Effect of the monocyte chemoattractant protein-1/CC chemokine receptor 2 system on nephrin expression in streptozotocin-treated mice and human cultured podocytes

Elena Tarabra, Sara Giunti, Federica Barutta, Gennaro Salvidio, Davina Burt, Giacomo Deferrari, Roberto Gambino, Daniela Vergola, Silvia Pinach, Paolo Cavallo Perin, Giovanni Camussi, Gabriella Gruden, Elena Tarabra, Sara Giunti, Federica Barutta, Gennaro Salvidio, Davina Burt, Giacomo Deferrari, Roberto Gambino, Daniela Vergola, Silvia Pinach, Paolo Cavallo Perin, Giovanni Camussi, Gabriella Gruden

Abstract

Objective: Monocyte chemoattractant protein-1 (MCP-1), a chemokine binding to the CC chemokine receptor 2 (CCR2) and promoting monocyte infiltration, has been implicated in the pathogenesis of diabetic nephropathy. To assess the potential relevance of the MCP-1/CCR2 system in the pathogenesis of diabetic proteinuria, we studied in vitro if MCP-1 binding to the CCR2 receptor modulates nephrin expression in cultured podocytes. Moreover, we investigated in vivo if glomerular CCR2 expression is altered in kidney biopsies from patients with diabetic nephropathy and whether lack of MCP-1 affects proteinuria and expression of nephrin in experimental diabetes.

Research design and methods: Expression of nephrin was assessed in human podocytes exposed to rh-MCP-1 by immunofluorescence and real-time PCR. Glomerular CCR2 expression was studied in 10 kidney sections from patients with overt nephropathy and eight control subjects by immunohistochemistry. Both wild-type and MCP-1 knockout mice were made diabetic with streptozotocin. Ten weeks after the onset of diabetes, albuminuria and expression of nephrin, synaptopodin, and zonula occludens-1 were examined by immunofluorescence and immunoblotting.

Results: In human podocytes, MCP-1 binding to the CCR2 receptor induced a significant reduction in nephrin both mRNA and protein expression via a Rho-dependent mechanism. The MCP-1 receptor, CCR2, was overexpressed in the glomerular podocytes of patients with overt nephropathy. In experimental diabetes, MCP-1 was overexpressed within the glomeruli and the absence of MCP-1 reduced both albuminuria and downregulation of nephrin and synaptopodin.

Conclusions: These findings suggest that the MCP-1/CCR2 system may be relevant in the pathogenesis of proteinuria in diabetes.

