Senescence is a nonproliferative survival state that cancer cells can enter to escape therapy. In addition to soluble factors, senescence cells secrete extracellular vesicles (EV), which are important mediators of intercellular communication. To explore the role of senescent cell (SC)–derived EVs (senEV) in inflammatory responses to senescence, we developed an engraftment-based senescence model in wild-type mice and genetically blocked senEV release in vivo, without significantly affecting soluble mediators. SenEVs were both necessary and sufficient to trigger immune-mediated clearance of SCs, thereby suppressing tumor growth. In the absence of senEVs, the recruitment of MHC-II+ antigen-presenting cells (APC) to the senescence microenvironment was markedly impaired. Blocking senEV release redirected the primary target of SC signaling from APCs to neutrophils. Comprehensive transcriptional and proteomic analyses identified six ligands specific to senEVs, highlighting their role in promoting APC–T cell adhesion and synapse formation. APCs activated CCR2+CD4+ TH17 cells, which seemed to inhibit B-cell activation, and CD4+ T cells were essential for preventing tumor recurrence. These findings suggest that senEVs complement the activity of secreted inflammatory mediators by recruiting and activating distinct immune cell subsets, thereby enhancing the efficient clearance of SCs. These conclusions may have implications not only for tumor recurrence but also for understanding senescence during de novo carcinogenesis. Consequently, this work could inform the development of early detection strategies for cancer based on the biology of cellular senescence.

Significance: Chemotherapy-treated senescent tumor cells release extracellular vesicles that trigger an immune response and suppress tumor recurrence.

See related commentary by Almeida and Melo, p. 833

Chemotherapeutic regimens such as paclitaxel and cisplatin exert their antitumor activity by switching cancer cells to a stable nonproliferative, metabolically active survival state generally referred to as cellular senescence (1). Senescent cells (SC) undergo characteristic changes, including an enlarged and flattened morphology and expression of distinct genes, including senescence-associated β-galactosidase (SA-β-Gal), p16 (CDKN2A), and p21 (CDKN1A) (2). Senescence induction leads to an immediate restriction in cancer cell propagation and tumor growth. However, pathologic accumulation of SCs leads to cancer recurrence (3, 4). How SCs interact with their microenvironment and the mechanisms that fail to prevent SC accumulation in aging tissues are still largely unknown.

The senescence-associated secretory phenotype (SASP) is a hallmark of senescence (5). The SASP is composed of several different families of soluble mediators involved in extracellular matrix proteolysis, chemotaxis, cell growth and differentiation, and immune regulation (5, 6). SASP factors are known to both induce paracrine senescence in neighboring cells and to elicit immune responses (1). Extracellular vesicles (EV) are important mediators of intercellular communication (7). SC-derived EVs (senEV) were recently identified as a component of the SC secretome that impacts the recruitment of immune cells (812). EVs are membrane-bound, nanometer-sized particles released by every cell type and carrying bioactive signaling molecules from the parental cells (13). Unfortunately, investigations on the roles of EVs in cancer and senescence have been hampered by the need to isolate them before intravenous reinfusion in animal models; these manipulations can introduce biases such as loss of EV diversity, misjudgment of the amount to reinfuse, and assumptions on EV biodistribution (14). Model systems that distinguish the contributions of senEVs from those of SASP factors would enable the study the EV functions during immune responses to cellular senescence.

Senescence surveillance is the immune-mediated clearance of SCs. Immune cell types involved in senescence surveillance include monocytes (15, 16), NK cells (16), and T cells (15). However, SASP-mediated recruitment of other immune cell subsets, such as myeloid-derived suppressor cells (16), can inhibit cytotoxic CD8 T-cell responses (3, 4) and promote tumor angiogenesis (1), thereby fostering tumor progression and recurrence. The dual role of the SASP on senescence surveillance may derive from confounding effects due to the presence of different amounts and types of senEVs in SASP studies. Approaches to distinguish between senEVs and the SASP would enable to clarify the paradoxical influence of cellular senescence in cancer.

In the present study, we hypothesized that senEVs play a key role in senescence surveillance. To test this hypothesis, we developed and validated an engraftment-based in vivo senescence model that minimizes the impact of senescence-inducing stimuli on immune cells while at the same time enabling functional studies of senEVs without impacting the SASP (1719).

Reagents

Paclitaxel from Taxus brevifolia >95% (cat. #T7402-1MG) and Cisplatin Pharmaceutical Secondary Standard (cat. #PHR1624-200MG), DMEM (cat. #D8062-6X500ML), dispase II (cat. #D4693-1G), BCA Protein Assay Kit (cat. #71285-3), insulin (cat. #I6634-50MG), hydrocortisone (cat. #H0888-10G), cholera toxin (cat. #C8052-.5MG), transferrin (cat. #T1147-100MG), Tri-Iodo-Thyronine (cat. #T5516-1 mg) were purchased from Sigma-Aldrich. Cellular Senescence Detection Kit (cat. #SG04-10) was purchased from Dojindo. Amicon ultra-15 centrifugal filters UFC910024 100 kDa (cat. #UFC910024), Licor IRDye 800CW streptavidin (cat. #NC0883593), trypsin–EDTA (cat. #25-200-056), and RIPA (10×) buffer (cat. #50-195-822) were purchased from Thermo Fisher Scientific. Phase-Flow Alexa Fluor 647 BrdU Kit (cat. #370706), 5-bromo-2-deoxyuridine (BrdU; cat. #423401), APC Anti-BrdU Antibody (cat. #364114), Annexin V Binding buffer (cat. #422201), FITC Annexin V (cat. #640945), True-Nuclear Transcription Factor Buffer Set (cat. #424401), and Zombie Aqua Fixable Viability Kit 500 tests (cat. #423102) were purchased from BioLegend. Cell lysis buffer (cat. #9803S) was purchased from Cell Signaling Technology. Anti-rabbit detection module for whole-exome sequencing, 12-230 kDa whole-exome sequencing separation module (cat. #DM-001), was purchased from ProteinSimple. Proteome Profiler Mouse Cytokine Array Kit, Panel A (cat. #ARY006) was purchased from R&D Systems. Penicillin/streptomycin solution (cat. #SV30010) was purchased from GE HealthCare. EGF (cat. #PHG0311) was purchased from Gibco. Flow count beads (cat. #C36950), Di 8 ANNEPS (cat. #D3167), pluronic F-127 (cat. #P3000MP), Halt Protease Inhibitor Cocktail (PI78430), High-Capacity cDNA Reverse Transcriptase Kit (#4368813), and Fast SYBR Green Master Mix (#4385616) were purchased from Thermo Fisher Scientific. Ultracentrifugation tubes 1 × 3 ½ (cat. #NC9146666) were purchased from Beckman Coulter. Flotillin-1 antibody (cat. #NBP2-75492) was purchased from Novus Biologicals). MACS buffer was prepared by adding 2 mmol/L EDTA (Thermo Fisher Scientific) and 0.5% BSA (Thermo Fisher Scientific) to 500 mL PBS. CD4 depleting antibody (cat. #GK1.5) was purchased from Bio X Cell. Oligos for RNAi (cat. #SR405972) were purchased from OriGene. A Key Resource Table is available as Supplementary Material (Supplementary Table S1).

Cell culture

Two oral epithelial squamous cell carcinoma cell lines were used in this study. MOC2 cells were obtained from Kerafast, Inc., and mEER+ cells were kindly provided by Dr. William Spanos (Sanford Research, Sioux Falls, SD). MOC2 carries the same mutations observed in human head and neck cancers, namely, Trp53, MAPK, and FAT (20), whereas mEER+ cells have been engineered to express HrasG12V and HPV-E6/E7 (21). MOC2 and mEER+ cell lines were cultured in E-media as previously described (22). Cell lines were tested for Mycoplasma. We prepared E-media by mixing DMEM with Ham’s F12 3:1 and supplementing with 10% FBS (from VWR), 0.5% penicillin/streptomycin solution,10 µL of 25 µg/µL hydrocortisone 16.8 µL of 0.25 µg/µL cholera toxin, 100 µL of 25 µg/µL transferrin, 250 µL of 10 µg/µL insulin, 3.4 µL of 0.2 µg/µL Tri-Iodo-Thyronine, and 250 µL of 10 µg/mL EGF.

Lentiviral vectors, gene transfer, and inhibition of EV release

Lentiviral vector (LV) production and transduction of MOC2 and mEER+ were performed as previously described (17, 23). Briefly, third-generation, self-inactivating LVs were produced by calcium phosphate precipitation. To compare cells expressing Rab35 dominant-negative mutant (Rab35-DN) with those expressing wild-type Rab35 (Rab35-WT), MOC2 and mEER+ carrying one copy of LV per cell (1CpC) were generated by titration of LVs on cells and purification of transduced cells from the LV dilution that yielded less than 20% positive cells. Purification was performed by magnetic sorting using dLNGFR, a truncated receptor commonly used as a surface marker (24). Cells were used as bulk populations, without cloning.

To complement our findings with Rab35-DN–expressing cells, Rab27a was targeted using siRNAs. SCs were treated with scrambled control oligos (siScr) or oligos targeting Rab27a (siRab27a) for 48 hours, along with paclitaxel treatment (see “Induction of senescence”). For oligo transfection, the kit protocol was followed. Briefly, 900 µL of RNAiMAX was diluted with half of the cell media required. Oligos (145 µL for siScr oligo and 50 µL for siRab27a oligo) were added to 30 mL cell media. The oligo and RNAiMAX dilutions were combined in a 1:1 ratio and incubated for 10 minutes, after which, the mixture was added dropwise to SCs in a 15-cm dish. At the endpoint, cell pellets and conditioned media were collected to determine Rab27a gene expression and EV inhibition levels.

Gene expression

RNA was isolated from SC pellets after siRNA oligo transfection using Qiagen RNeasy Column Purification Kit and was quantified using a NanoDrop spectrophotometer. cDNA was synthesized from 1,000 ng of RNA using High-Capacity cDNA Reverse Transcriptase Kit. Relative quantification of Rab27a was performed in triplicate with Fast SYBR Green Master Mix (Thermo Fisher Scientific) using the ΔΔCt method, with GAPDH as reference gene. The following primers were utilized for amplification of Rab27a and GAPDH (FWD: GAG​CAA​AGT​TTC​CTC​AAT​GTC​CG, REV: CTT​TCA​CTG​CCC​TCT​GGT​CTT​C and FWD: CAT​CAC​TGC​CAC​CCA​GAA​GAC​TG, REV :ATG​CCA​GTG​AGC​TTC​CCG​TTC​AG, respectively).

Induction of senescence

To optimize the induction of senescence in MOC2 or mEER+ using chemotherapeutic agents, a matrix of paclitaxel (0–1,000 nmol/L) and/or cisplatin (0–48 µmol/L) was used. MOC2 and mEER+ cells were expanded in E-media, plated at 20,000 cells per well in a 96-well plate (in quadruplicate), and maintained at 37°C and under 5% CO2 for 24 hours. Subsequently, the specified ranges of paclitaxel and/or cisplatin were added to the cells, and they were incubated for an additional 48 hours at 37°C in 5% CO2. As the stock solutions of paclitaxel and cisplatin were dissolved in DMSO, an equivalent amount of DMSO was added to the control wells.

Evaluation of senescence induction

SA-β-Gal

The analysis of cellular senescence was conducted using the cellular senescence detection kit SPiDER-β-Gal, and all steps were executed in accordance with the manufacturer’s instructions. Following the 48-hour treatment with paclitaxel and/or cisplatin, as previously outlined, the cells were washed with PBS and then incubated at 37°C with 5% CO2 for 1 hour after the addition of bafilomycin A1 solution. Subsequently, the cells were further incubated for an additional 30 minutes after the addition of SPiDER-β-Gal solution. After washing the cells with PBS, they were digested with EDTA-free trypsin, and the resulting cell suspension was collected in a round-bottom 96-well plate containing MACS buffer following centrifugation at 800 rpm for 5 minutes. Finally, a flow cytometer (BD Biosciences) was used for high-throughput detection, with excitation and emission set at 488 and 530 nm, respectively. For flow cytometry analysis, samples that were not treated with SPiDER-β-Gal were utilized to establish the threshold for the negative control.

Apoptosis/cell death

To assess the potential unwanted induction of apoptosis and/or cell death by paclitaxel and/or cisplatin, parallel plates were prepared. After removing the E-media containing paclitaxel and/or cisplatin, cells were washed with PBS, harvested by trypsin, and resuspended in 50 µL FITC Annexin V working solution [3% FITC Annexin V and 3% 7-aminoactinomycin D (7-AAD) in Annexin B Binding Buffer]. The cells were then incubated for 15 minutes at room temperature in the dark. Following this, the FITC Annexin V working solution was discarded, and the cells were resuspended in MACS buffer. Quantification was performed using a flow cytometer (BD Biosciences).

