathways, we have shown before the involvement of the RAGE pathway in the HIV-induced Aβ accumulation in HBMEC [26].
The HIV vs. HIV+Aβ comparison for the total proteome indicated substantial remodeling in the HIV+Aβ with 201 unique proteins as compared to 28 of such proteins in the HIV group. Consistent with HIV+Aβ-mediated EV release [18], the biological processes changed from “cell envelope organization” (Table 9) to mainly “vesicle-mediated transport,” “exocytosis,” and immune responses (Figure 2D and Table 10). The KEGG pathways also shifted to a diverse profile. “Endocytosis” was the most significant, followed by “focal adhesion” and “bacterial invasion of epithelial cells.” Several proteins were part of the carbohydrate metabolic pathways, such as the “pentose phosphate pathway,” “starch and sucrose metabolism,” and “proteoglycans in cancer” (Table 10).
Surprisingly, surface and total proteome analysis across different groups did not find any Aβ species in EVs, not even in samples that were isolated from Aβ-exposed HBMEC. This lack of Aβ identification could be related to technical issues, such as aggregation of Aβ, its insolubility, and possibly indigestibility by trypsin. The tryptic peptide used to quantify β-amyloid, LVFFAEDVGSNK, corresponding to amino acids 688–699, maps to all species of Aβ and full-length APP [27] and has been identified in the human CSF proteome [28]. In our study, no peptides mapping to the Aβ-generating region of APP were identified, even though APP was identified on the surface proteome. Similar obstacles were described in another proteomic study, in which Aβ was not identified in human AD brains. However, Aβ was detected by dot blot and ELISA from the same samples [29], supporting the notion that the lack of Aβ detection in the proteome was likely due to technical limitations.
Our previous studies demonstrated that treatment of HBMEC with Aβ could enrich EVs with this peptide, which can then be carried and delivered to different cells of the neurovascular unit [18,30]. In support of these findings, literature reports described Aβ as being present on the EV surface. For example, neuron-derived EVs accelerated Aβ fibril formation from monomeric Aβ, and this process was inhibited by cleavage of glycosphingolipid (GSL) glycans by endoglycoceramidase (EGCase) [31]. The same group also demonstrated that EV GSL-glycans were critical for Aβ binding in vitro and in vivo [15]. GSLs are found mainly in lipid rafts in the outer layer plasma membrane with their glycans facing outside; however, they are more abundant in EVs than in the parent cells [15]. Besides GSL, EVs were shown to bind Aβ through the prion protein (PrP) [14], a glycosylphosphatidylinositol-anchored protein in the outer leaflet of the neuron and neuron-derived EV membrane [32].
Some of the unique proteins identified in our HBMEC-derived EVs exhibit a substantial overlap with proteins detected by label-free proteomics in Aβ-enriched extracts from human AD brains [29], suggesting the relevance of EV proteins to Aβ pathology. The examples include ANXA5, FGB, LAMA5, and VIM found both in the total proteome of EVs in the HIV group and in Aβ-enriched extracts from human AD brains [29]. In addition, specific types of tubulins, such as TUBA1B and TUBB4B, were present, although they did not change in AD brains. Among the unique proteins in the HIV+Aβ group’s total proteome, FGG and HIST1H2BK, as well as tubulins TUBB and TUBB2A, were also enriched in extracts from AD brains [29]. In addition, HIST1H2BK has been one of the unique proteins in the EV total proteome from the Aβ group. In contrast, RNF213 was not identified in any of our EV samples, although it was unique to the AD brain samples and also found within the amyloid plaques [29]. One explanation for this phenomenon could be that RNF213 in the AD brain might not originate from brain endothelial cells.
Analysis for predicted significant functional interactions among the unique proteins produced several elaborate interaction maps (Figure 3 and Figure 4). It is striking to notice that several proteins on these maps act like “hubs” or centers by having a substantial number of connections to other proteins. Such “hubs” for the surface proteomes were SERPINE1 (PAI-1), GPC1, FERMT3 (Figure 3A), and ALDOA (Figure 3C). The most complex functional interaction maps were obtained for the total proteomes due to the high number of unique proteins. The identified “hubs” were RAC1, GAS6, SERPINE1, AGRN, APOB, and RAB5C (Figure 4A), as well as CDC42 and RAB1A (Figure 4B). Among these proteins, endothelial AGRN (agrin) was shown to be implicated in the brain Aβ pathology. For example, deletion of the Agrn gene from endothelial cells resulted in significantly increased Aβ levels in the mouse brain; however, overexpression of Agrn restored brain Aβ levels [33]. SERPINE1 (PAI-1) and GPC1 (glypican-1) may be additional important players in the Aβ pathology [34,35]. Indeed, GPC1, a heparan sulfate proteoglycan, localized mainly in detergent-insoluble, GSL-rich membrane domains, was shown to bind fibrillar Aβ in the human brain [36], further suggesting that protein “hubs” identified in the present study may be involved in EV-mediated Aβ pathology.
In summary, our results provide information, with an unprecedented resolution, on the brain endothelial surface and total EV proteome changes after HIV and Aβ exposure of the parent cells. The analyses identified protein–protein interaction networks, biological processes, pathways, and cellular localization. Overall, the obtained results factor for a better understanding of HBMEC-EV protein landscape changes induced by HIV and Aβ and their contribution to the HIV-associated Aβ pathology in the brain.

4. Materials and Methods

4.1. Cell Cultures

Primary human brain microvascular endothelial cells (HBMEC) used in the study were purchased from ScienCell Research laboratories (Carlsbad, CA, USA). HBMEC were isolated from human brain and cryopreserved at passage one. HBMEC were characterized by immunofluorescence with antibodies specific to vWF/Factor VIII and CD31 (PECAM). Cells were cultured on bovine plasma fibronectin (ScienCell)-coated dishes in endothelial cell medium (ECM). Specifically, 500 mL of basal ECM medium was supplemented with 25 mL of exosome-depleted fetal bovine serum (Exo-FBS; System Biosciences, Mountain View, CA, USA), 5 mL of endothelial cell growth supplement (ECGS, ScienCell), and 5 mL of penicillin/streptomycin solution (P/S, ScienCell). We initiated two separate cultures on 100 mm cell culture dishes to reduce the number of passages and subcultured the cells twice at the 1:4 ratio. This resulted in 32 confluent cultures, with the average cell number at the end of experiment of 9.065 × 107 cells/dish. Sixteen confluent cultures were used for EV surface proteomics, and 16 for EV total proteomics. The treatment groups were: 1) Control exposed to vehicle, 2) Aβ alone, 3) HIV alone, 4) HIV plus Aβ, with four samples/group.

4.2. HIV Infection and Aβ Treatment

HIV-1 stock was generated using human embryonic kidney (HEK) 293T cells (ATCC, Manassas, VA, USA) transfected with pYK-JRCSF plasmid containing full-length proviral DNA. Throughout the study, HBMEC were exposed to HIV particles at the p24 level of 30 ng/mL as previously reported [37]. Treatment was terminated by removing the cell culture media for EV isolation.
Aβ (1–40) was purchased from Anaspec (San Jose, CA, USA) and dissolved in PBS. Freshly solubilized Aβ solutions without pre-aggregation were used for experiments as such a form of Aβ was demonstrated to induce proinflammatory reactions in isolated rat brain microvessels [38]. Cells were treated with Aβ (1–40) at the concentration of 100 nM for 48 h in complete medium. Although uptake of Aβ by the BBB occurs rapidly [39], we terminated the treatment at 48 h to allow more EV to be secreted into the culture medium. Confluent HBMEC were exposed to HIV-1 or/and Aβ (1–40) for 48 h.

4.3. EV Isolation

EV isolation was performed using ExoQuick-TC precipitation solution (System Biosciences) from conditioned culture media according to the manufacturer’s specifications. Briefly, 10 mL culture media from confluent HBMEC cultures was centrifuged at 3000 g for 15 min to remove cells and debris, and then mixed thoroughly with 2 mL of Exo-Quick precipitation solution and incubated overnight at 4 °C. The next day, samples were centrifuged at 1500 g for 30 min, and the supernatants were removed and centrifuged again at 1500 g for 5 min. The EV pellets were stored at –80 °C and used for proteomics analysis. Separate EV samples were prepared for EV surface and total proteomics.

4.4. Proteomics

Sample Preparation. Each sample was processed by SDS-PAGE using a 10% Bis Tris NuPage mini-gel (Invitrogen, Waltham, MA, USA) in the MES buffer system. The migration windows (1 cm lane) were excised and processed by in-gel digestion with trypsin using a ProGest robot (DigiLab) with the following protocol: The samples were washed with 25 mM ammonium bicarbonate followed by acetonitrile, reduced with 10 mM dithiothreitol at 60 °C, followed by alkylation with 50 mM iodoacetamide at room temperature, digested with trypsin (Promega, Madison, WI, USA) at 37 °C for 4 h, and quenched with formic acid. The supernatants were then analyzed directly without further processing.
Mass Spectrometry. Half of each digested sample was analyzed by nano LC-MS/MS with a Waters NanoAcquity HPLC system interfaced to a ThermoFisher Q Exactive. Peptides were loaded on a trapping column and eluted over a 75 μm analytical column at 350 nL/min; both columns were packed with Luna C18 resin (Phenomenex, Torrance, CA, USA). The mass spectrometer was operated in data-dependent mode, with the Orbitrap operating at 70,000 FWHM and 17,500 FWHM for MS and MS/MS respectively. The fifteen most abundant ions were selected for MS/MS.
Data Processing. Data were searched using Mascot (Matrix Science, London, UK; version 2.6.0) with the following parameters: Enzyme: Trypsin/P; Databases: SwissProt Human (concatenated forward and reverse plus common contaminants); fixed modifications: Carbamidomethyl (C); variable modifications: Acetyl (N-term), deamidation (N,Q), oxidation (M), Pyro-Glu (N-term Q); mass values: Monoisotopic; peptide mass tolerance: 10 ppm; fragment mass tolerance: 0.02 Da; max missed cleavages: 2. Mascot DAT files were parsed into Scaffold (Proteome Software, version Scaffold 4.8.7, Proteome Software Inc., Portland, OR, USA) for validation, filtering, and to create a non-redundant list per sample. Data were filtered using a 1% protein and peptide FDR and required at least two unique peptides per protein. Protein probabilities were assigned by the Protein Prophet algorithm [40]. Proteins that contained similar peptides and could not be differentiated based on MS/MS analysis alone were grouped to satisfy the principles of parsimony. Proteins were annotated with GO terms from NCBI (downloaded on Sep 6, 2018) [41]. The normalized spectral abundance factor (NSAF) calculation contains the conversion to the spectral abundance factor (SAF) and subsequent normalized spectral abundance factor (NSAF). This was based on the equation: NSAF = (SpC/MW)/Σ(SpC/MW)N, where SpC = spectral counts, MW = protein molecular weight in kDa, and N = total number of proteins. NSAF values can be used to approximate the relative abundance of proteins within a given sample and the relative abundance of a given protein between samples. The different treatment groups were compared using the t-test, and p < 0.05 was considered significant.

