Jagged1/Notch2 controls kidney fibrosis via Tfam-mediated metabolic reprogramming

While Notch signaling has been proposed to play a key role in fibrosis, the direct molecular pathways targeted by Notch signaling and the precise ligand and receptor pair that are responsible for kidney disease remain poorly defined. In this study, we found that JAG1 and NOTCH2 showed the strongest correlation with the degree of interstitial fibrosis in a genome wide expression analysis of a large cohort of human kidney samples. RNA sequencing analysis of kidneys of mice with folic acid nephropathy, unilateral ureteral obstruction, or APOL1-associated kidney disease indicated that Jag1 and Notch2 levels were higher in all analyzed kidney fibrosis models. Mice with tubule-specific deletion of Jag1 or Notch2 (Kspcre/Jag1flox/flox, and Kspcre/Notch2flox/flox) had no kidney-specific alterations at baseline, but showed protection from folic acid induced kidney fibrosis. Tubule-specific genetic deletion of Notch1 and global knock-out of Notch3 had no effect on fibrosis. In vitro chromatin immunoprecipitation experiments and genome-wide expression studies identified the mitochondrial transcription factor A (Tfam) as a direct Notch target. Re-expression of Tfam in tubule cells prevented Notch-induced metabolic and profibrotic reprogramming. Kidney tubule specific deletion of Tfam resulted in perinatal lethality. In summary, Jag1/Notch2 plays a key role in kidney fibrosis development by regulating Tfam expression and metabolic reprogramming.

[1]  Jakub Toczek,et al.  Genetic deficiency or pharmacological inhibition of miR-33 protects from kidney fibrosis. , 2019, JCI insight.

[2]  M. Mukherjee,et al.  Notch Signaling in Kidney Development, Maintenance, and Disease , 2019, Biomolecules.

[3]  Chengxiang Qiu,et al.  Mitochondrial Damage and Activation of the STING Pathway Lead to Renal Inflammation and Fibrosis. , 2019, Cell metabolism.

[4]  K. Suszták,et al.  Going from acute to chronic kidney injury with FoxO3. , 2019, The Journal of clinical investigation.

[5]  Mingyao Li,et al.  Single-cell transcriptomics of the mouse kidney reveals potential cellular targets of kidney disease , 2018, Science.

[6]  K. Suszták,et al.  The Role of Peroxisome Proliferator-Activated Receptor γ Coactivator 1α (PGC-1α) in Kidney Disease. , 2018, Seminars in nephrology.

[7]  T. Yoo,et al.  PGC-1α Protects from Notch-Induced Kidney Fibrosis Development. , 2017, Journal of the American Society of Nephrology : JASN.

[8]  Chengxiang Qiu,et al.  Human Kidney Tubule-Specific Gene Expression Based Dissection of Chronic Kidney Disease Traits , 2017, EBioMedicine.

[9]  S. Rong,et al.  Antagonism of profibrotic microRNA-21 improves outcome of murine chronic renal allograft dysfunction. , 2017, Kidney international.

[10]  Patrick D. Dummer,et al.  Transgenic expression of human APOL1 risk variants in podocytes induces kidney disease in mice , 2017, Nature Medicine.

[11]  H. Pavenstädt,et al.  Developmental signalling pathways in renal fibrosis: the roles of Notch, Wnt and Hedgehog , 2016, Nature Reviews Nephrology.

[12]  P. Ruiz-Lozano,et al.  Notch-independent RBPJ controls angiogenesis in the adult heart , 2016, Nature Communications.

[13]  John N. Hutchinson,et al.  RNA Sequencing Identifies Novel Translational Biomarkers of Kidney Fibrosis. , 2016, Journal of the American Society of Nephrology : JASN.

[14]  M. Breyer,et al.  The next generation of therapeutics for chronic kidney disease , 2016, Nature Reviews Drug Discovery.

[15]  P. Moulos,et al.  Whole-transcriptome analysis of UUO mouse model of renal fibrosis reveals new molecular players in kidney diseases , 2016, Scientific Reports.

[16]  J. Bonventre,et al.  Kidney tubules: intertubular, vascular, and glomerular cross-talk , 2016, Current opinion in nephrology and hypertension.

