Skip to main page content
U.S. flag

An official website of the United States government

Dot gov

The . gov means it’s official. Federal government websites often end in . gov or . mil. Before sharing sensitive information, make sure you’re on a federal government site VSports app下载. .

Https

The site is secure. The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely V体育官网. .

. 2019 Apr 2;10(1):1492.
doi: 10.1038/s41467-019-09525-y.

"V体育ios版" Gut microbiota dependent anti-tumor immunity restricts melanoma growth in Rnf5-/- mice

Affiliations

Gut microbiota dependent anti-tumor immunity restricts melanoma growth in Rnf5-/- mice

Yan Li et al. Nat Commun. .

Abstract (VSports app下载)

Accumulating evidence points to an important role for the gut microbiome in anti-tumor immunity. Here, we show that altered intestinal microbiota contributes to anti-tumor immunity, limiting tumor expansion. Mice lacking the ubiquitin ligase RNF5 exhibit attenuated activation of the unfolded protein response (UPR) components, which coincides with increased expression of inflammasome components, recruitment and activation of dendritic cells and reduced expression of antimicrobial peptides in intestinal epithelial cells. Reduced UPR expression is also seen in murine and human melanoma tumor specimens that responded to immune checkpoint therapy. Co-housing of Rnf5-/- and WT mice abolishes the anti-tumor immunity and tumor inhibition phenotype, whereas transfer of 11 bacterial strains, including B. rodentium, enriched in Rnf5-/- mice, establishes anti-tumor immunity and restricts melanoma growth in germ-free WT mice. Altered UPR signaling, exemplified in Rnf5-/- mice, coincides with altered gut microbiota composition and anti-tumor immunity to control melanoma growth VSports手机版. .

PubMed Disclaimer

Conflict of interest statement

The authors declare no competing interests.

Figures (V体育2025版)

