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.

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.

Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
. 2016 Dec 8;1(20):e87446.
doi: 10.1172/jci.insight.87446.

Pentraxin-2 suppresses c-Jun/AP-1 signaling to inhibit progressive fibrotic disease

Affiliations

Pentraxin-2 suppresses c-Jun/AP-1 signaling to inhibit progressive fibrotic disease

Naoki Nakagawa et al. JCI Insight. .

Abstract

Pentraxin-2 (PTX-2), also known as serum amyloid P component (SAP/APCS), is a constitutive, antiinflammatory, innate immune plasma protein whose circulating level is decreased in chronic human fibrotic diseases. Here we show that recombinant human PTX-2 (rhPTX-2) retards progression of chronic kidney disease in Col4a3 mutant mice with Alport syndrome, reducing blood markers of kidney failure, enhancing lifespan by 20%, and improving histological signs of disease. Exogenously delivered rhPTX-2 was detected in macrophages but also in tubular epithelial cells, where it counteracted macrophage activation and was cytoprotective for the epithelium. Computational analysis of genes regulated by rhPTX-2 identified the transcriptional regulator c-Jun along with its activator protein-1 (AP-1) binding partners as a central target for the function of rhPTX-2. Accordingly, PTX-2 attenuates c-Jun and AP-1 activity, and reduces expression of AP-1-dependent inflammatory genes in both monocytes and epithelium. Our studies therefore identify rhPTX-2 as a potential therapy for chronic fibrotic disease of the kidney and an important inhibitor of pathological c-Jun signaling in this setting.

PubMed Disclaimer

Conflict of interest statement

R.M. Jack is the CSO, COO, and President of Promedior Inc. M.L. Lupher Jr. is a former member of Promedior Inc. J.S. Duffield holds stock options in Promedior Inc.

