scispace - formally typeset
Search or ask a question

Showing papers by "Jihwan Park published in 2018"


Journal ArticleDOI
18 May 2018-Science
TL;DR: It is inferred that inherited kidney diseases that arise from distinct genetic mutations but share the same phenotypic manifestation originate from the same differentiated cell type, and that the collecting duct in kidneys of adult mice generates a spectrum of cell types through a newly identified transitional cell.
Abstract: Our understanding of kidney disease pathogenesis is limited by an incomplete molecular characterization of the cell types responsible for the organ’s multiple homeostatic functions. To help fill this knowledge gap, we characterized 57,979 cells from healthy mouse kidneys by using unbiased single-cell RNA sequencing. On the basis of gene expression patterns, we infer that inherited kidney diseases that arise from distinct genetic mutations but share the same phenotypic manifestation originate from the same differentiated cell type. We also found that the collecting duct in kidneys of adult mice generates a spectrum of cell types through a newly identified transitional cell. Computational cell trajectory analysis and in vivo lineage tracing revealed that intercalated cells and principal cells undergo transitions mediated by the Notch signaling pathway. In mouse and human kidney disease, these transitions were shifted toward a principal cell fate and were associated with metabolic acidosis.

751 citations


Journal ArticleDOI
TL;DR: Kidney compartment–specific eQTL analysis goes beyond GWAS to reveal causal genes and pathways involved in renal disease development, and reduces Dab2 expression in renal tubules protected mice from CKD.
Abstract: Chronic kidney disease (CKD), a condition in which the kidneys are unable to clear waste products, affects 700 million people globally. Genome-wide association studies (GWASs) have identified sequence variants for CKD; however, the biological basis of these GWAS results remains poorly understood. To address this issue, we created an expression quantitative trait loci (eQTL) atlas for the glomerular and tubular compartments of the human kidney. Through integrating the CKD GWAS with eQTL, single-cell RNA sequencing and regulatory region maps, we identified novel genes for CKD. Putative causal genes were enriched for proximal tubule expression and endolysosomal function, where DAB2, an adaptor protein in the TGF-β pathway, formed a central node. Functional experiments confirmed that reducing Dab2 expression in renal tubules protected mice from CKD. In conclusion, compartment-specific eQTL analysis is an important avenue for the identification of novel genes and cellular pathways involved in CKD development and thus potential new opportunities for its treatment.

157 citations


Journal ArticleDOI
TL;DR: It is 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.
Abstract: 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. Transcript analysis of mouse kidney disease models, including folic-acid (FA)–induced nephropathy, unilateral ureteral obstruction (UUO), or apolipoprotein L1 (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 FA-induced kidney fibrosis. Tubule-specific genetic deletion of Notch1 and global knockout 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. Tubule–specific deletion of Tfam resulted in fibrosis. In summary, Jag1 and Notch2 play a key role in kidney fibrosis development by regulating Tfam expression and metabolic reprogramming.

45 citations


Posted ContentDOI
29 Mar 2018-bioRxiv
TL;DR: 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 and played a key role in kidney fibrosis development by regulating Tfam expression and metabolic reprogramming.
Abstract: 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.

