Unbiased Human Kidney Tissue Proteomics Identifies Matrix Metalloproteinase 7 as a Kidney Disease Biomarker

Significance Statement Although gene expression changes have been characterized in human diabetic kidney disease (DKD), unbiased tissue proteomics information for this condition is lacking. The authors conducted an unbiased aptamer-based proteomic analysis of samples from patients with DKD and healthy controls, identifying proteins with levels that associate with kidney function (eGFR) or fibrosis, after adjusting for key covariates. Overall, tissue gene expression only modestly correlated with tissue protein levels. Kidney protein and RNA levels of matrix metalloproteinase 7 (MMP7) strongly correlated with fibrosis and with eGFR. Single-cell RNA sequencing indicated that kidney tubule cells are an important source of MMP7. Furthermore, plasma MMP7 levels predicted future kidney function decline. These findings identify kidney tissue MMP7 as a biomarker of fibrosis and blood MMP7 as a biomarker for future kidney function decline. Background Diabetic kidney disease (DKD) is responsible for close to half of all ESKD cases. Although unbiased gene expression changes have been extensively characterized in human kidney tissue samples, unbiased protein-level information is not available. Methods We collected human kidney samples from 23 individuals with DKD and ten healthy controls, gathered associated clinical and demographics information, and implemented histologic analysis. We performed unbiased proteomics using the SomaScan platform and quantified the level of 1305 proteins and analyzed gene expression levels by bulk RNA and single-cell RNA sequencing (scRNA-seq). We validated protein levels in a separate cohort of kidney tissue samples as well as in 11,030 blood samples. Results Globally, human kidney transcript and protein levels showed only modest correlation. Our analysis identified 14 proteins with kidney tissue levels that correlated with eGFR and found that the levels of 152 proteins correlated with interstitial fibrosis. Of the identified proteins, matrix metalloprotease 7 (MMP7) showed the strongest association with both fibrosis and eGFR. The correlation between tissue MMP7 protein expression and kidney function was validated in external datasets. The levels of MMP7 RNA correlated with fibrosis in the primary and validation datasets. Findings from scRNA-seq pointed to proximal tubules, connecting tubules, and principal cells as likely cellular sources of increased tissue MMP7 expression. Furthermore, plasma MMP7 levels correlated not only with kidney function but also associated with prospective kidney function decline. Conclusions Our findings, which underscore the value of human kidney tissue proteomics analysis, identify kidney tissue MMP7 as a diagnostic marker of kidney fibrosis and blood MMP7 as a biomarker for future kidney function decline.

[1]  A. Galecki,et al.  Neuroblastoma suppressor of tumorigenicity 1 is a circulating protein associated with progression to end-stage kidney disease in diabetes , 2022, Science Translational Medicine.

[2]  Christopher D. Brown,et al.  Epigenomic and transcriptomic analyses define core cell types, genes and targetable mechanisms for kidney disease , 2022, Nature Genetics.

[3]  A. Galecki,et al.  Results of untargeted analysis using the SOMAscan proteomics platform indicates novel associations of circulating proteins with risk of progression to kidney failure in diabetes. , 2022, Kidney international.

[4]  E. Boerwinkle,et al.  APOL1 Kidney Risk Variants and Proteomics , 2022, Clinical journal of the American Society of Nephrology : CJASN.

[5]  Brad T. Sherman,et al.  DAVID: a web server for functional enrichment analysis and functional annotation of gene lists (2021 update) , 2022, Nucleic Acids Res..

[6]  N. Roetker,et al.  US Renal Data System 2021 Annual Data Report: Epidemiology of Kidney Disease in the United States , 2022, American Journal of Kidney Diseases.

[7]  M. Merchant,et al.  Advances in proteomic profiling of pediatric kidney diseases , 2022, Pediatric Nephrology.

[8]  N. Powe,et al.  New Creatinine- and Cystatin C-Based Equations to Estimate GFR without Race. , 2021, The New England journal of medicine.

