Next Article in Journal
Phytoplasmas: Molecular Characterization and Host–Pathogen Interactions
Previous Article in Journal
Stomata Are Driving the Direction of CO2-Induced Water-Use Efficiency Gain in Selected Tropical Trees in Fiji
Previous Article in Special Issue
Persistence of Correlations in Neurotransmitter Transport through the Synaptic Cleft
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Low Magnetic Field Exposure Alters Prostate Cancer Cell Properties

by
Sigrun Lange
1,*,
Jameel M. Inal
2,3,
Igor Kraev
4,
Dafydd Alwyn Dart
5 and
Pinar Uysal-Onganer
6,*
1
Pathobiology and Extracellular Vesicles Research Group, School of Life Sciences, University of Westminster, London W1W 6UW, UK
2
Cell Communication in Disease Pathology, School of Human Sciences, London Metropolitan University, 166-220 Holloway Road, London N7 8DB, UK
3
Biosciences Research Group, School of Life and Medical Sciences, University of Hertfordshire, Hatfield AL10 9EU, UK
4
Electron Microscopy Suite, Faculty of Science, Technology, Engineering and Mathematics, Open University, Milton Keynes MK7 6AA, UK
5
UCL Cancer Institute, University College London, Paul O’Gorman Building, 72 Huntley Street, London WC1E 6DD, UK
6
Cancer Mechanisms and Biomarkers Research Group, School of Life Sciences, College of Liberal Arts and Sciences, University of Westminster, 115 New Cavendish Street, London W1W 6UW, UK
*
Authors to whom correspondence should be addressed.
Biology 2024, 13(9), 734; https://doi.org/10.3390/biology13090734
Submission received: 30 August 2024 / Revised: 11 September 2024 / Accepted: 17 September 2024 / Published: 19 September 2024
(This article belongs to the Special Issue The Rules of Life Rethought: Latest Progress in Quantum Biology)

Abstract

:

Simple Summary

The effects of magnetic fields on health and disease have been subject to considerable research interest for the past few decades but still remain relatively poorly understood. Therefore, the identification of molecular and cellular pathways affected is of considerable importance. This study investigated the effects of very low magnetic field (LMF) exposure in a cellular model of prostate cancer (PCa), the second most common cancer diagnosed in men. Extracellular vesicles (EVs) are lipid structures released from and taken up by cells and play crucial roles in cell communication and in processes such as cancer spread via their protein and nucleic acid cargoes. Short-term (4 h) LMF exposure significantly altered the release profiles and protein content of EVs from PCa cells to a more pro-cancerous profile. We then investigated changes in several key micro-RNAs, which are regulators of cancer behaviour and indicators of cancer aggressiveness and metastasis. LMF exposure caused significant upregulation of three key oncogenic miRNAs (miR-155, miR-21, and miR-210) and significant downregulation of two key tumour-suppressive miRNAs (miR-126 and miR-200c) in the PCa cells. These changes were also associated with a significant increase in the cancer cells’ invasion capability, which is a key indicator of cancer aggressiveness. We further verified the metastatic ability of the cancer cells caused by the LMF exposure by assessing two metastasis-related proteins, matrix metalloproteinases MMP2 and MMP9, which both were significantly increased. We compared these findings with normal prostate cells, which showed fewer changes in response to LMF exposure. Our findings suggest that LMF exposure may promote a more aggressive cancer phenotype by modulating key molecular and cellular pathways, highlighting the potential therapeutic implications of magnetic field modulation in cancer treatment.

Abstract

Prostate cancer is the second most common neoplasia and fifth-leading cause of cancer death in men worldwide. Electromagnetic and magnetic fields have been classified as possible human carcinogens, but current understanding of molecular and cellular pathways involved is very limited. Effects due to extremely low magnetic/hypomagnetic fields (LMF) are furthermore poorly understood. Extracellular vesicles (EVs) are crucial mediators of cellular communication with multifaceted roles in cancer progression, including via transport and uptake of various protein and microRNA (miRNA) EV-cargoes. miRNAs regulate gene expression and are implicated in cancer-related processes such as proliferation, metastasis, and chemoresistance. This study investigated the effects of LMF exposure (20 nT) by magnetic shielding on the prostate cancer cell line PC3 compared to the prostate epithelial cell line PNT2 under short-term (4 h) conditions. We examined EV profiles following a 4 h LMF exposure alongside associated functional enrichment KEGG and GO pathways for the EV proteomes. The 4 h LMF exposure significantly reduced cellular EV release and modified PC3 EV cargoes to a more inflammatory and metastatic profile, with 16 Disease Pathways and 95 Human Phenotypes associated specifically with the LMF-treated PC3 EV proteomes. These included cancerous, metabolic, blood, skin, cardiac and skeletal Disease Pathways, as well as pain and developmental disorders. In the normal PNT2 cells, less EV protein cargo was observed following LMF exposure compared with cells not exposed to LMF, and fewer associated functional enrichment pathways were identified. This pointed to some differences in various cellular functions, ageing, defence responses, oxidative stress, and disease phenotypes, including respiratory, digestive, immune, and developmental pathways. Furthermore, we analysed alterations in matrix metalloproteinases (MMPs) and miRNAs linked to metastasis, as this is crucial in cancer aggressiveness. The 4 h LMF exposure caused a significant increase in MMP2 and MMP9, as well as in onco-miRs miR-155, miR-210, miR-21, but a significant reduction in tumour-suppressor miRs (miR-200c and miR-126) in the metastatic PC3 cells, compared with normal PNT2 cells. In addition, 4 h LMF exposure significantly induced cellular invasion of PC3 cells. Overall, our findings suggest that changes in magnetic field exposures modulate EV-mediated and miR-regulatory processes in PCa metastasis, providing a basis for exploring novel therapeutic strategies.

1. Introduction

Prostate cancer (PCa) is the second most prevalent cancer globally, contributing significantly to cancer-related mortality rates [1,2]. Various risk factors, including age, dietary habits, obesity, tobacco and alcohol use, disruptions in circadian rhythms, racial background, and sexual behaviour have been linked to an increased risk of developing PCa. Additionally, environmental and occupational exposures have been suggested to explain the differing epidemiological impacts of the disease across populations [3,4,5].
Previously, a potential association between childhood leukaemia and electromagnetic fields (EMFs) emitted by power lines has been highlighted, leading to their classification as possible human carcinogens [6]. Although the associated risk is considered low, this link was recognised several years ago and is now regarded as a preventable risk factor. However, establishing a definitive causal relationship and elucidating the underlying biological mechanisms has proven challenging, primarily due to limited support from animal and laboratory studies regarding the carcinogenic effects of magnetic, including low (LMF) magnetic fields. Consensus remains elusive on the mechanisms by which magnetic fields interact with biological systems and biomolecules beyond thermal interactions. Moreover, magnetic fields can potentially influence quantum systems within biological molecules, affecting the spin states of electrons and interactions with other molecules. These effects may have implications for various biological processes and enzymatic reactions; however, precise mechanisms by which LMFs interact with cellular processes remain unclear. The emerging field of quantum biology, though in its early stages, is rapidly expanding and gaining recognition. Quantum phenomena have been implicated in a variety of biological processes, including photosynthesis, navigation, enzyme catalysis, olfaction, and DNA mutation [7].
microRNAs (miRs/miRNAs) are single-stranded RNA molecules that can moderate gene expression at the post-transcriptional level and are estimated to control more than 60% of protein-encoding genes [8,9,10]. Furthermore, it has been proposed that ineffective cancer therapy is frequently due to a lack of current understanding of accurate molecular mechanisms that are involved in tumorigenesis. Recently, miRNA profiling and disease-specific miRNA signatures have been widely used for the detection of various cancers, such as pancreatic, prostate, and breast cancers, as they are secreted into body fluids, primarily packaged into EVs [11,12,13,14]. Work in our lab has highlighted the fact that several miRNAs are involved in biological processes such as proliferation, invasion, migration, metastasis, angiogenesis, and chemoresistance [14,15,16,17,18].
Extracellular vesicles (EVs) are 30–1000 nm lipid bilayer-enclosed structures released from cells and taken up by neighbouring cells. EVs play important roles in cellular communication via the transport of EV cargoes, including proteins, genetic material, and non-coding RNAs, some of which are microRNAs. EVs are key mediators in intra- and inter-tumour communication, can influence the tumour microenvironment, participate in the preparation of the pre-metastatic niche, and contribute to cancer aggressiveness [19,20]. The release profiles of EVs from cancer cells are, therefore, of considerable importance, both with respect to EV numbers released as well as EV signatures relating to changes in EV sub-populations and EV cargoes, including proteins and miRs [16,21,22]. Importantly, previous research has shown that exposure to extremely low-frequency strength magnetic fields modulates the numbers of EVs released from various cancer cells in vitro, also sensitising some cancer cells to chemotherapeutic treatment [23,24].
The current scientific literature lacks a full understanding of mechanisms explaining the interactions between electromagnetic and/or magnetic fields and biological material. Magnetic fields may influence the behaviour of biological molecules at the quantum level via their electron spin states with significant implications for processes where electron transfer is vital, such as mitochondrial ATP production or in enzymatic processes where transient electron spin states are generated. Additionally, magnetic fields may interact with other biomolecules, including DNA and RNA, although the specific physicochemical mechanisms remain unclear [25]. Further research into cellular and molecular mechanisms is, therefore, essential.
Matrix metalloproteinases (MMPs) comprise a group of twenty-four zinc-dependent extracellular endopeptidases, extensively expressed across various tissues and engaged in numerous biological functions. Their primary function is the degradation of all components of the extracellular matrix. Additionally, MMPs play crucial roles in inflammatory processes by modulating the synthesis and release of cytokines and chemokines. Furthermore, MMPs are associated with cellular growth, proliferation, and tissue remodelling [26]. Among these, MMP2 and MMP9 are the most significant cancer-associated zinc-dependent endopeptidases involved in the invasion and metastasis of various carcinomas and elevated expression levels of activated MMP2 or MMP9 have been correlated with metastasis in patients with PCa [27,28].
In this study, we used a known cancer model to examine the impact of LMF/hypomagnetic exposure (20 nT) by magnetic shielding on cell–cell communication and molecular mechanisms involved, focussing on EVs, miRs, and MMPs. We compared the PC3 prostate cancer cell line with the normal prostate epithelial cell line PNT2. We assessed the effects of 4 h magnetic shielding on EV signatures, identifying changes in EV release profiles, total EV protein cargoes, and associated KEGG and GO pathways. Based on those findings, we further assessed changes in key MMPs and selected miRs associated with metastasis.

2. Materials and Methods

2.1. Cell Culture

The prostate epithelial cell line PNT2 and the PCa cell line PC3 (ATCC, Manassas, VA, USA) were maintained in Roswell Park Memorial Institute (RPMI) 1640 (Gibco-Life Technologies, Carlsbad, CA, USA) with 10% (v/v) heat-inactivated foetal bovine serum (FBS; Pan Biotech, Aiedenbach, Germany) and penicillin–streptomycin (10,000 units penicillin/mL and 10 mg streptomycin/mL) (Pan Biotech, Germany) at 37 °C in a humidified 5% CO2 incubator (Heracell 150i, Thermo Fisher Scientific, Hemel Hempstead, UK).

2.2. Magnetic Shielding—Extremely Low Magnetic/Hypomagnetic Treatment

To investigate the effects of shielding biological samples from Earth’s magnetic field, modelling low magnetic field (LMF) exposure, we utilised an instrument from Magnetic Shields Ltd. (MSL, Staplehurst, Kent, UK), crafted from a metallic alloy known as mu-metal. This material attracts and deflects the geomagnetic field, creating an internal environment of an extremely low magnetic/hypomagnetic (20 nT) field. Cells were plated onto appropriate cell culture plates or flasks for subsequent assays and allowed to adhere overnight. After adherence, the cells were divided into two groups. The experimental LMF group was exposed to the magnetic shield by placing the cells inside the mu-metal instrument at room temperature for 4 h. The control group was kept on the bench outside the incubator at room temperature for the same duration without exposure to the magnetic shield. Following the 4 h exposure period, the cells were processed according to the specific requirements of the assays described below.

2.3. RNA Extraction and qRT-PCR

RNA was extracted from cells using Trizol (Sigma, Haverhill, UK), and RNA concentration and purity were measured using the NanoDrop spectrophotometer (Thermo Fisher Scientific, Hemel Hempstead, UK) at 260 nm and 280 nm absorbance. Reverse transcription of RNA to cDNA was carried out using a miRCURY LNA RT Kit (Qiagen, Manchester, UK) according to the manufacturer’s instructions. The miRCURY LNA miRNA SYBR Green (Qiagen, Manchester, UK) was used in conjunction with MystiCq microRNA qPCR primers for miRs 21 (hsa-miR-21-5p MIRAP00047), 155 (hsa-miR-155-5p MIRAP00202), 210 (hsa-miR-210 MIRAP00262), 126 (hsa-miR-126-5p MIRAP00142), and 200c (hsa-miR-200c-5p MIRAP00252), all from Sigma, Haverhill, UK. The resulting cDNA was used to assess the expression of miR-21, miR-155, miR-210, miR126, and miR-200c while RNU6 (F 5′-GCTTCGGCAGCACATATACTAAAAT-3; R 5′-CGCTTCACGAATTTGCGTGTCAT-3′) was used as a reference RNA for normalisation of miRNA expression levels, as described before [11,12]. Each experiment was repeated three times, and the relative expression levels of listed miRNAs were normalised with RNU6 expressions using the comparative cycle threshold method [29]. cDNAs for the analysis of MMP2 (F 5′-GAGAAGACATTCCTCAGAGACG-3′; R 5′-TGGGGAGGTTTACCCTATATGG-3′) and MMP9 (F 5′-GGACCCGAAGCGGACATTG-3′; R 5′-CGTCGTCGAAATGGGCATCT-3′) primers were used as described before and expressions were generated using qScript cDNA Supermix (Quantabio, London, UK) with incubations at 42 °C for 30 min and 85 °C for 5 min [30]. The gene expressions were analysed by using PrecisionPlus qPCR Master Mix (Primer Design, Eastleigh, UK) for RT-qPCR synthesis with the following thermocycling conditions for 40 cycles: 95 °C for 2 min, 95 °C for 10 s, and 60 °C for 60 s. Relative gene expression levels of MMPs were calculated with RNA polymerase II (RPII) (F 5′-GCACCACGTCCAATGACAT-3′ R; 5′-GTGCGGCTGCTTCCATAA-3′) as described before [11,18].