4.5. ExoCarta Database Search and Functional Enrichment Analysis

The list of the top 100 proteins most often identified in EVs was composed based on the ExoCarta EV proteomics database from different human cell types [19]. Enrichment in molecular functions of the identified EV proteins was analyzed using the Scaffold Proteome Software and STRING [42]. A gene ontology analysis study was carried out with the proteomic profiles obtained to identify overrepresentation profiles. Gene ontology was investigated at the levels of the biological process, KEGG pathways, and cell component. Textmining in STRING provided the most relevant publications for a particular enrichment. Kyoto Encyclopedia of Genes and Genomes (KEGG) established pathway maps representing molecular interactions, reactions, and relation networks for Metabolism, Genetic Information Processing, Environmental Information Processing, Cellular Processes, Organismal Systems, Human Diseases and Drug Development. KEGG PATHWAY is the reference database for pathway mapping in KEGG Mapper.

Supplementary Materials

The following are available online at https://www.mdpi.com/1422-0067/21/8/2741/s1.

Author Contributions

Conceptualization, I.E.A. and M.T.; Formal analysis, I.E.A. and B.B.S.; Funding acquisition, M.T.; Investigation, I.E.A. and B.B.S.; Supervision, I.E.A. and M.T.; Visualization, I.E.A.; Writing—original draft, I.E.A.; Writing—review & editing, M.T. All authors have read and agreed to the published version of the manuscript.

Funding

Supported by the Florida Department of Health grant 8AZ24 and the National Institutes of Health (NIH), grants MH072567, MH098891, HL126559, DA039576, DA040537, DA044579, and DA047157. We acknowledge support from the Miami Center for AIDS Research (CFAR) at the University of Miami Miller School of Medicine, which is funded by a grant (P30AI073961) from the National Institutes of Health (NIH). We would like to thank Joshua Zahner, undergraduate student at the University of Miami, for his help in sorting the unique protein lists.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

amyloid beta
ADAlzheimer’s disease
BBBBlood–brain barrier
ECGSEndothelial cell growth supplement
EVExtracellular vesicle
ELISAEnzyme-linked immunosorbent assay
HANDHIV-associated neurocognitive disorders;
HBMECHuman brain microvascular endothelial cells
HEK cellsHuman embryonic kidney cells
HIVHuman immunodeficiency virus type 1
PBSPhosphate buffered saline
PECAMPlatelet endothelial cell adhesion molecule
RAGEReceptor for advanced glycation end products

