2. Materials and methods
2.1. Chemicals and reagents
Exosome isolation kit, Revert-Aid first strand cDNA synthesis kit, DNaseI and Power SYBR green master mix were purchased from ThermoFisher Scientific. PKH26 red fluorescent cell linker midi kit, Trizol, poly-L-lysine, H2O2 and citrate buffer were from Sigma-Aldrich. RNeasy Micro Kit and RNA protect tissue reagent were from Qiagen. Anti-CD63 was purchased from Cloud Clone Corp., anti-Alix and anti-flotillin-1, from Abcam. Alexa Fluor 568 goat anti-rabbit secondary antibody was from Invitrogen and HRP-labelled anti-mouse antibody was from Santa Cruz Biotechnology. Vectashield with DAPI was purchased from Vector laboratories. DHE (dihydroethidium) was from Invitrogen Molecular Probes. Xylene was purchased from Fisher Scientific and absolute alcohol was from Merck. Crystal violet, DPX, slides, cover slips and other reagents were of the highest analytical grade and were obtained from the common source.
2.2. Experimental animals
Inbred strains of healthy BALB/c mice (22-25g) of either sex, aged 3-4 months old, were obtained from the animal house facility of Department of Zoology, University of Delhi. Animals were housed in propylene cages, fed with standard pellet diet and water ad libitum at a constant environment (at 18-26°C with 12h light/dark cycles). The study was performed in accordance with the guidelines for the care and use of laboratory animals with approval of the Institutional Animal Ethics Committee, University of Delhi and protocols approved by the CPCSEA, India (Approval number: DU/ZOOL/IAECR/2019/16/E-1/2021).
2.3. Maintenance of Dalton’s lymphoma (DL) in vivo
The DL cells were obtained from the Department of Biotechnology, Banaras Hindu University and maintained in the peritoneum of BALB/c mice by i.p. transplantation as described earlier [
30]. For experiments, growing DL cells were collected from the donor mice and immediately suspended in sterile phosphate buffer saline (PBS, 1X). The viability of cells was confirmed by the trypan blue staining and cell counting was done. The number of cells was adjusted to 1×10
6 cells/ml and i.p. injected in the healthy BALB/c mice.
2.4. Experimental groups
The mice, of either sex, were divided into two groups, with each group consisting of six mice. Group I represented the control group while group II animals were induced for DL development. The tumor was allowed to grow and mice were sacrificed through the cervical dislocation at day 18 post-tumor transplantation.
2.5. Cell culture and treatments
RAW264.7 cells (murine macrophage cell line) was obtained from the National cell repository NCCS, Pune, India. The cells were grown in Dulbecco’s modified eagle medium (DMEM) supplemented with 10% heat-inactivated fetal bovine serum (FBS) in a humidified incubator at 370C with 5% CO2. Cells were sub-cultured upon they reached 80% confluence. All the experiments were performed in sterile and endotoxin free conditions. To investigate the polarization of macrophages by lymphoma-derived exosomes, we included four groups. Group I - control, group II- cells treated with lymphoma-derived exosomes (100µg/ml), group III- lipopolysaccharide (LPS, 2µg/ml)-stimulated cells and group IV- co-treatment of exosomes and LPS.
2.6. Immunofluorescence for CD63
The ascites was collected from the peritoneum of DL mice and centrifuged at 1500g for 10 min at 40C. The pellet of DL cells was washed with PBS three times and smeared on a clean glass slide, air dried and fixed in methanol. For T cells, thymus was dissected out from control mice and single cell suspension was made in PBS. The cells were pellet down and washed with PBS. The pellet was resuspended in PBS and smeared on a clean glass slide, air dried and fixed in methanol. EDTA anticoagulated peripheral blood samples were smeared on glass slides, air dried and fixed in methanol. Fixed slides were then permeabilized with 4% paraformaldehyde for 20 min at 40C followed by blocking with 5% normal goat serum for 1 h. Thereafter, the slides were incubated with anti-CD63 primary antibody (1, 100) at 40C overnight. Slides were washed three times with PBST (0.2% Tween-20) and incubated with Alexa Fluor 568 anti-rabbit secondary antibody for 2 h at room temperature in the dark. The slides were washed with PBST and mounted in Prolong Vectashield® mounting medium with DAPI. The slides were then observed under a fluorescence microscope (Nikon) and analyzed using NIS Elements software.
2.7. Immunohistochemistry for CD63
Immunohistochemistry was performed to check the expression of CD63 protein in peritoneum, liver, spleen and lungs. Tissue samples were fixed in 10% neutral buffer formalin (NBF), embedded in paraffin and were cut into 5 μm sections. Sections were deparaffinized in xylene, rehydrated with graded ethanol (100%, 95%, 80%, 70% and 50%) and then transferred to tap water. The endogenous peroxidase activity was blocked by incubating the sections with 3% H2O2 for 20 min. Thereafter, the slides were heated in sodium citrate buffer (pH 6.0) solution at 950C for 20 min for antigen retrieval. Non-specific reactivity was blocked by incubating the slides with 5% normal goat serum for 1 h. The slides were washed three times in PBST and incubated with anti-CD63 primary antibody (1, 100) at 40C overnight in a humidified chamber. Thereafter, slides were washed and incubated with Alexa Fluor anti-rabbit secondary antibody (1, 200) for 2h at room temperature in the dark. The slides were washed with PBST and mounted in Prolong Vectashield® mounting medium with DAPI. The slides were observed under a confocal microscope (Leica) and analyzed using Leica LASX software.
2.8. Isolation of exosomes
For isolation of exosomes from serum, blood was collected through retro-orbital method from control and DL mice. The blood was allowed to stand for an hour at room temperature followed by centrifugation at 1500g for 10 min at 40C. The supernatant was collected and passed through a 0.22µm syringe filter. Thereafter, exosomes were isolated using Total exosome isolation kit (from serum) as per the manufacturer’s instruction. Briefly, the required volume of serum was mixed with 0.2 volume of reagent and incubated for 30 min at 40C followed by centrifugation at 10,000g for 10 min. The supernatant was discarded and the exosome pellet was resuspended in PBS and used for further downstream analysis.
