Next Article in Journal
How to Assess Pulmonary Circulation and Right Heart Chambers in Systemic Sclerosis Patients?
Previous Article in Journal
Clear Cell Sarcoma of Soft Tissues: Radiological Analysis of 14 Patients—MRI Findings Related to Metastatic Disease
Previous Article in Special Issue
Comparative Analysis of HPV Detection Efficiency: Evaluating Cobas 8800 Performance in Vaginal Self-Sampling versus Clinician-Collected Samples at a Regional Thai Hospital
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

From Cell Lines to Patients: Dissecting the Proteomic Landscape of Exosomes in Breast Cancer

1
Laboratory of Molecular Medicine, Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia
2
Institute of Medicine and Medical Technologies, Novosibirsk State University, 630090 Novosibirsk, Russia
3
Laboratory of Proteomics and Metabolomics, International Tomography Center, Siberian Branch of Russian Academy of Sciences, 630090 Novosibirsk, Russia
4
Department of Mammology, Novosibirsk Regional Clinical Oncological Dispensary, 630108 Novosibirsk, Russia
5
Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37203, USA
6
Laboratory of Tumor Biochemistry, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Science, 634028 Tomsk, Russia
7
Institute of Oncology and Neurosurgery, E. Meshalkin National Medical Research Center of the Ministry of Health of the Russian Federation, 630090 Novosibirsk, Russia
*
Author to whom correspondence should be addressed.
Diagnostics 2025, 15(8), 1028; https://doi.org/10.3390/diagnostics15081028
Submission received: 19 February 2025 / Revised: 7 April 2025 / Accepted: 8 April 2025 / Published: 17 April 2025
(This article belongs to the Special Issue Liquid Biopsy: Cancer Diagnostic Biomarkers of the Future)

Abstract

:
Background: Breast cancer (BC) is the most common cancer among women worldwide; therefore, the efforts of many scientists are aimed at finding effective biomarkers for this disease. It is known that exosomes are nanosized extracellular vesicles (EVs) that are released from various cell types, including cancer cells. Exosomes are directly involved in governing the physiological and pathological processes of an organism through the horizontal transfer of functional molecules (proteins, microRNA, etc.) from producing to receiving cells. Since the diagnosis and treatment of BC have been improved substantially with exosomes, in this study, we isolated breast carcinoma cell-derived exosomes, primary endotheliocyte-derived exosomes, and blood exosomes from BC patients (BCPs) in the first stage of disease and investigated their proteomic profiles. Methods: Exosomes were isolated from the samples by ultrafiltration and ultracentrifugation, followed by mass spectrometric and bioinformatics analyses of the data. The exosomal nature of vesicles was verified using transmission electron microscopy and flow cytometry. Results: Exosome proteins secreted by MCF-7 and BT-474 cells were found to form two clusters, one of which enhanced the malignant potential of cancer cells, while the other coincided with a cluster of HUVEC-derived exosome proteins. Despite the different ensembles of proteins in exosomes from the MCF-7 and BT-474 lines, the relevant portions of these proteins are involved in similar biological pathways. Comparison analysis revealed that more BC-associated proteins were found in the exosomal fraction of blood from BCPs than in the exosomal fraction of conditioned medium from cells mimicking the corresponding cancer subtype (89% and 81% for luminal A BC and MCF-7 cells and 86% and 80% for triple-positive BC and BT-474 cells, respectively). Conclusions: Tumor-associated proteins should be sought not in exosomes secreted by cell lines but in the composition of blood exosomes from cancer patients, while the contribution of endotheliocyte exosomes to the total pool of blood exosomes can be neglected.

1. Introduction

In recent decades, there has been a steady increase in the number of new cases of malignant neoplasms, which is primarily due to increased life expectancy, as well as changes in lifestyle and exposure to unfavorable environmental factors. Today, cancer ranks second in mortality, conceding only to cardiovascular diseases. In 2022, breast cancer (BC) accounted for 23.8% of all new cancer cases in women worldwide. In particular, there were 2.3 million new cases of BC and 666,000 deaths from BC worldwide in 2022 [1]. The incidence of BC continues to increase despite the success of mammographic screening (depending on the country, from 14.2% (Republic of Moldova) to 47.3% (Czech Republic) of patients are diagnosed at stage I) [2]. Obviously, early BC diagnosis will increase the effectiveness of anti-tumor therapy and the survival rate of tumor patients. Thus, there is an urgent need to identify more effective and non-invasive surrogate markers that can guide not only early diagnosis but also the selection of therapeutic strategies for individual patients and the accurate assessment of prognosis [3]. Furthermore, BC is characterized by considerable tissue heterogeneity, showing distinct clinical and biological features, which makes tumors respond differently to treatments and complicates their management [4]. Today, molecular profiles have been largely explored, providing a well-established classification of BCs into five well-settled subtypes: luminal A, luminal B Her2+, luminal B Her2-, basal-like, and human epidermal growth factor receptor 2 (Her2)-enriched [2,5]. These molecular subtypes of BC are established by tumor biopsy, which can cause the displacement of tumor cells, promoting metastasis and various pathological changes in breast tissue [6]. In addition, biopsy exhibits inaccuracy in determining the BC subtype and is not posed to track patient follow-up [4]. In this regard, a promising direction in molecular oncology has been the search for new tumor markers in the composition of extracellular vesicles (EVs) by liquid biopsy. Among EVs, exosomes, small vesicles 30–150 nm in diameter with a lipid bilayer membrane and tetraspanins CD9, CD63, and CD81 on their crown, are prominent [7,8]. The advantage of exosomes is the sorting of biologically active molecules (proteins, different types of RNA) at the maturation stage of these vesicles [9]. Recently, exosomes from breast cancer cell lines have been shown to be a rich source of breast cancer-related proteins and surface biomarkers and can be used for the diagnosis and prognosis of the disease [10,11]. However, when analyzing the content of exosomes circulating in the blood of cancer patients, it is necessary to take into account that in addition to tumor exosomes, exosomes from endotheliocytes and other non-tumor cells, as well as cells from the tumor microenvironment, are present in the blood.
To assess the diagnostic potential of blood exosomes and evaluate the contribution of endotheliocytes to the blood exosome proteome, we performed a differential analysis of exosomal proteomes from primary endotheliocytes, from two cell lines mimicking luminal A and luminal Her2-positive BC, and from the blood of patients with these BC subtypes.

2. Materials and Methods

2.1. Isolation and Cultivation of Human Umbilical Vein Endothelial Cells (HUVECs)

HUVECs were obtained from three donors. Each vein was washed sequentially with 50 mL phosphate-buffered saline (PBS) and 20 mL collagenase IV buffer (0.1% collagenase IV in buffer containing 1.5 mM HEPES, 14 mM NaCl, 0.4 mM KCl, 0.12 mM CaCl2, 0.04 mM MgSO4, and 0.76 mM D-glucose, pH 7.4) [12]. The vein was incubated with 0.1% collagenase IV solution at 37 °C for 15 min to release endothelial cells. The collagenase solution containing detached cells was collected and combined with an additional PBS wash of the vein. The pooled solution was centrifuged at 800× g for 10 min, and the cell pellet was resuspended in IMDM (Gibco, Aucland, New Zealand) supplemented with 10% fetal bovine serum (FBS) (Thermo Fisher Scientific, Waltham, MA, USA) and penicillin–streptomycin (100 μg/mL) (Thermo Fisher Scientific, Waltham, MA, USA). HUVECs were cultured in a CO2 incubator (5% CO2) at 37 °C, and adherent cells were washed the next day with fresh IMDM to remove residual blood cells. HUVECs were cultured to 70–80% confluence, and cells from the first passage were used for exosome isolation. For dissociation, cells were treated with 0.1% collagenase IV.

2.2. Cancer Cell Line Cultivation

MCF-7 (ATCC® HTB-22™) and BT-474 (ATCC® HTB-20™) BC cell lines were cultured in Dulbecco’s Modified Eagle Medium (DMEM) (Thermo Fisher Scientific, Waltham, MA, USA) supplemented with 10% FBS (Gibco, USA) and penicillin–streptomycin (100 μg/mL) (Thermo Fisher Scientific, Waltham, MA, USA) in a CO2 incubator (5% CO2) at 37 °C up to 70–80% confluence. Cells were subcultured with a solution of 0.25% trypsin in PBS with 5 mM EDTA.

