Abstract
Small extracellular vesicles (sEV) contribute to the crosstalk between tumor cells and stroma, but the underlying signals are elusive. Here, we show that sEV generated by breast cancer cells in hypoxic (sEVHYP), but not normoxic (sEVNORM) conditions activate NFκB in recipient normal mammary epithelial cells. This increases the production and release of inflammatory cytokines, promotes mitochondrial dynamics leading to heightened cell motility and disrupts 3D mammary acini architecture with aberrant cell proliferation, reduced apoptosis and EMT. Mechanistically, Integrin-Linked Kinase packaged in sEVHYP via HIF1α is sufficient to activate NFκB in the normal mammary epithelium, in vivo. Therefore, sEVHYP activation of NFκB drives multiple oncogenic steps of inflammation, mitochondrial dynamics, and mammary gland morphogenesis in a breast cancer microenvironment.
INTRODUCTION
Small (40–150 nm) extracellular vesicles (sEV) originating from the late endosomal trafficking machinery or via shedding of the plasma membrane [1] are important mediators of tumor-stroma crosstalk [2]. Released by virtually all cells, including breast cancer cells [3], sEV can package a diverse array of bioactive proteins and RNAs that affect multiple traits of a tumor microenvironment, including stemness [4], exit from dormancy [5] and inflammatory responses [6]. In addition, sEV have been linked to enhanced cell motility of both tumor and stromal components [7]. Mechanistically, this involves modulation of directional cell movements [8], integrin-dependent adhesion [9], and, more recently, deregulated mitochondrial dynamics with increased mitochondrial accumulation at the cortical cytoskeleton to fuel membrane requirements of cell motility [10].
Despite these advances and their clear disease-relevance [11], the requirements of sEV signaling, especially in the regulation of a tumor microenvironment, are still mostly unknown [12]. In particular, a role of sEV in pleiotropic inflammatory mechanisms, capable of influencing local and distant recurrences, as well as response to therapy, has not been widely investigated [13].
In this study, we examined the effect of breast cancer-derived sEV generated in hypoxic conditions [10], a common occurrence in a tumor microenvironment, on inflammatory reprogramming of normal recipient mammary epithelial cells.
RESULTS AND DISCUSSION
Hypoxic sEV-associated transcriptome.
We isolated sEV from HER2−/ER+/PR+ human breast adenocarcinoma MCF7 cells maintained under conditions of normoxia (sEVNORM) or hypoxia (sEVHYP). sEV quantified after each collection had comparable morphology (Supplementary Fig. S1A), yield (Supplementary Fig. S1B) and size distribution (Supplementary Fig. S1C), in agreement with previous findings [10]. sEVNORM and sEVHYP isolated from HER2−/ER−/PR− human breast adenocarcinoma MDA-231 or normal mammary epithelial MCF10A cells were also comparable (Supplementary Figure S1B). sEV production in these settings was associated with negligible (<5%) cell death (Supplementary Figure S1D). By RNA-Seq profiling [10], sEVHYP, but not sEVNORM produced by MCF-7 cells caused the upregulation of several NFκB target genes in recipient MCF10A cells (Fig. 1A), including TNFα, IFNγ, IL1β, IL6, IL17, RelA, and PRKCD (Supplementary Fig. S1E).
Fig. 1: Breast cancer-derived sEVHYP activate NFκB in MCF10A recipient cells.
A Bioinformatics analysis of NFκB target genes upregulated by sEVHYP in recipient MCF10A cells by RNA-Seq. B MCF10A cells treated with sEVNORM or sEVHYP for 24 h were analyzed for nuclear accumulation of p50 NFκB by fluorescence microscopy. Representative images. Scale bar, 25 μm. Ctrl, control. Cyan, nuclei; yellow, p50 NFκB. C The conditions are as in B and nuclear accumulation of p50 NFκB was quantified at the indicated increasing concentrations of sEVNORM or sEVHYP. Mean±SD (two independent experiments). D MCF10A cells treated with sEVNORM or sEVHYP were incubated with pXSC or transfected with p65 NFκB -directed siRNA (si-p65) and analyzed for expression of the indicated NFκB target genes, by qRT-PCR. Mean±SD (N=3). E Conditioned media from MCF10A cells treated as in D were analyzed for cytokine release by LEGENDplex. Mean±SD (N=4). F sEV-induced nuclear accumulation of p50 NFκB was quantified in the presence or absence of pXSC. For panels C-F, numbers correspond to p values by 1-way Anova with Tukey’s posttest.