Proliferation

Thymidine analog BrdU labeling was conducted following the manufacturer’s guidelines. Briefly, MOC2 and mEER+ squamous cell carcinoma lines were seeded at 20,000 cells per well in a 96-well plate and allowed to reach 70% confluence for 24 hours at 37°C with 5% CO2. The following day, cells were exposed to the chemotherapeutic agents and incubated for 48 hours. After washing the cells with media, a pulse of 5 µg BrdU per well was administered for 2, 4, or 24 hours, depending on the study’s objective. Subsequently, cells were washed, fixed, and permeabilized using the corresponding buffers, followed by treatment with DNase for 1 hour. After additional washing steps, cells were stained with 1 µg APC anti-BrdU antibody per well for 30 minutes and washed again, and signals were acquired on a flow cytometer (BD Biosciences). To quantify the residual proliferative potential (RPP) of SCs, we used the following formula:
in which PMAX is the fraction of proliferating cells at saturation (i.e., after 24 hours of paclitaxel treatment) and PPTX and PCTRL are the short-term (2 hours) proliferating fractions in paclitaxel or normal media, respectively. Therefore, the RPP represents a measure for the senescence-induced reduction in both proliferation rate and maximum proliferation.

Mice

Six- to eight-week-old female and male C57BL/6J mice weighting between 18 and 25 g for females and 20 to 30 g for males were purchased from Charles River Laboratories, housed under conventional SPF conditions, and provided with food and water ad libitum. Animal care was handled in accordance with the Guide for the Care and Use of Laboratory Animals of the Oregon Health and Science University (OHSU) and approved by the OHSU Institutional Animal Care and Use Committee. All animal studies, including implantation, tumor size assessment, anesthesia administration, surgery, and euthanasia, were carried out in the OHSU animal facility with a priority on minimizing stress and discomfort. Mice were housed in clean cages with a maximum of five mice per cage, furnished with bedding material, a nutritionally balanced diet, and access to clean water ad libitum. The temperature was maintained within the range of 20 to 26°C, and lighting adhered to a regular light–dark cycle of 12 hours each. Regular visual checks were conducted for health monitoring to promptly detect any signs of distress or illness. Anesthesia was induced using isoflurane, and euthanasia was performed in a CO2 chamber.

Engraftment of senescent SCs

For tissue engraftments, mice were anesthetized with inhalant isoflurane, and the success of anesthesia was confirmed by the lack of reaction to toe pinch. For intradermal senescence model, SCs were washed in 50 mL PBS to remove residual paclitaxel. A total of 0.5 × 106 MOC2 or 1 × 106 mEER+ suspended in 50 µL filtered PBS were injected into the flanks of the mice. For the oral senescence model, cells were injected into the central portion of the tongue in a volume of 20 µL. For the intradermal procedure, after removing hair using a body trimmer and shaving cream, the beveled tip of a needle with a gauge of 29G or 31G for males or females, respectively, was inserted face up at an angle of less than 10 degrees. The needle was advanced 2 to 3 mm into the tissue parallel to the skin while simultaneously lifting the needle to create a tenting effect. After injection of the cell suspension, a well-demarcated blister was a sign of successful intradermal placement. Mice in the control groups were injected in the same manner with an equivalent volume of filtered PBS. To track the site of injection for day 3 analyses, a black marker pen was used. Tumor volume was determined using a caliper, and the greater longitudinal diameter (length) and the greatest transverse diameter (width) were determined twice a week after injection. Tumor volume based on caliper measurement was calculated by the modified ellipsoidal formula as follows:
Mice were euthanized by CO2 asphyxiation just before tissue harvest. The site of injection was dissected and washed briefly in PBS and weighed. The time points indicated in the graphs as “days after challenge” mean days after injection of SCs. Quantification of tumor volumes in the oral mucosa (tongue tumors) was interrupted after two measurements because the mice could not drink and feed themselves anymore. Per Institutional Animal Care and Use Committee policies, we had to euthanize the cohorts.

Evaluation of immune response

Samples were dissociated using the following method: tissues were minced with scissors in a 1.5-mL tube in 100 µL dissociation media containing 2% FBS, 1× penicillin/streptomycin, 1× glutamine, 0.2 mg/mL collagenase IV, 2 mg/mL dispase, and 5 U/mL DNase I in Iscove’s Modified Dulbecco's Medium; after transferring to 15-mL conical tubes containing 5 mL dissociation media, minced tissues were incubated at 37°C with agitation at 500 rpm for 30 minutes in a heater–shaker. After dissociation, the tissues were filtered through a 100-µm strainer with 50 mL of MACS buffer and centrifuged at 500 g for 10 minutes. The resulting pellets were transferred to a deep 96-well plate on ice and mixed with prediluted surface antibodies and flow count beads. A fraction of the pellets was pooled and stained with corresponding antibodies for fluorescence minus o control samples. The plates were sealed with adhesive film and incubated on ice for 30 minutes in the dark. After incubation, the cells were washed twice with PBS and centrifuged at 500 g for 5 minutes. Subsequently, they were stained with Zombie Aqua dye and incubated for 15 minutes on ice in the dark. Following this, the cells were washed with PBS and gently vortexed to dissociate the pellets. The cells were then fixed and permeabilized using True-Nuclear Transcription Factor Buffer Set following the manufacturer’s guidelines. Briefly, the cells were fixed and incubated at room temperature for 30 minutes. Permeabilization was achieved by adding 1× perm buffer. After three washes in 1× perm buffer, the cells were gently vortexed. A mixture of intracellular antibodies in 1× perm buffer was added to the cells, and they were incubated overnight at 4°C in the dark. Following antibody incubation, the antibodies were removed by washing with perm buffer, and the cells were resuspended in MACS buffer. The readout was performed using a Fortessa flow cytometer.

Flow cytometry analysis of immune cells was performed according to the following gating:

First in vivo experiment:

  • Neutrophils: Zombie, CD45+, Ly6G+, and F4/80

  • MHC-II+ macrophages: Zombie, CD45+, Ly6G, F4/80+, CD80, and MHC-II+

  • Macrophages: Zombie, CD45+, Ly6G, and F4/80+

  • B cells: Zombie, CD45+, Ly6G, F4/80, B220+,CD3

  • Dendritic cells (DC): Zombie, CD45+, Ly6G, F4/80, B220, CD3, MHC-II++, and NK1.1

  • NK cells: Zombie, CD45+, Ly6G, F4/80, B220, CD3, MHC-II, and NK1.1+

  • NKT cells: Zombie, CD45+, Ly6G, F4/80, B220, CD3+, NK1.1+, and CD4

  • T cells: Zombie, CD45+, Ly6G, F4/80, B220, and CD3+

  • T regulatory cells: Zombie, CD45+, Ly6G, F4/80, B220, CD3+, and FoxP3+

  • T helper 1 cells: Zombie, CD45+, Ly6G, F4/80, B220, CD3+, CD4+, and Tbet+

Second in vivo experiment:

  • DCs: Zombie, CD45+, B220, F4/80, CD3, CD103+, XCR1+, and MHC-II+

  • B cells: Zombie, CD45+, B220+, F4/80, MHC-II+, CD3, NK1.1, and Ly6G

  • Activated B cells: Zombie, CD45+, B220+, F4/80, MHC-II+, CD3, NK1.1, Ly6G, CD80+, and CD86+

  • MHC-II+ macrophages: Zombie, CD45+, B220, F4/80+, MHC-II+, CD206, and CD80

EV purification

Ultrafiltration EV-depleted FBS was prepared as described elsewhere (25). In brief, regular FBS was subjected to centrifugation in Amicon Ultra-15 centrifugal filters for 55 minutes at 3,000 g. The resulting flow-through was collected and used to make E-media. Rab35-WT MOC2 (capable of EV production) and Rab35-DN MOC2 (exhibiting impaired EV production) were cultured in E-media containing EV-depleted FBS and paclitaxel for 72 hours. The absence of EVs in FBS did not impact senescence induction, nor did it change apoptosis or cell death. We used both E-media with and without FBS EVs, and we did not notice any difference. Supernatants were collected, normalized based on cell numbers, and then centrifuged at 2,000 rpm for 10 minutes to remove cell debris. The supernatant was transferred to ultracentrifugation tubes (Beckman Coulter) and subjected to centrifugation at 24,200 rpm at 20°C for 70 minutes in a SW28 rotor. Following centrifugation, the supernatant was discarded, and 35 mL of sterile, 0.2 µm filtered PBS was added to the pellet. The mixture was then centrifuged again at 24,200 rpm at 20°C for 70 minutes. After discarding the supernatants, the tubes were inverted and placed on tissue papers for 1 minute to ensure complete drainage of the tube walls. Subsequently, 50 µL of 0.2 µm filtered PBS was added to each tube. The contents were gently pipetted several times to collect EVs, which were then stored at −20°C until further use.

Transmission electron microscopy of EVs

Electron microscopy of EV preparations was done as previously described (23). Briefly, a 200 mesh–formvar, carbon-coated nickel grids were glow-discharged just before use. Five μL of each EV sample (1–5 mg/mL of protein) were placed on the grids and incubate for 5 minutes at room temperature. After washing with 3 × 30 µL of filtered PBS (1 minute/wash), the grids were fixed with 2% glutaraldehyde for 5 minutes at room temperature. After washing five times with water 2 minutes each wash, the grids were stained with UA 2% 3 minutes at room temperature.

Qualification of EVs

Western blotting

After successfully purifying EVs through ultracentrifugation, we proceeded with Western blot analysis using the Simple Western system (26) according to the manufacturer’s guidelines. For this purpose, EVs were lysed using 1× RIPA buffer containing proteinase inhibitor cocktail. The lysate was vortexed several times and mixed with 5× Fluorescent Master Mix. The resulting mixture was then denatured at 95°C for 5 minutes along with a ladder and subsequently transferred to ice. The final concentrations of EV lysate and rabbit anti–flotillin-1 were optimized at 0.2 mg/mL and 1:50, respectively.

Nanoparticle tracking analysis

The quantification of EVs was conducted using nanoparticle tracking analysis (NTA) instrument (ZetaView) using two independent methods: the first method utilized the properties of both light scattering and Brownian motion to obtain the nanoparticle size distribution of the sample in liquid suspension, and the second method evaluated the fluorescence of membrane-intercalating dyes (Di 8 ANNEPS) that have bound to the membrane of EVs. The sensitivity of ZetaView was calibrated to 70 using EV-free media diluted at a ratio of 1:1,000 in 0.22 µm filtered PBS. Subsequently, the number of particles in each sample was determined after diluting the samples at a 1:1,000 ratio in filtered PBS with the same calibration adjustment. For fluorescent-based quantification, a Di 8 ANNEPS working solution was prepared by combining 1% Di 8 ANNEPS and 0.25% pluronic F-127 in PBS. The supernatants containing EVs were then incubated with the Di 8 ANNEPS working solution in a 10:1 ratio for 10 minutes at room temperature. Following the incubation, the samples were diluted at a ratio of 1:1,000 in filtered PBS and subsequently applied to ZetaView, selecting the laser at 488 nm and the filter at 500 nm. Control samples (filtered PBS, filtered media, and diluted Di 8 ANNEPS working solution) were routinely used and yielded no particle counts.

Adoptive transfer of EVs from SCs

EVs from senescent MOC2 cells were collected as described above. Senescent MOC2 cells expressing Rab35-DN were mixed immediately before injection with senEVs (100 µg/mouse, about 25 µL) and implanted intradermally as described above (N = 10 mice per group). Additional transfers of senEVs (same dosing) were performed every 5 days, intradermally and near the site of SC injection. Control mice received senescent MOC2 cells expressing Rab35-DN mixed with PBS and subsequent PBS injections. The dosing was established based on the amount of senEVs released per day in vitro. To avoid batch effects, three separate senEV preparations were pooled, each yielding ∼4 mg/mL in 250 µL. Tumor volumes were measured as described above.

Mass spectrometry

Proteins were extracted using a modified Folch extraction method (27). Briefly, samples were extracted with chloroform/methanol (2:1, v/v) in a 4-fold excess to the sample volume to yield a hydrophobic layer containing lipids and a protein disc. Each layer was transferred to separate glass autosampler vials, and the protein pellet was washed with 1 mL of ice-cold methanol. Each sample was evaporated to dryness in vacuo and appropriately stored at −20°C until further processing and analysis.

Proteins were dissolved in 8 mol/L urea prepared in 50 mmol/L NH4HCO3, pH 8.0, containing 5 mmol/L dithiothreitol and reduced for 30 minutes at 37°C. Cysteine residues were alkylated for 30 minutes at 37°C with a final concentration of 10 mmol/L iodoacetamide from a 500 mmol/L stock solution. Samples were diluted 8-fold with 50 mmol/L NH4HCO3, pH 8.0, and 1 mol/L CaCl2 was added to a final concentration of 1 mmol/L. Proteins were digested with sequencing-grade modified trypsin (Promega) overnight at 37°C. Samples were submitted to solid-phase extraction using C18 spin columns (UltraMicroSpin columns, C18, 3- to 30-µg capacity; Nest Group), as previously described (28).

Peptides were separated on a capillary column (60 cm × 50 μm ID packed with C18, 3-μm particles) using a gradient of acetonitrile (mobile phase B) in water (mobile phase A), both supplemented with 0.1% formic acid. The elution was carried with the following gradient: 2 minutes, 8% B; 20 minutes, 12% B; 75 minutes, 30% B; 97 minutes, 45% B; 100 minutes, 95% B; 105 minutes, 95% B; 115 minutes, 1% B; and 120 minutes, 1% B. Eluting samples were analyzed on a Q-Exactive mass spectrometer (Thermo Fisher Scientific). Full-scan spectra were collected in a range of 300 to 1,800 m/z with a resolution of 70,000 at m/z 400. The 12 most intense parent ions with multiple charges were submitted to high-energy collision dissociation with 33% normalized collision energy at a resolution of 17,500. Each parent ion was fragmented once and then excluded for 30 seconds.