2.3. Isolation of Exosomes from Conditioned Medium

All cells were negative for mycoplasma infection, as confirmed by PCR analysis of the 16S mycoplasma ribosomal gene [13].
FBS was centrifuged at 100,000× g for 2 h at 4 °C to remove bovine exosomes. The supernatant was collected and used to prepare bovine exosome-depleted medium. Three days prior to cell harvesting, the culture medium was replaced with a depleted medium containing IMDM for HUVECs or DMEM for MCF-7 and BT-474 cells, a mixture of antibiotics, and 10% FBS devoid of bovine exosomes.
For the isolation of exosomes, after 72 h, conditioned medium was collected and subjected to two successive centrifugations at 300× g and 15,000× g for 10 min and 20 min, respectively to remove dead cells and cellular debris. To eliminate large EVs, the supernatant was filtered through 100 nm pore-size filters (Minisart high flow, 16553-K, Sartorius, Goettingen, Germany). Exosomes were isolated from the pre-cleared conditioned medium by centrifugation at 100,000× g for 90 min at 100,000× g at 4 °C. The pellets were suspended in 12 mL of PBS and again centrifuged for 90 min at 100,000× g at 4 °C. The washing stage was repeated two times. Then, the supernatant was removed, and exosomes were resuspended in 300 μL of PBS, aliquoted, frozen in liquid nitrogen, and stored at −80 °C.

2.4. Ethics Statement

The study protocol was approved by the Ethics Committee of the Institute of Chemical Biology and Fundamental Medicine. Written informed consent was obtained from every female. Human samples were obtained according to the principles expressed in the Declaration of Helsinki. Blood samples from untreated BC patients (BCPs) with from luminal A (n = 5, age range 56–61 years, median age 61) and triple-positive (n = 8, age range 44–69 years, median age 61) subtypes [2,5] (Table 1) were obtained from the Novosibirsk Regional Oncology Dispensary.

2.5. Exosome Isolation from Blood

Venous blood (9 mL) was collected by venipuncture in K3EDTA spray-coated vacutainers (Improvacuter, Guangzhou, China) and processed within one hour. To isolate total blood exosomes by ultrafiltration and differential ultracentrifugation, a previously described protocol was used [14]. The pellet containing blood exosomes was resuspended in 300 μL of PBS.

2.6. Characterization of Exosomes

The morphology and membrane integrity of the isolated exosomes was assessed by transmission electron microscopy (TEM), as described previously [15]. The initial volume of exosomes analyzed using TEM was 15 μL.
To evaluate the protein concentration of exosomes, a NanoOrange Protein Quantitation kit (NanoOrange Protein Quantitation Kit, Molecular Probes, San Jose, CA, USA) was used as described previously [16].
The presence of CD9, CD63, and CD81 tetraspanins in the exosomal crown was confirmed by flow cytometry, as described previously [16]. The median fluorescence intensity (MFI) of stained exosomes was analyzed and compared to the isotype control (BD bioscience, Heidelberg, Germany). Flow cytometry was performed using a Cytoflex instrument (Becman Coulter, BioBay, Suzhou, China) with CytExpert 2.0 Software.

2.7. Mass Spectrometry Analysis

For the identification of exosomal proteins by MALDI-TOF mass spectrometry, proteins were separated using SDS-disc electrophoresis, and then fragments of polyacrylamide gel containing the studied proteins were washed from SDS and subjected to trypsinolysis, as described previously [14]. Specifically, samples were applied to the gel in 5 replicates, with line widths of 6 mm each. After the separation of the proteins in the gel, each line was cut into pieces 5 mm thick, resulting in 20 pieces from each line. Peptides from each piece of gel were analyzed independently; a protein was considered reliably identified when it was detected in at least three out of five cases.
Peptide fragments of proteins were extracted from the gel, concentrated, and desalted on C18 ZipTips microcolumns (Milipore, Burlington, MA, USA). Next, 5 μL of a saturated solution of α-cyano-4-hydroxycinnamic acid in 70% acetonitrile was added and then spotted onto an MTP 384 ground steel target plate. After crystallization, the target was loaded into the mass spectrometer to obtain the protein molecular weight spectrum. The acquisition and registration of mass spectra were carried out on an Ultraflex III time-of-flight tandem mass spectrometer (MALDI-TOF/TOF spectrometer) (Bruker Daltonics, Bremen, Germany). To ensure the reliability and reproducibility of the results, all analyses were performed with five biological replicates for each sample.
Spectra were acquired using the following parameters: shots—150, laser frequency—66.7 Hz, laser attenuator offset—85%, laser attenuator range—21%, laser attenuator set—5_ularge, laser focus offset—0%, laser focus range—100%, and laser focus value—4%. The instrument was pre-calibrated for a mass range of 500–3800 kDa. The obtained spectra were converted into mass values using flexAnalysis software 3.4. Protein identification was performed by searching for appropriate candidates in the annotated NCBI and SwissProt databases using the Mascot program (Matrix Science Ltd., London, UK, Available online: www.matrixscience.com/search_form_select.html (accessed on 20 October 2024)) with the following search parameters: species—Homo sapiens, error tolerance—±300 ppm, maximum number of missed cleavages—2, fixed modifications—propionamide (C), variable modifications—oxidation (M), phospho (ST); the identification reliability was not lower than 95%.
The matching of at least two peptides comprising 9 or more amino acid residues was considered a reliable identification of minor proteins [17].

2.8. Bioinformatics Analysis

Data from the SwissProt database were translated into the UniProt database for further analysis using the Retrieve/ID mapping platform (Available online: https://www.uniprot.org (accessed on 11 November 2024)). Functional enrichment analysis of exosomal proteomes according to Gene Ontologies was conducted using STRING software 12.0 (Available online: https://www.string-db.org/ (accessed on 10 January 2025)). Cellular localization, molecular functions, and involvement in biological processes were determined using FunRich 3.13 software based on the Gene Ontologies (GO) component, GO function, and GO process categories. Involvement in biological pathways was assessed using the Reactome service (Available online: https://reactome.org/ (accessed on 11 January 2025)). For primary data processing and graph generation, Python 3.11 libraries such as Pandas 2.2.3, Numpy 2.2.0, Matplotlib 3.10.1, and Seaborn 0.13.2 were utilized.

3. Results

3.1. Characterization of Isolated Exosomes

To characterize exosomes isolated from conditioned medium and BCP blood, TEM and flow cytometry were used. TEM revealed the presence of clearly structured cup-shaped vesicles (40–95 nm) of low electron density with preserved membranes in all samples; the portion of vesicles with a size smaller than 30 nm was no more than 13% (Figure 1). We then confirmed that the exosomes isolated from all sources contained typical exosome protein markers, namely, tetraspanins CD9 and CD63. For this purpose, exosomes adsorbed onto aldehyde/sulfate latex beads using anti-CD9 antibodies were stained with FITC-labeled antibodies for the tetraspanin CD63. It was shown that the CD9 and CD63 tetraspanins were comparably represented in all vesicles (Figure 1).
Collectively, the obtained data reveal that the sEVs isolated from conditioned medium and blood have all properties of exosomes [18].

3.2. Annotation of Identified Proteins from HUVEC-Derived Exosomes

Since in addition to tumor exosomes, exosomes from normal cells, including those of endothelial origin, circulate in the blood of cancer patients, the first stage of this work involved the identification and characterization of exosome proteins secreted by primary endothelial cells from three donors. A total of 24 proteins were identified with high confidence (p < 0.05) using MALDI-TOF mass spectrometry in the HUVEC-derived exosomes (Table S1); of these, 29% (7 proteins) were common to the three groups (Figure 2A).
The majority (70%) of identified exosomal proteins from HUVECs were previously discovered in other studies using mass spectrometry and were annotated in the Vesiclepedia database [19] (the putative CNGA1-overlapping antisense gene protein is not included in the analysis because its gene is not known). Thus, about one-third of the exosomal proteins are identified in our study for the first time as a part of EVs; previously, they were not annotated in this database (Figure 2B, Table 2).
The protein–protein interaction (PPI) network was mainly concentrated on the relevance among ALB, CD9, CD63, CD81, ADAM10, and MMP9, all of which are major exosome proteins (Figure 3).
The molecular function GO analysis revealed that HUVEC-derived exosomal proteins were commonly enriched in functions such as “metallopeptidase activity” and “transporter activity” (Table 3).
Six predominant proteins (ALB, CD9, CD63, CD81, MMP9, PSMD10) were found to be significantly (p < 0.05) involved in biological pathways (Table 4). In particular, the common proteomic profiles of endothelial exosomes were enriched in terms related to biological processes such as “metabolism of RNA”, “metabolism of mRNA”, “adaptive immune system”, etc.

