WO2024063109A1 - Senescent cell remover - Google Patents
Senescent cell remover Download PDFInfo
- Publication number
- WO2024063109A1 WO2024063109A1 PCT/JP2023/034172 JP2023034172W WO2024063109A1 WO 2024063109 A1 WO2024063109 A1 WO 2024063109A1 JP 2023034172 W JP2023034172 W JP 2023034172W WO 2024063109 A1 WO2024063109 A1 WO 2024063109A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- cells
- tom
- cell
- mice
- senescent
- Prior art date
- Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
- Ceased
Links
Classifications
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K39/395—Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K45/00—Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P1/00—Drugs for disorders of the alimentary tract or the digestive system
- A61P1/16—Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P3/00—Drugs for disorders of the metabolism
- A61P3/06—Antihyperlipidemics
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P43/00—Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12Q—MEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
- C12Q1/00—Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
- C12Q1/68—Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
- C12Q1/6876—Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
Definitions
- the present invention relates to a senescent cell removing agent and a method for improving various diseases associated with aging using the same.
- the present invention also relates to a pharmaceutical composition for the treatment or prevention of non-alcoholic steatohepatitis (NASH).
- NASH non-alcoholic steatohepatitis
- Non-Patent Document 1 Accumulation of aging is one of the main causes of age-related inflammation and causes various age-related diseases. However, little is known about the molecular basis underlying this accumulation and its potential as a target to ameliorate the aging process.
- Rb2/p130 is the dominating pocket protein in the p53-p21 DNA damage response pathway leading to senescence. Oncogene 28, (2009). Fan, J. & Bertino, J. R. Functional roles of E2F in cell cycle regulation. Oncogene 14, (1997). Dyson, N., Guida, P., Muenger, K. & Harlow, E. Homologous sequences in adenovirus E1A and human papillomavirus E7 proteins mediate interaction with the same set of cellular proteins. Journal of Virology 66, (1992). Janky, R. et al. iRegulon: From a Gene List to a Gene Regulatory Network Using Large Motif and Track Collections.
- CD8+ tissue-resident memory T cells promote liver fibrosis resolution by inducing apoptosis of hepatic stellate cells. Nature Communications 12, (2021).
- K. et al. PD-1 immune checkpoint blockade reduces pathology and improves memory in mouse models of Alzheimer’s disease. Nature Medicine 22, (2016). Rosenzweig, N. et al. PD-1/PD-L1 checkpoint blockade harnesses monocyte-derived macrophages to combat cognitive impairment in a tauopathy mouse model. Nature Communications 10, (2019).
- Pfister, D. et al. NASH limits anti-tumour surveillance in immunotherapy-treated HCC. Nature 592, (2021). Martins, F. et al.
- the CSF1 receptor inhibitor pexidartinib reduces tissue macrophage levels without affecting glucose homeostasis in mice.
- the main object of the present invention is to provide a pharmaceutical composition and a senescent cell remover for use in the treatment or prevention of NASH.
- the present inventors have found that by inhibiting PD-L1 on the cell membrane from binding to PD-1, senescent cells are removed in NASH model animals, leading to fat accumulation and fibrosis in the liver.
- the inventors have completed the present invention by discovering that liver function is significantly reduced and that indicators of liver function are also improved accordingly.
- compositions and the like according to the present invention are as follows.
- NASH non-alcoholic steatohepatitis
- the substance is one selected from the group consisting of anti-PD-L1 antibodies and anti-PD-1 antibodies.
- the pharmaceutical composition of [1] above, wherein the substance is a low molecular compound that specifically binds to PD-L1.
- the pharmaceutical composition and senescent cell removal agent according to the present invention can reduce the proportion of p16-positive (p16 + ) senescent cells in the liver, and can further normalize the functions in the liver. Therefore, the pharmaceutical composition according to the present invention is effective in treating or preventing NASH.
- Example 1 Tom + cells (upper (panel) and the proportion of PD-L1 + cells in the Tom + cell population (lower panel).
- FIG. 2 is a diagram showing the median fluorescence intensity (MFI) of ProteoStat signals of protein aggregate staining analyzed by FACS in PD-L1 + and PPD-L1 ⁇ d-Sen HCA2 cells in Example 1. A representative scatter plot is shown in the right panel.
- MFI median fluorescence intensity
- Relative light units (RLU) per cell represent the luminescent signal for assessing chymotrypsin, trypsin, or caspase-like protease activity.
- Example 1 the indicated PD-L1 + population of cells was determined by FACS.
- Example 2 the indicated PD-L1 + populations (Quie: quiescent, n-Sen: nutlin3a-induced senescence, d-Sen: doxorubicin-induced senescence) of mouse lung fibroblasts (MPF) were determined by FACS.
- FIG. A representative histogram is shown in the left panel.
- the cytotoxicity of the indicated MPFs target cells was evaluated by incubation with mouse activated CD8 + T cells (effector cells) for 18 hours.
- FIG. 2 is a diagram showing a volcano plot of differentially expressed genes (DEGs) between n-Sen MPF and Quie MPF in Example 2.
- DEGs differentially expressed genes
- Red dots indicate DEGs identified by FDR ⁇ 0.05 and Log2FC>0.6.
- Figure 2 shows a dot plot of gene ontology (GO) enriched for biological process terms in DEGs upregulated in d-Sen MPF compared to Quie MPF in Example 2. Enriched terms were identified by FDR ⁇ 0.05.
- FIG. 3 shows the cytotoxicity of Quie MPF supplied by conditioned medium from Quie MPF or n-Sen MPF expressing doxycycline-inducible 3xFlag-I ⁇ B ⁇ (S32/36A) in Example 2.
- 2 is a volcano plot of transposable elements (TE) differentially expressed between n-Sen MPF and Quie MPF in Example 2. Blue dots (Sig.
- FIG. 2 is a diagram showing qPCR analysis of ERV expression in Tom ⁇ cells and Tom + /CD45 ⁇ cells selected from 6-month-old male p16-Tom mice in Example 2.
- 2B is a diagram showing the cytotoxicity of PD-L1 + and PD-L1 ⁇ n-Sen MPFs selected in Example 2, similar to FIG. 2b.
- FIG. 3 is a diagram showing UMAP visualization of liver sinusoidal endothelial cells (LSEC) of a 7-month-old p16-Tom mouse in Example 3.
- FIG. 3 shows a violin plot of the expression of Cd274 (PD-L1) in Tom + and Tom ⁇ LSECs in Example 3.
- the red dotted line represents the cutoff threshold for determining PD-L1 + and PD-L1 ⁇ cells.
- FIG. 3 is a diagram showing a volcano plot of DEGs between PD-L1 + cells and PD-L1 ⁇ cells in Example 3.
- FIG. DEGs were identified by FDR ⁇ 0.05 and Log2FC>0.15. Red dots (gray dots in the figure) represent important DEGs.
- FIG. 3 shows a category-gene net plot of DEGs between Tom + /PD-L1 + cells and Tom + /PD-L1 ⁇ cells containing the GO terms listed in FIG. 3d in Example 3.
- FIG. 7 is a diagram showing GSEA between Tom + /PD-L1 + LSEC and Tom + /PD-L1 ⁇ LSEC in Example 3. All GSEA terms were identified by FDR ⁇ 0.05.
- Example 4 the proportion of PD-L1 + cells in Tom + and Tom ⁇ cells in the indicated organs, as in Figure 4a.
- FIG. 4c shows representative images of livers from the indicated mice (left panel: H&E staining) and quantification of lipid droplet area (right panel) similar to FIG. 4c (scale Bar: 100 ⁇ m).
- FIG. 4 shows a representative fluorescence image (upper panel) of the liver of a p16-Tom mouse treated in the same manner as in FIG. 4e and the percentage of Tom + cells (lower panel) in Example 4.
- FIG. 1c is a diagram showing the gating strategy of the experiment shown in FIG. 1c in Example 1.
- HCA2 cells were isolated from Quie, replicative senescent cells (r-Sen), n-Sen, doxorubicin-induced senescent cells (d-Sen), and proliferative cells (Prolifer) using the indicated antibodies. Immunoblot of lysate.
- HCA2 cells were isolated from Quie,
- FIG. 2 is a diagram showing qPCR analysis of PD-L1 and CDC20 expression in selected G1, S, and G2/M HCA2 cells in Example 1. Results were expressed as relative fold change.
- FIG. 2 shows an immunoblot of lysate from HCA2 cells expressing mock, p130 and Flag-E2F97 using the indicated antibodies in Example 1.
- Figure 6c shows qPCR analysis of PD-L1 and CDC20 expression in HCA2 cells similar to Figure 6c.
- FIG. 3 shows an immunoblot of lysate from cells.
- FIG. 2 is a diagram showing a volcano plot of differentially expressed genes between PD-L1 + and PD-L1 ⁇ d-Sen HCA2 cells in Example 1. Blue dots indicate DEGs identified by FDR ⁇ 0.05. Red arrow indicates Cd274(PD-L1).
- FIG. 6f shows transcription factor (TF) candidates enriched in the promoter regions of genes positively correlated with PD-L1 (R 2 ⁇ 0.95) in the RNA-Seq results in FIG. 6f.
- the normalized enrichment score threshold was 3 or higher.
- FIG. 2 shows an immunoblot of lysates from sorted PD-L1 ⁇ and PD-L + 1d-Sen HCA2 cells using the indicated antibodies in Example 1. Relative quantification values of PD-L1 are labeled. 2 is an SA- ⁇ -Gal stained image of Quien, n-Sen, and d-Sen mouse lung fibroblasts (MPFs) in Example 2. Representative images are displayed in the right panel (scale bar: 500 ⁇ m).
- Figure 7a shows immunoblotting of lysates from MPF similar to Figure 7a using the indicated antibodies.
- FIG. 2b shows a representative histogram of the results shown in Figure 2b (right panel). The gating strategy for the in vitro T cell killing assay is shown in the left panel.
- FIG. 2D shows a categorical gene net plot of DEGs between n-Sen MPF and Quie MPF that are involved in GO results as in FIG. 2d.
- FIG. 6 is a diagram showing GSEA of RNA-Seq results between n-Sen MPF and Quie MPF in Example 2. All GSEA terms were identified by FDR ⁇ 0.05.
- FIG. 2 is a diagram showing qPCR analysis of SASP factor expression in n-Sen MPF with or without doxy-inducible 3 ⁇ Flag-I ⁇ B ⁇ (S32/36A) in Example 2.
- Example 2 the cytotoxicity of sorted PD-L1 + and PD-L1 - n-Sen MPFs (target cells) was evaluated for 18 hours with mouse activated CD8 + T cells (effector cells) derived from OT-1 mice. It was evaluated by incubating. During co-culture, ovalbumin (OVA) peptide 257-264 was added at 1 ⁇ g/mL to induce antigen-specific responses. A representative histogram is shown in the right panel.
- Figure 2 is an immunoblot of lysate from mock PD-L1 overexpressing (OE) MPF using the indicated antibodies in Example 2. A representative histogram of the results shown in Figure 2i.
- Example 2 mock or PD-L1 expressing n-Sen MPFs were treated with 10 ⁇ g/mL isotype IgG or ⁇ PD-1 during incubation with CD8 + T cells from OT-1 mice. It is a figure showing cytotoxicity. A representative histogram is shown in the right panel.
- Figure 3 shows UMAP visualization of Tom + population (top panel) and Cd274 (PD-L1) expression levels (bottom panel) in the indicated scRNA-Seq dataset and cell types in Example 3. .
- NASH samples were prepared from 7-month-old p16-Tom mice that were fed CDA-HFD for 6 months. Kidney samples were collected from 5.5 month old p16-Tom mice.
- FIG. 3 is a volcano plot of DEGs between PD-L1 + and PD-L1 - /Tom + cells in the indicated samples in Example 3. DEGs were identified by FDR ⁇ 0.05 and Log2FC>0.15. Red dots (gray dots in the figure) represent important DEGs.
- Figure 3 is a volcano plot of DEGs between PD-L1 + and PD-L1 - /Tom + cells in the indicated samples in Example 3. DEGs were identified by FDR ⁇ 0.05 and Log2FC>0.15.
- Red dots represent important DEGs.
- Figure 3 is a volcano plot of DEGs between PD-L1 + and PD-L1 - /Tom + cells in the indicated samples in Example 3.
- DEGs were identified by FDR ⁇ 0.05 and Log2FC>0.15.
- Red dots represent important DEGs.
- Example 3 GO terms enriched in upregulated DEGs of PD-L1 + /Tom + cells compared to PD-L1 ⁇ /Tom + cells in the indicated samples.
- 5 is a diagram showing the results of FACS analysis of splenic CD8 + T cells of mice treated with ⁇ PD-1 and/or ⁇ CD8 as in FIG. 4.
- FIG. The gating strategy and representative scatter plots are shown in the right panel. Representative images of the immunofluorescence staining shown in Figure 4a with the indicated treatments for lung, liver, and kidney (blue: DAPI; red: PD-L1; yellow: tdTomato; green: CD3; scale bar: 500 ⁇ m).
- Figure 1 shows the percentage of PD-L1 + /Tom + cells (upper panel) and PD-L1 - /Tom + cells from immunofluorescent stained sections of lung, liver, and kidney with the indicated treatments in Example 4.
- Fold change (FC) between the IgG group and the ⁇ PD-1 group without ⁇ CD8 treatment is shown in the figure.
- Representative images of immunofluorescence staining of IgG and ⁇ PD-1 treated liver shown in Figure 4a blue: DAPI; red: PD-L1; yellow: tdTomato; green: F40/80; scale bar: 500 ⁇ m) .
- FIG. 3 is a diagram showing the percentage of PD-L1 + cells in F40/80 ⁇ cells (left panel) and F40/80 + cells (right panel) from immunofluorescence-stained liver sections in Example 4.
- FIG. 4 is a diagram showing the percentage of PD-L1 + cells in Tom + /F40/80 ⁇ cells from an immunofluorescence-stained section of the liver in Example 4.
- FIG. 4 is a diagram showing the results of a motor ability test of young (3 months old) and old (17.5 months old) wild type mice in Example 4.
- FIG. 11e is a diagram showing the results of a mouse grip strength test similar to FIG. 11e. Similar to Figure 4e, wild animals were fed a normal diet (Normal) or an L-amino acid diet (CDA-HFD) containing 60 kcal of fat, 0.1% methionine, and no choline for 10 weeks together with ⁇ PD-1 treatment or ABT263 treatment.
- FIG. 3 shows activation and infiltration of hepatic CD8 + T cells analyzed by FACS from type mice. The gating strategy is shown in the right panel.
- FIG. 12 is a diagram showing the measurement results of serum levels of AST, ALT, and LDH in mice similar to FIG.
- FIG. 12a Figure 12a is Sirius red staining of mouse liver similar to Figure 12a. Representative images are shown in the top panel. The percentage of red area (collagen area) in each field was quantified and described as liver fibrosis (scale bar: 100 ⁇ m). Body weight and serum total cholesterol (T-CHO) values of mice similar to FIG. 12a.
- FIG. 13 is a diagram showing the results of a motor ability test of mice shown similarly to FIG. 13a. The dwell time from the start of rotation of the rotarod model until the mouse fell into the switch was recorded.
- FIG. 13A is a diagram showing the results of a grip strength test of a mouse shown similarly to FIG. 13a. 13A is a diagram showing the results of quantifying the alveolar size of the mouse shown in the same manner as FIG. 13a in Example 4.
- Figure 13e shows the measurement results of serum levels of AST, ALT, and LDH in mice shown in the same manner as in Figure 13e. Representative images of the liver from mice shown as in Figure 13e (bottom panel: H&E staining) and quantification of lipid droplet area (top panel) results. Sirius Red staining of mouse liver shown similarly to Figure 13e. Representative images are shown in the bottom panel (scale bar, 100 ⁇ m).
- senescent cells heterogeneously express the immune checkpoint PD-L1 and that PD-L1 + senescent cells accumulate with age in vivo.
- PD-L1 ⁇ cells are sensitive to T cell surveillance, whereas PD-L1 + cells are resistant even in the presence of senescence-associated secretory phenotype (SASP).
- SASP senescence-associated secretory phenotype
- single cell analysis of p16 + cells in vivo revealed that PD-L1 expression correlated with higher levels of SASP. Consistent with this, administration of anti-PD-1 antibodies to naturally aging or NASH-induced mice not only reduces the total number of p16 + cells in vivo but also reduces PD in an activated CD8 + T cell-dependent manner.
- the accumulation of senescent cells is suppressed by inhibiting the binding between PD-L1 and PD-1.
- PD-L1-positive (PD-L1 + ) senescent cells bind to PD-1 on CD8-positive T cells (CD8 + T cells), suppress the activity of CD8 + T cells, and perform immune surveillance.
- CD8 + T cells CD8-positive T cells
- CD8 + T cells CD8-positive T cells
- senescent cells p16 + cells
- ALT, and LDH serum levels can be improved.
- the pharmaceutical composition of the present embodiment is a pharmaceutical composition for treating or preventing NASH, which contains as an active ingredient a substance that inhibits PD-L1 on the cell membrane from binding to PD-1. By inhibiting the binding of PD-L1 on the cell membrane to PD-1, the pathological condition of NASH can be improved by removing senescent cells. Due to this mechanism of action, the pharmaceutical composition of this embodiment is effective in treating and preventing NASH.
- the active ingredient of the pharmaceutical composition of the present embodiment is not particularly limited as long as it is a substance that can inhibit PD-L1 on the cell membrane from binding to PD-1. It may be a substance that binds to PD-L1 and inhibits the binding to PD-1, or it may be a substance that binds to PD-1 and inhibits the binding to PD-L1. Since the effect of preventing or treating NASH can be obtained more effectively, the active ingredients of the pharmaceutical composition of this embodiment include PD-L1, which is a cell membrane protein of liver cells, and a cell membrane protein of CD8 + T cells. Preferably, it is a substance that inhibits binding to PD-1.
- Examples of the active ingredient of the pharmaceutical composition of the present embodiment include one or more selected from the group consisting of anti-PD-L1 antibodies and anti-PD-1 antibodies.
- the anti-PD-L1 antibody any known anti-PD-L1 antibody may be used, or it may be an antibody obtained by appropriately modifying a known antibody without impairing its affinity for PD-L1.
- the anti-PD-1 antibody any known anti-PD-1 antibody may be used, or it may be an antibody obtained by appropriately modifying a known antibody without impairing its affinity for PD-1. Improving the specificity and safety of antibodies is a technique that has been used for a long time, and can be carried out as appropriate using these techniques.
- anti-PD-L1 antibodies and anti-PD-1 antibodies that can be used in immune checkpoint inhibition therapy and the like can also be used as active ingredients in the pharmaceutical composition of the present embodiment.
- the active ingredient of the pharmaceutical composition of the present embodiment may be, for example, a low-molecular compound that specifically binds to PD-L1 or a low-molecular compound that specifically binds to PD-1. Further, the active ingredient of the pharmaceutical composition of the present embodiment may be a nucleic acid aptamer or a peptide that specifically binds to PD-L1 or PD-1. PD-L1 and low-molecular compounds that specifically bind to PD-1 can be obtained by screening various libraries. Furthermore, it is also possible to synthesize using the structural data of PD-L1 and PD-1.
- the active ingredient of the pharmaceutical composition of this embodiment also functions as an agent for removing senescent cells, in particular, an agent for removing p16-positive senescent cells.
- the pharmaceutical composition of this embodiment can be prepared by appropriately mixing a substance capable of inhibiting the binding of PD-L1 on cell membranes with PD-1, which is an active ingredient, and a pharmaceutically acceptable carrier. , can be prepared.
- the pharmaceutical composition can be manufactured by a method commonly used in the field of pharmaceutical manufacturing by using appropriate additives as necessary.
- Pharmaceutically acceptable carriers are diluents, excipients, binders, Solvents, etc.
- the carrier specifically, for example, water, physiological saline, various buffer solutions, etc. are used.