Figures

FIG. 1.
FIG. 1.
The CCR2 receptor is expressed by human podocytes. CCR2 protein expression was studied in human cultured podocytes by immunoblotting as described in research design and methods. Total proteins were separated by SDS gel electrophoresis, transferred to nitrocellulose membranes, and probed for the CCR2 receptor by immunoblotting using a rabbit anti-human CCR2 antibody. A representative immunoblot is shown of the specific band for CCR2 at ∼42 kDa. NC: negative control obtained by omitting the primary antibody. PC: positive control of total protein extracts from the monocyte cell line THP-1. PODO: total protein extracts from human podocytes.
FIG. 2.
FIG. 2.
MCP-1 reduces nephrin mRNA and protein expression via a CCR2-Rho-dependent mechanism in cultured human podocytes. A: Nephrin mRNA levels measured by real-time PCR in podocytes exposed to either vehicle or rh-MCP-1 (10 ng/ml) for 2 and 4 h. Results were corrected for the expression of the housekeeping gene glyceraldehydes-3-phosphate dehydrogenase and expressed as percentage decrease as compared with control subjects (n = 3, *P < 0.01 rh-MCP-1 at 2 h vs. control subjects). B: Podocytes were exposed to rh-MCP-1 (10 ng/ml) for 2, 4, 6, 12, and 24 h and (C) to rh-MCP-1 (0.1–1 to 10–100 ng/ml) for 4 h. Nephrin expression, assessed by immunofluorescence, was expressed as percentage change in RFI as compared with control subjects (n = 3 *P < 0.01 rh-MCP-1 at 2, 12, and 24 h over control subjects [□]; †P < 0.001 rh-MCP-1 at 4 and 6 h over control subjects; ‡P < 0.05 rh-MCP-1 at 1 ng/ml over control subjects; §P < 0.001 rh-MCP-1 at 10 ng/ml over control subjects). Representative immunofluorescence images are shown in D (vehicle) and E (rh-MCP-1 at 10 ng/ml for 4 h). Magnification ×400. F: Podocytes were exposed to MCP-1 (10 ng/ml) for 0, 10, 30, 60, 120, and 180 min (upper panel) and 10 min in the absence and/or in the presence of Y27632 (Y27 10 μmol/l), a specific ROCK inhibitor (lower panel). Both total and phosphorylayed MYPT1 were assessed by immunoblotting on total protein extracts. Representative blottings are shown. G: Podocytes were exposed to rh-MCP-1 (10 ng/ml) in the presence and in the absence of RS102895 (RS 6 μmol/l), a CCR2 receptor antagonist, and Y27632 (Y27 10 μmol/l), a specific ROCK inhibitor, added 60 min before rh-MCP-1. After 2 h incubation, nephrin mRNA levels were measured by real-time PCR, corrected for the expression of the housekeeping gene glyceraldehydes-3-phosphate dehydrogenase, and expressed as percentage change over control (n = 3, *P < 0.05 rh-MCP-1 vs. others). H: At 4 h, nephrin protein expression was assessed by indirect immunofluorescence using a low-light video camera and expressed as percentage change in RFI as compared to control subjects (n = 3; *P < 0.05 rh-MCP-1 vs. others).
FIG. 3.
FIG. 3.
MCP-1 effect on synaptopodin expression in cultured human podocytes. Podocytes were exposed either to rh-MCP-1 (10 ng/ml) (A) or vehicle (B) for 4 h, then synaptopodin expression assessed by immunofluorescence. Representative immunofluorescence images are shown (magnification ×800). C: Results were expressed as percentage change in RFI as compared with control subjects (n = 3 *NS rh-MCP-1 vs. control subjects). (A high-quality digital representation of this figure is available in the online issue.)
FIG. 4.
FIG. 4.
CCR2 staining of human glomeruli from control subjects and patients with diabetic nephropathy. CCR2 protein expression was evaluated in human glomeruli from control subjects (A and D) and diabetic patients with overt nephropathy (B and E) by immunohistochemistry as described in research design and methods. C: Nonspecific staining was determined by preabsorbing the anti-CCR2 antibody with a 10-fold excess of control peptide. F: Double immunofluorescence for CCR2 (F) and (G) the podocyte marker synaptopodin performed on the diabetic glomeruli showed colocalisation of the positive staining, as demonstrated by merging (H). Magnification ×400 (×80 D and E). Arrows and arrowhead indicate podocytes and mesangial cells, respectively. (A high-quality digital representation of this figure is available in the online issue.)
FIG. 5.
FIG. 5.
Glomerular staining for nephrin, synaptopodin, and ZO-1 in diabetic wild-type and MCP-1 knockout mice. Kidney paraffin sections from both diabetic and nondiabetic MCP-1+/+ and MCP-1−/− mice were stained for nephrin, synaptopodin, and ZO-1 by immunofluorescence as described in research design and methods. B, D, and F: Quantification of glomerular staining for nephrin (*P < 0.01 diabetic MCP-1+/+ vs. nondiabetic MCP-1+/+ mice; †P < 0.001 diabetic MCP-1−/− vs. diabetic MCP-1+/+ mice), synaptopodin (*P < 0.01 diabetic MCP-1−/− vs. diabetic MCP-1+/+ mice; †P < 0.05 diabetic MCP-1+/+ vs. nondiabetic MCP-1+/+ mice), and ZO-1 (P = NS). A, C, and E: Representative figures of nephrin, synaptopodin, and ZO-1 glomerular staining. Magnification ×400. (A high-quality digital representation of this figure is available in the online issue.)
FIG. 6.
FIG. 6.
Nephrin, synaptopodin, and ZO-1 expression in the renal cortex from diabetic wild-type and MCP-1 knockout mice. Nephrin (A), synaptopodin (B), and ZO-1 (C) expression was studied in renal cortex from both diabetic and nondiabetic MCP-1+/+ and MCP-1−/− mice by immunoblotting as described in research design and methods. Densitometry analysis and representative immunoblots are shown. *P < 0.05 diabetic versus others.
FIG. 7.
FIG. 7.
Morphology of podocyte foot process (transmission electron microscopy, ×7,000) in nondiabetic MCP-1+/+ (A), diabetic MCP-1+/+ (B), diabetic MCP-1−/− (C), and nondiabetic MCP-1−/− (D) mice 10 weeks after the onset of STZ-induced diabetes.

