FIGURE SUMMARY
Title

Telomere shortening produces an inflammatory environment that increases tumor incidence in zebrafish

Authors
Lex, K., Maia Gil, M., Lopes-Bastos, B., Figueira, M., Marzullo, M., Giannetti, K., Carvalho, T., Ferreira, M.G.
Source
Full text @ Proc. Natl. Acad. Sci. USA

Short telomeres promote tumorigenesis in a noncell autonomous manner. (A) Experimental setup for the generation of zebrafish chimeras. Donor cells are transplanted from a Tg(mitfa:HRASG12V; β-actin:GFP) embryo at the blastula stage into embryos resulting from an in cross of tert+/−; casper zebrafish. (B) Representative images of adult chimera zebrafish harboring melanoma in either WT or tert−/− recipients. (C) Melanoma occurrence over time in chimeric fish. tert−/− recipient fish have a higher risk of tumorigenesis than WT and het recipient fish. *P < 0.05.

tert−/− tissues increase melanoma invasiveness and progression. (A) Hematoxylin and eosin (H and E) images of melanoma arising in a WT (Upper) or tert−/− recipient fish. Strong infiltration into other tissues was typical in tert−/− fish but not in WT (arrowheads). (B) Melanomas were staged according to histopathology into benign lesions (melanosis), noninvasive and invasive malignant tumors (P = 0.0269; WT n = 9; tert−/− n = 10). (C) Analysis of malignant tumors for cellular atypia (P = 0.0278; WT n = 5; tert−/− n = 7). (D) Representative immunofluorescence images of PCNA-positive cells (red) in melanoma (green) developed in WT chimeras and in tert−/− chimeras. Dashed lines locate the skin (no green fluorescence), squares show the place of amplification, and arrows indicate PCNA-positive cells. (E) Quantification of PCNA-positive melanoma cells in WT and tert−/− chimeras. Data are represented as mean ± SEM. *P < 0.05; n = 3.

G2 tert−/− larvae with very short telomeres exhibit increased melanoma expansion. (A) Experimental design for melanoma allotransplants in zebrafish larvae. Melanoma tumors were dissected from Tg(mitfa:HRASG12V; β-actin:GFP) zebrafish and cells dissociated. HRAS melanoma cells were then injected into blood circulation of 2-dpf zebrafish larvae. Larvae were kept for 7-d-postinjection (7-dpi). (B) Representative images of HRAS melanoma cells spread (green) in WT or G2 tert−/− larvae at 7 dpi. (Scale bars: 0.5 mm.) (C) Time course of HRAS melanoma cells spread in a group of WT and G2 tert−/− larvae (P < 0.01 at 7 dpi; WT n = 10; G2 tert−/− n = 11). (D) Melanoma tumors are more disseminated in G2 tert−/− larvae (*P < 0.05; WT n = 32; G2 tert−/− n = 31). A linear regression of three time points (1, 4, and 7 dpi) was used to calculate the slope of melanoma expansion over time. Each dot represents one larvae allotransplant.

tert −/− tissues present increased levels of senescence (p15/16) and inflammation (TNF-α). (A) Representative immunofluorescence images of p15/16 and TNF-α in the skin near melanoma in chimeric WT and tert−/− zebrafish. Dashed lines locate the skin (no green fluorescence), and arrows indicate p15/16-positive cells. Squares with dashed lines show place of amplification in a sequential section, and squares with solid lines show place of amplification on that section. (B and C) Quantification of p15/16-positive cells and levels of TNF-α, respectively, in the skin of chimeric WT and tert−/− zebrafish (n = 3). (D–F) RT-qPCR analysis comparing the expression levels of cdkn2a/b (p15/16), cdkn1a (p21), and tnfa (TNF) of 4-dpf WT, G2 tert−/− larvae, 9-months WT, and tert−/− adult intestine tissue. Data are represented as mean ± SEM. (*P < 0.05; **P < 0.01 for n = 30).

G2 tert mutant cells induce high levels of senescence and systemic inflammation in host larvae. (A) Representative images of SA-β-Gal assay comparing WT and G2 tert−/− 4-dpf zebrafish embryos. Yolk sack staining is nonspecific. (B, Left) Scheme of chimera generation: G2 tert−/− blastula cells are transplanted into WT embryos (G2 tert−/− → WT). (B, Right) Marked senescence in 4-dpf WT embryos upon G2 tert−/− cell injection (C, Left) Scheme of chimera generation: WT or G2 tert−/− blastula cells transplanted into WT Tg(mpx:GFP) embryos with GFP-labeled neutrophils: WT → WT Tg(mpx:GFP) vs. G2 tert−/− → WT Tg(mpx:GFP). (C, Right) Representative images at 4 dpf, and neutrophils are shown in green. (Scale bars: 0.5 mm.) (D) Quantification of neutrophils at 4 dpf. Noninjected Tg(mpx:GFP) were used as controls. Each data point represents one zebrafish (**P > 0.01; noninjected n = 24; WT n = 33; tert−/− n = 25; ns, nonsignificant [P > 0.05]).

Increased tumor expansion in G2 tert−/− larvae is rescued by inflammation inhibition. (A) Allotransplants of primary tumor cells extracted from melanoma in adult fish into 2-dpf larvae that were treated with aspirin or Celecoxib (COX-2 selective inhibitor). (B) Representative images of melanoma expansion at 7 dpi upon aspirin treatment. (Scale bars: 0.5 mm.) (C) Time course of melanoma dissemination in WT and G2 tert−/− larvae under aspirin treatment. (D) Slope of HRAS melanoma spread among 1, 4, and 7 dpi. Comparison of tumor cell expansion in a WT or G2 tert−/− treated with aspirin (WT n = 26 and G2 tert−/− n = 29) or Celecoxib (WT n = 19 and G2 tert−/− n = 13). Each dot represents one larva allotransplant. ns, nonsignificant (P > 0.05).

Acknowledgments
This image is the copyrighted work of the attributed author or publisher, and ZFIN has permission only to display this image to its users. Additional permissions should be obtained from the applicable author or publisher of the image. Full text @ Proc. Natl. Acad. Sci. USA