2.4. EV Isolation and Characterisation

EV isolation was carried out according to established and previously published protocols [21,22], also adhering to the recommendations of the International Society of Extracellular Vesicle Research (ISEV; MISEV2023) [31]. Cells were cultured to a 70% confluence in T25 flasks, and the adherent cells were washed with sterile-filtered EV-free Dulbecco’s Phosphate-Buffered Saline (DPBS) before applying 5 mL of fresh cell culture medium per flask, omitting foetal bovine serum (FBS) for the duration of the 4 h experiment, to avoid contamination with FBS derived EVs. Negligible detection of EVs in the cell-free medium was confirmed by NTA. Following the 4 h LMF incubation (the control treatment was handled the same way but kept outside the LMF chamber), EVs were isolated from the cell culture supernatants (from the 3 flasks per experiment, containing 5 mL medium each) from each T25 flask as follows: First the supernatants were centrifuged at 4000× g for 30 min at 4 °C to remove cell debris, whereafter the supernatant was carefully collected by pipetting and centrifuged for 1 h at 100,000× g at 4 °C for the enrichment of total EVs, generating an EV pellet. The supernatants were carefully aspirated and discarded, and the isolated EV pellets were thereafter resuspended and washed in ice-cold sterile-filtered EV-free DPBS and centrifuged again at 100,000× g for 1 h at 4 °C. The final EV-enriched pellets were then resuspended in 100 μL sterile-filtered EV-free DPBS for further analysis. nNanoparticle tracking analysis (NTA) was carried out using the NS300 Nanosight (Malvern Panalytical Ltd., Malvern, UK), equipped with a sCMOS camera and a 405 nm diode laser, to enumerate the EVs and assess EV size profiles. Samples were diluted 1:100 in sterile-filtered EV-free DPBS, and the number of particles in the field of view was maintained in the range of 30–50 with a minimum concentration of samples at 5 × 107 particles/mL. The camera settings for recording were set at 13 and for post-processing of videos at setting 5, according to the manufacturer’s instructions (Malvern Panalytical Ltd.). Five 60 s videos were recorded per sample, and the replicate histograms were averaged using the NTA 3.4 software. Each experiment was repeated in three biological replicates. EVs were further characterised by the two surface markers CD63 (ab216130, Abcam, Cambridge, UK) and Flotillin-1 (ab41927, Abcam) using western blotting and imaged by transmission electron microscopy (TEM), according to previously published protocols [21,22]. Briefly, for TEM, EVs pellets were resuspended in 0.1 M sodium cacodylate buffer (pH 7.4), and a 3–5 µL drop of EVs suspension was applied onto a glow-discharged carbon film-supported TEM grid. After allowing the suspension to air dry for approximately 10 min, the grid was placed sample-side down onto a drop of 2.5% glutaraldehyde fixative solution (Agar Scientific Ltd., Stansted, UK) in 0.1 M sodium cacodylate buffer (pH 7.4) for 1 min at room temperature. The grid was then washed by placing it onto three separate drops of distilled water, removing excess water between each step using filter paper. Next, the grid was placed onto a drop of 2% aqueous uranyl acetate (Agar Scientific Ltd., Stansted, UK) for 1 min for staining. Excess stain was removed using filter paper, and the grid was air dried. Imaging of the EVs was performed using a JEOL JEM 1400 microscope (JEOL, Tokyo, Japan) operated at 80 kV, with magnifications ranging from 10,000× to 30,000×, with digital images captured using a 16-megapixel GATAN RIO 16 camera (AMETEK (GB) Limited, Leicester, UK).

2.5. Liquid Chromatography with Tandem Mass Spectrometry (LC-MS/MS) and STRING Protein–Protein Interaction Network Analysis

EV protein cargoes from isolated EV preparations of LMF exposed and control/untreated PC3 and PNT2 cells, respectively, were analysed for protein hits by LC-MS/MS. EVs were isolated from 3 × 5 mL culture medium as described above, from three T25 flasks per experimental group. Proteins were verified by SDS-PAGE and silver staining before running the EV protein isolates 0.5 cm into a 12% TGX gel and cutting each sample out as one band. The gel bands were then subjected to in-gel digestion followed by LC-MS/MS by Cambridge Proteomics (Cambridge, UK). In brief, automated LC-MS/MS analysis was carried out using a Dionex Ultimate 3000 RSLC nanoUPLC (Thermo Fisher Scientific Inc., Waltham, MA, USA) system in conjunction with a QExactive Orbitrap mass spectrometer (Thermo Fisher Scientific Inc., Waltham, MA, USA). Peptide separation was carried out using reverse-phase chromatography and a Thermo Scientific reverse-phase nano Easy-spray column (Thermo Fisher Scientific Inc). The LC eluent was sprayed into the mass spectrometer using an Easy-Spray source (Thermo Fisher Scientific Inc.). The m/z values of all eluting ions were measured in an Orbitrap mass analyser; data-dependent scans (selecting top 20) were employed for automatic isolation and generation of fragment ions using the HCD collision cell, measured using the Orbitrap analyser. Both singly charged ions as well as ions with unassigned charge states were excluded from selection for MS/MS. A dynamic exclusion window of 20 sec was also applied. Data were processed post-run using Proteome Discoverer (version 2.1., Thermo Scientific), converted to mgf files, and submitted to Mascot (Mascot search algorithm; Matrix Science, London, UK). Search for hits was carried out against the UniProt Homo_sapiens_20221011 database (226,953 sequences; 74,609,178 residues) with peptide and fragment mass tolerances respectively set at 20 ppm and 0.1 Da. The threshold value for significance was set at p < 0.05, and the peptide cut-off score was set at 35. To generate protein–protein interaction networks and associated functional enrichment pathway analysis, protein hits were fed into the STRING database (https://string-db.org/; accessed 19 April 2024) and analysed based on the Homo sapiens database. Settings were at medium confidence. Protein–protein interaction networks were generated in STRING for each experimental group and compared between the control and LMF-treated EV proteomes. STRING functional enrichment pathway analysis was used to identify shared and distinct Gene ontology (GO), Reactome, and STRING cluster pathways, as well as Disease–gene associations and Human Phenotype. Functional enrichment tables were downloaded from STRING as Excel files and the protein–protein interaction network images were downloaded as PNG files.

2.6. Assays for Cellular Invasion and Proliferation

Cell invasion assay was performed as follows: 5 × 105 cells were plated on Matrigel-coated transwell filters (Corning™ BioCoat™ Matrigel™ Invasion Chamber with Corning™ Matrigel Matrix; BD Biosciences, Wokingham, Berkshire, UK) in a chemotactic gradient of 1:10% FBS. After 4 h incubation either inside of the magnetic shield instrument (LMF) or outside as the control, the total number of invaded cells was determined by MTT assay (Abcam, Cambridge, UK) and further confirmed by crystal violet assay (Abcam, UK). In parallel, the same number of cells was plated and incubated for 4 h to determine the effect of LMF exposure on cell proliferation by MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide) assay. Absorbance was measured using CLARIOstar plate reader (BMG Labtech, Aylesbury, UK) at 540–590 nm and normalised according to the control (n = 3).

2.7. Data Analysis

All data were checked for normal distribution and analysed as means ± standard deviation (SD). Statistical significance was determined using a Student’s t-test or ANOVA with a Newman–Keuls post hoc analysis, as appropriate. Results were considered significant for p < 0.05. One-way ANOVA Bonferroni’s multiple comparisons test was performed using GraphPad Prism version 7.00 for Windows (GraphPad Software, La Jolla, CA, USA).

3. Results

In summary, this study determined the effects of 4 h magnetic shield (low magnetic/hypomagnetic) exposure on the prostate cancer PC3 cell line, compared to the normal (immortalised and nontumourigenic) prostate epithelial cell line PNT2. EV profiling was carried out for changes in EV numbers released and on EV protein cargoes. Based on these outcomes, further assessments were carried out for metastasis-associated MMPs and miRNAs, as well as changes in PC3 cell invasion and proliferation capacities.

3.1. EV Profiles from PC3 and PNT2 Cells Were Modified in Response to 4 h LMF Exposure

EVs obtained from PC3 and PNT2 cells were isolated by differential centrifugation and characterised using nanoparticle tracking analysis (NTA, representative figures are shown in Figure 1A–D), two EV-specific surface markers (CD63 and flotillin-1) by western blotting (Figure 1E) and transmission electron microscopy (TEM) (Figure 1F).
EVs were enumerated by NTA, assessing total EV numbers released from PC3 and PNT2 cells under control and LMF conditions (Figure 2A). Differences in EV subpopulations released were measured, considering small EVs ≤ 100 nm, medium EVs 101–200 nm, and large EVs > 200 nm (Figure 2B,C). Also, the mean size (Figure 2D) and modal size (Figure 2E) of EVs were assessed. Following 4 h LMF exposure, both PC3 and PNT2 cells showed a significant reduction in total EV numbers released (Figure 2A), and all EV sub-populations were significantly reduced following LMF exposure in PC3 cells (Figure 2B). In PNT2 cells, the numbers of medium-sized EVs (101–200 nm) and large EVs (>200 nm) were significantly reduced following LMF exposure (Figure 2C). EV mean size was reduced in PNT2 but not changed in PC3 cells following 4 h LMF exposure (Figure 2D). EV modal size showed a trend in increase (but not significantly) in PC3 LMF-treated cells, while EV modal size was reduced in PNT2 cells following LMF exposure (Figure 2D).

3.2. Proteomic EV Cargoes Showed a Shift to More Pro-Cancerous Signature in PC3 Cells Following 4 h LMF Exposure

EVs were isolated from the PC3 and PNT2 cell cultures according to methods described in Section 2.4. The protein content of EVs was assessed by SDS-PAGE with silver-staining for PC3-derived EVs from the control and LMF-treated cells (Figure 3A) and from PNT2-derived EVs from the control and LMF-treated cells, respectively (Figure 3B). Proteomic content was then analysed by LC-MS/MS, and numbers of shared and unique protein hits per experimental group are presented in the Venn diagrams for PC3 EVs (Figure 3C) and PNT2 EVs (Figure 3D), respectively (for full information on LC-MS/MS analysis see Supplementary Tables S1–S4).
The protein hits identified for EV cargoes from PC3 and PNT2 cells, comparing cell-derived EVs from control untreated to LMF exposed cells, respectively, are listed and summarised in Table 1. Proteins that were identified in the EV proteomes of the PC3 LMF treated cells only included Keratin II, Keratin 6A, Actin cytoplasmic 2, Immunoglobulin heavy chain variable region, Annexin 1, Haemoglobin subunit beta, Phosphopyruvate hydratase, Villin 2/Ezrin, Histone H2A.Z, Histone H2B, Serine protease 1, Fructose-bisphosphate aldolase, Interferon-induced transmembrane protein, Dermcidin, CD44 antigen, Triosephosphate isomerase, HSP90AA1 protein, Ventricular zone-expressed PH domain-containing protein, and Tyrosine 3-monooxygenase/tryptophan 5-monooxygenase activation protein eta.

3.3. Protein–Protein Interaction Network Analysis for EV Protein Cargoes, Comparing LMF Treated to Control Untreated PC3 Cells

Protein–protein interaction networks for EV protein cargoes were created in STRING (https://string-db.org/; accessed 19 April 2024) for the EVs isolated from PC3 cells, the control, and 4 h LMF treatment, respectively. A considerable change was observed in the protein interaction networks and associated functional enrichment pathway analysis for the PC3-derived EVs following 4 h LMF exposure (Figure 4A). Furthermore, Gene ontology (GO) analysis showed increased pathways associated with EV proteomes of LMF-treated cells (Figure 4B), including 31 Biological GO, 4 Molecular GO, and 14 Cellular GO pathways only identified following LMF treatment. In addition, an increase in disease-associated pathways was observed, with 16 DISEASE and 95 Human Phenotype (Monarch) pathways identified for protein cargoes of the EVs from LMF-treated PC3 cells. Details on the protein network annotations for the PC3-derived EVs are listed in Table 2.

3.4. Protein–Protein Interaction Network Analysis for EV Protein Cargoes, Comparing LMF Treated to Control Untreated PNT2 Cells

Protein–protein interaction networks for EV protein cargoes were created in STRING for the EVs isolated from PNT2 cells, from control and 4 h LMF treatment, respectively. Considerably fewer proteins were identified in the EV proteome of the LMF-treated PNT2 cells, as reflected in the differences in the protein-interaction networks (Figure 5A). Furthermore, functional enrichment analysis pathways were accordingly associated with the control EV proteome, compared to the EV proteome of the LMF-treated cells. This included 37 biological and 34 Cellular GO pathways, as well as 18 DISEASE and 55 Human Phenotype pathways for the control EV proteome. For the EV proteome of LMF-treated PNT2 cells, 30 Human Phenotypes were specific. Furthermore, several pathways were identified in both groups, as summarised in the Venn diagram in Figure 5B. For details on pathways, see Table 3.
A summary of the PC3 and PNT2 EV proteome-associated protein-interaction networks and functional enrichment analysis are presented in Figure 6. This shows Biological GO, Molecular Function GO, Cellular Component GO, KEGG and Reactome pathways, Disease–gene associations, Subcellular localisations, STRING and Human Phenotype pathways for PC3-EV-associated proteins (Figure 6A) and PNT2-EV-associated proteins (Figure 6B), respectively, indicating LMF-treated groups in red.

3.5. Expression Levels of Oncogenic and Tumour Suppressor miRNAs Were Differently Modulated in Response to LMF Exposure, Only in PC3 Cells

Following 4 h LMF exposure, effects on key oncomiRs and tumour suppressor miRs were investigated. Based on our previous and other studies, miR-21, miR-210, miR-155, miR-200c, and miR-126 are closely associated with the development of PCa at different stages [10,12,32,33,34,35]. When assessing oncomiRs expression levels (miR-155, miR-210, miR-21) and tumour-suppressor miRs (miR-200c and miR-126) comparing control with LMF-exposed cells, significant expression changes were observed in response to LMF exposure only in the metastatic PC3 cell line but not in the non-tumorigenic control PNT2 cell line (Figure 7A,B). Following 4 h LMF exposure, miR-155 showed a 57-fold increase (p < 0.0001) in PC3 cells, compared to only a 2-fold (ns) increase in PNT2 cells; miR-21 was 57-fold increased (p < 0.0001) in PC3 cells, while no significant change was observed in PNT2 cells, and miR-210 showed a 17-fold increase in PC3 cells (p < 0.0001), while no significant change was observed in PNT2 cells (Figure 7A). The 4 h LMF exposure resulted in a 1.25-fold decrease in tumour-suppressor miR-200c (p < 0.05) and a 2.5-fold decrease in miR-126 (p < 0.01) in PC3 cells, but no significant changes were observed for the PNT2 cells (Figure 7B).

3.6. LMF Exposure Enhanced the Expression Levels of Matrix Metalloproteinases (MMP2 and MMP9) in PC3 Cells

Following the EV proteome data analysis, which indicated an increase in metastasis-related proteins, we analysed the expression levels of MMP2 and MMP9 after exposing both cell lines to 4 h LMF treatment. We found that LMF exposure induced MMP2 and MMP9 mRNA levels significantly (p < 0.0001) in the PC3 cells by 25-fold and 20-fold, respectively; however, no significant changes were detected in the PNT2 cells (Figure 8A,B).
To confirm the effects of LMF exposure on PCa metastasis, a Matrigel invasion assay was carried out. The assay was conducted during a 4 h LMF exposure time window inside the magnetic field shield instrument at room temperature and compared to a control assay, without LMF exposure, also carried out at room temperature. Despite using a shorter incubation period of 4 h rather than a typical overnight incubation, the PC3 cells demonstrated significantly increased invasion (34% increase, p < 0.0001) when kept in the LMF chamber for 4 h, compared to cells kept outside the effects of LMF (Figure 9A). In parallel, a proliferation assay was performed, with no changes detected in cellular proliferation (Figure 9B).