- additives that can be used include adjuvants, diluents, excipients, binders, stabilizers, tonicity agents, buffers, solubilizing agents, suspending agents, preservatives, antifreeze agents, and cryoprotectants. agents, lyoprotectants, bacteriostatic agents, etc.
- the animal to which the pharmaceutical composition of the present embodiment is administered is not particularly limited as long as it is an animal for which treatment or prevention of NASH is required, and may be a human or a non-human animal. However, mammals are preferred. Non-human mammals include cows, pigs, horses, sheep, goats, monkeys, dogs, cats, rabbits, mice, rats, hamsters, guinea pigs, and the like. Since a more sufficient therapeutic effect on NASH can be expected, the animal to which the pharmaceutical composition of this embodiment is administered is preferably an animal that has not developed cancer other than liver cancer. Furthermore, the administration route for administering the pharmaceutical composition of the present embodiment to animals is not particularly limited, and may include oral administration, intravenous administration, enteral administration, intramuscular administration, subcutaneous administration, transdermal administration, Examples include nasal administration and pulmonary administration.
- the dosage per administration of the pharmaceutical composition of the present embodiment is not particularly limited as long as the active ingredient can achieve the effect of removing senescent cells, and the dosage may include the species, sex, and age of the animal to which it is administered. , weight, presence or absence of underlying disease, pathological condition, etc. can be appropriately determined. For example, when using an anti-PD-L1 antibody or anti-PD-1 antibody used in immune checkpoint inhibition therapy as an active ingredient, refer to the dosage used in immune checkpoint inhibition therapy, etc. , the dosage of the pharmaceutical composition of this embodiment can be determined.
- the dosage of the pharmaceutical composition of the present embodiment is not particularly limited as long as the active ingredient has the effect of removing senescent cells, but the administration period is short in order to suppress side effects. It is preferable.
- the pharmaceutical composition of the present embodiment is preferably administered multiple times within 3 weeks, and preferably multiple times within 2 weeks, to animals that have developed NASH and require treatment. More preferably, it is administered multiple times within one week, and particularly preferably only once.
- mice were placed in cages of 2 to 5 mice at an ambient temperature of 23 to 25°C in a humidity-controlled room, fed standard food (CA-1, manufactured by CLEA Japan), and kept on a 12-hour light-dark cycle ( The lighting was maintained from 08:00 to 20:00). Water was provided freely with access to environmental enrichment. All animals were treated in accordance with the Animal Experiment Guidelines and Institutional Laboratory Animal Management of the Institute of Medical Science, The University of Tokyo. All p16-Tom mice are heterozygous and were generated by crossing p16 Ink4a -Cre ERT2 mice (Non-Patent Document 10) with Rosa26-CAG-LSL-tdTomato mice (obtained from Jackson Laboratory).
- OT-1-TCR transgenic mice have already been disclosed (Non-Patent Documents 47, 48). In this mouse, most CD8 + T cells are reactive to OVA peptide/MHC class I complexes.
- OT-1 mice were housed in a specific pathogen-free facility at Keio University. All mice used were bred in-house. All p16-Tom mice received tamoxifen (TAM, 80 mg/kg BW) intraperitoneally daily at the indicated times for 5 days.
- TAM tamoxifen
- mice received 250 ⁇ g/dose of isotype control IgG (BioLegend, clone: RTK2758) for 3 days or anti-mouse PD-1 (BioLegend, clone: 29F.1A12) for 3 weeks. 8 intraperitoneal injections were given within 8 days, and mice were sacrificed 1 day after the last injection. To deplete CD8 + T cells, all mice were treated with 200 ⁇ g/dose of isotype control IgG (Bio X Cell, clone: 2A3) or anti-mouse CD8a (Bio X Cell, clone: 2A3) twice weekly. : 53-6.7) was intraperitoneally administered 14 times within 7 weeks (Non-Patent Document 49).
- mice For the induction of non-alcoholic steatohepatitis (NASH), 1.5-month-old p16-Cre ERT2 -tdTomato mice were fed a diet containing 60 kcal% fat, 0.1% methionine, and a choline-free L-amino acid diet (CDA). -HFD: A06071302, manufactured by Research Diets) for a total of 10 weeks. For senolytic reagent treatment, all mice received placebo (vehicle control) or ABT263 by oral gavage.
- NASH non-alcoholic steatohepatitis
- ABT263 (manufactured by Selleck) was suspended in ethanol: polyethylene glycol 400: Phosal 50 PG at a ratio of 10:30:60, and then 50 mg/kg BW was administered daily for 7 days per cycle. A total of 2 cycles were performed with a 2 week interval between cycles (Non-Patent Document 37).
- PLX3397 (Selleck) was suspended in 5:25:70 DMSO:PEG300: ddH2O and then administered orally by gavage at 40 mg/kg BW twice a week ( Non-patent document 50).
- Non-Patent Document 5 Cell culture, cell cycle synchronization, and treatment with various drugs were performed as previously described (Non-Patent Document 5).
- Early passage hHCA2 (Non-Patent Document 51), primary mouse lung fibroblasts (MPF), and HEK293T cells (obtained from RIKEN Cell Bank) were incubated with 10% fetal bovine serum (FBS) (Sigma) and 1 ⁇ penicillin.
- FBS fetal bovine serum
- the cells were cultured in DMEM (manufactured by Nacalai Tesque) supplemented with /streptomycin/amphotericin B (manufactured by Nacalai Tesque). All cells were cultured at 37° C.
- Non-Patent Document 52 For quiescence induction, cells were cultured in DMEM supplemented with 0.5% FBS for 3 days until cell cycle arrest was observed. For senescence induction with RO3306 and Nutlin3a, cells were synchronized to G2 phase by treatment with 9 ⁇ M RO3306 (Roche) for 16 to 20 hours, followed by treatment with 9 ⁇ M RO3306 and 5 ⁇ M Nutlin3a (Sigma-Aldrich). The cells were treated for 8 hours, and then 5 ⁇ M nutlin3a was administered for 2 days.
- doxorubicin for DNA damage aging, cells were treated with 100 nM doxorubicin (Sigma) for 24 hours. Cells were then treated with 100 nM BI-2536 until 12 days after induction to remove proliferating cells. Replicative senescent cells were generated by culturing nearly senescent HCA2 cells (41 passages). Doxycycline, epoxomicin, and bortezomib were used at concentrations of 1 ⁇ g/mL, 1 ⁇ M, and 1 ⁇ M, respectively. Cell number was determined by staining cells with 0.4% trypan blue dye and counting viable cells with a hemocytometer.
- Lentivirus-based shRNA constructs and Tet-on inducible lentivirus constructs were generated as previously described (53).
- PCR-generated EcoRI/NotI fragments containing human PD-L1, P130, Flag-E2F97, and NRF1 ⁇ 1-103 cDNAs were inserted into the CSII-CMV-IRES2-Bsd vector (Invitrogen).
- BamHI/NotI PCR fragments containing I ⁇ B ⁇ (S32/36A) and HPV16E7 cDNAs were inserted into the BamHI/NotI digested pENTR-1A vector (Invitrogen) containing the 3xFlag epitope.
- the obtained plasmid was mixed with the CSIV-TRE-RfA-UbC-Puro vector and reacted with Gateway TM LR Clonase TM Enzyme mix (manufactured by Thermo Fisher Scientific) to generate a lentiviral plasmid.
- Lentivirus generation and cell infection were performed as previously described (53).
- Lentiviruses expressing the respective genes were generated by cotransfection of HEK293T cells with pCMV-VSV-G-RSV-RevB, pCAG-HIVgp, and the respective CSII-CMV-IRES2-Bsd, or CSIV-TRE-RfA-UbC-Puro plasmids using calcium phosphate coprecipitation.
- HCA2 cells or MPF infected with the indicated viruses were treated with 10 ⁇ g/mL blasticidin (Gibco) or 2 ⁇ g/mL puromycin (Gibco) for 2 to 4 days.
- doxycycline Sigma-Aldrich
- RNA sequencing> For HCA2 samples, total RNA was extracted using ISOGEN (Nippon Gene) according to the manufacturer's instructions, and quality was assessed using an Agilent Bioanalyzer (Agilent Technologies, Santa Clara, Calif.). PolyA-tailed mRNA was bead-selected from 200 ⁇ g of total RNA using the NEBNext Poly(A) mRNA Magnetic Isolation Module (New England Biolabs, Ipswich, Mass.). RNA-seq libraries were prepared using the NEBNext Ultra II Directional RNA Library Prep Kit (New England Biolabs, Ipswich, Mass.) according to the manufacturer's instructions. Sequencing of 100 nucleotide long paired-end reads was performed on a HiSeq2500 (Illumina, San Diego, CA).
- Genomon (ver. 2.5.2), an in-house pipeline built at the Human Genome Center, University of Tokyo, was used to identify differentially expressed genes (https://github.com/Genomon-Project). Briefly, sequencing data were aligned to the human genome (hg19) using STAR aligner (v2.5.2a) (54). Raw gene counts were obtained from read alignments using HTSeq (v0.11.2) (55). Differential expression analysis was performed using DESeq2 (v1.22.2) (56). Genes with a B-H adjusted P value ⁇ 0.05 were considered significantly differentially expressed.
- 230 genes whose FPKM showed a positive correlation with PD-L1 (R 2 >0.95) were entered into iRegulon (v1.3) (Non-Patent Document 15) on the Cytoscape platform (v3.8.2).
- the 20 kb region centered on the TSS was considered as the promoter region of each gene, and the normalized enrichment score (NES) was set as the threshold with a cutoff of 3.
- Non-Patent Document 57 Sequencing data were aligned to the mouse reference genome (mm10) using Rsubread (v2.4.3) (Non-Patent Document 57). Raw counts were obtained from read alignments via refGene for gene counts and RepeatMasker annotations for TE, and further transferred to CPM by edgeR (v3.32.1) (58). Both the mm10 refGene and RepeatMasker annotation databases are from the University of California, Santa Cruz (UCSC) Genome Browser. After filtering low expressed genes with CPM ⁇ 0.1, differential expression was analyzed with a linear model using limma (v3.46.0) (Non-Patent Document 59).
- Non-Patent Document 60 Genes with log2FC > 0.6 and FDR ⁇ 0.05 adjusted by the BH method are considered as significant differentially expressed genes (DEGs), and are further analyzed using clusterProfiler (v3.18.1) (Non-Patent Document 60) was registered for gene ontology analysis using .
- GSEA gene set enrichment analysis
- a list of genes ranked in order by log2 fold change is input into clusterProfiler's pre-ranked GSEA function, and mouse hallmark gene sets are obtained from msigdb (v7.4.1) It was done. All figures were plotted with enrichplot (v1.10.2).
- In the classification of TEs significantly changed TEs with log2FC > 1 and FDR ⁇ 0.05 were further annotated by Dfam (Release 3.5) (Non-Patent Document 61).
- RNA sequence data analysis For analysis of Tom+/PD-L1+ and Tom+/PD-L1- cells of p16-Tom mice, processed droplet-based single-cell RNA-seq dataset 10 (GSE155182) available from GEO was used.
- Single-cell RNAseq data (Non-Patent Document 62) was processed using the Scanpy package (ver. 1.4.5.post1) on Python (ver. 3.6.8).
- the threshold for gating PD-L1+ cells was defined as a Cd274 UMI count greater than 0.
- DEG differentially expressed gene
- genes with FDR less than 0.05 and log2 fold change greater than 0.15 were identified using the dffxpy package (ver. 0.7.3).
- GSEA gene set enrichment analysis
- a list of genes ranked in order by log2 fold change was input into the pre-ranked GSEA function of clusterProfiler, and the mouse characteristic gene set was extracted from msigdb (v7.4.1). Obtained.
- RNA isolation and real-time PCR> Total RNA from cultured cells was extracted using ISOGEN (Nippon Gene) according to the manufacturer's instructions. For RNA from the selected primary cells, Single Cell RNA Purification Kit (manufactured by Norgen) was used according to the manufacturer's instructions. For qPCR analysis, cDNA was synthesized using ReverTra Ace qPCR RT Master Mix (manufactured by Toyobo). Real-time PCR amplification was performed in a 96-well optical reaction plate using THUNDERBIRD SYBR qPCR Master Mix (Toyobo) and StepOnePlus (Applied Biosystems). The relative expression level of each gene was determined by normalization to the ⁇ -actin expression level of each sample. All primers are listed in Table 1.
- ⁇ Histology> For immunofluorescence staining, kidneys, lungs, liver, and colon preserved in OCT compound were frozen sectioned and stained with DAPI. After imaging the fluorescent signals using INCell Analyzer 2500HS (manufactured by Cytiva), this instrument automatically counted Tom+ cells by recognizing DAPI signals as nuclei through IN Cell Developer Toolbox v1.9.2. The percentage was calculated. Tom+ signal was 10 times background. To determine the proportion of PD-L1+ cells among Tom+ cells, anti-PD-L1 (Abcam, clone: EPR20529) and anti-CD3 (Abcam, clone: KT3) staining was performed.
- PD-L1 intensity was quantified for each Tom+ cell, excluding CD3+ cells and cells exhibiting strong autofluorescence (such as oil droplets in NASH samples).
- PD-L1+ cells were recognized by PD-L1 intensity that was 1.8 times (for kidney) or 2.5 times (for lung and liver) greater than the median. 10-30 fields of each organ section were randomly selected and out-of-focus images were further excluded.
- the representative image in Figure 1b was observed on a Carl Zeiss LSM 710 NLO (Zeiss). All antibodies are listed in Tables 2 and 3.
- liver and lungs were fixed with paraffin, sectioned, and stained with H&E or Sirius Red.
- the stained tissue sections were observed using an Olympus BX51 microscope (manufactured by Olympus) under bright field illumination.
- Olympus BX51 microscope manufactured by Olympus
- Sirius Red area via ImageJ software, define the area using the Huang threshold, 1500, 2000, and 400, respectively, in three fields for each section. The average area over the pixels was measured.
- CD8+ T cells were plated in a 24-well plate precoated with 5 ⁇ g/mL anti-CD3 ⁇ (BioLegend, clone: 145-2C11) and 5 ⁇ g/mL anti-CD28 (BioLegend, clone: 37.51). , and cultured for 72 hours.
- Non-Patent Document 64 Activated CD8+ T cells were collected and co-cultured with target cells at a ratio of 5:1 for 18 hours.
- OVA 257-264 ovalbumin peptide
- SynPeptide ovalbumin peptide
- anti-CD45 manufactured by BioLegend, clone: 30-F11
- propidium iodide 50 ⁇ g/mL staining were performed to exclude effector cells, and cytotoxicity was evaluated.
- Cells were analyzed by BD FACSCanto TM II (BD Biosciences). All antibodies are listed in Tables 2 and 3.
- liver or spleen was harvested from mice and triturated with the flat end of a syringe over a 70 ⁇ m cell strainer in 3 mL of PBS. All cells that passed were collected in a tube and pelleted by centrifugation at 400 g for 5 minutes. After RBC lysis (manufactured by Thermo Fisher) and FcR blocking (manufactured by Miltenyi Biotec), cells were treated with anti-CD45 (manufactured by BioLegend, clone: 30-F11), anti-CD44 (manufactured by BioLegend, clone: IM7), and anti-CD3.
- RBC lysis manufactured by Thermo Fisher
- FcR blocking manufactured by Miltenyi Biotec
- ProteoStat staining cells were labeled with anti-PD-L1 (Abcam, clone: 28-8) and AF405-conjugated anti-rabbit IgG (Abcam), and then the cell suspension was added to PBS containing 4% formaldehyde. It was added dropwise and incubated at room temperature for 10 minutes. Cells were washed once with wash buffer, and the cell suspension was added dropwise to permeabilization solution (0.2% Triton X-100, 4% FBS in PBS) and incubated for 5 minutes at room temperature. After washing the cells with a washing buffer, they were stained with ProteoStat staining solution (1/5000-fold dilution) at room temperature for 30 minutes. AF405 and ProteoStat (PE-Texas Red channel) signals were analyzed using BD FACSAria SORP (manufactured by BD Biosciences). All antibodies are listed in Tables 2 and 3.
- ⁇ Proteasome activity assay> After sorting PD-L1+ cells and PD-L1- cells, 10,000 cells were seeded into a white 96-well plate. Chymotrypsin, trypsin, and caspase-like proteasome activities were tested using the Proteasome-Glo Cell-Based Assay kit (Promega) according to the manufacturer's instructions. The luminescence intensity was measured using FLUOstar OPTIMA (manufactured by BMG LABTECH).
- AST, ALT, LDH, and total cholesterol (T-CHO) levels Blood was collected from mice by cardiac puncture and left at room temperature for 15 min. To obtain serum, samples were centrifuged at 3400 g for 15 min at 4°C. Serum samples with severe hemolysis were excluded from further analysis of AST, ALT, and LDH (one sample from a mouse receiving normal diet and ABT263 treatment). Serum levels of AST, ALT, LDH, and T-CHO were measured by Fujifilm Wako Pure Chemical Corporation (Japan).
- Grip strength was measured using a BIO-GS3 grip strength testing device (manufactured by BIOSEB). I grabbed the mouse by its tail and had it grip a metal grid with its two front paws. As the grid is pulled backwards in a horizontal plane until it is released, the strength of the force displayed on the device is recorded.
- a rotarod model MK-630B (Muromachi Co., Ltd.) was used. The rotation speed was set to 30 rpm, and the time the mouse stayed on the rod from the start of rotation until it fell onto the magnetic switch was recorded. To prevent learning effects, both grip strength and rotarod tests were performed three times on the same day. The average value for each mouse was used for statistical inference.
- Example 1 The properties of senescent cells in vivo vary depending on their origin and stimulus, and certain types of senescent cells may accumulate with aging. Immune surveillance against incompetent cells, such as cancer cells, is negatively regulated by immune checkpoints. Therefore, it was thought that this might also apply to senescent cells. To address this question, we investigated the expression of several immune checkpoints in the human fibroblast cell line HCA2, a well-established in vitro model of aging fibroblasts. was analyzed.
- Non-Patent Document 5 nutlin-3a-induced cells
- d-Sen DNA damage-induced senescent cells
- PD-1 programmed cell death protein 1
- PD-L1 a receptor for PD-L1
- PD-L1 prevents cytotoxicity caused by attack of PD-1-expressing T cells, and therefore, PD-L1 expression in senescent cells may escape immune surveillance and lead to age-related accumulation of senescent cells. I hypothesized that there is.
- p16-CreERT2-tdTomato mouse model p16-Tom
- treated these cells p16 high cells
- tdTomato-expressing cells Tom + cells.
- Non-Patent Document 10 It was visualized by fluorescence imaging (Non-Patent Document 10).
- SASP senescence-associated secretory phenotype
- FIG. 1b Some Tom + cells expressing PD-L1 were detected (Fig. 1b), and the population of Tom + cells and PD-L1-expressing cells within Tom + cells was smaller than in the younger group (2 months old). It was significantly higher in the older age group (24-26 months). (Fig. 1c, Fig. 5c).
- PD-L1 levels were high in proliferating cells and low in quiescent and senescent cells (Fig. 6a).
- PD-L1 transcription was cell cycle dependent and was highest in S phase (Fig. 6b).
- p130 a dominant pocket protein that forms the DREAM complex and suppresses E2F activity in quiescent cells and senescent cells (Non-Patent Documents 11, 12), or the dominant-negative mutant E2F (E2F97) (Non-Patent Document 13). Both suppressed PD-L1 expression when overexpressed in proliferating cells ( Figures 6c and 6d).
- PD-L1 ⁇ and PD-L1 + d-Sen HCA2 cells were sorted for bulk RNA-seq analysis (Fig. 6f).
- the present inventors constructed a transcriptional regulatory network of 230 PD-L1 positively correlated genes (R 2 >0.95) using the transcription factor binding motif enrichment tool iRegulon (Non-Patent Documents 15, 16). ( Figure 6g).