For isolation of lymphoma-derived exosomes, ascites was collected from the peritoneum of DL mice and centrifuged at 1500g for 10 min at 40C. The supernatant was collected and passed through a 0.22µm syringe filter and exosomes were isolated using Total exosome isolation kit (from other body fluids) as per the manufacturer’s instruction. Briefly, the required volume of ascites was mixed with 0.5 volume of reagent and incubated for 30 min at room temperature. Then the sample was centrifuged at 10,000g for 10 min at room temperature. The supernatant was discarded and the exosome pellet was washed with PBS. The exosome pellet was resuspended in PBS and used for further downstream analysis.
2.9. Electron microscopy
For morphometric analysis of isolated exosomes, scanning electron microscopy (SEM) and transmission electron microscopy (TEM) were performed. Briefly, the exosome pellet was resuspended in 100µl of 2% paraformaldehyde. 5µl of suspension was deposited on a grid and was allowed to dry, coated with gold palladium and microscopy was performed.
2.10. Dynamic light scattering (DLS)
The size and dispersity of the isolated exosomes was determined by DLS using Malvern Zetasizer Nano ZS analyzer. 50μL of exosome suspension isolated from serum and DL-ascites were diluted to 750μL in PBS, added to disposable micro brand UV cuvettes for measurement and three scattering measurements were recorded. The graph for size distribution by intensity and count rate of each sample was determined using the Zetasizer analyzer.
2.11. Western blotting
DL cells, ascites and lymphoma-derived exosomes were lysed in RIPA lysis buffer containing protease inhibitor cocktail and centrifuged at 10,000g at 40C to obtain clear supernatant. Protein concentrations were estimated by BCA assay. Protein samples (30μg) were mixed with protein loading dye followed by heating for 5 min at 950C and centrifugation at 10,000g. The protein samples were then electrophoresed on 10% sodium dodecyl sulphate (SDS) polyacrylamide gels followed by their transfer onto nitrocellulose membrane. The membranes were blocked with 5% bovine serum albumin (BSA) and incubated with primary antibodies (anti-Alix and anti-flotillin-1) overnight at 40C followed by incubation with their respective secondary antibodies. The specific antibody-bound protein bands were detected with ECL under Amersham Imager 600 (GE Healthcare).
2.12. Liquid chromatography-mass spectrometry (LC-MS) analysis
The exosome pellet was lysed in Urea lysis buffer (8 M urea buffer in 50 mM ammonium bicarbonate pH 8.5 with protease inhibitor cocktail) and protein concentration was determined by BCA assay. Exosomal protein (200µg) was reduced with dithiothreitol (DTT) with a final concentration of 20mM and alkylated with iodoacetamide (IAA) with a final concentration of 40mM. Alkylation reaction was quenched by adding DTT to a final concentration of 10mM followed by trypsin digestion with a final protease to protein ratio of 1, 20 to 1, 100 (w/w). The peptides were fractionated by reverse phase nano-Liquid Chromatography (Thermo Scientific Easy-nLC 1200) on PepMap RSLC C18 (2µm, 75µm x 50cm) column. After separation, the peptides were analyzed by Q Exactive Orbitrap (Thermo Scientific). The tandem mass spectra were queried against the Mus musculus on UniProt (
https://www.uniprot.org/uniprotkb?facets=model_organism%3A10090&query=mus%20musculus) and analyzed using Proteome Discoverer 2.4. Thereafter, all the characterized proteins were analyzed for gene ontology using Gene Ontology Resource (
http://geneontology.org/) wherein Fisher’s exact test with FDR correction was used to assess the enrichment levels of proteins. Further, the protein-protein interaction was determined using the STRING database (
https://string-db.org/).
2.13. Exosome uptake assay
Exosomes were isolated from ascites and labelled with PKH26 dye according to the manufacturer’s protocol. Briefly, the exosome pellet was resuspended in 1ml of diluent C. Separately, 1ml of diluent C was mixed with 4μl of PKH26 dye. Immediately, the exosome suspension was mixed with the staining solution and incubated for 5 min. Subsequently, the reaction was stopped by adding an equal volume of DMEM supplemented with 2% exosome depleted-FBS or d-FBS. Labelled exosomes were further re-isolated using Exosome isolation kit (from cell culture media) as per manufacturer’s instructions. Briefly, 0.5volume of reagent was mixed with the sample and incubated at 40C overnight. Thereafter, the samples were centrifuged at 10,000g for 1hr at 40C. Supernatant was discarded and labelled exosome pellet was washed twice with PBS to remove unbound dye and resuspended in PBS. PKH labelled exosomes were quantified using BCA protein estimation kit and a total of 100μg of exosomes were incubated with RAW264.7 cells for different time points (30 min, 2hrs and 4hrs) and untreated cells were considered as control (0 min). After incubation time, the cells were washed with PBS thrice and fixed with methanol followed by rehydration with PBS for 5 min. The cells were mounted in Prolong Vectashield® mounting medium with DAPI and observed under a confocal microscope (Nikon).
2.14. Morphological analysis
To study the effect of lymphoma-derived exosomes on morphology of macrophages, crystal violet staining was performed. RAW 264.7 cells (5x103) were seeded in a 24 well plate and treated with exosomes and LPS, alone and combined. After 24h, the culture medium was removed and cells were washed with PBS. Cells were fixed with methanol followed by crystal violet staining. The morphology of cells was analysed under Nikon inverted microscope.
2.15. Phagocytic assay
To assess the phagocytic function of macrophages, 1x105 RAW264.7 cells per well were seeded in 6 well plate and treatment was given. After 24 hrs, the media was removed, cells were washed with PBS and heat killed yeast (0.5mg/ml) were flooded over macrophages in each well and incubated for 15 min to allow phagocytosis. Then cells were washed with PBS thrice and fixed in methanol and stained with crystal violet. Percent phagocytosis was calculated by counting the number of cells showing engulfment of yeast under Nikon inverted microscope.