LC-MS/MS data were processed with MaxQuant (29) by searching tandem mass spectra against the human sequences from UniProt Knowledgebase. The intensity-based absolute quantification method was used for quantification (30).

Protein array

The expression levels of cytokines and chemokines in cell supernatants were quantified using Proteome Profiler Mouse Array Kit. Supernatants from 80% confluent cells were collected and centrifuged at 1,000 rpm for 5 minutes. Array membranes were incubated and developed with supernatants mixed with biotinylated detection antibodies following the manufacturer’s instructions. For the detection of cytokines/chemokines, IRDye 800CW streptavidin was used, and arrays were scanned using a Li-Cor Odyssey imaging system, which enables near-IR fluorescence detection. The mean pixel intensity for each signal was analyzed using ImageJ.

Bulk and single-cell RNA sequencing

Bulk RNA sequencing (RNA-seq) libraries were prepared using TruSeq Stranded mRNA Library Prep Kit (Illumina). Briefly, RNA was profiled for integrity on a 2200 Bioanalyzer (Agilent). Poly(A)+ RNA was isolated using Oligo-dT magnetic beads and then fragmented using divalent cations and heat. Fragmented RNA was converted to double-stranded cDNA by random hexamer priming. The second strand was synthesized with dUTP in place of dTTP to prevent priming from this strand during amplification. The cDNA was ligated to adapters with dual unique indexes and amplified by limited rounds of PCR. The amplified material was cleaned using AMPure XP Beads (Beckman Coulter). The final library was profiled on a 4200 TapeStation (Illumina) and quantified by RT-PCR using NGS Library Quantification Kit (Roche/Kapa Biosystems) on a StepOnePlus RT-PCR workstation (Thermo Fisher Scientific/ABI).

Single-cell RNA-seq libraries were prepared using Chromium Next GEM Single Cell 3′ Reagent Kit v 3.1 with dual indexes (10x Genomics). Cells were counted and then processed through the Chromium Controller (10x Genomics) to produce amplified cDNAs with cell-specific tags. The cDNA was enzymatically fragmented and then size-selected using solid-phase reversible immobilization (SPRI) beads. Adapters were ligated to the cDNA, and samples were indexed using indexed amplification primers. Libraries were profiled on a 4200 TapeStation (Agilent) and quantified using RT-PCR with NGS Library Quantification Kit (Roche/Kapa Biosystems) on a StepOnePlus RT-PCR workstation (Thermo Fisher Scientific/ABI). Libraries were sequenced on a NovaSeq 6000 (Illumina) running RTA v3.4.4. BCL files were demultiplexed using bcl2fastq v2.20.0.422 (Illumina).

A custom reference genome was constructed using GRCm38 (Ensembl) and the reporter gene dLNGFR with genome annotations from GRC38.89.gtf. Briefly, the dLNGFR FASTA file was added to the end of the GRCm38 FASTA as a virtual chromosome using the Linux cat command. The GTF file was manually edited to add dLNGFR annotation. The FASTA and GTF files were then used to construct a reference genome using the STAR genomeGenerate runMode command. Cell Ranger 7.1.0 uses STAR 2.7.2a aligner for building and aligning to the reference genome. Bulk RNA libraries were aligned using STAR 2.5.0a.

Bulk RNA-seq analysis

Bulk RNA-seq analysis was performed with the DESeq2 (v1.42.0) R package (31). Raw count data were normalized using the variance stabilization transformation function, and differential expression was computed between sets of triplicate samples using DESeq2 with LFCshrinkage via apeglm (32).

scRNA-seq analysis

All analyses conducted in this study were carried out using R software (version R 4.1.2) and RStudio (version 2023.12.0+369). Single-cell RNA-seq data underwent analysis utilizing the Seurat R package (version 4.3) to convert matrix files into Seurat objects (33). We excluded cells of substandard quality (minimum number of unique genes = 350 and maximum mitochondrial RNA = 15%). After hashtag demultiplexing (Seurat), doublets were removed, and the data from the two independent experiments were merged. Both singlets and hashtag negative cells were retained for analysis, with hashtag negative cells used just for dimensionality reduction and unsupervised clustering but excluded from differential expression analysis. Unique molecular identifier (UMI) counts were normalized with the SCTransform approach to correct for depth of sequencing between cells (34). The total number of the quality control–passed cells in this study was 27,219. The top 2,000 highly variable genes were selected for dimensionality reduction using principal component analysis and uniform manifold approximation and projection. Initial cell clustering was performed with the top N principal components using the Louvian algorithm (“FindClusters” function) across a sweep of resolutions. We computed the silhouette width and root mean square error for each clustering resolution and used the value that maximized silhouette width and minimized root mean square error (33). The “FindAllMarkers” function was utilized to identify marker genes upregulated in each cluster and define their identity, resulting in the identification of 23 clusters, including 7 stromal clusters, 5 epithelial clusters, and 9 immune clusters. This approach failed to resolve DCs and B cells, so the cluster containing both was separated and reprocessed (variable feature identification and principal component analysis) to identify them, increasing the total number of clusters to 24. Additionally, the Seurat function “FindAllMarkers” was used to identify differentially expressed genes (DEG) for each cluster."

We used three metrics to differentiate between epithelial clusters composed of endogenous cells and the implanted MOC2 SCs; (i) imputed copy-number variation (CNV) distance from diploid, (ii) correlation to bulk MOC2 RNA-seq profiles, and (iii) expression of human transgene dLNGFR. For CNV analysis, we subset the scRNA-seq data to just the cells with either a “stromal” or “epithelial” lineage label and used the CopyKAT R package (v1.1.0) to infer CNVs from gene expression (35). The CopyKat algorithm was applied with “stromal” cells set as expected diploid using the “mm10” genome and default parameters. To identify outlier cells, we computed the distance from diploid as the logarithm of summed squared distance from diploid. To compute expression correlation to the bulk profile, we found the intersect of genes expressed in the scRNA-seq and bulk-RNAseq data and computed the single-cell Pearson correlation to mean expression bulk profiles for triplicate samples of paclitaxel-treated MOC2-25 (EV-proficient) and MOC2-27 (EV-deficient). For assessment of dLNGFR expression, this gene was added to our reference transcriptome prior to alignment, and dLNGFR was normalized with the SCTransform approach along with all endogenous mouse genes.

For intercellular communication analysis and network visualization of scRNA-seq data, we used the R package “CellChat” (v1.6.1), a public repository of ligands, receptors, cofactors, and their interactions (36). We utilized CellChatDB.mouse, a database containing literature-supported ligand–receptor interactions in mice. This database distinguishes between three classes of ligand–receptor interactions: secreted signaling, ECM–receptor, and cell–cell contact interactions. Seurat normalized gene expression was used to create cell chat objects from the pooled tumors for each condition (senEV-proficient and senEV-deficient), and then these interactions were compared to identify differences across the conditions. To identify signaling pathways between senEVs and their microenvironment, we focused our analysis specifically on membrane-bound ligand–receptor interactions (i.e., cell–cell contact interactions). A heatmap was used to detail the differential interaction numbers and strengths between the different subsets identified within the senescence microenvironment. Chord diagrams were used to focus on top membrane-bound ligand–receptor interactions after specifying the sender and recipients of interest. A bubble plot was used to compare communication probabilities mediated by specific ligand–receptor pairs (based on proteomic data) toward any immune cell subset. SCs and stem-like SCs displayed overlapping interaction partners, including themselves, IFN-activated neutrophils, basal epithelial cells, and Schwann cells, when expressing Rab35-WT. For this reason, we considered them as a single subset in following analyses.

To unbiasedly define SASP factors, we filtered the BioMart database (37) for genes containing a signal peptide and lacking a transmembrane domain.

CD4 T-cell depletion

To deplete CD4 T cells, 200 µg of anti-CD4 antibody (GK1.5) was administered intraperitoneally once per week. The first administration was performed a day before challenge with senescent MOC2 cells. More than 99% of CD4 T cells were depleted in circulation, as confirmed 4 days after the first GK1.5 treatment (using a different anti-CD4 clone).

Statistics

Statistical analyses of the results were performed using appropriate tests in GraphPad Prism version 10. Proteomic and RNA-seq data were analyzed using R. Adjusted P values <0.05 were defined as significant. Biological and technical replicates are reported for each experiment.

Data availability

Sequencing data has been deposited to Gene Expression Omnibus under accession numbers GSE262139 (bulk RNA-seq) and GSE262141 (scRNA-seq). Other data generated in this study are available upon request from the corresponding author.

A novel engraftment-based senescence model enables study of the senescence microenvironment

SenEVs are a novel and largely underappreciated component of the senescence secretome (38). To enable in vivo assessment of senEVs, we developed an engraftment-based approach that allows us to genetically control EV release specifically in SCs. We induced senescence in vitro by treating murine squamous cell carcinoma cell lines (MOC2 and mEER+) with chemotherapeutic agents (Fig. 1; Supplementary Fig. S1, respectively). We chose cisplatin and paclitaxel because they are standard-of-care for head and neck squamous cell carcinoma (HNSCC; ref. 39). To optimize the dose, we tested different concentrations of paclitaxel (0–750 nmol/L) and cisplatin (0–35 µmol/L) in a combinatorial manner, with each condition replicated in quadruplicate (Fig. 1A). After 48 hours of treatment, we quantified cellular senescence by measuring the senescence marker SA-β-Gal activity (40). A defining feature of cellular senescence is the exit from the cell cycle while maintaining cell viability. To confirm that paclitaxel treatment generates bona fide SCs, we assessed proliferation (by BrdU incorporation), apoptosis (by Annexin V), and cell death (by 7-AAD), or lack thereof. These experiments determined the optimal doses of paclitaxel and cisplatin to maximize induction of cellular senescence by minimize proliferation without inducing significant cell loss by apoptosis or cell death (Fig. 1A; Supplementary Fig. S1). Although maximum senescence was induced when treating MOC2 or mEER+ with 600 nmol/L paclitaxel and 4 µmol/L cisplatin, the addition of cisplatin increased cell death and reduced cell recovery, which would have a negative impact on the model. Therefore, cisplatin was excluded from the senescence-inducing treatment in subsequent experiments. Bulk RNA-seq data from cultured senescent and proliferating cells (MOC2) confirmed increased expression of the senescence markers Cdkn1a (p21), Cdkn2a (p16), and Glb1 (SA-β-Gal) and decreased expression of the proliferation marker Ki67 in paclitaxel-treated versus untreated MOC2 (Fig. 1B). SCs exhibit an enlarged and flattened morphology (41). SA-β-Gal activity correlated with morphologic changes of the cells after exposure to chemotherapeutic drugs (Supplementary Fig. S1). These results indicate that 600 nmol/L paclitaxel triggers treatment-induced senescence in vitro while preserving cell recovery.

Figure 1.

A novel engraftment-based senescence model enables study of the senescence microenvironment. A, Optimization of in vitro senescence induction in MOC2 squamous carcinoma cells through 48-hour treatment with specified concentrations of paclitaxel and cisplatin. Ideal conditions ensured elevated senescence levels (quantified by increased SA-β-Gal staining and reduced BrdU incorporation), minimized apoptosis/cell death (measured by Annexin V and 7-AAD staining, respectively), and maximization of cell recovery (quantified by cell counting; n = 4). The first column on the left of each heatmap has 0 nmol/L paclitaxel and therefore represents cisplatin only. The bottom row of each heatmap has 0 µmol/L cisplatin and therefore represents paclitaxel only. The rest of the heatmap shows the combination of the two chemotherapeutic agents. B, Differential expression of senescence [Cdkn1a (p21), Cdkn2a (p16), and Glb1 (SA-β-Gal)] and proliferation (Ki67) markers in paclitaxel-treated (Pac; 600 nmol/L) vs. untreated (UT) MOC2 cells in vitro analyzed by bulk RNA-seq (n = 3). C, Inhibition of SC (MOC2) population expansion by continuous 600 nmol/L paclitaxel treatment for 5 days with fresh media replenishment every 48 hours (n = 3). Error bars, SD. Statistical analysis conducted using repeated-measure one-way ANOVA with multiple testing correction. D, Inhibitory effect of paclitaxel treatment on MOC2 proliferation over a 24-hour period, resulting in 15% BrdU incorporation (∼7-fold reduction from 100% maximum BrdU incorporation; n = 3). Error bars, SD. Max, maximum. E, Effect of paclitaxel treatment on BrdU incorporation rate in MOC2 cells, demonstrating an 8-fold reduction following a 2-hour BrdU pulse (n = 3). F, Persistence of the senescent phenotype in vitro, as evidenced by unaltered SA-β-Gal activity for 7 days after paclitaxel removal (n = 3). G, Delay in tumor formation by paclitaxel-treated senescent MOC2 cells implanted orthotopically in the dermis of mice (1M per mouse, n = 10). H, Flow cytometric analysis of immune cell subsets infiltrating tumor or senescent MOC2 cell implants on day 3 post-challenge with 5E5 cells implanted orthotopically (n = 5). Statistical analysis conducted using nonparametric one-way ANOVA (Kruskal–Wallis test) with multiple testing correction. *, P ≤ 0.05; **, P ≤ 0.01; ***, P ≤ 0.001.

Figure 1.