- Non-Patent Documents 17, 18 the heterogeneity of PD-L1 expression in senescent cells may be due to individual This may be due to transcription-independent mechanisms such as proteasome activity in senescent cells. This idea was supported by the fact that levels of protein aggregates differ between senescent cells. In PD-L1 + senescent cells, chymotrypsin-like protease activity, trypsin-like protease activity, and caspase-like protease activity (Non-Patent Document 19) were lower than in PD-L1 ⁇ cells (Fig. 1f).
- Example 2 Next, we investigated whether PD-L1 functions as an immune checkpoint for T cell immunity during cellular senescence.
- Mouse activated primary CD8 + T cells were co-cultured as effector cells and primary mouse lung fibroblasts (MPF) as target cells, and a syngeneic in vitro T cell killing assay was performed to analyze cytotoxicity.
- Senescence induction was quantified by SA- ⁇ -gal staining and p16 expression in n-Sen and d-Sen MPFs ( Figures 7a and 7b). In MPF, heterogeneous expression of PD-L1 was observed in senescent cells but not in quiescent cells (Fig. 2a).
- senescent cells were more sensitive to T cells than quiescent cells in all induction methods (Fig. 2b, Fig. 7c).
- SASP is a typical phenotype of cellular aging (Non-Patent Document 22), and is also known to be involved in immune surveillance of senescent cells by attracting immune cells to damaged tissues (Non-Patent Document 23). ). Differences in transcriptional signatures between n-Sen MPF and Quie MPF were investigated by bulk RNA-seq (Fig. 2c).
- TEs transposable elements
- ERVs TE-encoded endogenous retroviral elements
- Non-Patent Document 26 a positive regulator of the MHC class I antigen processing machinery
- senescent cells a positive regulator of the MHC class I antigen processing machinery
- RNA-sequencing RNA-sequencing
- LSEC liver sinusoidal endothelial cells
- NASH nonalcoholic steatohepatitis
- NASH-associated macrophages NASH liver
- a rich set of cell types were enrolled, including vascular cells from normal kidney (Fig. 3a, Fig. 9a).
- Immune checkpoint blockade such as treatment with monoclonal antibodies against PD-1 or PD-L1 has contributed to significant advances in cancer immunotherapy over the past decade, redirecting T cells to PD-L1-expressing cancer cells. has been successfully used to treat various malignant tumors (Non-Patent Document 29).
- Non-Patent Document 29 To assess the effect of anti-PD-1 ( ⁇ PD-1) antibodies on senescent cell dynamics in vivo, we inoculated 7-month-old p16-Tom mice that had been labeled with TAM for senescent cells 2 weeks earlier. ⁇ PD-1 antibody or isotype control (IgG) was administered for 3 weeks ( Figure 4a).
- cytotoxic T cells were systemically reduced by anti-CD8 ( ⁇ CD8) antibody treatment throughout the experiment ( Figure 10a).
- the total number of Tom + cells in the lungs, liver and kidneys of mice injected with ⁇ PD-1 antibody was significantly lower than in the IgG group (Fig. 4a, Fig. 10b).
- ⁇ PD-1 antibody treatment decreased the PD-L1 + subpopulation in Tom + cells, but not in Tom ⁇ cells (Fig. 10b).
- ⁇ PD-1 treatment reduced the population of PD-L1 ⁇ /Tom + cells to a lesser extent than PD-L1 + /Tom + cells.
- Non-Patent Document 31 age-related increases in alveolar volume
- Non-Patent Document 32 hepatic lipidosis
- Non-Patent Document 33 decrease in grip strength
- Non-Patent Document 33 decrease in exercise capacity
- NASH choline-deficient L-amino acid defined high-fat diet
- Non-Patent Document 36 choline-deficient L-amino acid defined high-fat diet
- Figure 4e ⁇ PD-1 antibody or IgG for 3 weeks.
- the present inventors further found that the elimination of PD-L1 + senescent cells by immune checkpoint blockade (ICB) therapy is more effective than the senolytic drug ABT263 in the pathogenesis of NASH (37).
- IDB immune checkpoint blockade
- ⁇ PD-1 antibody and ABT263 ameliorated the pathogenesis, but the ⁇ PD-1 group showed more improvement in hepatic steatosis than the ABT263 group (Fig. 4e).
- body weight and serum total cholesterol (T-CHO) levels showed similar trends (Fig. 12d), suggesting that PD-L1 + senescent cell clearance increases LDL secretion from the liver and reduces lipid accumulation in the progression of NASH. suggested that it was possible.
- senescent cells are highly heterogeneous depending on the cell type and the cause of senescence induction in vivo. Therefore, senescent cells may exhibit heterogeneity even among cells of the same type.
- PD-L1 + senescent cells showed much more inflammatory transcriptome than PD- L1 ⁇ cells, even within the same cell type (LSEC, NAM, or renal vascular cells). Therefore, the accumulation of PD-L1 + senescent cells may have more deleterious consequences than a simple increase in the number of senescent cells with age. Therefore, selective inhibition of PD-L1 + senescence accumulation by ICB treatment is a more promising senescence treatment strategy than conventional senescence treatments.
- Non-Patent Documents 43, 44 It has been reported that in Alzheimer's disease models, blocking the PD-L1/PD-1 axis alleviates the disease state and improves cognitive impairment. It has been reported that relatively long-term (8 weeks) administration of ⁇ PD-1 antibody to a NASH model with 10 months of CDA-HFD induction increases the burden of hepatocellular carcinoma (HCC) (Non-patent literature). 45). Furthermore, long-term ⁇ PD-1 antibody administration often induced systemic autoimmune diseases (Non-Patent Document 46). However, these were not observed with our short-term treatment (3 weeks of ⁇ PD-1 antibody treatment followed by 10 weeks of CDA-HFD induction). Therefore, controlling immunotherapy in age-related diseases requires not only reducing the dose and frequency, but also considering the balance between enhanced immune clearance, resistance to acute inflammation, and tissue repair rate. It may be necessary to optimize the use of each treatment with consideration to reducing
Landscapes
- Health & Medical Sciences (AREA)
- Chemical & Material Sciences (AREA)
- Medicinal Chemistry (AREA)
- Life Sciences & Earth Sciences (AREA)
- Animal Behavior & Ethology (AREA)
- Veterinary Medicine (AREA)
- Public Health (AREA)
- Pharmacology & Pharmacy (AREA)
- General Health & Medical Sciences (AREA)
- Engineering & Computer Science (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Chemical Kinetics & Catalysis (AREA)
- General Chemical & Material Sciences (AREA)
- Organic Chemistry (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Epidemiology (AREA)
- Diabetes (AREA)
- Obesity (AREA)
- Immunology (AREA)
- Hematology (AREA)
- Microbiology (AREA)
- Mycology (AREA)
- Gastroenterology & Hepatology (AREA)
- Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
- Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
Abstract
Description
本発明は、老化細胞除去剤、及びこれを用いた各種老化に伴う疾患の改善方法に関する。また、本発明は、非アルコール性脂肪肝炎(NASH)の治療又は予防のための医薬組成物に関する。
本願は、2022年9月20日に米国に出願された米国特許第63/408105号に基づき優先権を主張し、その内容をここに援用する。
The present invention relates to a senescent cell removing agent and a method for improving various diseases associated with aging using the same. The present invention also relates to a pharmaceutical composition for the treatment or prevention of non-alcoholic steatohepatitis (NASH).
This application claims priority from US Patent No. 63/408105, filed in the United States on September 20, 2022, the contents of which are hereby incorporated by reference.
老化の蓄積は、加齢に伴う炎症の主な原因の1つであり、様々な加齢に関連した病気の原因となる(非特許文献1)。しかし、この蓄積の根底にある分子基盤と、老化プロセスを改善する標的としてのその可能性については、ほとんど知られていない。 Accumulation of aging is one of the main causes of age-related inflammation and causes various age-related diseases (Non-Patent Document 1). However, little is known about the molecular basis underlying this accumulation and its potential as a target to ameliorate the aging process.
いくつかの証拠は、加齢に伴い老化細胞が様々な組織に蓄積し、過剰な炎症を引き起こし、その結果、組織の恒常性の不均衡を引き起こすことを示唆している。この蓄積の簡単な説明は、テロメア短縮やDNA損傷などの老化誘発刺激が、年齢に依存して増加することである。がん遺伝子によって誘導された老化肝細胞と損傷によって誘導された老化星細胞が、それぞれ活性化T細胞とナチュラルキラー細胞によって排除されることが、最近報告されているため、免疫系障害も関与している可能性がある(非特許文献2及び3)。 しかし、免疫系が自然な老化中の細胞老化をどのように監視しているかについては、正確にはほとんどわかっていない。 Several lines of evidence suggest that senescent cells accumulate in various tissues with age, causing excessive inflammation and, consequently, tissue homeostasis imbalance. A simple explanation for this accumulation is an age-dependent increase in senescence-inducing stimuli, such as telomere shortening and DNA damage. Immune system impairment may also be involved, as it has been recently reported that oncogene-induced senescent hepatocytes and injury-induced senescent stellate cells are eliminated by activated T cells and natural killer cells, respectively (Non-Patent Documents 2 and 3). However, exactly how the immune system monitors cellular senescence during natural aging remains largely unknown.
本発明は、NASHの治療又は予防に用いられる医薬組成物、及び老化細胞除去剤を提供することを主たる目的とする。 The main object of the present invention is to provide a pharmaceutical composition and a senescent cell remover for use in the treatment or prevention of NASH.
本発明者らは、鋭意研究した結果、細胞膜上のPD-L1がPD-1と結合することを阻害することにより、NASHモデル動物において、老化細胞が除去され、肝臓での脂肪蓄積や線維化が有意に減少し、それに伴い肝機能の指標も改善することを見出し、本発明を完成させた。 As a result of extensive research, the present inventors have found that by inhibiting PD-L1 on the cell membrane from binding to PD-1, senescent cells are removed in NASH model animals, leading to fat accumulation and fibrosis in the liver. The inventors have completed the present invention by discovering that liver function is significantly reduced and that indicators of liver function are also improved accordingly.
本発明に係る医薬組成物等は、下記の通りである。
[1] 細胞膜上のPD-L1がPD-1と結合することを阻害する物質を有効成分とする、非アルコール性脂肪肝炎(NASH)の治療又は予防のための医薬組成物。
[2] 前記物質が、抗PD-L1抗体及び抗PD-1抗体からなる群より選択される一種である、前記[1]の医薬組成物。
[3] 前記物質が、PD-L1と特異的に結合する低分子化合物である、前記[1]の医薬組成物。
[4] 前記PD-L1は、肝臓細胞の細胞膜タンパク質であり、前記PD-1は、CD8陽性T細胞の細胞膜タンパク質である、前記[1]~[3]のいずれかの医薬組成物。
[5] NASHを発症した動物に、1回のみ、投与される、前記[1]~[4]のいずれかの医薬組成物。
[6] 前記NASHを発症した動物が、肝臓がん以外のがんを発症していない、前記[5]の医薬組成物。
[7] 抗PD-L1抗体及び抗PD-1抗体からなる群より選択される一種以上を有効成分とする、p16陽性老化細胞の除去剤。
Pharmaceutical compositions and the like according to the present invention are as follows.
[1] A pharmaceutical composition for the treatment or prevention of non-alcoholic steatohepatitis (NASH), which contains as an active ingredient a substance that inhibits the binding of PD-L1 on cell membranes to PD-1.
[2] The pharmaceutical composition of [1] above, wherein the substance is one selected from the group consisting of anti-PD-L1 antibodies and anti-PD-1 antibodies.
[3] The pharmaceutical composition of [1] above, wherein the substance is a low molecular compound that specifically binds to PD-L1.
[4] The pharmaceutical composition according to any one of [1] to [3], wherein the PD-L1 is a cell membrane protein of liver cells, and the PD-1 is a cell membrane protein of CD8-positive T cells.
[5] The pharmaceutical composition according to any one of [1] to [4] above, which is administered only once to an animal that has developed NASH.
[6] The pharmaceutical composition of [5] above, wherein the animal that has developed NASH has not developed cancer other than liver cancer.
[7] An agent for removing p16-positive senescent cells, which contains as an active ingredient one or more selected from the group consisting of anti-PD-L1 antibodies and anti-PD-1 antibodies.
本発明に係る医薬組成物及び老化細胞除去剤により、肝臓内のp16陽性(p16+)老化細胞の割合を減少させることができ、肝臓内の機能をより正常化させることができる。このため、本発明に係る医薬組成物は、NASHの治療又は予防に有効である。 The pharmaceutical composition and senescent cell removal agent according to the present invention can reduce the proportion of p16-positive (p16 + ) senescent cells in the liver, and can further normalize the functions in the liver. Therefore, the pharmaceutical composition according to the present invention is effective in treating or preventing NASH.
本発明者らは、老化細胞が免疫チェックポイントPD-L1を不均一に発現し、生体内で加齢とともにPD-L1+老化細胞が蓄積することを示した。PD-L1-細胞は、T細胞監視に対して感受性があるが、PD-L1+細胞は老化関連分泌表現型(SASP)の存在下でも耐性がある。興味深いことに、in vivoでのp16+細胞のシングルセル分析により、PD-L1発現がより高いレベルのSASPと相関していることが明らかになった。これと一致して、自然老化マウス又はNASH誘発マウスに抗PD-1抗体を投与すると、in vivoでp16+細胞の総数が減少するだけでなく、活性化されたCD8+T細胞依存的にPD-L1+集団も減少し、加齢に伴う病気の様々な症状が改善される。これらの結果は、PD-L1の不均一な発現が老化細胞の蓄積と老化に伴う炎症に重要な役割を果たしており、免疫チェックポイント阻害によるPD-L1+老化細胞の除去が、抗老化療法の有望な戦略となる可能性があることを示唆している。 We showed that senescent cells heterogeneously express the immune checkpoint PD-L1 and that PD-L1 + senescent cells accumulate with age in vivo. PD-L1 − cells are sensitive to T cell surveillance, whereas PD-L1 + cells are resistant even in the presence of senescence-associated secretory phenotype (SASP). Interestingly, single cell analysis of p16 + cells in vivo revealed that PD-L1 expression correlated with higher levels of SASP. Consistent with this, administration of anti-PD-1 antibodies to naturally aging or NASH-induced mice not only reduces the total number of p16 + cells in vivo but also reduces PD in an activated CD8 + T cell-dependent manner. -L1 + population also decreases, and various symptoms of age-related diseases are improved. These results suggest that the heterogeneous expression of PD-L1 plays an important role in senescent cell accumulation and aging-associated inflammation, and that the removal of PD-L1 + senescent cells by immune checkpoint inhibition may be an effective strategy for anti-aging therapy. This suggests that it may be a promising strategy.
後記実施例に示すように、PD-L1とPD-1の結合を阻害することにより、老化細胞(p16+細胞)の蓄積が抑制される。これは、PD-L1陽性(PD-L1+)の老化細胞は、CD8陽性T細胞(CD8+T細胞)上のPD-1と結合してCD8+T細胞の活性を抑制して免疫監視を回避しているが、PD-L1とPD-1の結合を阻害することにより、CD8+T細胞の活性を抑制することができず、PD-L1+老化細胞はCD8+T細胞により除去されるためである。 As shown in the Examples below, the accumulation of senescent cells (p16 + cells) is suppressed by inhibiting the binding between PD-L1 and PD-1. This is because PD-L1-positive (PD-L1 + ) senescent cells bind to PD-1 on CD8-positive T cells (CD8 + T cells), suppress the activity of CD8 + T cells, and perform immune surveillance. However, by inhibiting the binding of PD-L1 and PD-1, the activity of CD8 + T cells cannot be suppressed, and PD-L1 + senescent cells are eliminated by CD8 + T cells. It's for a reason.
NASHの病態には、細胞老化が重要な役割を果たしている。後記実施例に示すように、老化細胞(p16+細胞)の肝臓での存在量を低減させることにより、NASHの病態を改善させる、具体的には、肝臓の脂質量の増大、線維化、AST、ALT、及びLDHの血清レベルの上昇を改善させることができる。 Cellular aging plays an important role in the pathology of NASH. As shown in the Examples below, by reducing the amount of senescent cells (p16 + cells) in the liver, the pathological condition of NASH is improved. , ALT, and LDH serum levels can be improved.
本実施形態の医薬組成物は、細胞膜上のPD-L1がPD-1と結合することを阻害する物質を有効成分とする、NASHの治療又は予防のための医薬組成物である。細胞膜上のPD-L1がPD-1と結合することを阻害することにより、老化細胞を除去することで、NASHの病態を改善することができる。この作用機序により、本実施形態の医薬組成物は、NASHの治療や予防に有効である。 The pharmaceutical composition of the present embodiment is a pharmaceutical composition for treating or preventing NASH, which contains as an active ingredient a substance that inhibits PD-L1 on the cell membrane from binding to PD-1. By inhibiting the binding of PD-L1 on the cell membrane to PD-1, the pathological condition of NASH can be improved by removing senescent cells. Due to this mechanism of action, the pharmaceutical composition of this embodiment is effective in treating and preventing NASH.
本実施形態の医薬組成物の有効成分は、細胞膜上のPD-L1がPD-1と結合することを阻害することができる物質であれば、特に限定されるものではない。PD-L1と結合してPD-1との結合を阻害する物質であってもよく、PD-1と結合してPD-L1との結合を阻害する物質であってもよい。より効果的にNASHの予防又は治療の効果が得られるため、本実施形態の医薬組成物の有効成分としては、肝臓細胞の細胞膜タンパク質であるPD-L1と、CD8+T細胞の細胞膜タンパク質であるPD-1との結合を阻害する物質であることが好ましい。 The active ingredient of the pharmaceutical composition of the present embodiment is not particularly limited as long as it is a substance that can inhibit PD-L1 on the cell membrane from binding to PD-1. It may be a substance that binds to PD-L1 and inhibits the binding to PD-1, or it may be a substance that binds to PD-1 and inhibits the binding to PD-L1. Since the effect of preventing or treating NASH can be obtained more effectively, the active ingredients of the pharmaceutical composition of this embodiment include PD-L1, which is a cell membrane protein of liver cells, and a cell membrane protein of CD8 + T cells. Preferably, it is a substance that inhibits binding to PD-1.
本実施形態の医薬組成物の有効成分としては、例えば、抗PD-L1抗体及び抗PD-1抗体からなる群より選択される一種以上が挙げられる。抗PD-L1抗体としては、公知のいずれの抗PD-L1抗体を用いてもよく、公知の抗体をPD-L1に対する親和性を損なうことなく適宜改変させた抗体であってもよい。抗PD-1抗体としては、公知のいずれの抗PD-1抗体を用いてもよく、公知の抗体をPD-1に対する親和性を損なうことなく適宜改変させた抗体であってもよい。抗体の特異性や安全性の改善は、古くから行われている技術であり、これらの技術を利用して適宜行うことができる。また、例えば、免疫チェックポイント阻害療法等で使用され得る抗PD-L1抗体及び抗PD-1抗体も、本実施形態の医薬組成物の有効成分として用いることができる。 Examples of the active ingredient of the pharmaceutical composition of the present embodiment include one or more selected from the group consisting of anti-PD-L1 antibodies and anti-PD-1 antibodies. As the anti-PD-L1 antibody, any known anti-PD-L1 antibody may be used, or it may be an antibody obtained by appropriately modifying a known antibody without impairing its affinity for PD-L1. As the anti-PD-1 antibody, any known anti-PD-1 antibody may be used, or it may be an antibody obtained by appropriately modifying a known antibody without impairing its affinity for PD-1. Improving the specificity and safety of antibodies is a technique that has been used for a long time, and can be carried out as appropriate using these techniques. Furthermore, for example, anti-PD-L1 antibodies and anti-PD-1 antibodies that can be used in immune checkpoint inhibition therapy and the like can also be used as active ingredients in the pharmaceutical composition of the present embodiment.
