Volume 21, Issue 2, Pages (February 2013)

Slides:



Advertisements
Similar presentations
Cardiac-Specific Overexpression of HIF-1α Prevents Deterioration of Glycolytic Pathway and Cardiac Remodeling in Streptozotocin-Induced Diabetic Mice 
Advertisements

Volume 84, Issue 6, Pages (December 2013)
The Absence of Interleukin-6 Enhanced Arsenite-Induced Renal Injury by Promoting Autophagy of Tubular Epithelial Cells with Aberrant Extracellular Signal-Regulated.
MicroRNA-558 regulates the expression of cyclooxygenase-2 and IL-1β-induced catabolic effects in human articular chondrocytes  S.J. Park, E.J. Cheon,
Volume 65, Issue 1, Pages (January 2004)
Volume 66, Issue 6, Pages (December 2004)
Volume 77, Issue 6, Pages (March 2010)
Volume 24, Issue 7, Pages (July 2016)
Volume 78, Issue 3, Pages (August 2010)
Canonical Wnt/β-catenin signaling mediates transforming growth factor-β1-driven podocyte injury and proteinuria  Dan Wang, Chunsun Dai, Yingjian Li, Youhua.
Volume 84, Issue 6, Pages (December 2013)
Volume 85, Issue 2, Pages (January 2014)
HDAC Dependent Transcriptional Repression of Bmp-7 Potentiates TGF-β Mediated Renal Fibrosis in Obstructive Uropathy  Scott R. Manson, Joseph B. Song,
Tranilast attenuates connective tissue growth factor-induced extracellular matrix accumulation in renal cells  W. Qi, X. Chen, S. Twigg, T.S. Polhill,
Volume 76, Issue 1, Pages (July 2009)
Volume 78, Issue 4, Pages (August 2010)
Volume 82, Issue 6, Pages (September 2012)
Volume 85, Issue 2, Pages (January 2014)
MicroRNA-558 regulates the expression of cyclooxygenase-2 and IL-1β-induced catabolic effects in human articular chondrocytes  S.J. Park, E.J. Cheon,
Volume 71, Issue 3, Pages (February 2007)
Smad7 gene transfer inhibits peritoneal fibrosis
MicroRNA-29b Inhibits Diabetic Nephropathy in db/db Mice
Volume 67, Issue 3, Pages (March 2005)
Volume 84, Issue 2, Pages (August 2013)
IN-1130, a novel transforming growth factor-β type I receptor kinase (ALK5) inhibitor, suppresses renal fibrosis in obstructive nephropathy  J.-A. Moon,
Volume 60, Issue 2, Pages (August 2001)
Volume 86, Issue 3, Pages (September 2014)
Anti-fibrotic Effects of Synthetic Oligodeoxynucleotide for TGF-β1 and Smad in an Animal Model of Liver Cirrhosis  Jung-Yeon Kim, Hyun-Jin An, Woon-Hae.
Volume 88, Issue 3, Pages (September 2015)
Volume 58, Issue 6, Pages (December 2000)
Volume 84, Issue 1, Pages (July 2013)
Volume 79, Issue 9, Pages (May 2011)
Mast cells decrease renal fibrosis in unilateral ureteral obstruction
Volume 87, Issue 2, Pages (February 2015)
Volume 61, Issue 1, Pages (January 2002)
Albumin up-regulates the type II transforming growth factor-beta receptor in cultured proximal tubular cells1  Gunter Wolf, Regine Schroeder, Fuad N.
Volume 80, Issue 10, Pages (November 2011)
Volume 61, Issue 5, Pages (May 2002)
Volume 24, Issue 5, Pages (May 2016)
Heterozygous disruption of activin receptor–like kinase 1 is associated with increased renal fibrosis in a mouse model of obstructive nephropathy  José.
Role of Connective Tissue Growth Factor in Oval Cell Response During Liver Regeneration After 2-AAF/PHx in Rats  Liya Pi, Seh-Hoon Oh, Thomas Shupe, Bryon.
Volume 81, Issue 3, Pages (February 2012)
Volume 84, Issue 2, Pages (August 2013)
Volume 84, Issue 5, Pages (November 2013)
Volume 81, Issue 5, Pages (March 2012)
Hepatocyte growth factor suppresses interstitial fibrosis in a mouse model of obstructive nephropathy  Shinya Mizuno, Kunio Matsumoto, Toshikazu Nakamura 
Antagonistic Effect of the Matricellular Signaling Protein CCN3 on TGF-β- and Wnt- Mediated Fibrillinogenesis in Systemic Sclerosis and Marfan Syndrome 
Volume 23, Issue 8, Pages (August 2015)
Sustained Activation of Fibroblast Transforming Growth Factor-β/Smad Signaling in a Murine Model of Scleroderma  Shinsuke Takagawa, Gabriella Lakos, Yasuji.
1,25-dihydroxyvitamin D3 inhibits renal interstitial myofibroblast activation by inducing hepatocyte growth factor expression  Yingjian Li, Bradley C.
Overexpression of CD109 in the Epidermis Differentially Regulates ALK1 Versus ALK5 Signaling and Modulates Extracellular Matrix Synthesis in the Skin 
Volume 79, Issue 4, Pages (February 2011)
Jin H. Li, Xiao R. Huang, Hong-Jian Zhu, Richard Johnson, Hui Y. Lan 
Volume 92, Issue 3, Pages (September 2017)
Volume 66, Issue 5, Pages (November 2004)
Volume 19, Issue 8, Pages (August 2011)
Volume 85, Issue 2, Pages (January 2014)
Volume 78, Issue 7, Pages (October 2010)
Volume 82, Issue 2, Pages (July 2012)
Volume 61, Issue 1, Pages S94-S98 (January 2002)
Volume 56, Issue 2, Pages (August 1999)
M.-J. Wu, M.-C. Wen, Y.-T. Chiu, Y.-Y. Chiou, K.-H. Shu, M.-J. Tang 
Volume 67, Issue 6, Pages (June 2005)
LncRNA ZEB1-AS1 Was Suppressed by p53 for Renal Fibrosis in Diabetic Nephropathy  Juan Wang, Jian Pang, Huiling Li, Jie Long, Fang Fang, Junxiang Chen,
Volume 60, Issue 5, Pages (November 2001)
Molecular Therapy - Nucleic Acids
miR-29 Inhibits Bleomycin-induced Pulmonary Fibrosis in Mice
MicroRNA-125b Promotes Hepatic Stellate Cell Activation and Liver Fibrosis by Activating RhoA Signaling  Kai You, Song-Yang Li, Jiao Gong, Jian-Hong Fang,
Volume 22, Issue 6, Pages (June 2014)
miR-25 Tough Decoy Enhances Cardiac Function in Heart Failure
Presentation transcript:

Volume 21, Issue 2, Pages 388-398 (February 2013) Smad7 suppresses renal fibrosis via altering expression of TGF-β/Smad3-regulated microRNAs  Arthur C.K. Chung, Yuan Dong, Weiqin Yang, Xiang Zhong, Rong Li, Hui Y. Lan  Molecular Therapy  Volume 21, Issue 2, Pages 388-398 (February 2013) DOI: 10.1038/mt.2012.251 Copyright © 2013 The American Society of Gene & Cell Therapy Terms and Conditions

Figure 1 Delivery of Smad7 overexpression plasmids attenuates renal fibrosis and the expression levels of fibrotic markers in mice. (a) Histology and immunohistochemistry. (b) Quantitative analysis of immunohistochemical staining. (c) Representative western blots. Expression of collagen I, α-SMA, and phosphorylated Smad3 (P-Smad3) in ligated kidneys increases at day 7 after unilateral ureteral obstruction (UUO) but reduces after gene transfer of Smad7 overexpression plasmids (S7 plasmid). Note that the expression levels of total Smad3 (T-Smad3) are not affected during fibrosis. Owing to the cross-reactivity of the antibody, the lower bands are total Smad3 while the upper bands are total Smad2 (T-Smad2). Each bar represents the mean + SEM for at least eight mice. **P < 0.01, ***P < 0.001 as compared with normal mice; ##P < 0.01, ###P < 0.001 as compared with UUO mice with control plasmids (Ctl plasmid). α-SMA, α-smooth muscle actin. Molecular Therapy 2013 21, 388-398DOI: (10.1038/mt.2012.251) Copyright © 2013 The American Society of Gene & Cell Therapy Terms and Conditions

Figure 2 Smad7 overexpression attenuates the expression levels of fibrotic markers and alters the expression levels of fibrosis-related microRNAs. (a) Real-time PCR of expression of fibrotic markers in the unilateral ureteral obstruction (UUO) model. Expression of collagen I, fibronectin (Fn), and α-smooth muscle actin (α-SMA) in ligated kidneys increases after UUO but reduces after gene transfer of Smad7 overexpression plasmids (S7 plasmid). (b) Real-time PCR of expression of renal expression of miR-29b, miR-192, and miR-21 in the UUO model. (c–e) The combination of immunohistochemical staining with the anti-Smad7 antibody (red) and in situ hybridization with FITC-miR-29b, miR-192, and miR-21 (green). At Day7 of UUO, expression levels of miR-192 and miR-21 are elevated and miR-29b expression is reduced. During UUO, endogenous Smad7 expression is reduced in ligated kidneys. Smad7 overexpression restores miR-29b expression but abolishes expression of miR-21 and miR-192. No signal was observed when a scramble microRNA probe was used (data not shown). Each bar represents the mean + SEM for at least eight mice. ***P < 0.001 as compared with normal mice; ##P < 0.01, ###P < 0.001 as compared with UUO mice with control plasmids (Ctl plasmid). Ctl: control plasmid; S7: Smad7 overexpression plasmid. Molecular Therapy 2013 21, 388-398DOI: (10.1038/mt.2012.251) Copyright © 2013 The American Society of Gene & Cell Therapy Terms and Conditions