4. Discussion

PCa remains the second leading malignancy and the fifth cancer-related cause of death among men worldwide, with approximately 1.4 million new cases and 400,000 deaths [36]. Given its high incidence and mortality, this malignant disease represents an important public health problem [37]. Magnetic field effects and the influences of exposure to altered magnetic conditions are gaining increased attention in biological research [38], including in cancer [39]. To date, while a range of studies has been carried out, understanding of mechanisms is still limited, including exact roles in the regulation of cellular processes. Furthermore, short-term and long-term effects on mechanisms involved in cancer are relatively poorly understood. Therefore, studies identifying molecular and cellular pathways influenced by changes in magnetic field exposure are of great importance enhancing the current understanding of their contribution to disease processes and identifying possible therapeutic benefits.
The current study focussed on assessing the effects of a short (4 h) low magnetic/hypomagnetic field exposure (20 nT, using a magnetic shield instrument) on prostate cancer cell properties in vitro. Effects of such low magnetic field exposure (LMF) were assessed for changes in extracellular vesicle (EV) signatures, focussing on proteomic cargoes, with results indicating pro-metastatic changes. Therefore, further investigations focussed on cancer-associated MMPs as well as key oncogenic and tumour-suppressor miRNAs. Matrigel cell invasion assay confirmed that PC3 cell invasion increased in response to the 4 h LMF exposure. PC3 cell proliferation capacity was also assessed in response to the 4 h LMF treatment.
EVs were significantly reduced following a 4 h LMF exposure, and this was observed for small (<100 nm), medium (101–200 nm), and large (>200 nm) EVs in the PC3 cells, indicating that all EV subpopulations were influenced by LMF exposure. This may have considerable effects on cellular communication in prostate cancer, as generally increased EV release is associated with cancers, and EVs contribute significantly to metastatic processes [40,41]. Interestingly, a reduction in EV numbers was also observed for the PNT2 cell line, with a significant reduction in medium and large EVs but not small EVs. The different EV subpopulations have been subject to a wide range of studies, with a focus on small EVs and medium/large EVs in various cancer models in response to different treatments, while studies on the effects of changes in magnetic fields are very limited to date [23,24]. Therefore, the current findings are of considerable importance to gain an understanding of the roles of EV modulation in cellular communication in response to LMF exposure. Interestingly, previous research showed the enhanced release of calcium-stimulated EV release (medium/large EVs) from human monocytic leukaemia cells following 30 min pulsed LMF treatment at 0.3 μT [23]. This indicates possible differences between EV release profiles in response to LMF treatment between cancer cell lines, but also that LMF levels and time windows of LMF exposure may cause different effects of EV release. Importantly, changes in EV cargoes must also be considered, as EVs carry a range of protein, non-coding RNA, genetic, and other cargo. The focus of the current study was, therefore, also on EV protein cargoes, and interestingly, a considerable change in protein hits and increased protein hits were identified in PC3-derived EVs following the 4 h LMF exposure. While 13 protein hits were common between control and LMF-treated EV cargoes, 15 were unique to the controls, but there were 31 hits unique to EVs of the LMF-treated PC3 cells. Proteins that were only identified in the EV proteomes of the PC3 LMF-treated cells included Actin cytoplasmic 2, Cytoskeletal Keratins (Type I 16, Type II 5, Type II 6A and B), Immunoglobulin heavy chain variable region, Annexin 1, Haemoglobin subunit beta, Phosphopyruvate hydratase, Villin 2/Ezrin, Histone H2A.Z, Histone H2B, Serine protease 1, Fructose-bisphosphate aldolase, Interferon-induced transmembrane protein, Dermcidin, CD44 antigen, Triosephosphate isomerase, HSP90AA1 protein, Ventricular zone-expressed PH domain-containing protein, and Tyrosine 3-monooxygenase/tryptophan 5-monooxygenase activation protein eta. Several of these have been associated with cancer pathology, including metastasis, aggressiveness, and chemoresistance, also in PCa, and are briefly discussed below. Actins play multifaceted roles in podosomes and invadopodia formation [42]. Various keratins, including KRT5 and KRT6A, have been associated with PCa assessment and prognosis [43,44]. Ezrin has been identified as an indicator of metastasis via EV export [45] and as a circulating biomarker for PCa metastasis [46]. Histone H2A.Z is linked to the regulation of tumorigenesis, metastasis, and response to chemotherapy [47,48], while post-translational modifications of H2B are implicated in cancer initiation and progression [49]. Annexin 1 is a reported proteomic marker of PCa metastasis [50]. Fructose-bisphosphate aldolase has been identified in EV proteomes linked to PCa chemoresistance [51]. Interferon-induced transmembrane protein 1 belongs to a group of IFITM proteins that have been implicated in cancer aggressiveness and chemoresistance [52]. Dermcidin is associated with cancer survival, including in PCa [53]. CD44 is a cancer stem cell marker and indicative of PCa tumour initiation, drug resistance, metastasis, and recurrence [54]. HSP90AA1 is involved in PC3 necroptosis via mitochondrial fission pathways and ROS generation [55]. Tyrosine 3-monooxygenase/tryptophan 5-monooxygenase activation protein eta (YWHAH) is linked to EV-mediated activation of cancer-associated fibroblasts [56].
In relation to the EV proteomes, there was a marked increase in the numbers of associated functional protein pathways, based on STRING analysis. This included 5 STRING pathways and 1 KEGG pathway, and an increase in GO pathways with 31 Biological, 4 Molecular, and 14 Cellular GO pathways unique to the PC3 LMF-treated EVs. Interestingly, 16 Disease Pathways and furthermore 95 Human Phenotypes were unique to the PC3 LMF-treated EVs. These were linked to stress, cytoskeletal function, nitric oxide, antimicrobial activity, immune function (including the complement system), hypoxia responses, cancer, histone acetylation, and metabolism. Some of these will be discussed in relation to the published literature below.
Previous studies using magnetic shielding have, for example, studied the roles of hypomagnetic conditions in stimulating the proliferation of neural progenitor and stem cells associated with observed central nervous system dysfunction and developmental abnormalities in animals [57]. This correlates to some of the functional enrichment pathways identified in our current study. For example, the link to cardiac conditions identified here correlates to cardiovascular studies under hypomagnetic conditions in humans [58], changes in erythrocytes in a rat model [59], increase in human blood hemolysis [60], and increased embryo mortality and modified cardiac function, also associated to the circadian rhythm, in zebrafish [61]. Changes in immune response pathways relate to some published studies, including increased blood granulocytes [62] and increased neutrophil respiratory burst [63]. Effects on cytoskeleton organisation have been reported in cellular cancer models, relating to pathways identified here in relation to LMF exposure in PC3 cells [62]. Effects of hypomagnetic fields on DNA methylation have been reported in embryonic stem cells [64], and this pathway was identified here as associated with the control PC3 EV proteome but not following LMF exposure. Various defence pathways and bacterial pathways were identified in the EV proteomes. Some were shared between control and LMF-treated conditions. Effects of hypomagnetic field exposure have been reported, for example, on antibiotic resistance [65,66].
Interestingly, in the PNT2-derived EVs, considerably fewer protein hits were identified following LMF exposure compared to controls. This indicates a shift in protein export via EVs in normal cells in response to LMF effects and may be of considerable relevance for understanding influences on normal cellular processes. Notably, there was also a loss of many functional pathways associated with the changes in the EV proteomes of PNT2 cells following LMF exposure. For example, 14 STRING and 1 KEGG pathways were associated with the control cell EVs, but these were not present in the LMF-treated ones, while 6 and 2 other pathways were shared between both groups. A similar pattern was observed for GO pathways, with 37 Biological, 3 Molecular and 34 Cellular GO pathways associated with the controls, and further 13,3 and 13 shared, respectively. In the LMF-exposed PNT2 cells, there were only two unique Biological GO pathways. Furthermore, Reactome pathways showed 20 unique for the control, two shared, and three unique for the LMF-treated group. A considerable difference was also seen for Disease pathways, with 23 shared pathways but no unique ones for the LMF-treated group, while 18 unique ones were associated with the control EV PNT2 proteome. Human Phenotypes similarly indicated 81 shared pathways, 55 unique for the control PNT2 EV proteomes, but fewer, 20, for the LMF PNT2 EV proteomes. Pathways unique to the PNT2 LMF-treated group were related to corticotropin-releasing hormone signalling pathway, hematopoietic system disease, hemidesmosome assembly, cell junction organisation, and the uptake of dietary cobalamin into enterocytes. Human Phenotypes identified for the LMF group related to digestive, skeletal, musculoskeletal, conjunctiva, mucosal abnormalities, squamous cell carcinoma, various skin disorders, and furthermore, respiratory stress, and abnormal temperature regulation. This indicates that LMF exposure does add to some cellular stress responses. The loss of so many functional enrichment pathways may also indicate that many critical pathways may be negatively affected by LMF treatment in normal cells. It may also be postulated that the absence of various biological process pathway associations, including ageing, response to oxidative stress, cell differentiation, and immune response pathways, may have some positive effects on normal cells. Such speculations will require further investigations as effects of changes in magnetic fields will have multifaceted effects, and further time windows and ranges of fields will need to be explored, both on multiple cell types in vitro, as well as using in vivo models.
Interestingly, changes to oxidative stress regulation by reduced ROS were previously reported in response to hypomagnetic field exposure in rat hippocampal neurogenesis [67]. In some cancer cells, oxidative stress has been reported to be reduced in hypomagnetic conditions [68]. This may contribute to carcinogenic effects, which are reported in response to low magnetic fields [69]. In this context, different hypomagnetic conditions and time windows must also be considered. Our findings on identified functional networks also relate to studies reporting changes in skeletal muscle functioning, muscle metabolism regarding glucose and glycogen [57,70], and bone functioning/fragility [71]. Various developmental pathways were identified here as modified in EV cargoes. Previous studies in various models have shown links to hypomagnetic effects on developmental processes, including gamete quality, embryogenesis, teratogenic effects, and malformations [72,73,74,75]. Pain was one of the associated pathways identified for the EV proteome, and changes in pain sensitivity have been reported in mollusc models in response to electromagnetic fields [76]. While digestive-associated functional networks were identified in EV proteomes in this study, previous studies have reported no hypomagnetic effects on water and food intake in mice [62], but further studies will most likely be needed, both comparing different models as well as different LMF exposures.
Exposure of PC3 cells to LMF led to upregulation in miR-155, miR-21, and miR-210 in the current study. miR-155 is associated with inflammatory responses and has been linked to enhanced tumour growth and metastasis [77]. miR-21 promotes oncogenesis by targeting tumour-suppressor genes, thereby facilitating cell survival and proliferation [11,17]. miR-210, often upregulated under hypoxic conditions, aids cancer cells in adapting to low-oxygen environments and is correlated with increased tumour aggressiveness [78]. Conversely, LMF exposure of PC3 cells resulted in the current study in the downregulation in miR-126 and miR-200c. miR-126 inhibits angiogenesis by targeting VEGF signalling pathways, thereby suppressing tumour growth and metastasis [79]. Similarly, miR-200c plays a role in regulating EMT by targeting the transcription factors ZEB1 and ZEB2 [13,80]. Interestingly, LMF exposure did not result in significant changes in the expression of these miRNAs in PNT2 cells. The LMF-induced modulation of both oncogenic and tumour-suppressive microRNAs observed here in PC3 cells suggests that LMF exposure may contribute to a more aggressive cancer phenotype by influencing gene expression pathways associated with tumour progression. Further studies will be required to elucidate the underlying mechanisms and explore the potential of these microRNAs as therapeutic targets in PCa.
As some of the PC3 EV cargo protein hits in the LMF group indicated differences in metastatic associated pathways, further assessment was also carried out for selected key MMPs. MMP2 and MMP9 were both confirmed to be upregulated in PC3 cells following 4 h LMF exposure, while no significant changes were seen in the PNT2 cells. High expression levels of activated MMP2 or MMP9 have been associated with metastasis in patients with PCa [27,28,81,82]. Studies have indicated that serum levels of matrix MMP2 are correlated with the grading and malignancy of PCa [27,83]. It was suggested that MMP2 could be used as a molecular marker for PCa and may serve as a predictive indicator for the disease [84]. Additionally, the expression levels of MMP2, influenced by the regulation in associated pathways, have been shown to either promote or inhibit tumour cell invasion in PCa [85]. Elevated MMP9 expression was reported in PCa patients compared to those with benign prostatic hyperplasia [86,87]. Higher MMP9 expression levels are associated with an increased metastatic rate, and its inhibition may reduce the metastatic potential of PCa [88]. MMP9 plays a crucial role in multiple stages of cancer development, including reducing cancer cell apoptotic potential, promoting angiogenesis, and modulating the immune response to cancer cells [89]. However, the precise mechanism by which hormonal therapy influences MMP9 expression levels remains unclear, necessitating further investigation. In this current study, analysis of the EV cargo proteomics data, indicating a more aggressive signature, correlates with the findings that MMP2 and MMP9 mRNA levels were enhanced following the 4 h LMF exposure in PC3 cells. Indeed, the Matrigel invasion assay confirmed that LMF exposure increased cellular invasion capabilities while proliferation did not change within the 4 h exposure time.
Previous studies on hypomagnetic treatment of neuroblastoma cells showed wide-ranging changes in decreased gene expression associated with cell survival and cell death regulation [90]. It is also of interest that in neuronal models, hypomagnetic exposure induced proliferation rate [57] and this was also observed for neuroblastoma cells [91]. This may vary between cell types, as no effects on normal endothelial cells were observed in other studies [92]. Extremely low-frequency electromagnetic fields of 0.3 μT have been reported to reduce cancer cell migration, increase proliferation, and enhance uptake in cytotoxic drugs in PC12, THP-1, and HeLa cancer cell lines following 30 min LMF treatment [24]. However, proliferation was not affected in the PC3 or PNT2 cells in our current study following 4 h LMF (20 nT) exposure. Further time windows and ranges of LMF exposure may need to be explored in future studies to fully understand the effects of short- and longer-term LMF, as well as ranging LMF exposures on different cancer cells and cancer-type properties.
The effects of magnetic fields on health and disease are of great interest, and investigations into therapeutic benefits as well as disease risk are required to understand the complex phenomena associated with their still relatively poorly understood functions [93]. Furthermore, interest in utilising magnetic-based therapies in cancer includes, for example, the development of magnetic nanoparticles [94,95,96]. Various limitations for studies on the effects of changed magnetic field exposure, including low magnetic and hypomagnetic conditions, on cellular and organismal systems cannot be ignored and will need attention to move the field forwards. This includes different experimental setups, types of devices used, time windows and levels of exposure, and additional effects, including radical pairs and interference quantum effects [38]. These variables may make it hard to compare experiments between studies and also hamper the repeatability of experiments between research groups. Differing sensitivity of cellular mechanisms to magneto-biological effects may also vary between cell types and organ systems [93], and studies have indeed reported different outcomes between cell types, including cancer types, as also mentioned in our discussion above. Furthermore, the translatability from cellular to organismal level may be an additional challenge, although comparisons between some studies, including our current findings with the wider literature, are encouraging in this aspect. Influences of hypomagnetic conditions on living organisms and the increased interest in this research field, also in relation to future space missions, have recently been extensively reviewed by Sarimov et al. [38]. Further research in both in vitro and in vivo models is, therefore, of high priority and will aid the future standardisation and optimisation of methods. Importantly, in our current study, we report that even a short (4 h) exposure to magnetic shielding significantly modified cellular EV release profiles and induced notable molecular changes in pro-metastatic pathways at the protein and nucleic acid levels. Research into molecular and cellular mechanisms, including our findings here, will contribute to furthering the current understanding of hypomagnetic conditions at both cellular and organismal levels. This may aid in identifying disease-associated, but possibly also health-promoting, mechanisms with clinical relevance.