2.16. Reactive oxygen species (ROS) detection
RAW264.7 cells were seeded on glass coverslips placed in a 24 well plate and given treatment of exosomes and LPS, alone and combined. After 24 hrs, the media was removed, cells were washed with PBS and incubated with dihydroethidium (DHE). DHE, a reduced form of ethidium bromide, can passively diffuse into the cells and reacts with the superoxide anion to form a red fluorescent product, 2-hydroxyethidium. It is commonly used as the most specific fluorescent probe for superoxide detection. DHE was suspended in DMSO at a stock concentration of 10mM and diluted for a final working concentration of 10μM in PBS. After incubation, the cells were washed with PBS and coverslips were mounted in Prolong Vectashield® mounting medium with DAPI and observed under inverted fluorescence microscope (Nikon).
2.17. Nitrite estimation
Nitric oxide (NO) level in the culture supernatant was estimated by measuring nitrite concentration, the stable end product of NO, using Griess reagents. Briefly, 100µl culture conditioned medium and Griess reagent (1, 1 of 0.1% naphthylethylenediamine dihydrochloride and 1% of sulfanilamide in 2.5% phosphoric acid) were mixed and incubated for 10 min at room temperature. Thereafter, absorbance was taken at 540 nm using microplate reader (EPOCH2, BioTek). Sodium nitrite was taken as standard and unknown concentrations in the samples were extrapolated from the standard curve.
2.18. RNA extraction and real-time quantitative PCR
To assess the expression of genes at transcriptional level, total RNA was isolated from cells in different treatment groups using RNeasy Mini Kit as per the manufacturer’s protocol. The quality of RNA was checked by analyzing the absorbance ratio at 260/280 nm using Nanodrop. The RNA was treated with DNaseI and then reverse transcribed using cDNA synthesis kit. The cDNA was used to analyze the expression of NOS2, arginase-1, IL6, IL10, IL12, TGFβ, TNFα and IL1β by performing real time PCR (qPCR) using QuantStudio
™6 Flex System Thermal cycler. All sets of genes were run in duplicate with a reaction volume of 6µl using Power SYBR green master mix. The qPCR was run for 3min at 95
0C followed by 40 cycles, each consisting of 15s at 95
0C and 45s at 60
0C. The fold changes were calculated by 2
-(ΔΔCt) value and data were normalized by β-actin. The primers were designed using NCBI primer BLAST. Sequence of the primers used in the study are listed in
Table 1,
2.19. Statistical analysis
The statistical analysis was performed using GraphPad Prism 8 software by Student t-test for comparison between two groups and by one-way ANOVA followed by Tukey post hoc test for comparison between multiple groups. P≤0.05 was considered a statistically significant difference.
4. Discussion
Exponentially emerging reports have established the role of exosomes in cancer growth and its metastatic progression. Importantly, tumor-derived exosomes play a crucial role in mediating intercellular communication networks with stromal cells, including fibroblast cells and immune cells, at local and distant sites [
10,
79,
80,
81]. Indeed, immunosuppressive tumor microenvironment is known to be promoted by tumor-derived exosomes [
11,
12,
13,
14,
15]. Among various immunosuppressive cells, macrophages are the key immune cells whose enrichment within tumor microenvironment is associated with poor prognosis of cancer patients [
5,
17,
18]. Therefore, macrophages or tumor-associated macrophages are considered to be the key target in cancer immunotherapy. In recent years, although researchers have shown the impact of tumor-derived exosomes on M1/M2 activation state of macrophages, there are contrasting reports [
19,
22,
23,
25,
26]. Considering the pivotal role played by tumor-derived exosomes in mediating immunosuppressive environment and macrophages being the key players in governing prognosis of cancer in the host, there is an urgent need to decipher the cross-talk between tumor cells and macrophages via exosomes. In this line, we were interested in deciphering the effect of lymphoma-derived exosomes on macrophage activation. We observed increased exosome level in peripheral blood as well as in all the examined organs of lymphoma-bearing hosts. Further, we found that lymphoma-derived exosomes have immunomodulatory proteins which mediated a pro-tumoral change in macrophages by altering morphology, reducing phagocytic ability, increasing the expression of arginase-1 (M2-marker) and also by inducing the release of pro-tumoral cytokine profile.
Cancer cells are the transformed cells which show increased exosome biogenesis that carry oncogenic cargoes and alter the target cell behaviour towards pro-tumoral type [
7]. Indeed, tumor-derived exosomes play a crucial role in governing aggressiveness and metastatic potential of cancer cells [
5,
6,
7]. There are various molecular mechanisms which upregulate the exosome biogenesis machinery in cancer cells. Also, it has been revealed that inhibiting exosome machinery could reduce the pathogenicity of cancer [
7]. In the present study, we evaluated the exosome machinery in tumor cells wherein we observed increased level of exosomes in DL cells compared to the normal cells (
Figure 2A). Our result is in line with the existing reports which show increased production of exosomes by tumor cells [
7]. Once formed, the exosomes are released out of the cells and account for their increased level in peripheral blood. Importantly, various reports suggest a significantly high exosome level in systemic circulation of cancer patients than healthy individuals [
31]. Likewise, we observed increased count rate of exosomes in serum isolated from the DL-bearing group compared to the normal group (
Figure 2C(iii)) reflecting increased exosome level in the circulation of tumor-bearing hosts. Of note, we also observed increased uptake of exosomes by peripheral blood leukocytes in the DL-bearing group compared to the normal group (
Figure 2D). Although it has been revealed that increased exosome level in blood is not only contributed by tumor-derived exosomes, but, upon uptake, the immune cells further release exosomes either to suppress or promote the tumor growth, depending upon the molecular constituents of tumor-derived exosomes [
6]. However, in our study, whether the increased exosome level in blood is also contributed by immune cells or not needs to be further investigated.