3.3. Annotation of Protein Cargo from BC-Derived Exosomes

Using MALDI-TOF mass spectrometry, 34 and 42 proteins were identified with high reliability (p < 0.05) in the exosomes from BC cell lines BT-474 and MCF-7, respectively (Supplementary Tables S2 and S3). Of these, only 6% (five proteins) were common to both groups (Supplementary Tables S2 and S3, Figure 4A). It should be noted that the same proteins (CD9, CD24, CD63, CD81, MMP9) were common to exosomes of cancer cells and primary cells.
It should be noted that due to the small size of exosomes (30–150 nm) and their high RNA content, the cargo of these vesicles contains predominantly low-molecular-weight proteins rich in lysine and arginine. As a consequence, the proportion of reliably identified proteins meeting the new criteria for proteome investigation [17] is extremely low (trypsinolysis results in a large number of peptides with fewer than nine amino acid residues).
Approximately 65% of identified exosomal proteins from cancer cells were previously discovered in other studies using mass spectrometry and were annotated in the Vesiclepedia database (Figure 4B) (the putative uncharacterized protein FLJ45840 protein is not included in the analysis because its gene is not known). The non-deposited Vesiclepedia database proteins of exosomes secreted by MCF-7 and BT-474 cells are presented in Table 5 and Table 6, respectively.
For cancer-derived exosomes, the PPI network was mainly concentrated on the connections among UPF3B, SRSF6, SRSF5, RBMX, PRPF38A, KCNC4, RPS27I, VAMP8, EFNB2, CD9, CD63, CD81, ADAM10, and MMP9, among which 11 exosomal proteins formed two clusters: in addition to the cluster of major proteins identified by molecular cargo analysis of primary endotheliocytes, one cluster was identified that enhanced the malignant potential of MCF-7 and BT-474 cells (Figure 5).
The molecular function GO analysis revealed that MCF-7-derived exosomal proteins were commonly enriched in functions such as “calcium ion binding”, “metallopeptidase activity”, “RNA binding”, “ubiquitin-specific protease activity”, “GTPase activity”, “DNA binding”, etc. (Table 7).
Analysis of the protein cargo of exosomes secreted by MCF-7 cells showed the involvement of proteins in biological pathways such as “membrane trafficking”, “Clathrin-derived vesicle budding”, “trans-Golgi network vesicle budding”, “signaling by SCF-KIT”, “Notch signaling pathway”, “signaling by EGFR”, etc. (Table 8). It should be noted that five of the eight (63%) proteins in Table 6 were identified by PPI analysis as proteins interacting with a large number of other proteins (Figure 5).
The molecular functions of BT-474 cell-derived exosomal proteins were very similar to the functions of proteins secreted by MCF-7 exosomes. Specifically, bioinformatics analysis showed that the most common molecular functions of exosomal proteins were “metallopeptidase activity” and “RNA binding” (Table 9).
Similarly, bioinformatics analysis of proteins from BT-474 cell-secreted exosomes revealed their involvement in biological pathways such as “gene expression”, “mRNA splicing”, “mRNA processing”, “transcription”, etc. (Table 10). As with exosomal proteins from MCF-7 cells, in exosomes from BT-474 cells, five of the eight proteins (63%) presented in Table 10 were also identified by PPI analysis as proteins that interact with a large number of other proteins (Figure 5).
It was also shown that despite the different ensembles of proteins within exosomes from MCF-7 and BT-474 cell lines, these exosomal proteins are involved in many of the same biological pathways such as “post-elongation processing of intron-containing pre-mRNA”, “mRNA 3′-end processing”, “cleavage of growing transcript in the termination region”, “post-elongation processing of the transcript”, “RNA polymerase II transcription termination”, and “receptor–ligand binding initiates the second proteolytic cleavage of Notch receptor” (Table 8 and Table 10).

3.4. Comparative Proteomic Analysis of Exosomes in the Blood of BCPs with Luminal A and Triple-Positive Subtypes

Using MALDI-TOF mass spectrometry with high confidence (p < 0.05), we identified 74 and 110 proteins in exosomes from the blood of untreated BCPs with luminal A (n = 5, Table 1) and triple-positive (n = 8, Table 1) subtypes, respectively (Supplementary Tables S4 and S5). It should be noted that 8 out of 41 universal exosomal proteins were detected in half of the samples. The analysis of the identified exosomal proteins using the Vesiclepedia database revealed that among the 119 proteins that were previously found to be associated with vesicles, a total of 61 and 95 proteins were identified in the luminal A BC and triple-positive BC exosomes, respectively. Thus, 17% of proteins identified in exosomes from the blood of BCPs in this study were not previously annotated in the Vesiclepedia database (Figure 6). The non-deposited Vesiclepedia database proteins of BCP blood exosomes are presented in Table 11.
The molecular functions of BCP blood exosome proteins were similar to those of proteins within exosomes secreted by MCF-7 and BT-474 cells. In particular, bioinformatics analysis showed that the universal molecular functions of exosomal proteins were “calcium ion binding”, “DNA binding”, “metallopeptidase activity”, and “RNA binding” (Table 7, Table 9, and Table 12).
At the same time, the bioinformatic analysis also revealed terms unique to BCP blood exosome proteins such as “protease inhibitor activity”, “protein binding”, etc.
It should be noted that barrier-to-autointegration factor (BANF1) and zinc finger protein 638 (ZNF638) were found earlier as DNA-binding proteins in exosomes [20]. In general, these exosomal DNA-binding proteins have many molecular functions and are involved in important biological processes [21,22,23,24]. Thus, exosomes carry biologically active DNA-binding proteins, mainly in the form of internal contents from donor cells to recipient cells, causing changes in the behavior of recipient cells.
To determine whether the exosomal proteins we identified were found in breast neoplasms, the list of proteins was analyzed using FunRich 3.13 software to search a publicly available database for proteins in BC, namely, the Human Protein Atlas (HPA). It was shown that 66 BC-associated proteins from blood exosomes of BCPs with a luminal A subtype and 34 from MCF-7-derived exosomes were annotated in the HPA database; all 5 major exosomal proteins were also found in the HPA database (Figure 7A).
Similar results were obtained for protein cargo of exosomes from the blood of triple-positive BCPs and from BT-474 conditioned medium (Figure 7B). Thus, more BC-associated proteins were found in the composition of exosomes from the blood of BCPs than in the exosomes from the conditioned medium of cells mimicking the corresponding cancer subtype (89% and 81% for luminal A BC and MCF-7 cells and 86% and 80% for triple-positive BC and BT-474 cells, respectively).

