A Novel Signaling Pathway

The nicotinic acetylcholine receptor α1 (nAChRα1) was investigated as a potential fibrogenic molecule in the kidney, given reports that it may be an alternative urokinase (urokinase plasminogen activator; uPA) receptor in addition to the classical receptor uPAR. In a mouse obstructive uropathy model of chronic kidney disease, interstitial fibroblasts were identified as the primary cell type that bears nAChRα1 during fibrogenesis. Silencing of the nAChRα1 gene led to significantly fewer interstitial αSMA+ myofibroblasts (2.8 times decreased), reduced interstitial cell proliferation (2.6 times decreased), better tubular cell preservation (E-cadherin 14 times increased), and reduced fibrosis severity (24% decrease in total collagen). The myofibroblast-inhibiting effect of nAChRα1 silencing in uPA-sufficient mice disappeared in uPA-null mice, suggesting that a uPA-dependent fibroblastic nAChRα1 pathway promotes renal fibrosis. To further establish this possible ligand-receptor relationship and to identify downstream signaling pathways, in vitro studies were performed using primary cultures of renal fibroblasts. 35S-Labeled uPA bound to nAChRα1 with a Kd of 1.6 × 10−8 m, which was displaced by the specific nAChRα1 inhibitor d-tubocurarine in a dose-dependent manner. Pre-exposure of uPA to the fibroblasts inhibited [3H]nicotine binding. The uPA binding induced a cellular calcium influx and an inward membrane current that was entirely prevented by d-tubocurarine preincubation or nAChRα1 silencing. By mass spectrometry phosphoproteome analyses, uPA stimulation phosphorylated nAChRα1 and a complex of signaling proteins, including calcium-binding proteins, cytoskeletal proteins, and a nucleoprotein. This signaling pathway appears to regulate the expression of a group of genes that transform renal fibroblasts into more active myofibroblasts characterized by enhanced proliferation and contractility. This new fibrosis-promoting pathway may also be relevant to disorders that extend beyond chronic kidney disease.

[1]  D. Pawlak,et al.  Tissue factor and urokinase-type plasminogen activator system are related to the presence of cardiovascular disease in hemodialysis patients. , 2007, Thrombosis research.

[2]  A. Eddy,et al.  Mitogenic signaling of urokinase receptor-deficient kidney fibroblasts: actions of an alternative urokinase receptor and LDL receptor-related protein. , 2004, Journal of the American Society of Nephrology : JASN.

[3]  J. Luketich,et al.  Nicotine signals through muscle-type and neuronal nicotinic acetylcholine receptors in both human bronchial epithelial cells and airway fibroblasts , 2004, Respiratory research.

[4]  Yasuhiko Tomino,et al.  Synaptopodin orchestrates actin organization and cell motility via regulation of RhoA signalling , 2006, Nature Cell Biology.

[5]  D. Belin,et al.  A cellular binding site for the Mr 55,000 form of the human plasminogen activator, urokinase , 1985, The Journal of cell biology.

[6]  K. Preissner,et al.  Binding of Urokinase Plasminogen Activator to gp130 via a Putative Urokinase-Binding Consensus Sequence , 2003, Biological chemistry.

[7]  L Orci,et al.  Transforming growth factor beta stimulates collagen-matrix contraction by fibroblasts: implications for wound healing. , 1988, Proceedings of the National Academy of Sciences of the United States of America.

[8]  D. Dichek,et al.  Mechanisms of Cardiac Fibrosis Induced by Urokinase Plasminogen Activator* , 2006, Journal of Biological Chemistry.

[9]  M. Bokarewa,et al.  Urokinase-type plasminogen activator, an endogenous antibiotic. , 2005, The Journal of infectious diseases.

[10]  B. Eyden The myofibroblast: a study of normal, reactive and neoplastic tissues, with an emphasis on ultrastructure. Part 1--normal and reactive cells. , 2005, Journal of submicroscopic cytology and pathology.

[11]  P. du Toit,et al.  Does urokinase play a role in renal stone formation? , 1997, Medical hypotheses.

[12]  F. Strutz,et al.  Basic fibroblast growth factor expression is increased in human renal fibrogenesis and may mediate autocrine fibroblast proliferation. , 2000, Kidney international.

[13]  A. Eddy,et al.  Endogenous urokinase lacks antifibrotic activity during progressive renal injury. , 2007, American journal of physiology. Renal physiology.

[14]  Junwei Yang,et al.  Single injection of naked plasmid encoding hepatocyte growth factor prevents cell death and ameliorates acute renal failure in mice. , 2002, Journal of the American Society of Nephrology : JASN.

[15]  F. Blasi,et al.  An uncleavable uPAR mutant allows dissection of signaling pathways in uPA-dependent cell migration. , 2005, Molecular biology of the cell.

[16]  In Hye Lee,et al.  Ahnak Protein Activates Protein Kinase C (PKC) through Dissociation of the PKC-Protein Phosphatase 2A Complex* , 2008, Journal of Biological Chemistry.

[17]  Nili Jin,et al.  Gene silencing in mammalian cells by PCR‐based short hairpin RNA , 2003, FEBS letters.

[18]  K. Tracey,et al.  Cholinergic agonists attenuate renal ischemia-reperfusion injury in rats. , 2008, Kidney international.

[19]  P. Carmeliet,et al.  Urokinase receptor deficiency accelerates renal fibrosis in obstructive nephropathy. , 2003, Journal of the American Society of Nephrology : JASN.