In validation experiments, sEVHYP treatment increased NFκB transcriptional activity using a luminescence reporter gene (Supplementary Fig. S1F), promoted nuclear accumulation of the p50 subunit of NFκB in a dose-dependent fashion (Fig. 1B, C), and upregulated the mRNA levels of NFκB target cytokines, IL6, IL8, TNFα and MCP1 (Fig. 1D) in recipient MCF10A cells. This was associated with increased release of IL6 and TNFα in the supernatant of sEVHYP-treated MCF10A cells (Fig. 1E). In terms of specificity, a small molecule inhibitor of NFκB DNA binding, 1,4-phenylenebis(methylene)selenocyanate (pXSC) or siRNA silencing of the p65 subunit of NFκB prevented sEVHYP-induced NFκB transcriptional activity (Supplementary Fig. S1F), blocked nuclear accumulation of p50 NFκB (Fig. 1F), and suppressed inflammatory cytokine production (Fig. 1D) and release (Fig. 1E) to levels of control cultures. sEVNORM had no effect on NFκB signaling in the presence or absence of pXSC or p65 silencing (Fig. 1D–F, Supplementary Fig. S1F).
Requirements of ILK and HIFIα for sEVHYP induced NFκB activity.
We next looked at the composition of NFκB -inducing sEVHYP. Consistent with previous observations [10], sEVHYP produced by MCF7 or MDA-231 breast cancer cell types contained higher levels of Integrin-Linked Kinase (ILK), compared to sEVNORM, whereas no differences were observed in sEV produced by MCF10A cells, by flow cytometry (Supplementary Fig. S2A). Similarly, 3D reconstruction of confocal microscopy images from MCF7 cells showed increased co-localization of ILK with the sEV marker, CD63 in hypoxia, but not normoxia (Supplementary Fig. S2B, C). siRNA silencing of HIF1α in sEVHYP-producing MCF7 cells suppressed the co- localization of ILK with CD63, by confocal microscopy (Supplementary Fig. S2D), and reduced the levels of ILK in sEV, by Western blotting (Supplementary Fig. S2E, F). Similar results were obtained with an independent breast cancer cell type, T47D (Supplementary Fig. S2D). Conversely, HIF1α silencing did not affect ILK levels in sEVNORM, by confocal microscopy (Supplementary Fig. S2B–D) or Western blotting (Supplementary Fig. S2E, F). Other cellular (calnexin) or sEV (Flotillin 1 and TSG101) markers were also unaffected (Supplementary Fig. S2E).
Based on these data, we next asked if ILK and/or HIF1α were important in sEVHYP induction of NFκB. In these experiments, siRNA silencing of ILK or HIF1α in sEV-producing breast cancer cells suppressed nuclear accumulation of p50 NFκB (Supplementary Fig. S3A) and cytokine mRNA upregulation in recipient cells (Supplementary Fig. S3B). Small molecule inhibitors of ILK (Cpd22) or its downstream target, Akt (MK2206) gave similar results, preventing nuclear localization of p50 NFκB (Supplementary Fig. S3C) and upregulation of cytokine mRNA levels (Supplementary Fig. S3D), compared to controls. As an independent approach, transfection of MCF10A cells with WT ILK, but not loss of function L207W ILK mutant was sufficient to induce nuclear accumulation of p50 NFκB (Supplementary Fig. S4A, B), increase cytokine mRNA levels (Supplementary Fig. S4C) and upregulate NFκB target proteins, MMP9 and BclXL, by Western blotting (Supplementary Fig. S4D).
NFκB regulation of sEVHYP-induced mitochondrial-fueled cell motility.