Fig. 1
Fig. 1
Enhanced antitumor immune responses in Rnf5−/− mice. a Growth of YUMM1.5 (BrafV600E:PTEN−/−:Cdkn2a−/−) melanoma cells after subcutaneous injection of 106 cells into WT or Rnf5−/− mice (n = 5). b Quantification of tumor-infiltrating effector (CD44hi) CD4+ and CD8+ T cells and total CD45+ cells on day 24 after tumor injection (n = 5). c Frequencies of tumor-infiltrating TNF-α-, IFN-γ-, and IL-2-producing CD4+ and CD8+ T cells on day 24 after tumor inoculation (n = 5). d Quantification of tumor-infiltrating total DCs, pDCs, mDCs, and CD8α+ DCs on day 24 after tumor inoculation (n = 5). e Expression (mean fluorescence intensity, MFI) of MHC class II, CD40, CD80, and CD86 on tumor-infiltrating DCs (CD45+ CD11c+) on day 24 after tumor inoculation (n = 5). f Quantification of OT-I CD8+ T cells in tumor-draining lymph nodes (TdLN) and non-draining lymph nodes (ndLN) of CD45.1+ WT and Rnf5−/− mice injected with B16-OVA melanoma cells (WT, n = 6; Rnf5−/−, n = 5). g, h Growth of YUMM1.5 melanoma cells in mice injected i.p. with control IgG and anti-CD4 (g) or control IgG and anti-CD8 (h) depleting antibodies on days 0, 3, 6, 11, 16 (n = 9). FACS analysis revealed >90% depletion of blood CD4+ and CD8+ T cells on day 7 after tumor inoculation. i, Growth of YUMM1.5 melanoma cells in lethally irradiated bone marrow-reconstituted WT or Rnf5−/− mice (arrow indicates bone marrow donor → recipient; n = 7). Data are representative of three independent experiments (ae), two independent experiments (f, i) and one experiment (g, h) ≥5 mice per group. Graphs show the mean ± s.e.m. *P < 0.05, **P < 0.005, ***P < 0.001, ****P < 0.0001 by two-way ANOVA with Sidak’s correction (a, gi) or by two-tailed t-test or Mann–Whitney U test (bf)
Fig. 2
Fig. 2
Enhanced inflammasome and pathogen receptor signaling in Rnf5−/− mice. a NanoString analysis of PanCancer Immune Profiling genes in tumors from WT and Rnf5−/− mice. The heatmap shows 47 genes with >1.2-fold (P < 0.05) differences in expression between YUMM1.5 tumors from the two genotypes (n = 5) at 10 days after injection. b Growth of B16F10 melanoma cells after subcutaneous injection of 106 cells into WT, Rnf5−/−, MyD88/− and MyD88−/Rnf5−/ mice (WT, Rnf5−/−, n = 6; MyD88−/−, n = 5; MyD88−/− Rnf5−/−, n = 8). c qRT-PCR analysis of pathogen receptor mRNA levels in IECs from tumor-bearing WT or Rnf5−/− mice (n = 4). d qRT-PCR analysis of cytokine and chemokine mRNA levels in IECs isolated from the small intestines of tumor-bearing WT or Rnf5−/− mice (n = 4). Data are representative of three independent experiments (c, d), two independent experiments (b) or one experiment (a) ≥5 mice per group. Graphs show the mean ± s.e.m. *P < 0.05, **P < 0.005, ***P < 0.001, ****P < 0.0001 by two-tailed t test or Mann–Whitney U test (c, d) or two-way ANOVA with Sidak’s correction (b)
Fig. 3
Fig. 3
Gut microbiota control melanoma growth. a Principal component analysis (PCA) of all taxa enumerated in Rnf5−/− and WT fecal microbiota samples taken before (day 0) and 24 days after injection of YUMM1.5 tumor cells (n = 30). b Elimination of tumor growth suppression in Rnf5−/− mice by treatment with an antibiotic cocktail administered for 2 weeks prior to tumor cell injection (n = 5). c PCA of all taxa showing convergence of gut microbiota in WT and Rnf5−/− mice after co-housing (WT alone, n = 14; Rnf5−/ alone, n = 15; WT mixed, Rnf5/− mixed, n = 5). d Growth of YUMM1.5 melanoma cells in WT or Rnf5−/− mice housed alone or co-housed (mixed) for 4 weeks prior to tumor inoculation (alone, n = 15; mixed, n = 16). e Quantification of effector (CD44hi) CD4+ and CD8+ T cells, total CD45+ cells, and frequencies of IFN-γ + TNF-α-producing CD4+ and CD8+ T cells in tumors from WT or Rnf5−/− mice housed alone or co-housed for 4 weeks prior to tumor inoculation (n = 10). f Quantification of tumor-infiltrating total DCs, pDCs, and mDCs in WT or Rnf5−/− mice housed alone or co-housed for 4 weeks prior to tumor inoculation (n = 10). g A cladogram representation of taxa enriched in Rnf5−/− mice (red) microbiota and taxa enriched in WT mice (green) microbiota (n = 30). The background color indicates phylum; names of phyla are indicated. Data are representative of two independent experiments ≥5 mice per group. Graphs show the mean ± s.e.m. *P < 0.05, **P < 0.005, ***P < 0.001, ****P < 0.0001 by one-way ANOVA with Tukey’s (e, f) or two way ANOVA with Sidak’s (bd) correction for multiple comparisons
Fig. 4
Fig. 4
Oral administration of select bacterial strains enriched in Rnf5−/− mice to gnotobiotic mice enhances antitumor immune response. a YUMM1.5 tumor growth in germ-free (GF) mice undergoing oral gavage with WT or Rnf5−/− cecal contents 2 weeks prior to tumor implantation (n = 10). b Quantification of tumor-infiltrating CD45+ cells, effector (CD44hi) CD4+ and CD8+ T cells, total CD45+ cells, and frequencies of IFN-γ+TNF-α+-producing CD4+ and CD8+ T cells on day 24 after tumor injection in recipients treated as in (a) (n = 10). c YUMM1.5 tumor growth in GF mice undergoing oral gavage with either Altered Schaedler Flora (ASF) or ASF plus bacterial cocktail prior to tumor inoculation (ASF, n = 12; ASF + bacterial cocktail, n = 14). d Quantification of tumor-infiltrating effector (CD44hi) CD4+ and CD8+ T cells and frequencies of IFN-γ+TNF-α+-producing CD4+ and CD8+ T cells on day 21 after tumor injection in mice treated as in (c) (n = 8). e YUMM1.5 tumor growth in GF mice undergoing oral gavage with ASF or ASF plus B. rodentium prior to tumor inoculation (n = 15). f Quantification of tumor-infiltrating effector (CD44hi) CD4+ and CD8+ T cells and frequencies of IFN-γ+TNF-α+-producing CD8+ T cells on day 24 after tumor injection in mice treated as in (e) (n = 8). Data are one experiment ≥8 mice per group.Graphs are the mean ± s.e.m. *P < 0.05, **P < 0.005, ****P < 0.0001 by two-way ANOVA with Tukey’s or Sidak’s correction for multiple comparisons (a, c, e) or two-tailed t test or Mann–Whitney U test (b, d, f)