Several lines of evidence have shown that exosomes act as key drivers for pre-metastatic niche formation in various cancers [
33]. Importantly, tumor-derived exosomes, once they enter the circulation, reach at different sites throughout the body and establish an immunosuppressive pre-metastatic niche, where the tumor cells actually metastasize [
7]. For instance, Peinado et al 2012, observed organotropism of melanoma cell-derived exosomes in the interstitium of lungs, liver, spleen and bone marrow [
82]. In contrast, another report suggests the presence of tumor-derived exosomes in metastatic as well as non-metastatic organs of tumor-bearing hosts [
34]. In our study, we also observed increased exosome level in various tissues (peritoneum, liver, spleen and lungs) of DL-bearing group (
Figure 3). Likewise, several line of evidence showed that systemically administered exosomes accumulate mainly in the vital organs, such as in liver, spleen, lungs and gastrointestinal tract [
83]. Previously, we have shown that DL represents a systemic disease wherein we observed disturbed histoarchitecture and infiltration of leukocytes in most of the organs, including peritoneum, liver, spleen and lungs [
30]. Presence of exosomes in these organs suggest that the exosomes might be mediating a change in functioning of stromal cells as well as infiltrating immune cells to make the sites favorable for metastasis. There are reports that show tumor microenvironment-associated cells also release more exosomes than in normal environments to promote cell to cell communication [
84]. So, we speculate that increased systemic exosome load might be majorly contributed by tumor-derived exosomes while stromal cells and infiltrating immune cells may also be releasing some exosomes in response to tumor burden. We recently reviewed various mechanisms which affect exosome biogenesis machinery in cancer cells, among them, hypoxia is known to be the key trigger [
7]. Conversely, exosome release by cancer cells has been found to be reduced under hyperoxic conditions [
85]. Moreover, accumulating evidence suggests that exosome biogenesis under hypoxic conditions is considered to be universal because in such conditions non-transformed cells also release a high number of exosomes [
86,
87]. In view of this, we correlated our previous study, wherein we found high systemic ROS levels in DL mice [
30], and speculate that increased systemic exosome load could be due to high oxidative stress which might be causing immune cells as well as other stromal cells to release more exosomes.
Several lines of evidence have also shown the role of key exosomal cargoes in promoting various aspects of tumor growth and progression, including oncogenic transformation, stromal remodelling, angiogenesis, immune evasion and metastasis. For instance, in multiple myeloma, tumor-derived exosomes have been characterized to have VEGF, bFGF, HGF, MMP-9 and Serpin E1 [
10] while glioblastoma-derived exosomes carry VEGF, TGFβ, gelatinases and plasminogen activators to promote angiogenesis [
88]. Moreover, there are reports which show the presence of immunomodulatory proteins within tumor-derived exosomes, such as presence of TGFβ1, IL-10 and Fas Ligand in ovarian cancer cell-derived exosomes and presence of NKG2D ligands and TGFβ1 in prostate cancer, mesothelioma and B-lymphoblastoid cell-derived exosomes, which could mediate immune evasion [
15,
89]. In this line, we investigated the proteomic constituents of lymphoma-derived exosomes, which is not yet revealed, by LC-MS analysis. Although most of the existing studies have used culture conditioned media of cancer cell lines for isolation of their exosomes, we isolated lymphoma-derived exosomes from the ascites of growing DL cells in the in vivo system. This negotiated the differences caused by in vitro culture of tumor cells and thus provided a more rational approach to investigate the molecular constituents of these exosomes. Interestingly, we found various oncogenic proteins in proteome profile of lymphoma-derived exosomes (data not shown). Also, we observed that many of these proteins are known to be EMT-related markers in several cancer types (
Table 2). This suggests invasive and metastatic potential of lymphoma-derived exosomes. Moreover, we observed enrichment of these proteins in immunomodulatory pathways (
Figure 4E(iii)). These immunomodulatory proteins formed a strong interactome network principally constituted by complement proteins, such as C1q (a,b,c), C3, C4 and C9 (
Figure 5). The immunomodulatory potential of complement proteins has been very well-established (25, 26). Importantly, within the tumor microenvironment, different complement proteins are known to alter the activation state of various immune cells (25). Also, evidence suggests the presence of complement proteins with extracellular vesicles which may modulate the innate and adaptive immune response (27, 28). Moreover, we observed presence of proteasomal proteins, including PSMA1, PSME1, PSMB8, PSMB9 and PSMB10 (
Figure 5). Besides their role in proteasome-mediated catabolic processes, the functional enrichment analysis revealed their involvement in immune system processes. Previous studies also reported presence of proteasomal subunits within exosomes [
90]. For instance, Ding et al, 2020, revealed the presence of various proteasomal proteins, including PSMA1, in exosomes isolated from serum of gastric cancer patients [
90]. Altogether, our results revealed that lymphoma-derived exosomal proteins could modulate the immune response of the host. As we were focused on investigating the impact of these exosomes on macrophage activation, we further screened these immunomodulatory proteins for their impact on macrophage function. From literature, it was observed that the exosomal cargoes such as Hsp90α, Myh9, CFHR4 and galectin3 are known to promote the recruitment of macrophages at tumor site in different cancer types (
Table 3). For instance, Hsp90α stabilizes macrophage migration inhibitory factor (MIF) which consequently promote cancer growth via recruitment of macrophages. These recruited macrophages release high level of VEGF and thus show pro-tumoral behavior [
35]. Moreover, we observed proteins which could induce M1 polarization such as STAT1, vitronectin, clusterin, or M2 polarization, such as S100A8/A9, annexin, nidogen 1, MMP-8, cathepsin S, cathepsin L, either in tumor conditions or in other pathological conditions (
Table 3). Depending upon the abundance value of these proteins, exosomes would switch the polarized state of macrophages either towards M1 or M2 type. This suggests that macrophages might be recruited at the tumor site or pre-metastatic site by the lymphoma-derived exosomes wherein their polarization would have been altered in the tumor-bearing host. Of note, we also observed proteins which are known to be overexpressed by M2 macrophages, such as arginase-1, macrophage mannose receptor 1, TGFβi, Tgm2, Vps35, which implied that lymphoma-derived exosomes could induce M2 macrophage polarization (
Table 3).