5. Conclusions

This study identified novel molecular and cellular communication mechanisms affected by short-term low magnetic/hypomagnetic exposure by magnetic field shielding, with significant effects on cancer cells. The findings of this study indicated that short-term 4 h LMF (20 nT) exposure/magnetic shielding induced significant changes in prostate cancer cell properties in vitro. More pro-cancerous changes were observed in EV profiles, oncogenic miRNA expressions, and cellular invasion capabilities of PC3 cells without affecting proliferation. EV protein content was modified to a more pro-inflammatory and cancerous signature following 4 h LMF exposure, based on functional enrichment analysis. This correlated with the upregulation in oncogenic miRNAs miR-155, miR-21, and miR-210, alongside the downregulation in tumour-suppressive miRNAs, miR-126 and miR-200c, suggesting that LMF exposure may promote a more aggressive cancer phenotype by modulating gene expression pathways associated with tumour progression. Additionally, a significant increase in metalloproteinase MMP2 and MMP9 expression, which are linked to enhanced metastatic potential, further underscored the potential of LMF to influence PC3 cancer cell behaviour. Our findings report new mechanisms of LMF-induced cellular, molecular, and epigenetic changes and highlight the need to explore the potential implications of modulated magnetic field exposure on human health. Our study also emphasises the importance of investigating the differential effects of LMF on normal cells, as evidenced by the distinct EV proteome changes observed in PNT2 cells. Improving current understanding of downstream mechanisms due to altered magnetic fields is important in medical research, including cancer biology, with putative implications for future therapeutic strategies as biological reactions in cancer cells may be adapted to normal MF strength, and MF shielding may influence cellular behaviour.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/biology13090734/s1, Figure S1. Full western blot images for CD63 and Flot-1 on EVs. Table S1. PC3 control EV proteomes by LC-MS/MS analysis. Table S2. PC3 4 h LMF-treated EV proteomes by LC-MS/MS analysis. Table S3. PNT2 control EV proteomes by LC-MS/MS analysis. Table S4. PNT2 4 h LMF-treated EV proteomes by LC-MS/MS analysis.

Author Contributions

Conceptualization, S.L. and P.U.-O.; methodology, S.L., J.M.I., I.K., D.A.D. and P.U-O.; validation, S.L. and P.U.-O.; formal analysis, S.L. and P.U.-O.; investigation, S.L. and P.U.-O.; resources, S.L., J.M.I., I.K., D.A.D. and P.U.-O.; data curation, S.L. and P.U.-O.; writing—original draft preparation, S.L. and P.U.-O.; writing—review and editing, S.L., J.M.I., I.K., D.A.D. and P.U.-O.; visualization, S.L., I.K. and P.U.-O.; supervision, S.L. and P.U.-O.; project administration, S.L. and P.U.-O.; funding acquisition, S.L., J.M.I., I.K. and P.U.-O. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding..

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The original contributions presented in the study are included in the article/Supplementary Materials, further inquiries can be directed to the corresponding authors.