[9]  N. Chatterjee,et al.  Polygenic Risk Scores for Kidney Function and Their Associations with Circulating Proteome, and Incident Kidney Diseases , 2021, Journal of the American Society of Nephrology : JASN.

[10]  Rachel S. G. Sealfon,et al.  A reference tissue atlas for the human kidney , 2021, bioRxiv.

[11]  E. Boerwinkle,et al.  Proteins Associated with Risk of Kidney Function Decline in the General Population , 2021, Journal of the American Society of Nephrology : JASN.

[12]  K. Suszták,et al.  Renal Histologic Analysis Provides Complementary Information to Kidney Function Measurement for Patients with Early Diabetic or Hypertensive Disease , 2021, Journal of the American Society of Nephrology : JASN.

[13]  T. Libermann,et al.  Circulating proteins protect against renal decline and progression to end-stage renal disease in patients with diabetes , 2021, Science Translational Medicine.

[14]  E. Boerwinkle,et al.  The ARIC (Atherosclerosis Risk In Communities) Study: JACC Focus Seminar 3/8. , 2021, Journal of the American College of Cardiology.

[15]  Michael J. Randles,et al.  Identification of an Altered Matrix Signature in Kidney Aging and Disease , 2021, Journal of the American Society of Nephrology : JASN.

[16]  R. Townsend,et al.  The Role of Glomerular Epithelial Injury in Kidney Function Decline in Patients With Diabetic Kidney Disease in the TRIDENT Cohort , 2021, Kidney international reports.

[17]  Christopher D. Brown,et al.  Mapping the genetic architecture of human traits to cell types in the kidney identifies mechanisms of disease and potential treatments , 2020, bioRxiv.

[18]  J. Skupień,et al.  Profibrotic Circulating Proteins and Risk of Early Progressive Renal Decline in Patients With Type 2 Diabetes With and Without Albuminuria , 2020, Diabetes Care.

[19]  Youhua Liu,et al.  The Many Faces of Matrix Metalloproteinase-7 in Kidney Diseases , 2020, Biomolecules.

[20]  Stephen A. Williams,et al.  Plasma protein patterns as comprehensive indicators of health , 2019, Nature Medicine.

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

[22]  Sushrut S. Waikar,et al.  The single-cell transcriptomic landscape of early human diabetic nephropathy , 2019, Proceedings of the National Academy of Sciences.

[23]  C. Cervellati,et al.  TRAIL, OPG, and TWEAK in kidney disease: biomarkers or therapeutic targets? , 2019, Clinical science.

[24]  Jennifer K. Sun,et al.  A Signature of Circulating Inflammatory Proteins and Development of End Stage Renal Disease in Diabetes , 2019, Nature Medicine.

[25]  Z. Botta‐Dukát,et al.  Silhouette width using generalized mean—A flexible method for assessing clustering efficiency , 2018, bioRxiv.

[26]  Christopher D. Brown,et al.  Renal compartment-specific genetic variation analyses identify new pathways in chronic kidney disease , 2018, Nature Medicine.

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

[28]  R. V. van Lier,et al.  Proteomic Analyses of Human Regulatory T Cells Reveal Adaptations in Signaling Pathways that Protect Cellular Identity , 2018, Immunity.

[29]  Shu-Yan Liu,et al.  Clinical significance of serum interleukin‐8 and soluble tumor necrosis factor‐like weak inducer of apoptosis levels in patients with diabetic nephropathy , 2018, Journal of diabetes investigation.

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

[31]  Ben Ke,et al.  Corrigendum: Matrix Metalloproteinase-7 and Kidney Fibrosis , 2017, Front. Physiol..

[32]  Youhua Liu,et al.  Matrix Metalloproteinase-7 Is a Urinary Biomarker and Pathogenic Mediator of Kidney Fibrosis. , 2017, Journal of the American Society of Nephrology : JASN.

[33]  Yi Luan,et al.  Biopsy transcriptome expression profiling to identify kidney transplants at risk of chronic injury: a multicentre, prospective study , 2016, The Lancet.