[20]  M. Parsons,et al.  Investigation of nicotine binding to THP-1 cells: evidence for a non-cholinergic binding site. , 2001, Biochemical pharmacology.

[21]  Kelly A Kernan,et al.  Plasmin(ogen) promotes renal interstitial fibrosis by promoting epithelial-to-mesenchymal transition: role of plasmin-activated signals. , 2007, Journal of the American Society of Nephrology : JASN.

[22]  V. Tkachuk,et al.  Urokinase Plasminogen Activator in Injured Adventitia Increases the Number of Myofibroblasts and Augments Early Proliferation , 2006, Journal of Vascular Research.

[23]  J. Verheijen,et al.  Mitogenic Effects of Urokinase on Melanoma Cells Are Independent of High Affinity Binding to the Urokinase Receptor* , 1998, The Journal of Biological Chemistry.

[24]  N. Harbeck,et al.  uPA and PAI-1 in breast cancer: review of their clinical utility and current validation in the prospective NNBC-3 trial. , 2008, Advances in clinical chemistry.

[25]  K. Kivirikko,et al.  Modifications of a specific assay for hydroxyproline in urine. , 1967, Analytical biochemistry.

[26]  C. Schwaenen,et al.  Overexpression of urokinase receptor and cell surface urokinase-type plasminogen activator in the human vessel wall with different types of atherosclerotic lesions , 2004, Blood coagulation & fibrinolysis : an international journal in haemostasis and thrombosis.

[27]  B. Agranoff,et al.  Identification of a novel nicotinic acetylcholine receptor structural subunit expressed in goldfish retina , 1989, The Journal of cell biology.

[28]  Y. Gursky,et al.  Chemotactic effect of urokinase plasminogen activator: a major role for mechanisms independent of its proteolytic or growth factor domains. , 1999, Journal of receptor and signal transduction research.

[29]  M. Ploug,et al.  Structure—function relationships in the receptor for urokinase‐type plasminogen activator Comparison to other members of the Ly‐6 family and snake venom α‐neurotoxins , 1994 .

[30]  P. Greengard,et al.  Autoregulation of phosphorylation of the nicotinic acetylcholine receptor , 1994, The Journal of neuroscience : the official journal of the Society for Neuroscience.

[31]  A. Eddy,et al.  Urokinase and its receptors in chronic kidney disease. , 2008, Frontiers in bioscience : a journal and virtual library.

[32]  G. Dooijewaard,et al.  Blood Fibrinolysis and the Response to Desmopressin in Glomerulonephritis , 1994, Thrombosis and Haemostasis.

[33]  F. Di Virgilio,et al.  Intracellular free calcium abnormalities in fibroblasts from non-insulin-dependent diabetic patients with and without arterial hypertension. , 1997, Hypertension.

[34]  P. Grieco,et al.  Activation of urokinase receptor by a novel interaction between the connecting peptide region of urokinase and αvβ5 integrin , 2006, Journal of Cell Science.

[35]  P. Carmeliet,et al.  Urokinase receptor modulates cellular and angiogenic responses in obstructive nephropathy. , 2003, Journal of the American Society of Nephrology : JASN.

[36]  T. Astrup,et al.  The plasminogen activator in urine and the urinary trypsin inhibitor. , 1955, Scandinavian journal of clinical and laboratory investigation.

[37]  G. Lagoumintzis,et al.  Muscle and neuronal nicotinic acetylcholine receptors , 2007 .

[38]  V. Tkachuk,et al.  The Chemotactic Action of Urokinase on Smooth Muscle Cells Is Dependent on Its Kringle Domain , 2000, The Journal of Biological Chemistry.

[39]  A Bensimon,et al.  Monitoring S phase progression globally and locally using BrdU incorporation in TK(+) yeast strains. , 2001, Nucleic acids research.

[40]  J. Merlie,et al.  An acetylcholine receptor precursor alpha subunit that binds alpha-bungarotoxin but not d-tubocurare. , 1986, Proceedings of the National Academy of Sciences of the United States of America.

[41]  A. Mondino,et al.  uPA and uPAR in fibrinolysis, immunity and pathology. , 2004, Trends in immunology.

[42]  A. Eddy Progression in chronic kidney disease. , 2005, Advances in chronic kidney disease.

[43]  J. Massagué TGF-beta signal transduction. , 1998, Annual review of biochemistry.

[44]  J. Steinbach,et al.  Activation and block of mouse muscle‐type nicotinic receptors by tetraethylammonium , 2003, The Journal of physiology.

[45]  J. Schmidt,et al.  Expression of the acetylcholine receptor delta‐subunit gene in differentiating chick muscle cells is activated by an element that contains two 16 bp copies of a segment of the alpha‐subunit enhancer. , 1990, The EMBO journal.

[46]  E. Tamm,et al.  Single-cell contraction assay for human ciliary muscle cells. Effect of carbachol. , 1993, Investigative Ophthalmology and Visual Science.

[47]  Steven E. Wilson,et al.  Dynamics of the expression of intermediate filaments vimentin and desmin during myofibroblast differentiation after corneal injury. , 2009, Experimental eye research.

[48]  P. Carmeliet,et al.  Physiological consequences of loss of plasminogen activator gene function in mice , 1994, Nature.

[49]  J. Wolff,et al.  High levels of foreign gene expression in hepatocytes after tail vein injections of naked plasmid DNA. , 1999, Human gene therapy.

[50]  Roger E Bumgarner,et al.  Multifunctionality of PAI-1 in fibrogenesis: evidence from obstructive nephropathy in PAI-1-overexpressing mice. , 2005, Kidney international.