Acknowledgments

Thanks to the Cambridge Centre for Proteomics for the LC-MS/MS analysis. Thanks to the Guy Foundation for equipment used in this study.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Wang, L.; Lu, B.; He, M.; Wang, Y.; Wang, Z.; Du, L. Prostate Cancer Incidence and Mortality: Global Status and Temporal Trends in 89 Countries from 2000 to 2019. Front. Public Health 2022, 10, 811044. [Google Scholar] [CrossRef] [PubMed]
  2. Bergengren, O.; Pekala, K.R.; Matsoukas, K.; Fainberg, J.; Mungovan, S.F.; Bratt, O.; Bray, F.; Brawley, O.; Luckenbaugh, A.N.; Mucci, L.; et al. 2022 Update on Prostate Cancer Epidemiology and Risk Factors-A Systematic Review. Eur. Urol. 2023, 84, 191–206. [Google Scholar] [CrossRef] [PubMed]
  3. Berenguer, C.V.; Pereira, F.; Câmara, J.S.; Pereira, J.A.M. Underlying Features of Prostate Cancer-Statistics, Risk Factors, and Emerging Methods for Its Diagnosis. Curr. Oncol. 2023, 30, 2300–2321. [Google Scholar] [CrossRef] [PubMed]
  4. Pernar, C.H.; Ebot, E.M.; Wilson, K.M.; Mucci, L.A. The Epidemiology of Prostate Cancer. Cold Spring Harb. Perspect. Med. 2018, 8, a030361. [Google Scholar] [CrossRef]
  5. Mullins, J.K.; Loeb, S. Environmental exposures and prostate cancer. Urol. Oncol. 2012, 30, 216–219. [Google Scholar] [CrossRef]
  6. Brain, J.D.; Kavet, R.; McCormick, D.L.; Poole, C.; Silverman, L.B.; Smith, T.J.; Valberg, P.A.; Van Etten, R.A.; Weaver, J.C. Childhood leukemia: Electric and magnetic fields as possible risk factors. Environ. Health Perspect. 2003, 111, 962–970. [Google Scholar] [CrossRef]
  7. Mazzoccoli, G. Chronobiology Meets Quantum Biology: A New Paradigm Overlooking the Horizon? Front. Physiol. 2022, 13, 892582. [Google Scholar] [CrossRef]
  8. Turchinovich, A.; Weiz, L.; Langheinz, A.; Burwinkel, B. Characterization of extracellular circulating microRNA. Nucleic Acids Res. 2011, 39, 7223–7233. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  9. Mortoglou, M.; Tabin, Z.K.; Arisan, E.D.; Kocher, H.M.; Uysal-Onganer, P. Non-coding RNAs in pancreatic ductal adenocarcinoma: New approaches for better diagnosis and therapy. Transl Oncol. 2021, 14, 101090. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  10. Lanning, B.; Webber, J.; Uysal-Onganer, P.; Jiang, W.G.; Clayton, A.; Dart, D.A. Prostate Cancer Cell Extracellular Vesicles Increase Mineralisation of Bone Osteoblast Precursor Cells in an In Vitro Model. Biology 2021, 10, 318. [Google Scholar] [CrossRef]
  11. Mortoglou, M.; Miralles, F.; Arisan, E.D.; Dart, A.; Jurcevic, S.; Lange, S.; Uysal-Onganer, P. microRNA-21 Regulates Stemness in Pancreatic Ductal Adenocarcinoma Cells. Int. J. Mol. Sci. 2022, 23, 1275. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  12. Arisan, E.D.; Rencuzogullari, O.; Keskin, B.; Grant, G.H.; Uysal-Onganer, P. Inhibition on JNK Mimics Silencing of Wnt-11 Mediated Cellular Response in Androgen-Independent Prostate Cancer Cells. Biology 2020, 9, 142. [Google Scholar] [CrossRef] [PubMed]
  13. Alavanda, C.; Dirimtekin, E.; Mortoglou, M.; Arslan Ates, E.; Guney, A.I.; Uysal-Onganer, P. BRCA Mutations and MicroRNA Expression Patterns in the Peripheral Blood of Breast Cancer Patients. ACS Omega 2024, 9, 17217–17228. [Google Scholar] [CrossRef] [PubMed]
  14. Kosgodage, U.S.; Uysal-Onganer, P.; MacLatchy, A.; Kraev, I.; Chatterton, N.P.; Nicholas, A.P.; Inal, J.M.; Lange, S. Peptidylarginine Deiminases Post-Translationally Deiminate Prohibitin and Modulate Extracellular Vesicle Release and MicroRNAs in Glioblastoma Multiforme. Int. J. Mol. Sci. 2018, 20, 103. [Google Scholar] [CrossRef]
  15. Koushyar, S.; Uysal-Onganer, P.; Jiang, W.G.; Dart, D.A. Prohibitin Links Cell Cycle, Motility and Invasion in Prostate Cancer Cells. Int. J. Mol. Sci. 2023, 24, 9919. [Google Scholar] [CrossRef]
  16. Uysal-Onganer, P.; D’Alessio, S.; Mortoglou, M.; Kraev, I.; Lange, S. Peptidylarginine Deiminase Inhibitor Application, Using Cl-Amidine, PAD2, PAD3 and PAD4 Isozyme-Specific Inhibitors in Pancreatic Cancer Cells, Reveals Roles for PAD2 and PAD3 in Cancer Invasion and Modulation of Extracellular Vesicle Signatures. Int. J. Mol. Sci. 2021, 22, 1396. [Google Scholar] [CrossRef]
  17. Arisan, E.D.; Rencuzogullari, O.; Cieza-Borrella, C.; Miralles Arenas, F.; Dwek, M.; Lange, S.; Uysal-Onganer, P. MiR-21 Is Required for the Epithelial-Mesenchymal Transition in MDA-MB-231 Breast Cancer Cells. Int. J. Mol. Sci. 2021, 22, 1557. [Google Scholar] [CrossRef]
  18. Dirimtekin, E.; Mortoglou, M.; Alavanda, C.; Benomar Yemlahi, A.; Arslan Ates, E.; Guney, I.; Uysal-Onganer, P. miR-34a-FOXP1 Loop in Ovarian Cancer. ACS Omega 2023, 8, 27743–27750. [Google Scholar] [CrossRef]
  19. Cheng, L.; Hill, A.F. Therapeutically harnessing extracellular vesicles. Nat. Rev. Drug Discov. 2022, 21, 379–399. [Google Scholar] [CrossRef]
  20. Robinson, H.; Roberts, M.J.; Gardiner, R.A.; Hill, M.M. Extracellular vesicles for precision medicine in prostate cancer—Is it ready for clinical translation? Semin. Cancer Biol. 2023, 89, 18–29. [Google Scholar] [CrossRef]
  21. Kosgodage, U.S.; Trindade, R.P.; Thompson, P.R.; Inal, J.M.; Lange, S. Chloramidine/Bisindolylmaleimide-I-Mediated Inhibition of Exosome and Microvesicle Release and Enhanced Efficacy of Cancer Chemotherapy. Int. J. Mol. Sci. 2017, 18, 1007. [Google Scholar] [CrossRef] [PubMed]
  22. Uysal-Onganer, P.; MacLatchy, A.; Mahmoud, R.; Kraev, I.; Thompson, P.R.; Inal, J.M.; Lange, S. Peptidylarginine Deiminase Isozyme-Specific PAD2, PAD3 and PAD4 Inhibitors Differentially Modulate Extracellular Vesicle Signatures and Cell Invasion in Two Glioblastoma Multiforme Cell Lines. Int. J. Mol. Sci. 2020, 21, 1495. [Google Scholar] [CrossRef] [PubMed]
  23. Stratton, D.; Lange, S.; Inal, J.M. Pulsed extremely low-frequency magnetic fields stimulate microvesicle release from human monocytic leukaemia cells. Biochem. Biophys. Res. Commun. 2013, 430, 470–475. [Google Scholar] [CrossRef] [PubMed]
  24. Stratton, D.; Malibha-Pinchbeck, M.; Inal, J. Extremely low-frequency magnetic fields significantly enhance the cytotoxicity of methotrexate and can reduce migration of cancer cell lines via transiently induced plasma membrane damage. Biochem. Biophys. Res. Commun. 2022, 626, 192–199. [Google Scholar] [CrossRef] [PubMed]
  25. Dart, D.A.; Koushyar, S.; Uysal-Onganer, P. Exploring the potential link between prostate cancer and magnetic fields. Med. Hypotheses 2024, 189, 111384. [Google Scholar] [CrossRef]
  26. Davey, A.; McAuley, D.F.; O’Kane, C.M. Matrix metalloproteinases in acute lung injury: Mediators of injury and drivers of repair. Eur. Respir. J. 2011, 38, 959–970. [Google Scholar] [CrossRef]
  27. Gohji, K.; Fujimoto, N.; Hara, I.; Fujii, A.; Gotoh, A.; Okada, H.; Arakawa, S.; Kitazawa, S.; Miyake, H.; Kamidono, S.; et al. Serum matrix metalloproteinase-2 and its density in men with prostate cancer as a new predictor of disease extension. Int. J. Cancer 1998, 79, 96–101. [Google Scholar] [CrossRef]
  28. Moses, M.A.; Wiederschain, D.; Loughlin, K.R.; Zurakowski, D.; Lamb, C.C.; Freeman, M.R. Increased incidence of matrix metalloproteinases in urine of cancer patients. Cancer Res. 1998, 58, 1395–1399. [Google Scholar]
  29. Livak, K.J.; Schmittgen, T.D. Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method. Methods 2001, 25, 402–408. [Google Scholar] [CrossRef]
  30. Kato, T.; Fujita, Y.; Nakane, K.; Kojima, T.; Nozawa, Y.; Deguchi, T.; Ito, M. ETS1 promotes chemoresistance and invasion of paclitaxel-resistant, hormone-refractory PC3 prostate cancer cells by up-regulating MDR1 and MMP9 expression. Biochem. Biophys. Res. Commun. 2012, 417, 966–971. [Google Scholar] [CrossRef]
  31. Welsh, J.A.; Goberdhan DC, I.; O’Driscoll, L.; Buzas, E.I.; Blenkiron, C.; Bussolati, B.; Cai, H.; Di Vizio, D.; Driedonks TA, 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] [PubMed]
  32. Dart, D.A.; Bevan, C.L.; Uysal-Onganer, P.; Jiang, W.G. Analysis of androgen receptor expression and activity in the mouse brain. Sci. Rep. 2024, 14, 11115. [Google Scholar] [CrossRef] [PubMed]
  33. de Nóbrega, M.; Dos Reis, M.B.; Pereira, É.R.; de Souza, M.F.; de Syllos Cólus, I.M. The potential of cell-free and exosomal microRNAs as biomarkers in liquid biopsy in patients with prostate cancer. J. Cancer Res. Clin. Oncol. 2022, 148, 2893–2910. [Google Scholar] [CrossRef] [PubMed]
  34. Cavallari, I.; Ciccarese, F.; Sharova, E.; Urso, L.; Raimondi, V.; Silic-Benussi, M.; D’Agostino, D.M.; Ciminale, V. The miR-200 Family of microRNAs: Fine Tuners of Epithelial-Mesenchymal Transition and Circulating Cancer Biomarkers. Cancers 2021, 13, 5874. [Google Scholar] [CrossRef]
  35. Angel, C.Z.; Lynch, S.M.; Nesbitt, H.; McKenna, M.M.; Walsh, C.P.; McKenna, D.J. miR-210 is induced by hypoxia and regulates neural cell adhesion molecule in prostate cells. J. Cell. Physiol. 2020, 235, 6194–6203. [Google Scholar] [CrossRef]
  36. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA A Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef]
  37. Gandaglia, G.; Leni, R.; Bray, F.; Fleshner, N.; Freedland, S.J.; Kibel, A.; Stattin, P.; Van Poppel, H.; La Vecchia, C. Epidemiology and prevention of prostate cancer. Eur. Urol. Oncol. 2021, 4, 877–892. [Google Scholar] [CrossRef]
  38. Sarimov, R.M.; Serov, D.A.; Gudkov, S.V. Hypomagnetic Conditions and Their Biological Action (Review). Biology 2023, 12, 1513. [Google Scholar] [CrossRef]
  39. Aydemir, E.; Arslan, İ.İ.; Görkay, A.H. The Application of Electromagnetic Fields in Cancer. Adv. Exp. Med. Biol. 2024, 1450, 103–120. [Google Scholar] [CrossRef]
  40. Akoto, T.; Saini, S. Role of Exosomes in Prostate Cancer Metastasis. Int. J. Mol. Sci. 2021, 22, 3528. [Google Scholar] [CrossRef]
  41. Henrich, S.E.; McMahon, K.M.; Plebanek, M.P.; Calvert, A.E.; Feliciano, T.J.; Parrish, S.; Tavora, F.; Mega, A.; De Souza, A.; Carneiro, B.A.; et al. Prostate cancer extracellular vesicles mediate intercellular communication with bone marrow cells and promote metastasis in a cholesterol-dependent manner. J. Extracell. Vesicles 2020, 10, e12042. [Google Scholar] [CrossRef] [PubMed]
  42. Linder, S.; Cervero, P.; Eddy, R.; Condeelis, J. Mechanisms and roles of podosomes and invadopodia. Nat. Rev. Mol. Cell Biol. 2023, 24, 86–106. [Google Scholar] [CrossRef] [PubMed]
  43. Boran, C.; Kandirali, E.; Yilmaz, F.; Serin, E.; Akyol, M. Reliability of the 34βE12, keratin 5/6, p63, bcl-2, and AMACR in the diagnosis of prostate carcinoma. Urol. Oncol. 2011, 29, 614–623. [Google Scholar] [CrossRef] [PubMed]
  44. Guo, L.; Liu, Y.; Yang, T.; Wang, G.; Liu, J.; Li, S.; Liu, B.; Cai, J. CAV1 and KRT5 are potential targets for prostate cancer. Medicine 2023, 102, e36473. [Google Scholar] [CrossRef] [PubMed]
  45. Chang, Y.T.; Peng, H.Y.; Hu, C.M.; Tien, S.C.; Chen, Y.I.; Jeng, Y.M.; Chang, M.C. Pancreatic cancer-derived small extracellular vesical ezrin activates fibroblasts to exacerbate cancer metastasis through STAT3 and YAP-1 signaling pathways. Mol. Oncol. 2023, 17, 1628–1647. [Google Scholar] [CrossRef]
  46. Chen, Z.; Wang, J.; Lu, Y.; Lai, C.; Qu, L.; Zhuo, Y. Ezrin expression in circulating tumor cells is a predictor of prostate cancer metastasis. Bioengineered 2022, 13, 4076–4084. [Google Scholar] [CrossRef]
  47. Dryhurst, D.; Ausió, J. Histone H2A.Z deregulation in prostate cancer. Cause or effect? Cancer Metastasis Rev. 2014, 33, 429–439. [Google Scholar] [CrossRef]
  48. Yang, B.; Tong, R.; Liu, H.; Wu, J.; Chen, D.; Xue, Z.; Ding, C.; Zhou, L.; Xie, H.; Wu, J.; et al. H2A.Z regulates tumorigenesis, metastasis and sensitivity to cisplatin in intrahepatic cholangiocarcinoma. Int. J. Oncol. 2018, 52, 1235–1245. [Google Scholar] [CrossRef]
  49. Zhou, S.; Cai, Y.; Liu, X.; Jin, L.; Wang, X.; Ma, W.; Zhang, T. Role of H2B mono-ubiquitination in the initiation and progression of cancer. Bull. Du Cancer 2021, 108, 385–398. [Google Scholar] [CrossRef]
  50. Ronquist, K.G.; Ronquist, G.; Larsson, A.; Carlsson, L. Proteomic analysis of prostate cancer metastasis-derived prostasomes. Anticancer Res. 2010, 30, 285–290. [Google Scholar]
  51. Hishida, S.; Kawakami, K.; Fujita, Y.; Kato, T.; Takai, M.; Iinuma, K.; Nakane, K.; Tsuchiya, T.; Koie, T.; Miura, Y.; et al. Proteomic analysis of extracellular vesicles identified PI3K pathway as a potential therapeutic target for cabazitaxel-resistant prostate cancer. Prostate 2021, 81, 592–602. [Google Scholar] [CrossRef] [PubMed]
  52. Gómez-Herranz, M.; Taylor, J.; Sloan, R.D. IFITM proteins: Understanding their diverse roles in viral infection, cancer, and immunity. J. Biol. Chem. 2023, 299, 102741. [Google Scholar] [CrossRef] [PubMed]
  53. Stewart, G.D.; Lowrie, A.G.; Riddick, A.C.; Fearon, K.C.; Habib, F.K.; Ross, J.A. Dermcidin expression confers a survival advantage in prostate cancer cells subjected to oxidative stress or hypoxia. Prostate 2007, 67, 1308–1317. [Google Scholar] [CrossRef] [PubMed]
  54. Acikgoz, E.; Soner, B.C.; Ozdil, B.; Guven, M. CD133+/CD44+ prostate cancer stem cells exhibit embryo-like behavior patterns. Acta Histochem. 2021, 123, 151743. [Google Scholar] [CrossRef] [PubMed]
  55. Zhou, X.; Yeasmin Khusbu, F.; Xie, Y.; Yang, P. Emodin-Induced Necroptosis in Prostate Cancer Cells via the Mitochondrial Fission HSP90/MLKL/PGAM Pathway. Chem. Biodivers. 2023, 20, e202201130. [Google Scholar] [CrossRef]
  56. He, T.; Zhang, Q.; Xu, P.; Tao, W.; Lin, F.; Liu, R.; Li, M.; Duan, X.; Cai, C.; Gu, D.; et al. Extracellular vesicle-circEHD2 promotes the progression of renal cell carcinoma by activating cancer-associated fibroblasts. Mol. Cancer 2023, 22, 117. [Google Scholar] [CrossRef]
  57. Fu, J.-P.; Mo, W.-C.; Liu, Y.; Bartlett, P.F.; He, R.-Q. Elimination of the geomagnetic field stimulates the proliferation of mouse neural progenitor and stem cells. Protein Cell 2016, 7, 624–637. [Google Scholar] [CrossRef] [PubMed]
  58. Fu, J.-P.; Mo, W.-C.; Liu, Y.; He, R.-Q. Decline of cell viability and mitochondrial activity in mouse skeletal muscle cell in a hypomagnetic field. Bioelectromagnetics 2016, 37, 212–222. [Google Scholar] [CrossRef]
  59. Katiukhin, L.N. Rheological Properties of the Erythrocytes in Weakened Static Magnetic Field of the Earth In Vitro Study. J. Sci. Res. Rep. 2019, 22, 1–12. [Google Scholar] [CrossRef]
  60. Ciorba, D.; Morariu, V.V. Life in zero magnetic field. Iii. Activity of aspartate aminotransferase and alanine aminotransferase during in vitro aging of human blood. Electro-Magnetobiology 2009, 20, 313–321. [Google Scholar] [CrossRef]
  61. Krylov, V.; Machikhin, A.; Sizov, D.; Guryleva, A.; Sizova, A.; Zhdanova, S.; Tchougounov, V.; Burlakov, A. Influence of hypomagnetic field on the heartbeat in zebrafish embryos. Front. Physiol. 2022, 13, 1040083. [Google Scholar] [CrossRef] [PubMed]
  62. Mo, W.-C.; Fu, J.-P.; Ding, H.-M.; Liu, Y.; Hua, Q.; He, R.-Q. Hypomagnetic Field Alters Circadian Rhythm and Increases Algesia in Adult Male Mice. Prog. Biochem. Biophys. 2015, 42, 639–646. [Google Scholar] [CrossRef]