References

  1. Esiri, M.M.; Biddolph, S.C.; Morris, C.S. Prevalence of Alzheimer plaques in AIDS. J. Neurol. Neurosurg. Psychiatry 1998, 65, 29–33. [Google Scholar] [CrossRef] [PubMed]
  2. Rempel, H.C.; Pulliam, L. HIV-1 Tat inhibits neprilysin and elevates amyloid beta. Aids 2005, 19, 127–135. [Google Scholar] [CrossRef] [PubMed]
  3. Xu, J.; Ikezu, T. The comorbidity of HIV-associated neurocognitive disorders and Alzheimer’s disease: A foreseeable medical challenge in post-HAART era. J. Neuroimmune Pharmacol. Off. J. Soc. NeuroImmune Pharmacol. 2009, 4, 200–212. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Brew, B.J.; Crowe, S.M.; Landay, A.; Cysique, L.A.; Guillemin, G. Neurodegeneration and ageing in the HAART era. J. Neuroimmune Pharmacol. Off. J. Soc. NeuroImmune Pharmacol. 2009, 4, 163–174. [Google Scholar] [CrossRef]
  5. Achim, C.L.; Adame, A.; Dumaop, W.; Everall, I.P.; Masliah, E.; Neurobehavioral Research, C. Increased accumulation of intraneuronal amyloid beta in HIV-infected patients. J. Neuroimmune Pharmacol. Off. J. Soc. NeuroImmune Pharmacol. 2009, 4, 190–199. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Pulliam, L. HIV regulation of amyloid beta production. J. Neuroimmune Pharmacol. Off. J. Soc. NeuroImmune Pharmacol. 2009, 4, 213–217. [Google Scholar] [CrossRef]
  7. Green, D.A.; Masliah, E.; Vinters, H.V.; Beizai, P.; Moore, D.J.; Achim, C.L. Brain deposition of beta-amyloid is a common pathologic feature in HIV positive patients. Aids 2005, 19, 407–411. [Google Scholar] [CrossRef]
  8. Soontornniyomkij, V.; Moore, D.J.; Gouaux, B.; Soontornniyomkij, B.; Tatro, E.T.; Umlauf, A.; Masliah, E.; Levine, A.J.; Singer, E.J.; Vinters, H.V.; et al. Cerebral beta-amyloid deposition predicts HIV-associated neurocognitive disorders in APOE epsilon4 carriers. Aids 2012, 26, 2327–2335. [Google Scholar] [CrossRef] [Green Version]
  9. Steinbrink, F.; Evers, S.; Buerke, B.; Young, P.; Arendt, G.; Koutsilieri, E.; Reichelt, D.; Lohmann, H.; Husstedt, I.W.; German Competence Network, H.A. Cognitive impairment in HIV infection is associated with MRI and CSF pattern of neurodegeneration. Eur. J. Neurol. 2013, 20, 420–428. [Google Scholar] [CrossRef]
  10. Deane, R.; Zlokovic, B.V. Role of the blood-brain barrier in the pathogenesis of Alzheimer’s disease. Curr. Alzheimer Res. 2007, 4, 191–197. [Google Scholar] [CrossRef]
  11. Vella, L.J.; Sharples, R.A.; Nisbet, R.M.; Cappai, R.; Hill, A.F. The role of exosomes in the processing of proteins associated with neurodegenerative diseases. Eur. Biophys. J. EBJ 2008, 37, 323–332. [Google Scholar] [CrossRef] [PubMed]
  12. Kalani, A.; Tyagi, A.; Tyagi, N. Exosomes: Mediators of neurodegeneration, neuroprotection and therapeutics. Mol. Neurobiol. 2014, 49, 590–600. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Gupta, A.; Pulliam, L. Exosomes as mediators of neuroinflammation. J. Neuroinflammation 2014, 11, 68. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. An, K.; Klyubin, I.; Kim, Y.; Jung, J.H.; Mably, A.J.; O’Dowd, S.T.; Lynch, T.; Kanmert, D.; Lemere, C.A.; Finan, G.M.; et al. Exosomes neutralize synaptic-plasticity-disrupting activity of Abeta assemblies in vivo. Mol. Brain 2013, 6, 47. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Yuyama, K.; Sun, H.; Sakai, S.; Mitsutake, S.; Okada, M.; Tahara, H.; Furukawa, J.; Fujitani, N.; Shinohara, Y.; Igarashi, Y. Decreased amyloid-beta pathologies by intracerebral loading of glycosphingolipid-enriched exosomes in Alzheimer model mice. J. Biol. Chem. 2014, 289, 24488–24498. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Yuyama, K.; Sun, H.; Usuki, S.; Sakai, S.; Hanamatsu, H.; Mioka, T.; Kimura, N.; Okada, M.; Tahara, H.; Furukawa, J.; et al. A potential function for neuronal exosomes: Sequestering intracerebral amyloid-beta peptide. FEBS Lett. 2015, 589, 84–88. [Google Scholar] [CrossRef]
  17. Dinkins, M.B.; Dasgupta, S.; Wang, G.; Zhu, G.; He, Q.; Kong, J.N.; Bieberich, E. The 5XFAD Mouse Model of Alzheimer’s Disease Exhibits an Age-Dependent Increase in Anti-Ceramide IgG and Exogenous Administration of Ceramide Further Increases Anti-Ceramide Titers and Amyloid Plaque Burden. J. Alzheimer’s Dis. JAD 2015, 46, 55–61. [Google Scholar] [CrossRef] [Green Version]
  18. Andras, I.E.; Leda, A.; Contreras, M.G.; Bertrand, L.; Park, M.; Skowronska, M.; Toborek, M. Extracellular vesicles of the blood-brain barrier: Role in the HIV-1 associated amyloid beta pathology. Mol. Cell. Neurosci. 2017, 79, 12–22. [Google Scholar] [CrossRef] [Green Version]
  19. Keerthikumar, S.; Chisanga, D.; Ariyaratne, D.; Al Saffar, H.; Anand, S.; Zhao, K.; Samuel, M.; Pathan, M.; Jois, M.; Chilamkurti, N.; et al. ExoCarta: A Web-Based Compendium of Exosomal Cargo. J. Mol. Biol. 2016, 428, 688–692. [Google Scholar] [CrossRef] [Green Version]
  20. Mathivanan, S.; Fahner, C.J.; Reid, G.E.; Simpson, R.J. ExoCarta 2012: Database of exosomal proteins, RNA and lipids. Nucleic Acids Res. 2012, 40, D1241–D1244. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  21. Wang, Y.; Ledet, R.J.; Imberg-Kazdan, K.; Logan, S.K.; Garabedian, M.J. Dynein axonemal heavy chain 8 promotes androgen receptor activity and associates with prostate cancer progression. Oncotarget 2016, 7, 49268–49280. [Google Scholar] [CrossRef] [PubMed]
  22. Chauveau, C.; Rowell, J.; Ferreiro, A. A rising titan: TTN review and mutation update. Hum. Mutat. 2014, 35, 1046–1059. [Google Scholar] [CrossRef] [PubMed]
  23. Aurivillius, M.; Oymar, K.; Oxelius, V.A. Immunoglobulin heavy G2 chain (IGHG2) gene restriction in the development of severe respiratory syncytial virus infection. Acta Paediatr. 2005, 94, 414–418. [Google Scholar] [CrossRef] [PubMed]
  24. Savas, J.N.; Wang, Y.Z.; DeNardo, L.A.; Martinez-Bartolome, S.; McClatchy, D.B.; Hark, T.J.; Shanks, N.F.; Cozzolino, K.A.; Lavallee-Adam, M.; Smukowski, S.N.; et al. Amyloid Accumulation Drives Proteome-wide Alterations in Mouse Models of Alzheimer’s Disease-like Pathology. Cell Rep. 2017, 21, 2614–2627. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Kim, D.K.; Park, J.; Han, D.; Yang, J.; Kim, A.; Woo, J.; Kim, Y.; Mook-Jung, I. Molecular and functional signatures in a novel Alzheimer’s disease mouse model assessed by quantitative proteomics. Mol. Neurodegener. 2018, 13, 2. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Andras, I.E.; Eum, S.Y.; Huang, W.; Zhong, Y.; Hennig, B.; Toborek, M. HIV-1-induced amyloid beta accumulation in brain endothelial cells is attenuated by simvastatin. Mol. Cell. Neurosci. 2010, 43, 232–243. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Yu, L.; Petyuk, V.A.; Tasaki, S.; Boyle, P.A.; Gaiteri, C.; Schneider, J.A.; De Jager, P.L.; Bennett, D.A. Association of Cortical beta-Amyloid Protein in the Absence of Insoluble Deposits With Alzheimer Disease. JAMA Neurol. 2019, 76, 818–826. [Google Scholar] [CrossRef] [PubMed]
  28. Macron, C.; Lane, L.; Nunez Galindo, A.; Dayon, L. Deep Dive on the Proteome of Human Cerebrospinal Fluid: A Valuable Data Resource for Biomarker Discovery and Missing Protein Identification. J. Proteome Res. 2018, 17, 4113–4126. [Google Scholar] [CrossRef]
  29. Pedrero-Prieto, C.M.; Flores-Cuadrado, A.; Saiz-Sanchez, D.; Ubeda-Banon, I.; Frontinan-Rubio, J.; Alcain, F.J.; Mateos-Hernandez, L.; de la Fuente, J.; Duran-Prado, M.; Villar, M.; et al. Human amyloid-beta enriched extracts: Evaluation of in vitro and in vivo internalization and molecular characterization. Alzheimer’s Res. Ther. 2019, 11, 56. [Google Scholar] [CrossRef]
  30. Andras, I.E.; Toborek, M. Extracellular vesicles of the blood-brain barrier. Tissue Barriers 2016, 4, e1131804. [Google Scholar] [CrossRef] [Green Version]
  31. Yuyama, K.; Sun, H.; Mitsutake, S.; Igarashi, Y. Sphingolipid-modulated exosome secretion promotes clearance of amyloid-beta by microglia. J. Biol. Chem. 2012, 287, 10977–10989. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Lauren, J.; Gimbel, D.A.; Nygaard, H.B.; Gilbert, J.W.; Strittmatter, S.M. Cellular prion protein mediates impairment of synaptic plasticity by amyloid-beta oligomers. Nature 2009, 457, 1128–1132. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Rauch, S.M.; Huen, K.; Miller, M.C.; Chaudry, H.; Lau, M.; Sanes, J.R.; Johanson, C.E.; Stopa, E.G.; Burgess, R.W. Changes in brain beta-amyloid deposition and aquaporin 4 levels in response to altered agrin expression in mice. J. Neuropathol. Exp. Neurol. 2011, 70, 1124–1137. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Liu, R.M.; van Groen, T.; Katre, A.; Cao, D.; Kadisha, I.; Ballinger, C.; Wang, L.; Carroll, S.L.; Li, L. Knockout of plasminogen activator inhibitor 1 gene reduces amyloid beta peptide burden in a mouse model of Alzheimer’s disease. Neurobiol. Aging 2011, 32, 1079–1089. [Google Scholar] [CrossRef] [Green Version]