References

    1. Molitch ME, DeFronzo RA, Franz MJ, Keane WF, Mogensen CE, Parving HH, Steffes MW: the American Diabetes Association. Nephropathy in diabetes. Diabetes Care 2004; 27: S79– S83
    1. Li JJ, Kwak SJ, Jung DS, Kim JJ, Yoo TH, Ryu DR, Han SH, Choi HY, Lee JE, Moon SJ, Kim DK, Han DS, Kang SW: Podocyte biology in diabetic nephropathy. Kidney Int 2007; 106: S36– S42
    1. Wolf G, Chen S, Ziyadeh FN: From the periphery of the glomerular capillary wall toward the center of disease: podocyte injury comes of age in diabetic nephropathy. Diabetes 2005; 54: 1626– 1634
    1. Pavenstadt H, Kriz W, Kretzler M: Cell biology of the glomerular podocyte. Physiol Rev 2003; 83: 253– 307
    1. Kestila M, Lenkkeri U, Mannikko M, Lamerdin J, McCready P, Putaala H, Ruotsalainen V, Morita T, Nissinen M, Herva R, Kashtan CE, Peltonen L, Holmberg C, Olsen A, Tryggvason K: Positionally cloned gene for a novel glomerular protein—nephrin—is mutated in congenital nephrotic syndrome. Mol Cell 1998; 1: 575– 582
    1. Huh W, Kim DJ, Kim MK, Kim YG, Oh HY, Ruotsalainen V, Tryggvason K: Expression of nephrin in acquired human glomerular disease. Nephrol Dial Transplant 2002; 17: 478– 484
    1. Doublier S, Ruotsalainen V, Salvidio G, Lupia E, Biancone L, Conaldi PG, Reponen P, Tryggvason K, Camussi G: Nephrin redistribution on podocytes is a potential mechanism for proteinuria in patients with primary acquired nephrotic syndrome. Am J Pathol 2001; 158: 1723– 1731
    1. Benigni A, Gagliardini E, Tomasoni S, Abbate M, Ruggenenti P, Kalluri R, Remuzzi G: Selective impairment of gene expression and assembly of nephrin in human diabetic nephropathy. Kidney Int 2004; 65: 2193– 2200
    1. Doublier S, Salvidio G, Lupia E, Ruotsalainen V, Verzola D, Deferrari G, Camussi G: Nephrin expression is reduced in human diabetic nephropathy: evidence for a distinct role for glycated albumin and angiotensin II. Diabetes 2003; 52: 1023– 1030
    1. Cooper ME: Interaction of metabolic and haemodynamic factors in mediating experimental diabetic nephropathy. Diabetologia 2001; 44: 1957– 1972
    1. Kato S, Luyckx VA, Ots M, Lee KW, Ziai F, Troy JL, Brenner BM, MacKenzie HS: Renin-angiotensin blockade lowers MCP-1 expression in diabetic rats. Kidney Int 1999; 56: 1037– 1048
    1. Sassy-Prigent C, Heudes D, Mandet C: Early glomerular macrophage recruitment in streptozotocin-induced diabetic rats. Diabetes 2000; 49: 466– 475
    1. Chow FY, Nikolic-Paterson DJ, Ozols E, Atkins RC, Rollin BJ, Tesch GH: Monocyte chemoattractant protein-1 promotes the development of diabetic renal injury in streptozotocin-treated mice. Kidney Int 2006; 69: 73– 80
    1. Charo IF, Myers SJ, Herman A, Franci C, Connolly AJ, Coughlin SR: Molecular cloning and functional expression of two monocyte chemoattractant protein 1 receptors reveals alternative splicing of the carboxyl-terminal tails. Proc Natl Acad Sci U S A 1994; 91: 2752– 2756
    1. Viedt C, Vogel J, Athanasiou T, Shen W, Orth SR, Kubler W, Kreuzer J: Monocyte chemoattractant protein-1 induces proliferation and interleukin-6 production in human smooth muscle cells by differential activation of nuclear factor-kB and activator protein-1. Arterioscler Thromb Vasc Biol 2002; 22: 914– 920
    1. Weber KS, Nelson PJ, Grone HJ, Weber C: Expression of CCR2 by endothelial cells: implications for MCP-1 mediated wound injury repair and in vivo inflammatory activation of endothelium. Arterioscler Thromb Vasc Biol 1999; 19: 2085– 2093