[17]  B. Kishore,et al.  Regulation of Vascular and Renal Function by Metabolite Receptors. , 2016, Annual review of physiology.

[18]  M. Bhasin,et al.  PGC1α-dependent NAD biosynthesis links oxidative metabolism to renal protection , 2016, Nature.

[19]  K. Reidy,et al.  Sox9-Positive Progenitor Cells Play a Key Role in Renal Tubule Epithelial Regeneration in Mice. , 2016, Cell reports.

[20]  Kirill Alexandrov,et al.  Performance benchmarking of four cell‐free protein expression systems , 2015, Biotechnology and bioengineering.

[21]  K. Suszták,et al.  Epithelial Plasticity versus EMT in Kidney Fibrosis. , 2016, Trends in molecular medicine.

[22]  K. Shedden,et al.  Tissue transcriptome-driven identification of epidermal growth factor as a chronic kidney disease biomarker , 2015, Science Translational Medicine.

[23]  V. Haase,et al.  Molecular mechanisms of ischemic preconditioning in the kidney. , 2015, American journal of physiology. Renal physiology.

[24]  S. Weiss,et al.  Snail1-induced partial epithelial-to-mesenchymal transition drives renal fibrosis in mice and can be targeted to reverse established disease , 2015, Nature Medicine.

[25]  R. Nishinakamura,et al.  Notch1 and Notch2 in Podocytes Play Differential Roles During Diabetic Nephropathy Development , 2015, Diabetes.

[26]  R. Kalluri,et al.  Epithelial to Mesenchymal Transition induces cell cycle arrest and parenchymal damage in renal fibrosis , 2015, Nature Medicine.

[27]  W. Mitch,et al.  Migration of smooth muscle cells from the arterial anastomosis of arteriovenous fistulas requires Notch activation to form neointima , 2015, Kidney international.

[28]  Grégoire Pau,et al.  Differential effects of targeting Notch receptors in a mouse model of liver cancer , 2015, Hepatology.

[29]  Kumar Sharma,et al.  Defective fatty acid oxidation in renal tubular epithelial cells has a key role in kidney fibrosis development , 2014, Nature Medicine.

[30]  K. Suszták,et al.  Kick it up a notch: Notch signaling and kidney fibrosis , 2014, Kidney international supplements.

[31]  J. Bonventre Primary proximal tubule injury leads to epithelial cell cycle arrest, fibrosis, vascular rarefaction, and glomerulosclerosis , 2014, Kidney international supplements.

[32]  Emma R. Andersson,et al.  Therapeutic modulation of Notch signalling — are we there yet? , 2014, Nature Reviews Drug Discovery.

[33]  W. Mitch,et al.  Blocking Notch in endothelial cells prevents arteriovenous fistula failure despite CKD. , 2014, Journal of the American Society of Nephrology : JASN.

[34]  M. Takagi,et al.  Notch2 activation ameliorates nephrosis , 2014, Nature Communications.

[35]  H. Stunnenberg,et al.  Dynamic binding of RBPJ is determined by Notch signaling status. , 2013, Genes & development.

[36]  C. Ebens,et al.  Blockade of individual Notch ligands and receptors controls graft-versus-host disease. , 2013, The Journal of clinical investigation.

[37]  V. Taylor,et al.  Endocardial to Myocardial Notch-Wnt-Bmp Axis Regulates Early Heart Valve Development , 2013, PloS one.

[38]  K. Suszták,et al.  For Better or Worse: A Niche for Notch in Parietal Epithelial Cell Activation , 2013, Kidney international.

[39]  Na Liu,et al.  Role of epidermal growth factor receptor in acute and chronic kidney injury , 2013, Kidney international.

[40]  K. Suszták,et al.  Repair problems in podocytes: Wnt, Notch, and glomerulosclerosis. , 2012, Seminars in nephrology.

[41]  Johannes E. Schindelin,et al.  Fiji: an open-source platform for biological-image analysis , 2012, Nature Methods.

[42]  Katalin Susztak,et al.  Notch signaling in diabetic nephropathy. , 2012, Experimental cell research.

[43]  M. Mitobe,et al.  Reduced Klotho expression level in kidney aggravates renal interstitial fibrosis. , 2012, American journal of physiology. Renal physiology.