Consistent with recent findings [10], sEVHYP stimulated mitochondrial dynamics in MCF10A cells (Fig. 2A) resulting in increased rates of both mitochondrial fusion and fission (Fig. 2B). This was accompanied by redistribution of mitochondria to the cortical cytoskeleton of sEVHYP recipient cells (Fig. 2C, Supplementary Fig. S5B). NFκB inhibition with pXSC normalized mitochondrial dynamics in the presence of sEVHYP (Fig. 2A), restored the rate of mitochondrial fusion and fission events to levels of control cultures (Fig. 2B, Supplementary Fig. S5A) and prevented the redistribution of mitochondria to the cortical cytoskeleton (Fig. 2C, Supplementary Fig. S5B). Treatment of recipient cells with sEVNORM had no effect on mitochondrial dynamics or mitochondrial localization to the cortical cytoskeleton in the presence or absence of pXSC (Fig. 2B, C and Supplementary Fig. S5A, B).
Fig. 2: NFκB regulation of sEVHYP-dependent mitochondrial dynamics and cell motility.
A, B MCF10A cells treated with sEVHYP were incubated with or without pXSC and analyzed for changes in mitochondrial volume indicative of organelle fusion (>1.3-fold, positive y-scale) or fission (<0.7-fold, negative y-scale) over 40-sec intervals (A) and the number of mitochondrial fusion and fission events was quantified (B). Each line corresponds to a single cell. Mean±SD (N=3). Numbers correspond to p values by unpaired t-test. C MCF10A cells treated with sEVNORM or sEVHYP were imaged after 24 h by confocal microscopy and mitochondrial accumulation at the cortical cytoskeleton was quantified. Mean±SD (N=3). D MCF10A cells as in C were analyzed for single cell motility in 2D contour plots with quantification of speed of cell movements (Velocity, Vel, μm/min) and distance traveled by individual cells (Dis, μm). The cutoff velocities for slow (black, <0.53 μm/min)- or fast (red, >0.53 μm/min)-moving cells are indicated. E MCF10A cells transfected with siCtrl or si-p65 NFκB were treated with sEVNORM or sEVHYP and analyzed for directional cell migration in a wound closure assay at the indicated time intervals. Mean±SD (N=3). F MCF10A cells incubated with sEVNORM or sEVHYP were treated with pXSC or transfected with si-p65 NFκB and analyzed for migration on PET inserts.Mean±SD (N=3). For panels C and F, numbers correspond to p values by 1-way Anova with Tukey’s posttest.
Increased mitochondrial dynamics mediated by sEVHYP fuels directional cell motility [10, 14] and a role of NFκB in this response was next investigated. Treatment with pXSC or siRNA silencing of p65 NFκB suppressed single cell motility mediated by sEVHYP (Fig. 2D, Supplementary Fig. S5C), inhibited directional cell motility in a wound closure assay (Fig. 2E, Supplementary Fig. S5D) and blocked cell migration in a Transwell assay (Fig. 2F). In rescue experiments, reconstitution of recipient MCF10A cells with WT p65 NFκB (Supplementary Fig. S5E) normalized single cell motility and restored the speed of cell movements and the distance traveled by individual cells to levels of control transfectants (Supplementary Fig. S5F). Conversely, reconstitution with a loss-of-function T305D p65 NFκB mutant had no effect (Supplementary Fig. S5E, F).
Deregulation of epithelial morphogenesis by sEVHYP-NFκB signaling.
When tested in a model of 3D mammary gland formation [10], sEVHYP treatment increased MCF10A acini surface area and reduced acini circularity (Supplementary Fig. S6A, B). This was associated with extensive oncogenic changes, including nuclear accumulation of p50 NFκB, sustained upregulation of ILK, increased cell proliferation by Ki-67 staining and direct cell counting (Supplementary Fig. S6C) and impaired apoptosis with reduced caspase-3 expression and upregulation of survivin (Fig. 3A, B). sEVNORM had no effect on MCF10A acini size and circularity (Supplementary Fig. S6A, B) or oncogenic changes (Fig. 3A, B, Supplementary Fig. S6C). In addition, sEVHYP treatment induced hallmarks of EMT in MCF10A acini with upregulation of vimentin, N-cadherin and nuclear accumulation of SNAIL by fluorescence microscopy (Fig. 3A, B) as well as Western blotting (Supplementary Fig. S6E). Mechanistically, NFκB inhibition with pXSC restored MCF10A acini morphogenesis (Supplementary Fig. S6B), suppressed oncogenic changes (Fig. 3A, B), including cell proliferation (Supplementary Fig. S6D), and prevented EMT (Fig. 3A, B, Supplementary Fig. S6E) induced by sEVHYP. As an independent approach, small molecule inhibition of ILK or Akt with Cpd22 or MK2206, respectively, also prevented sEVHYP upregulation of N-cadherin and vimentin by Western blotting (Supplementary Fig. S6E).