15 citations


Posted ContentDOI
Rany M. Salem1, Jennifer N. Todd2, Jennifer N. Todd3, Jennifer N. Todd4, Niina Sandholm5, Joanne B. Cole4, Joanne B. Cole3, Joanne B. Cole2, Wei-Min Chen6, Darrel Andrews7, Marcus G. Pezzolesi8, Paul M. McKeigue9, Linda T Hiraki10, Chengxiang Qiu11, Viji Nair12, Chen Di Liao10, Jing Jing Cao10, Erkka Valo5, Suna Onengut-Gumuscu6, Adam M. Smiles13, Stuart J. McGurnaghan9, Jani K. Haukka5, Valma Harjutsalo, Eoin P. Brennan7, Natalie R. van Zuydam14, Natalie R. van Zuydam15, Emma Ahlqvist16, Ross Doyle7, Tarunveer S. Ahluwalia17, Maria Lajer17, Maria Hughes7, Jihwan Park11, Jan Skupien13, Athina Spiliopoulou9, Andrew S.K. Liu12, Rajasree Menon12, Carine M. Boustany-Kari18, Hyun Min Kang12, Robert G. Nelson19, Ronald Klein20, Barbara E.K. Klein20, Kristine E. Lee20, Xiaoyu Gao21, Michael Mauer22, Silvia Maeastroni, Maria Luiza Caramori22, Ian H. de Boer23, Rachel G. Miller24, Jingchuan Guo24, Andrew P. Boright10, David-Alexandre Trégouët25, David-Alexandre Trégouët26, Beata Gyorgy25, Beata Gyorgy26, Janet K. Snell-Bergeon27, David M. Maahs28, Shelley B. Bull29, Angelo J. Canty30, Colin N. A. Palmer31, Lars Stechemesser32, Bernhard Paulweber32, Raimund Weitgasser, Jelizaveta Sokolovska33, Vita Rovīte34, Valdis Pīrāgs33, Edita Prakapiene35, Lina Radzeviciene, Rasa Verkauskiene, Nicolae Mircea Panduru5, Nicolae Mircea Panduru36, Leif Groop5, Leif Groop16, Mark I. McCarthy14, Mark I. McCarthy15, Harvest F. Gu37, Harvest F. Gu38, Anna Möllsten39, Henrik Falhammar40, Henrik Falhammar38, Kerstin Brismar38, Kerstin Brismar40, Dcct, F Martin17, F Martin41, Peter Rossing24, Tina Costacou, Gianpaolo Zerbini42, Gianpaolo Zerbini43, Michel Marre44, Michel Marre45, Samy Hadjadj46, Amy J. McKnight5, Carol Forsblom46, Gareth J. McKay7, Catherine Godson46, A. Peter Maxwell12, Matthias Kretzler11, Katalin Susztak9, Helen M. Colhoun13, Andrzej Krolewski10, Andrew D. Paterson, Per-Henrik Groop6, Stephen S. Rich3, Stephen S. Rich2, Joel N. Hirschhorn, Jose C. Florez 
19 Dec 2018-bioRxiv
TL;DR: The 16 DKD-associated loci provide novel insights into the pathogenesis of DKD, identifying potential biological targets for prevention and treatment.
Abstract: Diabetic kidney disease (DKD) is a heritable but poorly understood complication of diabetes. To identify genetic variants predisposing to DKD, we performed genome-wide association analyses in 19,406 individuals with type 1 diabetes (T1D) using a spectrum of DKD definitions basedon albuminuria and renal function. We identified 16 genome-wide significant loci. The variant with the strongest association (rs55703767) is a common missense mutation in the collagen type IV alpha 3 chain (COL4A3) gene, which encodes a major structural component of the glomerular basement membrane (GBM) implicated in heritable nephropathies. The rs55703767 minor allele (Asp326Tyr) is protective against several definitions of DKD, including albuminuria and end-stage renal disease. Three other loci are in or near genes with known or suggestive involvement in DKD (BMP7) or renal biology (COLEC11 and DDR1). The 16 DKD-associated loci provide novel insights into the pathogenesis of DKD, identifying potential biological targets for prevention and treatment.

11 citations


Posted ContentDOI
Xuran Wang1, Jihwan Park1, Katalin Susztak1, Nan Zhang1, Mingyao Li1 
26 Jun 2018-bioRxiv
TL;DR: When applied to pancreatic islet and whole kidney expression data in human, mouse, and rats, MuSiC outperformed existing methods, especially for tissues with closely related cell types.
Abstract: We present MuSiC, a method that utilizes cell-type specific gene expression from single-cell RNA sequencing (RNA-seq) data to characterize cell type compositions from bulk RNA-seq data in complex tissues. When applied to pancreatic islet and whole kidney expression data in human, mouse, and rats, MuSiC outperformed existing methods, especially for tissues with closely related cell types. MuSiC enables characterization of cellular heterogeneity of complex tissues for identification of disease mechanisms.

3 citations


Posted ContentDOI
29 Jun 2018-bioRxiv
TL;DR: Genome-wide cytosine methylation and gene expression profiling showed that Dnmt1-mediated DNA methylation is essential for kidney development by preventing regression of progenitor cells into a primitive undifferentiated state and demethylation of transposable elements.
Abstract: Cytosine methylation (5mC) plays a key role in maintaining progenitor cell self-renewal and differentiation. Here, we analyzed the role of 5mC in kidney development by genome-wide methylation, expression profiling, and by systematic genetic targeting of DNA methyltransferases (Dnmt) and Ten-eleven translocation methylcytosine hydroxylases (Tet). In mice, nephrons differentiate from Six2+ progenitor cells, therefore we created animals with genetic deletion of Dnmt1, 3a, 3b, Tet1, and Tet2 in the Six2+ population (Six2Cre/Dnmt1flox/flox, Six2Cre/Dnmt3aflox/flox, Six2Cre/Dnmt3bflox/flox, Six2Cre/Tet2flox/flox and Tet1-/-). Animals with conditional deletion of Dnmt3a, 3b, Tet1 and Tet2 showed no significant structural or functional renal abnormalities. On the other hand, Six2Cre/Dnmt1flox/flox mice died within 24hrs of birth. Dnmt1 knock-out animals had small kidneys and significantly reduced nephron number. Genome-wide methylation analysis indicated marked loss of methylation mostly on transposable elements. RNA sequencing detected endogenous retroviral (ERV) gene transcripts and early embryonic genes. Increase in levels of interferon (and RIG-I signaling) and apoptosis (Trp53) in response to ERV activity likely contributed to the phenotype development. Once epithelial differentiation was established, loss of Dnmt1, 3a, 3b, Tet1 or Tet2 in glomerular epithelial cells did not lead to functional or structural differences at baseline or following toxic glomerular injury. Genome-wide cytosine methylation and gene expression profiling showed that Dnmt1-mediated DNA methylation is essential for kidney development by preventing regression of progenitor cells into a primitive undifferentiated state and demethylation of transposable elements.

2 citations