Epidermal growth factor receptor promotes glomerular injury and renal failure in rapidly progressive crescentic glomerulonephritis

Guillaume Bollée, Martin Flamant, Sandra Schordan, Cécile Fligny, Elisabeth Rumpel, Marine Milon, Eric Schordan, Nathalie Sabaa, Sophie Vandermeersch, Ariane Galaup, Anita Rodenas, Ibrahim Casal, Susan W Sunnarborg, David J Salant, Jeffrey B Kopp, David W Threadgill, Susan E Quaggin, Jean-Claude Dussaule, Stéphane Germain, Laurent Mesnard, Karlhans Endlich, Claude Boucheix, Xavier Belenfant, Patrice Callard, Nicole Endlich, Pierre-Louis Tharaux, Guillaume Bollée, Martin Flamant, Sandra Schordan, Cécile Fligny, Elisabeth Rumpel, Marine Milon, Eric Schordan, Nathalie Sabaa, Sophie Vandermeersch, Ariane Galaup, Anita Rodenas, Ibrahim Casal, Susan W Sunnarborg, David J Salant, Jeffrey B Kopp, David W Threadgill, Susan E Quaggin, Jean-Claude Dussaule, Stéphane Germain, Laurent Mesnard, Karlhans Endlich, Claude Boucheix, Xavier Belenfant, Patrice Callard, Nicole Endlich, Pierre-Louis Tharaux

Abstract

Rapidly progressive glomerulonephritis (RPGN) is a life-threatening clinical syndrome and a morphological manifestation of severe glomerular injury that is marked by a proliferative histological pattern ('crescents') with accumulation of T cells and macrophages and proliferation of intrinsic glomerular cells. We show de novo induction of heparin-binding epidermal growth factor-like growth factor (HB-EGF) in intrinsic glomerular epithelial cells (podocytes) from both mice and humans with RPGN. HB-EGF induction increases phosphorylation of the epidermal growth factor receptor (EGFR, also known as ErbB1) in mice with RPGN. In HB-EGF-deficient mice, EGFR activation in glomeruli is absent and the course of RPGN is improved. Autocrine HB-EGF induces a phenotypic switch in podocytes in vitro. Conditional deletion of the Egfr gene from podocytes of mice alleviates the severity of RPGN. Likewise, pharmacological blockade of EGFR also improves the course of RPGN, even when started 4 d after the induction of experimental RPGN. This suggests that targeting the HB-EGF-EGFR pathway could also be beneficial in treatment of human RPGN.

