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 CaCl
2, 0.04 mM MgSO
4, 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 CO
2 incubator (5% CO
2) 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 K
3EDTA 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.
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 mm
2, 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 (r
2 = 0.92), followed by human bone marrow (r
2 = 0.65) and lymphatic fluid EVs (r
2 = 0.64); these proteins correlated least with human cell line- (r
2 = 0.15) and tissue explant-derived EVs (r
2 = 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.