A novel engraftment-based senescence model enables study of the senescence microenvironment. A, Optimization of in vitro senescence induction in MOC2 squamous carcinoma cells through 48-hour treatment with specified concentrations of paclitaxel and cisplatin. Ideal conditions ensured elevated senescence levels (quantified by increased SA-β-Gal staining and reduced BrdU incorporation), minimized apoptosis/cell death (measured by Annexin V and 7-AAD staining, respectively), and maximization of cell recovery (quantified by cell counting; n = 4). The first column on the left of each heatmap has 0 nmol/L paclitaxel and therefore represents cisplatin only. The bottom row of each heatmap has 0 µmol/L cisplatin and therefore represents paclitaxel only. The rest of the heatmap shows the combination of the two chemotherapeutic agents. B, Differential expression of senescence [Cdkn1a (p21), Cdkn2a (p16), and Glb1 (SA-β-Gal)] and proliferation (Ki67) markers in paclitaxel-treated (Pac; 600 nmol/L) vs. untreated (UT) MOC2 cells in vitro analyzed by bulk RNA-seq (n = 3). C, Inhibition of SC (MOC2) population expansion by continuous 600 nmol/L paclitaxel treatment for 5 days with fresh media replenishment every 48 hours (n = 3). Error bars, SD. Statistical analysis conducted using repeated-measure one-way ANOVA with multiple testing correction. D, Inhibitory effect of paclitaxel treatment on MOC2 proliferation over a 24-hour period, resulting in 15% BrdU incorporation (∼7-fold reduction from 100% maximum BrdU incorporation; n = 3). Error bars, SD. Max, maximum. E, Effect of paclitaxel treatment on BrdU incorporation rate in MOC2 cells, demonstrating an 8-fold reduction following a 2-hour BrdU pulse (n = 3). F, Persistence of the senescent phenotype in vitro, as evidenced by unaltered SA-β-Gal activity for 7 days after paclitaxel removal (n = 3). G, Delay in tumor formation by paclitaxel-treated senescent MOC2 cells implanted orthotopically in the dermis of mice (1M per mouse, n = 10). H, Flow cytometric analysis of immune cell subsets infiltrating tumor or senescent MOC2 cell implants on day 3 post-challenge with 5E5 cells implanted orthotopically (n = 5). Statistical analysis conducted using nonparametric one-way ANOVA (Kruskal–Wallis test) with multiple testing correction. *, P ≤ 0.05; **, P ≤ 0.01; ***, P ≤ 0.001.

Close modal

We next quantified the proliferative ability of SCs to evaluate the durability of the paclitaxel-induced senescent phenotype. To this end, we analyzed the in vitro proliferation rate of MOC2 cells treated with 600 nmol/L paclitaxel (Fig. 1C–E). Continuous paclitaxel treatment blocked cell proliferation, with only a ∼10% cell loss on day 3 as compared with days 1 to 2 (Fig. 1C). To quantify the residual proliferative potential of SCs, we measured BrdU incorporation at saturation and after a short pulse while culturing SCs in presence of paclitaxel (Fig. 1D and E). Over a 24-hour period, BrdU incorporation plateaued at ∼15%, suggesting that paclitaxel alone was sufficient to reduce maximum proliferation of MOC2 ∼7-fold (down from 100% BrdU incorporation). To assess the rate of proliferation, we compared BrdU incorporation between paclitaxel-treated and untreated MOC2 cells after a short pulse with BrdU (2 hours; Fig. 1E). We observed an 8-fold reduction in the rate of BrdU incorporation, from 40% to 5%. These analyses indicate that paclitaxel treatment alone can reduce the residual proliferation potential of a fast-growing squamous carcinoma model by ∼2% (see “Materials and Methods”).

Because we aimed to implant SCs in vivo, without continuing paclitaxel treatment in mice, we assessed whether in vitro–generated SCs retain the senescence phenotype after removal of paclitaxel. To this end, we simulated in vivo conditions by removing paclitaxel after 2 days of treatment and quantified the persistence of senescence over time (Fig. 1F). We determined that over 7 days, SA-β-Gal activity did not decrease and remained present in ∼80% of the cells. These results suggest that the senescent state may be retained in vivo. To confirm this, we measured the tumor formation ability of our SC preparation in immunocompetent syngeneic mice (Fig. 1G). After implanting the same numbers of viable or senescent MOC2 cells, we observed that SCs were unable to form tumors for at least 2 weeks, indicating that paclitaxel treatment induces a functional state of cellular senescence. These results indicate that although our senescence model does not fully recapitulate all the events that take place in patients before and during recurrence (i.e., mutagenesis, immune escape, tumor growth, surgery, chemotherapy, and recurrence), it can faithfully model the final steps, and therefore it represents a useful tool to study tumor recurrence.

The removal of SCs is mediated by the immune system (42). The immune response against SCs is different from that elicited within the tumor microenvironment (43). To test whether our engraftment model of senescence recapitulates this difference, we studied the immune infiltrate recruited within 3 days of orthotopic injection of SCs (Fig. 1H). When we compared senescence and tumor models, we observed significant differences in their immune composition. As expected (15, 16), the fractions of T and NK cells were significantly increased within senescent tissues 3 days after challenge. Although not statistically significant, we observed a trend in macrophage subsets, with a relative increase in the fraction of MHC-II CD80+ CD206+ macrophages and a corresponding decrease in the MHC-II+ CD80 CD206 macrophages in presence of SCs. On the contrary, neutrophils and regulatory (FoxP3+) and type-1 helper (Tbet+) T cells were similarly recruited by both tumor cells and SCs. These results suggest that our senescence model recapitulates the immune responses observed in different in vivo models of cellular senescence (15).

SenEVs differ from tumor-derived EVs in morphology and protein composition

EVs are key mediators of intercellular communication (7). SenEVs may carry unique signals that impact the recruitment of immune cells (Fig. 1H; refs. 8, 9). To explore the role of senEVs in senescence surveillance (i.e., the immune-mediated removal of SCs; ref. 15), we characterized the protein composition of senEVs in our model system. We isolated EVs from senescent and proliferating squamous carcinoma cells using ultracentrifugation and compared their protein content, quantities, and size by spectrometry, NTA, and electron microscopy (Fig. 2A–C; Supplementary Fig. S2A). We found that the protein content released into EVs from SCs was ∼6-fold higher compared with nonsenescent squamous carcinoma cells (Fig. 2A). These results may be explained by either a higher rate of senEV release, larger average senEV size, higher density of protein cargo, or a combination of the above. To discriminate between these alternatives, we performed NTA. Whereas EV numbers were similar, we observed a 16% increase in median diameter (Fig. 2B), which corresponds to a 56% increase in EV volume (assuming a spherical shape). Electron microscopy analysis confirmed that senEVs are 16% larger than tumor EVs in both MOC2 and mEER+ cells (Fig. 2C). Immunoblot analysis for a pan-EV marker (flotillin-1; ref. 44) confirmed the presence of bona fide EVs (Fig. 2D). These findings suggest that senEVs exhibit a larger size and carry a greater protein payload in comparison with EVs from proliferating squamous carcinoma cells.

Figure 2.

SenEVs differ from tumor-derived EVs in morphology and protein composition. A, Increased EV protein content per million senescent MOC2 cells (600 nmol/L paclitaxel for 48 hours), determined to be 6-fold higher compared with nonsenescent cells (control), as assessed by NanoDrop (n = 3). EV isolation conducted from cell supernatant utilizing ultracentrifugation. Data are representative of both MOC2 and mEER+ cell lines. B, NTA of EVs from senescent MOC2 cells: A 16% increase in median diameter corresponding to a 56% increase in volume compared with EVs from proliferating cells (n = 5). No differences in EV numbers between proliferating (Ctrl) and SC-derived (paclitaxel, Pac) EVs (rightmost graph). C, Electron microscopy (EM) analysis of senescent EVs from both MOC2 and mEER+ cell lines confirms that senEVs are 16% larger than tumor-derived EVs. Data from >10 images per condition. D, Immunoblot for the pan-EV marker flotillin-1. E, Differential proteomic analysis of EVs isolated from senescent and proliferating squamous carcinoma cells reveals that 150 and 120 proteins exhibit significantly altered expression levels in EVs derived from SCs (MOC2 and mEER+, respectively) compared with EVs from proliferating cells (tEVs). FC, fold change. F, Pathway analysis (DAVID) of enriched proteins in senescent EVs from senescent MOC2 revealed significant associations with core cellular functions such as ribosomes, spliceosomes, DNA replication, and type-1 immunity (P adj < 0.01). G, Comparison of proteins enriched in SC-derived EVs (as compared with EVs from proliferating cells) shows a 30% overlap (31 out of 109) between two distinct SC types. Statistical tests were conducted utilizing the Mann–Whitney test or one-way ANOVA. Proteomic analysis employed multiple comparisons correction using the Benjamin, Krieger, and Yekkutieli method. *, P ≤ 0.05; **, P ≤ 0.01; ****, P ≤ 0.0001.

Figure 2.

SenEVs differ from tumor-derived EVs in morphology and protein composition. A, Increased EV protein content per million senescent MOC2 cells (600 nmol/L paclitaxel for 48 hours), determined to be 6-fold higher compared with nonsenescent cells (control), as assessed by NanoDrop (n = 3). EV isolation conducted from cell supernatant utilizing ultracentrifugation. Data are representative of both MOC2 and mEER+ cell lines. B, NTA of EVs from senescent MOC2 cells: A 16% increase in median diameter corresponding to a 56% increase in volume compared with EVs from proliferating cells (n = 5). No differences in EV numbers between proliferating (Ctrl) and SC-derived (paclitaxel, Pac) EVs (rightmost graph). C, Electron microscopy (EM) analysis of senescent EVs from both MOC2 and mEER+ cell lines confirms that senEVs are 16% larger than tumor-derived EVs. Data from >10 images per condition. D, Immunoblot for the pan-EV marker flotillin-1. E, Differential proteomic analysis of EVs isolated from senescent and proliferating squamous carcinoma cells reveals that 150 and 120 proteins exhibit significantly altered expression levels in EVs derived from SCs (MOC2 and mEER+, respectively) compared with EVs from proliferating cells (tEVs). FC, fold change. F, Pathway analysis (DAVID) of enriched proteins in senescent EVs from senescent MOC2 revealed significant associations with core cellular functions such as ribosomes, spliceosomes, DNA replication, and type-1 immunity (P adj < 0.01). G, Comparison of proteins enriched in SC-derived EVs (as compared with EVs from proliferating cells) shows a 30% overlap (31 out of 109) between two distinct SC types. Statistical tests were conducted utilizing the Mann–Whitney test or one-way ANOVA. Proteomic analysis employed multiple comparisons correction using the Benjamin, Krieger, and Yekkutieli method. *, P ≤ 0.05; **, P ≤ 0.01; ****, P ≤ 0.0001.

Close modal

To determine the protein composition of senEVs, we performed proteomic analyses on EVs isolated from senescent and proliferating MOC2 and mEER+ cells (Fig. 2E–G; Supplementary Table S2). We found 150 and 120 proteins significantly different in senEVs from MOC2 and mEER+ cells, respectively (as compared with EVs from proliferating squamous carcinoma cells; Fig. 2E). Pathway analysis showed that proteins enriched in senEVs were significantly associated with core cellular functions, including ribosomes, spliceosomes, and DNA replication but also with type-1 immunity (systemic lupus erythematosus; Fig. 2F). When we determined the common proteins enriched in senEVs from both cell lines (as compared with EVs from the respective proliferating cells), we found a 30% overlap (Fig. 2G). Among these proteins, ribosomal proteins were the largest family.

Inhibition of EV release does not impact the SASP

Several studies have reported a dual role for the SASP in both promoting and inhibiting cancer progression and recurrence (3, 4, 15, 16). These conflicting results may derive from the presence of different amounts and types of senEVs in SASP studies. To distinguish the role of senEVs in senescence surveillance from that of the SASP, we aimed to specifically inhibit senEV release. Inhibition of Rab27a is a commonly used method to block EV release (45). To test whether inhibition of Rab27a would stop release of EVs in the context of senescence, we used siRNA oligoes to successfully inhibit expression of Rab27a in SCs, as measured by qPCR (Supplementary Fig. S2B). However, two independent methods to assess inhibition of EV release from SCs showed that EVs were instead significantly increased (Supplementary Fig. S2C and S2D). These observations, together with the fact that the total protein content of senEVs was significantly decreased by Rab27a knockdown (Supplementary Fig. S2E), suggest that Rab27a in SCs may contribute to loading EV cargo proteins and that SCs attempt to compensate the diminished EV cargo by releasing higher numbers of senEVs.

We previously validated an alternative genetic tool to inhibit EV release from parental cells (1719). In this approach, Rab35-DN is expressed in parental cells. As control, Rab35-WT is used. To confirm that expression of Rab35-DN efficiently and specifically reduces EV release in both proliferating and senescent MOC2 and mEER+ cells, we performed immunoblot assays for the pan-EV marker flotillin-1 on purified EVs. We observed almost complete absence of EVs in both proliferating and senescent MOC2 expressing Rab35-DN (Fig. 3A; Supplementary Fig. S3A and S3B). We further validated inhibition of EV release using an independent technique and confirmed a 4- to 6-fold decrease of EV release from SCs expressing Rab35-DN (Fig. 3B). Comparison of EV diameters using membrane-intercalating fluorescent dyes (which allows distinguishing membrane-bound particles from nonvesicular particles) suggests that, notwithstanding some variability due to low EV numbers, the increase in size is associated with the senescence phenotype and not with the expression of Rab35-DN (Supplementary Fig. S3C). These results indicate that the profound inhibition of senEV release is suitable for in vivo functional studies.