Fig. 3: NFκB regulation of 3D mammary acini morphogenesis.
A MCF10A acini in 3D culture seeded in the presence of Ctrl, sEVNORM or sEVHYP with or without pXSC (added once at t=0) were analyzed by fluorescence microscopy 21 d after seeding (representative images; scale bar, 15 μm). B The conditions are as in A and the expression of p50 NFκB, ILK, Ki-67, cleaved (Cl) caspase 3, survivin, SNAIL and vimentin was quantified. Magenta, nuclei; yellow, individual protein of interest. MFI, mean fluorescence intensity. Mean±SD (N=3). Numbers correspond to p values by 1-way Anova with Tukey’s posttest.
Finally, we tested the potential relevance of this pathway, in vivo, and we isolated sEV from mouse breast adenocarcinoma AT3 cells under hypoxic or normoxic conditions. AT3-derived sEVNORM or sEVHYP had comparable size particle distribution (Supplementary Fig. S7A) and expressed sEV (flotillin, TSG101), but not cellular (calnexin, GM130) markers (Supplementary Fig. S7B). Injection of AT3-derived sEVHYP in the abdominal mammary gland of immunocompetent C57BL/6 mice was associated with increased ILK expression in the recipient epithelium after 6 weeks, by immunohistochemistry (IHC) (Fig. 4A, B). Consistent with the data above, sEVHYP injection in the normal mammary epithelium resulted in nuclear accumulation of p50 NFκB, in vivo, by immunofluorescence staining (Fig. 4C, D), as well as EMT, with upregulation of vimentin (Fig. 4A, B), decreased level of E-cadherin and nuclear localization of SNAIL (Fig. 4C, D).
Fig. 4: sEV-induced EMT in the mammary gland, in vivo.
A, B AT3-derived sEVHYP were injected in the abdominal mammary gland of C57BL/6 mice and tissue samples harvested after 6 weeks were analyzed by immunohistochemistry (A) and reactivity with antibodies to ILK or vimentin was quantified (B). Ctrl, control. Representative images. Scale bar, 100 μm. C, D The conditions are as in A and mammary gland tissues harvested after 6 weeks were analyzed by immunofluorescence (C, representative images) and changes in expression of p50 NFκB, ILK, E-cadherin and SNAIL were quantified (D). Scale bar, 50 μm. MFI, mean fluorescence intensity. Mean±SD. Numbers correspond to p values by unpaired t-test.
In sum, we have shown that sEV packaged by breast cancer cells in hypoxia [10] activate NFκB in a normal mammary epithelium, promoting inflammatory cytokine release, mitochondrial dynamics with heightened cell migration, disruption of 3D acini morphogenesis and EMT, in vivo. A role of sEV, let alone sEVHYP in NFκB activation has not been previously described. This pathway may be uniquely suited to globally alter a hypoxic breast cancer microenvironment, which occurs commonly in patients, favoring field cancerization [15] and oncogenic inflammatory changes [13]. Key effectors of this response included HIF1α, which has been linked to sEV-induced immunologic and metabolic changes in breast cancer [16] as well as ILK, leading to downstream activation of its pleiotropic oncogenic target, Akt [17]. Although its mechanism(s) of action continue to be debated [18], these data identify ILK as an important therapeutic target to disrupt sEV-induced oncogenic and inflammatory signaling during breast cancer development.
Supplementary Material
ACKNOWLEDGMENTS
We thank James Hayden and Frederick Keeney of the Wistar Imaging Core Shared Resource for assistance with time-lapse videomicroscopy and Sudheer Mulugu of the Electron Microscopy Resource Lab, Perelman School of Medicine, University of Pennsylvania for cryo-electron microscopy.
FUNDING
This work was supported by National Institutes of Health (NIH) grants P01 CA140043, R35 CA220446 (D.C.A.), R50 CA211199 (A.V.K.) and an award from the Mary Kay Ash Foundation (D.C.A.).
Footnotes
COMPETING INTERESTS
The authors declare that they have no competing interests.