4. Discussion

Tumor exosomes able to transport biologically active molecules (RNA, proteins, and metabolites) in recipient cells have been recognized as fundamental mediators of cell-to-cell communication in cancer, including BC. Since the molecular cargo of exosomes reflects the composition of the parent cell, in the field of molecular diagnostics, great potential is associated with the identification of tumor-specific signatures in the composition of exosomes for the development of a method for the diagnosis of malignant neoplasms by liquid biopsy [25,26]. One of the undoubted advantages of exosomal proteome research is the possibility of removing ballast proteins from blood plasma and increasing the concentration of tumor-specific proteins, including membrane proteins. It should be noted that the membrane of vesicles protects their contents from the action of proteases and nucleases, and vesicle preparations are stable and can be stored for a long time in laboratory conditions [27].
The search for tumor markers in exosomes is complicated by the high individual variability of the exosome proteome, even in the group of healthy donors. In a comparison of the proteomic profiles of blood plasma exosomes from fifteen clinically healthy donors, only 9 out of 109 identified exosomal proteins were present in all samples [28]. Our profiling of the proteome of HUVEC-derived exosomes partially confirms these data: only 29% of proteins within primary endotheliocytes were common to the three human umbilical vein donors.
To avoid the problem of low reproducibility between different samples, as well as to solve the problem of the amount of protein needed for mass spectrometry, most researchers work with cell culture exosomes, which is reflected in the Vesiclepedia database. As a result, the authors provide data on thousands of proteins in the composition of exosomes obtained from the conditioned media of various cell lines. In particular, Altelaar’s group successfully identified exosome proteomes from cell lines mimicking triple-negative (BT-549, Hs578T, LM2, MDA-MB-231), HER-2-positive (HCC1419, HCC1954, JIMT1, SKBR-3), and luminal A (MCF-7) BC subtypes [29], identifying 4648 proteins. However, the authors did not take into account the fact that if the size of exosomes is less than 150 nm, the vesicle cannot contain more than 100 proteins. Thus, there is no point in obtaining excessively large quantities of exosomes and describing proteins that can be detected in blood exosomes with very low probability.
Modern BC diagnostic methods (mammography, breast ultrasound, and MRI) can detect neoplasms larger than 0.5 cm; however, these tests are often ambiguous and have documented drawbacks [3]. For simplicity of calculation, we assume that only the surface cells of the tumor secrete exosomes to the external space. Then, considering that the surface area of a 5 mm diameter sphere-shaped tumor is 78.5 mm2, and taking into account the average diameter of a breast carcinoma cell of 20 µm, a quarter of a million cells will be located on the outer surface of the tumor. Taking into account the above calculations, in the current work, for the analysis of exosome proteins by mass spectrometry, the proteins in exosomes secreted by 500,000 MCF-7 or BT-474 cells were applied to a gel and separated. The obtained data on the proteomic profiles of exosomes from the conditioned media of breast carcinoma lines correspond to the number of exosomes secreted by the tumor at stage T1. A comparative analysis of proteins of secreted by primary endotheliocyte exosomes and breast carcinoma cell exosomes showed that it is possible to neglect the contribution of the endotheliocyte exosome proteome when searching for tumor markers in the composition of blood exosomes. Moreover, since the five proteins CD9, CD24, CD63, CD81, and MMP9 are universal and present in exosomes secreted by both primary endotheliocytes and breast carcinoma cells, they cannot be part of the diagnostic panels being developed.
To confirm that EVs are ideal diagnostic tools, Hoshino’s group analyzed the proteomes of EVs from different sources. It was shown that EV proteins from human plasma overlapped best with human serum-derived EVs (r2 = 0.92), followed by human bone marrow (r2 = 0.65) and lymphatic fluid EVs (r2 = 0.64); these proteins correlated least with human cell line- (r2 = 0.15) and tissue explant-derived EVs (r2 = 0.24), suggesting that the complexity of plasma and lymph EV proteomes may drive the divergence of tissue EV proteomes [30]. Our study also revealed an extremely weak correlation between exosome proteins secreted by MCF-7 cells and blood exosomes from patients with the luminal A subtype of BC, as well as between exosome proteins secreted by BT-474 cells and blood exosomes from patients with the triple-positive subtype of BC. However, when comparing protein profiles of blood exosomes from patients and tissue proteins, a significant coincidence of proteins was found. It should be noted that breast tumors are characterized by significant variability in their cellular composition, as well as histological, expression, and genotypic heterogeneity. In particular, the intratumoral morphologic heterogeneity of invasive breast carcinoma of a nonspecific type, the most common histologic form of BC (incidence rate up to 80%), has been described [31]. As a consequence, exosomes secreted by tumor cells have a more diverse composition than exosomes originating from MCF-7 or BT-474 cells. In addition, in the body, besides cancer cells, exosomes are secreted by other cells, in particular cells from the tumor microenvironment [32]. Thus, the exosomal content in the blood of BCPs at the T1 stage reflects the cancer-associated changes occurring not only in the developing primary tumor but also the tumor microenvironment. Despite previous studies searching for protein biomarkers within exosomes secreted by breast carcinoma cell lines [11,33,34], there is no consensus on exosomal tumor markers due to limited EV proteomic datasets from human samples and appropriate controls for data analysis and interpretation.
It should be noted that only three (GTSE1, VAV3, and SOCS3) proteins identified in our study were deposited simultaneously in Vesiclepedia and were shown to be strongly associated with BC according to the HPA database. Specifically, for the G2 and S phase-expressed-1 (GTSE1) protein, its expression is known to be significantly upregulated in BC tissues and cell lines, with high levels of this protein correlating with tumor prevalence and poor prognosis [35]. It has been shown that GTSE1 promotes tumor cell proliferation by activating the AKT signaling pathway and enhances metastasis through the regulation of the epithelial–mesenchymal transition [36].
Another protein, VAV3, has been identified as a critical player in the modulation of immune responses and cancer cell behavior. VAV3 has been shown to be involved in the activation of Rho/Rac signaling pathways that regulate cytoskeleton dynamics, cell migration, and invasion. VAV3 is known to be upregulated in various types of cancer, including breast cancer [37]. VAV3 has been found to be associated with poor prognosis and aggressive breast cancer subtypes [38].
SOCS3 is a critical negative regulator of cytokine signaling pathways, primarily modulating the Janus kinase/signal transducer and activator of transcription (JAK/STAT) axis. It achieves this through direct binding to phosphorylated tyrosine residues on activated receptors, thereby inhibiting STAT activation [39]. The dysregulation of SOCS3 expression is associated with aberrant JAK/STAT signaling, contributing to the development and progression of several cancers [40]. SOCS3 functions as a tumor suppressor in many contexts, where its downregulation correlates with enhanced tumor growth, angiogenesis, and immune evasion [41]. SOCS3 also exerts broader regulatory effects on other oncogenic pathways, including PI3K/AKT and MAPK signaling, by interacting with receptor tyrosine kinases and downstream effectors. These interactions modulate cell proliferation, survival, and migration [39].
Taken together, our results support the idea that tumor-associated proteins should be sought not in exosomes secreted by cell lines but in the composition of blood exosomes from cancer patients, while the contribution of endotheliocyte exosomes to the total pool of blood exosomes can be neglected.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/diagnostics15081028/s1, Table S1: Proteins identified in exosomes secreted by HUVECs; Table S2: Proteins identified in exosomes secreted by BT-474 cells; Table S3: Proteins identified in exosomes secreted by MCF-7 cells; Table S4: Exosomal proteins identified in the blood of luminal A BCPs; Table S5: Exosomal proteins identified in the blood of triple positive BCPs.

Author Contributions

Conceptualization, Y.T. and S.T.; methodology, A.S., L.Y., O.T., A.G. and N.Y.; formal analysis, A.S.; investigation, A.S., L.Y., K.P., O.T., A.G. and N.Y.; resources, Y.T.; data curation, S.T.; writing—original draft preparation, A.S., L.Y. and S.T.; writing—review and editing, Y.T. and S.T.; project administration, L.Y. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by a grant from the Russian Science Foundation, N23-25-00462, https://rscf.ru/project/23-25-00462/, accessed on 25 January 2023.

Institutional Review Board Statement

This study was conducted in accordance with the Declaration of Helsinki and was approved by the Local Ethics Committee of the Institute of Chemical Biology and Fundamental Medicine (the protocol N3 from 2 September 2024).

Informed Consent Statement

Informed consent was obtained from all subjects involved in the study.