To investigate the impact of lymphoma-derived exosomes on macrophage polarization, we did in vitro study using RAW264.7 cell line. Importantly, we observed frequent uptake of lymphoma-derived exosomes, i.e., within 30min., by macrophages which further increased in a time-dependent manner (
Figure 6A). Our observation was in line with other studies, wherein macrophages have been shown to be the key player in the rapid uptake of exosomes [
22]. Also, it is evident that monocytes show frequent uptake of exosomes compared to other PBMCs [
11] and thus suggest that macrophages could readily accept the biological signals via exosomes. Further, we observed that exosomes induced a morphological change in macrophages wherein they become elongated and resembled spindle-shaped (
Figure 6B). Our results were consistent with previous reports which showed a phenotypic change in macrophages when incubated with tumor-derived exosomes [
5,
24]. For instance, Piao et al, 2018, observed a similar elongated morphology in pro-tumoral macrophages upon uptake of breast cancer cell-derived exosomes [
5]. Also, it is suggested that M2 macrophages exhibit more elongated morphology compared to M1 type [
91]. In a study, monocytes when treated with cord blood-stem cell-derived exosomes become M2 macrophages and displayed an elongated, spindle-shaped morphology [
92]. These studies suggest that lymphoma-derived exosomes mediated a pro-tumoral M2 like phenotypic change in macrophages. However, in another study, it has been revealed that different sources of tumor-derived exosomes induce different morphologies in monocytes due to cytoskeletal rearrangements [
11]. Besides morphological alterations, macrophages are known to show differential phagocytic activity with respect to their different polarized state. For instance, activation of immunosuppressive M2 macrophages in tumor conditions are known to show reduced phagocytosis [
74,
75]. Likewise, we observed reduced phagocytic activity of macrophages when treated with lymphoma-derived exosomes. Importantly, these exosomes also inhibited the increased phagocytic response induced by LPS (
Figure 6C). This suggests the activation of pro-tumoral M2 macrophages by lymphoma-derived exosomes.
To further confirm the polarized state of macrophages in presence of lymphoma-derived exosomes, we investigated various other parameters. Particularly, we assessed hypoxic condition, a predominant factor known to play a crucial role in mediating immunosuppressive pro-tumoral microenvironment. Indeed, high ROS level is a key determinant of hypoxic conditions. Within intra-tumoral hypoxic regions, infiltrating immunosuppressive leukocytes, such as myeloid-derived suppressor cells (MDSCs) and TAMs, are known to regulate T cell function to inhibit tumor cell killing via ROS production [
76,
77]. Likewise, we observed a significantly high level of ROS in macrophages when activated by lymphoma-derived exosomes (
Figure 7A). Importantly, high ROS levels not only mediate T cell deregulation but also govern the differentiated state of macrophages towards M2 type within tumor-microenvironment [
76]. In view of this, we hypothesized that lymphoma-derived exosomes might be polarizing macrophages towards pro-tumoral M2 type. To confirm the polarized state of macrophages induced by lymphoma-derived exosomes, we assessed their arginine metabolism. Macrophage polarization is governed by two opposing metabolic pathways for a single amino acid, arginine. Arginine can be metabolized either to nitric oxide (NO) and citrulline by nitric oxide synthase (NOS2) of M1 macrophages or to ornithine and urea by arginase-1 of M2 macrophages [
78]. We observed that macrophages in presence of exosomes produced only basal levels of NO. Importantly, presence of exosomes inhibited the effect of LPS for production of heightened levels of NO in macrophages (
Figure 7B). This result was further corroborated with increased expression of arginase-1 while NOS2 expression was unaltered in macrophages activated by exosomes (
Figure 7C & 7D). Importantly, LPS-induced NOS2 expression was found to be significantly inhibited in presence of exosomes (
Figure 7C) and thus explained the reduction in LPS-induced NO level in presence of exosomes compared LPS-stimulated group. Several lines of evidence have evaluated the M1 or M2 polarized state of macrophages based on the expression level of arginase-1 and NOS2 [
5,
19,
22,
26]. For instance, Bardi et al, 2018, demonstrated that melanoma-derived exosomes promoted mixed polarized state of macrophages (M1 and M2) as they expressed NOS2 as well as arginase-1 [
26]. Piao et al, 2017, evaluated that M2 polarized macrophages express arginase-1 when activated by tumor-derived exosomes and they further associated their polarization with lymph node metastasis [
5]. In view of the existing studies, we suggest that lymphoma-derived exosomes induced M2 polarized state in macrophages. Although, we also observed arginase-1 expression in LPS-stimulated macrophages (
Figure 7D) which is suggested to be a counter regulatory response wherein LPS induces NOS2 expression earlier than arginase-1 expression [
93]. Of note, enhanced level of arginase-1 in macrophages is not only important for immunosuppressive M2 polarization but it also weakens the NO-mediated cytotoxic response against the tumor cells. Arginase-1 not only enables the production of polyamines important for tumor cell proliferation but it also limits the substrate availability for the activity of NOS2 enzyme, consequently the NO level and thus prevents tumor cell death [
78].