REFERENCES
- 1.Jaiswal R, Sedger LM. Intercellular Vesicular Transfer by Exosomes, Microparticles and Oncosomes - Implications for Cancer Biology and Treatments. Front Oncol 2019; 9: 125. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Kalluri R, LeBleu VS. The biology, function, and biomedical applications of exosomes. Science 2020; 367. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.You S, Barkalifa R, Chaney EJ, Tu H, Park J, Sorrells JE et al. Label-free visualization and characterization of extracellular vesicles in breast cancer. Proc Natl Acad Sci U S A 2019; 116: 24012–24018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Zhao LJ, Li YY, Zhang YT, Fan QQ, Ren HM, Zhang C et al. Lysine demethylase LSD1 delivered via small extracellular vesicles promotes gastric cancer cell stemness. EMBO Rep 2021; 22: e50922. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Sansone P, Savini C, Kurelac I, Chang Q, Amato LB, Strillacci A et al. Packaging and transfer of mitochondrial DNA via exosomes regulate escape from dormancy in hormonal therapy-resistant breast cancer. Proc Natl Acad Sci U S A 2017; 114: E9066- E9075. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Marar C, Starich B, Wirtz D. Extracellular vesicles in immunomodulation and tumor progression. Nat Immunol 2021; 22: 560–570. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Sung BH, Parent CA, Weaver AM. Extracellular vesicles: Critical players during cell migration. Dev Cell 2021; 56: 1861–1874. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Sung BH, von Lersner A, Guerrero J, Krystofiak ES, Inman D, Pelletier R et al. A live cell reporter of exosome secretion and uptake reveals pathfinding behavior of migrating cells. Nat Commun 2020; 11: 2092. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Singh A, Fedele C, Lu H, Nevalainen MT, Keen JH, Languino LR. Exosome-mediated Transfer of alphavbeta3 Integrin from Tumorigenic to Nontumorigenic Cells Promotes a Migratory Phenotype. Mol Cancer Res 2016; 14: 1136–1146. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Bertolini I, Ghosh JC, Kossenkov AV, Mulugu S, Krishn SR, Vaira V et al. Small Extracellular Vesicle Regulation of Mitochondrial Dynamics Reprograms a Hypoxic Tumor Microenvironment. Dev Cell 2020; 55: 163–177 e166. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Ma Y, Dong S, Li X, Kim BYS, Yang Z, Jiang W. Extracellular Vesicles: An Emerging Nanoplatform for Cancer Therapy. Front Oncol 2020; 10: 606906. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Lucien F, Leong HS. The role of extracellular vesicles in cancer microenvironment and metastasis: myths and challenges. Biochem Soc Trans 2019; 47: 273–280. [DOI] [PubMed] [Google Scholar]
- 13.Baram T, Rubinstein-Achiasaf L, Ben-Yaakov H, Ben-Baruch A. Inflammation-Driven Breast Tumor Cell Plasticity: Stemness/EMT, Therapy Resistance and Dormancy. Front Oncol 2020; 10: 614468. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Caino MC, Seo JH, Aguinaldo A, Wait E, Bryant KG, Kossenkov AV et al. A neuronal network of mitochondrial dynamics regulates metastasis. Nat Commun 2016; 7: 13730. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Curtius K, Wright NA, Graham TA. An evolutionary perspective on field cancerization. Nat Rev Cancer 2018; 18: 19–32. [DOI] [PubMed] [Google Scholar]
- 16.Chen F, Chen J, Yang L, Liu J, Zhang X, Zhang Y et al. Extracellular vesicle-packaged HIF-1alpha-stabilizing lncRNA from tumour-associated macrophages regulates aerobic glycolysis of breast cancer cells. Nat Cell Biol 2019; 21: 498–510. [DOI] [PubMed] [Google Scholar]
- 17.Hannigan G, Troussard AA, Dedhar S. Integrin-linked kinase: a cancer therapeutic target unique among its ILK. Nat Rev Cancer 2005; 5: 51–63. [DOI] [PubMed] [Google Scholar]
- 18.Vaynberg J, Fukuda K, Lu F, Bialkowska K, Chen Y, Plow EF et al. Non-catalytic signaling by pseudokinase ILK for regulating cell adhesion. Nat Commun 2018; 9: 4465. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.