Figures

Figure 1
Figure 1. rhPTX-2 localizes to interstitial macrophages and proximal epithelial cells and protects Col4a3–/– mice from kidney disease progression.
Experimental schema indicating rhPTX-2 delivery from 3.5 weeks after birth and (A) analysis at 42 days (6 weeks) and 63 days (9 weeks) or (B) continued to 94 days (14 weeks). (C) Split-panel confocal images of 9-week-old Col4a3–/– kidney cortex showing distribution of rhPTX-2 colocalized with proximal tubular epithelial cells (lotus lectin [LTL], arrows) and macrophages (F4/80, arrowheads). Scale bars: 50 μm. Proportions of (D) hPTX-2+LTL+ proximal tubular epithelial cells and (E) hPTX2+F4/80+ macrophages at 9 weeks after twice weekly i.p. injections of rhPTX-2. (F) Plasma BUN levels at 9 weeks. (G) Urine albumin concentration normalized to urine creatinine at 9 weeks. (H) Body weights from start of treatment to death. (I) Kaplan-Meier survival curve. n = 6–12/group. *P < 0.05, ***P < 0.001 (ANOVA with post hoc testing for multiple comparisons and Gehan-Breslow-Wilcoxon test for survival). VEH, vehicle; n.d., not detected.
Figure 2
Figure 2. rhPTX-2 prevents glomerulosclerosis, glomerular basement membrane accumulation, and podocyte loss.
(A) Representative images of kidney cortex at 9 weeks of age showing periodic acid–Schiff– stained (PAS-stained) images of glomeruli with glomerulosclerosis (white arrows); WT-1 immunofluorescence for podocytes within glomeruli (dotted lines); electron microscopy (EM) images of glomerular capillary loops showing severe glomerular basement membrane (GBM) thickening with humps, and podocyte effacement (loss of processes) in vehicle-treated Col4a3–/– mice. rhPTX-2–treated Col4a3–/– mice show areas of preserved basement membrane and partial foot process effacement, but small classical basement membrane humps persist. U, urinary space; P, podocyte; Pp, podocyte processes; L, capillary lumen; EC, endothelial cell. (B) Glomerulosclerosis score ranged from none (0) to >75% of glomeruli affected (4). (C) WT-1+ podocytes per glomerular cross-section. (D) GBM thickness. Scale bars: 50 μm (light) or 500 nm (EM). n = 6–12/group. *P < 0.05, **P < 0.01 (ANOVA with post hoc testing for multiple comparisons). VEH, vehicle.
Figure 3
Figure 3. rhPTX-2 prevents tubular epithelial injury and interstitial fibrosis in Col4a3–/– kidneys.
(A) PAS-stained and Sirius red–stained images of kidney cortex. Note severe tubular injury with proteinaceous casts and loss of polarity and brush border of epithelium in Col4a3–/– mice (upper center panel). These pathological features are much milder following rhPTX-2 treatment. (B) Tubular injury score (C) Morphometric analysis of interstitial fibrosis detected by Sirius red staining. Scale bars: 50 μm. n = 6–12/group. **P < 0.01 (ANOVA with post hoc testing for multiple comparisons). VEH, vehicle.
Figure 4
Figure 4. Epithelial cell death, myofibroblasts, and macrophage cell expansion are all attenuated by rhPTX-2.
(A) Images of apoptotic tubular epithelial cells (TUNEL+), macrophages (F4/80+), myeloid cells (CD11b+), and myofibroblasts (α-SMA+) in kidney at 9 weeks. Insets show a glomerulus labeled for CD11b expression. (B) Tubular apoptosis, (C) macrophages, (D) myeloid cells, and (E) myofibroblasts. (F) CD11b+ cells per glomerular tuft. (G and H) Q-PCR measuring Acta2 and Col1a1 transcripts in whole kidney tissue. (I) Quantification of peritubular capillary density. Scale bars: 50 μm. n = 6–12/group. *P < 0.05, **P < 0.01 (ANOVA with post hoc testing for multiple comparisons). Avg, average; VEH, vehicle.
Figure 5
Figure 5. rhPTX-2 reduces the population of M1-type activated macrophages in Col4a3–/– mice.
(A) Images of kidney M1 (CD86+) and M2 (CD206/mannose receptor+) macrophages at 9 weeks. G, glomerulus. (B and D) Numbers and percentages of CD86+ or CD206+ macrophages in tissue sections. (C and EI) Q-PCR measuring M1-type (CD86, Trem1, Tnfa) and M2-like (CD206, Arg1) markers, and Il10 in whole kidney. Scale bars: 50 μm. n = 6–12/group. *P < 0.05, **P < 0.01 (ANOVA with post hoc testing for multiple comparisons). VEH, vehicle.
Figure 6
Figure 6. Gene set enrichment analysis identifies attenuated c-Jun signaling as a direct and central consequence of PTX-2 treatment.