Figure 3.

Expression of Rab35-DN does not inhibit the SASP. A, Effective and targeted reduction of EV release detected in both proliferating and senescent MOC2 cells expressing Rab35-DN using immunoblot assay for the pan-EV marker flotillin-1. EV preparations were normalized by cell number. UT, untransduced. B, Detection of reduced EV release in both proliferating and senescent MOC2 cells expressing Rab35-DN utilizing NTA, which assesses EV numbers and size via light scatter (left) and fluorescence (right) measurements. C, SASP factors are not decreased by Rab35-DN expression, as evidenced by quantification of 40 secreted factors using mouse cytokine array on the senescent MOC2 cell culture supernatant. D, Bulk RNA-seq of Rab35-WT and Rab35-DN senescent MOC2 cells (left) and of proliferating and senescent MOC2 cells (right) confirms that Rab35-DN expression has minimal impact on the global SASP (red), defined as genes containing a signal peptide and lacking a transmembrane domain (left), and that changes in gene profile after Rab35-DN expression are minor compared with changes induced by senescence stimuli (right). Significance level set at adjusted P <0.01. Statistical tests by two-way ANOVA with multiple testing correction. *, P ≤ 0.05. FC, fold change.

Figure 3.

Expression of Rab35-DN does not inhibit the SASP. A, Effective and targeted reduction of EV release detected in both proliferating and senescent MOC2 cells expressing Rab35-DN using immunoblot assay for the pan-EV marker flotillin-1. EV preparations were normalized by cell number. UT, untransduced. B, Detection of reduced EV release in both proliferating and senescent MOC2 cells expressing Rab35-DN utilizing NTA, which assesses EV numbers and size via light scatter (left) and fluorescence (right) measurements. C, SASP factors are not decreased by Rab35-DN expression, as evidenced by quantification of 40 secreted factors using mouse cytokine array on the senescent MOC2 cell culture supernatant. D, Bulk RNA-seq of Rab35-WT and Rab35-DN senescent MOC2 cells (left) and of proliferating and senescent MOC2 cells (right) confirms that Rab35-DN expression has minimal impact on the global SASP (red), defined as genes containing a signal peptide and lacking a transmembrane domain (left), and that changes in gene profile after Rab35-DN expression are minor compared with changes induced by senescence stimuli (right). Significance level set at adjusted P <0.01. Statistical tests by two-way ANOVA with multiple testing correction. *, P ≤ 0.05. FC, fold change.

Close modal

A caveat of interfering with vesicular trafficking via Rab35 is that some SASP factors may be decreased as well, thereby confounding the interpretation of in vivo data. To test whether Rab35-DN expression in SCs inhibits SASP components, we quantified 40 different secreted factors by cytokine array on SC culture supernatants (Fig. 3C). Of these 40 mediators, 8 were expressed at detectable levels (G-CSF, GM-CSF, CXCL10, CXCL1, M-CSF, CCL2, CXCL2, and TIMP1), and are all well-established SASP factors (46). Surprisingly, two of these eight SASP factors (CCL2 and CXCL2) were expressed at higher levels in Rab35-DN SCs. An additional six mediators (IL1α, IL5, IL17, CCL5, CXCL12, and TNFα) were expressed at low but detectable levels and were unaffected by expression of Rab35-DN. To confirm these observations, we performed bulk RNA-seq of proliferating and senescent MOC2 from cultures (Fig. 3D; Supplementary Fig. S3D; Supplementary Table S3). We focused our analysis on those genes exclusively featuring signal peptides and lacking transmembrane domains, categorizing them as secretory genes. The analysis of DEGs between SCs expressing Rab35-DN and SCs expressing Rab35-WT revealed that most secretory genes were not differentially expressed, with only three being downregulated more than 2-fold (Ccl28, Inhbb, and Chrd; Fig. 3D). Noting that these three genes are not commonly found among SASP factors, these findings indicate that expression of Rab35-DN does not impede the release of SASP factors.

SenEVs recruit antigen-presenting cells and are necessary to inhibit cancer recurrence

Whereas the role of the SASP in intercellular communication is well established (6), the specific role of senEVs in communicating with the SC microenvironment is still largely unclear. Although current approaches using the isolation and injection of exogenous EVs (derived from cell cultures or biofluids) in animal models allow precise control over pharmacokinetic and pharmacodynamic parameters, it remains uncertain whether the insights gained from exogenously administered EVs sufficiently address key aspects of EV biology (47). To overcome these limitations, we took advantage of our engraftment-based senescence model to investigate native senEVs, i.e., senEVs released by SCs living within a tissue. To this end, we challenged mice orthotopically with Rab35-WT or Rab35-DN SCs (Fig. 4A). We observed that in the absence of senEVs, tumor recurrence was significantly accelerated, as compared with mice receiving Rab35-WT SCs (Fig. 4A). To assess whether expression of Rab35-DN interferes with paclitaxel-induced senescence, we compared SA-β-Gal activity and cell expansion in vitro (Supplementary Fig. S4A and S4B). We found that Rab35 manipulation did not influence senescence induction (Supplementary Fig. S4A) and did not rescue the blockade of cell expansion at the population level (Supplementary Fig. S4B), pointing to the fact that inhibition of senEV release does not interfere with induction of senescence by paclitaxel treatment. To test whether inhibition of tumor recurrence by EVs is specific to senescence, we tested whether expression of Rab35-DN impacts in vivo growth of untreated, nonsenescent MOC2 cells. As compared with MOC2 expressing Rab35-WT, we observed no differences in tumor growth of Rab35-DN expressing MOC2 (Supplementary Fig. S4C), indicating that the inhibition of disease progression is a property specific of senEVs. These findings suggest that the release of senEVs inhibits tumor recurrence, possibly via non–cell-autonomous mechanisms.

Figure 4.

SC-derived EVs recruit APCs and are necessary and sufficient to inhibit cancer recurrence. A, Rab35-DN expression in SCs promotes tumor recurrence in C57BL/6J mice challenged orthotopically (dermis or tongue) with 5E5 SCs (three independent experiments, two different SC types, n = 10). B and C, Rab35-DN expression in SCs promotes accumulation of senescent MOC2 cells 3 days after engraftment, as measured by flow cytometry (n = 5; B), and by single-cell sequencing (n = 3; C), utilizing the transgenic marker dLNGFR. Data are representative of two independent experiments. D, Quantification of immune cell infiltrates by flow cytometry 3 days after implantation of Rab35-WT or Rab35-DN senescent MOC2 cells (data are representative of two independent experiments; see Supplementary Fig. S4D). E, Administration of senEVs to mice challenged with Rab35-DN senescent MOC2 cells reverts tumor recurrence (N = 10). Statistical tests by one-way ANOVA, two-way ANOVA, or the Mann–Whitney test. Multiple testing correction applied (Sidak). *, P ≤ 0.05; **, P ≤ 0.01; ***, P ≤ 0.001; ****, P ≤ 0.0001.

Figure 4.

SC-derived EVs recruit APCs and are necessary and sufficient to inhibit cancer recurrence. A, Rab35-DN expression in SCs promotes tumor recurrence in C57BL/6J mice challenged orthotopically (dermis or tongue) with 5E5 SCs (three independent experiments, two different SC types, n = 10). B and C, Rab35-DN expression in SCs promotes accumulation of senescent MOC2 cells 3 days after engraftment, as measured by flow cytometry (n = 5; B), and by single-cell sequencing (n = 3; C), utilizing the transgenic marker dLNGFR. Data are representative of two independent experiments. D, Quantification of immune cell infiltrates by flow cytometry 3 days after implantation of Rab35-WT or Rab35-DN senescent MOC2 cells (data are representative of two independent experiments; see Supplementary Fig. S4D). E, Administration of senEVs to mice challenged with Rab35-DN senescent MOC2 cells reverts tumor recurrence (N = 10). Statistical tests by one-way ANOVA, two-way ANOVA, or the Mann–Whitney test. Multiple testing correction applied (Sidak). *, P ≤ 0.05; **, P ≤ 0.01; ***, P ≤ 0.001; ****, P ≤ 0.0001.

Close modal

To better understand the initial host responses to cellular senescence, we studied our engraftment-based model 3 days after challenge with SCs (i.e., before tumor formation). We took advantage of a genetic marker (dLNGFR) expressed along with Rab35 variants to track SCs in vivo (Fig. 4B). We found that interfering with senEV release led to 4-fold higher accumulation of SCs on day 3, as compared with Rab35-WT SCs (Fig. 4B). This observation was confirmed by two independent single-cell sequencing experiments, also performed on day 3 after SC implantation (Fig. 4C). To test whether senEVs have a role in SC removal through recruitment of immune cells, we repeated the experiment above and quantified the immune infiltrate early after SC engraftment by flow cytometry (Fig. 4D; Supplementary Fig. S4D). Among the immune subsets we queried were CD4 and CD8 T cells, their type-1 effector subsets expressing T-bet, T regulatory cells, NK and NKT cells, myeloid cells (including neutrophils, DCs, macrophages, and their subsets), and B cells. We observed that in the presence of senEVs, specific immune cell subsets were significantly increased (∼50%; P < 0.05), including infiltrating DCs, B cells, and MHC-II+ macrophages (Fig. 4D; Supplementary Fig. S4D). These immune cell subsets were related only by the fact that they are all professional antigen-presenting cells (APC), indicating that senEVs are necessary to recruit APCs and that the absence of senEVs increases accumulation of SCs in tissues.

To test whether senEVs are necessary and sufficient to counter tumor recurrence, we performed a complementation experiment in which we reintroduced senEVs along with SCs unable to release them. To this end, we implanted senescent MOC2 cells expressing Rab35-DN intradermally in two groups of mice. One group received the SCs mixed with 100 µg of senEVs and additional intradermal administrations of senEVs (100 µg) every 5 days near the SC injection site. We selected the amount of senEVs based on how many EVs SCs release daily in vitro. Control mice received the same treatment with PBS (Fig. 4E). Strikingly, provision of senEVs restored tumor recurrence to levels observed in untransduced SCs (Fig. 1G) and in SCs expressing Rab35-WT (Fig. 4A). Together, these results indicate that the presence of senEVs is sufficient and necessary to counter tumor recurrence.

Single-cell sequencing identifies three distinct subsets of SCs in vivo

To better understand the signals that senEVs deliver to immune cells during senescence surveillance, we profiled the senescence microenvironment at the single-cell level, 3 days after engraftment of Rab35-WT or Rab35-DN SCs, and prior to tumor formation. To collect enough cells for profiling, we performed the experiment twice by pooling three mice per group every time. We corrected for differences in sequencing depth using SCTransform (48, 49), which successfully removed batch effects (Supplementary Fig. S5A). We classified three major lineages, namely, immune (Ptprc, S100a8, S100a9, and Cd14), epithelial (Epcam, Krt5, Krt19, and dLNGFR), and stromal (Pdgfra, Pdgfrb, and Pecam1; Supplementary Fig. S5B), based on the DEGs in each lineage compared with the others (Fig. 5A). To distinguish between different immune cell subsets, we subclustered immune lineage cells separately (Fig. 5B). This procedure allowed us to identify a diverse combination of lymphoid and myeloid immune cells recruited to the site of senescence, including two distinct subsets of macrophages characterized by expression of C1q and Spp1, DCs, monocytes, TH17 cells, four subsets of neutrophils, and B cells (Supplementary Table S4). The Spp1+ macrophage subset corresponds to the MHC-II+ macrophages identified by flow cytometry (Supplementary Fig. S6). These results confirmed the presence of the APC subsets we observed by flow cytometric analyses (Fig. 4D; Supplementary Fig. S4D).

Figure 5.

APCs recruited by senEVs require TH17 cells to inhibit tumor recurrence. A, Identification of three major cellular lineages within the senescence microenvironment through single-cell sequencing: immune, epithelial, and stromal cells, identified based on DEGs in each lineage (merged data of two independent experiments). B, Subclustering of cells belonging to the immune lineage distinguishes between different immune cell subsets. C and D, Identification of engrafted SC subset among epithelial lineage cells. Proliferating MOC2 cells, stem-like senescent MOC2 cells, and senescent MOC2 cells were defined based on markers of proliferation (Mki67), stemness (Sox9, Hmga2, Ly6a/Sca1, and Krt19), and senescence (Cdkn1a and Cdkn2a). Prolif., proliferative; Sen., senescent; Stem, stem-like. E, Visualizing cell–cell communications between SCs and immune cells mediated by membrane-bound ligand–receptor interactions using chord diagrams. Each sector in the chord diagrams is an arrow depicting the direction of the ligand–receptor signaling. Mac, macrophage; Mono, monocyte; Neu, neutrophil. F, Identification of membrane ligands specifically present on SC-derived EVs. Communications emanating from SCs that were enriched in presence of senEVs (differential membrane signaling between Rab35-WT vs. Rab35-DN senescent MOC2 cells; left) were intersected with the senEV proteome (right). This analysis was restricted to membrane-bound ligands. The resulting short-list (right) represents putative ligands specifically present on senEVs and not on the SC surface. Right, bubble plot identifies the cellular targets within the senescent microenvironment. G, Visualizing cell–cell communications between APCs and TH17 cells mediated by membrane-bound ligand–receptor interactions using chord diagrams. Each sector in the chord diagrams is an arrow depicting the direction of the ligand–receptor signaling. H, Tumor recurrence by Rab35-WT senescent MOC2 cells is increased in the absence of CD4 T cells to levels like those observed in Rab35-DN senescent MOC2 cells. I, Schematic of the proposed mechanism of EVs in senescence surveillance. *, P ≤ 0.05. I, Created in BioRender.com (Pucci, F.; https://app.biorender.com/illustrations/66c3d10ff323a174357c3c4e?slideId=3a8de68c-e682-4f15-87f2-1ea5986d7a89).