Figures

Figure 1. Induction of renal HB-EGF synthesis…
Figure 1. Induction of renal HB-EGF synthesis leads to glomerular activation of EGFR during RPGN
(a) Representative image of in situ hybridization study in NTS-injected wild-type (Hbegf (+/+)) animals, showing proHB-EGF expression in glomeruli (G), especially in parietal glomerular epithelial cells (Pec), in podocytes (day 4) and in crescents (Cr) (day 8). White arrow indicates abundant proHB-EGF mRNA expression in areas where tuft-capsular podocyte bridges were present. Scale bar (orange), 50 μm. (b) Quantification by real-time RT-PCR of proHB-EGF mRNA in freshly isolated podocytes on day 6 after nephrotoxic serum injection (NTS) and in podocytes from non-injected control mice (CT) (n=3 per group). * P<0.05 versus controls. (c) Western blot analysis of phosphorylated EGFR and total EGFR in the renal cortex from non-challenged controls (CT), wild-type (Hbegf (+/+)) mice infused with NTS ((+/+)NTS), HB-EGF deficient mice infused with NTS ((−/−)NTS), and from Hbegf (+/+) animals that were given intraperitoneal injections of the EGFR tyrosine kinase inhibitor AG1478 ((+/+)NTS+AG1478). Values reported are means ± sem. (n=6–8 per group). (d) Immunofluorescence staining for phosphoEGFR in renal cortex from controls (CT), NTS-injected Hbegf (+/+) mice ((+/+)NTS), HB-EGF deficient mice infused with NTS ((−/−)NTS), and from NTS-injected Hbegf (+/+) animals treated by AG1478 ((+/+)NTS+AG1478), on day 8 after NTS administration. * P<0.05 versus controls at baseline, ** P<0.01 versus controls at baseline. ## P<0.01 versus mice treated with vehicle only. Pec: parietal glomerular epithelial cells; G: glomerulus; Cr: crescent.
Figure 2. HB-EGF induces a migratory phenotype…
Figure 2. HB-EGF induces a migratory phenotype in podocytes in vitro
(a) Podocyte outgrowth over 6 days from decapsulated glomeruli of Hbegf (+/+) or Hbegf (−/−) mice (arrow). Cells are stained for WT-1 expression. (b) Outgrowth area from glomeruli of Hbegf (−/−) mice in the absence (light blue bar) or presence of the EGFR inhibitor AG1478 (500 nM) (AG, dark blue bar) and from glomeruli. Sparse outgrowth from glomeruli of Hbegf (−/−) mice in the absence (light grey bar) was rescued by addition of 30 nM HB-EGF (black bar). (c) Schematic drawing of podocyte outgrowth from isolated glomeruli, which was used as a combined migration/proliferation assay to assess the ability of crescent formation in vitro. Podocytes are in a stationary state (blue color) on the surface of capillary loops (grey circle), when glomeruli are plated. Subsequently, podocytes assume a migratory phenotye (orange color), characterized by apical protrusions, by attachment and by migration on the substratum. Later stages of outgrowth also involve proliferation. (d) Representative image of F-actin reorganisation and formation of ring-like actin structures (RiLiS) induced by HB-EGF (30 nM for 7 min) in differentiated podocytes, in the absence or presence of AG1478 (500 nM). The effect of HB-EGF to induce apical protrusions is abrogated in the presence of AG1478 (500 nM). (e) Quantitative analysis of RiLiS formation in differentiated podocytes. HB-EGF (30 nM) was added in the absence (Ctl) or presence of inhibitors: AG - AG1478 (500 nM), Wo - Wortmannin (100 nM), LY - LY294002 (30 μM), PD - PD98059 (25 μM), SB - SB203580 (25 μM). (f) BrdU incorporation in differentiated podocytes over 48 h. (g) Distance of migration of differentiated podocytes within 8 h in the wound assay. Data are means ± SEM (n=3–4 experiments). * P<0.05 vs. untreated Hbegf (+/+) glomeruli in (b) and vs. HB-EGF alone in (e–g). Scale bar : 300 μm in (a) and 30 μm in (d).
Figure 3. Deletion of Hbegf gene prevents…
Figure 3. Deletion of Hbegf gene prevents fatal renal destruction
