Obstructive nephropathy: Insights from genetically engineered animals

Slides:



Advertisements
Similar presentations
- + cellular infiltration myofibroblast accumulation tubular atrophy
Advertisements

Molecular mechanims of CKD progression following obstructive uropathy Mordi Muorah Clinical Fellow INSERM U845 – Equipe Fabiola Terzi Hopital Necker Enfants-
In vitro branching tubulogenesis: Implications for developmental and cystic disorders, nephron number, renal repair, and nephron engineering  Hiroyuki.
Biologic therapy of inflammatory bowel disease
Volume 78, Issue 3, Pages (August 2010)
Extracellular Vesicles in Cancer: Cell-to-Cell Mediators of Metastasis
NOD-like and Toll-like receptors or inflammasomes contribute to kidney disease in a canonical and a non-canonical manner  Hans-Joachim Anders, Maciej.
Jan-Hendrik B. Hardenberg, Andrea Braun, Michael P. Schön 
Biologic therapy of inflammatory bowel disease
Figure 1 Overview of canonical TGF-β/Smad signalling in tissue fibrosis Figure 1 | Overview of canonical TGF-β/Smad signalling in tissue fibrosis. Once.
Tumor necrosis factor: Biology and therapeutic inhibitors
Chadrick E. Denlinger, MD, John S. Ikonomidis, MD, PhD, Carolyn E
Macrophage heterogeneity, phenotypes, and roles in renal fibrosis
Volume 66, Issue 2, Pages (February 2017)
Bhagelu Ram Achyut, Li Yang  Gastroenterology 
Molecular mechanisms of diabetic renal hypertrophy
Kidney disease and vitamin D levels: 25-hydroxyvitamin D, 1,25-dihydroxyvitamin D, and VDR activation  Adriana S. Dusso  Kidney International Supplements 
Volume 93, Issue 1, Pages (January 2018)
Renoprotective effects of vitamin D analogs
Figure 4 TNFSF inflammatory activities in tissue cells
TGF-² Signaling Pathway in Lung Adenocarcinoma Invasion
JAK/STAT signaling in renal diseases
Mechanisms of Tissue Injury in Lupus Nephritis
Hans-Joachim Anders, Volker Vielhauer, Detlef Schlöndorff 
Paricalcitol attenuates cyclosporine-induced kidney injury in rats
Susan E. Quaggin, András Kapus  Kidney International 
Figure 4 Mechanisms of leptin function on kidney injury
Volume 68, Issue 1, Pages (July 2005)
Volume 80, Issue 10, Pages (November 2011)
Jo-Ellen Murphy, Caroline Robert, Thomas S. Kupper 
Dietmar M.W. Zaiss, William C. Gause, Lisa C. Osborne, David Artis 
Heterozygous disruption of activin receptor–like kinase 1 is associated with increased renal fibrosis in a mouse model of obstructive nephropathy  José.
Volume 81, Issue 3, Pages (February 2012)
Volume 81, Issue 12, Pages (June 2012)
Pro-endometriotic niche in endometriosis
Inflammatory health effects of indoor and outdoor particulate matter
Wnt/β-catenin signaling and kidney fibrosis
Psoriasis and Systemic Inflammatory Diseases: Potential Mechanistic Links between Skin Disease and Co-Morbid Conditions  Batya B. Davidovici, Naveed Sattar,
Volume 84, Issue 5, Pages (November 2013)
Hepatocyte growth factor suppresses interstitial fibrosis in a mouse model of obstructive nephropathy  Shinya Mizuno, Kunio Matsumoto, Toshikazu Nakamura 
Volume 64, Pages S39-S45 (October 2003)
Thrombosis and Inflammatory Bowel Disease
Volume 70, Issue 7, Pages (October 2006)
Anti-inflammatory Agents: Present and Future
The renin–angiotensin–aldosterone system in peritoneal dialysis: is what is good for the kidney also good for the peritoneum?  Sharon J. Nessim, Jeffrey.
Reciprocal functions of hepatocyte growth factor and transforming growth factor-β1 in the progression of renal diseases: A role for CD44?  Sandrine Florquin,
Volume 64, Pages S64-S70 (October 2003)
Volume 57, Issue 1, Pages (January 2000)
Volume 92, Issue 3, Pages (September 2017)
Blockade of NFκB activation and renal inflammation by ultrasound-mediated gene transfer of Smad7 in rat remnant kidney  Yee-Yung Ng, Chun-Cheng Hou, Wansheng.
Osteopontin in diabetic nephropathy: signpost or road?
Nitric oxide and vascular remodeling: Spotlight on the kidney
Pathogenesis of idiopathic pulmonary fibrosis (IPF).
Robert L. Chevalier, Michael S. Forbes, Barbara A. Thornhill 
Volume 78, Issue 7, Pages (October 2010)
CD4+ T cells: a potential player in renal fibrosis
Tubulointerstitial damage and progression of renal failure
EMT and proteinuria as progression factors
Smad3 and Extracellular Signal-Regulated Kinase 1/2 Coordinately Mediate Transforming Growth Factor-β-Induced Expression of Connective Tissue Growth Factor.
Endothelial cell activation
NODding off in acute kidney injury with progranulin?
Volume 76, Issue 5, Pages (September 2009)
Treating myocardial ischemia-reperfusion injury by targeting endothelial cell transcription  Edward M Boyle, MD, Timothy G Canty, MD, Elizabeth N Morgan,
Volume 70, Issue 2, Pages (July 2006)
Macrophages, Immunity, and Metabolic Disease
Tumor necrosis factor: Biology and therapeutic inhibitors
Notch signaling from tumor cells: A new mechanism of angiogenesis
Renoprotection with vitamin D: Specific for diabetic nephropathy?
Dietmar M.W. Zaiss, William C. Gause, Lisa C. Osborne, David Artis 
Predicting Risk of Radiation-Induced Lung Injury
Presentation transcript:

Obstructive nephropathy: Insights from genetically engineered animals Jean-Loup Bascands, Joost P. Schanstra  Kidney International  Volume 68, Issue 3, Pages 925-937 (September 2005) DOI: 10.1111/j.1523-1755.2005.00486.x Copyright © 2005 International Society of Nephrology Terms and Conditions

Figure 1 Overview of the different stages of the development of obstructive nephropathy. Rapidly after unilateral ureteral obstruction (UUO) cytokines and growth factors secreted by tubular epithelial cells induce cell infiltration (mainly macrophages) that in their turn also secrete growth factors and cytokines inducing a disequilibrium between apoptosis and proliferation of tubular cells, as well as inducing fibroblast activation and proliferation. Fibroblasts either infiltrate from the circulation into the interstitium, appear by epithelial-mesenchymal transition (EMT) or appear by proliferation of the few resident fibroblasts. Activated fibroblasts secrete the extracellular matrix (ECM) that is starting to accumulate into the interstitium as soon as myofibroblasts appear. As the obstruction continuous, ECM deposition becomes massive and uncontrolled apoptosis of tubular cells results in tubular atrophy. The involvement of different molecules in these stages of obstructive nephropathy, studied using genetically modified animals, is described in the text and in the subsequent figures. Kidney International 2005 68, 925-937DOI: (10.1111/j.1523-1755.2005.00486.x) Copyright © 2005 International Society of Nephrology Terms and Conditions

Figure 2 Hypothetic representation of cellular infiltration in unilateral ureteral obstruction (UUO) based on data obtained with knockout mice. UUO induces macrophage infiltration in the tubulointerstitium. In UUO, the interaction between L-selectin and sulfatide seems to mediate the initial contact (rolling) between macrophages and the vascular endothelium. These rolling macrophages are exposed to adhesion molecules like chemokines and osteopontin expressed on endothelial cells. The chemokines bind to chemokine receptors (in UUO mainly the CCR1) on the macrophages. Osteopontin binds to the CD44 receptor. This results in firm adhesion and transendothelial migration of macrophages. The role of other important molecules [including intercellular adhesion molecule-1 (ICAM-1), vascular cell adhesion molecule-1 (VCAM-1), and monocyte chemoattractant protein-1 (MCP-1)] involved in cellular infiltration shown to be induced in UUO has not been studied yet and should give a more complete picture of cellular infiltration in UUO. Kidney International 2005 68, 925-937DOI: (10.1111/j.1523-1755.2005.00486.x) Copyright © 2005 International Society of Nephrology Terms and Conditions