(A) Principal component analysis of kidney transcriptome. Treatment of Col4a3–/– mice with rhPTX-2 partly reverses separation from the uninjured Col4a3+/+ animals, indicating rhPTX-2 therapy reduced transcriptional perturbation. (B) Summary of pathway analysis (GSEA) depicted as heatmaps of individual leading-edge genes (rows) and individual mice (columns). Enriched gene sets fell into 4 broad functional categories induced by disease: mitochondrion/oxidoreductase activity, metabolism, inflammation/immunity, and tissue remodeling. Within each category, PTX-2 treatment of Col4a3–/– mice partly restored disease-induced gene expression changes to their unperturbed baseline. Note only AP-1 signaling (blue box) was suppressed below baseline levels by rhPTX-2. (C) Abbreviated protein-protein interaction network predicted by STRING analysis of the top 100 leading-edge genes identified by GSEA and downregulated by rhPTX-2 treatment. Line color denotes type of evidence: green: neighborhood, red: gene fusion, blue: co-occurrence, black: coexpression, purple: experiments, cyan: databases, yellow: text mining, and lilac: homology. (D) Western blots showing the effect of disease and PTX-2 treatment on p–S63-c-Jun, total c-Jun, and C/EBP in kidney tissue. VEH, vehicle.
Figure 7
Figure 7. rhPTX-2 directly attenuates c-Jun and AP-1 complex signaling in human monocytes.
(A and B) Time-course Western blots (A) and densitometry quantification (B) showing c-Jun phosphorylation (p–S63-c-Jun) and total c-Jun levels from 15 to 60 minutes after rhPTX-2 and/or TNF-α stimulation in human iPSC-derived monocytes, normalized to GAPDH. Note that rhPTX-2 attenuates both total and p–c-Jun in response to TNF-α. Data represent 3 experiments. (C) Effect of concentrations of PTX-2 or AP-1 inhibitor SR11302 on TNF-α–induced AP1/NF-κB promoter activity in human monocytes. (D) Effect of concentrations of PTX-2 on TNF-α–induced AP-1 promoter activity alone, in human monocytes. (E) rhPTX-2–induced changes in the secretion of factors associated with fibrosis by primary human monocytes cultured from 12 to 120 hours. Presence of c-Jun and AP-1 binding sites in the promoters of these genes is indicated, based on the GeneCards database. n = 3–8/group (*P < 0.05, **P < 0.01, ***P < 0.001 (ANOVA with post hoc testing for multiple comparisons). VEH, vehicle; AlkPhos, alkaline phosphatase.
Figure 8
Figure 8. rhPTX-2 binds to proximal tubule epithelial cell membrane, inhibits c-Jun signaling, and protects against cellular stress.
(A) 3D reconstructed confocal images of representative individual human PTECs stained for rhPTX-2 (red) and clathrin, LAMP-2, or caveolin (green) showing x, y, and z axes. The left panels indicate the surface distribution of hPTX-2 following rhPTX-2 incubation for 30 minutes on ice (arrowheads). The right panels show the distribution of internalized rhPTX-2 following incubation 30 minutes at 37°C. (B and C) Time-course Western blots showing c-Jun phosphorylation (P-S63 c-Jun) and total c-Jun levels from 15 to 60 minutes after rhPTX-2 and/or plasma shock in human fetal kidney proximal tubule epithelial cells, normalized to GAPDH. Data represent 3 experiments. (D) Images and graph quantifying nuclear p–c-JUN immunofluorescence staining intensity in human PTECs in response to plasma and PTX-2. (E) Images and quantification of E-cadherin and vimentin expression by human PTECs in resting culture conditions or in response to human plasma or TGF-β1 for 24 hours. Note that 1 hour pretreatment of rhPTX-2 markedly blocked these responses. (F) Quantification of mitochondrial dysfunction by MitoSOX (left) and apoptosis by activated caspase-3 (right) detection 24 hours after human PTECs were challenged with human plasma or TGF-β1, with or without 1-hour rhPTX-2 pretreatment. (G) Inhibitory effect of rhPTX-2 on factors secreted by human PTECs in response to TGF-β1 stimulation. Fold change versus vehicle-treated PTECs and percent inhibition by rhPTX-2 are shown, and presence of c-Jun binding sites in promoters are noted. Scale bars: 25 μm. n = 3–6/group. *P < 0.05, **P < 0.01 (ANOVA with post hoc testing for multiple comparisons). VEH, vehicle.

References

    1. Pilling D, Buckley CD, Salmon M, Gomer RH. Inhibition of fibrocyte differentiation by serum amyloid P. J Immunol. 2003;171(10):5537–5546. doi: 10.4049/jimmunol.171.10.5537. - DOI - PMC - PubMed
    1. Castaño AP, et al. Serum amyloid P inhibits fibrosis through Fc gamma R-dependent monocyte-macrophage regulation in vivo. Sci Transl Med. 2009;1(5):5ra13. - PMC - PubMed
    1. Duffield JS, Lupher ML. PRM-151 (recombinant human serum amyloid P/pentraxin 2) for the treatment of fibrosis. Drug News Perspect. 2010;23(5):305–315. doi: 10.1358/dnp.2010.23.5.1444206. - DOI - PubMed
    1. Botto M, et al. Amyloid deposition is delayed in mice with targeted deletion of the serum amyloid P component gene. Nat Med. 1997;3(8):855–859. doi: 10.1038/nm0897-855. - DOI - PubMed
    1. Bickerstaff MC, et al. Serum amyloid P component controls chromatin degradation and prevents antinuclear autoimmunity. Nat Med. 1999;5(6):694–697. doi: 10.1038/9544. - DOI - PubMed

Publication types