  35. Bi Oh, S.; Suh, N.; Kim, I.; Lee, J.Y. Impacts of aging and amyloid-beta deposition on plasminogen activators and plasminogen activator inhibitor-1 in the Tg2576 mouse model of Alzheimer’s disease. Brain Res. 2015, 1597, 159–167. [Google Scholar] [CrossRef]
  36. Watanabe, N.; Araki, W.; Chui, D.H.; Makifuchi, T.; Ihara, Y.; Tabira, T. Glypican-1 as an Abeta binding HSPG in the human brain: Its localization in DIG domains and possible roles in the pathogenesis of Alzheimer’s disease. FASEB J. Off. Publ. Fed. Am. Soc. Exp. Biol. 2004, 18, 1013–1015. [Google Scholar]
  37. Andras, I.E.; Toborek, M. HIV-1 stimulates nuclear entry of amyloid beta via dynamin dependent EEA1 and TGF-beta/Smad signaling. Exp. Cell Res. 2014, 323, 66–76. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. Paris, D.; Townsend, K.P.; Obregon, D.F.; Humphrey, J.; Mullan, M. Pro-inflammatory effect of freshly solubilized beta-amyloid peptides in the brain. Prostaglandins Other Lipid Mediat. 2002, 70, 1–12. [Google Scholar] [CrossRef]
  39. Yamada, K.; Hashimoto, T.; Yabuki, C.; Nagae, Y.; Tachikawa, M.; Strickland, D.K.; Liu, Q.; Bu, G.; Basak, J.M.; Holtzman, D.M.; et al. The low density lipoprotein receptor-related protein 1 mediates uptake of amyloid beta peptides in an in vitro model of the blood-brain barrier cells. J. Biol. Chem. 2008, 283, 34554–34562. [Google Scholar] [CrossRef] [Green Version]
  40. Nesvizhskii, A.I.; Keller, A.; Kolker, E.; Aebersold, R. A statistical model for identifying proteins by tandem mass spectrometry. Anal. Chem. 2003, 75, 4646–4658. [Google Scholar] [CrossRef]
  41. Ashburner, M.; Ball, C.A.; Blake, J.A.; Botstein, D.; Butler, H.; Cherry, J.M.; Davis, A.P.; Dolinski, K.; Dwight, S.S.; Eppig, J.T.; et al. Gene ontology: Tool for the unification of biology. The Gene Ontology Consortium. Nat. Genet. 2000, 25, 25–29. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Szklarczyk, D.; Morris, J.H.; Cook, H.; Kuhn, M.; Wyder, S.; Simonovic, M.; Santos, A.; Doncheva, N.T.; Roth, A.; Bork, P.; et al. The STRING database in 2017: Quality-controlled protein-protein association networks, made broadly accessible. Nucleic Acids Res. 2017, 45, D362–D368. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Extracellular vesicle (EV)-specific markers in the surface and total proteomes of human brain microvascular endothelial cells (HBMEC)-derived EVs. Venn diagram showing the overlap between the HBMEC-EV surface proteome (283 proteins) (A) or the HBMEC-EV total proteome (501 proteins) (B) and the top 100 EV marker proteins from ExoCarta. Cellular component enrichment of the identified surface (C) and total (D) EV proteomes. The identified EV proteins were enriched for cellular component using the Scaffold software.
Figure 1. Extracellular vesicle (EV)-specific markers in the surface and total proteomes of human brain microvascular endothelial cells (HBMEC)-derived EVs. Venn diagram showing the overlap between the HBMEC-EV surface proteome (283 proteins) (A) or the HBMEC-EV total proteome (501 proteins) (B) and the top 100 EV marker proteins from ExoCarta. Cellular component enrichment of the identified surface (C) and total (D) EV proteomes. The identified EV proteins were enriched for cellular component using the Scaffold software.
Ijms 21 02741 g001aIjms 21 02741 g001b
Figure 2. Enrichment for biological processes of the identified unique EV proteins. Scaffold software was used to enrich for the main biological processes for the identified unique EV proteins. The upper Venn diagrams show the compared groups with the number of their unique and shared proteins. The lower pie charts depict the enriched biological processes corresponding to the unique lists highlighted in yellow. The number of proteins in a particular biological process category is also provided. (A) Surface proteome, control vs. HIV. (B) Total proteome, control vs. HIV. (C) Surface proteome, HIV vs. HIV+ amyloid beta (Aβ). (D) Total proteome, HIV vs. HIV+Aβ. Combined graph for the biological processes in the EV unique surface (E) and total (F) proteomes. The number of unique proteins corresponding to the main biological processes in the different comparisons is illustrated on the graph.
Figure 2. Enrichment for biological processes of the identified unique EV proteins. Scaffold software was used to enrich for the main biological processes for the identified unique EV proteins. The upper Venn diagrams show the compared groups with the number of their unique and shared proteins. The lower pie charts depict the enriched biological processes corresponding to the unique lists highlighted in yellow. The number of proteins in a particular biological process category is also provided. (A) Surface proteome, control vs. HIV. (B) Total proteome, control vs. HIV. (C) Surface proteome, HIV vs. HIV+ amyloid beta (Aβ). (D) Total proteome, HIV vs. HIV+Aβ. Combined graph for the biological processes in the EV unique surface (E) and total (F) proteomes. The number of unique proteins corresponding to the main biological processes in the different comparisons is illustrated on the graph.
Ijms 21 02741 g002aIjms 21 02741 g002bIjms 21 02741 g002c
Figure 3. Protein–protein interactions between the identified unique proteins of the EV surface proteome. Venn diagrams illustrating the type of comparison and the number of identified unique proteins (highlighted). (A) Protein–protein interactions (PPI) (STRING) among the unique surface proteins in the control group. Only interactions with the highest confidence are shown with a minimum required interaction score of 0.900 (PPI enrichment p-value: 6.59 × 10−7; the network has significantly more interactions than expected). Known interactions: From curated databases (turquoise), experimentally determined (pink); predicted interactions: Gene neighborhood (green), gene fusions (red), gene co-occurrence (blue); other interactions: Textmining (light green), co-expression (black), protein homology (purple). (B) No interactions with highest confidence were identified in STRING among the three unique proteins identified in the HIV group. Predicted functional partners of dynein heavy chain 8, axonemal (DNAH8) (upper map) and titin (TTN) (lower map). Only the first shell of five interactions with the highest confidence is shown. Color code of the interaction lines as described in (A). (C) Protein–protein interactions among the unique proteins in the HIV+Aβ group. Only interactions with the highest confidence are shown (PPI enrichment p-value: 0.00158; the network has significantly more interactions than expected). Color code of the interaction lines as described in (A).
Figure 3. Protein–protein interactions between the identified unique proteins of the EV surface proteome. Venn diagrams illustrating the type of comparison and the number of identified unique proteins (highlighted). (A) Protein–protein interactions (PPI) (STRING) among the unique surface proteins in the control group. Only interactions with the highest confidence are shown with a minimum required interaction score of 0.900 (PPI enrichment p-value: 6.59 × 10−7; the network has significantly more interactions than expected). Known interactions: From curated databases (turquoise), experimentally determined (pink); predicted interactions: Gene neighborhood (green), gene fusions (red), gene co-occurrence (blue); other interactions: Textmining (light green), co-expression (black), protein homology (purple). (B) No interactions with highest confidence were identified in STRING among the three unique proteins identified in the HIV group. Predicted functional partners of dynein heavy chain 8, axonemal (DNAH8) (upper map) and titin (TTN) (lower map). Only the first shell of five interactions with the highest confidence is shown. Color code of the interaction lines as described in (A). (C) Protein–protein interactions among the unique proteins in the HIV+Aβ group. Only interactions with the highest confidence are shown (PPI enrichment p-value: 0.00158; the network has significantly more interactions than expected). Color code of the interaction lines as described in (A).
Ijms 21 02741 g003aIjms 21 02741 g003b
Figure 4. Protein–protein interactions in the identified unique proteins of the EV total proteome. Venn diagrams illustrating the type of comparison and the number of identified unique proteins (highlighted). (A) Protein–protein interactions among the unique proteins in the HIV group. Only interactions with the highest confidence are shown (PPI enrichment p-value: 1.0 × 10−16; the network has significantly more interactions than expected). (B) Protein–protein interactions among the unique proteins in the HIV+Aβ group. Only interactions with the highest confidence are shown (PPI enrichment p-value: 1.45 × 10−7; the network has significantly more interactions than expected). Color code of the interaction lines as described in Figure 3A.