[34]  F. Kronenberg,et al.  Multinational Assessment of Accuracy of Equations for Predicting Risk of Kidney Failure: A Meta-analysis. , 2016, JAMA.

[35]  A. Quyyumi,et al.  Soluble Urokinase Receptor and Chronic Kidney Disease. , 2015, The New England journal of medicine.

[36]  M. Sánchez-Niño,et al.  TWEAK and the progression of renal disease: clinical translation. , 2014, Nephrology, dialysis, transplantation : official publication of the European Dialysis and Transplant Association - European Renal Association.

[37]  O. Troyanskaya,et al.  Defining cell-type specificity at the transcriptional level in human disease , 2013, Genome research.

[38]  P. Kellman,et al.  MultiContrast Delayed Enhancement (MCODE) improves detection of subendocardial myocardial infarction by late gadolinium enhancement cardiovascular magnetic resonance: a clinical validation study , 2012, Journal of Cardiovascular Magnetic Resonance.

[39]  R. Bilous,et al.  KDOQI Clinical Practice Guideline for Diabetes and CKD: 2012 Update. , 2012, American journal of kidney diseases : the official journal of the National Kidney Foundation.

[40]  T. Mayadas,et al.  Circulating TNF receptors 1 and 2 predict ESRD in type 2 diabetes. , 2012, Journal of the American Society of Nephrology : JASN.

[41]  Youhua Liu,et al.  Matrix metalloproteinase-7 as a surrogate marker predicts renal Wnt/β-catenin activity in CKD. , 2012, Journal of the American Society of Nephrology : JASN.

[42]  Youhua Liu Cellular and molecular mechanisms of renal fibrosis , 2011, Nature Reviews Nephrology.

[43]  M. Selbach,et al.  Global quantification of mammalian gene expression control , 2011, Nature.

[44]  N. Tangri,et al.  A predictive model for progression of chronic kidney disease to kidney failure. , 2011, JAMA.

[45]  P. Boutros,et al.  Molecular markers of injury in kidney biopsy specimens of patients with lupus nephritis. , 2011, The Journal of molecular diagnostics : JMD.

[46]  Rachel Ostroff,et al.  Aptamer-Based Multiplexed Proteomic Technology for Biomarker Discovery , 2010, PLoS ONE.

[47]  Jai Radhakrishnan,et al.  Pathologic classification of diabetic nephropathy. , 2010, Journal of the American Society of Nephrology : JASN.

[48]  D. Celermajer,et al.  Serum MMP-7 is increased in diabetic renal disease and diabetic diastolic dysfunction. , 2010, Diabetes research and clinical practice.

[49]  C. Schmid,et al.  A new equation to estimate glomerular filtration rate. , 2009, Annals of internal medicine.

[50]  S. Horvath,et al.  WGCNA: an R package for weighted correlation network analysis , 2008, BMC Bioinformatics.

[51]  T. Werner,et al.  Improved Elucidation of Biological Processes Linked to Diabetic Nephropathy by Single Probe-Based Microarray Data Analysis , 2008, PloS one.

[52]  M. Fasshauer,et al.  Serum levels of the atherosclerosis biomarker sTWEAK are decreased in type 2 diabetes and end-stage renal disease. , 2008, Atherosclerosis.

[53]  M. Sánchez-Niño,et al.  The cytokine TWEAK modulates renal tubulointerstitial inflammation. , 2008, Journal of the American Society of Nephrology : JASN.

[54]  Gary D. Bader,et al.  An automated method for finding molecular complexes in large protein interaction networks , 2003, BMC Bioinformatics.

[55]  Z. Werb,et al.  New functions for the matrix metalloproteinases in cancer progression , 2002, Nature Reviews Cancer.

[56]  Y. Hsu,et al.  TWEAK, a New Secreted Ligand in the Tumor Necrosis Factor Family That Weakly Induces Apoptosis* , 1997, The Journal of Biological Chemistry.