Fig. 5
Fig. 5
IEC of Rnf5−/− mice activate immune response and change anti-microbial peptide (AMP) expression. a Villi length and crypt depth calculated from H&E-stained sections of intestines from WT or Rnf5−/− mice (WT, n = 30; Rnf5−/−, n = 32). b qRT-PCR analysis of AMPs mRNA levels in IECs from small intestine of naive WT or Rnf5−/− mice (n = 6). c Representative images (left) and quantification (right) of cleaved caspase-3 immunostained small intestine organoids from tumor-bearing WT or Rnf5−/− mice (n = 3). Scale bar = 100μm. Graph shows percentage of cleaved caspase-3 + cells per immunostained organoid (n = 12 fields). d Intracellular IFN-γ and TNF-α staining of p14 CD8+ T cells incubated for 72 h with 2 μg/ml GP33 peptide recognized by the TCR of P14 and bone marrow-derived dendritic cells (BMDCs) that were incubated with medium alone (no stimulation) or with conditioned medium (CM) from shControl or shRNF5 MODE-K cells. e Representative images (left) and quantification (right) of CD11c+ cell immunostaining in the small intestine of WT or Rnf5−/− mice on day 24 after injection of YUMM1.5 cells. Scale bar = 50μm (n = 4). f Frequencies of total DCs and pDCs in Peyer’s patches from WT and Rnf5−/− mice on day 10 after YUMM1.5 cell injection (n = 6). g 10 days after tumor injection, DCs from GALT, dLN, and ndLN were isolated, pooled per group (n = 10 mice/group), and were incubated with OT-1 CD8+ T cells stimulated with 2 μg/ml OVA peptide (SINFEKL). Intracellular IFN-γ and TNF-α of OT-1 CD8+ T cells were detected. Data are representative of three independent experiments (b, d) and two independent experiments (a, c, e, f, g) ≥3 mice per group. Graphs show the mean ± s.e.m. *P < 0.05, **P < 0.005, ***P < 0.001, ****P < 0.0001 by two-tailed t test or Mann–Whitney U test (a, b, c, e, f) or one-way ANOVA with Tukey’s (d) correction for multiple comparisons
Fig. 6
Fig. 6
IEC of Rnf5−/− mice activate immune responses and changes AMPs expression by IRE1α/sXBP1 signaling. a Western blot analysis of the indicated proteins in lysates of MODE-K-shCon and MODE-K-shRNF5 cells treated with thapsigargin (TG, 1 μM) for indicated time. b qRT-PCR analysis of mRNA levels of sXBP1, XBP1 and target genes in small intestine-derived IECs from naive WT or Rnf5−/− mice (n = 4). c Representative micrographs of immunohistochemical (IHC) staining of BiP in the jejunum, ileum, and colon of YUMM1.5 tumor-bearing WT or Rnf5−/− mice (scale bar = 25 μm). Lower graphs show quantification of IHC staining (n = 12 fields per group). Staining was scored semi-quantitatively based on staining intensity (0, 1, 2, or 3) multiplied by the percentage of positively stained cells (0–100), to give a maximum IHC score of 300. d Quantification of BiP and CHOP IHC staining in ileum sections from WT and Rnf5−/− mice co-housed for 4 weeks prior to tumor inoculation (n = 12). e qRT-PCR analysis of sXBP1 mRNA levels in MODE-K-shCon and MODE-K-shRNF5 cells treated with the indicated concentration of MKC-4485 for 24 h (n = 3). f MFI of MHC class I and II on bone marrow-derived dendritic cells (BMDCs) incubated with media alone (no stimulation), conditioned media (CM) from shControl or shRNF5 MODE-K cells treated with MKC-4485 (n = 4). g qRT-PCR analysis of Defa1 mRNA levels in MODE-K-shCon and MODE-K-shRNF5 cells (n = 3). h qRT-PCR analysis of AMPs mRNA levels in IECs in MODE-K-shCon and MODE-K-shXBP1 cells (n = 3). i, qRT-PCR analysis of Defa1 mRNA levels in MODE-K cells treated with 2 μM MKC-4485 for 24 h (n = 3). Data are representative of three independent experiments (b, ei) and two independent experiments (a, c, d) ≥3 mice per group. Graphs show the mean ± s.e.m. *P < 0.05, **P < 0.005, ***P < 0.001, ****P < 0.0001 by two-tailed t test or Mann–Whitney U test (b, c, gi) or one-way ANOVA with Tukey’s correction for multiple comparisons (df)
Fig. 7
Fig. 7
Altered UPR in melanoma responders of anti-CTLA-4 immune therapy. a Analysis of ER stress markers genes in non-responders (NR) (n = 16) and responders (R) (n = 14) to immune checkpoint therapy in melanoma samples from MGH/DFCC (USA). b Analysis of ER stress markers genes in NR (n = 16) and R (n = 9) to anti-CTLA-4 immunotherapy in melanoma samples from Zurich Hospital (Switzerland). c Kaplan–Meier curves for progression-free survival probability in melanoma patients demonstrating low (n = 15 patients) versus high sXBP1(n = 14 patients) in tumor specimens obtained before immune checkpoint therapy at MGH/DFCC. d Kaplan–Meier estimates of progression-free survival probability in melanoma patients demonstrating low (n = 14 patients) versus high ATF4 (n = 16 patients) in tumor specimens obtained before immune checkpoint therapy in melanoma samples at MGH/DFCC. e Representative micrographs of IHC staining of BiP after anti-PD-1 immunotherapy in melanoma samples obtained at Rambam Health Care Center (Israel) (scale bar = 25 μm). f IHC score of BiP staining in NR (n = 10) and R (n = 11) to anti-PD-1 immunotherapy in melanoma samples from Rambam Health Care Center (Israel). g, h Kaplan–Meier estimates of (g) overall survival and (h) disease-free survival probability in melanoma patients demonstrating low (<25%, n = 12 patients) versus high melanoma cell expression of BiP (>25%, n = 9 patients) in tumor biospecimens obtained before initiation of systemic anti-PD-1 antibody treatment in melanoma samples from Rambam Health Care Center (Israel). i IHC staining of BiP in melanoma samples from select patients prior to, on and after immunotherapy treatment from MGH/DFCC (USA). Scale bar = 50 μm. Graphs show the mean ± s.e.m. *P < 0.05, **P < 0.005, ***P < 0.001, ****P < 0.0001 by two-tailed t-test or Mann–Whitney U test (a, b, f) or log-rank test (c, d, g, h)