Figure 4. Protein–protein interactions in the identified unique proteins of the EV total proteome. Venn diagrams illustrating the type of comparison and the number of identified unique proteins (highlighted). (A) Protein–protein interactions among the unique proteins in the HIV group. Only interactions with the highest confidence are shown (PPI enrichment p-value: 1.0 × 10−16; the network has significantly more interactions than expected). (B) Protein–protein interactions among the unique proteins in the HIV+Aβ group. Only interactions with the highest confidence are shown (PPI enrichment p-value: 1.45 × 10−7; the network has significantly more interactions than expected). Color code of the interaction lines as described in Figure 3A.
Ijms 21 02741 g004aIjms 21 02741 g004b
Table 1. List of the top 100 ExoCarta proteins present in the brain endothelial extracellular vesicle (EV) surface (S) and total (T) proteome. Bold, top 100 ExoCarta proteins present in S or T; bold and red, proteins present in both S and T.
Table 1. List of the top 100 ExoCarta proteins present in the brain endothelial extracellular vesicle (EV) surface (S) and total (T) proteome. Bold, top 100 ExoCarta proteins present in S or T; bold and red, proteins present in both S and T.
Gene SymbolDetected in SDetected in T
1CD9+
2HSPA8++
3PDCD6IP++
4GAPDH++
5ACTB++
6ANXA2++
7CD63+
8SDCBP++
9ENO1++
10HSP90AA1++
11TSG101+
12PKM++
13LDHA++
14EEF1A1++
15YWHAZ++
16PGK1++
17EEF2++
18ALDOA++
19HSP90AB1++
20ANXA5++
21FASN++
22YWHAE++
23CLTC++
24CD81+
25ALB++
26VCP++
27TPI1++
28PPIA++
29MSN++
30CFL1++
31PRDX1++
32PFN1++
33RAP1B++
34ITGB1++
35HSPA5++
36SLC3A2+
37HIST1H4A++
38GNB2
39ATP1A1+
40YWHAQ++
41FLOT1
42FLNA++
43CLIC1++
44CDC42++
45CCT2++
46A2M++
47YWHAG++
48TUBA1B++
49RAC1+
50LGALS3BP++
51HSPA1A++
52GNAI2++
53ANXA1++
54RHOA
55MFGE8+
56PRDX2+
57GDI2++
58EHD4+
59ACTN4++
60YWHAB
61RAB7A+
62LDHB++
63GNAS
64TFRC
65RAB5C+
66ARF1
67ANXA6++
68ANXA11+
69ACTG1
70KPNB1++
71EZR+
72ANXA4
73ACLY++
74TUBA1C
75RAB14+
76HIST2H4A
77GNB1++
78UBA1++
79THBS1++
80RAN++
81RAB5A
82PTGFRN++
83CCT5++
84CCT3+
85BSG+
86AHCY++
87RAB5B
88RAB1A+
89LAMP2+
90ITGA6
91HIST1H4B
92GSN++
93FN1++
94YWHAH+
95TUBA1A+
96TKT
97TCP1++
98STOM+
99SLC16A1
100RAB8A
Table 2. List of the unique proteins in the EV surface proteome.
Table 2. List of the unique proteins in the EV surface proteome.
Control vs. HIVHIV vs. HIV+Aβ
Control UniqueHIV UniqueHIV UniqueHIV+Aβ Unique
1433E GPC6 TGM1 TITIN
DYH8
IGHG2
TITIN
NIN
DYH8
CAP1
ARPC4
IGHG2
1433E GPC1TCPA
1433G GSTP1 TIG1 1433G GPC6TCPB
1433T IGL1 TIMP3 1A34GSTA5TCPE
1A34 ITA3 TPM4 1B15GSTP1TCPH
5NTD ITAV TRFE 5NTDHMCN1TCPZ
6PGD ITB1 TSP4 ACLYIGL1TIG1
ACLY K2C6B UBB ACTCIMB1TPM4
ACTC LAMA1 UGPA ADA10ITA3TRFE
ALDOA LCAT URP2 AL9A1ITA5TSP4
AMPN LDHA VINC ALDOAITAVUBA1
AMY1 LDHB WDR1 AMY1ITB1UBB
ANXA1 LOXL2 WNT5A ANXA1LDHAUGPA
APOA4 LRC17 ARF3LDHBURP2
ARF3 LRP1 ARP2LOXL2VINC
ARGI1 LTBP1 ARP3LRC17WDR1
ARP2 MIME ASPMLRP1WNT5A
ARP3 MMP2 ATL1LTBP1
ATS13 MPRI ATX2MIME
C1S MYL6 B4GA1MMP2
CASPE NID2 C1SNID2
CCD80 P3H1 CAZA1P3H1
CFAH PAI1 CCD80PAI1
CHIA PCOC1 CDC42PDC6I
CLIC1 PDC6I CHIAPDIA3
CO4A2 PDIA3 CHSS2PGK1
CO5A2 PGK1 CISYPGM1
CO7 PLEC CLIC1PLEC
CO7A1 PLOD3 CLUSPLOD3
COBA1 PPIA CO4A2PPIA
COF1 PRDX4 CO5A2PRDX2
COFA1 PYGB CO7A1PRDX6
COMP RAB1B COBA1PUR6
EF1G RACK1 COF1PYGB
ENOB RAP1B COFA1PYGL
EXT1 RLA0 COMPRACK1
EXT2 RS16 EF1GRAP1B
F13A S10A9 EXT1RGN
FA11 SDCB1 F13ARIMB1
FAS SEPR FA11RL12
FBLN1 SPB12 FASS10A9
FBN1SPR1B FBLN1SDCB1
FBN2 SPR2E FBN1SERA
FLNB SRCRL FBN2SERPH
FLNC SRPX2 FLNBSPR2E
FPRP SULF1 FLNCSRCRL
FRIH SULF2 FPRPSRPX2
FSCN1 SYTC FRIHSULF1
GAS6 TAGL2 FRILSULF2
GNAI2 TBA1A FSCN1SYTC
GPC1 TCPD GDIBTAGL2
Table 3. List of the unique proteins in the EV total proteome.
Table 3. List of the unique proteins in the EV total proteome.
Control vs. HIV
Control UniqueHIV Unique
ACTC
MYH1
TAU
1433T CD81 GDIB MVP S10AB URP2
1433Z CD82 GELS MYH9 SAHH VIME
1A24 CLH1 GGCT MYL6 SCRB2 VINC
5NTD CLIC1 GNAI2 MYOF SDC4 VPS35
6PGD CO1A2 GPC6 NID1 SDCB1 VTNC
A4 CO3A1 GRP78 NID2 SEPR VWF
ACLY CO4A1 GSLG1 NNMT SERPH WDR1
ACTN1 CO4A2 GTR1 OLFL3 SND1 WNT5A
ACTN4 CO5 H31 PAI1 SNED1 ZA2G
ADA10 CO5A1 H4 PDC6I SPTB2
AEBP1 CO5A2 HEP2 PDIA3 SPTN1
AGRIN CO6A2 HS90A PFKAP SRCRL
AHNK CO7A1 HS90B PGK1 SRGN
ALDOA CO9 HSP7C PGS1 SRPX
AMPN COEA1 HTRA1 PGS2 SRPX2
AMY1 COFA1 IF4A1 PKP1 SULF1
ANT3 COIA1 IGHA1 PLEC SULF2
ANX11 COMP ITA3 PLMN SYDC
ANXA1 CYTA ITA4 PLOD1 SYTC
ANXA5 DPYL2 ITA5 PLOD3 TAGL2
ANXA6 DYHC1 ITAV PLS1 TBA1B
AP2A1 EF1A1 ITB1 PLS3 TBB4B
AP2M1 EF2 ITIH1 PPIB TCPA
APLP2 EHD1 ITIH3 PRC2A TCPB
APOA4 EHD2 ITIH4 PRDX1 TCPD
APOB EMIL1 KLK7 PRDX6 TCPH
APOE ENOA KPYM PROF1 TCPQ
ARF3 ENPL LAMA1 PSB5 TCPZ
ARF4 EXT1 LAMA2 PTX3 TENA
ARGI1 EXT2 LAMA4 PXDN TERA
ARP2 F13A LAMA5 RAB5C TGM1
ARPC2 FA5 LAMP1 RAB7A TGM2
AT1A1 FAS LAMP2 RAC1 TGM3
ATL1 FAT1 LDHA RACK1 THBG
ATS12 FBLN1 LDHB RAN THRB
ATS13 FBN1 LEG1 RAP1B THY1
ATX2 FBX50 LORI RB11A TIG1
B4GA1 FETA LOXL2 RHOC TITIN
BGH3 FIBB LRC17 RIMB1 TLN1
BMP1 FILA LRP1 RL10A TPIS
C1QT3 FLNA LTBP1 RL12 TPM4
C1S FLNB LTBP2 RL13A TRFE
CAP1 FLNC LYSC RL27 TRFL
CASPE FPRP MAMC2 RL6 TSN14
CATA FSCN1 MARCS RS16 TSP 1
CCD80 G3P MFGM RS3 TSP 2
CD151 GALK1 MMP2 RS4X TSP 3
CD44 GAS6 MOES RS8 TTYH3
CD59 GBB1 MOT4 RSSA UBA1
CD63 GBG12 MRC2 S10A9 UBB
HIV vs. HIV+Aβ
HIV uniqueHIV+Aβ unique
AHNK 1433E CO8B MIME RL3
ARGI1 1433F COF1 MOB1B RL7
ATX2 1433G COPB2 MPRI RL7A
B4GA1 1B40 COR1A MRP RLA0
CASPE 2AAA COR1C MXRA5 RS11
CATA 4F2 CTL1 MYH16 RS18
CYTA ACTC CTL2 NDKA RS2
FBX50 AK1A1 CTND1 NEP RS20
FILA AL9A1 CYFP1 NIBL1 RS25
GGCT ALS DHX9 NOTC3 RS3A
HORN ANGL2 DSG4 NRP1 RS9
IGHA1 ANGL4 DX39B OLM2B RTN4
K1C13 ANM1 ECM1 P3H1 RUVB1
KLK7 ANR31 ECM2 PAMR1 SC23A
LORI AP1G1 EEA1 PARVB SCUB3
LYSC AP2B1 EF1G PCOC1 SEM3C
MYOF APOM EGLN PDIA1 SEP11
PLS1 ARF6 EHD4 PDIA6 SEPT2
PRC2A ARP3 EIF3A PDLI5 SERA
RIMB1 ARPC4 EZRI PGFRB SLIT2
RL27 ASSY FA10 PGM1 STOM
S10A9 AT1B3 FA11 PIP SVEP1
SNED1 ATPA FBN2 PLOD2 SYFB
SPB12 ATPB FIBG PP1B SYHC
TGM1 ATS7 FRIH PPIA SYK
TGM3 B4GT5 G6PD PRS23 SYRC
TITIN BASI G6PI PRS8 SYSC
ZA2G BASP1 GANAB PSA3 TARSH
BGAL H13 PSA6 TBB2A
C1R H2A1 PSD11 TBB5
C1TC H2B1K PSD12 TCPE
CAD23 HGFL PSD13 TCPG
CALR HHIP PSMD1 TGFB1
CAND1 HMCN1 PSMD2 TICN1
CAPZB HNRPK PSMD3 TIE1
CAV1 IGSF8 PUR6 TIMP3
CAZA1 ILK PYGB TS101
CBPN IMB1 PYGL TSN6
CCBE1 IPO5 QSOX1 TSN9
CD9 IPO7 RAB10 TSP4
CDC42 IQGA1 RAB14 UACA
CEMIP KCRM RAB1A UGDH
CFAH KR101 RAB2A VAT1
CHIA KR111 RALA VDAC1
CHSS2 KRA11 RELN VDAC2
CISY LAMB2 RGN VGFR1
CLUS LIS1 RL14 XPO1
CNTN1 LMNA RL18 XPO2
CO7 LRC15 RL18A XPP1
CO8A LUM RL22 XRCC6
XYLT1
Table 4. Biological processes, KEGG pathways, and PMIDs for the EV surface unique proteins in the control group for the control vs. HIV comparison.
Table 4. Biological processes, KEGG pathways, and PMIDs for the EV surface unique proteins in the control group for the control vs. HIV comparison.
Gene Ontology (GO) Terms for Biological Processes
10 Most Significant Results per FDR (for all GO Terms, See Supplemental Table S1A)
Term descriptionObsBgrFDRMatching proteins in the network
Extracellular structure organization163392.01 × 10−10APOA4,COMP,FBLN1,FBN1,FBN2,GAS6,LAMA1,LCAT,LOXL2,MMP2,NID2,PLOD3,PRDX4,SERPINE1,SULF1,SULF2
Extracellular matrix organization142964.25 × 10−9COMP,FBLN1,FBN1,FBN2,GAS6,LAMA1,LOXL2,MMP2,NID2,PLOD3,PRDX4,SERPINE1,SULF1,SULF2
Organonitrogen compound metabolic process4252813.69 × 10 -6ACLY,AICDA,ALDOA,ANXA1,APOA4,C1S,CHIA,EEF1G,EXT1,EXT2,F13A1,FBLN1,FBN1,GAS6,GNB2L1,GPC1,GPC6,GSTP1,IGF2R,IGLL1,KRT1,LCAT,LDHA,LDHB,LEPRE1,LOXL2,LRP1,LTBP1,MMP2,MSRB1,PDIA3,PGD,PGK1,PLOD3,PPIA,PRDX4,RAB1B,SULF1,SULF2,TGM1,UBB,WNT5A
Immune response2215606.48 × 10−6ACLY,ACTR3,AICDA,ALDOA,ANXA1,APOA4,C1S,CHIA,FAS,FLNB,GAS6,GSTP1,IGF2R,IGLL1,KRT1,LRP1,MSRB1,PPIA,PRDX4,PYGB,RAP1B,WNT5A
Vesicle-mediated transport2316996.48 × 10−6ACLY,ACTR3,ALDOA,ANXA1,ARF3,F13A1,FERMT3,GAS6,GSTP1,IGF2R,IGLL1,KRT1,LOXL2,LRP1,PPIA,PRDX4,PYGB,RAB1B,RAP1B,SERPINE1,TIMP3,UBB,WDR1
Regulated exocytosis156916.48 × 10−6ACLY,ALDOA,F13A1,FERMT3,GAS6,GSTP1,IGF2R,KRT1,PPIA,PRDX4,PYGB,RAP1B,SERPINE1,TIMP3,WDR1
Positive regulation of biological process4254596.48 × 10−6ACLY,ACTC1,ACTR3,AICDA,ANXA1,APOA4,C1S,CHIA,CLIC1,FAS,FBLN1,FBN1,FBN2,FERMT3,FSCN1,GAS6,GNAI2,GNB2L1,GPC1,GSTP1,IGF2R,IGLL1,KRT1,LDHA,LEPRE1,LOXL2,LRP1,MMP2,PDIA3,PPIA,RAB1B,RAP1B,SERPINE1,SRPX2,SULF1,SULF2,TGM1,THBS4,TIMP3,UBB,WDR1,WNT5A
Anatomical structure development4050856.48 × 10−6ACTC1,AICDA,ANXA1,APOA4,COMP,EXT1,EXT2,FAS,FBLN1,FBN1,FBN2,FERMT3,FLNB,FLNC,FSCN1,GAS6,GNB2L1,GPC1,GSTP1,IGF2R,KRT1,LDHA,LEPRE1,LOXL2,LRP1,LTBP1,MMP2,MYL6,PGK1,PLOD3,PRDX4,RAP1B,SERPINE1,SRPX2,SULF1,SULF2,TGM1,UBB,WDR1,WNT5A
Response to stimulus5178246.48 × 10−6ACLY,ACTC1,ACTR3,AICDA,ALDOA,ANXA1,APOA4,C1S,CHIA,CLIC1,EEF1G,EXT1,EXT2,F13A1,FAS,FBLN1,FBN1,FERMT3,FLNB,FSCN1,GAS6,GNAI2,GNB2L1,GPC1,GPC6,GPRC5A,GSTP1,IGF2R,IGLL1,KRT1,LAMA1,LDHA,LOXL2,LRP1,LTBP1,MMP2,MSRB1,PDIA3,PGK1,PLOD3,PPIA,PRDX4,PYGB,RAP1B,SERPINE1,SULF1,SULF2,THBS4,TIMP3,UBB,WNT5A
Positive regulation of cellular process3948987.40 × 10−6ACLY,ACTR3,AICDA,ANXA1,APOA4,CHIA,CLIC1,FAS,FBLN1,FBN1,FBN2,FERMT3,FSCN1,GAS6,GNAI2,GNB2L1,GPC1,GSTP1,IGF2R,IGLL1,LDHA,LEPRE1,LOXL2,LRP1,MMP2,PDIA3,PPIA,RAB1B,RAP1B,SERPINE1,SRPX2,SULF1,SULF2,TGM1,THBS4,TIMP3,UBB,WDR1,WNT5A