    1. Banisadr G, Queraud-Lesaux F, Boutterin MC, Pelaprat D, Zalc B, Rostene W, Haour F, Parsadaniantz SM: Distribution, cellular localization and functional role of CCR2 chemokine receptors in adult rat brain. J Neurochem 2002; 81: 257– 269
    1. Moore BB, Kolodsick JE, Thannickal VJ, Cooke K, Moore TA, Hogaboam C, Wilke CA, Toews GB: CCR2-mediated recruitment of fibrocytes to the alveolar space after fibrotic injury. Am J Pathol 2005; 166: 675– 684
    1. Spinetti G, Wang M, Monticone R, Zhang J, Zhao D, Lakatta EG: Rat aortic MCP-1 and its receptor CCR2 increase with age and alter vascular smooth muscle cell function. Arterioscler Thromb Vasc Biol 2004; 24: 1397– 1402
    1. Giunti S, Pinach S, Arnaldi L, Viberti G, Perin PC, Camussi G, Gruden G: The MCP-1/CCR2 system has direct proinflammatory effects in human mesangial cells. Kidney Int 2006; 69: 856– 863
    1. Giunti S, Tesch GH, Pinach S, Burt DJ, Cooper ME, Cavallo-Perin P, Camussi G, Gruden G: Monocyte chemoattractant protein-1 has prosclerotic effects both in a mouse model of experimental diabetes and in vitro in human mesangial cells. Diabetologia 2008; 51: 198– 207
    1. Burt D, Salvidio G, Tarabra E, Barutta F, Pinach S, Dentelli P, Camussi G, Cavallo Perin P, Gruden G: The monocyte chemoattractant protein-1/cognate CC chemokine receptor 2 system affects cell motility in cultured human podocytes. Am J Pathol 2007; 171: 1789– 1799
    1. Kolavennu V, Zeng L, Peng H, Wang Y, Danesh FR: Targeting of ρ-A/ROCK signaling ameliorates progression of diabetic nephropathy independent of glucose control. Diabetes 2008; 57: 714– 723
    1. Rossing P, Astrup AS, Smidt UM, Parving HH: Monitoring kidney function in diabetic nephropathy. Diabetologia 1994; 37: 708– 712
    1. Mundel P, Heid HW, Mundel TM, Kruger M, Reiser J, Kriz W: Synaptopodin: an actin-associated protein in telencephalic dendrites and renal podocytes. J Cell Biol 1997; 139: 193– 204
    1. Breyer MD, Bottinger E, Brosius FC, Coffman TM, Harris RC, Heilig CW, Sharma K: Mouse models of diabetic nephropathy. J Am Soc Nephrol 2005; 16: 27– 45
    1. Takemoto M, Asker N, Gerhardt H, Lundkvist A, Johansson BR, Saito Y, Betsholtz C: A new method for large scale isolation of kidney glomeruli from mice. Am J Pathol; 2002; 161: 799– 805
    1. Mirzadegan T, Diehl F, Ebi B, Bhakta S, Polsky I, McCarley D, Mulkins M, Weatherhead GS, Lapierre JM, Dankwardt J, Morgans D, Jr, Wilhelm R, Jarnagin K: Identification of the binding site for a novel class of CCR2b chemokine receptor antagonists: binding to a common chemokine receptor motif within the helical bundle. J Biol Chem 2000; 275: 25562– 25571
    1. Uehata M, Ishizaki T, Satoh H, Ono T, Kawahara T, Morishita T, Tamakawa H, Yamagami K, Inui J, Maekawa M, Narumiya S: Calcium sensitization of smooth muscle mediated by a ρ-associated protein kinase in hypertension. Nature 1997; 389: 990– 994
    1. Tesch GH: MCP-1/CCL2: a new diagnostic marker and therapeutic target for progressive renal injury in diabetic nephropathy. Am J Physiol Renal Physiol 2008; 294: F697– F701
    1. Kanamori H, Matsubara T, Mima A, Sumi E, Nagai K, Takahashi T, Abe H, Iehara N, Fukatsu A, Okamoto H, Kita T, Doi T, Arai H: Inhibition of MCP-1/CCR2 pathway ameliorates the development of diabetic nephropathy. Biochem Biophys Res Commun 2007; 360: 772– 777
    1. Han SY, So GA, Jee YH, Han KH, Kang YS, Kim HK, Kang SW, Han DS, Han JY, Cha DR: Effect of retinoic acid in experimental diabetic nephropathy. Immunol Cell Biol 2004; 82: 568– 576
    1. Tylaska LA, Boring L, Weng W, Aiello R, Charo IF, Rollins BJ, Gladue RP: CCR2 regulates the level of MCP-1/CCL2 in vitro and at inflammatory sites and controls T cell activation in response to alloantigen. Cytokine 2002; 18: 184– 190