[44]  K. Suszták,et al.  Notch in the kidney: development and disease , 2012, The Journal of pathology.

[45]  M. Bhasin,et al.  PGC-1α promotes recovery after acute kidney injury during systemic inflammation in mice. , 2011, The Journal of clinical investigation.

[46]  H. Schnaper,et al.  Tubulointerstitial injury and the progression of chronic kidney disease , 2011, Pediatric Nephrology.

[47]  K. Suszták,et al.  The story of Notch and chronic kidney disease , 2011, Current opinion in nephrology and hypertension.

[48]  M. Gessler,et al.  Epithelial Notch signaling regulates interstitial fibrosis development in the kidneys of mice and humans. , 2010, The Journal of clinical investigation.

[49]  S. Buch,et al.  Activation of Notch signaling pathway in HIV-associated nephropathy , 2010, AIDS.

[50]  K. Suszták,et al.  Expression of Notch pathway proteins correlates with albuminuria, glomerulosclerosis, and renal function. , 2010, Kidney international.

[51]  K. Suszták,et al.  Getting a Notch Closer to Understanding Diabetic Kidney Disease , 2010, Diabetes.

[52]  Feng-Sheng Wang,et al.  Modulation of Notch-1 Signaling Alleviates Vascular Endothelial Growth Factor–Mediated Diabetic Nephropathy , 2010, Diabetes.

[53]  R. Foley,et al.  Chronic kidney disease awareness, screening and prevention: Rationale for the design of a public education program , 2010, Nephrology.

[54]  F. Costantini,et al.  Patterning a complex organ: branching morphogenesis and nephron segmentation in kidney development. , 2010, Developmental cell.

[55]  R. Foley Temporal trends in the burden of chronic kidney disease in the United States , 2010, Current opinion in nephrology and hypertension.

[56]  T. Honjo,et al.  Two opposing roles of RBP-J in Notch signaling. , 2010, Current topics in developmental biology.

[57]  Lawrence Wang,et al.  Effect of Notch activation on the regenerative response to acute renal failure. , 2010, American journal of physiology. Renal physiology.

[58]  Raphael Kopan,et al.  The Canonical Notch Signaling Pathway: Unfolding the Activation Mechanism , 2009, Cell.

[59]  H. Kuwana,et al.  Expression and function of the Delta-1/Notch-2/Hes-1 pathway during experimental acute kidney injury. , 2008, Kidney international.

[60]  K. Suszták,et al.  The Notch pathway in podocytes plays a role in the development of glomerular disease , 2008, Nature Medicine.

[61]  F. Gonzalez,et al.  PPARα Protects Proximal Tubular Cells from Acute Fatty Acid Toxicity , 2007 .

[62]  Robert A. Dean,et al.  Effects of a γ-secretase inhibitor in a randomized study of patients with Alzheimer disease , 2006, Neurology.

[63]  P. Tariot,et al.  Effects of a gamma-secretase inhibitor in a randomized study of patients with Alzheimer disease. , 2006, Neurology.

[64]  Robert D. Kirch,et al.  Jagged1 signals in the postnatal subventricular zone are required for neural stem cell self‐renewal , 2005, The EMBO journal.

[65]  Phil Jones,et al.  Notch signaling regulates the differentiation of post-mitotic intestinal epithelial cells. , 2005, Genes & development.

[66]  J. Zavadil,et al.  Integration of TGF‐β/Smad and Jagged1/Notch signalling in epithelial‐to‐mesenchymal transition , 2004 .

[67]  J. Zavadil,et al.  Integration of TGF-beta/Smad and Jagged1/Notch signalling in epithelial-to-mesenchymal transition. , 2004, The EMBO journal.

[68]  B. Mccright,et al.  Notch signaling in kidney development , 2003, Current opinion in nephrology and hypertension.

[69]  D. Podolsky,et al.  Intestinal Trefoil Factor Induces Decay-Accelerating Factor Expression and Enhances the Protective Activities Against Complement Activation in Intestinal Epithelial Cells1 , 2001, The Journal of Immunology.

[70]  K. Nath,et al.  Tubulointerstitial changes as a major determinant in the progression of renal damage. , 1992, American journal of kidney diseases : the official journal of the National Kidney Foundation.