M1/M2 activation of macrophages represent two extreme polarized states. While M1 macrophages are known to inhibit tumor growth, M2 macrophages, or TAMs, are known to promote tumor growth and metastasis via releasing pro-tumoral factors and immunosuppressive cytokines [
23]. Cytokines are low molecular weight soluble proteins that act in autocrine or paracrine manner to mediate intercellular communication. Principally, they are synthesized by immune cells which then regulate cell survival, differentiation as well as the overall immune response in the host [
94]. In particular, M2 macrophages are immunosuppressive cells which release anti-inflammatory cytokines, such as IL10 and TGFβ, and are known to promote the stemness and migratory properties of cancer cells [
8,
95,
96]. Also, it has been revealed that tumor-derived exosomes induce high level of IL10 and TGFβ in macrophages and thus mediate their polarization towards M2 type [
8,
19,
96,
97]. Similarly, we observed increased expression of these immunosuppressive cytokines (
Figure 8) which thus suggest activation of M2 macrophages. Besides these anti-inflammatory cytokines, it is evident that TAMs also release certain pro-inflammatory cytokines, such as IL6, IL1β and TNFα, with different sources of exosomes and show the pro-tumoral behavior [
22,
25,
26]. For instance, Linton et al, 2018 observed that pancreatic cancer cell-derived exosomes induced release of IL6, IL1β and TNFα in M2 polarized macrophages [
25]. In contrast, few reports also evaluated increased expression of these cytokines by M1 macrophages. However, these M1 macrophages showed the pro-tumoral role and promoted malignant properties of cancer cells [
22,
26]. Intriguingly, we observed increased expression of IL6 and IL1β while no significant change in the TNFα expression was found in macrophages activated by lymphoma-derived exosomes (
Figure 8). Importantly, in our study, the LPS-stimulated group showed the highest expression of IL6, IL1β and TNFα, however in presence of exosomes, the heightened expression of these cytokines was found to be significantly reduced (
Figure 8). It could be explained by the fact that the mechanism for the production of these inflammatory cytokines by M1 and M2 macrophages is different. For instance, LPS-induced IL6 production is principally mediated by NFĸB activation [
98]; on the other hand, in TAMs, IL6 production is known to be induced via p38-MAPK pathway [
99]. So, we speculate that lymphoma-derived exosomes might have inhibited the LPS-induced pathway for pro-inflammatory cytokine production. Additionally, we observed remarkably no difference in the expression profile of the key pro-inflammatory cytokine, IL12, in presence of exosomes. Importantly, we found significantly reduced levels of LPS-induced IL12 in presence of lymphoma-derived exosomes compared to LPS treatment alone (
Figure 8). This showed the effectiveness of lymphoma-derived exosomes to inhibit the LPS-induced M1 polarization of macrophages. Taken together, our results substantiated the pro-tumoral M2-specific cytokine profile in macrophages activated by lymphoma-derived exosomes.
Figure 1.
Schematic representation of experimental design.
Figure 1.
Schematic representation of experimental design.
Figure 2.
Exosome level increases in tumor cells and in peripheral blood of DL-bearing mice. A) Representative immunofluorescence images of T cells and DL cells stained with CD63 antibody (magnification, X 200) and A(i) represents the mean fluorescence intensity analysed by ImageJ software. SEM imaging of exosomes isolated from serum of B(i) control and B(ii) DL mice. C(i) and (ii) represent the size distribution of exosomes in control and DL mice, respectively, by dynamic light scattering (DLS). C(iii) Bar graph represents the count rate of exosomes in the sample determined by DLS. D) Immunofluorescence staining of blood smear of control and DL mice with antibody directed against CD63 (magnification, X 400) and A(i) represents the mean fluorescence intensity analysed by ImageJ software. Data are shown from one of the three independent experiments with a similar pattern of results and are expressed as mean ±SD. The statistical significance between control and tumor group was determined by Student t-test where *p<0.05 and ***p<0.001). .
Figure 2.
Exosome level increases in tumor cells and in peripheral blood of DL-bearing mice. A) Representative immunofluorescence images of T cells and DL cells stained with CD63 antibody (magnification, X 200) and A(i) represents the mean fluorescence intensity analysed by ImageJ software. SEM imaging of exosomes isolated from serum of B(i) control and B(ii) DL mice. C(i) and (ii) represent the size distribution of exosomes in control and DL mice, respectively, by dynamic light scattering (DLS). C(iii) Bar graph represents the count rate of exosomes in the sample determined by DLS. D) Immunofluorescence staining of blood smear of control and DL mice with antibody directed against CD63 (magnification, X 400) and A(i) represents the mean fluorescence intensity analysed by ImageJ software. Data are shown from one of the three independent experiments with a similar pattern of results and are expressed as mean ±SD. The statistical significance between control and tumor group was determined by Student t-test where *p<0.05 and ***p<0.001). .
Figure 3.
Exosome level increases systemically in tumor-bearing mice. Representative immunofluorescence images of peritoneum, liver, spleen and lungs stained with anti-CD63 antibody (magnification, X 600, scale bar, 50µm). Bar graphs show the quantitative analysis of immunofluorescence data analysed by ImageJ software. Data are shown from one of the three independent experiments with a similar pattern of results and are expressed as mean ±SD. The statistical significance between the control and tumor group was determined by Student t-test where ***p≤0.001.
Figure 3.
Exosome level increases systemically in tumor-bearing mice. Representative immunofluorescence images of peritoneum, liver, spleen and lungs stained with anti-CD63 antibody (magnification, X 600, scale bar, 50µm). Bar graphs show the quantitative analysis of immunofluorescence data analysed by ImageJ software. Data are shown from one of the three independent experiments with a similar pattern of results and are expressed as mean ±SD. The statistical significance between the control and tumor group was determined by Student t-test where ***p≤0.001.
Figure 4.
GO enrichment analysis of lymphoma-derived exosomal proteins characterized by LC-MS analysis. Lymphoma-derived exosomes were assessed for morphology using electron microscopy, Ai) scanning electron microscopy (SEM) and Aii) transmission electron microscopy (TEM), scale bar, 200nm. The size distribution of exosomes was determined by B) dynamic light scattering (DLS) and presence of exosomes was confirmed by C) immunoblotting using exosome-specific markers, Alix and flotillin-1. D(i) and D(ii) Venn diagrams depict the presence of overlap proteins in lymphoma-derived exosomes with the proteins present in ExoCarta and EMTome database, respectively. Bar graphs represent GO enrichment analysis of the identified proteins in lymphoma-derived exosomes in the E(i) cellular component, E(ii) molecular function and E(iii) biological process using Gene ontology resource. Top 20 significantly enriched GO terms (p<0.05) are displayed.