本実施形態の医薬組成物の有効成分としては、例えば、PD-L1に特異的に結合する低分子化合物や、PD-1に特異的に結合する低分子化合物であってもよい。また、本実施形態の医薬組成物の有効成分としては、PD-L1やPD-1に特異的に結合する核酸アプタマーやペプチドであってもよい。PD-L1やPD-1に特異的に結合する低分子化合物等は、各種のライブラリからスクリーニングして求めることができる。また、PD-L1やPD-1の構造データを利用して合成することもできる。 The active ingredient of the pharmaceutical composition of the present embodiment may be, for example, a low-molecular compound that specifically binds to PD-L1 or a low-molecular compound that specifically binds to PD-1. Further, the active ingredient of the pharmaceutical composition of the present embodiment may be a nucleic acid aptamer or a peptide that specifically binds to PD-L1 or PD-1. PD-L1 and low-molecular compounds that specifically bind to PD-1 can be obtained by screening various libraries. Furthermore, it is also possible to synthesize using the structural data of PD-L1 and PD-1.
細胞膜上のPD-L1がPD-1と結合することを阻害することにより、肝臓内のp16陽性(p16+)老化細胞の割合を減少させることができる。このため、本実施形態の医薬組成物の有効成分は、老化細胞除去剤、特に、p16陽性老化細胞の除去剤としても機能する。 By inhibiting the binding of PD-L1 on the cell membrane to PD-1, the proportion of p16-positive (p16 + ) senescent cells in the liver can be reduced. Therefore, the active ingredient of the pharmaceutical composition of this embodiment also functions as an agent for removing senescent cells, in particular, an agent for removing p16-positive senescent cells.
本実施形態の医薬組成物は、有効成分である細胞膜上のPD-L1がPD-1と結合することを阻害することができる物質と、薬学的に許容される担体とを適宜混合することにより、調製できる。医薬組成物の製造は、必要に応じて適宜添加剤を用いることにより、医薬品の製造の分野で通常用いられる方法により行うことができる。 The pharmaceutical composition of this embodiment can be prepared by appropriately mixing a substance capable of inhibiting the binding of PD-L1 on cell membranes with PD-1, which is an active ingredient, and a pharmaceutically acceptable carrier. , can be prepared. The pharmaceutical composition can be manufactured by a method commonly used in the field of pharmaceutical manufacturing by using appropriate additives as necessary.
薬学的に許容しうる担体とは、投与対象に有害な生理学的反応を引き起こさず、かつ薬効成分等の他の成分と有害な相互作用を生じないような希釈剤、賦形剤、結合剤、溶媒等である。当該担体としては、具体的には、例えば、水、生理食塩水、各種緩衝液等が用いられる。また、使用できる添加剤としては、アジュバント、希釈剤、賦形剤、結合剤、安定剤、等張化剤、緩衝剤、溶解補助剤、懸濁化剤、保存剤、凍害防止剤、凍結保護剤、凍結乾燥保護剤、静菌剤等が挙げられる。 Pharmaceutically acceptable carriers are diluents, excipients, binders, Solvents, etc. As the carrier, specifically, for example, water, physiological saline, various buffer solutions, etc. are used. In addition, additives that can be used include adjuvants, diluents, excipients, binders, stabilizers, tonicity agents, buffers, solubilizing agents, suspending agents, preservatives, antifreeze agents, and cryoprotectants. agents, lyoprotectants, bacteriostatic agents, etc.
本実施形態の医薬組成物が投与される動物は、NASHの治療や予防が求められる動物であれば、特に限定されるものではなく、ヒトであってもよく、ヒト以外の動物であってもよいが、哺乳類であることが好ましい。非ヒト哺乳類動物としては、ウシ、ブタ、ウマ、ヒツジ、ヤギ、サル、イヌ、ネコ、ウサギ、マウス、ラット、ハムスター、モルモット等が挙げられる。NASHに対するより充分な治療効果が期待できる点から、本実施形態の医薬組成物が投与される動物としては、肝臓がん以外のがんを発症していない動物であることが好ましい。また、本実施形態の医薬組成物を動物に投与する際の投与経路は、特に限定されるものではなく、経口投与、経静脈投与、経腸投与、筋肉内投与、皮下投与、経皮投与、経鼻投与、経肺投与等が挙げられる。 The animal to which the pharmaceutical composition of the present embodiment is administered is not particularly limited as long as it is an animal for which treatment or prevention of NASH is required, and may be a human or a non-human animal. However, mammals are preferred. Non-human mammals include cows, pigs, horses, sheep, goats, monkeys, dogs, cats, rabbits, mice, rats, hamsters, guinea pigs, and the like. Since a more sufficient therapeutic effect on NASH can be expected, the animal to which the pharmaceutical composition of this embodiment is administered is preferably an animal that has not developed cancer other than liver cancer. Furthermore, the administration route for administering the pharmaceutical composition of the present embodiment to animals is not particularly limited, and may include oral administration, intravenous administration, enteral administration, intramuscular administration, subcutaneous administration, transdermal administration, Examples include nasal administration and pulmonary administration.
本実施形態の医薬組成物の1回当たりの投与量は、有効成分による老化細胞除去効果が得られる量であれば、特に限定されるものではなく、投与される動物の生物種、性別、年齢、体重、基礎疾患の有無、病態等を考慮して適宜決定することができる。例えば、免疫チェックポイント阻害療法等で使用されている抗PD-L1抗体又は抗PD-1抗体を有効成分とする場合には、免疫チェックポイント阻害療法等で使用されている投与量を参考にして、本実施形態の医薬組成物の投与量を決定することができる。 The dosage per administration of the pharmaceutical composition of the present embodiment is not particularly limited as long as the active ingredient can achieve the effect of removing senescent cells, and the dosage may include the species, sex, and age of the animal to which it is administered. , weight, presence or absence of underlying disease, pathological condition, etc. can be appropriately determined. For example, when using an anti-PD-L1 antibody or anti-PD-1 antibody used in immune checkpoint inhibition therapy as an active ingredient, refer to the dosage used in immune checkpoint inhibition therapy, etc. , the dosage of the pharmaceutical composition of this embodiment can be determined.
本実施形態の医薬組成物の用法用量は、有効成分による老化細胞除去効果が得られる量であれば、特に限定されるものではないが、副作用が抑えられる点から、投与期間は短期間であることが好ましい。例えば、本実施形態の医薬組成物は、NASHを発症しその治療が必要な動物に対して、その投与期間が3週間以内に複数回の投与であることが好ましく、2週間以内に複数回の投与であることがより好ましく、1週間以内に複数回の投与であることがさらに好ましく、1回のみ投与されることが特に好ましい。 The dosage of the pharmaceutical composition of the present embodiment is not particularly limited as long as the active ingredient has the effect of removing senescent cells, but the administration period is short in order to suppress side effects. It is preferable. For example, the pharmaceutical composition of the present embodiment is preferably administered multiple times within 3 weeks, and preferably multiple times within 2 weeks, to animals that have developed NASH and require treatment. More preferably, it is administered multiple times within one week, and particularly preferably only once.
次に、実施例等により本発明をさらに詳細に説明するが、本発明はこれらの例によって限定されるものではない。 Next, the present invention will be explained in more detail with reference to examples, but the present invention is not limited to these examples.
[材料と方法] [Materials and Methods]
<マウス>
マウスは、湿度管理された室内で、周囲温度23~25℃のケージに2~5匹ずつ入れ、標準的な餌(CA-1、日本クレア社製)を与えて、12時間の明暗サイクル(08:00~20:00点灯)で維持した。水は、環境エンリッチメントへのアクセスを自由に提供された。全ての動物は、東京大学医科学研究所の動物実験ガイドライン及び施設内実験動物管理に従って扱われた。全てのp16-Tomマウスはヘテロ接合性であり、p16Ink4a-CreERT2マウス(非特許文献10)とRosa26-CAG-LSL-tdTomatoマウス(Jackson Laboratoryより入手)を交配することによって生まれた。同じ実験の全てのグループで、年齢が一致し、同じ系統の雄マウスを使用した。 C57BL/6 OT-1-TCRトランスジェニックマウスは、既に開示されている(非特許文献47、48)。このマウスでは、ほとんどのCD8+T細胞がOVAペプチド/MHCクラスI複合体に対して反応性である。OT-1マウスは、慶応義塾大学の特定病原体フリー施設で飼育された。使用したマウスは、全て社内で交配させたものである。全てのp16-Tomマウスは、タモキシフェン(TAM、80mg/kg BW)を5日間、指定された時間に毎日腹腔内投与した。ICB処理では、全てのマウスに、250μg/用量のアイソタイプコントロールIgG(BioLegend社製、クローン: RTK2758) を3日間、又は、抗マウスPD-1(BioLegend社製、クローン: 29F.1A12) を3週間以内に8回、腹腔内投与し、そして最後の注射から1日後にマウスを屠殺した。CD8+T細胞を枯渇させるために、全てのマウスに、週に2回、200μg/用量のアイソタイプ コントロールIgG(Bio X Cell社製、クローン: 2A3) 又は抗マウスCD8a (Bio X Cell社製、クローン: 53-6.7) を7週間以内に14回、腹腔内投与した(非特許文献49)。
<Mouse>
Mice were placed in cages of 2 to 5 mice at an ambient temperature of 23 to 25°C in a humidity-controlled room, fed standard food (CA-1, manufactured by CLEA Japan), and kept on a 12-hour light-dark cycle ( The lighting was maintained from 08:00 to 20:00). Water was provided freely with access to environmental enrichment. All animals were treated in accordance with the Animal Experiment Guidelines and Institutional Laboratory Animal Management of the Institute of Medical Science, The University of Tokyo. All p16-Tom mice are heterozygous and were generated by crossing p16 Ink4a -Cre ERT2 mice (Non-Patent Document 10) with Rosa26-CAG-LSL-tdTomato mice (obtained from Jackson Laboratory). Age-matched male mice of the same strain were used in all groups in the same experiment. C57BL/6 OT-1-TCR transgenic mice have already been disclosed (Non-Patent Documents 47, 48). In this mouse, most CD8 + T cells are reactive to OVA peptide/MHC class I complexes. OT-1 mice were housed in a specific pathogen-free facility at Keio University. All mice used were bred in-house. All p16-Tom mice received tamoxifen (TAM, 80 mg/kg BW) intraperitoneally daily at the indicated times for 5 days. For ICB treatment, all mice received 250 μg/dose of isotype control IgG (BioLegend, clone: RTK2758) for 3 days or anti-mouse PD-1 (BioLegend, clone: 29F.1A12) for 3 weeks. 8 intraperitoneal injections were given within 8 days, and mice were sacrificed 1 day after the last injection. To deplete CD8 + T cells, all mice were treated with 200 μg/dose of isotype control IgG (Bio X Cell, clone: 2A3) or anti-mouse CD8a (Bio X Cell, clone: 2A3) twice weekly. : 53-6.7) was intraperitoneally administered 14 times within 7 weeks (Non-Patent Document 49).
非アルコール性脂肪性肝炎(NASH)の誘導のために、生後1.5か月p16ーCreERT2-tdTomatoマウスを、脂肪60kcal%、メチオニン0.1%、コリン無添加のL-アミノ酸食(CDA-HFD:A06071302、Research Diets社製)で維持することを合計10週間続けた。老化細胞破壊試薬治療のために、全てのマウスに、プラセボ(ビヒクル対照)又はABT263を強制経口投与した。 ABT263 (Selleck社製) をエタノール:ポリエチレングリコール400:Phosal 50 PGを10:30:60で懸濁し、その後1サイクルあたり7日間、50mg/kg BWを毎日投与した。サイクル間に2週間の間隔を置いて合計2サイクルを実施した(非特許文献37)。CSF1R阻害のために、PLX3397(Selleck社製)を、DMSO:PEG300:ddH2Oが5:25:70の溶媒に懸濁し、次いで、週に2回、40mg/kg BWを強制経口投与した(非特許文献50)。 For the induction of non-alcoholic steatohepatitis (NASH), 1.5-month-old p16-Cre ERT2 -tdTomato mice were fed a diet containing 60 kcal% fat, 0.1% methionine, and a choline-free L-amino acid diet (CDA). -HFD: A06071302, manufactured by Research Diets) for a total of 10 weeks. For senolytic reagent treatment, all mice received placebo (vehicle control) or ABT263 by oral gavage. ABT263 (manufactured by Selleck) was suspended in ethanol: polyethylene glycol 400: Phosal 50 PG at a ratio of 10:30:60, and then 50 mg/kg BW was administered daily for 7 days per cycle. A total of 2 cycles were performed with a 2 week interval between cycles (Non-Patent Document 37). For CSF1R inhibition, PLX3397 (Selleck) was suspended in 5:25:70 DMSO:PEG300: ddH2O and then administered orally by gavage at 40 mg/kg BW twice a week ( Non-patent document 50).
<細胞培養>
細胞培養、細胞周期の同期、及び様々な薬物による処理は、既述(非特許文献5)のように実行された。初期継代hHCA2(非特許文献51)、初代マウス肺線維芽細胞 (MPF)、及びHEK293T細胞(RIKEN Cell Bankから入手)を、10 % ウシ胎児血清(FBS)(Sigma社製)及び1×ペニシリン/ストレプトマイシン/アムホテリシンB(ナカライテスク社製)を補充したDMEM(ナカライテスク社製)中で培養した。全ての細胞は、酸素正常状態で5% CO2下、37℃で培養されたが、MPFのみが低酸素条件(3~5% O2)で培養された(非特許文献52)。静止誘導の場合、細胞は、0.5% FBSを補充したDMEM中で3日間、細胞周期の停止が観察されるまで培養された。RO3306 と Nutlin3aによる老化誘導では、9μMのRO3306(Roche社製)で16~20時間処理することで、細胞をG2期に同期させ、その後9μMのRO3306と5μMのNutlin3a(Sigma-Aldrich社製) で8時間処理し、その後、5μMのnutlin3aを2日間投与した。 DNA損傷老化のために、細胞を100nMのドキソルビシン(Sigma社製)で24時間処理した。次いで、細胞を、誘導後12日まで100nMのBI-2536で処理して、増殖細胞を除去した。複製老化細胞は、ほぼ老化したHCA2細胞の培養 (41継代) によって作製された。ドキシサイクリン、エポキソミシン、ボルテゾミブは、それぞれ、1μg/mL、1μM、1μMの濃度で使用された。細胞数は、細胞を0.4% トリパンブルー色素で染色し、血球計で生細胞を計数することによって決定した。
<Cell culture>
Cell culture, cell cycle synchronization, and treatment with various drugs were performed as previously described (Non-Patent Document 5). Early passage hHCA2 (Non-Patent Document 51), primary mouse lung fibroblasts (MPF), and HEK293T cells (obtained from RIKEN Cell Bank) were incubated with 10% fetal bovine serum (FBS) (Sigma) and 1× penicillin. The cells were cultured in DMEM (manufactured by Nacalai Tesque) supplemented with /streptomycin/amphotericin B (manufactured by Nacalai Tesque). All cells were cultured at 37° C. under 5% CO 2 in normoxic conditions, but only MPFs were cultured in hypoxic conditions (3-5% O 2 ) (Non-Patent Document 52). For quiescence induction, cells were cultured in DMEM supplemented with 0.5% FBS for 3 days until cell cycle arrest was observed. For senescence induction with RO3306 and Nutlin3a, cells were synchronized to G2 phase by treatment with 9 μM RO3306 (Roche) for 16 to 20 hours, followed by treatment with 9 μM RO3306 and 5 μM Nutlin3a (Sigma-Aldrich). The cells were treated for 8 hours, and then 5 μM nutlin3a was administered for 2 days. For DNA damage aging, cells were treated with 100 nM doxorubicin (Sigma) for 24 hours. Cells were then treated with 100 nM BI-2536 until 12 days after induction to remove proliferating cells. Replicative senescent cells were generated by culturing nearly senescent HCA2 cells (41 passages). Doxycycline, epoxomicin, and bortezomib were used at concentrations of 1 μg/mL, 1 μM, and 1 μM, respectively. Cell number was determined by staining cells with 0.4% trypan blue dye and counting viable cells with a hemocytometer.
<プラスミド構築>
レンチウイルスベースのshRNAコンストラクト及びTet-on誘導性レンチウイルスコンストラクトは、既述(非特許文献53)のとおり生成された。 レンチウイルスコンストラクトを構築するには、ヒト PD-L1、P130、Flag-E2F97、NRF1Δ1-103のcDNAを含むPCRで生成したEcoRI/NotIフラグメントを、CSII-CMV-IRES2-Bsdベクター(Invitrogen社製)に挿入した。Tet-on誘導性レンチウイルスコンストラクトを構築するために、IκBα(S32/36A)及びHPV16E7のcDNAを含むBamHI/NotI PCRフラグメントを、BamHI/NotIで消化された3xFlagエピトープを含む pENTR-1Aベクター (Invitrogen社製)に挿入した。得られたプラスミドを CSIV-TRE-RfA-UbC-Puro ベクターと混合し、GatewayTM LR ClonaseTM Enzyme mix(Thermo Fisher Scientific社製)と反応させて、レンチウイルスプラスミドを生成した。
<Plasmid construction>
Lentivirus-based shRNA constructs and Tet-on inducible lentivirus constructs were generated as previously described (53). To construct lentivirus constructs, PCR-generated EcoRI/NotI fragments containing human PD-L1, P130, Flag-E2F97, and NRF1Δ1-103 cDNAs were inserted into the CSII-CMV-IRES2-Bsd vector (Invitrogen). To construct Tet-on inducible lentivirus constructs, BamHI/NotI PCR fragments containing IκBα (S32/36A) and HPV16E7 cDNAs were inserted into the BamHI/NotI digested pENTR-1A vector (Invitrogen) containing the 3xFlag epitope. The obtained plasmid was mixed with the CSIV-TRE-RfA-UbC-Puro vector and reacted with Gateway ™ LR Clonase ™ Enzyme mix (manufactured by Thermo Fisher Scientific) to generate a lentiviral plasmid.
<レンチウイルスの生産と感染>
レンチウイルスの生成及び細胞への感染は、既述(非特許文献53)に実行された。それぞれの遺伝子を発現するレンチウイルスは、pCMV-VSV-G-RSV-RevB、pCAG-HIVgp、及びそれぞれのCSII-CMV-IRES2-Bsd、又はCSIV-TRE-RfA-UbC-Puroプラスミドを、HEK293T細胞にリン酸カルシウム共沈法を使用したコトランスフェクションをすることによって生成された。示されたウイルスに感染したHCA2細胞又はMPFを、10μg/mLのブラストサイジン(Gibco社製) 又は2μg/mLのピューロマイシン(Gibco社製)で2~4日間処理した。CSIVプラスミドシステムの誘導発現のために、ドキシサイクリン(Sigma-Aldrich社製)を1μg/mLの濃度で培地に添加した。
<Lentivirus production and infection>
Lentivirus generation and cell infection were performed as previously described (53). Lentiviruses expressing the respective genes were generated by cotransfection of HEK293T cells with pCMV-VSV-G-RSV-RevB, pCAG-HIVgp, and the respective CSII-CMV-IRES2-Bsd, or CSIV-TRE-RfA-UbC-Puro plasmids using calcium phosphate coprecipitation. HCA2 cells or MPF infected with the indicated viruses were treated with 10 μg/mL blasticidin (Gibco) or 2 μg/mL puromycin (Gibco) for 2 to 4 days. For inducible expression of the CSIV plasmid system, doxycycline (Sigma-Aldrich) was added to the medium at a concentration of 1 μg/mL.