(a) Survival curve for challenged Hbegf (+/+) and Hbegf (−/−) mice. In all cases, death was associated with severe renal damage with macroscopic hematuria (blood leakage into the urine) and animals died from renal failure (with 100% crescentic and necrotizing lesions at autopsy). (b) Masson Trichrome staining of kidneys and proportion of crescentic glomeruli in control mice and in NTS-injected Hbegf (+/+) and Hbegf (−/−) mice (day 8 post NTS) (Cr: crescents, G: glomeruli, Tc: tubules with proteinaceous casts) Scale bar, 50μm. (c) Ascites score as an index of albumin plasma loss and water and sodium retention, (d) albuminuria and (e) blood urea nitrogen concentrations in NTS-challenged Hbegf (+/+) and Hbegf (−/−) animals on day 8 post NTS, and in unchallenged controls (CT). Values reported are means ± sem. (n=9–12 per group). * P<0.05 versus controls at baseline, ** P<0.01 versus baseline, *** P<0.001 versus baseline. # P<0.05 versus NTS-treated (+/+), ## P<0.01 versus NTS-treated (+/+).
Figure 4. Selective deletion of Egfr from…
Figure 4. Selective deletion of Egfr from podocytes protects from RPGN
(a) Albuminuria (b) blood urea nitrogen concentration and (c) proportion of crescentic glomeruli in Pod-Tet on-Cre Egfrwt/wt and Pod-Tet on-Cre EgfrloxP/loxP mice, 8 days after NTS-induced RPGN (P<0.05 for all comparisons). (d) Survival curve of challenged Pod-Tet on-Cre EgfrloxP/loxP and littermate control mice in a severe model of RPGN. (* P<0.01). (e) Ultrastructural analysis of podocytes by transmission electron microscopy in NTS-treated Pod-Tet on-Cre Egfrwt/wt and Pod-Tet on-Cre EgfrloxP/loxP mice. More severe foot process effacement and irregular thickening of the GBM is visible in Pod-Tet on-Cre Egfrwt/wt animals. Scale bars: upper panels 2μm, lower panels 1μm.
Figure 5. Delayed EGFR tyrosine kinase inhibition…
Figure 5. Delayed EGFR tyrosine kinase inhibition stops the development of crescentic RPGN
(a) Quantification by western blot analysis of phosphorylated EGFR and total EGFR in the renal cortex from non-challenged controls (CT), NTS-injected mice treated with vehicle alone, and NTS-injected mice treated with erlotinib either started twelve hours before administration of NTS (days 0–14) or in a curative protocol, started four days later (d4–14). Mice were euthanised after 14 days of RPGN. (b) Blood urea nitrogen concentration and (c) proportion of crescentic glomeruli in CT in the different groups of mice as in (a). Data are means ± sem, (n=9 mice per group). ** P<0.01 versus controls at baseline (CT), *** P<0.001 versus CT, ## P<0.01 versus vehicle, ## P<0.001 versus vehicle. (d) Ultrastructural analysis of podocytes by transmission electron microscopy in erlotinib-treated and vehicle-treated mice, five days after injection of NTS. Scale bar 2 μm. (e) Masson trichrome staining of renal cortex from a mouse treated with erlotinib (day 4–14) (left panel) and a vehicle-treated mouse (right panel) on day 14. Ne: necrotic glomerular lesions, Cr: cellular crescents, Tc: tubular proteinaceous casts, Infilt: diffuse CD3-positive cell infiltrates (Infilt) seen in vehicle-treated mice. Scale bar 100 μm.
Figure 6. HB-EGF expression is induced in…
Figure 6. HB-EGF expression is induced in human crescentic glomerulonephritis
Representative images of immunostaining for HB-EGF using monoclonal sc-74526 antibody in sections of kidney biopsies from 8 random subjects diagnosed with non crescentic glomerulopathies (upper panels), including diabetic nephropathy, amyloidosis, minimal change disease (MCD), focal segmental glomerulosclerosis (FSGS), IgA nephropathy (IgAN), and membranous nephropathy (MN). Similarly, lower panels show immunostaining for HB-EGF in renal biopsies from 8 random subjects RPGN of various etiologies, including lupus nephritis, microscopic polyangiitis (MP), endocarditis (End), Goodpasture disease (Gp), and Wegener disease (Wg). Scale bar 50 μm.