KEGG Pathways
Term DescriptionObsBgrFDRMatching Proteins in the Network
Proteoglycans in cancer71950.00093FAS,FLNB,FLNC,GPC1,MMP2,TIMP3,WNT5A
Focal adhesion61970.0053COMP,FLNB,FLNC,LAMA1,RAP1B,THBS4
Glycolysis / Gluconeogenesis4680.0054ALDOA,LDHA,LDHB,PGK1
HIF-1 signaling pathway4980.0155ALDOA,LDHA,PGK1,SERPINE1
Cholesterol metabolism3480.0195APOA4,LCAT,LRP1
Malaria3470.0195COMP,LRP1,THBS4
10 Most Significant PMID Publications per FDR
Term IDTerm DescriptionObsBgrFDRMatching Proteins in the Network
PMID:21654676(2011) D-glucuronyl C5-epimerase suppresses small-cell lung cancer cell proliferation in vitro and tumour growth in vivo.8621.79 × 10−5EXT1,EXT2,FAS,GPC1,GPC6,MMP2,SERPINE1,TIMP3
PMID:22393382(2012) In vitro phenotypic, genomic and proteomic characterization of a cytokine-resistant murine Beta-TC3 cell line.7422.32 × 10−5ALDOA,FAS,GSTP1,LDHA,LDHB,PDIA3,PRDX4
PMID:25829250(2015) Insights into the key roles of proteoglycans in breast cancer biology and translational medicine.101562.32 × 10−5EXT1,FBLN1,FBN1,GPC1,GPC6,MMP2,SULF1,SULF2,TIMP3,WNT5A
PMID:26779482(2015) The Extracellular Matrix in Bronchopulmonary Dysplasia: Target and Source.7412.32 × 10−5FBLN1,FBN1,FBN2,LOXL2,LTBP1,PLOD3,SULF2
PMID:23143224(2013) The biology of the extracellular matrix: Novel insights.6285.53 × 10−5COMP,FBN1,FBN2,LTBP1,MMP2,TIMP3
PMID:24223867(2013) Lactate-modulated induction of THBS-1 activates transforming growth factor (TGF)-beta2 and migration of glioma cells in vitro.6317.90 × 10−5COMP,LDHA,LDHB,MMP2,SERPINE1,THBS4
PMID:26076122(2015) Interactions of signaling proteins, growth factors and other proteins with heparan sulfate: Mechanisms and mysteries.6317.90 × 10−5EXT1,EXT2,GPC1,GPC6,SULF1,SULF2
PMID:20236620(2010) Unraveling the mechanism of elastic fiber assembly: The roles of short fibulins.6338.27 × 10−5FBLN1,FBN1,FBN2,LOXL2,LTBP1,TIMP3
PMID:20140087(2010) Comprehensive identification and modified-site mapping of S-nitrosylated targets in prostate epithelial cells.81038.31 × 10−5ALDOA,ANXA1,CLIC1,FLNB,FLNC,PDIA3,PGK1,PLEC
PMID:27513329(2016) Differential Expression Pattern of THBS1 and THBS2 in Lung Cancer: Clinical Outcome and a Systematic-Analysis of Microarray Databases.7658.31 × 10−5COMP,FBLN1,FBN1,MMP2,NID2,SULF1,THBS4
Table 5. Biological processes and PMIDs for the EV surface unique proteins in the HIV group for the HIV vs. HIV+Aβ comparison.
Table 5. Biological processes and PMIDs for the EV surface unique proteins in the HIV group for the HIV vs. HIV+Aβ comparison.
Gene Ontology (GO) Terms for Biological Processes
Term DescriptionObsBgrFDRMatching Proteins in the Network
Cytoskeleton organization59538.35 × 10−5ARPC4,CAP1,DNAH8,NIN,TTN
Supramolecular fiber organization43830.00011ARPC4,CAP1,NIN,TTN
Actin filament organization32000.0011ARPC4,CAP1,TTN
Cellular protein-containing complex assembly48320.0012ARPC4,DNAH8,NIN,TTN
Actin polymerization or depolymerization2430.0031ARPC4,CAP1
Protein polymerization2830.0058ARPC4,NIN
Localization552330.0296ARPC4,CAP1,DNAH8,NIN,TTN
PMID Publications
Term IDTerm DescriptionObsBgrFDRMatching Proteins in the Network
PMID:21050039(2010) Titin A-band-specific monoclonal antibody Tit1 5H1.1. Cellular Titin as a centriolar protein in non-muscle cells.220.0016NIN,TTN
PMID:22985877(2012) Epitope of titin A-band-specific monoclonal antibody Tit1 5 H1.1 is highly conserved in several Fn3 domains of the titin molecule. Centriole staining in human, mouse and zebrafish cells.260.0037NIN,TTN
PMID:26655833(2016) The centrosome is an actin-organizing centre.2120.0081ARPC4,NIN
PMID:27094867(2016) Mutations in human C2CD3 cause skeletal dysplasia and provide new insights into phenotypic and cellular consequences of altered C2CD3 function.2270.027NIN,TTN
PMID:29255378(2017) The human, F-actin-based cytoskeleton as a mutagen sensor.2350.0353DNAH8,TTN
Table 6. Biological processes, KEGG pathways, and PMIDs for the EV surface unique proteins in the HIV+Aβ group for the HIV vs. HIV+Aβ comparison.
Table 6. Biological processes, KEGG pathways, and PMIDs for the EV surface unique proteins in the HIV+Aβ group for the HIV vs. HIV+Aβ comparison.
Gene ontology (GO) Terms for Biological Processes
10 Most Significant Results per FDR (for All GO Terms, See Supplementary Table S2A)
Term DescriptionObsBgrFDRMatching Proteins in the Network
Immune effector process189279.47 × 10−6ACLY,ACTR3,AICDA,ALDOA,C1S,CDC42,GSTP,IGLL1,KPNB1,KRT1,LRP1,PGM1,PPIA,PRDX6,PYGB,PYGL,RAP1B,WDR1
Leukocyte-mediated immunity156329.47 × 10−6ACLY,AICDA,ALDOA,C1S,GSTP1,IGLL1,KPNB1,KRT1,PGM1,PPIA,PRDX6,PYGB,PYGL,RAP1B,WDR1
Vesicle-mediated transport2316999.70 × 10−6ACLY,ACTR3,ALDOA,ANXA1,ARF3,CDC42,F13A1,FERMT3,GSTP1,IGLL1,KPNB1,KRT1,LOXL2,LRP1,PGM1,PPIA,PRDX6,PYGB,PYGL,RAP1B,SERPINE1,UBB,WDR1
Extracellular matrix organization112969.70 × 10−6COMP,FBLN1,FBN1,FBN2,LOXL2,MMP2,NID2,PLOD3,SERPINE1,SULF1,SULF2
Regulated exocytosis156911.15 × 10−5ACLY,ALDOA,F13A1,FERMT3,GSTP1,KPNB1,KRT1,PGM1,PPIA,PRDX6,PYGB,PYGL,RAP1B,SERPINE1,WDR1
Response to stimulus5178241.20 × 10−5ACLY,ACTC1,ACTR3,AICDA,ALDOA,ANXA1,C1S,CDC42,CHIA,CLIC1,EEF1G,EXT1,F13A1,FAS,FBLN1,FBN1,FERMT3,FLNB,FSCN1,GNB2L1,GPC1,GPC6,GPRC5A,GSTP1,HMCN1,IGLL1,KPNB1,KRT1,LDHA,LOXL2,LRP1,LTBP1,MMP2,PDIA3,PGK1,PGM1,PHGDH,PLOD3,PPIA,PRDX2,PRDX6,PYGB,PYGL,RAP1B,SERPINE1,SULF1,SULF2,THBS4,UBA1,UBB,WNT5A
Negative regulation of cellular response to growth factor stimulus81371.59 × 10−5FBN1,FBN2,GPC1,LTBP1,SULF1,SULF2,UBB,WNT5A
Immune system process2623702.26 × 10−5ACLY,ACTR3,AICDA,ALDOA,ANXA1,C1S,CDC42,CHIA,FAS,FLNB,GPC1,GSTP1,IGLL1,KPNB1,KRT1,LRP1,PDIA3,PGM1,PPIA,PRDX6,PYGB,PYGL,RAP1B,UBB,WDR1,WNT5A
Carbohydrate metabolic process124572.26 × 10−5ALDOA,AMY1B,CHIA,EXT1,FBN1,LDHA,LDHB,PGK1,PGM1,PYGB,PYGL,RGN
Immune response2115602.26 × 10−5ACLY,ACTR3,AICDA,ALDOA,ANXA1,C1S,CHIA,FAS,FLNB,GSTP1,IGLL1,KPNB1,KRT1,LRP1,PGM1,PPIA,PRDX6,PYGB,PYGL,RAP1B,WNT5A
KEGG Pathways
Term DescriptionObsBgrFDRMatching Proteins in the Network
Glycolysis / Gluconeogenesis5680.00077ALDOA,LDHA,LDHB,PGK1,PGM1
Proteoglycans in cancer71950.00077CDC42,FAS,FLNB,FLNC,GPC1,MMP2,WNT5A
Focal adhesion61970.0035CDC42,COMP,FLNB,FLNC,RAP1B,THBS4
Pentose phosphate pathway3300.0068ALDOA,PGM1,RGN
Starch and sucrose metabolism3330.0071PGM1,PYGB,PYGL
Metabolic pathways1312500.0095ACLY,ALDOA,CHIA,EXT1,LDHA,LDHB,PGK1,PGM1,PHGDH,PRDX6,PYGB,PYGL,RGN
HIF-1 signaling pathway4980.0095ALDOA,LDHA,PGK1,SERPINE1
Glucagon signaling pathway41000.0095LDHA,LDHB,PYGB,PYGL
Malaria3470.0104COMP,LRP1,THBS4
Carbon metabolism41160.011ALDOA,PGK1,PHGDH,RGN
Fluid shear stress and atherosclerosis41330.0164GPC1,GSTA5,GSTP1,MMP2
Biosynthesis of amino acids3720.0233ALDOA,PGK1,PHGDH
Platinum drug resistance3700.0233FAS,GSTA5,GSTP1
Necroptosis41550.0233FAS,PPIA,PYGB,PYGL
Complement and coagulation cascades3780.0251C1S,F13A1,SERPINE1
Salmonella infection3840.0288CDC42,FLNB,FLNC
MAPK signaling pathway52930.0307CDC42,FAS,FLNB,FLNC,RAP1B
AGE-RAGE signaling pathway in diabetic complications3980.0388CDC42,MMP2,SERPINE1
Human papillomavirus infection53170.0388CDC42,COMP,FAS,THBS4,WNT5A
Propanoate metabolism2320.0405LDHA,LDHB
Leukocyte transendothelial migration31120.0476CDC42,MMP2,RAP1B
Primary immunodeficiency2370.0481AICDA,IGLL1
10 Most Significant PMID Publications per FDR
Term IDTerm DescriptionObsBgrFDRMatching Proteins in the Network
PMID:23823696(2013) Isobaric Tagging-Based Quantification for Proteomic Analysis: A Comparative Study of Spared and Affected Muscles from mdx Mice at the Early Phase of Dystrophy.8421.26 × 10−6ACLY,ALDOA,ANXA1,EEF1G,LDHB,PGM1,PPIA,PRDX2
PMID:29250190(2017) Role of exosomes in hepatocellular carcinoma cell mobility alteration.7348.40 × 10−6ANXA1,CLIC1,FBLN1,LRP1,PPIA,PYGB,PYGL
PMID:20140087(2010) Comprehensive identification and modified-site mapping of S-nitrosylated targets in prostate epithelial cells.91039.47 × 10−6ALDOA,ANXA1,CLIC1,FLNB,FLNC,KPNB1,PDIA3,PGK1,PLEC
PMID:29360750(2018) Proteomic Analysis of Secretomes of Oncolytic Herpes Simplex Virus-Infected Squamous Cell Carcinoma Cells.7379.47 × 10−6ACLY,ANXA1,FBN1,FLNC,FSCN1,MMP2,PRDX2
PMID:26779482(2015) The Extracellular Matrix in Bronchopulmonary Dysplasia: Target and Source.7411.08 × 10−5FBLN1,FBN1,FBN2,LOXL2,LTBP1,PLOD3,SULF2
PMID:24142637(2013) Gastric autoantigenic proteins in Helicobacter pylori infection.7502.96 × 10−5ACTR3,GSTP1,LDHB,PDIA3,PRDX2,PRDX6,WDR1
PMID:26184160(2015) A Review: Proteomics in Nasopharyngeal Carcinoma.8832.96 × 10−5ANXA1,CLIC1,KRT1,MMP2,PPIA,PRDX2,PRDX6,SERPINE1
PMID:26918450(2016) A nuclear-directed human pancreatic ribonuclease (PE5) targets the metabolic phenotype of cancer cells.8893.71 × 10−5ACLY,CLIC1,GPC1,GPC6,LDHA,PGM1,PHGDH,WNT5A
PMID:24223867(2013) Lactate-modulated induction of THBS-1 activates transforming growth factor (TGF)-beta2 and migration of glioma cells in vitro.6315.98 × 10−5COMP,LDHA,LDHB,MMP2,SERPINE1,THBS4