"V体育官网入口" References

    1. Sharma P, Allison JP. Immune checkpoint targeting in cancer therapy: toward combination strategies with curative potential. Cell. 2015;161:205–214. doi: 10.1016/j.cell.2015.03.030. - DOI (VSports手机版) - PMC - PubMed
    1. Reddy SM, Reuben A, Wargo JA. Influences of BRAF inhibitors on the immune microenvironment and the rationale for combined molecular and immune targeted therapy. Curr. Oncol. Rep. 2016;18:42. doi: 10.1007/s11912-016-0531-z. - DOI - PMC - PubMed
    1. Sivan A, et al. Commensal Bifidobacterium promotes antitumor immunity and facilitates anti-PD-L1 efficacy. Science. 2015;350:1084–1089. doi: 10.1126/science.aac4255. - DOI (VSports最新版本) - PMC - PubMed
    1. Vetizou M, et al. Anticancer immunotherapy by CTLA-4 blockade relies on the gut microbiota. Science. 2015;350:1079–1084. doi: 10.1126/science.aad1329. - DOI (VSports) - PMC - PubMed
    1. Kuang E, Qi J, Ronai Z. Emerging roles of E3 ubiquitin ligases in autophagy. Trends Biochem. Sci. 2013;38:453–460. doi: 10.1016/j.tibs.2013.06.008. - DOI - PMC - PubMed

Publication types

MeSH terms

VSports最新版本 - Substances