    1. Bussolati B, Deregibus MC, Fonsato V, Doublier S, Spatola T, Procida S, Di Carlo F, Camussi G: Statins prevent oxidized LDL-induced injury of glomerular podocytes by activating the phosphatidylinositol 3-kinase/AKT-signaling pathway. J Am Soc Nephrol 2005; 16: 1936– 1947
    1. Langham RG, Kelly DJ, Cox AJ, Thomson NM, Holthofer H, Zaoui P, Pinel N, Cordonnier DJ, Gilbert R: Proteinuria and the expression of the podocyte slit diaphragm protein, nephrin, in diabetic nephropathy: effects of angiotensin converting enzyme inhibition. Diabetologia 2002; 45: 1572– 1576
    1. Benigni A, Zoja C, Tomasoni S, Campana M, Corna D, Zanchi C, Gagliardini E, Garofano E, Rottoli D, Ito T, Remuzzi G: Transcriptional regulation of nephrin gene by peroxisome proliferator-activated receptor-γ agonist: molecular mechanism of the antiproteinuric effect of pioglitazone. J Am Soc Nephrol 2006; 17: 1624– 1632
    1. Ren S, Xin C, Beck KF, Saleem MA, Mathieson P, Pavenstädt H, Pfeilschifter J, Huwiler A: PPAR-α activation upregulates nephrin expression in human embryonic kidney epithelial cells and podocytes by a dual mechanism. Biochem Biophys Res Commun 2005; 338: 1818– 1824
    1. Stamatovic SM, Keep RF, Kunkel SL, Andjelkovic AV: Potential role of MCP-1 in endothelial cell tight junction ‘opening’: signaling via ρ and ρ kinase. J Cell Sci 2003; 116: 4615– 4628
    1. Peng F, Wu D, Gao B, Ingram AJ, Zhang B, Chorneyko K, McKenzie R, Krepinsky JC: ρ-A/ρ-kinase contribute to the pathogenesis of diabetic renal disease. Diabetes 2008; 57: 1683– 1692
    1. Kolavennu V, Zeng L, Peng H, Wang Y, Danesh FR: Targeting of ρ-A/ROCK signaling ameliorates progression of diabetic nephropathy independent of glucose control. Diabetes 2008; 57: 714– 23
    1. Rao VH, Meehan DT, Delimont D, Delimont D, Nakajima M, Wada T, Gratton MA, Cosgrove D: Role for macrophage metalloelastase in glomerular basement membrane damage associated with Alport syndrome. Am J Pathol 2006; 169: 32– 46
    1. Rincon-Choles H, Vasylyeva TL, Pergola PE, Bhandari B, Bhandari K, Zhang JH, Wang W, Gorin Y, Barnes JL, Abboud HE: ZO-1 expression and phosphorylation in diabetic nephropathy. Diabetes 2006; 55: 894– 900
    1. Benigni A, Gagliardini E, Tomasoni S, Abbate M, Ruggenenti P, Kalluri R, Remuzzi G: Selective impairment of gene expression and assembly of nephrin in human diabetic nephropathy. Kidney Int 2004; 65: 2193– 2200
    1. Zhang Z, Sun L, Wang Y, Ning G, Minto AW, Kong J, Quigg RJ, Li YC: Renoprotective role of the vitamin D receptor in diabetic nephropathy. Kidney Int 2008; 73: 163– 171
    1. Zhang Z, Zhang Y, Ning G, Deb DK, Kong J, Li YC: Combination therapy with AT1 blocker and vitamin D analog markedly ameliorates diabetic nephropathy: blockade of compensatory renin increase. Proc Natl Acad Sci U S A 2008; 105: 15896– 15901
    1. Sung SH, Ziyadeh FN, Wang A, Pyagay PE, Kanwar YS, Chen S: Blockade of vascular endothelial growth factor signaling ameliorates diabetic albuminuria in mice. J Am Soc Nephrol 2006; 17: 3093– 3104
    1. Kalluri R: Proteinuria with and without renal glomerular podocyte effacement. J Am Soc Nephrol 2006; 17: 2383– 2389
    1. Utimura R, Fujihara CK, Mattar AL, Malheiros DM, Noronha IL, Zatz R: Mycophenolate mofetil prevents the development of glomerular injury in experimental diabetes. Kidney Int 2003; 63: 209– 216
    1. Rodríguez-Iturbe B, Quiroz Y, Shahkarami A, Li Z, Vaziri ND: Mycophenolate mofetil ameliorates nephropathy in the obese Zucker rat. Kidney Int 2005; 68: 1041– 1047

Source: PubMed

3
S'abonner