  63. Novikov, V.V.; Yablokova, E.V.; Fesenko, E.E. The Effect of a “Zero” Magnetic Field on the Production of Reactive Oxygen Species in Neutrophils. Biophysics 2018, 63, 365–368. [Google Scholar] [CrossRef]
  64. Baek, S.; Choi, H.; Park, H.; Cho, B.; Kim, S.; Kim, J. Effects of a hypomagnetic field on DNA methylation during the differentiation of embryonic stem cells. Sci. Rep. 2019, 9, 1333. [Google Scholar] [CrossRef] [PubMed]
  65. Poiata, A.; Creanga, D.E.; Morariu, V.V. Life in zero magnetic field. V. E. coli resistance to antibiotics. Electromagn. Biol. Med. 2009, 22, 171–182. [Google Scholar] [CrossRef]
  66. Ilyin, V.K.; Orlov, O.I.; Morozova, Y.A.; Skedina, M.A.; Vladimirov, S.K.; Plotnikov, E.V.; Artamonov, A.A. Prognostic model for bacterial drug resistance genes horizontal spread in space-crews. Acta Astronaut. 2022, 190, 388–394. [Google Scholar] [CrossRef]
  67. Zhang, B.; Wang, L.; Zhan, A.; Wang, M.; Tian, L.; Guo, W.; Pan, Y. Long-term exposure to a hypomagnetic field attenuates adult hippocampal neurogenesis and cognition. Nat. Commun. 2021, 12, 1174. [Google Scholar] [CrossRef]
  68. Jagetia, G.C.; Martino, C.F.; Castello, P.R. Modulation of Hydrogen Peroxide Production in Cellular Systems by Low Level Magnetic Fields. PLoS ONE 2011, 6, e22753. [Google Scholar] [CrossRef]
  69. Juutilainen, J.; Herrala, M.; Luukkonen, J.; Naarala, J.; Hore, P.J. Magnetocarcinogenesis: Is there a mechanism for carcinogenic effects of weak magnetic fields? Proc. R. Soc. B Biol. Sci. 2018, 285, 20180590. [Google Scholar] [CrossRef]
  70. Hu, P.; Mo, W.-C.; Fu, J.-P.; Liu, Y.; He, R.Q. Long-term hypogeomagnetic field exposure reduces muscular mitochondrial function and exercise capacity in adult male mice. Prog. Biochem. Biophys. 2020, 47, 426–438. [Google Scholar] [CrossRef]
  71. Jia, B.; Xie, L.; Zheng, Q.; Yang, P.F.; Zhang, W.J.; Ding, C.; Qian, A.R.; Shang, P.A. A Hypomagnetic Field Aggravates Bone Loss Induced by Hindlimb Unloading in Rat Femurs. PLoS ONE 2014, 9, e105604. [Google Scholar] [CrossRef] [PubMed]
  72. Wan, G.-J.; Jiang, S.-L.; Zhao, Z.-C.; Xu, J.-J.; Tao, X.-R.; Sword, G.A.; Gao, Y.-B.; Pan, W.-D.; Chen, F.-J. Bio-effects of near-zero magnetic fields on the growth, development and reproduction of small brown planthopper, Laodelphax striatellus and brown planthopper, Nilaparvata lugens. J. Insect Physiol. 2014, 68, 7–15. [Google Scholar] [CrossRef] [PubMed]
  73. Ogneva, I.V.; Usik, M.A.; Burtseva, M.V.; Biryukov, N.S.; Zhdankina, Y.S.; Sychev, V.N.; Orlov, O.I. Drosophila melanogaster Sperm under Simulated Microgravity and a Hypomagnetic Field: Motility and Cell Respiration. Int. J. Mol. Sci. 2020, 21, 5985. [Google Scholar] [CrossRef] [PubMed]
  74. Fesenko, E.E.; Mezhevikina, L.M.; Osipenko, M.A.; Gordon, R.Y.; Khutzian, S.S. Effect of the “zero” Magnetic Field on Early Embryogenesis in Mice. Electromagn. Biol. Med. 2010, 29, 1–8. [Google Scholar] [CrossRef] [PubMed]
  75. Mo, W.-C.; Liu, Y.; Cooper, H.M.; He, R.-Q. Altered development of Xenopus embryos in a hypogeomagnetic field. Bioelectromagnetics 2012, 33, 238–246. [Google Scholar] [CrossRef] [PubMed]
  76. Temour’yants, N.A.; Kostyuk, A.S.; Tumanyants, K.N. Dynamics and Infradian Rhythmics of Thermal/Pain Sensitivity of the Helix Mollusc under the Action of Electromagnetic Fields. Neurophysiology 2011, 42, 276–285. [Google Scholar] [CrossRef]
  77. Dart, D.A.; Uysal-Onganer, P.; Jiang, W.G. Prostate-specific PTen deletion in mice activates inflammatory microRNA expression pathways in the epithelium early in hyperplasia development. Oncogenesis 2017, 6, 400. [Google Scholar] [CrossRef]
  78. Lian, M.; Mortoglou, M.; Uysal-Onganer, P. Impact of Hypoxia-Induced miR-210 on Pancreatic Cancer. Curr. Issues Mol. Biol. 2023, 45, 9778–9792. [Google Scholar] [CrossRef]
  79. Jalil, A.T.; Abdulhadi, M.A.; Al-Ameer, L.R.; Abbas, H.A.; Merza, M.S.; Zabibah, R.S.; Fadhil, A.A. The emerging role of microRNA-126 as a potential therapeutic target in cancer: A comprehensive review. Pathol. Res. Pract. 2023, 248, 154631. [Google Scholar] [CrossRef]
  80. Brabletz, S.; Brabletz, T. The ZEB/miR-200 feedback loop--a motor of cellular plasticity in development and cancer? EMBO Rep. 2010, 11, 670–677. [Google Scholar] [CrossRef]
  81. Pinheiro, L.C.L.; Pereira, É.R.; Francelino, A.L.; Guembarovski, A.F.M.L.; Fuganti, P.E.; de Oliveira, K.B.; Miqueloto, C.A.; Serpeloni, J.M.; Guembarovski, R.L. Metalloproteinase 9 immunostaining profile is positively correlated with tumor grade, extraprostatic extension and biochemical recurrence in prostate cancer. Pathol Res Pract. 2024, 253, 155024. [Google Scholar] [CrossRef] [PubMed]
  82. Mandel, A.; Larsson, P.; Sarwar, M.; Semenas, J.; Syed Khaja, A.S.; Persson, J.L. The interplay between AR, EGF receptor and MMP-9 signaling pathways in invasive prostate cancer. Mol Med. 2018, 24, 34. [Google Scholar] [CrossRef] [PubMed]
  83. Zhang, L.; Shi, J.; Feng, J.; Klocker, H.; Lee, C.; Zhang, J. Type IV collagenase (matrix metalloproteinase-2 and -9) in prostate cancer. Prostate Cancer Prostatic Dis. 2004, 7, 327–332. [Google Scholar] [CrossRef]
  84. dos Reis, S.T.; Villanova, F.E.; Andrade, P.M.; Pontes, J., Jr.; de Sousa-Canavez, J.M.; Sañudo, A.; Antunes, A.A.; Dall’oglio, M.F.; Srougi, M.; Moreira Leite, K.R. Matrix metalloproteinase-2 polymorphism is associated with prognosis in prostate cancer. Urol. Oncol. 2010, 28, 624–627. [Google Scholar] [CrossRef] [PubMed]
  85. Lovaas, J.D.; Zhu, L.; Chiao, C.Y.; Byles, V.; Faller, D.V.; Dai, Y. SIRT1 enhances matrix metalloproteinase-2 expression and tumor cell invasion in prostate cancer cells. Prostate 2013, 73, 522–530. [Google Scholar] [CrossRef] [PubMed]
  86. Zhong, W.D.; Han, Z.D.; He, H.C.; Bi, X.C.; Dai, Q.S.; Zhu, G.; Ye, Y.K.; Liang, Y.X.; Qin, W.J.; Zhang, Z.; et al. CD147, MMP-1, MMP-2 and MMP-9 protein expression as significant prognostic factors in human prostate cancer. Oncology 2008, 75, 230–236. [Google Scholar] [CrossRef]
  87. Trudel, D.; Fradet, Y.; Meyer, F.; Têtu, B. Matrix metalloproteinase 9 is associated with Gleason score in prostate cancer but not with prognosis. Hum. Pathol. 2010, 41, 1694–1701. [Google Scholar] [CrossRef]
  88. Ma, F.; Wang, Z.; Abdularab, A.; Dai, L.; Niu, Y.; Jiang, N. Matrix metalloproteinase 9 and prostate cancer risk: A meta-analysis of 1059 participants. Minerva Urol. E Nefrol. Ital. J. Urol. Nephrol. 2017, 69, 324–329. [Google Scholar] [CrossRef]
  89. Nelson, A.R.; Fingleton, B.; Rothenberg, M.L.; Matrisian, L.M. Matrix metalloproteinases: Biologic activity and clinical implications. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 2000, 18, 1135–1149. [Google Scholar] [CrossRef]
  90. Mo, W.; Liu, Y.; Bartlett, P.F.; He, R. Transcriptome profile of human neuroblastoma cells in the hypomagnetic field. Sci. China Life Sci. 2014, 57, 448–461. [Google Scholar] [CrossRef]
  91. Roemer, K.; Mo, W.-C.; Zhang, Z.-J.; Liu, Y.; Bartlett, P.F.; He, R.-Q. Magnetic Shielding Accelerates the Proliferation of Human Neuroblastoma Cell by Promoting G1-Phase Progression. PLoS ONE 2013, 8, e54775. [Google Scholar] [CrossRef]
  92. Eldashev, I.S.; Shchegolev, B.F.; Surma, S.V.; Belostotskaya, G.B. Influence of low-intensity magnetic fields on the development of satellite muscle cells of a newborn rat in primary culture. Biophysics 2011, 55, 765–770. [Google Scholar] [CrossRef]
  93. Sarimov, R.M.; Serov, D.A.; Gudkov, S.V. Biological Effects of Magnetic Storms and ELF Magnetic Fields. Biology 2023, 12, 1506. [Google Scholar] [CrossRef] [PubMed]
  94. Singh, A.; Kumar, P.; Pathak, S.; Jain, K.; Garg, P.Y.; Pant, M.; Mahapatro, A.K.; Rath, D.; Wang, L.; Kim, S.-K.; et al. A threefold increase in SAR performance for magnetic hyperthermia by compositional tuning in zinc-substituted iron oxide superparamagnetic nanoparticles with superior biocompatibility. J. Alloys Compd. 2023, 968, 171868. [Google Scholar] [CrossRef]
  95. Singh, A.; Kumar, P.; Pathak, S.; Jain, K.; Garg, P.; Pant, M.; Mahapatro, A.K.; Singh, R.K.; Rajput PMKim, S.-K.; Maurya, K.K.; et al. Tailored nanoparticles for magnetic hyperthermia: Highly stable aqueous dispersion of Mn-substituted magnetite superparamagnetic nanoparticles by double surfactant coating for improved heating efficiency. J. Alloys Compd. 2024, 976, 172999. [Google Scholar] [CrossRef]
  96. Li, B.; Zu, M.; Jiang, A.; Cao, Y.; Wu, J.; Shahbazi, M.A.; Shi, X.; Reis, R.L.; Kundu, S.C.; Xiao, B. Magnetic natural lipid nanoparticles for oral treatment of colorectal cancer through potentiated antitumor immunity and microbiota metabolite regulation. Biomaterials 2024, 307, 122530. [Google Scholar] [CrossRef]
Figure 1. EVs isolated from PC3 and PNT2 cells. (AD) NTA profiles of PC3 and PNT2 EVs, comparing control and 4 h magnetic field shielding (LMF). (E) Western blotting confirms two EV-specific surface markers, CD63 and Flotillin-1 (see Supplementary Figure S1 for full western blot). (F) Representative transmission electron microscopy (TEM) images of isolated EV are shown with scale bars indicated in µm or nm.
Figure 1. EVs isolated from PC3 and PNT2 cells. (AD) NTA profiles of PC3 and PNT2 EVs, comparing control and 4 h magnetic field shielding (LMF). (E) Western blotting confirms two EV-specific surface markers, CD63 and Flotillin-1 (see Supplementary Figure S1 for full western blot). (F) Representative transmission electron microscopy (TEM) images of isolated EV are shown with scale bars indicated in µm or nm.
Biology 13 00734 g001
Figure 2. EV profiling from PC3 and PNT2 cells, comparing control and 4 h LMF exposure. (A) Total EV numbers released from the cells; (B) EV subpopulations from PC3 cells, control and LMF exposed; (C) EV subpopulations from PNT2 cells, control and LMF exposed; (D) mean size of EVs released from PC3 and PNT2 cells, showing controls and LMF exposed; (E) modal sizes of EVs from PC3 and PNT2 cells, showing control and LMF exposed; * p < 0.05.
Figure 2. EV profiling from PC3 and PNT2 cells, comparing control and 4 h LMF exposure. (A) Total EV numbers released from the cells; (B) EV subpopulations from PC3 cells, control and LMF exposed; (C) EV subpopulations from PNT2 cells, control and LMF exposed; (D) mean size of EVs released from PC3 and PNT2 cells, showing controls and LMF exposed; (E) modal sizes of EVs from PC3 and PNT2 cells, showing control and LMF exposed; * p < 0.05.
Biology 13 00734 g002
Figure 3. Proteomic analysis of EV cargoes from 4 h LMF-treated (and control untreated) PC3 and PNT2 cells. (A) EV protein content, as assessed by SDS-PAGE and silver staining, from PC3-derived EVs, showing control untreated (CTR) and LMF-treated cell-derived EVs. (B) Protein content from PNT2-derived EVs, showing control untreated (CTR) and LMF-treated cell-derived EVs. (C,D) Venn diagrams showing shared and specific proteins identified via LC-MS/MS in the EVs of PC3 (C) and PNT2 (D) cells, respectively, comparing control conditions to the 4 h LMF treatment.
Figure 3. Proteomic analysis of EV cargoes from 4 h LMF-treated (and control untreated) PC3 and PNT2 cells. (A) EV protein content, as assessed by SDS-PAGE and silver staining, from PC3-derived EVs, showing control untreated (CTR) and LMF-treated cell-derived EVs. (B) Protein content from PNT2-derived EVs, showing control untreated (CTR) and LMF-treated cell-derived EVs. (C,D) Venn diagrams showing shared and specific proteins identified via LC-MS/MS in the EVs of PC3 (C) and PNT2 (D) cells, respectively, comparing control conditions to the 4 h LMF treatment.
Biology 13 00734 g003
Figure 4. Protein–protein interaction network analysis for PC3-derived EV protein cargoes. (A) Protein interaction networks are shown for EV protein cargoes from PC3 control and PC3 LMF-treated cells, respectively. (B) The Venn diagram summarises numbers of shared and group-specific functional enrichment pathways associated with the EV proteomes from control and LMF-treated PC3 cells. For a full list of pathways, see Table 2.
Figure 4. Protein–protein interaction network analysis for PC3-derived EV protein cargoes. (A) Protein interaction networks are shown for EV protein cargoes from PC3 control and PC3 LMF-treated cells, respectively. (B) The Venn diagram summarises numbers of shared and group-specific functional enrichment pathways associated with the EV proteomes from control and LMF-treated PC3 cells. For a full list of pathways, see Table 2.
Biology 13 00734 g004
Figure 5. Protein–protein interaction network analysis for PNT2-derived EV protein cargoes. (A) Protein–protein interaction networks are shown for EV protein cargoes from PNT2 control untreated and PNT2 LMF-treated cells, respectively. (B) The Venn diagram summarises numbers of shared and group-specific functional enrichment analysis pathways associated with the EV proteomes from control untreated and LMF-treated PNT2 cells. For a full list of pathways, see Table 3.
Figure 5. Protein–protein interaction network analysis for PNT2-derived EV protein cargoes. (A) Protein–protein interaction networks are shown for EV protein cargoes from PNT2 control untreated and PNT2 LMF-treated cells, respectively. (B) The Venn diagram summarises numbers of shared and group-specific functional enrichment analysis pathways associated with the EV proteomes from control untreated and LMF-treated PNT2 cells. For a full list of pathways, see Table 3.
Biology 13 00734 g005
Figure 6. Summary for the functional protein enrichment analysis of protein cargoes identified in EVs derived from (A) PC3 and (B) PNT2 cells, following 4 h LMF exposure (in red), compared with controls (in grey).
Figure 6. Summary for the functional protein enrichment analysis of protein cargoes identified in EVs derived from (A) PC3 and (B) PNT2 cells, following 4 h LMF exposure (in red), compared with controls (in grey).
Biology 13 00734 g006
Figure 7. Comparative reverse transcription–quantitative polymerase chain reaction (RT-qPCR) analysis of expression levels of miR-155, miR-210, miR-21, miR-200c, and miR-126. (A) Relative expressions of oncogenic miRs: miR-155, miR-21, miR-210 in PNT2 and PC3 cells following 4 h LMF exposure. (B) Relative expression levels of tumour suppressor miRs: miR-200c and miR-126 in PNT2 and PC3 cells following 4 h LMF exposure. The column graphs represent the average of three replicates of RNA isolated from each sample. Data were normalised according to RNU6 expression by fold analysis (n = 3, p < 0.05 for all). Exact p-values are indicated (* p ≤ 0.05; ** p ≤ 0.01; **** p ≤ 0.0001); error bars indicate standard deviation (SD).