Figure 3 Proposed role of epithelial-mesenchymal transition (EMT) in ureteral obstruction. Unilateral ureteral obstruction (UUO) induces EMT which contributes significantly (36%) to the appearance of interstitial myofibroblasts. Myofibroblasts are the main source of profibrotic and inflammatory cytokines leading to matrix accumulation. The remainder of the fibroblasts comes from bone marrow (14% to 15%) and probably local fibroblast proliferation and activation98. EMT involves four key events leading to epithelial cell migration and invasion into the interstitium and transition into fibroblasts49. Interestingly one of these events, tubular basement membrane degradation, is blocked in tissue plasminogen activator (tPA) knockout mice (in contrast to what was expected for a tPA knockout) thereby blocking EMT and subsequent tubulointerstitial fibrosis. Abbreviations are MMP-9, matrix metalloproteinase-9; TGF-β, transforming growth factor-β; α-SMA, α-smooth muscle actin. Kidney International 2005 68, 925-937DOI: (10.1111/j.1523-1755.2005.00486.x) Copyright © 2005 International Society of Nephrology Terms and Conditions

Figure 4 The role and connections between the renin-angiotensin and kinin-kallikrein system and proteolytic systems in unilateral ureteral obstruction (UUO). The profibrotic role of angiotensin II (AII) in UUO (see Figure 5) is counterbalanced by the production of bradykinin. It is proposed that during UUO, bradykinin is activating the G protein-coupled B2 receptor which activates the proteolytic plasminogen activator (PA), combined tissue and urokinase plasminogen activator activity) system [either directly or by inhibition of plasminogen activator inhibitor-1 (PAI-1)] which in turn switches on matrix metalloproteinases (MMPs) promoting matrix degradation (bold dashed line). Angiotensin-converting enzyme (ACE) inhibition thus blocks production of profibrotic Ang II and promotes accumulation of the antifibrotic peptide bradykinin. The role of the proteolytic enzymes in matrix degradation in UUO is depicted in the gray area in figure. Both pro- and antifibrotic properties of this system have now been proposed: (1) in UUO it was shown that plasmin can generate both profibrotic transforming growth factor-β (TGF-β) and antifibrotic hepatocyte growth factor (HGF), (2) activation of MMPs degrades the extracellular matrix (ECM) but stimulates endothelial-mesencymal transition (EMT), and (3) PAI-1 has been shown, besides its role to inhibit PA activity, to stimulate cellular infiltration in UUO. Kidney International 2005 68, 925-937DOI: (10.1111/j.1523-1755.2005.00486.x) Copyright © 2005 International Society of Nephrology Terms and Conditions

Figure 5 The central role for nuclear factor-κB (NF-κB) in angiotensin II profibrotic effects in unilateral ureteral obstruction (UUO). Angiotensin II (AII) accumulation during UUO stimulates two G protein-coupled receptors, the angiotensin type 1 (AT1) and angiotensin type 2 (AT2). Although AT2 knockout mice display less tubulointerstitial fibrosis, stimulation of this receptor and the AT1 receptor induces NF-κB activation which translocates to the nucleus and activates a vast panel of proinflammatory and profibrotic cytokines leading to sustained inflammation and matrix production. NF-κB activation is also leading to at least two positive autocrine regulation loops by the increased expression of tumor necrosis factor-α (TNF-α) and angiotensinogen. IκB is inhibitor of κB. Kidney International 2005 68, 925-937DOI: (10.1111/j.1523-1755.2005.00486.x) Copyright © 2005 International Society of Nephrology Terms and Conditions

Figure 6 Proposed schematic representation of the role of transforming growth factor-β (TGF-β) in unilateral ureteral obstruction (UUO). TGF-β has been shown to be an important regulator of many of the processes leading to tubulointerstitial fibrosis, including epithelial-mesencymal transition (EMT), cellular infiltration, and apoptosis leading to tubular atrophy and interstitial accumulation of extracellular matrix (ECM) proteins. Binding of TGF-β to the transmembrane serine/threonine kinase receptor type II (RII) leads to the recruitment phosphorylation and activation of the type I receptor kinase (RI), which, in turn, phosphorylates Smad2 and Smad 3 proteins thus able to bind Smad4 and translocate to the nucleus where they regulate the transcription of target genes by binding to their specific promoter sequences. In addition to this classic TGF-β signaling pathway there is now increasing evidence that Smad-independent pathways like activation of RhoA and p38 mitogen-activated protein kinases (MAPKs) are involved in EMT. Smad3 knockout mice are protected against UUO-induced tubulointerstitial fibrosis and showed the existence of a TGF-β autocrine loop during UUO. Furthermore, UUO on knockout mice showed that the production of active TGF-β is controlled by integrins, plasmin, and decorin. Kidney International 2005 68, 925-937DOI: (10.1111/j.1523-1755.2005.00486.x) Copyright © 2005 International Society of Nephrology Terms and Conditions