Figure 4.
GO enrichment analysis of lymphoma-derived exosomal proteins characterized by LC-MS analysis. Lymphoma-derived exosomes were assessed for morphology using electron microscopy, Ai) scanning electron microscopy (SEM) and Aii) transmission electron microscopy (TEM), scale bar, 200nm. The size distribution of exosomes was determined by B) dynamic light scattering (DLS) and presence of exosomes was confirmed by C) immunoblotting using exosome-specific markers, Alix and flotillin-1. D(i) and D(ii) Venn diagrams depict the presence of overlap proteins in lymphoma-derived exosomes with the proteins present in ExoCarta and EMTome database, respectively. Bar graphs represent GO enrichment analysis of the identified proteins in lymphoma-derived exosomes in the E(i) cellular component, E(ii) molecular function and E(iii) biological process using Gene ontology resource. Top 20 significantly enriched GO terms (p<0.05) are displayed.
Figure 5.
Interactome network of the immunomodulatory proteins identified in lymphoma-derived exosomes by LC-MS analysis. The proteins enriched in immunomodulatory processes were screened from the top 20 GO enriched biological processes.
Figure 5.
Interactome network of the immunomodulatory proteins identified in lymphoma-derived exosomes by LC-MS analysis. The proteins enriched in immunomodulatory processes were screened from the top 20 GO enriched biological processes.
Figure 6.
Uptake of lymphoma-derived exosomes mediate a morphological change in macrophages and reduces their phagocytic activity in the in vitro system. A) Fluorescence images showing the time kinetics for the uptake of lymphoma-derived exosomes by macrophages. The exosomes were pre-labelled with PKH dye and incubated with macrophages for different time points. The red fluorescence represents the uptake of pre-labelled exosomes by macrophages, (magnification, x 400, scale bar, 20µm). B) Representative images showing the morphological change in macrophages in different treatment groups by crystal violet staining (magnification, x 200, scale bar, 50µm). C) Bar graph represents the percent phagocytosis shown by macrophages in different treatment groups. The results are expressed as the mean ± SD from three independent experiments. Statistical significance between the groups was determined by one-way ANOVA followed by Tukey post hoc test where *p<0.05, ***p<0.001 vs control group and ###p<0.001 vs LPS-stimulated group.
Figure 6.
Uptake of lymphoma-derived exosomes mediate a morphological change in macrophages and reduces their phagocytic activity in the in vitro system. A) Fluorescence images showing the time kinetics for the uptake of lymphoma-derived exosomes by macrophages. The exosomes were pre-labelled with PKH dye and incubated with macrophages for different time points. The red fluorescence represents the uptake of pre-labelled exosomes by macrophages, (magnification, x 400, scale bar, 20µm). B) Representative images showing the morphological change in macrophages in different treatment groups by crystal violet staining (magnification, x 200, scale bar, 50µm). C) Bar graph represents the percent phagocytosis shown by macrophages in different treatment groups. The results are expressed as the mean ± SD from three independent experiments. Statistical significance between the groups was determined by one-way ANOVA followed by Tukey post hoc test where *p<0.05, ***p<0.001 vs control group and ###p<0.001 vs LPS-stimulated group.
Figure 7.
Lymphoma-derived exosomes increase the ROS levels and upregulate arginase-1 expression in macrophages. A) Representative images showing ROS level assessed by DHE staining in different treatment groups (magnification, x 200, scale bar, 50µm). A(i) Bar graph represents the mean fluorescence intensity of ROS analysed by ImageJ software. B) Bar graph represents the nitrite level assessed by Griess reaction in different treatment groups. C) and D) Representative mRNA expression of NOS2 and arginase-1, respectively, assessed by quantitative RT-PCR in different treatment groups. β-actin was used as an internal control. The results are expressed as the mean ± SD from three independent experiments. Statistical significance between the groups was determined by one-way ANOVA followed by Tukey post hoc test where *p<0.05, ***p<0.001 vs control group and ##p<0.01, ###p<0.001 vs LPS-stimulated group.
Figure 7.
Lymphoma-derived exosomes increase the ROS levels and upregulate arginase-1 expression in macrophages. A) Representative images showing ROS level assessed by DHE staining in different treatment groups (magnification, x 200, scale bar, 50µm). A(i) Bar graph represents the mean fluorescence intensity of ROS analysed by ImageJ software. B) Bar graph represents the nitrite level assessed by Griess reaction in different treatment groups. C) and D) Representative mRNA expression of NOS2 and arginase-1, respectively, assessed by quantitative RT-PCR in different treatment groups. β-actin was used as an internal control. The results are expressed as the mean ± SD from three independent experiments. Statistical significance between the groups was determined by one-way ANOVA followed by Tukey post hoc test where *p<0.05, ***p<0.001 vs control group and ##p<0.01, ###p<0.001 vs LPS-stimulated group.
Figure 8.
Lymphoma-derived exosomes induce an M2-specific cytokine profile in macrophages in vitro. Representative mRNA expression of IL10, IL6, TNFα, TGFβ, IL1β and IL12 assessed by quantitative RT-PCR in different treatment groups. β-actin was used as an internal control. The results are expressed as the mean ± SD from three independent experiments. Statistical significance between the groups was determined by one-way ANOVA followed by Tukey post hoc test where *p<0.05, **p<0.01, ***p<0.001 vs control group and #p<0.05, ##p<0.01, ###p<0.001 vs LPS-stimulated group.
Figure 8.
Lymphoma-derived exosomes induce an M2-specific cytokine profile in macrophages in vitro. Representative mRNA expression of IL10, IL6, TNFα, TGFβ, IL1β and IL12 assessed by quantitative RT-PCR in different treatment groups. β-actin was used as an internal control. The results are expressed as the mean ± SD from three independent experiments. Statistical significance between the groups was determined by one-way ANOVA followed by Tukey post hoc test where *p<0.05, **p<0.01, ***p<0.001 vs control group and #p<0.05, ##p<0.01, ###p<0.001 vs LPS-stimulated group.