Figure 7. Comparative reverse transcription–quantitative polymerase chain reaction (RT-qPCR) analysis of expression levels of miR-155, miR-210, miR-21, miR-200c, and miR-126. (A) Relative expressions of oncogenic miRs: miR-155, miR-21, miR-210 in PNT2 and PC3 cells following 4 h LMF exposure. (B) Relative expression levels of tumour suppressor miRs: miR-200c and miR-126 in PNT2 and PC3 cells following 4 h LMF exposure. The column graphs represent the average of three replicates of RNA isolated from each sample. Data were normalised according to RNU6 expression by fold analysis (n = 3, p < 0.05 for all). Exact p-values are indicated (* p ≤ 0.05; ** p ≤ 0.01; **** p ≤ 0.0001); error bars indicate standard deviation (SD).
Biology 13 00734 g007
Figure 8. Comparative reverse transcription–quantitative polymerase chain reaction (RT-qPCR) analysis of expression levels of MMP2 and MMP9. (A) Relative expressions of MMP2 in PNT2 and PC3 cells following the LMF exposures. (B) Relative expression level of MMP9 in PNT2 and PC3 cells following the LMF exposures. The column graphs represent the average of three replicates of RNA isolated from each sample. Data normalised according to RNU6 expression by fold analysis. (n = 3, p < 0.05 for all). Exact p-values are indicated (**** p ≤ 0.0001); error bars indicate standard deviation (SD).
Figure 8. Comparative reverse transcription–quantitative polymerase chain reaction (RT-qPCR) analysis of expression levels of MMP2 and MMP9. (A) Relative expressions of MMP2 in PNT2 and PC3 cells following the LMF exposures. (B) Relative expression level of MMP9 in PNT2 and PC3 cells following the LMF exposures. The column graphs represent the average of three replicates of RNA isolated from each sample. Data normalised according to RNU6 expression by fold analysis. (n = 3, p < 0.05 for all). Exact p-values are indicated (**** p ≤ 0.0001); error bars indicate standard deviation (SD).
Biology 13 00734 g008
Figure 9. LMF exposure (4 h) induced cellular invasion of PC3 cells but did not affect cell proliferation. (A) PC3 cells were plated on Matrigel-coated transwell filters, and the extent of invasion was determined following a 4 h LMF exposure and compared to the control assay. The results are plotted as invasion (%), which is the percentage of invaded cells compared to the total number of cells seeded (n = 3; **** p ≤ 0.0001). (B) The total cell number/proliferation did not change during the experiment (n = 3; ns); error bars indicate SD. Scale bars indicate 650 μm for all images that are representative of triplicates.
Figure 9. LMF exposure (4 h) induced cellular invasion of PC3 cells but did not affect cell proliferation. (A) PC3 cells were plated on Matrigel-coated transwell filters, and the extent of invasion was determined following a 4 h LMF exposure and compared to the control assay. The results are plotted as invasion (%), which is the percentage of invaded cells compared to the total number of cells seeded (n = 3; **** p ≤ 0.0001). (B) The total cell number/proliferation did not change during the experiment (n = 3; ns); error bars indicate SD. Scale bars indicate 650 μm for all images that are representative of triplicates.
Biology 13 00734 g009
Table 1. Protein hits identified in EVs isolated from PC3 cells, from control untreated (ctrl) and 4 h LMF exposure groups, respectively. A tick (V) indicates that the protein hit was present in the EV proteome. Protein IDs and names are shown, and additionally, gene names are included for some hits as indicated in italics.
Table 1. Protein hits identified in EVs isolated from PC3 cells, from control untreated (ctrl) and 4 h LMF exposure groups, respectively. A tick (V) indicates that the protein hit was present in the EV proteome. Protein IDs and names are shown, and additionally, gene names are included for some hits as indicated in italics.
Protein IDProtein NamePC3
ctrl
PC3
LMF
PNT2
ctrl
PNT2
LMF
H6VRG0
KRT1
Keratin, type II cytoskeletal 1 V
H6VRG2
KRT1
Keratin, type II cytoskeletal 1 VVV
P02533Keratin, type I cytoskeletal 14VVVV
P08779Keratin, type I cytoskeletal 16 VVV
P13647Keratin, type II cytoskeletal 5 VVV
B4E1T1
KRT5
cDNA FLJ54081, highly similar
to Keratin, type II cytoskeletal 5
VV
P04259Keratin, type II cytoskeletal 6B V V
A0A0S2Z428HCG2039812, KRT6A VVV
A0A804GS07Actin, cytoplasmic 2 VV
Q6GMX6
IGH@
IGH@ proteinVV
P0DOX5Immunoglobulin gamma-1 heavy chain VV
A0A384NYT8
TUBB
Tubulin beta chainVV
P05787-2
KRT8
Isoform 2 of Keratin, type II cytoskeletal 8VVV
A0A0K0K1H8
HEL-S-71p
SerotransferrinVVVV
A0A5E4Uncharacterised protein V
A0A024R5Z7Annexin 2VVV
A0A4D5RA86
ANXA1
Annexin 1 V
A0A087WVQ9Elongation factor 1-alpha VV
A0A087WV01Elongation factor 1-alphaV
A0A5C2GAZ2IGH + IGL
c262_heavy_IGHV3-15_IGHD4-17_IGHJ4
VV
Q2TSD0
V9HVZ4
GAPDH
Glyceraldehyde-3-phosphate dehydrogenaseVVV
A0A0B4J1Y9Immunoglobulin heavy variable 3–72 V
B3KPS3
TUBA1C
Tubulin alpha chainVV
A0A2R3Z0D6
HBB
Haemoglobin subunit beta V
Q0VAS5
HIST1H4H
Histone H4VV
A0A7S5BYV3IGH c429_heavy_IGHV1-24_IGHD1-7_IGHJ4 V
E2DRY6
ENO1
Phosphopyruvate hydratase V
A0A2U8J951
IgH
Ig heavy chain variable region V
B2R6J2
VIL2
cDNA, FLJ92973, highly similar to Homo sapiens villin 2 (ezrin) (VIL2), mRNA V
Q0KKI6Immunoglobulin light chain (Fragment)VV
A0A286YES1
IGHG3
Immunoglobulin heavy constant gamma 3 V
P0C0S5Histone H2A.Z VV
H0Y8D1Serine protease 1 VVV
A0A0M4FNU3
ALDOA
Fructose-bisphosphate aldolase V
A0A0K2BMD8
HBA2
Mutant haemoglobin alpha 2 globin chainVV V
A0A024R5Z9Pyruvate kinaseVVV
A0A0S2Z4D4Proteolipid protein 1 isoform 1 V
A0A024R210
IFITM1
Interferon-induced transmembrane protein 1 (9–27) V
A0A2R8Y619Histone H2B type 2-K1 V
P81605-2
DCD
Isoform 2 of Dermcidin VV
A0A385KNS5
CD44
CD44 antigen V
Q2QD09
TPI1
Triosephosphate isomerase V
Q2VPJ6
HSP90AA1
HSP90AA1 protein V
Q14D04-2
VEPH1
Isoform 2 of Ventricular
zone-expressed PH domain-containing protein homolog 1
V
A0A024R1K7Tyrosine 3-monooxygenase/tryptophan 5-monooxygenase activation protein, eta polypeptide V
H6VRF8
KRT1
Keratin, type II cytoskeletal 1V VV
A0A2R8Y793Actin, cytoplasmic 1V
P0C0S8Histone H2A type 1V
A0A024R4F1Phosphopyruvate hydrataseV V
P0DOY2Immunoglobulin lambda constant 2V
A0A481SHK9
HBB
Haemoglobin subunit betaV
A0A024RCJ2Histone H2BV
A0A024RA28
HNRNPA2B1
Heterogeneous nuclear ribonucleoprotein A2/B1V
Q9BS19Epididymis secretory sperm binding proteinV
A0A0A0MRQ5Peroxiredoxin-1V
A0A286YFJ8Immunoglobulin heavy constant gamma 4V
A0A5C2GPU9IG c1228_heavy_IGHV3-33_IGHD1-1_IGHJ4V
Q9Y5H4-2
PCDHGA1
Isoform 2 of Protocadherin gamma-A1V
A0A2R8Y5P0RadixinV
P48668Keratin, type II cytoskeletal 6C, KRT6C VV
P15924Desmoplakin V
B4DKV4
KRT6B
cDNA FLJ60647, highly similar to Keratin, type II cytoskeletal 6B V
Q04695Keratin, type I cytoskeletal 17, KRT17 V
Q86YZ3Hornerin V
Q02413Desmoglein-1 V
P12035Keratin, type II cytoskeletal 3 V
A0A0C4DGB6Albumin V
Q0IIN1
KRT77
Keratin 77 V
A0A024R952Plakophilin 1 (Ectodermal dysplasia/skin fragility syndrome) V
Q86Y46Keratin, type II cytoskeletal 73 V
Q8N1N4Keratin, type II cytoskeletal 78 V
A0A494C0J7TGc domain-containing protein V
A0A1U9X8X5
CDSN
Corneodesmosin V
Q9HB00Desmocollin 1 V
Q5D862Filaggrin-2, FLG2 V
Q5K634SCCA2/SCCA1 fusion protein isoform 1 V
P05089-2
ARG1
Isoform 2 of Arginase-1, ARG1 V
Q3SYB5
SERPINB12
Serpin B12 V
P47929Galectin-7, LGALS7B V
A0A087WYS6Proteasome (Prosome, macropain) subunit, alpha type, 8 V
A0A384P5Q0Catalase V
E7DVW5Fatty acid binding protein 5
(Psoriasis-associated)
V
P05109Protein S100-A8 V
A0JNT2
KRT83
KRT83 V
B4DF70
PRDX2
cDNA FLJ60461, highly similar to Peroxiredoxin-2 V
Q6KB66-3
KRT80
Isoform 3 of Keratin, type II
cytoskeletal 80, KRT80
V
P42357-2
HAL
Isoform 2 of Histidine ammonia-lyase V
A0A024RC29
DSC3
Desmocollin 3 V
B0AZM8
TGM1
cDNA, FLJ79468, highly similar to Protein-glutamine gamma-glutamyltransferase K V
A0A2R8YD45Tripeptidyl-peptidase 1, TPP1 V
Q9NZT1Calmodulin-like protein 5 V
A0A7P0TAI0
HSPA5
78 kDa glucose-regulated protein V
A0A0B4J259Lysozyme C V
J3KSD8Bleomycin hydrolase (Fragment) V
A0A2R8Y5E5Glutathione S-transferase P, GSTP1 V
A0A248RGE3
RPS27A
Ubiquitin-40S ribosomal protein S27a (Fragment) V
A0A024RD80Heat shock protein 90kDa alpha (Cytosolic), class B member 1, HSP90AB1 V
A0A024R8D7Lipocalin 1 (Tear prealbumin), isoform CRA_a V
A0A087WVQ6Clathrin heavy chain, CLTC V
A0A0K2BMD8
HBA2
Mutant haemoglobin alpha 2 globin chain V
A0A087WUB9Beta-catenin-like protein 1, CTNNBL1 V
A0A0S2Z3L4
CTSD
Cathepsin D isoform 2 (Fragment), CTSD V
Q5T750Skin-specific protein 32 V
A0A0U1RQT9Synaptophysin-like protein 1 (Fragment), SYPL1 V
Q9Y3R4Sialidase-2, NEU2 V
B4DGC3
APOD
Apolipoprotein D V
F5GX11Proteasome subunit alpha
type-1, PSMA1
V
A0A024RAM2Glutaredoxin (Thioltransferase)
P0DOX8
IGL1
Immunoglobulin lambda-1 light
Chain, IGL1
V
B4DTN4N6-adenosine-methyltransferase
catalytic subunit, METTL3
V
A0A140VK00Testicular tissue
protein Li 227
V
A0A1B4WRL5
HBB
Beta globin (Fragment) V
B3VL17Beta globin (Fragment) V
A0A1B0GVI3
KRT10
Keratin, type I cytoskeletal 10 KRT10 VV
P02790Hemopexin VV
B4DE59
A0A024R1X8
JUP
Junction plakoglobin VV
A1A508
PRSS3
PRSS3 protein VV
Q86W19
PRSS1
Protease serine 1 (Fragment), PRSS1 V
A0A024RAY2Keratin 18 V
B2R4M6
S100A9
Protein S100 VV
Table 2. Functional enrichment pathway analysis for EV protein cargoes derived from PC3 cell, control untreated, and LMF-treated groups, respectively. A tick (V) indicates that the pathway was present for the EV proteome.
Table 2. Functional enrichment pathway analysis for EV protein cargoes derived from PC3 cell, control untreated, and LMF-treated groups, respectively. A tick (V) indicates that the pathway was present for the EV proteome.
STRING Cluster PathwaysPC3 ctrlPC3 LMF
Pachyonychia congenita and Epidermolysis bullosa simplex Dowling–Meara type V
Formation of the cornified envelope and Serpin, conserved site V
Phosphoglycerate kinase and Aerobic glycolysis V
Glycolysis and sugar phosphatase activity V
Carbon metabolism and Pyruvate metabolism V
KEGG PathwaysPC3 ctrlPC3 LMF
African trypanosomiasisVV
Glycolysis/GluconeogenesisVV
MalariaV
Systemic lupus erythematosusVV
Biosynthesis of amino acidsVV
Viral myocarditisV
HIF-1 signalling pathwayVV
PhagosomeV
Pathogenic E.coli infectionVV
Carbon metabolismVV
AlcoholismV
Tight junctionV
Viral carcinogenesisVV
Salmonella infectionV
Amyotrophic lateral sclerosisV
S. aureus infection V
Biological Process GOPC3 ctrlPC3 LMF
Hydrogen peroxidase catabolic processV
Retina homeostasisV
Nitric oxide transport V
Positive regulation of plasminogen activation V
Positive regulation of vesicle function V
Canonical glycolysis V
Glycolytic process V
Killing of host by symbiont cells V
Intermediate filament organisation V
Keratinisation V
Keratinocyte differentiation V
Complement activation V
Glucose metabolic process V
Hexose metabolic process V
Antimicrobial humoral response V
Humoral immune response V
Epidermis development V
Supramolecular fibre organisation V
Defence response to bacterium V
Epithelial cell differentiation V
Monocarboxylic acid metabolic process V
Cytoskeleton organisation V
Defence response to other organism V
Innate immune response V
Epithelium development V
Response to other organism V
Defence response V
Immune response V
Immune system process V
Response to external stimulus V
Organelle organisation V
Response to stress V
Cellular component organisation V
Wiki Pathways
Aerobic glycolysisVV
Glycolysis and gluconeogenesisVV
Glycolysis in senescenceVV
Metabolic reprogramming in colon cancerVV
Pathogenic Escherichia coli infectionVV
Clear cell renal cell carcinoma pathwaysVV
Cori cycle V
Sudden infant death syndrome (SIDS) susceptibility pathways V
VEGFA-VEGFR2 signalling V
HIF1A and PPARG regulation of glycolysis V
Corticotropin-releasing hormone signalling pathway V
Molecular Function GOPC3 ctrlPC3 LMF
Structural constituent of cytoskeletonVV
Cadherin bindingVV
Structural molecule activityVV
Protein bindingVV
Phospholipidase A2 inhibitor activity V
Structural constituent of skin epidermis V
Disordered domain-specific binding V
Protein dimerization activity V
Cellular Component GOPC3 ctrlPC3 LMF
Haptoglobin–haemoglobin complexVV
Haemoglobin complexVV
Endocytic vesicle lumenVV
CENP-A containing nucleosomeV
Blood microparticleV
Ficolin-1-rich granule lumenVV
Nuclear matrixV
NucleosomeV
Keratin filamentVV
Cortical cytoskeletonV
Secretory granule lumenVV
Cell cortexV
Endocytic vesicleV
Chromosomal regionV
Extracellular exosomeVV
Secretory granuleVV
Polymeric cytoskeletal fibreVV
Supramolecular fibreVV
Extracellular spaceVV
VesicleVV
Cytoplasmic vesicleVV
CytoskeletonVV
Intracellular non-membrane-bounded organelleV
CytosolVV
Protein containing complexVV
Extrinsic component of external side of plasma membrane V
Immunoglobulin complex, circulating V
M band V
Cornified envelope V
Myelin sheath V
Blood microparticle V
Intermediate filamentVV
Basal plasma membrane V
Basolateral plasma membrane V
Extrinsic component of membrane V
Collagen-containing extracellular matrix V
Apical plasma membrane V
Side of membrane V
Cell surface V
Intracellular non-membrane bound organelle V
Disease–Gene AssociationsPC3 ctrlPC3 LMF
AmyloidosisVV
Cutaneous T-cell lymphomaV
non-Hodgkin lymphomaVV
Primary cutaneous amyloidosisV
Familial visceral amyloidosisVV
Skin carcinomaV
Borst–Jadassohn intraepidermal carcinomaVV
Hematopoietic system diseaseVV
CarcinomaVV
Seborrheic keratosisVV
Mycosis fungoidesVV
Alpha thalassemiaV
Blood protein diseaseV
Organ system cancerV
Hepatocellular carcinomaV
Inherited metabolic disorder V
Alpha thalassemia V
Keratosis V
Palmoplantar keratosis V
Pachyonychia congenita V
Nonepidermolytic palmoplantar keratoderma V
Congenital haemolytic anaemia V
Primary cutaneous amyloidosis V
Autosomal dominant disease V
Epidermolysis bullosa simplex Dowling–Meara type V
Epidermolysis bullosa simplex with mottled pigmentation V
Focal nonepidermolytic palmoplantar keratoderma V
Basal cell carcinoma V
Skin disease V
Steatocystoma multiplex V
Blood protein disease V
Reactome PathwaysPC3 ctrlPC3 LMF
Erythrocytes take up oxygen and release carbon dioxideVV
Scavenging of heme from plasmaVV
Erythrocytes take up carbon dioxide and release oxygenVV
Chaperone mediated AutophagyVV
Prefoldin-mediated transfer of substrate to CCT/TricV
RHO GTPases activate IQGAPsV
Recyclin pathway of L1V
RNA polymerase I promoter openingV
GluconeogenesisVV
Packaging of telomere endsV
DNA methylationV
Activated PKN1 stimulates transcription of androgen receptorV
Gene and protein expression by JAK-STAT signallingV
SIRT1 negatively regulates rRNA expressionV
Cleavage of damaged purineV
Recognition and association of DNA glycosylase with site containing an affected purineV
B-WICH complex positively regulates rRNA expressionV
HDACs deacetylate histonesV
Assembly of the ORC complex at the origin of replicationV
Diseases of programmed cell deathV
GlycolysisVV
HCMV early eventsV
HATs acetylate histonesV
HCMV late eventsV
Formation of the cornified envelopeVV
AutophagyVV
Factors involved in megakaryocyte development and plateletsV
RHO GTPase effectorsV
Neutrophil degranulationVV
M phaseVV
Cellular responses to stressVV
Vesicle-mediated transportV
Infectious diseaseV
HemostasisV
Developmental biologyVV
Innate immune systemVV
DiseaseV
Post-translational protein modificationV