Data Availability Statement

The data presented in this study are available on request from the corresponding author.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Bray, F.; Laversanne, M.; Sung, H.; Ferlay, J.; Siegel, R.L.; Soerjomataram, I.; Jemal, A. Global cancer statistics 2022: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 2024, 74, 229–263. [Google Scholar] [CrossRef] [PubMed]
  2. Pu, Q.; Gao, H. The Role of the Tumor Microenvironment in Triple-Positive Breast Cancer Progression and Therapeutic Resistance. Cancers 2023, 15, 5493. [Google Scholar] [CrossRef] [PubMed]
  3. Shefer, A.; Yalovaya, A.; Tamkovich, S. Exosomes in breast cancer: Involvement in tumor dissemination and prospects for liquid biopsy. Int. J. Mol. Sci. 2022, 23, 8845. [Google Scholar] [CrossRef] [PubMed]
  4. Bandini, E.; Rossi, T.; Scarpi, E.; Gallerani, G.; Vannini, I.; Salvi, S.; Azzali, I.; Melloni, M.; Salucci, S.; Battistelli, M.; et al. Early Detection and Investigation of Extracellular Vesicles Biomarkers in Breast Cancer. Front. Mol. Biosci. 2021, 8, 732900. [Google Scholar] [CrossRef]
  5. Johnson, K.S.; Conant, E.F.; Soo, M.S. Molecular Subtypes of Breast Cancer: A Review for Breast Radiologists. J. Breast Imaging 2021, 3, 12–24. [Google Scholar] [CrossRef]
  6. Bilous, M. Breast core needle biopsy: Issues and controversies. Mod. Pathol. 2010, 23 (Suppl. S2), S36–S45. [Google Scholar] [CrossRef]
  7. Lee, Y.; Ni, J.; Wasinger, V.C.; Graham, P.; Li, Y. Comparison Study of Small Extracellular Vesicle Isolation Methods for Profiling Protein Biomarkers in Breast Cancer Liquid Biopsies. Int. J. Mol. Sci. 2023, 24, 15462. [Google Scholar] [CrossRef]
  8. Yunusova, N.V.; Tugutova, E.A.; Tamkovich, S.N.; Kondakova, I.V. The role of exosomal tetraspanins and proteases in tumor progression. Biochem. Suppl. Ser. B Biomed. Chem. 2018, 12, 191–202. [Google Scholar]
  9. Neagu, A.-N.; Whitham, D.; Bruno, P.; Morrissiey, H.; Darie, C.A.; Darie, C.C. Omics-Based Investigations of Breast Cancer. Molecules 2023, 28, 4768. [Google Scholar] [CrossRef]
  10. Risha, Y.; Minic, Z.; Ghobadloo, S.M.; Berezovski, M.V. The proteomic analysis of breast cell line exosomes reveals disease patterns and potential biomarkers. Sci. Rep. 2020, 10, 13572. [Google Scholar] [CrossRef]
  11. Gangoda, L.; Liem, M.; Ang, C.S.; Keerthikumar, S.; Adda, C.G.; Parker, B.S.; Mathivanan, S. Proteomic Profiling of Exosomes Secreted by Breast Cancer Cells with Varying Metastatic Potential. Proteomics 2017, 17, 1600370. [Google Scholar] [CrossRef] [PubMed]
  12. Stepanova, A.O.; Laktionov, P.P.; Cherepanova, A.V.; Chernonosova, V.S.; Shevelev, G.Y.; Zaporozhchenko, I.A.; Karaskov, A.M.; Laktionov, P.P. General Study and Gene Expression Profiling of Endotheliocytes Cultivated on Electrospun Materials. Materials 2019, 12, 4082. [Google Scholar] [CrossRef] [PubMed]
  13. Cherepanova, A.V.; Bushuev, A.V.; Kharkova, M.V.; Vlassov, V.V.; Laktionov, P.P. DNA inhibits dsRNA-induced secretion of pro-inflammatory cytokines by gingival fibroblasts. Immunobiology 2013, 218, 272–280. [Google Scholar] [CrossRef]
  14. Tutanov, O.; Orlova, E.; Proskura, K.; Grigor’eva, A.; Yunusova, N.; Tsentalovich, Y.; Alexandrova, A.; Tamkovich, S. Proteomic Analysis of Blood Exosomes from Healthy Females and Breast Cancer Patients Reveals an Association between Different Exosomal Bioactivity on Non-tumorigenic Epithelial Cell and Breast Cancer Cell Migration In Vitro. Biomolecules 2020, 10, 495. [Google Scholar] [CrossRef]
  15. Tamkovich, S.; Tutanov, O.; Efimenko, A.; Grigor’eva, A.; Ryabchikova, E.; Kirushina, N.; Vlassov, V.; Tkachuk, V.; Laktionov, P. Blood Circulating Exosomes Contain Distinguishable Fractions of Free and Cell-Surface-Associated Vesicles. Curr. Mol. Med. 2019, 19, 273–285. [Google Scholar] [CrossRef]
  16. Tamkovich, S.N.; Yunusova, N.V.; Tugutova, E.; Somov, A.K.; Proskura, K.V.; Kolomiets, L.A.; Stakheeva, M.N.; Grigor’eva, A.E.; Laktionov, P.P.; Kondakova, I.V. Protease cargo in circulating exosomes of breast cancer and ovarian cancer patients. Asian Pac. J. Cancer Prev. 2019, 20, 255–262. [Google Scholar] [CrossRef]
  17. Deutsch, E.W.; Lane, L.; Overall, C.M.; Bandeira, N.; Baker, M.S.; Pineau, C.; Moritz, R.L.; Corrales, F.; Orchard, S.; Van Eyk, J.E.; et al. Human Proteome Project Mass Spectrometry Data Interpretation Guidelines 3.0. J. Proteome Res. 2019, 18, 4108–4116. [Google Scholar] [CrossRef]
  18. Welsh, J.A.; Goberdhan, D.C.I.; O’Driscoll, L.; Buzas, E.I.; Blenkiron, C.; Bussolati, B.; Cai, H.; Di Vizio, D.; Driedonks, T.A.P.; Erdbrügger, U.; et al. Minimal information for studies of extracellular vesicles (MISEV2023): From basic to advanced approaches. J. Extracell. Vesicles. 2024, 13, e12404. [Google Scholar] [CrossRef]
  19. Vesiclepedia Database. Available online: www.microvesicles.org (accessed on 30 November 2024).
  20. Tutanov, O.; Shtam, T.; Grigor’eva, A.; Tupikin, A.; Tsentalovich, Y.; Tamkovich, S. Blood Plasma Exosomes Contain Circulating DNA in Their Crown. Diagnostics 2022, 12, 854. [Google Scholar] [CrossRef]
  21. Zheng, J.; Liu, C.; Shi, J.; Wen, K.; Wang, X. AIM2 Inhibits the Proliferation, Invasion and Migration, and Promotes the Apoptosis of Osteosarcoma Cells by Inactivating the PI3K/AKT/mTOR Signaling Pathway. Mol. Med. Rep. 2022, 25, 53. [Google Scholar] [CrossRef]
  22. Corona, R.I.; Seo, J.H.; Lin, X.; Hazelett, D.J.; Reddy, J.; Fonseca, M.A.S.; Abassi, F.; Lin, Y.G.; Mhawech-Fauceglia, P.Y.; Shah, S.P.; et al. Non-Coding Somatic Mutations Converge on the PAX8 Pathway in Ovarian Cancer. Nat. Commun. 2020, 11, 2020. [Google Scholar] [CrossRef] [PubMed]
  23. Yang, M.; Wang, A.; Li, C.; Sun, J.; Yi, G.; Cheng, H.; Liu, X.; Wang, Z.; Zhou, Y.; Yao, G.; et al. Methylation-Induced Silencing of ALDH2 Facilitates Lung Adenocarcinoma Bone Metastasis by Activating the MAPK Pathway. Front. Oncol. 2020, 10, 1141. [Google Scholar] [CrossRef] [PubMed]
  24. Du, C.; Ma, X.; Meruvu, S.; Hugendubler, L.; Mueller, E. The Adipogenic Transcriptional Cofactor ZNF638 Interacts with Splicing Regulators and Influences Alternative Splicing. J. Lipid Res. 2014, 55, 1886–1896. [Google Scholar] [CrossRef] [PubMed]
  25. Bates, M.; Mohamed, B.M.; Lewis, F.; O’Toole, S.; O’Leary, J.J. Biomarkers in high grade serous ovarian cancer. Biochim. Biophys. Acta Rev. Cancer. 2024, 1879, 189224. [Google Scholar] [CrossRef]
  26. Bao, H.; Min, L.; Bu, F.; Wang, S.; Meng, J. Recent advances of liquid biopsy: Interdisciplinary strategies toward clinical decision-making. Interdiscip. Med. 2023, 1, e20230021. [Google Scholar] [CrossRef]
  27. Zhang, Y.; Bi, J.; Huang, J.; Tang, Y.; Du, S.; Li, P. Exosome: A Review of Its Classification, Isolation Techniques, Storage, Diagnostic and Targeted Therapy Applications. Int. J. Nanomedicine 2020, 15, 6917–6934. [Google Scholar] [CrossRef]
  28. Bastos-Amador, P.; Royo, F.; Gonzalez, E.; Conde-Vancells, J.; Palomo-Diez, L.; Borras, F.E.; Falcon-Perez, J.M. Proteomic analysis of microvesicles from plasma of healthy donors reveals high individual variability. J. Proteomics 2012, 75, 3574–3584. [Google Scholar] [CrossRef]