Table 1.
List of primer sequence used for qPCR.
Table 1.
List of primer sequence used for qPCR.
Gene |
Forward |
Reverse |
Arginase-1 |
CTTAGAGATTATCGGAGCGCCT |
AAGTTTTTCCAGCAGACCAGC |
NOS2 |
ACAACAGGAACCTACCAGCTC |
TACAGTTCCGAGCGTCAAAGA |
IL6 |
CTTCTTGGGACTGATGCTGGT |
GCCATTGCACAACTCTTTTCTCA |
IL10 |
TGAGGCGCTGTCATCGATTT |
TGGCCTTGTAGACACCTTGG |
IL1β |
GCCACCTTTTGACAGTGATGAG |
ATGTGCTGCTGCGAGATTTG |
IL12 |
TGTGGAATGGCGTCTCTGTC |
AGTTCAATGGGCAGGGTCTC |
TGFβ |
ATGCTAAAGAGGTCACCCGC |
ACTGCTTCCCGAATGTCTGA |
TNFα |
ACGCTGATTTGGTGACCAGG |
CCCGTAGGGCGATTACAGTC |
β-actin |
CTTCTTGGGTATGGAATCCTG |
GTAATCTCCTTCTGCATCCTG |
Fibronectin |
Lymphocyte cytosolic protein 1 |
Complement factor H |
Sulfhydryl oxidase 1 |
Filamin α |
Vimentin |
Moesin |
Alpha actinin 1α |
Keratin 5 |
Keratin 15 |
Xanthine dehydrogenase/oxidase |
Epidermal growth factor receptor |
Serine protease HTRA1 |
Nidogen-1 |
Keratin 8 |
Protein S100-A9 |
Platelet-activating factor acetylhydrolase |
Fibrillar collagen NC1 domain-containing protein |
Collagen α-1(I) chain |
Keratin 19 |
Keratin 14 |
Junction plakoglobin |
Keratin 16 |
Transforming growth factor-β-induced (TGFβi) |
Fibulin-1 |
Laminin subunit Ƴ-2 |
Fibrillar collagen NC1 domain-containing protein |
Protein S100-A8 |
Extracellular matrix protein 1 |
Desmoplakin |
Ectonucleotide pyrophosphatase 2 |
Laminin B1 |
Filamin-β |
Coronin |
D-3-phosphoglycerate dehydrogenase |
ZnMc domain-containing protein (MMP3) |
Fibulin-2 |
Periostin isoform M2 |
Procollagen-lysine 5-dioxygenase |
Kallikrein related-peptidase 10 |
Basement membrane-specific heparan sulfate proteoglycan core protein |
Procollagen C-endopeptidase enhancer 1 |
Decorin |
Low density lipoprotein receptor-related protein 1 |
Guanine nucleotide-binding protein subunit β-4 |
Thrombospondin 1 |
Prolyl 3-hydroxylase 2 |
Collagen, type IV, α2 |
Platelet-activating factor acetylhydrolase IB subunit α |
Heme oxygenase (biliverdin-producing) (HMOX1) |
Neuropilin 1 |
Doublecortin like kinase 1 |
Protein-glutamine Ƴ-glutamyltransferase 2 |
Collagen, type VI, α3 |
Signal transducer and activator of transcription (STAT1) |
Table 3.
List of exosomal proteins that affect either macrophage recruitment or macrophage polarization.
Table 3.
List of exosomal proteins that affect either macrophage recruitment or macrophage polarization.
Proteins affecting recruitment of macrophages |
References |
Heat shock protein 90α (Hsp90α) |
[35] |
Myosin-9 (Myh9) |
[36] |
Complement factor H-related 4 (CFHR4) |
[37] |
Galectin 3 |
[38] |
Actin-related protein 2/3 complex subunit |
[39] |
Proteins associated with M1 macrophage polarization |
References |
Pigment epithelium-derived factor |
[40] |
Adenylyl cyclase-associated protein |
[41] |
ADP-ribosylation factor 3 |
[42] |
Vitronectin |
[43] |
Clusterin |
[44] |
Signal transducer and activator of transcription 1 (STAT1) |
[45] |
ATP-citrate synthase |
[46] |
Proteins associated with M2 macrophage polarization |
References |
Fibronectin |
[47] |
Receptor of activated protein C kinase 1 |
[48] |
Adhesion G protein-coupled receptor E5 |
[49] |
Myeloperoxidase |
[50] |
Neutrophilic granule protein |
[51] |
Nidogen 1 |
[52] |
Nicotinamide phosphoribosyl transferase |
[53] |
Nucleolin |
[54] |
Cathepsin B |
[55] |
Protein S100A8 |
[56] |
Plasma protease C1 inhibitor |
[57] |
α-1-acid glycoprotein |
[58] |
Protein S100A9 |
[56] |
Ras-related C3 botulinum toxin substrate 2 |
[59] |
Heme oxygenase |
[60] |
ZnMc domain-containing protein |
[61] |
Annexin |
[62] |
α-2-HS-glycoprotein |
[63] |
Vitamin K-dependent protein S |
[64] |
Procathepsin L |
[55] |
Nidogen |
[52] |
Proteasome subunit β5 |
[65] |
14-3-3 protein € |
[66] |
Arginase 1 |
[67] |
Macrophage mannose receptor 1 (Mrc1) |
[67] |
Chitinase-like protein 3 |
[68] |
Protein-glutamine Ƴ-glutamyltransferase 2 (Tgm2) |
[67] |
α actinin 1 A |
[69] |
Vacuolar protein sorting-associated protein 35 (Vps35) |
[67] |
Apolipoprotein E (ApoE) |
[70] |
Cytoplasmic FMR1-interacting protein (Cyfip1) |
[71] |
Transferrin receptor protein 1 |
[71] |
Transforming growth factor-beta-induced protein (TGFβi) |
[71] |
Lipoprotein lipase |
[71] |
Lipopolysaccharide binding protein |
[71] |