Figure 5.

APCs recruited by senEVs require TH17 cells to inhibit tumor recurrence. A, Identification of three major cellular lineages within the senescence microenvironment through single-cell sequencing: immune, epithelial, and stromal cells, identified based on DEGs in each lineage (merged data of two independent experiments). B, Subclustering of cells belonging to the immune lineage distinguishes between different immune cell subsets. C and D, Identification of engrafted SC subset among epithelial lineage cells. Proliferating MOC2 cells, stem-like senescent MOC2 cells, and senescent MOC2 cells were defined based on markers of proliferation (Mki67), stemness (Sox9, Hmga2, Ly6a/Sca1, and Krt19), and senescence (Cdkn1a and Cdkn2a). Prolif., proliferative; Sen., senescent; Stem, stem-like. E, Visualizing cell–cell communications between SCs and immune cells mediated by membrane-bound ligand–receptor interactions using chord diagrams. Each sector in the chord diagrams is an arrow depicting the direction of the ligand–receptor signaling. Mac, macrophage; Mono, monocyte; Neu, neutrophil. F, Identification of membrane ligands specifically present on SC-derived EVs. Communications emanating from SCs that were enriched in presence of senEVs (differential membrane signaling between Rab35-WT vs. Rab35-DN senescent MOC2 cells; left) were intersected with the senEV proteome (right). This analysis was restricted to membrane-bound ligands. The resulting short-list (right) represents putative ligands specifically present on senEVs and not on the SC surface. Right, bubble plot identifies the cellular targets within the senescent microenvironment. G, Visualizing cell–cell communications between APCs and TH17 cells mediated by membrane-bound ligand–receptor interactions using chord diagrams. Each sector in the chord diagrams is an arrow depicting the direction of the ligand–receptor signaling. H, Tumor recurrence by Rab35-WT senescent MOC2 cells is increased in the absence of CD4 T cells to levels like those observed in Rab35-DN senescent MOC2 cells. I, Schematic of the proposed mechanism of EVs in senescence surveillance. *, P ≤ 0.05. I, Created in BioRender.com (Pucci, F.; https://app.biorender.com/illustrations/66c3d10ff323a174357c3c4e?slideId=3a8de68c-e682-4f15-87f2-1ea5986d7a89).

Close modal

To identify the implanted SCs among the single cell pool, we used three different approaches: correlation with bulk RNA-seq profiles of SCs from culture, expression of the transgenic marker dLNGFR (coexpressed along with WT or DN Rab35), and CNV analysis (Supplementary Fig. S7). This examination identified three clusters of epithelial lineage cells as originating from the engrafted SCs (Fig. 5C). We named these three clusters based on their relative upregulation of markers related to “senescence,” “stemness,” and “proliferation” (Fig. 5D). Notably, all three SC clusters were found in both senEV-proficient and -deficient microenvironments, with SCs constituting the largest fraction (62%), followed by SCs expressing the stemness markers Sox9, Hmga2, Ly6a (Sca1), and Krt19 (Fig. 5D; refs. 5053). These results agree with reports showing that SCs can transform into cancer stem cells (5457) and highlight the usefulness of this model system to further study that process.

SenEVs display adhesion molecules that can bind to both APCs and T cells

The presence of senEVs had a significant impact on which signaling pathways were engaged in immune cells (Supplementary Fig. S6). To query immune cell–SC communications via EVs, we utilized the R package CellChat to impute likely ligand–receptor signaling from gene expression and distinguish secreted and membrane-bound ligand–receptor interactions (36). As compared with senEV-deficient, the senEV-proficient senescence microenvironment displayed significant changes in the strength and number of putative ligand–receptor interactions among the 24 cell types (Supplementary Fig. S8A). When expressing Rab35-DN, SCs and stem-like SCs mostly signaled to monocytes, T cells, neutrophils, B cells, luminal epithelial cells, and lymphatic endothelial cells (Supplementary Fig. S8A). These results indicate that the senescence microenvironment is highly affected by senEVs.

To focus on membrane-bound ligands (i.e., those on the surface of SCs and senEVs), we excluded SASP signaling (see Materials and Methods). When we assessed senescence microenvironments in the presence of senEVs, we observed that SCs signaled via membrane-bound ligands mainly with MHC-II+ APCs, i.e., B cells, DCs, and Spp1+ macrophages (Fig. 5E). In these interactions, the top membrane-bound ligand–receptor pair we identified was App-Cd74, followed by homotypic interactions via Cdh1. Strikingly, when senEVs were absent, the interaction partners of SCs shifted to neutrophil subsets (Fig. 5E). SC-Neu interactions in the absence of senEVs were based upon homotypic Pecam1 (CD31) interactions and Cdh5-Cdh1, followed by App-Cd74 and homotypic Esam interactions. Consistently, IFN-activated neutrophils and C1q+ macrophages (i.e., MHC-II–negative) were both increased in senEV-deficient senescence microenvironments (Supplementary Fig. S8B and S8C). These results suggest that senEVs promote SC engagement with APCs.

To distinguish between membrane-bound ligands on senEVs versus those on their parental cells (i.e., SCs), we took advantage of our senEV proteomic profile (Fig. 2E–G). First, we calculated the communications originating from SCs that were enriched in the presence of senEVs. This analysis was restricted to membrane-bound ligands (Supplementary Fig. S9A). Then, we intersected that list with the list of proteins detected on senEVs (Fig. 5F). The resulting short-list represented putative ligands specifically present on senEVs and not on the SC surface. This analysis yielded six membrane-bound ligands present on senEVs and engaging with cells within the senescence microenvironment: Cdh1, Dsg2, F11r, Ncam1, Ocln, and Pvr. These proteins are involved in adhesion and were found on both SCs and stem-like SCs. Interaction analysis suggested that senEVs are capable of binding to both MHC-II+ APC and TH17 cells through these adhesion-related proteins (Fig. 5F). These results suggest that senEVs may enhance the likelihood of endocytosis of SC antigens, potentially supporting T-cell engagement by APCs.

APCs recruited by senEVs require TH17 cells to inhibit tumor recurrence

Given the observed shift in SC–immune cell interactions away from APCs when senEVs are absent (Fig. 5E), and the resulting increase in tumor formation (Fig. 4A), we hypothesized that adaptive immune responses may have been impacted as well. To test this hypothesis, we queried the signaling pathways from APCs to TH17 cells (Fig. 5G; Supplementary Fig. S8D). We found that in senEV-proficient senescence microenvironments, TH17 cells have the potential to interact with all three MHC-II+ APC subsets via cell surface signaling (Fig. 5G). Moreover, TH17 cells expressed the activation marker CD69 (Supplementary Table S4), suggesting that they received antigen-specific stimulation (58). In senEV-deficient senescence microenvironments, DC cross-talk with TH17 cells used different membrane-bound ligand–receptor interactions (Fig. 5G), whereas B cells and Spp1+ macrophages displayed an immunoregulatory role by targeting TH17 cells with secreted signals (Supplementary Fig. S8D). These analyses suggest that APCs recruited by senEVs may prime and activate TH17 cells.

B–T cell coactivation is central to adaptive immune responses (59). In senEV-proficient senescence microenvironments, B-cell activation was restrained by the inhibitory coreceptors CD22 and Siglecg and by the release of Galectin-9 (Lgals9) from TH17 cells (Supplementary Fig. S8E and S8F; refs. 60, 61). B–T cell communication via soluble mediators was inferred to be mostly unidirectional, from TH17 cells to B cells (Supplementary Fig. S8F). These analyses suggest that TH17 cells may inhibit germinal center formation and that B cells may act mainly as APCs during the initial phases of senescence surveillance.

To experimentally validate the role of CD4 T cells in senescence surveillance, we performed antibody-mediated CD4 T-cell depletion (Supplementary Fig. S9B). CD4 depletion removes all CD4 T-cell subsets, including TH17 cells. We observed that CD4 T cells were necessary to control tumor recurrence in senEV-proficient senescence microenvironments (Fig. 5H). These results are in line with the reported role of this T-cell subset in senescence surveillance (15) and suggest that CD4 T cells are downstream of senEVs in the immune response to senescence (Fig. 5I).

Murine models have been instrumental in studying cellular senescence across various diseases and physiologic processes (62). They led to the identification of key senescence markers, understanding the role of senescence in disease progression, and exploring potential therapeutic interventions (1). However, challenges remain in identifying and characterizing SCs in vivo due to the lack of shared markers (63). Transgenic models of senescence are particularly susceptible to the uncertainty around SC biomarkers and require lengthy characterization of the expression patterns of the gene construct utilized. Nongenetic methods used to induce senescence in wild-type animals have systemic effects; for example, hydrodynamic (tail vein) injection of large volumes of plasmids expressing oncogenes (such as mutant Ras; refs. 15, 16) or administration of cytotoxic drugs such as bleomycin (64) cannot control which cell types become senescent. Of note, administration of cytotoxic/cytostatic drugs induces broad damage to tissues and alters immune cell development (65), which complicates the study of immune responses to SCs. In this study, we addressed these limitations by validating an engraftment-based senescence model that recapitulates therapy-induced senescence and tumor recurrence in patients with HNSCC. Although our model system does not fully recapitulate the pathogenesis of HNSCC, we think that our approach specifically models the recurrence stage, in which residual tumor cells accidentally left behind after surgery become senescent in vivo, during chemotherapy cycles.

Our approach allows to track the fate of implanted SCs in vivo by insertion of a genetic marker (dLNGFR) before senescence induction. The engraftment-based senescence model described here enables tracking of SC fates and functional studies via gene overexpression or knockdown; we took advantage of this feature to inhibit the release of senEVs. Lastly, our model allows us to tightly control which cells are induced into senescence without affecting immune cells.

The role of EVs in cellular senescence is currently understudied and poorly understood. Some studies reported that senEVs promote tumor cell proliferation in vitro (66, 67). Whether these effects are also in place in vivo and how the senescence microenvironment modulates tumor cell–autonomous effects is unclear. Our results suggest that the non–cell-autonomous effects of senEVs may overrule their direct signaling to (pre)cancerous cells, ultimately establishing senEVs as key to inhibit tumor formation and recurrence. This conceptual advance in our understanding of the senescence microenvironment is in line with the large body of knowledge accumulated around the impact of the tumor microenvironment in cancer (68). Our results suggest that senEVs are a distinct subset of EVs and differ from them in both morphology and protein content. Importantly, the unique set of proteins carried by senEVs may be involved in recruiting and activating immune cells.

In this study, we engrafted genetically engineered SCs at orthotopic locations to investigate the specific role of senEVs in senescence surveillance without interfering with the SASP. Although small changes in five soluble mediators (Ccl2, Cxcl2, Ccl28, Inhbb, and Chrd) were detected after Rab35-DN expression, our complementation experiment indicates that senEVs are sufficient and necessary to counter tumor recurrence. Nonetheless, TH17 cells expressed CCR2, which likely mediated their recruitment into the senescence microenvironment (69). These results suggest that senEVs and SASP factors play complementary but nonoverlapping roles in senescence surveillance.

The persistence of SCs in epithelial tissues is detrimental; it is commonly found in precursor lesions, leading to HNSCC in patients (70, 71), and it can drive cancer recurrence after chemoradiation regimens (3, 4). Our functional studies indicate that senEVs have a key role in the processes that mediate removal of SCs. This is important because our work shows that accumulation of SCs in the absence of senEV signaling leads to increased cancer recurrence. Our analyses suggest that specific immune cells (i.e., MHC-II+ APCs) need to be recruited to promptly clear SCs and avoid propagation of the senescence phenotype to neighboring cells. Our data suggest that a potential mechanism for this phenomenon may involve neutrophils, which were increased in the absence of senEVs and have been reported to spread senescence to neighboring cells (72). On the other hand, APCs like macrophages and DCs, along with IFN signaling, contribute to antitumor immunity (73). Our data show that APCs release cytokines (including Ccl2, Ccl6, and Ccl7) able to recruit TH17 cells via CCR2. Once in the senescence microenvironment, TH17 cells signaled with APCs via CD86-CD28, likely leading to T-cell activation. Consistently, depletion of CD4 T cells (the parental subset of TH17 cells) fully abolished senEV protective ability, suggesting that TH17 cells are downstream of senEVs in the immune response to senescence. Additional studies using depletion strategies more specific for TH17 cell will shed more light on their mechanism of action in senescence surveillance.