PMID:20236620(2010) Unraveling the mechanism of elastic fiber assembly: The roles of short fibulins.6337.46 × 10−5FBLN1,FBN1,FBN2,HMCN1,LOXL2,LTBP1
Table 7. PMIDs for the EV total unique proteins in the control group for the control vs. HIV comparison.
Table 7. PMIDs for the EV total unique proteins in the control group for the control vs. HIV comparison.
The 10 Most Significant PMID Publications According to FDR
Term IDTerm DescriptionObsBgrFDRMatching Proteins in the Network
PMID:19812696(2009) Cancer genomics identifies regulatory gene networks associated with the transition from dysplasia to advanced lung adenocarcinomas induced by c-Raf-1.31540.0084ACTC1,MAPT,MYH1
PMID:20587776(2010) Mathematical modeling of endocytic actin patch kinetics in fission yeast: disassembly requires release of actin filament fragments.2120.0086ACTC1,MYH1
PMID:25275480(2014) Urethral dysfunction in female mice with estrogen receptor Beta deficiency.2100.0086ACTC1,MYH1
PMID:22406440(2012) Deferiprone reduces amyloid-Beta and tau phosphorylation levels but not reactive oxygen species generation in hippocampus of rabbits fed a cholesterol-enriched diet.2150.0088ACTC1,MAPT
PMID:10931867(2000) Distinct families of Z-line targeting modules in the COOH-terminal region of nebulin.2250.0099ACTC1,MYH1
PMID:11994316(2002) The NH2-terminal peptide of alpha-smooth muscle actin inhibits force generation by the myofibroblast in vitro and in vivo.2260.0099ACTC1,MYH1
PMID:14557251(2003) Skeletal myosin heavy chain function in cultured lung myofibroblasts.2260.0099ACTC1,MYH1
PMID:17908293(2007) Identification of genes differentially expressed during prenatal development of skeletal muscle in two pig breeds differing in muscularity.2520.0099ACTC1,MYH1
PMID:19291799(2009) Fast-twitch sarcomeric and glycolytic enzyme protein loss in inclusion body myositis.2360.0099MAPT,MYH1
PMID:19325835(2008) Myosin assembly, maintenance and degradation in muscle: Role of the chaperone UNC-45 in myosin thick filament dynamics.2440.0099ACTC1,MYH1
Table 8. Biological processes, KEGG pathways, and PMIDs for the EV total unique proteins in the HIV group for the control vs. HIV comparison.
Table 8. Biological processes, KEGG pathways, and PMIDs for the EV total unique proteins in the HIV group for the control vs. HIV comparison.
Gene Ontology (GO) Terms for Biological Processes10 Most Significant Results per FDR (for All GO Terms, See Supplementary Table S4A)
Term DescriptionObsBgrFDRMatching Proteins in the Network
Vesicle-mediated transport5716991.02 × 10−18ACLY,ACTN1,ACTN4,ALDOA,ANXA1,ANXA11,ANXA5,AP2A1,AP2M1,APLP2,APOB,APOE,ARF3,ARF4,ARPC2,CAP1,CD44,CD59,CD63,CD81,EEF2,EHD1,EHD2,F13A1,FERMT3,FLNA,GAS6,ITIH3,ITIH4,KRT1,LAMP1,LAMP2,LOXL2,LRP1,MFGE8,MRC2,MVP,MYH9,PKP1,PRDX6,PTX3,RAB5C,RAB7A,RAC1,RAP1B,SERPINE1,SPTBN1,SRGN,SRPX,TGM2,THBS1,TLN1,TTN,UBB,VPS35,VWF,WDR1
Extracellular structure organization283397.06 × 10−17AGRN,APOA4,APOB,APOE,BMP1,CD44,COMP,DCN,FBLN1,FBN1,GAS6,HTRA1,KLK7,LAMA1,LAMA2,LAMA4,LAMA5,LOXL2,MMP2,NID1,NID2,PLOD3,PXDN,SERPINE1,SULF1,SULF2,THBS1,VWF
Platelet degranulation201292.26 × 10−16ACTN1,ACTN4,ALDOA,ANXA5,APLP2,CD63,F13A1,FERMT3,FLNA,GAS6,ITIH3,ITIH4,LAMP2,SERPINE1,SRGN,THBS1,TLN1,TTN,VWF,WDR1
Regulated exocytosis356911.19 × 10−15ACLY,ACTN1,ACTN4,ALDOA,ANXA5,APLP2,CAP1,CD44,CD59,CD63,EEF2,F13A1,FERMT3,FLNA,GAS6,ITIH3,ITIH4,KRT1,LAMP1,LAMP2,MVP,PKP1,PRDX6,PTX3,RAB5C,RAB7A,RAC1,RAP1B,SERPINE1,SRGN,THBS1,TLN1,TTN,VWF,WDR1
Extracellular matrix organization252962.14 × 10−15AGRN,BMP1,CD44,COMP,DCN,FBLN1,FBN1,GAS6,HTRA1,KLK7,LAMA1,LAMA2,LAMA4,LAMA5,LOXL2,MMP2,NID1,NID2,PLOD3,PXDN,SERPINE1,SULF1,SULF2,THBS1,VWF
Cellular component organization8951632.93 × 10−14ACTN1,ACTN4,AGRN,ALDOA,ANXA1,ANXA6,AP2A1,AP2M1,APOA4,APOB,APOE,ARF4,ARPC2,ATL1,ATXN2,BMP1,CAP1,CD151,CD44,CD59,COMP,DCN,EHD1,EHD2,EXT1,FAS,FAT1,FBLN1,FBN1,FERMT3,FLNA,FLNB,FLNC,FSCN1,GAS6,GGCT,HIST1H4F,HTRA1,KLK7,KRT1,LAMA1,LAMA2,LAMA4,LAMA5,LAMP2,LOXL2,LTBP2,MFGE8,MMP2,MSRB1,MYH9,MYOF,NID1,NID2,PKP1,PLEC,PLOD3,PLS1,PLS3,PTGFRN,PXDN,RAB7A,RAC1,RAN,RHOC,SDC4,SEMG1,SERPINE1,SGCG,SLC25A6,SPAG1,SPTBN1,SRGN,SRPX,SULF1,SULF2,TGM1,TGM2,TGM3,THBS1,THY1,TLN1,TPM4,TTN,UBB,VPS35,VWF,WDR1,WNT5A
Secretion by cell379592.43 × 10−13ACLY,ACTN1,ACTN4,ALDOA,ANXA1,ANXA5,APLP2,CAP1,CD44,CD59,CD63,EEF2,F13A1,FERMT3,FLNA,GAS6,ITIH3,ITIH4,KRT1,LAMP1,LAMP2,LTBP2,MVP,PKP1,PRDX6,PTX3,RAB5C,RAB7A,RAC1,RAP1B,SERPINE1,SRGN,THBS1,TLN1,TTN,VWF,WDR1
Response to stimulus10778246.96 × 10−12ACLY,ACTN4,AFP,AGRN,AHCY,AICDA,ALDOA,ANXA1,ANXA11,ANXA5,ANXA6,AP2A1,AP2M1,APLP2,APOA4,APOB,APOE,ARF4,ARPC2,AZGP1,C1S,CAP1,CD151,CD44,CD59,CD63,CD81,CD82,CLIC1,DCN,EEF2,EHD1,EHD2,EXT1,EXT2,F13A1,FAS,FBLN1,FBN1,FERMT3,FLNA,FLNB,FSCN1,GAS6,GGCT,GNAI2,GNB2L1,GPC6,GPRC5A,HIST1H4F,HSPA5,ITIH4,KRT1,LAMA1,LAMA2,LAMA5,LAMP1,LAMP2,LDHA,LOXL2,LRP1,LTBP1,LTBP2,MMP2,MRC2,MSRB1,MVP,MYH9,MYOF,NNMT,PDIA3,PGK1,PKP1,PLOD1,PLOD3,POLR3G,PRDX1,PRDX6,PTX3,PXDN,RAB5C,RAB7A,RAC1,RAN,RAP1B,RHOC,SDC4,SEMG1,SERPINE1,SLC25A6,SPTBN1,SRGN,SRPX,STK33,SULF1,SULF2,TGM2,THBS1,THRB,THY1,TLN1,TTN,UBA1,UBB,VPS35,VWF,WNT5A
Localization8352339.26 × 10−11ACLY,ACTN1,ACTN4,AGRN,ALDOA,ANXA1,ANXA11,ANXA5,ANXA6,AP2A1,AP2M1,APLP2,APOA4,APOB,APOE,ARF3,ARF4,ARPC2,ATXN2,AZGP1,CAP1,CD151,CD44,CD59,CD63,CD81,CLIC1,EEF2,EHD1,EHD2,F13A1,FAT1,FBN1,FERMT3,FLNA,FLNB,FSCN1,GAS6,GPC6,HSPA5,ITIH3,ITIH4,KRT1,LAMA5,LAMP1,LAMP2,LOXL2,LRP1,LTBP1,LTBP2,MFGE8,MRC2,MVP,MYH9,PKP1,PLOD3,PLS1,PRDX6,PTX3,RAB5C,RAB7A,RAC1,RAN,RAP1B,RHOC,SDC4,SERPINE1,SLC25A6,SPTBN1,SRGN,SRPX,SRPX2,TGM2,THBS1,THY1,TLN1,TTN,TTYH3,UBB,VPS35,VWF,WDR1,WNT5A
Anatomical structure development8050855.90 × 10−10ACTN1,AEBP1,AFP,AGRN,AICDA,ANXA1,AP2A1,APOA4,APOB,APOE,ARF4,ATL1,BMP1,C6orf58,CAP1,CD151,CD44,COMP,DCN,EEF2,EHD1,EXT1,EXT2,FAS,FAT1,FBLN1,FBN1,FERMT3,FLNA,FLNB,FLNC,FSCN1,GAS6,GNB2L1,HSPA5,HTRA1,KLK7,KRT1,LAMA2,LAMA5,LDHA,LOXL2,LRP1,LTBP1,MFGE8,MMP2,MYH9,MYL6,MYOF,NID1,NNMT,PGK1,PKP1,PLOD1,PLOD3,PLS3,PPIB,PRDX1,RAC1,RAP1B,RHOC,SDC4,SERPINE1,SGCG,SPTBN1,SRGN,SRPX2,SULF1,SULF2,TGM1,TGM2,TGM3,THBS1,THBS3,THRB,THY1,TTN,UBB,WDR1,WNT5A
KEGG Pathways
Term DescriptionObsBgrFDRMatching Proteins in the Network
Focal adhesion171971.23 × 10−10ACTN1,ACTN4,COMP,FLNA,FLNB,FLNC,LAMA1,LAMA2,LAMA4,LAMA5,RAC1,RAP1B,THBS1,THBS2,THBS3,TLN1,VWF
ECM-receptor interaction12815.47 × 10−10AGRN,CD44,COMP,LAMA1,LAMA2,LAMA4,LAMA5,SDC4,THBS1,THBS2,THBS3,VWF
Proteoglycans in cancer121953.88 × 10−6CD44,CD63,DCN,FAS,FLNA,FLNB,FLNC,MMP2,RAC1,SDC4,THBS1,WNT5A
Phagosome101451.40 × 10−5COMP,LAMP1,LAMP2,MRC2,RAB5C,RAB7A,RAC1,THBS1,THBS2,THBS3
Amoebiasis8944.01 × 10−5ACTN1,ACTN4,LAMA1,LAMA2,LAMA4,LAMA5,RAB5C,RAB7A
Malaria6478.88 × 10−5CD81,COMP,LRP1,THBS1,THBS2,THBS3
Salmonella infection7840.00016ARPC2,FLNA,FLNB,FLNC,MYH9,RAB7A,RAC1
Endocytosis102420.00054AP2A1,AP2M1,ARF3,ARPC2,EHD1,EHD2,RAB5C,RAB7A,UBB,VPS35
Leukocyte transendothelial migration71120.0007ACTN1,ACTN4,GNAI2,MMP2,RAC1,RAP1B,THY1
Human papillomavirus infection113170.00079COMP,FAS,LAMA1,LAMA2,LAMA4,LAMA5,THBS1,THBS2,THBS3,VWF,WNT5A
PI3K-Akt signaling pathway103480.0069COMP,LAMA1,LAMA2,LAMA4,LAMA5,RAC1,THBS1,THBS2,THBS3,VWF
Complement and coagulation cascades5780.0069C1S,CD59,F13A1,SERPINE1,VWF
Cholesterol metabolism4480.0088APOA4,APOB,APOE,LRP1
Toxoplasmosis51090.0226GNAI2,LAMA1,LAMA2,LAMA4,LAMA5
Glycolysis / Gluconeogenesis4680.0259ALDOA,LDHA,LDHB,PGK1
p53 signaling pathway4680.0259CD82,FAS,SERPINE1,THBS1
Platelet activation51230.0308FERMT3,GNAI2,RAP1B,TLN1,VWF
10 Most Significant PMID Publications per FDR
Term IDTerm DescriptionObsBgrFDRMatching Proteins in the Network
PMID:29250190(2017) Role of exosomes in hepatocellular carcinoma cell mobility alteration.17341.84 × 10−18ACTN1,ANXA1,ANXA11,ANXA5,ANXA6,APOB,APOE,CAP1,CLIC1,FBLN1,FLNA,ITIH4,LRP1,MFGE8,NID1,RAN,TLN1
PMID:24009881(2012) Quantitative proteomics of extracellular vesicles derived from human primary and metastatic colorectal cancer cells.211619.74 × 10−14AHCY,ANXA1,ANXA11,ANXA5,ANXA6,ARF3,ARPC2,CD44,CD63,CD81,FSCN1,KRT1,LAMP1,MFGE8,MYH9,MYL6,PGK1,PTGFRN,RAB5C,RAB7A,VPS35
PMID:19948009(2009) Proteomic analysis of blastema formation in regenerating axolotl limbs.222211.76 × 10−12ANXA1,ANXA11,ANXA5,ANXA6,DCN,EEF2,FBN1,FLNB,GNB2L1,MVP,MYH9,MYL6,MYOF,PDIA3,PLS3,PRDX1,PXDN,RAN,SND1,TTN,UBA1,WNT5A
PMID:24392111(2014) Proteomic analysis of C2C12 myoblast and myotube exosome-like vesicles: a new paradigm for myoblast-myotube cross talk?16791.87 × 10−12ALDOA,ANXA5,CD44,CD63,CD81,CD82,EEF2,FLNC,LAMP1,LAMP2,LDHA,MYOF,PGK1,TLN1,TTN,VPS35
PMID:27605433(2016) Secreted primary human malignant mesothelioma exosome signature reflects oncogenic cargo.171075.36 × 10−12ACLY,ANXA1,ANXA6,CD44,CD63,CD81,CD82,FAT1,GNB2L1,LAMA1,LAMP1,MFGE8,MMP2,PLS3,SULF1,THBS1,VPS35
PMID:22897585(2012) Rat mammary extracellular matrix composition and response to ibuprofen treatment during postpartum involution by differential GeLC-MSMS analysis.13421.24 × 10−11AGRN,ANXA1,ANXA11,ANXA5,ANXA6,CD44,DCN,FBN1,LAMA1,LAMA2,LAMA4,LAMA5,VWF