Figure 3 Overexpression of Smad7 reduces transforming growth factor-β1 (TGF-β1)-induced expression of fibrotic markers and alters expression levels of miR-29b, miR-21, and miR-192 in rat mesangial cells (MCs) and tubular epithelial cells (TECs). (a) Representative western blots of expression of fibrotic markers in TECs at 24 hour after treatment with TGF-β1. (b) Representative western blots of expression of fibrotic markers in MCs at 24 hour after treatment with TGF-β1. (c) Real-time PCR analysis of expression of fibrotic markers in TECs. (d) Real-time PCR analysis of expression of fibrotic markers in MCs. (e) Real-time PCR analysis of microRNA expression in TECs. (f) Real-time PCR analysis of microRNA expression in MCs. Overexpression of Smad7 in MCs and TECs suppresses mRNA and protein expression of collagen I (Col I), fibronectin (Fn), and α-smooth muscle actin (α-SMA) induced by TGF-β1 after Dox treatment. MCs and TECs transfected with Smad7 overexpression plasmids restores miR-29b expression, and inhibits the induction of miR-21 and miR-192 by TGF-β1. Each bar represents the mean + SEM for at least three independent experiments. *P < 0.05, **P < 0.01, ***P < 0.001 as compared with time 0; #P < 0.05, ##P < 0.01, ###P < 0.001 as compared with without Dox treatment. Molecular Therapy 2013 21, 388-398DOI: (10.1038/mt.2012.251) Copyright © 2013 The American Society of Gene & Cell Therapy Terms and Conditions

Figure 4 Overexpression of Smad7 restores miR-29b expression, and suppresses the induction of miR-21 and miR-192 after advanced glycated end products (AGE) and angiotensin II (AngII) treatments in rat mesangial cells (MCs) and tubular epithelial cells (TECs). (a) Real-time PCR analysis of microRNA expression in TECs after treatment of AGE (33 µg/ml). (b) Real-time PCR analysis of microRNA expression in MCs after treatment of AGE (33 µg/ml). (c) Real-time PCR analysis of microRNA expression in TECs after treatment of AngII (1.0 µmol/l). (d) Real-time PCR analysis of microRNA expression in MCs after treatment of AngII (1.0 µmol/l). Treatment with Ang II and AGE in MCs and TECs induce expression of miR-21 and miR-192 but suppress miR-29b. In MCs and TECs transfected with Smad7 overexpression plasmids restores miR-29b expression, and inhibits the induction of miR-21 and miR-192 by AGE and AngII. No effect is observed when BSA control or no AngII is used in the absence of Dox. BSA, bovine serum albumin (33 µg/ml) as a control of AGE treatment without Dox treatment. No AngII, absence of AngII as a control of AngII treatment without Dox treatment. Each bar represents the mean + SEM for at least 3 independent experiments. *P < 0.05, **P < 0.01, ***P < 0.001 as compared with time 0; #P < 0.05, ##P < 0.01, ###P < 0.001 as compared with no Dox treatment. Molecular Therapy 2013 21, 388-398DOI: (10.1038/mt.2012.251) Copyright © 2013 The American Society of Gene & Cell Therapy Terms and Conditions

Figure 5 Disruption of Smad7 gene in mice promotes renal fibrosis by increasing renal collagen I deposition and altering the expression of miR-21 and miR-29b at day 7 after unilateral ureteral obstruction (UUO). (a) Immunohistochemical analysis of collagen I. (i) A normal kidney from an untreated wild-type (WT) mouse, (ii) a normal kidney from an untreated Smad7ΔE1 mouse, (iii) a UUO kidney from a WT mouse, (iv) a UUO kidney from a Smad7ΔE1 mouse. (b) Real-time PCR results of miR-21 and miR-29b expression in Smad7 WT/ΔE1 kidneys with or without UUO at day 7. Note that both untreated WT and Smad7ΔE1 mice show little accumulation of collagen I within the interstitium (i and ii), but severe renal fibrosis with an abundant collagen I accumulation within the tubulointerstitium is developed in WT mice with UUO (iii), which is significantly enhanced in Smad7ΔE1 mice (iv). The slides were counterstained with hematoxylin for the nuclei. Each bar represents the mean + SEM for a group of eight mice. **P < 0.01, ***P < 0.001 as compared with the normal mice. ##P < 0.01 as compared with the WT mice with UUO. Magnification ×400. Molecular Therapy 2013 21, 388-398DOI: (10.1038/mt.2012.251) Copyright © 2013 The American Society of Gene & Cell Therapy Terms and Conditions