SenEVs carried several cell adhesion molecules that may mediate the uptake of SC antigens by MHC-II+ APCs and may strengthen the immunologic synapse between APCs and T cells during senescence surveillance. As compared with tumor microenvironments, we observed that NK and T cells were increased in senescence microenvironments, which is consistent with previous reports (15, 16). While the microenvironment of SCs expressing Rab35-DN quickly becomes a tumor microenvironment, the early time point chosen for analyses (3 days) means that both Rab35-WT and Rab35-DN groups largely represent senescent microenvironments. It is likely that NK and T cells may follow these initial immune responses. Further studies will clarify the time course of SC populations and immune surveillance during tumor recurrence. Overall, these results indicate that senEVs play a major role in senescence surveillance. The ability of SCs to release EVs complements the activity of the SASP in recruiting and activating distinct immune cell subsets for efficient removal of SCs. More work will be necessary to understand whether, after uptake, presentation of senEV-bound antigens and generation of antigen-specific immune responses are required to remove SCs.

Virus-like particles from endogenous retrovirus can be found within EV preparations obtained from several mouse tumor cell lines (74). Our proteomic analysis of senEVs detected high levels of envelop protein from murine leukemia virus, which was present in both senEVs and tumor EVs. Still, it is possible that virus-like particles may have contributed to the observed effects on tumor recurrence.

Although NTA may suggest that there may be a small percentage of EVs that are still released by SCs expressing Rab35-DN, immunoblot analysis for flotilin-1, a bona fide pan-EV marker (44), indicates a nearly complete (>95%) suppression of EV release. Unfortunately, NTA methods are affected by high background due to the inherent inability of scatter-based measurements to distinguish membrane-bound particles from nonvesicular particles and the potential presence of micelle-forming dyes during fluorescent-based measurements (44). We concluded that even if EV secretion was not completely abated, the level of inhibition achieved was suitable for in vivo functional studies.

Rab35 is involved in cytokinesis (75), which may impact residual tumor cells’ fitness. The reported role of Rab35 in human tumorigenesis is associated with constitutive-active mutants, which activate the PI3K/AKT pathway (76). Although this study used a DN mutant of Rab35 (i.e., a constitutively inactive GTPase that also interferes with the normal function of WT Rab35), and therefore our observations should not be affected by cell-autonomous mechanisms, there is also the possibility that DN mutants of Rab35 may possess tumorigenic potential. More studies are needed to clarify this point.

Single-cell sequencing data can display significant variability from batch to batch. To account and compensate for this variability, we repeated the single-cell analysis twice and performed data transformation, which is a process that removes batch effects. Nonetheless, residual differences may have impacted the results and masked smaller differences.

A limitation of this work is that, in patients, the immune system is also exposed to senescence-inducing stimuli like chemoradiation therapy. To better understand these dynamics, it is important to study the interactions between SCs and host immune cells in stages, beginning with how immune cells respond to SCs under normal physiologic conditions, as we have done in this study. Additional work will explore how senescence-inducing therapies may impact these baseline immune reactions (77).

Although not fully capturing the complete pathogenesis of human HNSCC and its clinical course (de novo carcinogenesis, growth and clinical manifestation, surgical removal, treatment of minimal residual disease, and recurrence), our conclusions may still be directly relevant for understanding senescence during de novo carcinogenesis and recurrence, as senescence can be induced by either oncogene activation or chemotherapy. Therefore, our findings could also help develop novel cancer early-detection strategies based on the biology of cellular senescence (78).

M. Hoare reports grants from AstraZeneca and Pfizer and personal fees from Boston Scientific, Sirtex, Quotient Therapeutics, and Ensocell outside the submitted work. No disclosures were reported by the other authors.

T. Ziglari: Data curation, software, formal analysis, validation, investigation, visualization, methodology, writing–original draft. N.L. Calistri: Data curation, software, formal analysis, visualization, methodology, writing–review and editing. J.M. Finan: Formal analysis, investigation, writing–review and editing. D.S. Derrick: Software, visualization. E.S. Nakayasu: Investigation, writing–review and editing. M.C. Burnet: Investigation. J.E. Kyle: Investigation, writing–review and editing. M. Hoare: Conceptualization, supervision, funding acquisition. L.M. Heiser: Supervision, writing–review and editing. F. Pucci: Conceptualization, resources, formal analysis, supervision, funding acquisition, investigation, visualization, methodology, writing–original draft, writing–review and editing.

We thank Drs. Sheila Stewart (Washington University, St. Louis, MO) for useful discussion and Robert Searles, Director of the Massively Parallel Sequencing Shared Resource at the OHSU, for help and advice with sequencing and genome alignment. We thank the OHSU Multiscale Microscopy Core for help with electron microscopy. This work was funded by the department of Otolaryngology startup grant (F. Ferdinando), the International Alliance for Cancer Early Detection, an alliance between Cancer Research UK (C52489/A29681 to F. Ferdinando, M. Hoare), Canary Center at Stanford University, the University of Cambridge, OHSU Knight Cancer Institute, University College London, and the University of Manchester, a NIH T32 Fellowship in “Integrated Training in Quantitative and Experimental Cancer Systems Biology” CA254888 (T. Ziglari), and Exploratory Research Seed Grant funding from the OHSU School of Medicine. This research is affiliated with the PMedIC joint research collaboration between the OHSU and Pacific Northwest National Laboratories (PNNL). The proteomics mass spectrometry analysis was performed at the Environmental Molecular Sciences Laboratory, U.S. Department of Energy National Scientific User Facility, located at the Pacific Northwest National Laboratory, operated under contract DE-AC05-76RL01830.

Note: Supplementary data for this article are available at Cancer Research Online (http://cancerres.aacrjournals.org/).