Immune systemV
Type I hemidesmosome assembly V
HSF1 activation V
Binding and uptake in ligands by scavenger receptors V
RHO-GTPases activate PNKs V
Metabolism of carbohydrates V
Immune system V
Human PhenotypePC3 ctrlPC3 LMF
Palmoplantar blistering V
Blistering by anatomical locationV
OnychogryphosisV
Lower limb painV
Nail dystrophyV
HyperhidrosisV
Palmoplantar keratodermaV
Hoarse voiceV
Nail dysplasiaV
Steatocystoma multiplexV
Eruptive vellus hair cystV
Abnormal fingernail morphologyV
Linear arrays of macular hyperkeratosis in flexural areasV
Hypohidrosis or hyperhidrosisV
Neoplasm of the skinV
Onychogryphosis of toenailsV
Abnormality of the digestive systemV
Angular cheilitisV
Hyperplastic callus formationV
Onychogryphosis of fingernailV
ParonychiaV
Abnormality of skin morphologyV
PainV
Abnormality of temperature regulationV
Palmoplantar hyperhidrosisV
Constitutional symptomV
Epidermoid cystV
Abdominal symptomV
Neoplasm by anatomical siteV
PainV
Ear painV
Thickened skinV
Sign or symptomV
Fingernail dysplasiaV
Oral leukoplakiaV
Follicular hyperkeratosisV
White lesion of the oral mucosaV
JaundiceV
Mottled pigmentation of the trunk and proximal extremitiesV
Discrete 2 to 5 mm hyper- and hypopigmented maculesV
AlopeciaV
Abnormality of digestive system physiologyV
Focal friction-related palmoplantar hyperkeratosisV
Generalised reticulate brown pigmentationV
Localised skin lesionV
Natal toothV
Pain in head and neck regionV
CholestasisV
Punctate palmoplantar hyperkeratosisV
Skin fragility with non-scarring blisteringV
Smooth tongueV
Foot painV
Genital blisteringV
Hyperkeratotic papuleV
FeverV
Acute episodes of neuropathic symptomsV
Reticulated skin pigmentationV
Depigmentation/hyperpigmentation of skinV
Aplasia cutis congenita on trunk or limbsV
Hypomelanotic maculeV
Generalised abnormality of skinV
Abnormality of the skeletal systemV
Chronic haemolytic anaemiaV
Erythematous papuleV
Dermatological manifestations of systemic disordersV
Upper limb painV
CholelithiasisV
Spotty hyperpigmentationV
Spotty hypopigmentationV
Diffuse palmoplantar hyperkeratosisV
Erosion of oral mucosaV
Abnormal circulating protein concentrationV
Lower limb amyotrophyV
Abnormal hair quantityV
Nonspherocytic haemolytic anaemiaV
Lamina lucida cleavageV
Abnormality of blood and blood-forming tissuesV
Feeding difficultiesV
Anaemia of inadequate productionV
Abnormal oral mucosa morphologyV
AnaemiaV
Abnormal skeletal morphologyV
Oral mucosal blistersV
Decreased body weightV
Female reproductive system diseaseV
Cutaneous photosensitivityV
Abnormal hair morphologyV
Abnormality of the respiratory systemV
Congestive heart failureV
Absent toenailV
Abnormality of the immune systemV
CholecystitisV
Abnormality of the musculoskeletal systemV
Ovarian endometrioid carcinomaV
Normocytic anaemiaV
Table 3. Functional enrichment pathway analysis for EV protein cargoes derived from PNT2 cells, control untreated, and LMF-treated, respectively. A tick (V) indicates that the pathway was present for the EV proteome.
Table 3. Functional enrichment pathway analysis for EV protein cargoes derived from PNT2 cells, control untreated, and LMF-treated, respectively. A tick (V) indicates that the pathway was present for the EV proteome.
STRING Cluster PathwaysPNT2 ctrlPNT2 LMF
Formation of the cornified envelope and Autosomal recessive congenital ichthyosisV
Formation of the cornified envelope and Serpin, conserved siteVV
Desmosome and Ichthyosis vulgarisVV
Keratinisation and Cornified envelopeV
Mixed, incl. Pachyonychia congenita and Epidermolysis bullosa simplex Dowling–Meara typeV
Pachyonychia congenita and Epidermolysis bullosa simplex Dowling–Meara typeVV
Mixed, incl. Pachyonychia congenita and Netherton syndromeV
Mixed, incl. Ichthyosis vulgaris and Bullous congenital ichthyosiform erythrodermaV
Pachyonychia congenita and Epidermolysis bullosa simplex Dowling–Meara typeVV
Mixed, incl. S100/CaBP-9k-type, calcium binding, subdomain, and Cystatin superfamilyV
Naxos disease and Subcorneal pustular dermatosisV
Ichthyosis vulgaris and Epidermolytic acanthomaVV
S100/CaBP-9k-type, calcium binding, subdomain, and AnnexinV
S-100/ICaBP-type calcium binding domainV
Keratin filament and Keratin, type IV
Detoxification of ROS and mRNA, protein, and metabolite induction pathway by cyclosporin AV
Mixed, incl. COVID-19, thrombosis and anticoagulation, and Scavenging of heme from plasmaV
Mixed, incl. Glutathione metabolism and Detoxification of Reactive Oxygen SpeciesV
Alcoholic pancreatitis and TyphusVV
Cell adhesive protein binding involved in bundle of His cell-Purkinje myocyte communicationV
KEGG Pathways:PNT2 ctrlPNT LMF
Oestrogen signalling pathwayVV
Staphylococcus aureus infectionVV
Biosynthesis of amino acidsV
Biological Process GOPNT2 ctrlPNT LMF
Intermediate filament organisationVV
Intermediate filament cytoskeleton organisation V
Epidermis developmentVV
Keratinocyte differentiationVV
KeratinisationVV
Skin developmentVV
Epithelial cell differentiationVV
Supramolecular fibre organisationVV
Epithelium developmentV
Tissue developmentV
Cellular oxidant detoxificationV
Response to toxic substanceV
Multicellular organismal processV
Anatomical structure developmentVV
Cell differentiationV
Developmental processV
Cytoskeleton organisationVV
Animal organ developmentV
Peptide cross-linkingV
Retina homeostasisV
Humoral immune responseVV
Immune response V
Cell–cell adhesionV
Multicellular organismal homeostasisV
Antimicrobial humoral responseVV
Response to biotic stimulusV
Cellular processV
Response to reactive oxygen speciesV
Peptidyl-cysteine S-nitrosylationV
Hydrogen peroxide catabolic processV
Catabolic processV
Biological process involved in interspecies interaction between organismsV
Response to other organismV
Establishment of skin barrierV
Cell adhesionV
Defence response to other organismVV
Immune system processV
Cellular catabolic processV
Response to external stimulusVV
Tissue homeostasisV
Homeostatic processV
AgeingV
Response to oxidative stressV
Glucose metabolic processV
Peptidyl-cysteine S-trans-nitrosylationV
Neutrophil aggregationV
Defence response to fungusV
Cell envelope organisationV
Defence responseV
Defence response to bacteriumV
Response to bacteriumV
Sequestering of zinc ionV
Wiki PathwaysPNT2 ctrlPNT LMF
Aerobic glycolysisV
Glycolysis in senescenceV
Network map of SARS-CoV-2 signalling pathwayV
Corticotropin-releasing hormone signalling pathway V
Molecular Function GOPNT2 ctrlPNT LMF
Structural constituent of skin epidermisVV
Structural molecule activityVV
Antioxidant activityV
Fatty acid bindingV
Structural constituent of cytoskeletonVV
Calcium ion bindingV
Cellular Component GOPNT2 ctrlPNT LMF
Extracellular spaceVV
Extracellular exosomeVV
Extracellular regionV
VesicleVV
Cornified envelopeVV
Intermediate filament cytoskeletonV
Intermediate filamentVV
Keratin filamentVV
Secretory granuleVV
ficolin-1-rich granuleV
Secretory granule lumenV
Polymeric cytoskeletal fiberV
Cytoplasmic vesicleV
DesmosomeV
Supramolecular fiberV
ficolin-1-rich granule lumenV
CytosolVV
Tertiary granuleV
CytoskeletonVV
CytoplasmV
MelanosomeV
Collagen-containing extracellular matrixVV
Blood microparticleVV
Tertiary granule lumenV
ficolin-1-rich granule membraneV
Vacuolar lumenV
Endomembrane systemV
Azurophil granule lumenV
OrganelleV
LysosomeV
Specific granule lumenV
Endocytic vesicle lumenVV
Keratohyalin granuleV
Membrane-bounded organelleV
Intracellular organelleV
Intracellular non-membrane-bounded organelleVV
Intracellular anatomical structureV
Cell peripheryV
Cell–cell junctionV
Proteasome core complex, alpha-subunit complexV
Fascia adherensV
Secretory granule membraneV
Haptoglobin–haemoglobin complexV
Cytoplasmic vesicle membraneV
Haemoglobin complexV
Bounding membrane of organelleV
Endocytic vesicleV
Disease–Gene AssociationsPNT2 ctrlPNT LMF
KeratosisVV
Palmoplantar keratosisVV
Integumentary system diseaseV
Skin diseaseVV
AmyloidosisV
Pachyonychia congenitaVV
Familial visceral amyloidosisV
Nonepidermolytic palmoplantar keratodermaVV
Autosomal dominant diseaseVV
Bullous skin diseaseVV
DermatitisV
AcanthomaVV
PemphigusV
Steatocystoma multiplexVV
Borst–Jadassohn intraepidermal carcinomaVV
Subcorneal pustular dermatosisV
CarcinomaVV
Epidermolysis bullosaV
Seborrheic keratosisVV
Mycosis fungoidesVV
Basal cell carcinomaVV
Skin cancerV
Hair diseaseV
IchthyosisV
Inherited metabolic disorderV
Autosomal genetic diseaseVV
DiseaseV
Genetic diseaseV
Monogenic diseaseV
Arrhythmogenic right ventricular cardiomyopathyV
Primary cutaneous amyloidosisVV
Epidermolysis bullosa simplex Dowling–Meara typeVV
Epidermolysis bullosa simplex with mottled pigmentationVV
Focal nonepidermolytic palmoplantar keratodermaVV
Epidermolytic acanthomaVV
Bullous congenital ichthyosiform erythrodermaVV
Disease of anatomical entityVV
Immune system diseaseV
Epidermolytic hyperkeratosisVV
Stomach cancerV
Naxos diseaseV
Autoimmune disease of skin and connective tissueV
Hematopoietic system disease V
Reactome PathwaysPNT2 ctrlPNT LMF
Formation of the cornified envelopeVV
Neutrophil degranulationV
Developmental BiologyV
Innate Immune SystemV
Immune SystemV
Cellular response to chemical stressV
Scavenging of heme from plasmaVV
Cellular responses to stressV
Chaperone-Mediated AutophagyV
Apoptotic cleavage of cell adhesion proteinsV
Antimicrobial peptidesV
ER-Phagosome pathwayV
ApoptosisV
Transport of small moleculesV
The role of GTSE1 in G2/M progression after G2 checkpointV
Metal sequestration by antimicrobial proteinsV
Detoxification of Reactive Oxygen SpeciesV
PCP/CE pathwayV
Class I MHC-mediated antigen processing and presentationV
RUNX1 regulates transcription of genes involved in differentiation of HSCsV
Erythrocytes take up oxygen and release carbon dioxideV
Transport of fatty acidsV
Type I hemidesmosome assembly V
Cell junction organisation V
Uptake of dietary cobalamins into enterocytes V
Human Phenotype (Monarch)PNT2 ctrlPNT LMF
Abnormal blistering of the skinVV
Palmoplantar keratodermaVV
HyperkeratosisV
Nail dystrophyVV
Palmoplantar blisteringVV
Abnormal epidermal morphologyV
Epidermal thickeningV
Blistering by anatomical locationVV
Epidermal acanthosisVV
Follicular hyperkeratosisVV
Abnormality of the nailV
Hypohidrosis or hyperhidrosisVV
AlopeciaVV
Nail dysplasiaVV
CheilitisV
Angular cheilitisVV
HyperhidrosisVV
OnychogryphosisVV
Abnormal hair quantityVV
ErythemaVV
Lower limb painVV
Scaling skinVV
Abnormality of skin physiologyV
Natal toothVV
Steatocystoma multiplexVV
Eruptive vellus hair cystVV
Linear arrays of macular hyperkeratosis in flexural areasVV
Neoplasm of the skinVV
Abnormality of the skinV
Onychogryphosis of toenailsVV
Abnormality of skin morphologyVV
Hyperplastic callus formationVV
Recurrent skin infectionsVV
PruritusV
Onychogryphosis of fingernailVV
Abnormal fingernail morphologyVV
Inflammatory abnormality of the skinVV
Abnormal oral mucosa morphologyVV
ParonychiaVV
Fragile skinVV
Palmoplantar hyperhidrosisVV
Epidermoid cystVV
Hoarse voiceVV
Absent toenailVV
Ear painVV
Fingernail dysplasiaVV
Abnormality of the respiratory systemVV
Abnormality of immune system physiologyVV
Abnormality of the dentitionVV
Sparse hairV
ErythrodermaV
Oral leukoplakiaVV
White lesion of the oral mucosaVV
Skin erosionVV
Increased inflammatory responseV
Decreased body weightVV
Localised skin lesionVV
AcantholysisV
ParakeratosisV
Constitutional symptomVV
Aplasia cutis congenitaVV
Abnormality of the immune systemV
Generalised abnormality of skinVV
Cutaneous photosensitivityVV
Pain in head and neck regionVV
Neoplasm by anatomical siteVV
Alopecia universalisV
OrthokeratosisV
Abnormality of the handV
Abnormality of the lower limbV
SepsisV
OnycholysisV
Failure to thriveV
Abnormality of the faceVV
Sign or symptomV
PainVV
Abnormality of nail colourV
Recurrent infectionsV
Abnormality of metabolism/homeostasisV
Growth abnormalityVV
Mottled pigmentation of the trunk and proximal extremitiesVV
Discrete 2 to 5 mm hyper- and hypopigmented maculesVV
Palmoplantar scaling skinV
Abnormal circulating transferrin concentrationV
Unusual infectionV
Abnormality of limbsV
Abnormal oral cavity morphologyV
Impaired myocardial contractilityV
Congenital bullous ichthyosiform erythrodermaVV
Focal friction-related palmoplantar hyperkeratosisVV
Localised epidermolytic hyperkeratosisV
Generalised reticulate brown pigmentationVV
Abnormal immunoglobulin levelV
Congenital ichthyosiform erythrodermaV
PainVV
Congenital alopecia totalisV
Punctate palmoplantar hyperkeratosisVV
Skin fragility with non-scarring blisteringVV
Widely spaced toesV
Smooth tongueVV
4–5 finger syndactylyV
Abdominal symptomVV
Foot painVV
Genital blisteringVV
Chapped lipV
Hyperkeratotic papuleVV
Phenotypic abnormalityVV
Skin plaqueVV
Acute episodes of neuropathic symptomsVV
Reticulated skin pigmentationVV
Depigmentation/hyperpigmentation of skinV
Aplasia cutis congenita on trunk or limbsVV
Hypomelanotic maculeVV
Tapered distal phalanges of fingerV
Hypovolemic shockV
Abnormal circulating metabolite concentrationV
Abnormal circulating protein concentrationVV
Aplasia/Hypoplasia of the eyebrowV
Abnormality of hair textureV
3–4 finger syndactylyV
Erythematous papuleVV
Obsolete Bilateral external ear deformityV
Abnormal cellular phenotypeV
HypernatremiaV
Patchy palmoplantar hyperkeratosisV
Upper limb painVV
Abnormality of digestive system physiologyVV
Sparse scalp hairV
Spotty hyperpigmentationVV
Spotty hypopigmentationV
Diffuse palmoplantar hyperkeratosisVV
Right ventricular cardiomyopathyV
Erosion of oral mucosaVV
Increased neuronal autofluorescent lipopigmentV
Lamina lucida cleavageVV
Mitten deformityV
Autoamputation of digitsV
Abnormal dermoepidermal hemidesmosome morphologyV
Abnormality of the digestive system V
Abnormality of the skeletal system V
Abnormality of the musculoskeletal system V
Abnormal musculoskeletal physiology V
Conjunctival hamartoma V
Abnormal skeletal morphology V
Oral mucosal blisters V
Heat intolerance V
Squamous cell carcinoma of the skin V
Abnormal conjunctiva morphology V
Bronchomalacia V
Milia V
Abnormal epiglottis morphology V
Abnormality of temperature regulation V
Ridged nail V
Dystrophic toenail V
Distal lower limb amyotrophy V
Atrophic scars V
Respiratory distress V
Poor appetite V
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

Lange, S.; Inal, J.M.; Kraev, I.; Dart, D.A.; Uysal-Onganer, P. Low Magnetic Field Exposure Alters Prostate Cancer Cell Properties. Biology 2024, 13, 734. https://doi.org/10.3390/biology13090734

AMA Style

Lange S, Inal JM, Kraev I, Dart DA, Uysal-Onganer P. Low Magnetic Field Exposure Alters Prostate Cancer Cell Properties. Biology. 2024; 13(9):734. https://doi.org/10.3390/biology13090734

Chicago/Turabian Style

Lange, Sigrun, Jameel M. Inal, Igor Kraev, Dafydd Alwyn Dart, and Pinar Uysal-Onganer. 2024. "Low Magnetic Field Exposure Alters Prostate Cancer Cell Properties" Biology 13, no. 9: 734. https://doi.org/10.3390/biology13090734

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