References

    1. Jennette JC, Thomas DB. Crescentic glomerulonephritis. Nephrol Dial Transplant. 2001;16(Suppl 6):80–82.
    1. Feng L, et al. Heparin-binding EGF-like growth factor contributes to reduced glomerular filtration rate during glomerulonephritis in rats. J Clin Invest. 2000;105:341–350.
    1. Yoshizumi M, et al. Tumor necrosis factor increases transcription of the heparin-binding epidermal growth factor-like growth factor gene in vascular endothelial cells. J Biol Chem. 1992;267:9467–9469.
    1. Kume N, Gimbrone MA., Jr. Lysophosphatidylcholine transcriptionally induces growth factor gene expression in cultured human endothelial cells. J Clin Invest. 1994;93:907–911.
    1. Marikovsky M, et al. Appearance of heparin-binding EGF-like growth factor in wound fluid as a response to injury. Proc Natl Acad Sci U S A. 1993;90:3889–3893.
    1. Higashiyama S, Abraham JA, Miller J, Fiddes JC, Klagsbrun M. A heparin-binding growth factor secreted by macrophage-like cells that is related to EGF. Science. 1991;251:936–939.
    1. Blotnick S, Peoples GE, Freeman MR, Eberlein TJ, Klagsbrun M. T lymphocytes synthesize and export heparin-binding epidermal growth factor-like growth factor and basic fibroblast growth factor, mitogens for vascular cells and fibroblasts: differential production and release by CD4+ and CD8+ T cells. Proc Natl Acad Sci U S A. 1994;91:2890–2894.
    1. Neale TJ, Tipping PG, Carson SD, Holdsworth SR. Participation of cell-mediated immunity in deposition of fibrin in glomerulonephritis. Lancet. 1988;2:421–424.
    1. Tipping PG, Holdsworth SR. T cells in glomerulonephritis. Springer Semin Immunopathol. 2003;24:377–393.
    1. Segerer S, et al. Expression of the chemokine monocyte chemoattractant protein-1 and its receptor chemokine receptor 2 in human crescentic glomerulonephritis. J Am Soc Nephrol. 2000;11:2231–2242.
    1. Lloyd CM, et al. RANTES and monocyte chemoattractant protein-1 (MCP-1) play an important role in the inflammatory phase of crescentic nephritis, but only MCP-1 is involved in crescent formation and interstitial fibrosis. J Exp Med. 1997;185:1371–1380.
    1. Besse-Eschmann V, Le Hir M, Endlich N, Endlich K. Alteration of podocytes in a murine model of crescentic glomerulonephritis. Histochem Cell Biol. 2004;122:139–149.
    1. Moeller MJ, et al. Podocytes populate cellular crescents in a murine model of inflammatory glomerulonephritis. J Am Soc Nephrol. 2004;15:61–67.
    1. Le Hir M, et al. Podocyte bridges between the tuft and Bowman's capsule: an early event in experimental crescentic glomerulonephritis. J Am Soc Nephrol. 2001;12:2060–2071.
    1. Thorner PS, Ho M, Eremina V, Sado Y, Quaggin S. Podocytes contribute to the formation of glomerular crescents. J Am Soc Nephrol. 2008;19:495–502.
    1. Bariety J, Bruneval P. Activated parietal epithelial cells or dedifferentiated podocytes in FSGS: can we make the difference? Kidney Int. 2006;69:194.
    1. Schiwek D, et al. Stable expression of nephrin and localization to cell-cell contacts in novel murine podocyte cell lines. Kidney Int. 2004;66:91–101.
    1. Simpson KJ, et al. Identification of genes that regulate epithelial cell migration using an siRNA screening approach. Nat Cell Biol. 2008
    1. Hancock WW, Atkins RC. Cellular composition of crescents in human rapidly progressive glomerulonephritis identified using monoclonal antibodies. Am J Nephrol. 1984;4:177–181.
    1. Lin F, et al. Respective roles of decay-accelerating factor and CD59 in circumventing glomerular injury in acute nephrotoxic serum nephritis. J Immunol. 2004;172:2636–2642.
    1. Peoples GE, et al. T lymphocytes that infiltrate tumors and atherosclerotic plaques produce heparin-binding epidermal growth factor-like growth factor and basic fibroblast growth factor: a potential pathologic role. Proc Natl Acad Sci U S A. 1995;92:6547–6551.
    1. Lee D, et al. Epiregulin is not essential for development of intestinal tumors but is required for protection from intestinal damage. Mol Cell Biol. 2004;24:8907–8916.
    1. Osherov N, Levitzki A. Epidermal-growth-factor-dependent activation of the src-family kinases. Eur J Biochem. 1994;225:1047–1053.
    1. Faul C, Asanuma K, Yanagida-Asanuma E, Kim K, Mundel P. Actin up: regulation of podocyte structure and function by components of the actin cytoskeleton. Trends Cell Biol. 2007;17:428–437.
    1. Lee YJ, Shin SJ, Lin SR, Tan MS, Tsai JH. Increased expression of heparin binding epidermal growth-factor-like growth factor mRNA in the kidney of streptozotocin-induced diabetic rats. Biochem Biophys Res Commun. 1995;207:216–222.
    1. Paizis K, et al. Heparin-binding epidermal growth factor-like growth factor in experimental models of membranous and minimal change nephropathy. Kidney Int. 1998;53:1162–1171.
    1. Khong TF, et al. Inhibition of heparin-binding epidermal growth factor-like growth factor increases albuminuria in puromycin aminonucleoside nephrosis. Kidney Int. 2000;58:1098–1107.
    1. Levy JB, Turner AN, Rees AJ, Pusey CD. Long-term outcome of anti-glomerular basement membrane antibody disease treated with plasma exchange and immunosuppression. Ann Intern Med. 2001;134:1033–1042.