1.
Calcinotto
A
,
Kohli
J
,
Zagato
E
,
Pellegrini
L
,
Demaria
M
,
Alimonti
A
.
Cellular senescence: aging, cancer, and injury
.
Physiol Rev
2019
;
99
:
1047
78
.
2.
Casella
G
,
Munk
R
,
Kim
KM
,
Piao
Y
,
De
S
,
Abdelmohsen
K
, et al
.
Transcriptome signature of cellular senescence
.
Nucleic Acids Res
2019
;
47
:
7294
305
.
3.
Demaria
M
,
O’Leary
MN
,
Chang
J
,
Shao
L
,
Liu
S
,
Alimirah
F
, et al
.
Cellular senescence promotes adverse effects of chemotherapy and cancer relapse
.
Cancer Discov
2017
;
7
:
165
76
.
4.
Fletcher-Sananikone
E
,
Kanji
S
,
Tomimatsu
N
,
Di Cristofaro
LFM
,
Kollipara
RK
,
Saha
D
, et al
.
Elimination of radiation-induced senescence in the brain tumor microenvironment attenuates glioblastoma recurrence
.
Cancer Res
2021
;
81
:
5935
47
.
5.
Hernandez-Segura
A
,
Nehme
J
,
Demaria
M
.
Hallmarks of cellular senescence
.
Trends Cell Biol
2018
;
28
:
436
53
.
6.
Salama
R
,
Sadaie
M
,
Hoare
M
,
Narita
M
.
Cellular senescence and its effector programs
.
Genes Dev
2014
;
28
:
99
114
.
7.
van Niel
G
,
Carter
DRF
,
Clayton
A
,
Lambert
DW
,
Raposo
G
,
Vader
P
.
Challenges and directions in studying cell-cell communication by extracellular vesicles
.
Nat Rev Mol Cell Biol
2022
;
23
:
369
82
.
8.
Basisty
N
,
Kale
A
,
Jeon
OH
,
Kuehnemann
C
,
Payne
T
,
Rao
C
, et al
.
A proteomic atlas of senescence-associated secretomes for aging biomarker development
.
PLoS Biol
2020
;
18
:
e3000599
.
9.
Kavanagh
EL
,
Lindsay
S
,
Halasz
M
,
Gubbins
LC
,
Weiner-Gorzel
K
,
Guang
MHZ
, et al
.
Protein and chemotherapy profiling of extracellular vesicles harvested from therapeutic induced senescent triple negative breast cancer cells
.
Oncogenesis
2017
;
6
:
e388
.
10.
Terlecki-Zaniewicz
L
,
Lämmermann
I
,
Latreille
J
,
Bobbili
MR
,
Pils
V
,
Schosserer
M
, et al
.
Small extracellular vesicles and their miRNA cargo are anti-apoptotic members of the senescence-associated secretory phenotype
.
Aging (Albany NY)
2018
;
10
:
1103
32
.
11.
Borghesan
M
,
Fafián-Labora
J
,
Eleftheriadou
O
,
Carpintero-Fernández
P
,
Paez-Ribes
M
,
Vizcay-Barrena
G
, et al
.
Small extracellular vesicles are key regulators of non-cell autonomous intercellular communication in senescence via the interferon protein IFITM3
.
Cell Rep
2019
;
27
:
3956
71 e6
.
12.
Kadota
T
,
Fujita
Y
,
Yoshioka
Y
,
Araya
J
,
Kuwano
K
,
Ochiya
T
.
Emerging role of extracellular vesicles as a senescence-associated secretory phenotype: insights into the pathophysiology of lung diseases
.
Mol Aspects Med
2018
;
60
:
92
103
.
13.
Mathieu
M
,
Martin-Jaular
L
,
Lavieu
G
,
Théry
C
.
Specificities of secretion and uptake of exosomes and other extracellular vesicles for cell-to-cell communication
.
Nat Cell Biol
2019
;
21
:
9
17
.
14.
Pucci
F
,
Pittet
MJ
.
Molecular pathways: tumor-derived microvesicles and their interactions with immune cells in vivo
.
Clin Cancer Res
2013
;
19
:
2598
604
.
15.
Kang
TW
,
Yevsa
T
,
Woller
N
,
Hoenicke
L
,
Wuestefeld
T
,
Dauch
D
, et al
.
Senescence surveillance of pre-malignant hepatocytes limits liver cancer development
.
Nature
2011
;
479
:
547
51
.
16.
Eggert
T
,
Wolter
K
,
Ji
J
,
Ma
C
,
Yevsa
T
,
Klotz
S
, et al
.
Distinct functions of senescence-associated immune responses in liver tumor surveillance and tumor progression
.
Cancer Cell
2016
;
30
:
533
47
.
17.
Pucci
F
,
Garris
C
,
Lai
CP
,
Newton
A
,
Pfirschke
C
,
Engblom
C
, et al
.
SCS macrophages suppress melanoma by restricting tumor-derived vesicle-B cell interactions
.
Science
2016
;
352
:
242
6
.
18.
Hsu
C
,
Morohashi
Y
,
Yoshimura
S
,
Manrique-Hoyos
N
,
Jung
S
,
Lauterbach
MA
, et al
.
Regulation of exosome secretion by Rab35 and its GTPase-activating proteins TBC1D10A-C
.
J Cell Biol
2010
;
189
:
223
32
.
19.
Yang
L
,
Peng
X
,
Li
Y
,
Zhang
X
,
Ma
Y
,
Wu
C
, et al
.
Long non-coding RNA HOTAIR promotes exosome secretion by regulating RAB35 and SNAP23 in hepatocellular carcinoma
.
Mol Cancer
2019
;
18
:
78
.
20.
Onken
MD
,
Winkler
AE
,
Kanchi
KL
,
Chalivendra
V
,
Law
JH
,
Rickert
CG
, et al
.
A surprising cross-species conservation in the genomic landscape of mouse and human oral cancer identifies a transcriptional signature predicting metastatic disease
.
Clin Cancer Res
2014
;
20
:
2873
84
.
21.
Vermeer
DW
,
Coppock
JD
,
Zeng
E
,
Lee
KM
,
Spanos
WC
,
Onken
MD
, et al
.
Metastatic model of HPV+ oropharyngeal squamous cell carcinoma demonstrates heterogeneity in tumor metastasis
.
Oncotarget
2016
;
7
:
24194
207
.
22.
Hoover
AC
,
Spanos
WC
,
Harris
GF
,
Anderson
ME
,
Klingelhutz
AJ
,
Lee
JH
.
The role of human papillomavirus 16 E6 in anchorage-independent and invasive growth of mouse tonsil epithelium
.
Arch Otolaryngol Head Neck Surg
2007
;
133
:
495
502
.
23.
Hamilton
N
,
Claudio
NM
,
Armstrong
RJ
,
Pucci
F
.
Cell surface labeling by engineered extracellular vesicles
.
Adv Biosyst
2020
;
4
:
e2000007
.
24.
Amendola
M
,
Venneri
MA
,
Biffi
A
,
Vigna
E
,
Naldini
L
.
Coordinate dual-gene transgenesis by lentiviral vectors carrying synthetic bidirectional promoters
.
Nat Biotechnol
2005
;
23
:
108
16
.
25.
Kornilov
R
,
Puhka
M
,
Mannerström
B
,
Hiidenmaa
H
,
Peltoniemi
H
,
Siljander
P
, et al
.
Efficient ultrafiltration-based protocol to deplete extracellular vesicles from fetal bovine serum
.
J Extracell Vesicles
2018
;
7
:
1422674
.
26.
Nguyen
U
,
Squaglia
N
,
Boge
A
,
Fung
PA
.
The Simple Western: a gel-free, blot-free, hands-free Western blotting reinvention
.
Nat Methods
2011
;
8
.
v
vi
.
27.
Nakayasu
ES
,
Nicora
CD
,
Sims
AC
,
Burnum-Johnson
KE
,
Kim
YM
,
Kyle
JE
, et al
.
MPLEx: a robust and universal protocol for single-sample integrative proteomic, metabolomic, and lipidomic analyses
.
mSystems
2016
;
1
:
e00043-16
.
28.
Matos Baltazar
L
,
Nakayasu
ES
,
Sobreira
TJ
,
Choi
H
,
Casadevall
A
,
Nimrichter
L
, et al
.
Antibody binding alters the characteristics and contents of extracellular vesicles released by histoplasma capsulatum
.
mSphere
2016
;
1
:
e00085-15
.
29.
Cox
J
,
Mann
M
.
MaxQuant enables high peptide identification rates, individualized p.p.b.-range mass accuracies and proteome-wide protein quantification
.
Nat Biotechnol
2008
;
26
:
1367
72
.
30.
Schwanhäusser
B
,
Busse
D
,
Li
N
,
Dittmar
G
,
Schuchhardt
J
,
Wolf
J
, et al
.
Global quantification of mammalian gene expression control
.
Nature
2011
;
473
:
337
42
.
31.
Love
MI
,
Huber
W
,
Anders
S
.
Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2
.
Genome Biol
2014
;
15
:
550
.
32.
Zhu
A
,
Ibrahim
JG
,
Love
MI
.
Heavy-tailed prior distributions for sequence count data: removing the noise and preserving large differences
.
Bioinformatics
2019
;
35
:
2084
92
.
33.
Hao
Y
,
Hao
S
,
Andersen-Nissen
E
,
Mauck
WM
3rd
,
Zheng
S
,
Butler
A
, et al
.
Integrated analysis of multimodal single-cell data
.
Cell
2021
;
184
:
3573
87 e29
.
34.
Hafemeister
C
,
Satija
R
.
Normalization and variance stabilization of single-cell RNA-seq data using regularized negative binomial regression
.
Genome Biol
2019
;
20
:
296
.
35.
Gao
R
,
Bai
S
,
Henderson
YC
,
Lin
Y
,
Schalck
A
,
Yan
Y
, et al
.
Delineating copy number and clonal substructure in human tumors from single-cell transcriptomes
.
Nat Biotechnol
2021
;
39
:
599
608
.
36.
Jin
S
,
Guerrero-Juarez
CF
,
Zhang
L
,
Chang
I
,
Ramos
R
,
Kuan
C-H
, et al
.
Inference and analysis of cell-cell communication using CellChat
.
Nat Commun
2021
;
12
:
1088
.
37.
Durinck
S
,
Spellman
PT
,
Birney
E
,
Huber
W
.
Mapping identifiers for the integration of genomic datasets with the R/Bioconductor package biomaRt
.
Nat Protoc
2009
;
4
:
1184
91
.
38.
Estévez-Souto
V
,
Da Silva-Álvarez
S
,
Collado
M
.
The role of extracellular vesicles in cellular senescence
.
FEBS J
2023
;
290
:
1203
11
.
39.
de Souza Viana
L
,
de Aguiar Silva
FC
,
Andrade Dos Anjos Jacome
A
,
Calheiros Campelo Maia
D
,
Duarte de Mattos
M
,
Arthur Jacinto
A
, et al
.
Efficacy and safety of a cisplatin and paclitaxel induction regimen followed by chemoradiotherapy for patients with locally advanced head and neck squamous cell carcinoma
.
Head Neck
2016
;
38
(
Suppl 1
):
E970
80
.
40.
Itahana
K
,
Campisi
J
,
Dimri
GP
.
Methods to detect biomarkers of cellular senescence: the senescence-associated beta-galactosidase assay
.
Methods Mol Biol
2007
;
371
:
21
31
.
41.
González-Gualda
E
,
Baker
AG
,
Fruk
L
,
Muñoz-Espín
D
.
A guide to assessing cellular senescence in vitro and in vivo
.
FEBS J
2021
;
288
:
56
80
.
42.
Kale
A
,
Sharma
A
,
Stolzing
A
,
Desprez
PY
,
Campisi
J
.
Role of immune cells in the removal of deleterious senescent cells
.
Immun Ageing
2020
;
17
:
16
.
43.
Gieseck
RL
,
Wilson
MS
,
Wynn
TA
.
Type 2 immunity in tissue repair and fibrosis
.
Nat Rev Immunol
2018
;
18
:
62
76
.
44.
Welsh
JA
,
Goberdhan
DCI
,
O’Driscoll
L
,
Buzas
EI
,
Blenkiron
C
,
Bussolati
B
, et al
.
Minimal information for studies of extracellular vesicles (MISEV2023): from basic to advanced approaches
.
J Extracell Vesicles
2024
;
13
:
e12404
.
45.
Ostrowski
M
,
Carmo
NB
,
Krumeich
S
,
Fanget
I
,
Raposo
G
,
Savina
A
, et al
.
Rab27a and Rab27b control different steps of the exosome secretion pathway
.
Nat Cell Biol
2010
;
12
:
19
30 sup pp 1–13
.
46.
Guccini
I
,
Revandkar
A
,
D’Ambrosio
M
,
Colucci
M
,
Pasquini
E
,
Mosole
S
, et al
.
Senescence reprogramming by TIMP1 deficiency promotes prostate cancer metastasis
.
Cancer Cell
2021
;
39
:
68
82.e9
.
47.
Verweij
FJ
,
Balaj
L
,
Boulanger
CM
,
Carter
DRF
,
Compeer
EB
,
D’Angelo
G
, et al
.
The power of imaging to understand extracellular vesicle biology in vivo
.
Nat Methods
2021
;
18
:
1013
26
.
48.
Choudhary
S
,
Satija
R
.
Comparison and evaluation of statistical error models for scRNA-seq
.
Genome Biol
2022
;
23
:
27
.
49.
Korsunsky
I
,
Millard
N
,
Fan
J
,
Slowikowski
K
,
Zhang
F
,
Wei
K
, et al
.
Fast, sensitive and accurate integration of single-cell data with Harmony
.
Nat Methods
2019
;
16
:
1289
96
.
50.
Taniguchi
S
,
Tanaka
Y
,
Elhance
A
,
Oshimori
N
.
A mechanistic basis for the malignant progression of salivary gland tumors
.
iScience
2021
;
24
:
103508
.
51.
Upadhyay
G
.
Emerging role of lymphocyte antigen-6 family of genes in cancer and immune cells
.
Front Immunol
2019
;
10
:
819
.
52.
Murakami
K
,
Umemura
N
,
Adachi
M
,
Motoki
M
,
Ohkoshi
E
.
ABCG2, CD44 and SOX9 are increased with the acquisition of drug resistance and involved in cancer stem cell activities in head and neck squamous cell carcinoma cells
.
Exp Ther Med
2022
;
24
:
722
.
53.
Saha
SK
,
Kim
K
,
Yang
GM
,
Choi
HY
,
Cho
SG
.
Cytokeratin 19 (KRT19) has a role in the reprogramming of cancer stem cell-like cells to less aggressive and more drug-sensitive cells
.
Int J Mol Sci
2018
;
19
:
1423
.
54.
Duy
C
,
Li
M
,
Teater
M
,
Meydan
C
,
Garrett-Bakelman
FE
,
Lee
TC
, et al
.
Chemotherapy induces senescence-like resilient cells capable of initiating AML recurrence
.
Cancer Discov
2021
;
11
:
1542
61
.
55.
Nacarelli
T
,
Fukumoto
T
,
Zundell
JA
,
Fatkhutdinov
N
,
Jean
S
,
Cadungog
MG
, et al
.
NAMPT inhibition suppresses cancer stem-like cells associated with therapy-induced senescence in ovarian cancer
.
Cancer Res
2020
;
80
:
890
900
.
56.
Deschênes-Simard
X
,
Parisotto
M
,
Rowell
MC
,
Le Calvé
B
,
Igelmann
S
,
Moineau-Vallée
K
, et al
.
Circumventing senescence is associated with stem cell properties and metformin sensitivity
.
Aging Cell
2019
;
18
:
e12889
.
57.
Milanovic
M
,
Fan
DNY
,
Belenki
D
,
Däbritz
JHM
,
Zhao
Z
,
Yu
Y
, et al
.
Senescence-associated reprogramming promotes cancer stemness
.
Nature
2018
;
553
:
96
100
.
58.
Cibrián
D
,
Sánchez-Madrid
F
.
CD69: from activation marker to metabolic gatekeeper
.
Eur J Immunol
2017
;
47
:
946
53
.
59.
Qi
H
,
Kastenmüller
W
,
Germain
RN
.
Spatiotemporal basis of innate and adaptive immunity in secondary lymphoid tissue
.
Annu Rev Cell Dev Biol
2014
;
30
:
141
67
.
60.
Meyer
SJ
,
Linder
AT
,
Brandl
C
,
Nitschke
L
.
B cell siglecs-news on signaling and its interplay with ligand binding
.
Front Immunol
2018
;
9
:
2820
.
61.
Cao
A
,
Alluqmani
N
,
Buhari
FHM
,
Wasim
L
,
Smith
LK
,
Quaile
AT
, et al
.
Galectin-9 binds IgM-BCR to regulate B cell signaling
.
Nat Commun
2018
;
9
:
3288
.
62.
Beck
J
,
Horikawa
I
,
Harris
C
.
Cellular senescence: mechanisms, morphology, and mouse models
.
Vet Pathol
2020
;
57
:
747
57
.
63.
Gorgoulis
V
,
Adams
PD
,
Alimonti
A
,
Bennett
DC
,
Bischof
O
,
Bishop
C
, et al
.
Cellular senescence: defining a path forward
.
Cell
2019
;
179
:
813
27
.
64.
Aoshiba
K
,
Tsuji
T
,
Nagai
A
.
Bleomycin induces cellular senescence in alveolar epithelial cells
.
Eur Respir J
2003
;
22
:
436
43
.
65.
Pich
O
,
Cortes-Bullich
A
,
Muiños
F
,
Pratcorona
M
,
Gonzalez-Perez
A
,
Lopez-Bigas
N
.
The evolution of hematopoietic cells under cancer therapy
.
Nat Commun
2021
;
12
:
4803
.
66.
Takasugi
M
,
Okada
R
,
Takahashi
A
,
Virya Chen
D
,
Watanabe
S
,
Hara
E
.
Small extracellular vesicles secreted from senescent cells promote cancer cell proliferation through EphA2
.
Nat Commun
2017
;
8
:
15729
.
67.
Al Suraih
MS
,
Trussoni
CE
,
Splinter
PL
,
LaRusso
NF
,
O’Hara
SP
.
Senescent cholangiocytes release extracellular vesicles that alter target cell phenotype via the epidermal growth factor receptor
.
Liver Int
2020
;
40
:
2455
68
.
68.
Pittet
MJ
,
Michielin
O
,
Migliorini
D
.
Clinical relevance of tumour-associated macrophages
.
Nat Rev Clin Oncol
2022
;
19
:
402
21
.
69.
Kara
EE
,
McKenzie
DR
,
Bastow
CR
,
Gregor
CE
,
Fenix
KA
,
Ogunniyi
AD
, et al
.
CCR2 defines in vivo development and homing of IL-23-driven GM-CSF-producing Th17 cells
.
Nat Commun
2015
;
6
:
8644
.
70.
Bascones-Martínez
A
,
López-Durán
M
,
Cano-Sánchez
J
,
Sánchez-Verde
L
,
Díez-Rodríguez
A
,
Aguirre-Echebarría
P
, et al
.
Differences in the expression of five senescence markers in oral cancer, oral leukoplakia and control samples in humans
.
Oncol Lett
2012
;
3
:
1319
25
.
71.
George
B
,
Horn
D
,
Bayo
P
,
Zaoui
K
,
Flechtenmacher
C
,
Grabe
N
, et al
.
Regulation and function of Myb-binding protein 1A (MYBBP1A) in cellular senescence and pathogenesis of head and neck cancer
.
Cancer Lett
2015
;
358
:
191
9
.
72.
Van Avondt
K
,
Strecker
JK
,
Tulotta
C
,
Minnerup
J
,
Schulz
C
,
Soehnlein
O
.
Neutrophils in aging and aging-related pathologies
.
Immunol Rev
2023
;
314
:
357
75
.
73.
Marin
I
,
Boix
O
,
Garcia-Garijo
A
,
Sirois
I
,
Caballe
A
,
Zarzuela
E
, et al
.
Cellular senescence is immunogenic and promotes antitumor immunity
.
Cancer Discov
2023
;
13
:
410
31
.
74.
Cocozza
F
,
Martin-Jaular
L
,
Lippens
L
,
Di Cicco
A
,
Arribas
YA
,
Ansart
N
, et al
.
Extracellular vesicles and co-isolated endogenous retroviruses from murine cancer cells differentially affect dendritic cells
.
EMBO J
2023
;
42
:
e113590
.
75.
Gibieža
P
,
Prekeris
R
.
Rab GTPases and cell division
.
Small GTPases
2018
;
9
:
107
15
.
76.
Wheeler
DB
,
Zoncu
R
,
Root
DE
,
Sabatini
DM
,
Sawyers
CL
.
Identification of an oncogenic RAB protein
.
Science
2015
;
350
:
211
7
.
77.
Duggal
NA
,
Niemiro
G
,
Harridge
SDR
,
Simpson
RJ
,
Lord
JM
.
Can physical activity ameliorate immunosenescence and thereby reduce age-related multi-morbidity?
Nat Rev Immunol
2019
;
19
:
563
72
.
78.
Paez-Ribes
M
,
González-Gualda
E
,
Doherty
GJ
,
Muñoz-Espín
D
.
Targeting senescent cells in translational medicine
.
EMBO Mol Med
2019
;
11
:
e10234
.