  29. Rontogianni, S.; Synadaki, E.; Li, B.; Liefaard, M.C.; Lips, E.H.; Wesseling, J.; Wu, W.; Altelaar, M. Proteomic profiling of extracellular vesicles allows for human breast cancer subtyping. Commun. Biol. 2019, 2, 325. [Google Scholar] [CrossRef]
  30. Hoshino, A.; Kim, H.S.; Bojmar, L.; Gyan, K.E.; Cioffi, M.; Hernandez, J.; Zambirinis, C.P.; Rodrigues, G.; Molina, H.; Heissel, S.; et al. Extracellular Vesicle and Particle Biomarkers Define Multiple Human Cancers. Cell 2020, 182, 1044–1061.e18. [Google Scholar] [CrossRef]
  31. Gerashchenko, T.S.; Zavyalova, M.V.; Denisov, E.V.; Krakhmal, N.V.; Pautova, D.N.; Litviakov, N.V.; Vtorushin, S.V.; Cherdyntseva, N.V.; Perelmuter, V.M. Intratumoral Morphological Heterogeneity of Breast Cancer as an Indicator of the Metastatic Potential and Tumor Chemosensitivity. Acta Naturae 2017, 9, 56–67. [Google Scholar] [CrossRef]
  32. Eguchi, T.; Sheta, M.; Fujii, M.; Calderwood, S.K. Cancer extracellular vesicles, tumoroid models, and tumor microenvironment. Semin. Cancer Biol. 2022, 86, 112–126. [Google Scholar] [CrossRef] [PubMed]
  33. Kugeratski, F.G.; Hodge, K.; Lilla, S.; McAndrews, K.M.; Zhou, X.; Hwang, R.F.; Zanivan, S.; Kalluri, R. Quantitative proteomics identifies the core proteome of exosomes with syntenin-1 as the highest abundant protein and a putative universal biomarker. Nat. Cell Biol. 2021, 23, 631–641. [Google Scholar] [CrossRef] [PubMed]
  34. Gelsomino, L.; Barone, I.; Caruso, A.; Giordano, F.; Brindisi, M.; Morello, G.; Accattatis, F.M.; Panza, S.; Cappello, A.R.; Bonofiglio, D.; et al. Proteomic Profiling of Extracellular Vesicles Released by Leptin-Treated Breast Cancer Cells: A Potential Role in Cancer Metabolism. Int. J. Mol. Sci. 2022, 23, 12941. [Google Scholar] [CrossRef] [PubMed]
  35. Bao, Z.; Cheng, J.; Zhu, J.; Ji, S.; Gu, K.; Zhao, Y.; Yu, S.; Meng, Y. Using Weighted Gene Co-Expression Network Analysis to Identify Increased MND1 Expression as a Predictor of Poor Breast Cancer Survival. Int. J. Gen. Med. 2022, 15, 4959–4974. [Google Scholar] [CrossRef]
  36. Lin, F.; Xie, Y.J.; Zhang, X.K.; Huang, T.J.; Xu, H.F.; Mei, Y.; Liang, H.; Hu, H.; Lin, S.T.; Luo, F.F.; et al. GTSE1 is involved in breast cancer progression in p53 mutation-dependent manner. J. Exp. Clin. Cancer Res. 2019, 38, 152. [Google Scholar] [CrossRef]
  37. Ojala, V.K.; Knittle, A.M.; Kirjalainen, P.; Merilahti, J.A.M.; Kortesoja, M.; Tvorogov, D.; Vaparanta, K.; Lin, S.; Kast, J.; Pulliainen, A.T.; et al. The guanine nucleotide exchange factor VAV3 participates in ERBB4-mediated cancer cell migration. J. Biol. Chem. 2020, 295, 11559–11571. [Google Scholar] [CrossRef]
  38. Chang, Y.T.; Hong, Z.J.; Tsai, H.H.; Feng, A.C.; Huang, T.Y.; Yu, J.C.; Hsu, K.F.; Huang, C.C.; Lin, W.Z.; Chu, C.M.; et al. Hub metastatic gene signature and risk score of breast cancer patients with small tumor sizes using WGCNA. Breast Cancer 2024, 31, 1114–1129. [Google Scholar] [CrossRef]
  39. Thacker, G.; Henry, S.; Nandi, A.; Debnath, R.; Singh, S.; Nayak, A.; Susnik, B.; Boone, M.M.; Zhang, Q.; Kesmodel, S.B.; et al. Immature natural killer cells promote progression of triple-negative breast cancer. Sci. Transl. Med. 2023, 15, eabl4414. [Google Scholar] [CrossRef]
  40. Chu, J.; Hu, X.C.; Li, C.C.; Li, T.Y.; Fan, H.W.; Jiang, G.Q. KLF14 alleviated breast cancer invasion and M2 macrophages polarization through modulating SOCS3/RhoA/Rock/STAT3 signaling. Cell Signal 2022, 92, 110242. [Google Scholar] [CrossRef]
  41. Li, X.; Peng, B.; Li, J.; Tian, M.; He, L. Unleashing Breast Cancer Progression: miR-455-5p’s Targeting of SOCS3 Drives Proliferation, Migration, and Invasion. Protein Pept. Lett. 2023, 30, 992–1000. [Google Scholar] [CrossRef]
Figure 1. Exosome characterization by TEM and flow cytometry analysis. Overview of exosome preparation and expression of CD63 on CD9-positive vesicles obtained from (A) conditioned medium from HUVECs; (B) conditioned medium from MCF-7 cells; (C) conditioned medium from BT-474 cells; (D) blood from patients with luminal A BC; and (E) blood from patients with triple-positive BC. Scale bars correspond to 100 nm. Electron microscopy, negative staining by phosphotungstate acid. For flow cytometry, mean MFI values are shown. Negative control (F) (latex beads labeled with anti-CD9 and anti-CD63 FITC antibody).
Figure 1. Exosome characterization by TEM and flow cytometry analysis. Overview of exosome preparation and expression of CD63 on CD9-positive vesicles obtained from (A) conditioned medium from HUVECs; (B) conditioned medium from MCF-7 cells; (C) conditioned medium from BT-474 cells; (D) blood from patients with luminal A BC; and (E) blood from patients with triple-positive BC. Scale bars correspond to 100 nm. Electron microscopy, negative staining by phosphotungstate acid. For flow cytometry, mean MFI values are shown. Negative control (F) (latex beads labeled with anti-CD9 and anti-CD63 FITC antibody).
Diagnostics 15 01028 g001
Figure 2. Venn–Euler diagrams of proteins in exosomes from HUVECs from three donors (A) and overlap with the Vesiclepedia database (B).
Figure 2. Venn–Euler diagrams of proteins in exosomes from HUVECs from three donors (A) and overlap with the Vesiclepedia database (B).
Diagnostics 15 01028 g002
Figure 3. PPI network of 18 proteins from HUVEC-derived exosomes. PPI networks were plotted with STRING (Available online: http://string-db.org/ (accessed on 10 January 2025)) with the following settings: minimum interaction score—high confidence (0.400); active interaction sources—text mining, experiments, databases, co-expression.
Figure 3. PPI network of 18 proteins from HUVEC-derived exosomes. PPI networks were plotted with STRING (Available online: http://string-db.org/ (accessed on 10 January 2025)) with the following settings: minimum interaction score—high confidence (0.400); active interaction sources—text mining, experiments, databases, co-expression.
Diagnostics 15 01028 g003
Figure 4. Venn–Euler diagrams of proteins in exosomes from HUVECs, BT-474 cells, and MCF-7 cells (A) and overlap with the Vesiclepedia database (B).
Figure 4. Venn–Euler diagrams of proteins in exosomes from HUVECs, BT-474 cells, and MCF-7 cells (A) and overlap with the Vesiclepedia database (B).
Diagnostics 15 01028 g004
Figure 5. PPI network of 65 proteins from cancer cell-derived exosomes. PPI networks were plotted with STRING (Available online: http://string-db.org/, accessed on 10 January 2025) with the following settings: minimum interaction score—high confidence (0.400); active interaction sources—text mining, experiments, databases, co-expression.
Figure 5. PPI network of 65 proteins from cancer cell-derived exosomes. PPI networks were plotted with STRING (Available online: http://string-db.org/, accessed on 10 January 2025) with the following settings: minimum interaction score—high confidence (0.400); active interaction sources—text mining, experiments, databases, co-expression.
Diagnostics 15 01028 g005
Figure 6. Venn–Euler diagram of proteins in exosomes from the blood of BCPs with luminal A and triple-positive subtypes and those in the Vesiclepedia database.