    1. Salama AD, Levy JB, Lightstone L, Pusey CD. Goodpasture's disease. Lancet. 2001;358:917–920.
    1. Topham PS, et al. Lack of chemokine receptor CCR1 enhances Th1 responses and glomerular injury during nephrotoxic nephritis. J Clin Invest. 1999;104:1549–1557.
    1. Ng YY, et al. Glomerular epithelial-myofibroblast transdifferentiation in the evolution of glomerular crescent formation. Nephrol Dial Transplant. 1999;14:2860–2872.
    1. Nikolic-Paterson DJ, Atkins RC. The role of macrophages in glomerulonephritis. Nephrol Dial Transplant. 2001;16(Suppl 5):3–7.
    1. Boucher A, Droz D, Adafer E, Noel LH. Relationship between the integrity of Bowman's capsule and the composition of cellular crescents in human crescentic glomerulonephritis. Lab Invest. 1987;56:526–533.
    1. Kalluri R, Danoff TM, Okada H, Neilson EG. Susceptibility to anti-glomerular basement membrane disease and Goodpasture syndrome is linked to MHC class II genes and the emergence of T cell-mediated immunity in mice. J Clin Invest. 1997;100:2263–2275.
    1. Guettier C, et al. Immunohistochemical demonstration of parietal epithelial cells and macrophages in human proliferative extra-capillary lesions. Virchows Arch A Pathol Anat Histopathol. 1986;409:739–748.
    1. Levy JB, Pusey CD. Anti-glomerular basement membrane disease. In: Warrell DA, Cox TM, Firth JD, editors. Oxford Textbook of Medicine. 4th Edition Oxford University Press; Oxford: 2003.
    1. Polihronis M, Murphy BF, Pearse MJ, Power DA. Heparin-binding epidermal growth factor-like growth factor, an immediate-early gene for mesangial cells, is up-regulated in the Thy-1.1 model. Exp Nephrol. 1996;4:271–278.
    1. Takemura T, et al. Heparin-binding EGF-like growth factor is expressed by mesangial cells and is involved in mesangial proliferation in glomerulonephritis. J Pathol. 1999;189:431–438.
    1. Triantafyllopoulou A, et al. Proliferative lesions and metalloproteinase activity in murine lupus nephritis mediated by type I interferons and macrophages. Proc Natl Acad Sci U S A. 2010;107:3012–3017.
    1. Chugh S, et al. Aminopeptidase A: a nephritogenic target antigen of nephrotoxic serum. Kidney Int. 2001;59:601–613.
    1. Englert C, et al. WT1 suppresses synthesis of the epidermal growth factor receptor and induces apoptosis. EMBO J. 1995;14:4662–4675.
    1. Elenius K, Paul S, Allison G, Sun J, Klagsbrun M. Activation of HER4 by heparin-binding EGF-like growth factor stimulates chemotaxis but not proliferation. Embo J. 1997;16:1268–1278.
    1. Mishra R, Leahy P, Simonson MS. Gene expression profiling reveals role for EGF-family ligands in mesangial cell proliferation. Am J Physiol Renal Physiol. 2002;283:F1151–1159.
    1. Kim HS, et al. Identification of novel Wilms' tumor suppressor gene target genes implicated in kidney development. J Biol Chem. 2007;282:16278–16287.
    1. Wassef L, Kelly DJ, Gilbert RE. Epidermal growth factor receptor inhibition attenuates early kidney enlargement in experimental diabetes. Kidney Int. 2004;66:1805–1814.
    1. Advani A, et al. Inhibition of the Epidermal Growth Factor Receptor Preserves Podocytes and Attenuates Albuminuria in Experimental Diabetic Nephropathy. Nephrology (Carlton) 2011
    1. Paizis K, et al. Heparin-binding epidermal growth factor-like growth factor is expressed in the adhesive lesions of experimental focal glomerular sclerosis. Kidney Int. 1999;55:2310–2321.
    1. Jackson LF, et al. Defective valvulogenesis in HB-EGF and TACE-null mice is associated with aberrant BMP signaling. Embo J. 2003;22:2704–2716.
    1. Luetteke NC, et al. TGF alpha deficiency results in hair follicle and eye abnormalities in targeted and waved-1 mice. Cell. 1993;73:263–278.
    1. Mann GB, et al. Mice with a null mutation of the TGF alpha gene have abnormal skin architecture, wavy hair, and curly whiskers and often develop corneal inflammation. Cell. 1993;73:249–261.
    1. Moeller MJ, Sanden SK, Soofi A, Wiggins RC, Holzman LB. Podocyte-specific expression of cre recombinase in transgenic mice. Genesis. 2003;35:39–42.
    1. Novak A, Guo C, Yang W, Nagy A, Lobe CG. Z/EG, a double reporter mouse line that expresses enhanced green fluorescent protein upon Cre-mediated excision. Genesis. 2000;28:147–155.
    1. Shigehara T, et al. Inducible podocyte-specific gene expression in transgenic mice. J Am Soc Nephrol. 2003;14:1998–2003.
    1. Eremina V, et al. VEGF inhibition and renal thrombotic microangiopathy. N Engl J Med. 2008;358:1129–1136.
    1. Lee TC, Threadgill DW. Generation and validation of mice carrying a conditional allele of the epidermal growth factor receptor. Genesis. 2009;47:85–92.
    1. Le Jan S, et al. Angiopoietin-like 4 is a proangiogenic factor produced during ischemia and in conventional renal cell carcinoma. Am J Pathol. 2003;162:1521–1528.
    1. Reiter JL, et al. Comparative genomic sequence analysis and isolation of human and mouse alternative EGFR transcripts encoding truncated receptor isoforms. Genomics. 2001;71:1–20.
    1. Garrett TP, et al. Crystal structure of a truncated epidermal growth factor receptor extracellular domain bound to transforming growth factor alpha. Cell. 2002;110:763–773.

Source: PubMed

3
Předplatit