<バルクRNAシーケンス>
HCA2サンプルの場合、メーカーの指示に従ってISOGEN (Nippon Gene社製)を使用して全RNAを抽出し、Agilent Bioanalyzer (Agilent Technologies社製、Santa Clara, CA)を使用して品質を評価した。NEBNext Poly(A) mRNA Magnetic Isolation Module (New England Biolabs社製、Ipswich, MA) を使用して、PolyAテール付きmRNAを200μgの全RNAからビーズで選択した。RNA-seqライブラリは、NEBNext Ultra II Directional RNA Library Prep Kit (New England Biolabs社製、Ipswich, MA)を製造業者の指示に従って使用して調製した。100ヌクレオチド長のペアエンドリードの配列決定は、HiSeq2500 (Illumina社製、San Diego, CA) で実行された。
<Bulk RNA sequencing>
For HCA2 samples, total RNA was extracted using ISOGEN (Nippon Gene) according to the manufacturer's instructions, and quality was assessed using an Agilent Bioanalyzer (Agilent Technologies, Santa Clara, Calif.). PolyA-tailed mRNA was bead-selected from 200 μg of total RNA using the NEBNext Poly(A) mRNA Magnetic Isolation Module (New England Biolabs, Ipswich, Mass.). RNA-seq libraries were prepared using the NEBNext Ultra II Directional RNA Library Prep Kit (New England Biolabs, Ipswich, Mass.) according to the manufacturer's instructions. Sequencing of 100 nucleotide long paired-end reads was performed on a HiSeq2500 (Illumina, San Diego, CA).
東京大学ヒトゲノム解析センターに構築された社内パイプラインである Genomon (ver. 2.5.2) は、差次的に発現する遺伝子の同定に使用された(https://github.com/Genomon-Project)。簡単に言うと、STAR aligner (v2.5.2a)(非特許文献54)を使用して、配列決定データをヒトゲノム(hg19)にアラインメントした。HTSeq(v0.11.2)(非特許文献55)を使用して、リードアラインメントから生の遺伝子数を取得した。DESeq2(v1.22.2)(非特許文献56)を使用して、差次的発現解析を実行した。B-H法で調整された P値<0.05の遺伝子は、有意に差次的に発現していると見なされる。転写調節ネットワークの構築のために、FPKMがPD-L1と正の相関を示す(R2>0.95)230個の遺伝子を Cytoscape platform (v3.8.2)上のiRegulon(v1.3)(非特許文献15)に入力した。TSSを中心とした20kbを各遺伝子のプロモーター領域とみなし、正規化濃縮スコア(NES)を閾値として3をカットオフとした。 Genomon (ver. 2.5.2), an in-house pipeline built at the Human Genome Center, University of Tokyo, was used to identify differentially expressed genes (https://github.com/Genomon-Project). Briefly, sequencing data were aligned to the human genome (hg19) using STAR aligner (v2.5.2a) (54). Raw gene counts were obtained from read alignments using HTSeq (v0.11.2) (55). Differential expression analysis was performed using DESeq2 (v1.22.2) (56). Genes with a B-H adjusted P value < 0.05 were considered significantly differentially expressed. To construct the transcriptional regulatory network, 230 genes whose FPKM showed a positive correlation with PD-L1 (R 2 >0.95) were entered into iRegulon (v1.3) (Non-Patent Document 15) on the Cytoscape platform (v3.8.2). The 20 kb region centered on the TSS was considered as the promoter region of each gene, and the normalized enrichment score (NES) was set as the threshold with a cutoff of 3.
静止及びd-Sen MPFサンプルの場合、メーカーの指示に従って RNeasy Mini Kit (QIAGEN社製)を使用してRNAを抽出した。NEBNext rRNA Depletion Kit (New England Biolabs社製)を使用して、全RNA(1μg)からのrRNAの除去を実行した。 RNA-seqライブラリは、NEBNext Ultra II Directional RNA Library Prep Kit (New England Biolabs社製)を使用して調製した。線状dsDNAライブラリを一本鎖環状DNAライブラリに変換した後 (Universal Library Conversion Kit, MGI Tech, Guangdong, China)、プールされたライブラリを、ペアエンドモード(2×100bp)を使用してDNBSEQ-G400RS(MGI Tech社製)で配列決定した。 For static and d-Sen MPF samples, RNA was extracted using the RNeasy Mini Kit (QIAGEN) according to the manufacturer's instructions. rRNA was removed from total RNA (1 μg) using NEBNext rRNA Depletion Kit (manufactured by New England Biolabs). The RNA-seq library was prepared using NEBNext Ultra II Directional RNA Library Prep Kit (New England Biolabs). After converting the linear dsDNA library into a single-stranded circular DNA library (Universal Library Conversion Kit, MGI Tech, Guangdong, China), the pooled library was converted to DNBSEQ-G400RS ( Sequence was determined using MGI Tech).
配列決定データは、Rsubread (v2.4.3)(非特許文献57)を使用してマウスリファレンスゲノム(mm10)にアライメントされた。生のカウントは、遺伝子カウントの場合は refGene、TEの場合はRepeatMasker アノテーションを介して、リードアライメントから取得され、さらにedgeR (v3 .32.1)(非特許文献58)によってCPMに転送された。mm10 refGeneとRepeatMaskerアノテーションデータベースはどちらも、カリフォルニア大学サンタ クルーズ校(UCSC)のゲノムブラウザーからのものである。CPM 0.1未満の低発現遺伝子をフィルタリングした後、示差的発現は、limma (v3.46.0)(非特許文献59) を使用した線形モデルで分析された。B-H法で調整されたlog2FC > 0.6及びFDR<0.05の遺伝子は、有意な示差的発現遺伝子(DEG)とみなされ、さらに、clusterProfiler (v3.18.1)(非特許文献60)を使用した遺伝子オントロジー分析に登録された。遺伝子セット濃縮分析(GSEA)の場合、log2倍率変化による順序でランク付けされた遺伝子リストが、clusterProfilerの事前ランク付けGSEA関数に入力され、マウスのホールマーク遺伝子セットがmsigdb (v7.4.1) から取得された。全ての図は、enrichplot (v1.10.2) によってプロットされた。TEの分類では、log2FC>1及びFDR<0.05の大幅に変化したTEに Dfam (リリース 3.5) (非特許文献61)によってさらにアノテーションが付けられた。 Sequencing data were aligned to the mouse reference genome (mm10) using Rsubread (v2.4.3) (Non-Patent Document 57). Raw counts were obtained from read alignments via refGene for gene counts and RepeatMasker annotations for TE, and further transferred to CPM by edgeR (v3.32.1) (58). Both the mm10 refGene and RepeatMasker annotation databases are from the University of California, Santa Cruz (UCSC) Genome Browser. After filtering low expressed genes with CPM <0.1, differential expression was analyzed with a linear model using limma (v3.46.0) (Non-Patent Document 59). Genes with log2FC > 0.6 and FDR < 0.05 adjusted by the BH method are considered as significant differentially expressed genes (DEGs), and are further analyzed using clusterProfiler (v3.18.1) (Non-Patent Document 60) was registered for gene ontology analysis using . For gene set enrichment analysis (GSEA), a list of genes ranked in order by log2 fold change is input into clusterProfiler's pre-ranked GSEA function, and mouse hallmark gene sets are obtained from msigdb (v7.4.1) It was done. All figures were plotted with enrichplot (v1.10.2). In the classification of TEs, significantly changed TEs with log2FC > 1 and FDR < 0.05 were further annotated by Dfam (Release 3.5) (Non-Patent Document 61).
<シングルセルRNAシーケンスデータ解析>
p16-TomマウスのTom+/PD-L1+及びTom+/PD-L1- 細胞の解析には、GEOから入手可能な処理済み液滴ベースのシングルセルRNAシーケンスデータセット10(GSE155182)を使用した。Python (ver. 3.6.8) 上のScanpy package (ver. 1.4.5.post1)を使用して、シングルセルRNAseqデータ(非特許文献62)を処理した。PD-L1+ 細胞をゲーティングするための閾値は、0より大きいCd274 UMIカウントとして定義された。差次的発現遺伝子(DEG)解析では、dffxpy package (ver. 0.7.3)を使用して、FDRが0.05未満、log2倍変化が0.15を超える遺伝子を特定した。遺伝子セット濃縮分析(GSEA)の場合、log2倍数変化による順序でランク付けされた遺伝子リストが、clusterProfiler の事前ランク付けGSEA関数に入力され、マウスの特徴的な遺伝子セットが msigdb (v7.4.1) から取得された。
<Single cell RNA sequence data analysis>
For analysis of Tom+/PD-L1+ and Tom+/PD-L1- cells of p16-Tom mice, processed droplet-based single-cell RNA-seq dataset 10 (GSE155182) available from GEO was used. Single-cell RNAseq data (Non-Patent Document 62) was processed using the Scanpy package (ver. 1.4.5.post1) on Python (ver. 3.6.8). The threshold for gating PD-L1+ cells was defined as a Cd274 UMI count greater than 0. For differentially expressed gene (DEG) analysis, genes with FDR less than 0.05 and log2 fold change greater than 0.15 were identified using the dffxpy package (ver. 0.7.3). For gene set enrichment analysis (GSEA), a list of genes ranked in order by log2 fold change was input into the pre-ranked GSEA function of clusterProfiler, and the mouse characteristic gene set was extracted from msigdb (v7.4.1). Obtained.
<RNAの分離とリアルタイムPCR>
培養細胞からの全RNAは、ISOGEN(Nippon Gene社製)を使用して製造業者の指示に従って抽出された。選別された初代細胞からのRNAについては、製造業者の指示に従って Single Cell RNA Purification Kit (Norgen社製)を利用した。qPCR分析では、ReverTra Ace qPCR RT Master Mix (Toyobo社製)を使用してcDNAを合成した。リアルタイムPCR増幅は、THUNDERBIRD SYBR qPCR Master Mix (Toyobo社製) 及び StepOnePlus (Applied Biosystems社製)を使用して、96ウェル光学反応プレートで実行された。各遺伝子の相対発現量は、各サンプルのβ-アクチン発現量に対する正規化によって決定された。全てのプライマーは、表1にリストされている。
<RNA isolation and real-time PCR>
Total RNA from cultured cells was extracted using ISOGEN (Nippon Gene) according to the manufacturer's instructions. For RNA from the selected primary cells, Single Cell RNA Purification Kit (manufactured by Norgen) was used according to the manufacturer's instructions. For qPCR analysis, cDNA was synthesized using ReverTra Ace qPCR RT Master Mix (manufactured by Toyobo). Real-time PCR amplification was performed in a 96-well optical reaction plate using THUNDERBIRD SYBR qPCR Master Mix (Toyobo) and StepOnePlus (Applied Biosystems). The relative expression level of each gene was determined by normalization to the β-actin expression level of each sample. All primers are listed in Table 1.
<タンパク質抽出とイムノブロッティング>
全細胞溶解物の場合、細胞を Laemmli緩衝液(2% SDS、10% グリセロール、5% 2-メルカプトエタノール、0.002% ブロモフェノールブルー、及び62.5mM Tris HCl、pH 6.8)で直接溶解した。全セルライセートをSDS-PAGEで分離し、PVDF膜(Immobileon-P; Millipore社製)に転写し、ECL検出システムを使用して指定の抗体によるイムノブロッティングを行った。β-アクチン抗体 (C-4、Santa Cruz社製)をローディングコントロールとして使用し、Amersham Imager 680 (Cytiva社製) を使用して画像をキャプチャした。全ての抗体は、表2及び3にリストされている。
Protein extraction and immunoblotting
For whole cell lysates, cells were directly lysed in Laemmli buffer (2% SDS, 10% glycerol, 5% 2-mercaptoethanol, 0.002% bromophenol blue, and 62.5 mM Tris HCl, pH 6.8). Whole cell lysates were separated by SDS-PAGE, transferred to PVDF membranes (Immobileon-P; Millipore), and immunoblotted with the indicated antibodies using the ECL detection system. β-actin antibody (C-4, Santa Cruz) was used as a loading control, and images were captured using an Amersham Imager 680 (Cytiva). All antibodies are listed in Tables 2 and 3.
<組織学>
免疫蛍光染色のために、OCT化合物中に保存された腎臓、肺、肝臓、及び結腸を凍結切片にし、DAPIで染色した。INCell Analyzer 2500HS (Cytiva社製)を使用して蛍光シグナルを画像化した後、この機器により、IN Cell Developer Toolbox v1.9.2 を通じてDAPIシグナルを核として認識することで、Tom+細胞を自動的にカウントしてパーセンテージを計算した。Tom+シグナルはバックグラウンドの10倍であった。Tom+細胞中のPD-L1+細胞の割合を決定するために、抗PD-L1(Abcam社製、クローン: EPR20529) 及び抗CD3(Abcam社製、クローン: KT3) 染色を実施した。PD-L1の強度は、各Tom+細胞について定量化されたが、CD3+細胞及び強い自己蛍光を示す細胞(NASHサンプルの油滴など) は除外された。PD-L1+細胞は、中央値よりも1.8倍 (腎臓の場合) 又は2.5倍 (肺及び肝臓の場合) 大きいPD-L1強度によって認識された。各臓器切片の10~30のフィールドをランダムに選択し、焦点の合っていない画像をさらに除外した。図1bの代表的な画像は、Carl Zeiss LSM 710 NLO (Zeiss社製)で観察された。全ての抗体は、表2及び3にリストされている。
<Histology>
For immunofluorescence staining, kidneys, lungs, liver, and colon preserved in OCT compound were frozen sectioned and stained with DAPI. After imaging the fluorescent signals using INCell Analyzer 2500HS (manufactured by Cytiva), this instrument automatically counted Tom+ cells by recognizing DAPI signals as nuclei through IN Cell Developer Toolbox v1.9.2. The percentage was calculated. Tom+ signal was 10 times background. To determine the proportion of PD-L1+ cells among Tom+ cells, anti-PD-L1 (Abcam, clone: EPR20529) and anti-CD3 (Abcam, clone: KT3) staining was performed. PD-L1 intensity was quantified for each Tom+ cell, excluding CD3+ cells and cells exhibiting strong autofluorescence (such as oil droplets in NASH samples). PD-L1+ cells were recognized by PD-L1 intensity that was 1.8 times (for kidney) or 2.5 times (for lung and liver) greater than the median. 10-30 fields of each organ section were randomly selected and out-of-focus images were further excluded. The representative image in Figure 1b was observed on a Carl Zeiss LSM 710 NLO (Zeiss). All antibodies are listed in Tables 2 and 3.
組織化学的染色では、肝臓と肺をパラフィンで固定し、切片にし、H&E又はシリウスレッドで染色した。染色された組織切片を、明視野照明下でOlympus BX51顕微鏡(Olympus社製)を用いて観察した。ImageJ ソフトウェアを介して、肺胞サイズ、脂肪滴面積、及びシリウスレッド面積を定量化するために、Huang閾値を使用して面積を定義し、各セクションの3つのフィールドでそれぞれ1500、2000、及び40ピクセルを超える平均面積を測定した。 For histochemical staining, the liver and lungs were fixed with paraffin, sectioned, and stained with H&E or Sirius Red. The stained tissue sections were observed using an Olympus BX51 microscope (manufactured by Olympus) under bright field illumination. To quantify alveolar size, lipid droplet area, and Sirius Red area via ImageJ software, define the area using the Huang threshold, 1500, 2000, and 400, respectively, in three fields for each section. The average area over the pixels was measured.
<In vitro T細胞殺傷アッセイ>
C57BL/6又はOT-1マウスから脾臓を採取し、3mLのPBS中で、70μmセルストレーナー上でシリンジの平らな端で粉砕した。通過した全ての細胞をチューブに収集し、400gで5分間遠心分離してペレット化した。細胞ペレットを2%FBS及び1mM EDTAを含有する1.5mLのPBSに再懸濁し、次いで細胞濃度を血球計により測定した。 CD8+T細胞は、EasySepTM Mouse CD8+ T Cell Isolation Kit (STEMCELL Technologies社製) を製造業者の指示に従って使用して単離した。その後、6 ×105個のCD8+T細胞を、5μg/mL 抗CD3α(BioLegend社製、クローン: 145-2C11) 及び5μg/mL 抗CD28(BioLegend社製、クローン: 37.51)でプレコートした24ウェルプレートで、72時間培養した。初代マウスCD8+T細胞を、10% ウシ胎児血清(FBS)(Sigma社製)、1×ペニシリン/ストレプトマイシン/アムホテリシンB(nacalai tesque社製)、2mM L-グルタミン(Thermo Fisher Scientific社製)、50μM 2-メルカプトエタノール、1×インスリン-トランスフェリン-セレン(Thermo Fisher Scientific社製)、及び30U/mL 組換えマウスIL-2 (PeproTech社製)を補充したRPMI(nacalai tesque社製)で培養した(非特許文献63)。活性化されたCD8+T細胞を収集し、標的細胞と5:1の比率で18時間共培養した(非特許文献64)。OT-1マウスから単離したT細胞との共培養中に、抗原特異的応答を活性化するために、オボアルブミンペプチド(OVA 257-264、SynPeptide社製)を1μg/mL供給した。共培養後、抗CD45(BioLegend社製、クローン: 30-F11) 及びヨウ化プロピジウム(50μg/mL)染色を行って、エフェクター細胞を除外し、細胞毒性をそれぞれ評価した。細胞は、BD FACSCantoTM II (BD Biosciences社製)によって分析された。全ての抗体は、表2及び3にリストされている。
<In vitro T cell killing assay>
Spleens were harvested from C57BL/6 or OT-1 mice and triturated with the flat end of a syringe over a 70 μm cell strainer in 3 mL of PBS. All cells that passed were collected in a tube and pelleted by centrifugation at 400 g for 5 minutes. The cell pellet was resuspended in 1.5 mL of PBS containing 2% FBS and 1 mM EDTA, and the cell concentration was then determined by a hemocytometer. CD8+ T cells were isolated using the EasySep ™ Mouse CD8+ T Cell Isolation Kit (STEMCELL Technologies) according to the manufacturer's instructions. Then, 6 × 10 CD8+ T cells were plated in a 24-well plate precoated with 5 μg/mL anti-CD3α (BioLegend, clone: 145-2C11) and 5 μg/mL anti-CD28 (BioLegend, clone: 37.51). , and cultured for 72 hours. Primary mouse CD8+ T cells were incubated with 10% fetal bovine serum (FBS) (Sigma), 1x penicillin/streptomycin/amphotericin B (Nacalai Tesque), 2mM L-glutamine (Thermo Fisher Scientific), and 50μM 2- Cultured in RPMI (manufactured by Nacalai Tesque) supplemented with mercaptoethanol, 1× insulin-transferrin-selenium (manufactured by Thermo Fisher Scientific), and 30 U/mL recombinant mouse IL-2 (manufactured by PeproTech) (non-patent literature). 63). Activated CD8+ T cells were collected and co-cultured with target cells at a ratio of 5:1 for 18 hours (Non-Patent Document 64). During co-culture with T cells isolated from OT-1 mice, 1 μg/mL of ovalbumin peptide (OVA 257-264, SynPeptide) was supplied to activate antigen-specific responses. After co-culture, anti-CD45 (manufactured by BioLegend, clone: 30-F11) and propidium iodide (50 μg/mL) staining were performed to exclude effector cells, and cytotoxicity was evaluated. Cells were analyzed by BD FACSCanto ™ II (BD Biosciences). All antibodies are listed in Tables 2 and 3.