Figure 6. Venn–Euler diagram of proteins in exosomes from the blood of BCPs with luminal A and triple-positive subtypes and those in the Vesiclepedia database.
Diagnostics 15 01028 g006
Figure 7. Venn–Euler diagram of exosomal proteins from cancer cells and blood samples from BCPs with luminal A (A) and triple-positive (B) subtypes compared with the HPA database, composed using QuickGO 2.0 and FunRich 3.13 software.
Figure 7. Venn–Euler diagram of exosomal proteins from cancer cells and blood samples from BCPs with luminal A (A) and triple-positive (B) subtypes compared with the HPA database, composed using QuickGO 2.0 and FunRich 3.13 software.
Diagnostics 15 01028 g007
Table 1. Clinical characteristics of untreated BCPs.
Table 1. Clinical characteristics of untreated BCPs.
SubtypeHormonal StatusHER2/Neo StatusAgeTNMKi-67GInfiltrative Ductal Carcinoma
Luminal A
n = 5
ER+ PR+Negative6110010–15%2Yes
6110012–14%2Yes
562005–10%2Yes
592005–10%2Yes
612105%2Yes
Triple-positive
n = 8
ER+ PR+Positive5210010%2Yes
6210010–15%2Yes
6610015%2Yes
681005–10%2Yes
691005–10%2Yes
442005%2Yes
612x *015–17%2Yes
6720020–25%2Yes
* no information available.
Table 2. HUVEC-secreted exosomal proteins not deposited in the Vesiclepedia database.
Table 2. HUVEC-secreted exosomal proteins not deposited in the Vesiclepedia database.
Uniprot IDProtein NameGene Name
A0A0A0MS01Probable non-functional T-cell receptor gamma variable 10TRGV10
A0A1B0GUA6Putative coiled-coil domain-containing protein 195CCDC195
MTRNR2L13Humanin-like 13S4R3P1
Q8IU53Protein CASC2, isoforms 1/2CASC2
Q96GV9Macrophage immunometabolism regulatorMACIR
Q9BXU9Calcium-binding protein 8CALN1
Q9HBF5Suppressor of tumorigenicity 20 proteinST20
Table 3. Analysis of the molecular functions in which HUVEC-derived exosomal proteins are involved.
Table 3. Analysis of the molecular functions in which HUVEC-derived exosomal proteins are involved.
Molecular Functionsp-ValueGene Name
Calcium ion binding0.1514CALN1
Carboxypeptidase activity0.0184CPA3
Catalytic activity0.3793CD81
Defense/immunity protein activity0.0525GNLY
Glutathione transferase activity0.0184GSTT2
GTPase activity0.1790RAB3D
Inward rectifier channel0.0123KCNJ5
Metallopeptidase activity0.0035ADAM10
MMP9
RNA binding0.2786RBM10
Transporter activity0.0902ALB
GABARAPL2
Ubiquitin-specific protease activity0.2857PSMD10
Table 4. Analysis of the biological pathways in which HUVEC-derived exosomal proteins are involved.
Table 4. Analysis of the biological pathways in which HUVEC-derived exosomal proteins are involved.
Biological Pathwaysp-ValueGene Name
a6b1 and a6b4 integrin signaling0.0437CD9
Adaptive immune system0.0341CD81
PSMD10
Alpha4 beta1 integrin signaling events0.0412CD81
Bile acid and bile salt metabolism0.0338ALB
HDL-mediated lipid transport0.0189ALB
Lipoprotein metabolism0.0338ALB
Metabolism of mRNA0.0333PSMD10
Metabolism of RNA0.0466PSMD10
Osteopontin-mediated events0.0363MMP9
Platelet degranulation0.0351CD63
Recycling of bile acids and salts0.0139ALB
Transport of organic anions0.0127ALB
Transport of vitamins, nucleosides, and related molecules0.0375ALB
Table 5. MCF-7 cell-secreted exosomal proteins not deposited in the Vesiclepedia database.
Table 5. MCF-7 cell-secreted exosomal proteins not deposited in the Vesiclepedia database.
Uniprot IDProtein NameGene Name
P46527Cyclin-dependent kinase inhibitor 1BCDKN1B
P59037Putative uncharacterized protein encoded by LINC00313LINC00313
P61236Protein yippee-like 3YPEL3
Q17RP2Tigger transposable element-derived protein 6TIGD6
Q32M92Uncharacterized protein C15orf32C15orf32
Q5SZD1Uncharacterized protein C6orf141C6orf141
Q5T035Putative uncharacterized protein FAM120A2PC9orf129
Q86UT8Centrosomal AT-AC splicing factorCENATAC
Q8IX90Spindle and kinetochore-associated protein 3SKA3
Q8N111Cell cycle exit and neuronal differentiation protein 1CEND1
Q8N7M0Dynein light chain Tctex-type 5DYNLT5
Q8TF44C2 calcium-dependent domain-containing protein 4CC2CD4C
Q96KF7Small integral membrane protein 8SMIM8
Q9Y584Mitochondrial import inner membrane translocase subunit Tim22TIMM22
Table 6. BT-474 cell-secreted exosomal proteins not deposited in the Vesiclepedia database.
Table 6. BT-474 cell-secreted exosomal proteins not deposited in the Vesiclepedia database.
Uniprot IDProtein NameGene Name
A0A0B4J271T-cell receptor alpha variable 12–3TRAV12-3
A6NLC8TATA-box binding protein associated factor 11 like protein 2TAF11L2
O15537RetinoschisinRS1
P0DMT0MyoregulinMRLN
P57077MAP3K7 C-terminal-like proteinMAP3K7CL
Q16384Protein SSX1SSX1
Q5TZK3No nameFAM74A4
Q5TZK3No nameFAM74A6
Q96MW1Coiled-coil domain-containing protein 43CCDC43
Q9BZI7Regulator of nonsense transcripts 3BUPF3B
Q9HAW0Transcription factor IIIB 50 kDa subunitBRF2
Table 7. Analysis of the molecular functions in which MCF-7 cell-derived exosomal proteins are involved.
Table 7. Analysis of the molecular functions in which MCF-7 cell-derived exosomal proteins are involved.
Molecular Functionsp-ValueGene Name
Acyltransferase activity0.1879BLOC1S1
Auxiliary transport protein activity0.5822VAMP8
Calcium ion binding0.0122CCBE1
KCNIP3
NPTX2
Catalytic activity0.7539CD81
Cell adhesion molecule activity0.6068MPZ
Defense/immunity protein activity0.1467ORM1
DNA binding0.5097ZNF114
ZNF648
Endopeptidase activity0.0052ATG4C
GTPase activity0.1129DIRAS1
GEM
Hydrolase activity0.4114CANT1
Isomerase activity0.1104PPIA
Kinase regulator activity0.0580CDKN1B
Metallopeptidase activity0.0282ADAM10
MMP9
Protein serine/threonine kinase activity0.5453PRKCH
Protein tyrosine/serine/threonine phosphatase activity0.0917PTPMT1
Receptor signaling complex scaffold activity0.5698BAG2
Ribonuclease activity0.1173REXO2
RNA binding0.2453CSTF1
SRSF6
Structural constituent of ribosome0.3272RPS27
Structural molecule activity0.5052TEKT3
Transcription factor activity0.8934CRX
Transcription regulator activity0.8905PARP15
Transporter activity0.7813TIMM22
Ubiquitin-specific protease activity0.2559COPS5
RBBP6
Voltage-gated ion channel activity0.2873KCNC4
Table 8. Analysis of the biological pathways in which MCF-7 cell-derived exosomal proteins are involved *.
Table 8. Analysis of the biological pathways in which MCF-7 cell-derived exosomal proteins are involved *.
Biological Pathwaysp-ValueGene Name
AKT phosphorylates targets in the cytosol0.0241CDKN1B
Clathrin-derived vesicle budding0.0035VAMP8
BLOC1S1
Cleavage of growing transcript in the termination region0.0058SRSF6
Integrins in angiogenesis0.0126MMP9
Membrane trafficking0.0210VAMP8
BLOC1S1
mRNA 3′-end processing0.0037SRSF6
mRNA splicing0.0330SRSF6
mRNA splicing—major pathway0.0330SRSF6
Notch signaling pathway0.0260ADAM10
CDKN1B
Notch-mediated HES/HEY network0.0260ADAM10
CDKN1B
Platelet activation, signaling, and aggregation0.0056CD63
PRKCH
Platelet degranulation0.0025CD63
Post-elongation processing of intron-containing pre-mRNA0.0037SRSF6
Post-elongation processing of the transcript0.0058SRSF6
Receptor–ligand binding initiates the second proteolytic cleavage of Notch receptor0.0241ADAM10
Response to elevated platelet cytosolic Ca2+0.0048CD63