Figure 6 Delivery of miR-192 mimic re-induces transforming growth factor-β1 (TGF-β1)-induced expression of fibrotic markers in Smad7 overexpressing tubular epithelial cells (TECs). (a) Real-time PCR analysis of miR-192 in TECs at 24 hours after TGF-β1 treatment (i) with all samples (ii) without miR-192 mimic. (b) Representative western blots of fibrotic markers. (c) Quantitative analysis of western blots. (d) Real-time PCR analysis of fibrotic markers. Note that (a-i) TECs transfected with miR-192 mimics contain an excessive amount of the mature form of miR-192. (a-ii) TECs transfected with Smad7 overexpression plasmids show a reduction of TGF-β1-induced miR-192 expression. After treatment with TGF-β1 for 24 hours, overexpression of Smad7 in TECs does not totally suppress TGF-β1-induced expression of collagen I (Col I), fibronectin (Fn), and α-SMA after the delivery of miR-192 mimic. Each bar represents the mean + SEM for at least three independent experiments. *P < 0.05, **P < 0.01, ***P < 0.001 as compared with time 0; #P < 0.05, ##P < 0.01, ###P < 0.001 as compared with no delivery of miR-192 mimic. ††P < 0.01, †††P < 0.001 as compared with no Dox treatment. SCR, scramble microRNA; α-SMA, α-smooth muscle actin. Molecular Therapy 2013 21, 388-398DOI: (10.1038/mt.2012.251) Copyright © 2013 The American Society of Gene & Cell Therapy Terms and Conditions

Figure 7 Delivery of miR-21 mimic re-induces transforming growth factor-β1 (TGF-β1)-induced expression of fibrotic markers in Smad7 overexpressing tubular epithelial cells (TECs). (a) Real-time PCR analysis of miR-21 in TECs at 24 hours after TGF-β1 treatment (i) with all samples (ii) without miR-21 mimic only. (b) Representative western blots of fibrotic markers. (c) Quantitative analysis of western blots. (d) Real-time PCR analysis of fibrotic markers. Note that (a-i) TECs transfected with miR-21 mimics contain an excessive amount of the mature form of miR-21. (a-ii) TECs transfected with Smad7 overexpression plasmids show a reduction of TGF-β1-induced miR-21 expression. After treatment with TGF-β1 for 24 hours, overexpression of Smad7 in TECs partly suppresses TGF-β1-induced expression of collagen I (Col I), fibronectin (Fn), and α-SMA after the delivery of miR-21 mimic. Each bar represents the mean + SEM for at least three independent experiments. *P < 0.05, **P < 0.01, ***P < 0.001 as compared with time 0; #P < 0.05, ##P < 0.01, ###P < 0.001 as compared with no delivery of miR-21 mimic. ††P < 0.01, †††P < 0.001 as compared with no Dox treatment. SCR: scramble microRNA; α-SMA, α-smooth muscle actin. Molecular Therapy 2013 21, 388-398DOI: (10.1038/mt.2012.251) Copyright © 2013 The American Society of Gene & Cell Therapy Terms and Conditions

Figure 8 Delivery of anti-miR-29b re-induces transforming growth factor-β1 (TGF-β1)-induced expression of fibrotic markers in Smad7 overexpressing tubular epithelial cells (TECs). (a) Real-time PCR analysis of miR-29b in TECs at 24 hours after TGF-β1 treatment (i) with all samples (ii) without anti-miR-29b only. (b) Representative western blots of fibrotic markers. (c) Quantitative analysis of western blots. (d) Real-time PCR analysis of fibrotic markers. Note that (a–i) TECs transfected with anti-miR-29b suppresses the expression of the mature form of miR-29b. TECs transfected with Smad7 overexpression plasmids show a restoration of TGF-β1-suppresed miR-29b expression. After treatment with TGF-β1 for 24 hours, overexpression of Smad7 in TECs partly suppresses TGF-β1-induced expression of collagen I (Col I), fibronectin (Fn), and α-SMA after the delivery of anti-miR-29b. Each bar represents the mean + SEM for at least three independent experiments. *P < 0.05, **P < 0.01, ***P < 0.001 as compared with time 0; #P < 0.05, ##P < 0.01, ###P < 0.001 as compared with no delivery of miR-21 mimic. ††P < 0.01, †††P < 0.001 as compared with no Dox treatment. SCR: scramble microRNA; α-SMA, α-smooth muscle actin. Molecular Therapy 2013 21, 388-398DOI: (10.1038/mt.2012.251) Copyright © 2013 The American Society of Gene & Cell Therapy Terms and Conditions