<フローサイトメトリーとソーティング>
in vivo 老化細胞から肺、肝臓、腎臓の PD-L1集団を測定するために、若齢 (生後 2か月)及び老齢(24~26か月) のp16-Tomマウスを、TAM投与の2週間後に屠殺した。アバーチン(Sigma社製)による麻酔後、マウスに心臓から血管系を介してPBSを灌流した。肺と腎臓を鋭利な刃で細断し、リベラーゼ溶液(RPMI(nacalai tesque社製)中、肺の場合は50μg/mL リベラーゼTM(Sigma社製)、腎臓の場合はリベラーゼTH(Sigma社製)、2Kunitz単位/mL DNase I(Sigma社製)、及び10mM HEPES(nacalai tesque社製))で37℃、1時間消化した。消化中、組織懸濁液を18G及び21Gの針に順次通して、残留組織をさらに均質化した。肝臓を鋭利な刃で細断し、コラゲナーゼ溶液(HBSS(+)(和光社製)中、1mg/mL コラゲナーゼIV (Thermo Fisher Scientific社製)、1mg/mL プロナーゼ(Sigma社製)、及び1Kunitz単位/mL DNase I(Sigma社製))で、37℃、20分間振とうして消化した。消化後、全てのサンプルを70μmのセルストレーナーに通し、400gで5分間遠心分離してペレット化した。RBC溶解(Thermo Fisher社製)及び FcRブロッキング(Miltenyi Biotec社製) の後、細胞を抗PD-L1(Proteintech社製、カタログ: 17952-1-AP)で、4℃ で1時間染色し、次いで、AF647(Thermo Fisher Scientific社製)コンジュゲート抗ウサギIgGと、4℃で30分間インキュベートした。免疫細胞と死細胞は、それぞれ抗CD45染色(BioLegend社製、クローン: 30-F11)とDAPI(0.1μg/mL)染色によって除外された。蛍光シグナルは、BD FACSCantoTM II (BD Biosciences社製)を使用して分析された。
<Flow cytometry and sorting>
To measure lung, liver, and kidney PD-L1 populations from in vivo senescent cells, young (2 months old) and old (24-26 months old) p16-Tom mice were treated with TAM for 2 weeks. It was later slaughtered. After anesthesia with Avertin (Sigma), the mice were perfused with PBS via the vascular system from the heart. The lungs and kidneys were shredded with a sharp blade and placed in Liberase solution (RPMI (manufactured by Nacalai Tesque) at 50 μg/mL Liberase TM (manufactured by Sigma) for lungs and Liberase TH (manufactured by Sigma) for kidneys. , 2 Kunitz units/mL DNase I (manufactured by Sigma), and 10 mM HEPES (manufactured by Nacalai Tesque) at 37°C for 1 hour. During digestion, the tissue suspension was passed sequentially through 18G and 21G needles to further homogenize the remaining tissue. The liver was shredded with a sharp blade and mixed with 1 mg/mL collagenase IV (Thermo Fisher Scientific), 1 mg/mL pronase (Sigma), and 1 Kunitz unit in collagenase solution (HBSS (+) (Wako)). /mL DNase I (manufactured by Sigma)) at 37°C for 20 minutes with shaking. After digestion, all samples were passed through a 70 μm cell strainer and pelleted by centrifugation at 400 g for 5 min. After RBC lysis (Thermo Fisher) and FcR blocking (Miltenyi Biotec), cells were stained with anti-PD-L1 (Proteintech, catalog: 17952-1-AP) for 1 hour at 4°C and then incubated. , AF647 (manufactured by Thermo Fisher Scientific) conjugated anti-rabbit IgG for 30 minutes at 4°C. Immune cells and dead cells were excluded by anti-CD45 staining (manufactured by BioLegend, clone: 30-F11) and DAPI (0.1 μg/mL) staining, respectively. Fluorescent signals were analyzed using a BD FACSCanto ™ II (BD Biosciences).
培養細胞内のPD-L1集団を決定するために、細胞をトリプシン処理によって剥離した。細胞を洗浄緩衝液(PBS中、4% FBS)で再懸濁した後、細胞をHCA2用の抗PD-L1(Abcam社製、クローン:28-8)又はMPS用の抗PD-L1(Proteintech社製、カタログ:17952-1-AP)とともに、4℃で1時間インキュベートし、その後AF647(Thermo Fisher Scientific社製)とコンジュゲートした抗ウサギIgGとともに4℃で30分間インキュベートした。 他の免疫チェックポイントについては、細胞を、抗CD80(BioLegend社製、クローン: W17149D)、抗CD86(BioLegend社製、クローン: BU63)、抗CD276(BioLegend社製、クローン: MIH42)、抗B7-H4 (BioLegend社製、 クローン:MIH43)、抗CD270(BioLegend社製、クローン:122)、抗ガレクチン-9(BioLegend社製、クローン:9M1-3)とインキュベートした。死細胞を除外するためにDAPI(0.1μg/mL)染色を実行し、BD FACSCantoTM II (BD Biosciences社製) を使用して蛍光シグナルを分析した。 To determine the PD-L1 population in cultured cells, cells were detached by trypsinization and resuspended in wash buffer (4% FBS in PBS), after which the cells were incubated with anti-PD-L1 for HCA2 (Abcam, clone: 28-8) or anti-PD-L1 for MPS (Proteintech, catalog: 17952-1-AP) at 4°C for 1 h, followed by incubation with anti-rabbit IgG conjugated to AF647 (Thermo Fisher Scientific) for 30 min at 4°C. For other immune checkpoints, cells were incubated with anti-CD80 (BioLegend, clone: W17149D), anti-CD86 (BioLegend, clone: BU63), anti-CD276 (BioLegend, clone: MIH42), anti-B7-H4 (BioLegend, clone: MIH43), anti-CD270 (BioLegend, clone: 122), and anti-galectin-9 (BioLegend, clone: 9M1-3). DAPI (0.1 μg/mL) staining was performed to exclude dead cells, and the fluorescent signals were analyzed using a BD FACSCanto ™ II (BD Biosciences).
肝臓浸潤又は脾臓の CD8+T細胞分析では、肝臓又は脾臓をマウスから採取し、3mLのPBS中で、70μmセルストレーナー上でシリンジの平らな端で粉砕した。通過した全ての細胞をチューブに収集し、400gで5分間遠心分離してペレット化した。RBC溶解(Thermo Fisher社製) 及びFcRブロッキング (Miltenyi Biotec社製) の後、細胞を抗CD45(BioLegend社製、クローン: 30-F11)、抗CD44 (BioLegend社製、クローン: IM7)、抗CD3(BioLegend社製、クローン:145-2C11)、及び抗CD8(BioLegend社製、クローン:53-6.7)で染色した。死細胞は、ヨウ化プロピジウム染色(2 μg/mL)により除去した。蛍光シグナルは、BD FACSCantoTM II (BD Biosciences社製) を使用して分析された。 For liver infiltration or splenic CD8+ T cell analysis, liver or spleen was harvested from mice and triturated with the flat end of a syringe over a 70 μm cell strainer in 3 mL of PBS. All cells that passed were collected in a tube and pelleted by centrifugation at 400 g for 5 minutes. After RBC lysis (manufactured by Thermo Fisher) and FcR blocking (manufactured by Miltenyi Biotec), cells were treated with anti-CD45 (manufactured by BioLegend, clone: 30-F11), anti-CD44 (manufactured by BioLegend, clone: IM7), and anti-CD3. (manufactured by BioLegend, clone: 145-2C11) and anti-CD8 (manufactured by BioLegend, clone: 53-6.7). Dead cells were removed by propidium iodide staining (2 μg/mL). Fluorescent signals were analyzed using a BD FACSCanto ™ II (BD Biosciences).
ProteoStat染色では、細胞を抗PD-L1(Abcam社製、クローン: 28-8)及びAF405コンジュゲート抗ウサギIgG (Abcam社製) で標識した後、細胞懸濁液を4% ホルムアルデヒドを含むPBSに滴下し、室温で10分間インキュベートした。細胞を洗浄緩衝液で1回洗浄し、細胞懸濁液を透過化溶液(PBS中、0.2% Triton X-100、4% FBS)に滴下し、室温で5分間インキュベートした。当該細胞を洗浄緩衝液で洗浄した後、ProteoStat染色液(1/5000倍希釈)で、室温で30分間染色した。AF405及びProteoStat(PE-Texas Redチャネル) シグナルは、BD FACSAria SORP (BD Biosciences社製)を使用して分析された。全ての抗体は、表2及び3にリストされている。 For ProteoStat staining, cells were labeled with anti-PD-L1 (Abcam, clone: 28-8) and AF405-conjugated anti-rabbit IgG (Abcam), and then the cell suspension was added to PBS containing 4% formaldehyde. It was added dropwise and incubated at room temperature for 10 minutes. Cells were washed once with wash buffer, and the cell suspension was added dropwise to permeabilization solution (0.2% Triton X-100, 4% FBS in PBS) and incubated for 5 minutes at room temperature. After washing the cells with a washing buffer, they were stained with ProteoStat staining solution (1/5000-fold dilution) at room temperature for 30 minutes. AF405 and ProteoStat (PE-Texas Red channel) signals were analyzed using BD FACSAria SORP (manufactured by BD Biosciences). All antibodies are listed in Tables 2 and 3.
<プロテアソーム活性アッセイ>
PD-L1+細胞とPD-L1-細胞を選別した後、10,000個の細胞を白色の96ウェルプレートに播種した。キモトリプシン、トリプシン、及びカスパーゼ様プロテアソーム活性は、Proteasome-Glo Cell-Based Assay kit (Promega社製) を使用して、製造業者の説明書に従ってテストされた。発光強度は、FLUOstar OPTIMA (BMG LABTECH社製)を用いて測定した。
<Proteasome activity assay>
After sorting PD-L1+ cells and PD-L1- cells, 10,000 cells were seeded into a white 96-well plate. Chymotrypsin, trypsin, and caspase-like proteasome activities were tested using the Proteasome-Glo Cell-Based Assay kit (Promega) according to the manufacturer's instructions. The luminescence intensity was measured using FLUOstar OPTIMA (manufactured by BMG LABTECH).
<血清中のAST、ALT、LDH、総コレステロール(T-CHO)レベルの測定>
心臓穿刺によりマウスから血液を採取し、室温で15分間放置した。血清を得るために、サンプルを 4℃、3400gで15分間遠心分離した。重度の溶血を伴う血清サンプルは、AST、ALT、及び LDHのさらなる分析から除外される(通常の食餌とABT263処理を受けたマウスからの1つのサンプル)。AST、ALT、LDH、及びT-CHOの血清レベルは、富士フイルム和光純薬株式会社 (日本) によって測定された。
<Measurement of serum AST, ALT, LDH, and total cholesterol (T-CHO) levels>
Blood was collected from mice by cardiac puncture and left at room temperature for 15 min. To obtain serum, samples were centrifuged at 3400 g for 15 min at 4°C. Serum samples with severe hemolysis were excluded from further analysis of AST, ALT, and LDH (one sample from a mouse receiving normal diet and ABT263 treatment). Serum levels of AST, ALT, LDH, and T-CHO were measured by Fujifilm Wako Pure Chemical Corporation (Japan).
<握力測定とパフォーマンステスト>
握力は、BIO-GS3握力試験装置(BIOSEB社製)により測定した。マウスの尻尾を掴んで、2本の前足で金属グリッドを掴ませた。グリッドが解放されるまで水平面内で後方に引っ張ると、デバイスに表示される力の強さが記録される。
<Grip strength measurement and performance test>
Grip strength was measured using a BIO-GS3 grip strength testing device (manufactured by BIOSEB). I grabbed the mouse by its tail and had it grip a metal grid with its two front paws. As the grid is pulled backwards in a horizontal plane until it is released, the strength of the force displayed on the device is recorded.
コーディネーションの測定には、ロータロッドモデル MK-630B (室町社製) を使用した。 回転速度を30rpmに設定し、回転を開始してからマウスが磁気スイッチ上に落ちるまでのロッド上の滞在時間を記録した。学習効果を防ぐために、握力とロータロッドの両方のテストを同じ日に3回実施した。各マウスの平均値を統計的推論に使用した。 To measure coordination, a rotarod model MK-630B (Muromachi Co., Ltd.) was used. The rotation speed was set to 30 rpm, and the time the mouse stayed on the rod from the start of rotation until it fell onto the magnetic switch was recorded. To prevent learning effects, both grip strength and rotarod tests were performed three times on the same day. The average value for each mouse was used for statistical inference.
<統計と再現性>
動物実験では、同じ遺伝子型を持つ全ての雄マウスが各グループにランダムに割り当てられ、各実験計画で、同じ年齢依存のスケジュールに従って独立して行った。サンプルサイズは、パイロット研究によって事前に決定されたものではない。画像解析装置を使用して同じ基準で自動解析が行われたため、この研究ではブラインド設計は採用されなかった。特に断りのない限り、データは平均値±SEMとして表示される。2つのグループ間の比較は、アンペア両側スチューデントt検定によって行われた。1変数データの多重比較は、一元配置分散分析(ANOVA)とその後の事後Tukey’s検定又はダネット検定によって実行された。多変数データの多重比較は、二元配置分散分析とそれに続くSidakの多重比較検定によって実行された。比較の一貫性を保つために、全ての図の有意性は次のように示される: *P<0.05、**P<0.01、***P<0.001、****P<0.0001。 全ての代表的な結果について、3回又は複数回の独立した実験が実行され、同様の結果が得られた。
<Statistics and reproducibility>
In animal experiments, all male mice with the same genotype were randomly assigned to each group and performed independently according to the same age-dependent schedule in each experimental design. The sample size was not predetermined by a pilot study. A blind design was not employed in this study, as automated analysis was performed using an image analyzer and with the same criteria. Data are presented as mean ± SEM unless otherwise noted. Comparisons between two groups were made by an unpaired two-tailed Student's t test. Multiple comparisons of univariate data were performed by one-way analysis of variance (ANOVA) followed by post hoc Tukey's or Dunnett's test. Multiple comparisons of multivariate data were performed by two-way analysis of variance followed by Sidak's multiple comparison test. To maintain consistency in comparisons, significance in all figures is indicated as follows: *P<0.05, **P<0.01, ***P<0.001, *** *P<0.0001. For all representative results, three or more independent experiments were performed with similar results.
[実施例1]
生体内における老化細胞の性質は、その起源や刺激によって異なり、加齢とともに特定の種類の老化細胞が蓄積する場合がある。癌細胞などの不適格な細胞に対する免疫監視は、免疫チェックポイントによって負に制御される。したがって、これは老化細胞にも当てはまるのではないかと考えられた。この疑問に対処するために、本発明者らは、老化線維芽細胞の十分に確立された in vitro モデルであるヒト線維芽細胞株HCA2におけるいくつかの免疫チェックポイント(非特許文献4)の発現を分析した。興味深いことに、飢餓誘発静止(Quie)と比較して、ナットリン3a誘発細胞(n-Sen)(非特許文献5)及びDNA損傷誘発老化細胞(d-Sen)では、プログラムされたデスリガンド1(PD-L1)集団の発現が見られ有意かつ特異的に上昇した唯一のものでした (図1a、図5a)。
[Example 1]
The properties of senescent cells in vivo vary depending on their origin and stimulus, and certain types of senescent cells may accumulate with aging. Immune surveillance against incompetent cells, such as cancer cells, is negatively regulated by immune checkpoints. Therefore, it was thought that this might also apply to senescent cells. To address this question, we investigated the expression of several immune checkpoints in the human fibroblast cell line HCA2, a well-established in vitro model of aging fibroblasts. was analyzed. Interestingly, compared to starvation-induced quiescence (Quie), nutlin-3a-induced cells (n-Sen) (Non-Patent Document 5) and DNA damage-induced senescent cells (d-Sen) show that programmed death ligand 1 ( PD-L1) population was the only one whose expression was significantly and specifically elevated (Figure 1a, Figure 5a).
PD-L1の受容体であるプログラム細胞死タンパク質1(PD-1)の発現は、マウスT細胞において、年齢とともに増加する(非特許文献6~9)。PD-L1は、PD-1発現T細胞の攻撃によって引き起こされる細胞毒性を防ぐため、老化細胞におけるPD-L1の発現が、免疫監視を逃れて加齢に伴う老化細胞の蓄積につながる可能性があると仮説を立てた。in vivo で老化細胞におけるPD-L1の発現を調べるために、p16-CreERT2-tdTomatoマウスモデル(p16-Tom)を使用し、これらの細胞(p16high細胞)をtdTomato発現細胞(Tom+細胞) として蛍光イメージングによって可視化した(非特許文献10)。肺、肝臓、腎臓では、Tom+非免疫細胞は一般にTom-細胞と比較して、老化関連分泌表現型(SASP)関連遺伝子の発現上昇を示し、Tom+細胞が老化細胞の少なくとも一部であることを示唆している(図5b)。PD-L1を発現するいくつかのTom+細胞が検出され(図1b)、Tom+細胞及びTom+細胞内のPD-L1発現細胞の集団は、若齢のグループ(生後2か月)よりも老齢のグループ(24~26か月)の方が有意に高かった。(図1c、図5c)。高齢マウスの Tom- 細胞におけるPD-L1細胞集団の増加は肝臓と腎臓でも観察されたが、肺では観察されなかった(図5d)。老化細胞の in vivo 動態をさらに特徴付けるために、生後2.5か月でこれらの細胞をタモキシフェン(TAM)でパルス標識し、その後、生後3、6、及び9か月でマウスを屠殺した(図1d)。肺、肝臓、腎臓のTom+細胞に占めるPD-L1+細胞の割合は、生後9か月群の方が生後 3か月群及び生後6か月群よりも高く、加齢に伴いPD-L1+細胞が蓄積する傾向が示唆された。 Expression of programmed cell death protein 1 (PD-1), a receptor for PD-L1, increases with age in mouse T cells (Non-Patent Documents 6 to 9). PD-L1 prevents cytotoxicity caused by attack of PD-1-expressing T cells, and therefore, PD-L1 expression in senescent cells may escape immune surveillance and lead to age-related accumulation of senescent cells. I hypothesized that there is. To examine the expression of PD-L1 in senescent cells in vivo, we used the p16-CreERT2-tdTomato mouse model (p16-Tom) and treated these cells (p16 high cells) as tdTomato-expressing cells (Tom + cells). It was visualized by fluorescence imaging (Non-Patent Document 10). In the lung, liver, and kidney, Tom + non-immune cells generally show increased expression of senescence-associated secretory phenotype (SASP)-related genes compared to Tom- cells, and Tom + cells are at least a subset of senescent cells . This suggests that (Figure 5b). Some Tom + cells expressing PD-L1 were detected (Fig. 1b), and the population of Tom + cells and PD-L1-expressing cells within Tom + cells was smaller than in the younger group (2 months old). It was significantly higher in the older age group (24-26 months). (Fig. 1c, Fig. 5c). An increase in PD-L1 cell population in Tom- cells of aged mice was also observed in liver and kidney, but not in lung (Fig. 5d). To further characterize the in vivo dynamics of senescent cells, we pulse-labeled these cells with tamoxifen (TAM) at 2.5 months of age and then sacrificed the mice at 3, 6, and 9 months of age (Figure 1d). The proportion of PD-L1 + cells among Tom + cells in the lungs, liver, and kidneys was higher in the 9-month-old group than in the 3-month-old and 6-month-old groups; A tendency for + cells to accumulate was suggested.
老化細胞におけるPD-L1発現の制御機構を明らかにするために、我々は in vitro 老化モデルを使用した。PD-L1レベルは、増殖細胞では高く、静止細胞及び老化細胞では低かった(図6a)。PD-L1転写は細胞周期に依存しており、S期で最も高かった(図6b)。DREAM複合体を形成し、静止細胞及び老化細胞でE2F活性を抑制するドミナント ポケット タンパク質であるp130(非特許文献11、12)、又はドミナントネガティブ変異体E2F(E2F97)(非特許文献13)は。どちらも増殖細胞で過剰発現すると PD-L1発現を抑制した(図6c及び6d)。これと一致して、ポケットタンパク質ファミリーを阻害するヒトパピローマウイルスタイプ16E7タンパク質(非特許文献14)の過剰発現は、老化誘導からの経過時間に関係なくPD-L1発現を回復した(図6e)。したがって、E2Fの活性が非常に低いため、静止細胞及び老化細胞におけるPD-L1の発現が抑制される可能性がある。 To clarify the regulatory mechanism of PD-L1 expression in senescent cells, we used an in vitro aging model. PD-L1 levels were high in proliferating cells and low in quiescent and senescent cells (Fig. 6a). PD-L1 transcription was cell cycle dependent and was highest in S phase (Fig. 6b). p130, a dominant pocket protein that forms the DREAM complex and suppresses E2F activity in quiescent cells and senescent cells (Non-Patent Documents 11, 12), or the dominant-negative mutant E2F (E2F97) (Non-Patent Document 13). Both suppressed PD-L1 expression when overexpressed in proliferating cells (Figures 6c and 6d). Consistent with this, overexpression of the human papillomavirus type 16E7 protein, which inhibits the pocket protein family (14), restored PD-L1 expression regardless of the time elapsed since senescence induction (Fig. 6e). Therefore, the very low activity of E2F may suppress PD-L1 expression in quiescent and senescent cells.