RNA polymerase II transcription0.0297SRSF6
RNA polymerase II transcription termination0.0058SRSF6
Signaling by EGFR0.02810.0281ADAM10
CDKN1B
Signaling by SCF-KIT0.0133CDKN1B
MMP9
Trans-Golgi network vesicle budding0.0035VAMP8
BLOC1S1
* biological pathways universal to exosomal proteins secreted by cancer cells are denoted in italics.
Table 9. Analysis of the molecular functions in which BT-474 cell-derived exosomal proteins are involved.
Table 9. Analysis of the molecular functions in which BT-474 cell-derived exosomal proteins are involved.
Molecular Functionsp-ValueGene Name
Calcium ion binding0.2343CALML5
Catalytic activity0.5393CD81
Cell adhesion molecule activity0.4032CLEC2B
Metallopeptidase activity0.0092ADAM10
MMP9
Oxidoreductase activity0.2072NDUFB1
Receptor binding0.1696EFNB2
RNA binding0.0151RBMX
SRSF5
UPF3B
Storage protein0.0043FTL
Transcription factor activity0.7099BRF2
Transcription regulator activity0.7055SSX1
Ubiquitin-specific protease activity0.4213SUMO3
Table 10. Analysis of the biological pathways in which BT-474 cell-derived exosomal proteins are involved *.
Table 10. Analysis of the biological pathways in which BT-474 cell-derived exosomal proteins are involved *.
Biological Pathwaysp-ValueGene Name
Cleavage of growing transcript in the termination region0.0134SRSF5
UPF3B
CXCR4-mediated signaling events0.0424MMP9
Formation and maturation of mRNA transcript0.0396SRSF5
RBMX
UPF3B
Gene expression0.0037SRSF5
RBMX
UPF3B
Hypoxic and oxygen homeostasis regulation of HIF-1α0.0426BNIP3
mRNA 3′-end processing0.0085SRSF5
UPF3B
mRNA processing0.0260SRSF5
RBMX
UPF3B
mRNA splicing0.0094SRSF5
RBMX
UPF3B
Post-elongation processing of intron-containing pre-mRNA0.0085SRSF5
UPF3B
Post-elongation processing of the transcript0.0134SRSF5
UPF3B
Processing of capped intron-containing pre-mRNA0.0186SRSF5
RBMX
UPF3B
Receptor–ligand binding initiates the second proteolytic cleavage of Notch receptor0.0366ADAM10
Respiratory electron transport0.0388NDUFB1
RNA polymerase II transcription termination0.0134SRSF5
UPF3B
Transcription0.0359BRF2
SRSF5
UPF3B
Transport of mature mRNA derived from an intron-containing transcript0.0185SRSF5
UPF3B
* biological pathways universal to exosome proteins secreted by cancer cells are highlighted in italics.
Table 11. BCP blood exosomal proteins not deposited in the Vesiclepedia database.
Table 11. BCP blood exosomal proteins not deposited in the Vesiclepedia database.
Uniprot IDProtein NameGene NameBC Subtype
A0A1B0GVM6Uncharacterized protein C11orf97C11orf97TP
A0A589T-cell receptor beta variableTRBV4-3Lum A
O14543Suppressor of cytokine signaling 3SOCS3TP
O43557Tumor necrosis factor ligand superfamily member 14TNFSF14Lum A
O60397Putative cytochrome c oxidase subunit 7A3, mitochondrialCOX7A2P2TP
P0CE72Oncomodulin-1OCMLum A
P0DUQ2PRAME family member 9PRAMEF9Lum A
P16233Pancreatic triacylglycerol lipasePNLIPLum A
P48167Glycine receptor subunit betaGLRBLum A
P49761Dual specificity protein kinase CLK3CLK3Lum A&TP
P56373P2X purinoceptor 3P2RX3Lum A&TP
Q13424Alpha-1-syntrophinSNTA1Lum A
Q15776Zinc finger protein with KRAB and SCAN domains 8ZKSCAN8Lum A&TP
Q2M218Zinc finger protein 630ZNF630TP
Q52M93Zinc finger protein 585BZNF585BTP
Q53FZ2Acyl-coenzyme A synthetase ACSM3, mitochondrialACSM3TP
Q5CZ79Ankyrin repeat domain-containing protein 20BANKRD20A8PTP
Q5VZ18SH2 domain-containing adapter protein ESHETP
Q7RTT3Putative protein SSX9SSX9PTP
Q8IUS5Epoxide hydrolase 4EPHX4TP
Q8NDD1Uncharacterized protein C1orf131C1orf131Lum A
Q8NEY8Periphilin-1PPHLN1TP
Q9H4Q4PR domain zinc finger protein 12PRDM12Lum A
Q9Y4E5E3 SUMO-protein ligase ZNF451ZNF451Lum A&TP
Table 12. Analysis of the molecular functions in which BCP blood exosomal proteins are involved.
Table 12. Analysis of the molecular functions in which BCP blood exosomal proteins are involved.
Molecular Functionsp-ValueGene Name
Amidinotransferase activity0.0204GATM
ATPase activity0.5344KIF20B
Calcium ion binding0.3618KCNIP3
OCM
Catalytic activity0.0298CD81
EHHADH
ACADM
CKMT2
HIF1AN
HMOX1
MMAB
MTHFD1
Cell adhesion molecule activity0.9153MAEA
Chaperone activity0.5802TOR3A
Complement activity0.0156CLU
C3
Complement binding0.0204CFH
Cysteine-type peptidase activity0.2711CAPN2
Cytokine activity0.5213IL16
Cytoskeletal protein binding0.4409SPTBN2
ERC2
Defense/immunity protein activity0.0654AMBP
AHSG
DNA binding0.2907BANF1
ZNF638
FAM50A
ZNF585B
ZNF630
PRDM12
DNA-directed RNA polymerase activity0.1806POLR2D
Extracellular ligand-gated ion channel activity0.2456P2RX3
GTPase activator activity0.6293RABGAP1L
GTPase activity0.7842RAB24
Guanyl-nucleotide exchange factor activity0.5376VAV3
Heat shock protein activity0.0146CRYAA
CRYAB
Hydrolase activity0.7538HAGH
Intracellular ligand-gated ion channel activity0.1863GLRB
Ligase activity0.1786ACSM3
FARSB
Lipase activity0.1223PNLIP
Metallopeptidase activity0.1520ADAM10
BMP1
Motor activity0.1019KIF3B
KIFC3
Phospholipase activity0.2660PLB1
Protease inhibitor activity0.0040A2M
SERPI
NA1
ITIH4
SERPINB7
Protein binding0.1290FGG
FGB
FGA
Protein serine/threonine kinase activity0.6131ATR
MYO3B
Protein serine/threonine phosphatase activity0.2610PPM1A
Protein threonine/tyrosine kinase activity0.1750CLK3
Protein tyrosine kinase activity0.2298ZAP70
Receptor activity0.9182IGF2R
Receptor binding0.2208BMP7
TNFSF14
Receptor signaling complex scaffold activity0.3787SOCS3
SNTA1
FBF1
RNA binding0.9210SKIV2L
Structural constituent of cytoskeleton0.2410GSN
PCNT
Structural constituent of ribosome0.0864RPL28
MRPL52
UBA52
Structural molecule activity0.1132KRT1
KRT6A
KRT6B
PPHLN1
Transcription factor activity0.9810PIBF1
ZKSCAN8
Transcription regulator activity0.9281PHB2
ZNF451
ZNF622
Transporter activity0.0002ABCB7
ALB
APOA1
COG4
EXOC8
HBB
HP
HPX
ITPR2
RANBP3
TTR
TF
VPS13A
Voltage-gated ion channel activity0.2233CACNA1A
CACNG8
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Shefer, A.; Yanshole, L.; Proskura, K.; Tutanov, O.; Yunusova, N.; Grigor’eva, A.; Tsentalovich, Y.; Tamkovich, S. From Cell Lines to Patients: Dissecting the Proteomic Landscape of Exosomes in Breast Cancer. Diagnostics 2025, 15, 1028. https://doi.org/10.3390/diagnostics15081028

AMA Style

Shefer A, Yanshole L, Proskura K, Tutanov O, Yunusova N, Grigor’eva A, Tsentalovich Y, Tamkovich S. From Cell Lines to Patients: Dissecting the Proteomic Landscape of Exosomes in Breast Cancer. Diagnostics. 2025; 15(8):1028. https://doi.org/10.3390/diagnostics15081028

Chicago/Turabian Style

Shefer, Aleksei, Lyudmila Yanshole, Ksenia Proskura, Oleg Tutanov, Natalia Yunusova, Alina Grigor’eva, Yuri Tsentalovich, and Svetlana Tamkovich. 2025. "From Cell Lines to Patients: Dissecting the Proteomic Landscape of Exosomes in Breast Cancer" Diagnostics 15, no. 8: 1028. https://doi.org/10.3390/diagnostics15081028

APA Style

Shefer, A., Yanshole, L., Proskura, K., Tutanov, O., Yunusova, N., Grigor’eva, A., Tsentalovich, Y., & Tamkovich, S. (2025). From Cell Lines to Patients: Dissecting the Proteomic Landscape of Exosomes in Breast Cancer. Diagnostics, 15(8), 1028. https://doi.org/10.3390/diagnostics15081028

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