Kunio Matsumoto, Toshikazu Nakamura  Kidney International 

Slides:



Advertisements
Similar presentations
Volume 56, Issue 4, Pages (October 1999)
Advertisements

Hepatocyte growth factor in renal failure: Promise and reality
In vitro branching tubulogenesis: Implications for developmental and cystic disorders, nephron number, renal repair, and nephron engineering  Hiroyuki.
Vascular Endothelial Growth Factor (VEGF)-Mediated Angiogenesis Is Associated with Enhanced Endothelial Cell Survival and Induction of Bcl-2 Expression 
Inflammatory cytokines in acute renal failure
Volume 78, Issue 11, Pages (December 2010)
HDAC Dependent Transcriptional Repression of Bmp-7 Potentiates TGF-β Mediated Renal Fibrosis in Obstructive Uropathy  Scott R. Manson, Joseph B. Song,
Volume 68, Issue 6, Pages (December 2005)
Regulation of mesangial cell proliferation
Hepatocyte growth factor in renal failure: Promise and reality
Toll-like receptor activation: from renal inflammation to fibrosis
Obstructive nephropathy: Insights from genetically engineered animals
Exosomes/microvesicles as a mechanism of cell-to-cell communication
Volume 63, Issue 2, Pages (February 2003)
Volume 86, Issue 3, Pages (September 2014)
Volume 91, Issue 5, Pages (May 2017)
TGF-β isoforms in renal fibrogenesis
EGFR signaling in renal fibrosis
Volume 82, Issue 10, Pages (November 2012)
Volume 93, Issue 1, Pages (January 2018)
Epidermal growth factor receptor inhibition attenuates early kidney enlargement in experimental diabetes  Lesley Wassef, Darren J. Kelly, Richard E. Gilbert 
Volume 84, Issue 6, Pages (December 2013)
Volume 54, Issue 4, Pages (October 1998)
JAK/STAT signaling in renal diseases
Volume 65, Issue 6, Pages (June 2004)
Paricalcitol attenuates cyclosporine-induced kidney injury in rats
Activation of hepatocyte growth factor receptor, c-met, in renal tubules is required for renoprotection after acute kidney injury  Dong Zhou, Roderick.
Volume 82, Issue 9, Pages (November 2012)
Volume 68, Issue 1, Pages (July 2005)
Endogenous hepatocyte growth factor ameliorates chronic renal injury by activating matrix degradation pathways  Youhua Liu, Krupa Rajur, Evelyn Tolbert,
Karl A. Nath, Anthony J. Croatt, Jill J. Haggard, Joseph P. Grande 
Hepatocyte growth factor suppresses interstitial fibrosis in a mouse model of obstructive nephropathy  Shinya Mizuno, Kunio Matsumoto, Toshikazu Nakamura 
AOPPs and the progression of kidney disease
Met as a therapeutic target in HCC: Facts and hopes
Volume 70, Issue 7, Pages (October 2006)
Volume 126, Issue 3, Pages (March 2004)
1,25-dihydroxyvitamin D3 inhibits renal interstitial myofibroblast activation by inducing hepatocyte growth factor expression  Yingjian Li, Bradley C.
Volume 66, Issue 6, Pages (December 2004)
Reciprocal functions of hepatocyte growth factor and transforming growth factor-β1 in the progression of renal diseases: A role for CD44?  Sandrine Florquin,
Deletion of the epidermal growth factor receptor in renal proximal tubule epithelial cells delays recovery from acute kidney injury  Jianchun Chen, Jian-Kang.
Jin H. Li, Xiao R. Huang, Hong-Jian Zhu, Richard Johnson, Hui Y. Lan 
Volume 92, Issue 3, Pages (September 2017)
Saulo Klahr, Jeremiah J. Morrissey  Kidney International 
Volume 41, Issue 6, Pages (December 2004)
Diarmaid Dominic Houlihan, Philip Noel Newsome  Gastroenterology 
Karsten Bartels, Almut Grenz, Holger K. Eltzschig  Kidney International 
Volume 63, Issue 5, Pages (May 2003)
Volume 59, Issue 2, Pages (February 2001)
Volume 69, Issue 12, Pages (June 2006)
Volume 83, Issue 4, Pages (April 2013)
Acute renal failure: From renal physiology to the renal transcriptome
CD4+ T cells: a potential player in renal fibrosis
Matrix metalloproteinases and matrix receptors in progression and reversal of kidney disease: therapeutic perspectives  Pierre Ronco, Christos Chatziantoniou 
Volume 65, Issue 6, Pages (June 2004)
Florian E. Togel, Christof Westenfelder 
Volume 56, Issue 5, Pages (November 1999)
Jinhua Tang, Na Liu, Shougang Zhuang  Kidney International 
Stephen O'Neill, Jeremy Hughes  Kidney International 
Molecular mechanisms of renal hypertrophy: Role of p27Kip1
Volume 76, Issue 5, Pages (September 2009)
Volume 55, Issue 2, Pages (February 1999)
David A. Ferenbach, David C. Kluth, Jeremy Hughes  Kidney International 
Volume 70, Issue 2, Pages (July 2006)
Cell cycle regulation: Repair and regeneration in acute renal failure
Volume 75, Issue 6, Pages (March 2009)
T-PA promotes glomerular plasmin generation and matrix degradation in experimental glomerulonephritis  Masashi Haraguchi, Wayne A. Border, Yufeng Huang,
Volume 56, Issue 4, Pages (October 1999)
Volume 21, Issue 2, Pages (February 2013)
Volume 18, Issue 7, Pages (July 2010)
Renoprotection with vitamin D: Specific for diabetic nephropathy?
Presentation transcript:

Hepatocyte growth factor: Renotropic role and potential therapeutics for renal diseases  Kunio Matsumoto, Toshikazu Nakamura  Kidney International  Volume 59, Issue 6, Pages 2023-2038 (June 2001) DOI: 10.1046/j.1523-1755.2001.00717.x Copyright © 2001 International Society of Nephrology Terms and Conditions

Figure 1 Schematic structures. (A) Prohepatocyte growth factor (HGF) and mature HGF. (B) Typical biological activities of HGF mediated by c-Met/HGF receptor and intracellular signal transducers which associate with tyrosine-phosphorylated c-Met. Kidney International 2001 59, 2023-2038DOI: (10.1046/j.1523-1755.2001.00717.x) Copyright © 2001 International Society of Nephrology Terms and Conditions

Figure 2 (A) Mitogenic action of HGF (•) and epidermal growth factor (EGF; ○) on cultured rabbit renal tubular cells. DNA synthesis of renal tubular cells cultured in the absence or presence of HGF or EGF was measured. (B) Induction of branching tubulogenesis by HGF in MDCK renal epithelial cells grown in a collagen gel. Cells were grown in the absence or presence of 10 ng/mL HGF. (C) Potential roles of HGF in epithelial–stromal and endothelial–mesangial interactions in the kidney. Kidney International 2001 59, 2023-2038DOI: (10.1046/j.1523-1755.2001.00717.x) Copyright © 2001 International Society of Nephrology Terms and Conditions

Figure 3 Possible pathways mediated by HGF for renal regeneration in response to acute renal injury. (A) Following renal injuries, HGF expression is up-regulated in stromal cells in the kidney and in distant intact organs such as the lung, liver, and spleen. The right upper panel shows induction of HGF mRNA in the lung following unilateral nephrectomy (UN) in rats. (B) Regulation of c-Met receptor activation in noninjured and injured tissues. Change in cell–cell contact or junctional communication due to tissue injury may possibly regulate c-Met receptor function, such that HGF exerts its biological activities in the injured tissue-specific manner. Kidney International 2001 59, 2023-2038DOI: (10.1046/j.1523-1755.2001.00717.x) Copyright © 2001 International Society of Nephrology Terms and Conditions

Figure 4 Possible involvement of counterbalance between HGF and transforming growth factor-β (TGF-β) in the pathogenesis of chronic renal failure/renal fibrosis. In the beginning of chronic injury, HGF expression is up-regulated, and the compensatory regenerative responses occur. Repetitive injury results in overexpression TGF-β, a potent fibrogenic cytokine that induces extracellular matrix (ECM) accumulation and epithelial and endothelial apoptosis. Since TGF-β suppresses HGF expression, HGF decreases in a reciprocal manner to the increase in TGF-β level during the progression of chronic renal failure. Kidney International 2001 59, 2023-2038DOI: (10.1046/j.1523-1755.2001.00717.x) Copyright © 2001 International Society of Nephrology Terms and Conditions

Figure 5 Preventive effect of HGF on acute renal failure induced by administration of cisplatin (A) or HgCl2 (B). Acute renal failure was induced by cisplatin (15 mg/kg body weight; A) or HgCl2 (7 mg/kg body weight; B). Mice were injected with HGF (250 μg/kg body weight) at 0.5 hours prior to administration of cisplatin or HgCl2 and 6, 12, 24, 36, 48 and 72 hours or 6, 12, 24, 36, and 48 hours after the administration of cisplatin or HgCl2, respectively. BUN (▪) and serum creatinine () at 96 (cisplatin) or 72 hours (HgCl2) were measured. Each value represents the mean ± SD174. Kidney International 2001 59, 2023-2038DOI: (10.1046/j.1523-1755.2001.00717.x) Copyright © 2001 International Society of Nephrology Terms and Conditions

Figure 6 Therapeutic effect of HGF on chronic renal failure/renal fibrosis in ICGN mice. (A) Changes in TGF-β, collagen accumulation, tubular apoptosis, and tubular proliferation by HGF administration in kidneys of ICGN mice, as determined by enzyme immunoassay (TGF-β) and immunohistochemical analysis (collagen type I, apoptosis, and BrdU-uptake). (B) Changes in BUN and serum creatinine (SCr) levels, without (○) or with (•) HGF-treatment. When BUN levels reached 40 mg/dL, HGF (500 μg/kg) or saline alone was given daily for 28 days108,152. Kidney International 2001 59, 2023-2038DOI: (10.1046/j.1523-1755.2001.00717.x) Copyright © 2001 International Society of Nephrology Terms and Conditions

Figure 7 Putative mechanism for therapeutic effects of HGF on chronic renal failure/renal fibrosis. The supplement of HGF suppresses TGF-β expression and enhances remodeling of renal tissues, including ECM degradation and epithelial and endothelial cell proliferation in fibrotic kidneys. HGF may prevent cell death in epithelial and endothelial cells. Kidney International 2001 59, 2023-2038DOI: (10.1046/j.1523-1755.2001.00717.x) Copyright © 2001 International Society of Nephrology Terms and Conditions