培養細胞では、老化細胞の6~10%のみがPD-L1を発現していたが、静止細胞ではその不均一な発現は観察されなかった(図1a)。PD-L1-及びPD-L1+ d-Sen HCA2細胞をバルクRNA-seq解析用に分類した(図6f)。本発明者らは、転写因子結合モチーフ濃縮ツールiRegulon(非特許文献15、16)を使用して、230個のPD-L1正相関遺伝子(R2>0.95)の転写制御ネットワークを構築した(図6g)。予測されたマスター調節因子の候補のうち、E2F1結合モチーフのみがPD-L1のプロモーター領域に濃縮されており、弱いE2F1活性がPD-L1+老化細胞に残存し、PD-L1転写をわずかに誘導する可能性があることが示唆された。この弱いE2F1活性は、老化細胞のG1/S移行を引き起こすには十分ではない可能性がある。しかし、これだけでは、PD-L1+細胞におけるかなり高いレベルのPD-L1タンパク質発現を説明できない(図6h)。これに関連して、PD-L1+老化細胞でも大量のタンパク質凝集体が検出された(図1e)。プロテアソーム分解ががん細胞におけるPD-L1の安定性の調節に重要な役割を果たしていること(非特許文献17、18)を考えると、老化細胞におけるPD-L1発現の不均一性は、個々の老化細胞におけるプロテアソーム活性などの転写非依存性メカニズムによるものである可能性がある。この考えは、老化細胞間でタンパク質凝集体のレベルが異なるという事実によって裏付けられた。PD-L1+老化細胞では、キモトリプシン様プロテアーゼ活性、トリプシン様プロテアーゼ活性、及びカスパーゼ様プロテアーゼ活性(非特許文献19)が、PD-L1-細胞よりも低かった(図1f)。エポキソミシンやボルテゾミブなどのプロテアソーム阻害剤で老化細胞を処理すると、PD-L1及びPD-L1+老化細胞集団の発現が増加した(図1g~図1h)。老化細胞におけるプロテアソーム活性を増強するために、本発明者らは、構成的に活性なNRF1変異体(NRFΔ1-103)(非特許文献20、21)の発現が、PD-L1発現に及ぼす影響を調べた。NRFΔ1-103の過剰発現により、PD-L1+老化細胞の数が大幅に減少した (図1i)。これらの結果は、老化細胞におけるPD-L1の不均一な蓄積が、少なくとも部分的にはE2F1媒介転写とプロテアソーム活性の低下によるものであることを示唆している。 In cultured cells, only 6-10% of senescent cells expressed PD-L1, whereas its heterogeneous expression was not observed in quiescent cells (Fig. 1a). PD-L1 − and PD-L1 + d-Sen HCA2 cells were sorted for bulk RNA-seq analysis (Fig. 6f). The present inventors constructed a transcriptional regulatory network of 230 PD-L1 positively correlated genes (R 2 >0.95) using the transcription factor binding motif enrichment tool iRegulon (Non-Patent Documents 15, 16). (Figure 6g). Among the predicted master regulator candidates, only E2F1-binding motifs are enriched in the promoter region of PD-L1, and weak E2F1 activity remains in PD-L1 + senescent cells and slightly induces PD-L1 transcription. It was suggested that there is a possibility that This weak E2F1 activity may not be sufficient to trigger the G1/S transition of senescent cells. However, this alone cannot explain the significantly higher level of PD-L1 protein expression in PD-L1 + cells (Fig. 6h). In this context, large amounts of protein aggregates were also detected in PD-L1 + senescent cells (Fig. 1e). Considering that proteasomal degradation plays an important role in regulating PD-L1 stability in cancer cells (Non-Patent Documents 17, 18), the heterogeneity of PD-L1 expression in senescent cells may be due to individual This may be due to transcription-independent mechanisms such as proteasome activity in senescent cells. This idea was supported by the fact that levels of protein aggregates differ between senescent cells. In PD-L1 + senescent cells, chymotrypsin-like protease activity, trypsin-like protease activity, and caspase-like protease activity (Non-Patent Document 19) were lower than in PD-L1 − cells (Fig. 1f). Treatment of senescent cells with proteasome inhibitors such as epoxomicin and bortezomib increased the expression of PD-L1 and PD-L1 + senescent cell populations (Figures 1g-1h). To enhance proteasome activity in senescent cells, we investigated the effect of expression of a constitutively active NRF1 mutant (NRFΔ1-103) (Non-Patent Documents 20, 21) on PD-L1 expression. Examined. Overexpression of NRFΔ1-103 significantly reduced the number of PD-L1 + senescent cells (Fig. 1i). These results suggest that the heterogeneous accumulation of PD-L1 in senescent cells is due, at least in part, to reduced E2F1-mediated transcription and proteasome activity.
[実施例2]
次に、PD-L1が細胞老化におけるT細胞免疫に対する免疫チェックポイントとして機能するかどうかを調べた。マウス活性化初代CD8+T細胞をエフェクター細胞として、初代マウス肺線維芽細胞(MPF)を標的細胞として共培養し、細胞毒性を分析するために同系in vitro T細胞殺傷アッセイを実施した。老化誘導は、n-Sen及び d-Sen MPFにおけるSA-β-gal染色とp16発現によって定量された(図7a及び7b)。MPFでは、老化細胞ではPD-L1の不均一な発現が観察されたが、静止細胞では観察されなかった(図2a)。共培養後、老化細胞は、全ての誘導方法において、静止細胞よりもT細胞に対してより感受性が高かった(図2b、図7c)。SASPは細胞老化の代表的な表現型(非特許文献22)であり、免疫細胞を損傷組織に誘引することで老化細胞の免疫監視に関与していることも知られている(非特許文献23)。n-Sen MPFとQuie MPF間の転写シグネチャの違いを、バルクRNA-seqによって調べた(図2c)。
[Example 2]
Next, we investigated whether PD-L1 functions as an immune checkpoint for T cell immunity during cellular senescence. Mouse activated primary CD8 + T cells were co-cultured as effector cells and primary mouse lung fibroblasts (MPF) as target cells, and a syngeneic in vitro T cell killing assay was performed to analyze cytotoxicity. Senescence induction was quantified by SA-β-gal staining and p16 expression in n-Sen and d-Sen MPFs (Figures 7a and 7b). In MPF, heterogeneous expression of PD-L1 was observed in senescent cells but not in quiescent cells (Fig. 2a). After co-culture, senescent cells were more sensitive to T cells than quiescent cells in all induction methods (Fig. 2b, Fig. 7c). SASP is a typical phenotype of cellular aging (Non-Patent Document 22), and is also known to be involved in immune surveillance of senescent cells by attracting immune cells to damaged tissues (Non-Patent Document 23). ). Differences in transcriptional signatures between n-Sen MPF and Quie MPF were investigated by bulk RNA-seq (Fig. 2c).
遺伝子オントロジー(GO)及び遺伝子セットエンリッチメント解析(GSEA)により、炎症、サイトカイン産生、T細胞活性化、及び抗原提示に関連するn-Sen MPFのタームが非常に豊富であることが明らかになった(図2d、図7d、図7e)。静止細胞はT細胞の細胞毒性を示さなかったが、老化細胞からの馴化培地でのインキュベーションにより、細胞毒性がわずかではあるが有意に増加した(図2e)。重要なことに、非分解性 IκBα(S32/36A)を発現する老化細胞からの馴化培地とのインキュベーションは、T細胞の細胞毒性を増加させず、老化細胞馴化培地による細胞毒性の増強は、NF-κB依存性であることが示唆された (図 1、図2e、図7f~図7h)。 Gene ontology (GO) and gene set enrichment analysis (GSEA) revealed that terms in n-Sen MPF related to inflammation, cytokine production, T cell activation, and antigen presentation are highly enriched. (Fig. 2d, Fig. 7d, Fig. 7e). Although resting cells did not exhibit T cell cytotoxicity, incubation with conditioned medium from senescent cells resulted in a small but significant increase in cytotoxicity (Fig. 2e). Importantly, incubation with conditioned medium from senescent cells expressing non-degradable IκBα (S32/36A) did not increase cytotoxicity of T cells, and enhancement of cytotoxicity by senescent cell conditioned medium was due to NF -κB dependence was suggested (Fig. 1, Fig. 2e, Fig. 7f to Fig. 7h).
最近、転移因子(TE)及びTEにコードされた内因性レトロウイルスエレメント(ERV)が、ネオアンチゲンとしてMHCクラス I に提示され、腫瘍固有の免疫原性に重要な役割を果たしていることが示された(非特許文献24、25)。RNA-seq分析により、老化細胞では44個のTEと20個のERVが転写活性化されていることが明らかになった(図2f)。それらのうち、いくつかのERVは、Tom-細胞と比較して、p16-Tomマウスから選別されたTom+細胞で上方制御されていることが判明した(図2g)。さらに、MHCクラスI抗原プロセシング機構(非特許文献26)の正の調節因子であるI型インターフェロンシグナル伝達は、老化細胞において増強された(図2d)。したがって、老化細胞は、一般に、炎症誘発性サイトカインの分泌と内因性抗原の提示を介して、細胞傷害性T細胞免疫を活性化する可能性がある。 Recently, it has been shown that transposable elements (TEs) and TE-encoded endogenous retroviral elements (ERVs) are presented in MHC class I as neoantigens and play an important role in tumor-specific immunogenicity. (Non-patent Documents 24, 25). RNA-seq analysis revealed that 44 TEs and 20 ERVs were transcriptionally activated in senescent cells (Fig. 2f). Among them, some ERVs were found to be upregulated in Tom+ cells sorted from p16-Tom mice compared to Tom- cells (Fig. 2g). Furthermore, type I interferon signaling, a positive regulator of the MHC class I antigen processing machinery (Non-Patent Document 26), was enhanced in senescent cells (Fig. 2d). Therefore, senescent cells generally have the potential to activate cytotoxic T cell immunity through secretion of proinflammatory cytokines and presentation of endogenous antigens.
さらに、活性化されたT細胞は、PD-L1-老化細胞に対して有意に細胞毒性を誘導したが、PD-L1+老化細胞に対しては誘導しなかった(図2h、図8a)。この細胞毒性が抗原提示に依存しているかどうかを判断するために、OT-1マウスのT細胞を用いて実験を繰り返したところ、オボアルブミン(OVA)ペプチド処理により老化細胞に対する細胞毒性が劇的に増加することがわかった(図8b)。これは、老化細胞に対するCD8+T細胞の細胞毒性が、抗原提示に依存していることを示唆している。PD-L1+老化細胞に対するこれらの増加は、PD-L1-細胞に対する増加よりも著しく小さい程度であった(図8b)。これと一致して、PD-L1の過剰発現は、老化細胞における細胞傷害性を有意に抑制したが、これは、中和抗PD-1抗体(αPD-1)とのインキュベーションによりPD-1受容体をブロックすることで回復した(図2i、図8c、図8d)。同様の結果が、OT-1マウスのOVAペプチド活性化T細胞でも得られた(図8e)。これらの結果は、T細胞免疫から逃れるためには、老化細胞におけるPD-L1発現が必要であることを示唆している。 Furthermore, activated T cells significantly induced cytotoxicity against PD-L1 − senescent cells, but not against PD-L1 + senescent cells (Fig. 2h, Fig. 8a). To determine whether this cytotoxicity is dependent on antigen presentation, we repeated the experiment using T cells from OT-1 mice and found that ovalbumin (OVA) peptide treatment resulted in dramatic cytotoxicity toward senescent cells. (Figure 8b). This suggests that the cytotoxicity of CD8 + T cells toward senescent cells is dependent on antigen presentation. These increases for PD-L1 + senescent cells were of a significantly smaller extent than for PD-L1 − cells (Fig. 8b). Consistent with this, overexpression of PD-L1 significantly suppressed cytotoxicity in senescent cells, which was inhibited by incubation with a neutralizing anti-PD-1 antibody (αPD-1). It was recovered by blocking the body (Fig. 2i, Fig. 8c, Fig. 8d). Similar results were obtained with OVA peptide-activated T cells from OT-1 mice (Fig. 8e). These results suggest that PD-L1 expression in senescent cells is required for escape from T cell immunity.
[実施例3]
特定の細胞型におけるin vivoの生理学的表現型に対するPD-L1+老化細胞の影響を決定するために、p16-Tomマウスによって提供されるシングルセルRNAシーケンス(scRNA-seq)データセットを調べた(非特許文献10)。各データセットでは、さらなる分析のために、(1)正常肝臓及び非アルコール性脂肪性肝炎(NASH)肝臓からの肝臓類洞内皮細胞(LSEC)、(2)NASH肝臓からのNASH関連マクロファージ(NAM)、及び(3)正常な腎臓からの血管細胞など、豊富な細胞タイプのセットが登録された(図3a、図9a)。Tom+細胞とTom-細胞は、Cd274の転写物に基づいてPD-L1+及びPD-L1-として区別された (図3b、図9b)。PD-L1+及びPD-L1- LSECにおいて示差的に発現された遺伝子(DEG)は、Tom+及びTom- LSECで別々に同定された(図3c)。Tom- LSECのDEGの数(1個のアップDEG、2個のダウンDEG)と比較して、Tom+ LSECでははるかに多数のDEG(71個のアップDEG、9個のダウン DEG)が検出された。LSECは、p16Ink4aの上方制御とともに、高齢マウスにおいて中程度の炎症性であることが示されている(非特許文献27、28)。GO分析は、PD-L1+/Tom+ LSECから上方制御されたDEG(71遺伝子)が、サイトカイン応答と炎症性シグナル伝達に富んでいることを示した(図3d、図3e)。さらに、GSEAの結果は、インターフェロン、TNF-α、及び炎症性シグナル伝達が、PD-L1-/Tom+ LSECよりもPD-L1+/Tom+ LSECでより活性化されていることを明らかにした(図3f)。炎症サインは、PD-L1+/Tom+ NASH LSEC、NAM、及び腎血管細胞でも、豊富であった(図5c~図5h)。これらの結果は、炎症促進などの老化によって誘導される表現型が、PD-L1+老化細胞集団の存在及び選択的蓄積と、正の相関があることを示唆した。
[Example 3]
To determine the impact of PD-L1 + senescent cells on in vivo physiological phenotypes in specific cell types, we examined a single-cell RNA-sequencing (scRNA-seq) dataset provided by p16-Tom mice ( Non-Patent Document 10). For each dataset, we included (1) liver sinusoidal endothelial cells (LSEC) from normal liver and nonalcoholic steatohepatitis (NASH) liver, (2) NASH-associated macrophages (NAM) from NASH liver, and ), and (3) a rich set of cell types were enrolled, including vascular cells from normal kidney (Fig. 3a, Fig. 9a). Tom + and Tom − cells were differentiated as PD-L1 + and PD-L1 − based on Cd274 transcripts (Fig. 3b, Fig. 9b). Differentially expressed genes (DEGs) in PD-L1 + and PD-L1 - LSECs were identified separately in Tom + and Tom - LSECs (Fig. 3c). Compared to the number of DEGs in Tom − LSEC (1 up DEG, 2 down DEGs), a much larger number of DEGs (71 up DEGs, 9 down DEGs) were detected in Tom + LSEC. Ta. LSECs have been shown to be moderately inflammatory in aged mice, with upregulation of p16 Ink4a (27, 28). GO analysis showed that DEGs (71 genes) upregulated from PD-L1 + /Tom + LSECs were enriched in cytokine responses and inflammatory signaling (Fig. 3d, Fig. 3e). Furthermore, GSEA results revealed that interferon, TNF-α, and inflammatory signaling were more activated in PD-L1 + /Tom + LSECs than in PD-L1 − /Tom + LSECs. (Fig. 3f). Inflammatory signatures were also abundant in PD-L1 + /Tom + NASH LSECs, NAMs, and renal vascular cells (Figures 5c-5h). These results suggested that aging-induced phenotypes, such as pro-inflammation, were positively correlated with the presence and selective accumulation of PD-L1 + senescent cell populations.
[実施例4]
PD-1又はPD-L1に対するモノクローナル抗体による治療などの免疫チェックポイント遮断は、過去10年間にわたるがん免疫療法の大幅な進歩に貢献し、T細胞をPD-L1発現がん細胞にリダイレクトすることでさまざまな悪性腫瘍の治療に成功している(非特許文献29)。 in vivo での老化細胞の動態に対する抗 PD-1(αPD-1)抗体の効果を評価するために、2週間前に老化細胞用のTAMで標識した生後7か月のp16-Tomマウスに 、αPD-1抗体又はアイソタイプ コントロール(IgG)を3週間投与した(図4a)。さらに、αPD-1抗体の効果がCD8+ T細胞によるものであるかどうかを検証するために、実験全体を通して、抗CD8(αCD8)抗体処理によって細胞傷害性T細胞を全身的に減少させた(図10a)。αPD-1抗体を注射したマウスの肺、肝臓、腎臓のTom+細胞総数は、IgGグループよりも有意に低かった(図4a、図10b)。さらに、αPD-1抗体処理により、Tom+細胞ではPD-L1+亜集団が減少したが、Tom-細胞では減少しなかった(図10b)。さらに、αPD-1治療によりPD-L1-/Tom+細胞の集団がPD-L1+/Tom+細胞よりも少ない程度に減少したことに気づいた。これは、αPD-1抗体がPD-L1+/Tom+細胞を優先的に標的とすることを示す(図10c)。ただし、これらの減少効果は主にCD8+T細胞の減少によって回復したが、αPD-1は肺のTom+細胞をわずかに減少させる傾向があり、これは統計的に有意ではなかった(図4a)。これらの結果は、細胞傷害性T細胞による免疫が、実際に細胞老化の免疫監視に関与していることを示した。マクロファージのサブセットもp16を発現し、細胞老化のいくつかの特徴を備えているため、上記のp16-Tomマウスの肝臓でF4/80を発現するTom+細胞を他のTom+細胞から区別した(図11a)。 肝臓内のTom+細胞の大部分はF4/80陰性であり(図11b)、F4/80陽性細胞と陰性細胞の両方におけるTom+細胞の割合は、αPD-1抗体処理下では低かった(図11b、図11c)。さらに、αPD-1抗体治療により、F4/80陰性Tom+細胞におけるPD-L1+部分集団が減少した(図11d)。
[Example 4]
Immune checkpoint blockade, such as treatment with monoclonal antibodies against PD-1 or PD-L1, has contributed to significant advances in cancer immunotherapy over the past decade, redirecting T cells to PD-L1-expressing cancer cells. has been successfully used to treat various malignant tumors (Non-Patent Document 29). To assess the effect of anti-PD-1 (αPD-1) antibodies on senescent cell dynamics in vivo, we inoculated 7-month-old p16-Tom mice that had been labeled with TAM for senescent cells 2 weeks earlier. αPD-1 antibody or isotype control (IgG) was administered for 3 weeks (Figure 4a). Furthermore, to verify whether the effect of αPD-1 antibody was due to CD8 + T cells, cytotoxic T cells were systemically reduced by anti-CD8 (αCD8) antibody treatment throughout the experiment ( Figure 10a). The total number of Tom + cells in the lungs, liver and kidneys of mice injected with αPD-1 antibody was significantly lower than in the IgG group (Fig. 4a, Fig. 10b). Furthermore, αPD-1 antibody treatment decreased the PD-L1 + subpopulation in Tom + cells, but not in Tom − cells (Fig. 10b). Furthermore, we noticed that αPD-1 treatment reduced the population of PD-L1 − /Tom + cells to a lesser extent than PD-L1 + /Tom + cells. This indicates that the αPD-1 antibody preferentially targets PD-L1 + /Tom + cells (Figure 10c). However, these reducing effects were mainly reversed by the reduction of CD8 + T cells, whereas αPD-1 tended to slightly reduce Tom + cells in the lungs, which was not statistically significant (Fig. 4a) ). These results indicated that immunity by cytotoxic T cells is indeed involved in immune surveillance of cellular aging. We distinguished F4/80-expressing Tom + cells from other Tom + cells in the livers of p16-Tom mice as described above, as a subset of macrophages also express p16 and possess some hallmarks of cellular senescence ( Figure 11a). The majority of Tom + cells in the liver were F4/80 negative (Fig. 11b), and the proportion of Tom + cells in both F4/80 positive and negative cells was lower under αPD-1 antibody treatment (Fig. 11b, Figure 11c). Furthermore, αPD-1 antibody treatment reduced the PD-L1 + subpopulation in F4/80 negative Tom + cells (Fig. 11d).
in vivo で老化細胞の除去を標的とすることは、さまざまな老化及び加齢関連障害を改善することが報告されていることから、αPD-1抗体媒介老化細胞の除去が老化表現型も改善するかどうかをテストした。本発明者らは、加齢に伴う肺胞容積の増加(非特許文献31)、肝リピドーシス(非特許文献32)、握力の低下(非特許文献33)、及び運動能力の低下(非特許文献34)が、老齢(17.5か月齢)の野生型マウスにαPD-1抗体を注射することにより、IgG治療したグループと比較して有意に改善されることを発見した。(図4c、図4d、図11e、図11f)。そのような影響は、若い(3か月齢)マウスでは観察されなかった。 αPD-1 antibody-mediated senescent cell ablation also ameliorates the senescent phenotype, as targeting senescent cell ablation in vivo has been reported to ameliorate various aging and age-related disorders. I tested whether. The present inventors have discovered that age-related increases in alveolar volume (Non-Patent Document 31), hepatic lipidosis (Non-Patent Document 32), decrease in grip strength (Non-Patent Document 33), and decrease in exercise capacity (Non-Patent Document 33) (34) found that injecting old (17.5 months old) wild-type mice with αPD-1 antibody significantly improved the results compared to the IgG-treated group. (Fig. 4c, Fig. 4d, Fig. 11e, Fig. 11f). No such effects were observed in young (3 month old) mice.