PMID:27770278(2017) Comprehensive proteome profiling of glioblastoma-derived extracellular vesicles identifies markers for more aggressive disease.14633.75 × 10−11ACTN4,ANXA1,CCT6A,CD44,EHD1,HSPA5,LAMA4,MMP2,MVP,MYH9,RAB5C,RAB7A,UBA1,VPS35
PMID:22159717(2012) The matrisome: in silico definition and in vivo characterization by proteomics of normal and tumor extracellular matrices.14643.97 × 10−11AGRN,ANXA1,ANXA11,ANXA5,ANXA6,DCN,FBN1,LOXL2,LTBP2,NID1,NID2,SRPX,THBS1,VWF
PMID:25201077(2015) Proteomics of apheresis platelet supernatants during routine storage: Gender-related differences.161065.20 × 10−-11ACTN1,APOB,APOE,ARPC2,C1S,FERMT3,FLNA,ITIH4,LDHA,MMP2,MYL6,PRDX6,SRGN,THBS1,TLN1,VWF
PMID:28071719(2017) Quantitative proteomic profiling of the extracellular matrix of pancreatic islets during the angiogenic switch and insulinoma progression.13541.20 × 10−10ANXA1,ANXA11,ANXA5,ANXA6,DCN,FBN1,LAMA1,LAMA2,LAMA4,LAMA5,NID1,NID2,THBS2
Table 9. Biological processes and PMIDs for the EV total unique proteins in the HIV group for the HIV vs. HIV+Aβ comparison.
Table 9. Biological processes and PMIDs for the EV total unique proteins in the HIV group for the HIV vs. HIV+Aβ comparison.
Gene ontology (GO) Terms for Biological Processes
Term DescriptionObsBgrFDRMatching Proteins in the Network
Cell envelope organization230.0017TGM1,TGM3
10 Most Significant PMID Publications per FDR
Term IDTerm DescriptionObsBgrFDRMatching Proteins in the Network
PMID:22329734(2012) Expression profile of cornified envelope structural proteins and keratinocyte differentiation-regulating proteins during skin barrier repair.3140.0016KLK7,TGM1,TGM3
PMID:11093806(2000) Transglutaminase-3, an esophageal cancer-related gene.220.0136TGM1,TGM3
PMID:11562168(2001) Crystallization and preliminary X-ray analysis of human transglutaminase 3 from zymogen to active form.220.0136TGM1,TGM3
PMID:11980702(2002) Three-dimensional structure of the human transglutaminase 3 enzyme: binding of calcium ions changes structure for activation.220.0136TGM1,TGM3
PMID:12850301(2003) Analysis of epidermal-type transglutaminase (transglutaminase 3) in human stratified epithelia and cultured keratinocytes using monoclonal antibodies.230.0136TGM1,TGM3
PMID:14508061(2003) A model for the reaction mechanism of the transglutaminase 3 enzyme.220.0136TGM1,TGM3
PMID:14645372(2004) Structural basis for the coordinated regulation of transglutaminase 3 by guanine nucleotides and calciummagnesium.220.0136TGM1,TGM3
PMID:14987256(2004) Identification of calcium-inducible genes in primary keratinocytes using suppression-subtractive hybridization.280.0136KLK7,TGM1
PMID:15084592(2004) Crystal structure of transglutaminase 3 in complex with GMP: structural basis for nucleotide specificity.220.0136TGM1,TGM3
PMID:15172109(2004) Transglutaminase activity and transglutaminase mRNA transcripts in gerbil brain ischemia.230.0136TGM1,TGM3
Table 10. Biological processes, KEGG pathways, and PMIDs for the EV total unique proteins in the HIV+Aβ group for the HIV vs. HIV+Aβ comparison.
Table 10. Biological processes, KEGG pathways, and PMIDs for the EV total unique proteins in the HIV+Aβ group for the HIV vs. HIV+Aβ comparison.
Gene Ontology (GO) Terms for Biological Processes
10 Most Significant Results per FDR (for All GO Terms, See Supplementary Table S5A)
Term DescriptionObsBgrFDRMatching Proteins in the Network
Vesicle-mediated transport4116992.01 × 10−13ACTR3,AP1G1,AP2B1,ARF6,ARPC4,CALR,CAND1,CAPZB,CAV1,CD9,CDC42,COPB2,ECM1,EEA1,EHD4,IGF2R,KPNB1,MME,NME1,PDIA6,PGM1,PPIA,PSMD1,PSMD2,PSMD3,PYGB,PYGL,QSOX1,RAB10,RAB14,RAB1A,RAB2A,RALA,SLC44A2,SOD1,STOM,SYK,TGFB1,TIMP3,VAT1,XRCC6
Localization6652334.37 × 10−11ACTR3,AP1G1,AP2B1,APOM,ARF6,ARPC4,CALR,CAND1,CAPZB,CAV1,CD9,CDC42,COPB2,CSE1L,DHX9,ECM1,EEA1,EHD4,FBN2,IGF2R,IGSF8,ILK,IPO5,IPO7,KPNB1,LMNA,MME,NME1,NRP1,PAFAH1B1,PDIA6,PGM1,PIP,PPIA,PSMD1,PSMD2,PSMD3,PYGB,PYGL,QSOX1,RAB10,RAB14,RAB1A,RAB2A,RALA,RELN,RNF128,RPL14,RTN4,SLC3A2,SLC44A1,SLC44A2,SLIT2,SOD1,SPOCK1,STOM,SYK,TGFB1,THBS4,TIMP3,VAT1,VDAC1,VDAC2,WLS,XPO1,XRCC6
Secretion3010708.12 × 10−11CAND1,CAV1,CD9,ECM1,IGF2R,KPNB1,MME,NME1,PAFAH1B1,PGM1,PPIA,PSMD1,PSMD2,PSMD3,PYGB,PYGL,QSOX1,RAB10,RAB14,RAB1A,RALA,SLC44A2,SOD1,STOM,SYK,TGFB1,TIMP3,VAT1,WLS,XRCC6
Transport5741301.80 × 10−10ACTR3,AP1G1,AP2B1,APOM,ARF6,ARPC4,CALR,CAND1,CAPZB,CAV1,CD9,CDC42,COPB2,CSE1L,DHX9,ECM1,EEA1,EHD4,IGF2R,IPO5,IPO7,KPNB1,LMNA,MME,NME1,NRP1,PAFAH1B1,PDIA6,PGM1,PIP,PPIA,PSMD1,PSMD2,PSMD3,PYGB,PYGL,QSOX1,RAB10,RAB14,RAB1A,RAB2A,RALA,RPL14,SLC3A2,SLC44A1,SLC44A2,SOD1,STOM,SYK,TGFB1,TIMP3,VAT1,VDAC1,VDAC2,WLS,XPO1,XRCC6
Secretion by cell289591.80 × 10−10CAND1,CD9,ECM1,IGF2R,KPNB1,MME,PAFAH1B1,PGM1,PPIA,PSMD1,PSMD2,PSMD3,PYGB,PYGL,QSOX1,RAB10,RAB14,RAB1A,RALA,SLC44A2,SOD1,STOM,SYK,TGFB1,TIMP3,VAT1,WLS,XRCC6
Regulated exocytosis246912.67 × 10−10CAND1,CD9,ECM1,IGF2R,KPNB1,MME,PGM1,PPIA,PSMD1,PSMD2,PSMD3,PYGB,PYGL,QSOX1,RAB10,RAB14,SLC44A2,SOD1,STOM,SYK,TGFB1,TIMP3,VAT1,XRCC6
Exocytosis257743.25 × 10−10CAND1,CD9,ECM1,IGF2R,KPNB1,MME,PGM1,PPIA,PSMD1,PSMD2,PSMD3,PYGB,PYGL,QSOX1,RAB10,RAB14,RALA,SLC44A2,SOD1,STOM,SYK,TGFB1,TIMP3,VAT1,XRCC6
Neutrophil activation involved in immune response194891.06 × 10−8CAND1,IGF2R,KPNB1,MME,PGM1,PPIA,PSMD1,PSMD2,PSMD3,PYGB,PYGL,QSOX1,RAB10,RAB14,SLC44A2,STOM,SYK,VAT1,XRCC6
Myeloid leukocyte activation205741.48 × 10−8CAND1,IGF2R,KPNB1,MME,PGM1,PPIA,PSMD1,PSMD2,PSMD3,PYGB,PYGL,QSOX1,RAB10,RAB14,SLC44A2,STOM,SYK,TGFB1,VAT1,XRCC6
Neutrophil degranulation184854.95 × 10−8CAND1,IGF2R,KPNB1,MME,PGM1,PPIA,PSMD1,PSMD2,PSMD3,PYGB,PYGL,QSOX1,RAB10,RAB14,SLC44A2,STOM,VAT1,XRCC6
KEGG Pathways
Term descriptionObsBgrFDRMatching Proteins in the Network
Endocytosis102420.00016AP2B1,ARF6,ARPC4,CAPZB,CAV1,CDC42,EEA1,EHD4,IGF2R,RAB10
Focal adhesion81970.0012CAV1,CDC42,ILK,LAMB2,PARVB,PPP1CB,RELN,THBS4
Bacterial invasion of epithelial cells5720.003ARPC4,CAV1,CDC42,ILK,SEPT2
Pentose phosphate pathway3300.0278G6PD,PGM1,RGN
Starch and sucrose metabolism3330.0278PGM1,PYGB,PYGL
Proteoglycans in cancer61950.0278CAV1,CDC42,LUM,PPP1CB,TGFB1,TIMP3
Proteasome3430.0347PSMD1,PSMD2,PSMD3
Necroptosis51550.0347PPIA,PYGB,PYGL,VDAC1,VDAC2
Fc gamma R-mediated phagocytosis4890.0347ARF6,ARPC4,CDC42,SYK
Amino sugar and nucleotide sugar metabolism3480.0396CHIA,PGM1,UGDH
HTLV-I infection62500.0396CALR,NRP1,TGFB1,VDAC1,VDAC2,XPO1
10 Most Significant PMID Publications per FDR
Term IDTerm DescriptionObsBgrFDRMatching Proteins in the Network
PMID:11149929(2001) The phagosome proteome: insight into phagosome functions.9473.12 × 10−6ARF6,CALR,DFFA,P4HB,RAB10,RAB14,RAB2A,STOM,VDAC1
PMID:17892558(2007) Quantifying raft proteins in neonatal mouse brain by ‘tube-gel’ protein digestion label-free shotgun proteomics.10836.99 × 10−6ACTC1,BASP1,CAV1,CNTN1,RAB10,RAB14,RAB1A,RAB2A,SLC3A2,VDAC1
PMID:22578496(2012) Harnessing the power of the endosome to regulate neural development.7350.00014ARF6,EEA1,EHD4,NRP1,RAB14,RTN4,WLS
PMID:24009881(2012) Quantitative proteomics of extracellular vesicles derived from human primary and metastatic colorectal cancer cells.111610.00014ACTR3,CAPZB,CD9,EHD4,ILK,RAB10,RALA,SLC3A2,SLC44A1,SYK,UGDH
PMID:27770278(2017) Comprehensive proteome profiling of glioblastoma-derived extracellular vesicles identifies markers for more aggressive disease.8630.00016ACTR3,CALR,ECM1,IGF2R,IPO5,PSMD2,RAB10TGFB1
PMID:26205348(2015) Fluoxetine increases plasticity and modulates the proteomic profile in the adult mouse visual cortex.6220.00023AP1G1,CDC42,NME1,SOD1,VDAC1,VDAC2
PMID:20140087(2010) Comprehensive identification and modified-site mapping of S-nitrosylated targets in prostate epithelial cells.91030.00024DHX9,HNRNPK,KPNB1,LMNA,P4HB,PDIA6,RTN4,VDAC1,VDAC2
PMID:27549615(2016) Genome-wide association study to identify potential genetic modifiers in a canine model for Duchenne muscular dystrophy.6230.00024LMNA,PAMR1,PPIA,PSMD2,SLIT2,THBS4
PMID:23170974(2012) Integrated miRNA, mRNA and protein expression analysis reveals the role of post-transcriptional regulation in controlling CHO cell growth rate.6270.00044HNRNPK,RAB10,RAB14,RAB1A,RAB2A,RPL14
PMID:24505448(2014) Characterisation of four LIM protein-encoding genes involved in infection-related development and pathogenicity by the rice blast fungus Magnaporthe oryzae.6280.00047CDC42,ILK,LMNA,PHGDH,RAB2A,XRCC6

Share and Cite

MDPI and ACS Style

András, I.E.; Sewell, B.B.; Toborek, M. HIV-1 and Amyloid Beta Remodel Proteome of Brain Endothelial Extracellular Vesicles. Int. J. Mol. Sci. 2020, 21, 2741. https://doi.org/10.3390/ijms21082741

AMA Style

András IE, Sewell BB, Toborek M. HIV-1 and Amyloid Beta Remodel Proteome of Brain Endothelial Extracellular Vesicles. International Journal of Molecular Sciences. 2020; 21(8):2741. https://doi.org/10.3390/ijms21082741

Chicago/Turabian Style

András, Ibolya E., Brice B. Sewell, and Michal Toborek. 2020. "HIV-1 and Amyloid Beta Remodel Proteome of Brain Endothelial Extracellular Vesicles" International Journal of Molecular Sciences 21, no. 8: 2741. https://doi.org/10.3390/ijms21082741

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop