Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Review Article
  • Published:

The tissue proteome in the multi-omic landscape of kidney disease

Abstract

Kidney research is entering an era of ‘big data’ and molecular omics data can provide comprehensive insights into the molecular footprints of cells. In contrast to transcriptomics, proteomics and metabolomics generate data that relate more directly to the pathological symptoms and clinical parameters observed in patients. Owing to its complexity, the proteome still holds many secrets, but has great potential for the identification of drug targets. Proteomics can provide information about protein synthesis, modification and degradation, as well as insight into the physical interactions between proteins, and between proteins and other biomolecules. Thus far, proteomics in nephrology has largely focused on the discovery and validation of biomarkers, but the systematic analysis of the nephroproteome can offer substantial additional insights, including the discovery of mechanisms that trigger and propagate kidney disease. Moreover, proteome acquisition might provide a diagnostic tool that complements the assessment of a kidney biopsy sample by a pathologist. Such applications are becoming increasingly feasible with the development of high-throughput and high-coverage technologies, such as versatile mass spectrometry-based techniques and protein arrays, and encourage further proteomics research in nephrology.

Key points

  • Proteomics analyses all the physical and chemical properties of proteins using a wide variety of technical approaches.

  • The versatility of proteome capture modes gives deep mechanistic insights into clinical samples and animal models of glomerular disease.

  • Both biomarker discovery and biopsy interrogation are key applications that have been propelled by increasingly simplified and sensitive sample preparation methods.

  • Given their diversified landscape, the analysis of post-translational modifications can uncover novel mechanisms of protein regulation.

  • Computational integration and mathematical modelling of molecular processes — tools largely developed for oncology — can be adapted to kidney proteomics research.

  • An increasingly clinical focus of proteomics analyses in combination with other omics technologies will aid or even drive novel discoveries of disease mechanisms and biomarkers in kidney disease.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: The proteome in the omics landscape.
Fig. 2: Basic principles of mass spectrometry-derived proteomics workflows.
Fig. 3: Pathophysiological protein signatures in kidney disease.
Fig. 4: Podocyte proteome and the potential of multi-layered proteomics.
Fig. 5: Modelling strategies and integration of previous knowledge.
Fig. 6: Signalling models of proteome-guided pharmacological intervention.

Similar content being viewed by others

References

  1. Lindenmeyer, M. T. & Kretzler, M. Renal biopsy-driven molecular target identification in glomerular disease. Pflugers Arch. 469, 1021–1028 (2017).

    CAS  PubMed  Google Scholar 

  2. Kiryluk, K. et al. Precision medicine for acute kidney injury (AKI): redefining AKI by agnostic kidney tissue interrogation and genetics. Semin. Nephrol. 38, 40–51 (2018).

    PubMed  PubMed Central  Google Scholar 

  3. Saez-Rodriguez, J., Rinschen, M. M., Floege, J. & Kramann, R. Big science and big data in nephrology. Kidney Int. 95, 1326–1337 (2019).

    PubMed  Google Scholar 

  4. Smith, L. M. & Kelleher, N. L. Proteoform: a single term describing protein complexity. Nat. Methods 10, 186–187 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  5. Liu, Y., Beyer, A. & Aebersold, R. On the dependency of cellular protein levels on mRNA abundance. Cell 165, 535–550 (2016).

    CAS  PubMed  Google Scholar 

  6. Olsen, J. V. & Mann, M. Status of large-scale analysis of post-translational modifications by mass spectrometry. Mol. Cell Proteom. 12, 3444–3452 (2013).

    CAS  Google Scholar 

  7. Overington, J. P., Al-Lazikani, B. & Hopkins, A. L. How many drug targets are there? Nat. Rev. Drug Discov. 5, 993–996 (2006).

    CAS  PubMed  Google Scholar 

  8. Bache, N. et al. A novel LC system embeds analytes in pre-formed gradients for rapid, ultra-robust proteomics. Mol. Cell Proteom. 17, 2284–2296 (2018).

    CAS  Google Scholar 

  9. Aebersold, R. & Mann, M. Mass-spectrometric exploration of proteome structure and function. Nature 537, 347–355 (2016).

    CAS  PubMed  Google Scholar 

  10. Wilhelm, M. et al. Mass-spectrometry-based draft of the human proteome. Nature 509, 582–587 (2014).

    CAS  PubMed  Google Scholar 

  11. Kim, M.-S. et al. A draft map of the human proteome. Nature 509, 575–581 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Hoyer, K. J. R., Dittrich, S., Bartram, M. P. & Rinschen, M. M. Quantification of molecular heterogeneity in kidney tissue by targeted proteomics. J. Proteom. 193, 85–92 (2019).

    CAS  Google Scholar 

  13. Aebersold, R. & Mann, M. Mass spectrometry-based proteomics. Nature 422, 198–207 (2003).

    CAS  PubMed  Google Scholar 

  14. Hayek, S. S. et al. A tripartite complex of suPAR, APOL1 risk variants and αvβ3 integrin on podocytes mediates chronic kidney disease. Nat. Med. 23, 945–953 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Beck, L. H. et al. M-type phospholipase A2 receptor as target antigen in idiopathic membranous nephropathy. N. Engl. J. Med. 361, 11–21 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Hobeika, L., Barati, M. T., Caster, D. J., McLeish, K. R. & Merchant, M. L. Characterization of glomerular extracellular matrix by proteomic analysis of laser-captured microdissected glomeruli. Kidney Int. 91, 501–511 (2017).

    CAS  PubMed  Google Scholar 

  17. Byron, A. et al. Glomerular cell cross-talk influences composition and assembly of extracellular matrix. J. Am. Soc. Nephrol. 25, 953 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Nagaraj, N. et al. Deep proteome and transcriptome mapping of a human cancer cell line. Mol. Syst. Biol. 7, 548 (2011).

    PubMed  PubMed Central  Google Scholar 

  19. Schwanhäusser, B. et al. Global quantification of mammalian gene expression control. Nature 473, 337–342 (2011).

    PubMed  Google Scholar 

  20. Rinschen, M. M. et al. Quantitative deep mapping of the cultured podocyte proteome uncovers shifts in proteostatic mechanisms during differentiation. Am. J. Physiol. Cell Physiol. 311, C404–C417 (2016).

    PubMed  Google Scholar 

  21. Boerries, M. et al. Molecular fingerprinting of the podocyte reveals novel gene and protein regulatory networks. Kidney Int. 83, 1052–1064 (2013).

    CAS  PubMed  Google Scholar 

  22. Rinschen, M. M. et al. A Multi-layered quantitative in vivo expression atlas of the podocyte unravels kidney disease candidate genes. Cell Rep. 23, 2495–2508 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  23. Geyer, P. E., Holdt, L. M., Teupser, D. & Mann, M. Revisiting biomarker discovery by plasma proteomics. Mol. Syst. Biol. 13, 942 (2017).

    PubMed  PubMed Central  Google Scholar 

  24. Martini, S., Eichinger, F., Nair, V. & Kretzler, M. Defining human diabetic nephropathy on the molecular level: integration of transcriptomic profiles with biological knowledge. Rev. Endocr. Metab. Disord. 9, 267–274 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Niewczas, M. A. et al. A signature of circulating inflammatory proteins and development of end-stage renal disease in diabetes. Nat. Med. 25, 805–813 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Pedigo, C. E. et al. Local TNF causes NFATc1-dependent cholesterol-mediated podocyte injury. J. Clin. Invest. 126, 3336–3350 (2016).

    PubMed  PubMed Central  Google Scholar 

  27. Nagaraj, N. & Mann, M. Quantitative analysis of the intra- and inter-individual variability of the normal urinary proteome. J. Proteome Res. 10, 637–645 (2011).

    CAS  PubMed  Google Scholar 

  28. Hogan, M. C. et al. Identification of biomarkers for PKD1 using urinary exosomes. J. Am. Soc. Nephrol. 26, 1661–1670 (2015).

    CAS  PubMed  Google Scholar 

  29. Salih, M. et al. Proteomics of urinary vesicles links plakins and complement to polycystic kidney disease. J. Am. Soc. Nephrol. 27, 3079–3092 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  30. Diedrich, B. & Dengjel, J. Insights into autosomal dominant polycystic kidney disease by quantitative mass spectrometry-based proteomics. Cell Tissue Res. 369, 41–51 (2017).

    CAS  PubMed  Google Scholar 

  31. Menezes, L. F. & Germino, G. G. Systems biology of polycystic kidney disease: a critical review. Wiley Interdiscip. Rev. Syst. Biol. Med. 7, 39–52 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Klein, J. et al. Fetal urinary peptides to predict postnatal outcome of renal disease in fetuses with posterior urethral valves (PUV). Sci. Transl Med. 5, 198ra106 (2013).

    PubMed  Google Scholar 

  33. Klein, J. et al. Urinary peptidomics provides a noninvasive humanized readout of diabetic nephropathy in mice. Kidney Int. 90, 1045–1055 (2016).

    CAS  PubMed  Google Scholar 

  34. Krochmal, M. et al. Urinary peptidomics analysis reveals proteases involved in diabetic nephropathy. Sci. Rep. 7, 15160 (2017).

    PubMed  PubMed Central  Google Scholar 

  35. Nkuipou-Kenfack, E. et al. Identification of ageing-associated naturally occurring peptides in human urine. Oncotarget 6, 34106–34117 (2015).

    PubMed  PubMed Central  Google Scholar 

  36. Merchant, M. L. et al. Urinary peptidome may predict renal function decline in type 1 diabetes and microalbuminuria. J. Am. Soc. Nephrol. 20, 2065–2074 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Van, J. A. D. et al. Peptidomic analysis of urine from youths with early type 1 diabetes reveals novel bioactivity of uromodulin peptides in vitro. Mol. Cell Proteom. 19, 501–517 (2020).

    CAS  Google Scholar 

  38. Tofte, N. et al. Early detection of diabetic kidney disease by urinary proteomics and subsequent intervention with spironolactone to delay progression (PRIORITY): a prospective observational study and embedded randomised placebo-controlled trial. Lancet Diabetes Endocrinol. 8, 301–312 (2020).

    CAS  PubMed  Google Scholar 

  39. Krochmal, M., Schanstra, J. P. & Mischak, H. Urinary peptidomics in kidney disease and drug research. Expert Opin. Drug Discov. 13, 259–268 (2018).

    CAS  PubMed  Google Scholar 

  40. Frantzi, M., Mischak, H. & Latosinska, A. Clinical proteomics on the path toward implementation: first promises delivered. Proteom. Clin. Appl. 13, e1800094 (2019).

    Google Scholar 

  41. Sethi, S. & Theis, J. D. Pathology and diagnosis of renal non-AL amyloidosis. J. Nephrol. 31, 343–350 (2017).

    PubMed  Google Scholar 

  42. Sethi, S. et al. Apolipoprotein CII amyloidosis associated with p.Lys41Thr mutation. Kidney Int. Rep. 3, 1193–1201 (2018).

    PubMed  PubMed Central  Google Scholar 

  43. Qi, W. et al. Pyruvate kinase M2 activation may protect against the progression of diabetic glomerular pathology and mitochondrial dysfunction. Nat. Med. 23, 753–762 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Slavov, N. Unpicking the proteome in single cells. Science 367, 512–513 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Koehler, S. et al. Proteome analysis of isolated podocytes reveals stress responses in glomerular sclerosis. J. Am. Soc. Nephrol. 31, 544–559 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Himmerkus, N. et al. Viewing cortical collecting duct function through phenotype-guided single-tubule proteomics. Function 1, zqaa007 (2020).