細胞老化は、NASH(非特許文献35)の病因において重要な役割を果たしている。NASHは、マウスに、コリン欠乏L-アミノ酸規定高脂肪食(CDA-HFD)を与えることによって誘発される(非特許文献36)。生後1.5か月のマウスに通常の食餌又はCDA-HFDを7週間与え、その後、αPD-1抗体又はIgGを3週間注射した(図4e)。本発明者らはさらに、NASHの病因において(非特許文献37)、免疫チェックポイント阻害(ICB)治療によるPD-L1+老化細胞の除去が、老化細胞破壊治療薬であるABT263よりも有効であるかどうかを調査した。通常の食事を与えられたマウスの肝臓では、CD8+ T細胞がαPD-1抗体投与により中程度に活性化されたが、NASH状態ではαPD-1抗体投与とは無関係に強く活性化される一方、肝臓へのT細胞浸潤はNASH状態でのみαPD-1抗体投与により増強されることが、判明した(図12a)。CDA-HFDを与えられたマウスは、通常の食餌を与えられたマウスよりも肝臓の脂質低下、線維化、AST、ALT、及びLDHの血清レベルが高く、CDA-HFDがNASHの病因をうまく誘導したことを示す(図4e、図12b、図12c)。αPD-1抗体とABT263は両方とも病因を改善したが、αPD-1グループはABT263グループよりも脂肪肝の改善を示した(図4e)。さらに、体重と血清総コレステロール(T-CHO)レベルも同様の傾向を示し(図12d)、PD-L1+老化細胞クリアランスが肝臓からのLDL分泌を増加させ、NASHの進行における脂質蓄積を減少させる可能性があることを示唆した。 Cellular senescence plays an important role in the pathogenesis of NASH (35). NASH is induced by feeding mice a choline-deficient L-amino acid defined high-fat diet (CDA-HFD) (Non-Patent Document 36). 1.5-month-old mice were fed normal diet or CDA-HFD for 7 weeks and then injected with αPD-1 antibody or IgG for 3 weeks (Figure 4e). The present inventors further found that the elimination of PD-L1 + senescent cells by immune checkpoint blockade (ICB) therapy is more effective than the senolytic drug ABT263 in the pathogenesis of NASH (37). We investigated whether In the livers of mice fed a normal diet, CD8 + T cells were moderately activated by αPD-1 antibody administration, whereas in NASH conditions they were strongly activated independent of αPD-1 antibody administration. It was found that T cell infiltration into the liver was enhanced by αPD-1 antibody administration only in the NASH state (Fig. 12a). Mice fed CDA-HFD had higher hepatic hypolipidemia, fibrosis, and serum levels of AST, ALT, and LDH than mice fed a normal diet, demonstrating that CDA-HFD successfully induces NASH pathogenesis. (Fig. 4e, Fig. 12b, Fig. 12c). Both αPD-1 antibody and ABT263 ameliorated the pathogenesis, but the αPD-1 group showed more improvement in hepatic steatosis than the ABT263 group (Fig. 4e). Moreover, body weight and serum total cholesterol (T-CHO) levels showed similar trends (Fig. 12d), suggesting that PD-L1 + senescent cell clearance increases LDL secretion from the liver and reduces lipid accumulation in the progression of NASH. suggested that it was possible.
ICB投与により上記のTom+マクロファージの一部が枯渇したため、ICB投与による疾患状態の改善が主にマクロファージ枯渇によるものであるかどうかをさらに明らかにするために、PLX3397(CSF1R阻害剤)をマウスに投与した。結果は、PLX3397の投与が、老齢マウスの肺胞サイズ、握力、又は運動能力に有意な影響を及ぼさないことを示した(図13a~13d)。マクロファージの活性化がNASHの進行に関与していることが報告されている(非特許文献38)。実際、CSF1R阻害は、血清AST、ALT、LDH、肝リピドーシス、及び線維症を含むNASH病理のいくつかの指標を改善することが判明した(図13e~13h)。重要なことに、αPD-1抗体と PLX3397 の同時治療により、肝線維症を除くNASH病状がさらに改善された。これらの結果は、NASH進行に対するPD-1の影響は、炎症性マクロファージ集団の除去によるものではないことを示唆している。αPD-1抗体の効果が老化細胞の減少によるものであることを確認するために、NASH誘導後のTAM投与によりp16-Tomマウスの老化細胞を標識した。NASH誘発Tom+細胞は、αPD-1処理により大幅に減少した(図4f)。これらの結果は、老化とNASHに依存する老化細胞の蓄積には、細胞性免疫から逃れるためのPD-L1/PD-1軸が関与していることを示唆している。したがって、ICBの投与は、T細胞への抑制シグナルを弱め、免疫監視を再活性化し、老化細胞を除去し、加齢に関連するさまざまな表現型を改善する可能性がある。 Because ICB administration depleted a portion of the Tom + macrophages mentioned above, we administered PLX3397 (a CSF1R inhibitor) to mice to further clarify whether the amelioration of the disease state by ICB administration was mainly due to macrophage depletion. administered. The results showed that administration of PLX3397 did not significantly affect alveolar size, grip strength, or exercise capacity in aged mice (Figures 13a-13d). It has been reported that macrophage activation is involved in the progression of NASH (Non-Patent Document 38). Indeed, CSF1R inhibition was found to improve several indicators of NASH pathology, including serum AST, ALT, LDH, hepatic lipidosis, and fibrosis (Figures 13e-13h). Importantly, co-treatment of αPD-1 antibody and PLX3397 further improved NASH pathology except liver fibrosis. These results suggest that the effect of PD-1 on NASH progression is not due to removal of the inflammatory macrophage population. To confirm that the effect of αPD-1 antibody was due to the reduction of senescent cells, senescent cells of p16-Tom mice were labeled by TAM administration after NASH induction. NASH-induced Tom + cells were significantly reduced by αPD-1 treatment (Fig. 4f). These results suggest that aging and NASH-dependent accumulation of senescent cells involve the PD-L1/PD-1 axis to escape from cell-mediated immunity. Therefore, administration of ICB may attenuate inhibitory signals to T cells, reactivate immune surveillance, eliminate senescent cells, and ameliorate various age-related phenotypes.
結論として、今回の結果は、老化細胞の免疫監視にはCD8+ T細胞が関与しており、PD-L1/PD-1免疫チェックポイントが老化細胞の年齢依存的な蓄積に少なくとも部分的に関与していることを示唆している。 最近、加齢に伴う神経変性患者や百寿者において、特定のTCRを持つCD8+ T細胞及び細胞傷害性 CD4+T細胞のクローン増殖が発見された(非特許文献39、40)。さらに、CD8+メモリーT細胞のクローン性は、糖尿病や肝線維症などの加齢に伴う疾患の誘発によって拡大する(非特許文献41、42)。これら一連の証拠は、MHC-Iでの抗原提示と適応免疫系の活性化が、一般に、加齢に伴う疾患の進行中に起こる可能性があることを示唆している。 In conclusion, our results suggest that CD8 + T cells are involved in the immune surveillance of senescent cells and that the PD-L1/PD-1 immune checkpoint is at least partially involved in the age-dependent accumulation of senescent cells. suggests that it is. Recently, clonal expansion of CD8 + T cells and cytotoxic CD4 + T cells with specific TCRs has been discovered in patients with age-related neurodegeneration and centenarians (Non-Patent Documents 39, 40). Furthermore, the clonality of CD8 + memory T cells is expanded by the induction of age-related diseases such as diabetes and liver fibrosis (Non-Patent Documents 41, 42). These lines of evidence suggest that antigen presentation at MHC-I and activation of the adaptive immune system may commonly occur during the progression of age-related diseases.
本発明者らの以前のシングルセル分析は、老化細胞が細胞の種類と生体内での老化誘導の原因に応じて非常に不均一であることを示唆した。したがって、同じ種類の細胞であっても老化細胞は不均一性を示す可能性がある。本研究では、同じ細胞型(LSEC、NAM、又は腎血管細胞)であっても、PD-L1+老化細胞はPD-L1-細胞よりもはるかに多くの炎症性トランスクリプトームを示した。したがって、PD-L1+老化細胞の蓄積は、加齢に伴う老化細胞の数の単純な増加よりも有害な結果をもたらす可能性がある。したがって、ICB治療によるPD-L1+老化蓄積の選択的阻害は、従来の老化治療よりも有望な老化治療戦略である。これに関して、アルツハイマー病モデルでは、PD-L1/PD-1軸をブロックすると病状が軽減され、認知障害が改善されることが報告されている(非特許文献43、44)。10か月のCDA-HFD誘導を伴う NASHモデルにαPD-1抗体を比較的長期間(8週間) 投与すると、肝細胞癌(HCC)の負担が増加することが報告されている(非特許文献45)。さらに、長期にわたるαPD-1抗体投与は、全身性自己免疫疾患を誘発することが多かった(非特許文献46)。しかし、これらは我々の短期治療(3週間のαPD-1抗体治療とそれに続く10週間のCDA-HFD誘導)では観察されなかった。したがって、加齢関連疾患における免疫療法を制御するには、投与量と頻度を減らすだけでなく、免疫クリアランスの強化、急性炎症への耐性、組織修復速度のバランスを考慮し、投与量と頻度を減らすことを考慮して、各治療法の使用を最適化する必要があるかもしれない。 Our previous single-cell analysis suggested that senescent cells are highly heterogeneous depending on the cell type and the cause of senescence induction in vivo. Therefore, senescent cells may exhibit heterogeneity even among cells of the same type. In this study, PD-L1 + senescent cells showed much more inflammatory transcriptome than PD- L1− cells, even within the same cell type (LSEC, NAM, or renal vascular cells). Therefore, the accumulation of PD-L1 + senescent cells may have more deleterious consequences than a simple increase in the number of senescent cells with age. Therefore, selective inhibition of PD-L1 + senescence accumulation by ICB treatment is a more promising senescence treatment strategy than conventional senescence treatments. In this regard, it has been reported that in Alzheimer's disease models, blocking the PD-L1/PD-1 axis alleviates the disease state and improves cognitive impairment (Non-Patent Documents 43, 44). It has been reported that relatively long-term (8 weeks) administration of αPD-1 antibody to a NASH model with 10 months of CDA-HFD induction increases the burden of hepatocellular carcinoma (HCC) (Non-patent literature). 45). Furthermore, long-term αPD-1 antibody administration often induced systemic autoimmune diseases (Non-Patent Document 46). However, these were not observed with our short-term treatment (3 weeks of αPD-1 antibody treatment followed by 10 weeks of CDA-HFD induction). Therefore, controlling immunotherapy in age-related diseases requires not only reducing the dose and frequency, but also considering the balance between enhanced immune clearance, resistance to acute inflammation, and tissue repair rate. It may be necessary to optimize the use of each treatment with consideration to reducing
Claims (7)
Applications Claiming Priority (2)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| US202263408105P | 2022-09-20 | 2022-09-20 | |
| US63/408,105 | 2022-09-20 |
Publications (1)
| Publication Number | Publication Date |
|---|---|
| WO2024063109A1 true WO2024063109A1 (en) | 2024-03-28 |
Family
ID=90454601
Family Applications (1)
| Application Number | Title | Priority Date | Filing Date |
|---|---|---|---|
| PCT/JP2023/034172 Ceased WO2024063109A1 (en) | 2022-09-20 | 2023-09-20 | Senescent cell remover |
Country Status (1)
| Country | Link |
|---|---|
| WO (1) | WO2024063109A1 (en) |
Citations (1)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| JP2020525021A (en) * | 2017-06-29 | 2020-08-27 | ファンダシオン デル セクトル プーブリコ エスタタル セントロ ナショナル デ インベスティゲーショネス オンコロジカス カルロス 3(エフ エス ピー クニオ) | Identification and elimination of damaged and/or senescent cells |
-
2023
- 2023-09-20 WO PCT/JP2023/034172 patent/WO2024063109A1/en not_active Ceased
Patent Citations (1)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| JP2020525021A (en) * | 2017-06-29 | 2020-08-27 | ファンダシオン デル セクトル プーブリコ エスタタル セントロ ナショナル デ インベスティゲーショネス オンコロジカス カルロス 3(エフ エス ピー クニオ) | Identification and elimination of damaged and/or senescent cells |
Non-Patent Citations (5)
| Title |
|---|
| NAKANISHI, MAKOTO: "Immune surveillance of senescence and senolysis", CELL / SAIBO, NYU SAIENSUSHA - NEW SCIENCE COMP., JP, vol. 55, no. 2, 1 February 2023 (2023-02-01), JP , pages 96 - 97, XP009554150, ISSN: 1346-7557 * |
| OMORI, SATOTAKA ET AL.: "Future perspective of p16 reporter mouse models", RINSHO MENEKI ARERUGIKA = CLINICAL IMMUNOLOGY & ALLERGOLOGY, JP, vol. 75, no. 6, 1 January 2021 (2021-01-01), JP , pages 753 - 758, XP009554154, ISSN: 1881-1930 * |
| WANG TEH-WEI, JOHMURA YOSHIKAZU, SUZUKI NARUMI, OMORI SATOTAKA, MIGITA TOSHIRO, YAMAGUCHI KIYOSHI, HATAKEYAMA SEIRA, YAMAZAKI SATO: "Blocking PD-L1–PD-1 improves senescence surveillance and ageing phenotypes", NATURE, SPRINGER NATURE LIMITED, vol. 611, no. 7935, 10 November 2022 (2022-11-10), pages 358 - 364, XP093150465, ISSN: 0028-0836, DOI: 10.1038/s41586-022-05388-4 * |
| WANG, TEH-WEI ET AL.: "2S07e-05 Immune Checkpoint Inhibitor-Based Aging Cell Removal and Individual Aging Regulation", PROCEEDINGS OF THE 94TH MEETING OF THE JAPANESE BIOCHEMICAL SOCIETY; NOVEMBER 3-5, 2021, JAPANESE BIOCHEMICAL SOCIETY, JP, vol. 94, 1 January 2021 (2021-01-01) - 5 November 2021 (2021-11-05), JP, pages 1, XP009553758 * |
| WANG, TEH-WEI ET AL.: "Involvement of the PD-L1-PD-1 system in aging", RINSHO MENEKI ARERUGIKA = CLINICAL IMMUNOLOGY & ALLERGOLOGY, JP, vol. 80, no. 1, 1 July 2023 (2023-07-01), JP , pages 94 - 100, XP009554155, ISSN: 1881-1930 * |
Similar Documents
| Publication | Publication Date | Title |
|---|---|---|
| Wang et al. | Blocking PD-L1–PD-1 improves senescence surveillance and ageing phenotypes | |
| Wei et al. | miRNA-181a over-expression in mesenchymal stem cell-derived exosomes influenced inflammatory response after myocardial ischemia-reperfusion injury | |
| Bailey et al. | Pharmacologic inhibition of lysine-specific demethylase 1 as a therapeutic and immune-sensitization strategy in pediatric high-grade glioma | |
| Majumder et al. | Shifts in podocyte histone H3K27me3 regulate mouse and human glomerular disease | |
| Ye et al. | YAP1-mediated suppression of USP31 enhances NFκB activity to promote sarcomagenesis | |
| Hill et al. | Activation of NF-κB drives the enhanced survival of adipose tissue macrophages in an obesogenic environment | |
| Chen et al. | LncRNA Airn maintains LSEC differentiation to alleviate liver fibrosis via the KLF2-eNOS-sGC pathway | |
| Kim et al. | Histone deficiency and accelerated replication stress in T cell aging | |
| Lin et al. | Angiopoietin-like proteins stimulate HSPC development through interaction with notch receptor signaling | |
| Yue-Chun et al. | Vagus nerve plays a pivotal role in CD4+ T cell differentiation during CVB3-induced murine acute myocarditis | |
| Chen et al. | NF-κB p65 and SETDB1 expedite lipopolysaccharide-induced intestinal inflammation in mice by inducing IRF7/NLR-dependent macrophage M1 polarization | |
| EP3261647B1 (en) | Compositions and methods of treating fanconi anemia | |
| Boyd et al. | Nonhematopoietic cells represent a more rational target of in vivo hedgehog signaling affecting normal or acute myeloid leukemia progenitors | |
| US20150272970A1 (en) | Targeting chemotherapy agent resistance in cancer | |
| Liu et al. | POLR2A blocks osteoclastic bone resorption and protects against osteoporosis by interacting with CREB1 | |
| Wang et al. | ARC is a critical protector against inflammatory bowel disease (IBD) and IBD-associated colorectal tumorigenesis | |
| Gao et al. | The contribution of neuropilin‐1 in the stability of CD4+ CD25+ regulatory T cells through the TGF‐β1/Smads signaling pathway in the presence of lipopolysaccharides | |
| WO2024063109A1 (en) | Senescent cell remover | |
| Hou et al. | Decreased ZONAB expression promotes excessive transdifferentiation of alveolar epithelial cells in hyperoxia-induced bronchopulmonary dysplasia | |
| US20130072844A1 (en) | Use of entrained neutrophils to treat metastatic and micrometastatic disease in at risk patients | |
| JP2022513082A (en) | Use of IRE1α-XBP1 signaling pathway biomarkers to regulate immune response | |
| WO2023205802A2 (en) | Senolytic targets of senescent cells | |
| WO2022196747A1 (en) | Therapy for heart failure and concomitant diseases thereof, therapeutic agent, and diagnostic method | |
| US20220220206A1 (en) | Treating chronic liver disease | |
| Umphred-Wilson et al. | The ESCRT protein CHMP5 promotes T cell leukemia by controlling BRD4-p300-dependent transcription |
Legal Events
| Date | Code | Title | Description |
|---|---|---|---|
| 121 | Ep: the epo has been informed by wipo that ep was designated in this application |
Ref document number: 23868230 Country of ref document: EP Kind code of ref document: A1 |
|
| NENP | Non-entry into the national phase |
Ref country code: DE |
|
| 122 | Ep: pct application non-entry in european phase |
Ref document number: 23868230 Country of ref document: EP Kind code of ref document: A1 |
|
| NENP | Non-entry into the national phase |
Ref country code: JP |