Circulating urokinase receptor as a cause of focal segmental glomerulosclerosis

Journal name:
Nature Medicine
Volume:
17,
Pages:
952–960
Year published:
DOI:
doi:10.1038/nm.2411
Received
Accepted
Published online

Abstract

Focal segmental glomerulosclerosis (FSGS) is a cause of proteinuric kidney disease, compromising both native and transplanted kidneys. Treatment is limited because of a complex pathogenesis, including unknown serum factors. Here we report that serum soluble urokinase receptor (suPAR) is elevated in two-thirds of subjects with primary FSGS, but not in people with other glomerular diseases. We further find that a higher concentration of suPAR before transplantation underlies an increased risk for recurrence of FSGS after transplantation. Using three mouse models, we explore the effects of suPAR on kidney function and morphology. We show that circulating suPAR activates podocyte β3 integrin in both native and grafted kidneys, causing foot process effacement, proteinuria and FSGS-like glomerulopathy. Our findings suggest that the renal disease only develops when suPAR sufficiently activates podocyte β3 integrin. Thus, the disease can be abrogated by lowering serum suPAR concentrations through plasmapheresis, or by interfering with the suPAR–β3 integrin interaction through antibodies and small molecules targeting either uPAR or β3 integrin. Our study identifies serum suPAR as a circulating factor that may cause FSGS.

At a glance

Figures

  1. suPAR measurement in the serum of subjects with glomerular disease.
    Figure 1: suPAR measurement in the serum of subjects with glomerular disease.

    For transplant subjects, the suPAR values were measured from pretransplantation serum unless otherwise indicated. Data is presented as means ± s.e.m. (a) Serum suPAR concentration in subjects with glomerular disease and healthy human subjects. MN, membranous nephropathy. *P < 0.05 for FSGS versus MN and preeclampsia; #P < 0.001 for FSGS versus healthy, MCD relapse, and MCD remission. Note that the values highlighted with red or green dots in the healthy subject and FSGS columns are identical twin pairs; in each case, one is healthy and has a twin brother with FSGS. (b) Serum suPAR in different population of subjects with primary FSGS. **P < 0.01 for recurrent FSGS versus nonrecurrent FSGS and nontransplant primary FSGS, respectively. (c) Serum suPAR concentrations after transplantation. #P < 0.001. (d) Correlation analysis of pretransplantation suPAR with proteinuria after transplantation. Pearson r = 0.16, P = 0.50. (e) Correlation analysis of pretransplantation suPAR with eGFR. Pearson r = 0.36, P = 0.16. (f) Correlation analysis of suPAR after transplantation with eGFR. Pearson r = 0.10, P = 0.58.

  2. suPAR binds to and activates [beta]3 integrin on podocytes.
    Figure 2: suPAR binds to and activates β3 integrin on podocytes.

    (a) Western blot showing that suPAR binds β3 integrin (representative of three experiments). EIF1B-GFP, encoding a translation initiation factor, and Raver-Flag encoding a ribonucleoprotein served as negative binding controls. β3 integrin is encoded by Itgb3. S, sPlaurWT (encoding suPAR); M, PlaurWT (encoding membrane-bound uPAR); IP, immunoprecipitation. (b) AP5 immunostaining of differentiated human podocytes incubated with suPAR-rich recurrent FSGS serum (rec-FSGS serum), co-treated with the monoclonal antibody to human uPAR (uPAR mAb), and with cycloRGDfv, a small molecule that blocks β3 integrin activity. AP5-specific antibody detects the active form of β3 integrin. Bovine serum, negative control; suPAR, recombinant human suPAR protein. (c) Immunohistochemistry of AP5 on kidney biopsies from patients with glomerular disease. Top, representative AP5 staining in the glomerulus of subjects with FSGS. Bottom, the percentage (mean ± s.e.m.) of AP5-positive glomeruli. *P < 0.05 for primary FSGS versus control; **P < 0.01 for recurrent FSGS versus control. (d) Double immunofluorescent staining in glomeruli of kidney grafts for AP5 (green) and the podocyte marker synaptopodin (red). Top and bottom left, AP5 in the graft glomerulus 2 h after reperfusion in recurrent and nonrecurrent transplant biopsies (n = 2 per group). Top right, AP5 signal in recurrent transplant biopsies (n = 3) and nonrecurrent grafts (n = 5). Bottom right, normal kidney sections (n = 2) and biopsies from acute T cell–mediated rejections (n = 3) served as controls. Scale bars, 30 μm.

  3. suPAR serum concentrations and podocyte [beta]3 integrin activity determine treatment response to plasmapheresis in recurrent FSGS.
    Figure 3: suPAR serum concentrations and podocyte β3 integrin activity determine treatment response to plasmapheresis in recurrent FSGS.

    (a) Human podocytes incubated with different pooled serum samples and assayed for β3 integrin activity. MFI, mean fluorescence intensity. *P < 0.05 for nonrecurrent FSGS versus normal subjects, ***P < 0.001 for recurrent versus nonrecurrent FSGS or versus healthy subjects. The respective suPAR concentration of the pooled sera is marked in red. NS, normal (healthy) subject; NR, nonrecurrent FSGS; REC, recurrent FSGS (representative of three experiments). (b) Pharmacological modulation of β3 integrin activity in podocytes. **P < 0.01 for cylcoRGDfv co-treated cells versus recurrent FSGS serum alone; ***P < 0.001 for uPAR-specific mAb co-treated cells versus recurrent FSGS serum alone. (c) suPAR in serum from subjects with recurrent FSGS (n = 4) before and after a course of plasmapheresis. **P < 0.01. (d) Effect of plasmapheresis on β3 integrin activity in podocytes incubated with recurrent FSGS serum (n = 6), collected before and after serial treatment with plasmapheresis. ***P < 0.001. (eh) Clinical cases of recurrent FSGS. Top graphs show serum suPAR, urine protein/creatinine ratio (g/g) and individual plasmapheresis treatment as indicated by arrows and plotted over time (d) from before (−1) to after transplantation. Bottom graphs and images show podocyte β3 integrin activity measured by FACS (left) and immunofluorescence (right) as a result of incubation with pretransplantation serum, or with the after-transplantation serum collected after repetitive plasmapheresis treatments. As a reference, the mean concentration of AP5 from a is marked as a dashed line. (e,f) Patients who obtained full remission after pheresis. (g,h) Patients who did not achieve remission after pheresis. Scale bars, 30 μm. Whiskers in plots of AP5 activity and serum suPAR show minimum to maximum.

  4. suPAR activates [beta]3 integrin and causes foot process effacement in Plaur-/- mouse kidneys and albuminuria in Plaur-/- mice.
    Figure 4: suPAR activates β3 integrin and causes foot process effacement in Plaur−/− mouse kidneys and albuminuria in Plaur−/− mice.

    (a) Injection (i.v.) of high doses of recombinant mouse suPAR into Plaur−/− mice (n = 4 per group) induces proteinuria. **P < 0.01 for mice injected with 20 μg of suPAR at 24 h versus mice injected with other doses or versus other time points. (b) Injection (i.v.) of high doses of recombinant suPAR deposits into podocytes. Green, uPAR; red, synaptopodin (Synpo). (c) AP5 activity induced in the podocytes of high-dosage suPAR-injected Plaur−/− mice (n = 4). Green, AP5; red, Synpo. (d,e) LPS induced endogenous suPAR in wild-type mice (n = 6). (d) Serum suPAR concentrations in LPS-treated mice. ***P < 0.001 for LPS-injected mice at 24 h versus PBS control, and versus LPS-injected mice at 0 h. **P < 0.01 for LPS-injected mice at 48 h versus at 0 h. (e) Urinary suPAR concentrations. ***P < 0.001 for LPS-injected mice at 48 h versus 0 h, and versus PBS control at any time point. **P < 0.01 for LPS-injected mice at 24 h versus 0 h. (f) Generation of a hybrid-kidney mouse model. (g) Electron microscope analysis of the PBS (n = 3) or LPS (n = 5) treated hybrid kidney. (h) uPAR expression in the native or Plaur−/− kidneys from the hybrid-kidney mice with or without LPS treatment. Scale bars, 30 μm in b,c and h; 250 nm in g. Error bars, means ± s.e.m. in a; means ± s.d. in d,e.

  5. Sustained overexpression of suPAR in the blood of wild-type mice leads to an FSGS-like glomerulopathy.
    Figure 5: Sustained overexpression of suPAR in the blood of wild-type mice leads to an FSGS-like glomerulopathy.

    (a) Generation of β3 integrin binding–deficient suPAR mutants. (b) Serum suPAR concentrations in the sPlaurWT engineered mice. *P < 0.05 at day 7 versus day 0 (before initial electroporation) (c) Urinary suPAR in sPlaurWT engineered mice. ***P < 0.001 for days 7, 14 and 28 versus day 0; *P < 0.05 for day 28 versus day 7. (n = 4 in each group). (d) Albuminuria in sPlaurWT and sPlaurE134A mice. *P < 0.05 for sPlaurWT mice at day 7 versus before treatment or versus sPlaurE134A mice at day 7. **P < 0.01 for sPlaurWT engineered mice at day 14 versus before treatment or versus sPlaurE134A treated mice at day 7 or 14. (e) Kidney EM analysis of sPlaur engineered mice. Podocyte damage is reflected by relating the length of effaced foot process (FP) to the total length of the glomerular basement membrane (GBM) analyzed. Scale bars, 1 μm for upper image, 250 nm for lower image. **P < 0.01. (f) Histochemistry and light microscopy of the kidney from sPlaur engineered mice. PAS, periodic acid–Schiff. Scale bars, 30 μm. (g) Histopathological alteration of the kidneys was semiquantitatively scored. *P < 0.05. Error bars, means ± s.e.m. in d; means ± s.d. in b,c,e and g.

  6. Administration of blocking antibody to uPAR ameliorates suPAR-caused kidney damage.
    Figure 6: Administration of blocking antibody to uPAR ameliorates suPAR-caused kidney damage.

    (n = 4 in each group). (a) Proteinuria in the antibody treated sPlaurWT mice. *P < 0.05 for sPlaurWT mice receiving isotype control at day 7 versus before initial electroporation at day 0 or versus mice treated with antibody to uPAR at day 7; ***P < 0.01 for sPlaurWT mice receiving isotype control at day 21 versus at day 0 or versus antibody to uPAR-treated mice at day 21. (b) Morphological examination of the antibody treated sPlaurWT kidney. Scale bars, 30 μm. (c) Pathology score. *P < 0.05. (d) Electron microscopic analysis of the antibody treated kidney from sPlaurWT engineered mice. **P < 0.01 for IgG isotype control versus uPAR-specific antibody–treated sPlaurWT engineered mice with respect to the ratio of effaced foot process (FP) to total GBM length measured. Scale bar, 360 nm. Error bars, means ± s.e.m.

Accession codes

Referenced accessions

GenBank/EMBL/DDBJ

References

  1. Kitiyakara, C., Eggers, P. & Kopp, J.B. Twenty-one-year trend in ESRD due to focal segmental glomerulosclerosis in the United States. Am. J. Kidney Dis. 44, 815825 (2004).
  2. Baum, M.A. Outcomes after renal transplantation for FSGS in children. Pediatr. Transplant. 8, 329333 (2004).
  3. Senggutuvan, P. et al. Recurrence of focal segmental glomerulosclerosis in transplanted kidneys: analysis of incidence and risk factors in 59 allografts. Pediatr. Nephrol. 4, 2128 (1990).
  4. Hickson, L.J. et al. Kidney transplantation for primary focal segmental glomerulosclerosis: outcomes and response to therapy for recurrence. Transplantation 87, 12321239 (2009).
  5. Ponticelli, C. & Glassock, R.J. Post-transplant recurrence of primary glomerulonephritis. Clin. J. Am. Soc. Nephrol. 5, 23632372 (2010).
  6. Tryggvason, K., Patrakka, J. & Wartiovaara, J. Hereditary proteinuria syndromes and mechanisms of proteinuria. N. Engl. J. Med. 354, 13871401 (2006).
  7. Mathieson, P.W. Proteinuria and immunity—an overstated relationship? N. Engl. J. Med. 359, 24922494 (2008).
  8. Savin, V.J. et al. Galactose binds to focal segmental glomerulosclerosis permeability factor and inhibits its activity. Transl. Res. 151, 288292 (2008).
  9. Hoyer, J.R. et al. Recurrence of idiopathic nephrotic syndrome after renal transplantation. J. Am. Soc. Nephrol. 12, 19942002 (2001).
  10. Artero, M.L. et al. Plasmapheresis reduces proteinuria and serum capacity to injure glomeruli in patients with recurrent focal glomerulosclerosis. Am. J. Kidney Dis. 23, 574581 (1994).
  11. Haas, M. et al. Plasma immunadsorption treatment in patients with primary focal and segmental glomerulosclerosis. Nephrol. Dial. Transplant. 13, 20132016 (1998).
  12. Kemper, M.J., Wolf, G. & Muller-Wiefel, D.E. Transmission of glomerular permeability factor from a mother to her child. N. Engl. J. Med. 344, 386387 (2001).
  13. Glassock, R.J. Circulating permeability factors in the nephrotic syndrome: A fresh look at an old problem. J. Am. Soc. Nephrol. 14, 541543 (2003).
  14. Savin, V.J. et al. Circulating factor associated with increased glomerular permeability to albumin in recurrent focal segmental glomerulosclerosis. N. Engl. J. Med. 334, 878883 (1996).
  15. Sharma, M., Sharma, R., McCarthy, E.T. & Savin, V.J. “The FSGS factor”: enrichment and in vivo effect of activity from focal segmental glomerulosclerosis plasma. J. Am. Soc. Nephrol. 10, 552561 (1999).
  16. Bruneau, S. et al. Potential role of soluble ST2 protein in idiopathic nephrotic syndrome recurrence following kidney transplantation. Am. J. Kidney Dis. 54, 522532 (2009).
  17. McCarthy, E.T., Sharma, M. & Savin, V.J. Circulating permeability factors in idiopathic nephrotic syndrome and focal segmental glomerulosclerosis. Clin. J. Am. Soc. Nephrol. 5, 21152121 (2010).
  18. Wei, C. et al. Modification of kidney barrier function by the urokinase receptor. Nat. Med. 14, 5563 (2008).
  19. Blasi, F. & Carmeliet, P. uPAR: A versatile signaling orchestrator. Nat. Rev. Mol. Cell Biol. 3, 932943 (2002).
  20. Smith, H.W. & Marshall, C.J. Regulation of cell signaling by uPAR. Nat. Rev. Mol. Cell Biol. 11, 2336 (2010).
  21. Sier, C.F. et al. The level of urokinase-type plasminogen activator receptor is increased in the serum of ovarian cancer patients. Cancer Res. 58, 18431849 (1998).
  22. Sidenius, N. et al. Serum level of soluble urokinase-type plasminogen activator receptor is a strong and independent predictor of survival in human immunodeficiency virus infection. Blood 96, 40914095 (2000).
  23. Young, B.C., Levine, R.J. & Karumanchi, S.A. Pathogenesis of preeclampsia. Annu. Rev. Pathol. 5, 173192 (2010).
  24. Ploug, M. & Ellis, V. Structure-function relationship in the receptor for urokinase-type plasminogen activator. Comparison to other members of the Ly-6 family and snake venom a-neurotoxins. FEBS Lett. 349, 163168 (1994).
  25. Kristensen, P., Eriksen, J., Blasi, F. & Dano, K. Two alternatively spliced mouse urokinase receptor mRNAs with different histological localization in the gastrointestinal tract. J. Cell Biol. 115, 17631771 (1991).
  26. Hada, Y. et al. Selective purification and characterization of adiponectin multimer species from human plasma. Biochem. Biophys. Res. Commun. 356, 487493 (2007).
  27. D'Alessio, S. & Blasi, F. The urokinase receptor as an entertainer of signal transduction. Front. Biosci. 14, 45754587 (2009).
  28. Selleri, C. et al. In vivo activity of the cleaved form of soluble urokinase receptor: a new hematopoietic stem/progenitor cell mobilizer. Cancer Res. 66, 1088510890 (2006).
  29. Pampori, N. et al. Mechanisms and consequences of affinity modulation of integrin αVβ3 detected with a novel patch-engineered monovalent ligand. J. Biol. Chem. 274, 2160921616 (1999).
  30. Honda, S. et al. Topography of ligand-induced binding sites, including a novel cation-sensitive epitope (AP5) at the amino terminus, of the human integrin beta 3 subunit. J. Biol. Chem. 270, 1194711954 (1995).
  31. Saleem, M.A. et al. A conditionally immortalized human podocyte cell line demonstrating nephrin and podocin expression. J. Am. Soc. Nephrol. 13, 630638 (2002).
  32. Zaidel-Bar, R., Ballestrem, C., Kam, Z. & Geiger, B. Early molecular events in the assembly of matrix adhesions at the leading edge of migrating cells. J. Cell Sci. 116, 46054613 (2003).
  33. Mundel, P. et al. Synaptopodin: an actin-associated protein in telencephalic dendrites and renal podocytes. J. Cell Biol. 139, 193204 (1997).
  34. Ghiggeri, G.M., Carraro, M. & Vincenti, F. Recurrent focal segmental glomerulosclerosis in the era of genetics of podocyte proteins: theory and therapy. Nephrol. Dial. Transplant. 19, 10361040 (2004).
  35. Dekkers, P.E., ten Hove, T., te Velde, A.A., van Deventer, S.J. & van Der Poll, T. Upregulation of monocyte urokinase plasminogen activator receptor during human endotoxemia. Infect. Immun. 68, 21562160 (2000).
  36. Crowley, S.D. et al. Glomerular type 1 angiotensin receptors augment kidney injury and inflammation in murine autoimmune nephritis. J. Clin. Invest. 119, 943953 (2009).
  37. Hoyer, J.R. et al. Recurrence of idiopathic nephrotic syndrome after renal transplantation. Lancet 2, 343348 (1972).
  38. Gohh, R.Y. et al. Preemptive plasmapheresis and recurrence of FSGS in high-risk renal transplant recipients. Am. J. Transplant. 5, 29072912 (2005).
  39. Beck, L.H. Jr. et al. M-type phospholipase A2 receptor as target antigen in idiopathic membranous nephropathy. N. Engl. J. Med. 36, 1121 (2009).
  40. Clement, L.C. et al. Podocyte-secreted angiopoietin-like-4 mediates proteinuria in glucocorticoid-sensitive nephrotic syndrome. Nat. Med. 17, 117122 (2011).
  41. Zhang, S.Y. et al. c-mip impairs podocyte proximal signaling and induces heavy proteinuria. Sci. Signal. 18, ra39 (2010).
  42. Einecke, G. et al. A molecular classifier for predicting future graft loss in late kidney transplant biopsies. J. Clin. Invest. 120, 18621872 (2010).
  43. Vijayan, K.V., Goldschmidt-Clermont, P.J., Roos, C. & Bray, P.F. The PIA2 polymorphism of integrin β3 enhances outside-in signaling and adhesive functions. J. Clin. Invest. 105, 793802 (2000).
  44. Salido, E. et al. The polymorphism of the platelet glycoprotein IIIA gene as a risk factor for acute renal allograft rejection. J. Am. Soc. Nephrol. 10, 25992605 (1999).
  45. Chiras, T. et al. Platelet GPIIIa polymorphism HPA-1 (PLA1/2) is associated with hypertension as the primary cause for end-stage renal disease in hemodialysis patients from Greece. In Vivo 23, 177181 (2009).
  46. Marszal, J. & Saleem, M.A. The bioactivity of plasma factors in focal segmental glomerulosclerosis. Nephron Exp. Nephrol. 104, e1e5 (2006).
  47. Tjwa, M. et al. Membrane-anchored uPAR regulates the proliferation, marrow pool size, engraftment, and mobilization of mouse hematopoietic stem/progenitor cells. J. Clin. Invest. 119, 10081018 (2009).

Download references

Author information

  1. These authors contributed equally to this work.

    • Shafic El Hindi,
    • Jing Li &
    • Alessia Fornoni

Affiliations

  1. Department of Medicine, Miller School of Medicine, University of Miami, Miami, Florida, USA.

    • Changli Wei,
    • Shafic El Hindi,
    • Jing Li,
    • Alessia Fornoni,
    • Dony Maiguel,
    • Vineet Gupta,
    • David Roth &
    • Jochen Reiser
  2. Diabetes Research Institute, Miller School of Medicine, University of Miami, Miami, Florida, USA.

    • Alessia Fornoni
  3. Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA.

    • Nelson Goes &
    • Boris Nikolic
  4. Department of Surgery, Miller School of Medicine, University of Miami, Miami, Florida, USA.

    • Junichiro Sageshima,
    • George Burke &
    • Phillip Ruiz
  5. Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA.

    • S Ananth Karumanchi
  6. Department of Pediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.

    • Hui-Kim Yap
  7. University of Bristol, Children's Renal Unit, Bristol Royal Hospital for Children, Bristol, UK.

    • Moin Saleem
  8. Department of Surgery, Columbia University, New York, New York, USA.

    • Qingyin Zhang
  9. Department of Pediatrics, Stanford University, Stanford, California, USA.

    • Abanti Chaudhuri &
    • Minnie M Sarwal
  10. The Wallace H. Coulter Center for Translational Research, Miller School of Medicine, University of Miami, Miami, Florida, USA.

    • Pirouz Daftarian
  11. Department of Ophthalmology, Miller School of Medicine, University of Miami, Miami, Florida, USA.

    • Pirouz Daftarian
  12. Servicio de Nefrologia and Centre for Biomedical Research on Rare Diseases (CIBERER), Hospital Universitario de Canarias, Canary Islands, Spain.

    • Eduardo Salido &
    • Armando Torres
  13. Division of Nephrology, SUNY Downstate Medical Center, Brooklyn, New York, USA.

    • Moro Salifu
  14. Center for Pediatric and Adolescent Medicine, University of Heidelberg, Heidelberg, Germany.

    • Franz Schaefer
  15. Department of Nephrology and Endocrinology, University of Heidelberg, Heidelberg, Germany.

    • Christian Morath,
    • Vedat Schwenger &
    • Martin Zeier
  16. Renal Research Laboratory, Fondazione IRCCS Ospedale Maggiore Policlinico & Fondazione D'Amico per la Ricerca sulle Malattie Renali, Milan, Italy.

    • Maria Pia Rastaldi
  17. Department of Pathology, Miller School of Medicine, University of Miami, Miami, Florida, USA.

    • Phillip Ruiz

Contributions

J.R. conceived the study. J.R. and C.W. designed the experiments, coordinated the study, analyzed the data and wrote the manuscript. C.W., S.E.H., J.L., D.M., Q.Z., B.N., P.D., V.G. performed the experiments. A.F., N.G., G.B., J.S., S.A.K., H.-K.Y., M.Saleem, A.C., E.S., A.T., M.Salifu, M.M.S., F.S., C.M., V.S., M.Z., D.R., M.P.R., P.R., J.R. contributed to clinical samples and clinical information. M.P.R. and P.R. provided pathology service.

Competing financial interests

The authors declare no competing financial interests.

Corresponding author

Correspondence to:

Author details

Supplementary information

PDF files

  1. Supplementary Text and Figures (688K)

    Supplementary Figures 1–3, Supplementary Tables 1–3 and Supplementary Methods

Additional data