    PubMed  PubMed Central  Google Scholar 

  47. Limbutara, K., Chou, C.-L. & Knepper, M. A. Quantitative proteomics of All 14 renal tubule segments in rat. J. Am. Soc. Nephrol. 31, 1255–1266 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Rinschen, M. M., Limbutara, K., Knepper, M. A., Payne, D. M. & Pisitkun, T. From molecules to mechanisms: functional proteomics and its application to renal tubule physiology. Physiol. Rev. 98, 2571–2606 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  49. Sung, C.-C. et al. RNA-Seq and protein mass spectrometry in microdissected kidney tubules reveal signaling processes initiating lithium-induced nephrogenic diabetes insipidus. Kidney Int. 96, 363–377 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  50. Carr, S. A. et al. Targeted peptide measurements in biology and medicine: best practices for mass spectrometry-based assay development using a fit-for-purpose approach. Mol. Cell. Proteom. 13, 907–917 (2014).

    CAS  Google Scholar 

  51. Guo, T. et al. Rapid mass spectrometric conversion of tissue biopsy samples into permanent quantitative digital proteome maps. Nat. Med. 21, 407–413 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  52. Andeen, N. K., Yang, H.-Y., Dai, D.-F., MacCoss, M. J. & Smith, K. D. DnaJ homolog subfamily B member 9 is a putative autoantigen in fibrillary GN. J. Am. Soc. Nephrol. 29, 231–239 (2018).

    CAS  PubMed  Google Scholar 

  53. Dasari, S. et al. DnaJ heat shock protein family B member 9 is a novel biomarker for fibrillary GN. J. Am. Soc. Nephrol. 29, 51–56 (2018).

    CAS  PubMed  Google Scholar 

  54. Merchant, M. L. et al. Proteomic analysis identifies distinct glomerular extracellular matrix in collapsing focal segmental glomerulosclerosis. J. Am. Soc. Nephrol. 31, 1883–1904 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  55. Hubner, N. C. & Mann, M. Extracting gene function from protein-protein interactions using quantitative BAC InteraCtomics (QUBIC). Methods 53, 453–459 (2011).

    CAS  PubMed  Google Scholar 

  56. Kohli, P. et al. Label-free quantitative proteomic analysis of the YAP/TAZ interactome. Am. J. Physiol. Cell Physiol. 306, C805–C818 (2014).

    CAS  PubMed  Google Scholar 

  57. Klein, J. B. Applying proteomics to detect early signs of chronic kidney disease: where has the magic gone? Expert. Rev. Proteom. 14, 387–390 (2017).

    CAS  Google Scholar 

  58. Francis, J. M., Beck, L. H. & Salant, D. J. Membranous nephropathy: a journey from bench to bedside. Am. J. Kidney Dis. 68, 138–147 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  59. Tomas, N. M. et al. Thrombospondin type-1 domain-containing 7A in idiopathic membranous nephropathy. N. Engl. J. Med. 371, 2277–2287 (2014).

    PubMed  PubMed Central  Google Scholar 

  60. Sethi, S. et al. Exostosin 1/exostosin 2-associated membranous nephropathy. J. Am. Soc. Nephrol. 30, 1123–1136 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  61. Laghmani, K. et al. Polyhydramnios, transient antenatal Bartter’s syndrome, and MAGED2 mutations. N. Engl. J. Med. 374, 1853–1863 (2016).

    CAS  PubMed  Google Scholar 

  62. Legrand, A. et al. Prevalence of Novel MAGED2 Mutations in antenatal Bartter syndrome. Clin. J. Am. Soc. Nephrol. 13, 242–250 (2018).

    PubMed  Google Scholar 

  63. Grahammer, F. et al. A flexible, multilayered protein scaffold maintains the slit in between glomerular podocytes. JCI Insight 1, e86177 (2016).

    PubMed Central  Google Scholar 

  64. Rinschen, M. M. et al. The ubiquitin ligase Ubr4 controls stability of podocin/MEC-2 supercomplexes. Hum. Mol. Genet. 25, 1328–1344 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  65. Anders, U. et al. SPRi-MALDI MS: characterization and identification of a kinase from cell lysate by specific interaction with different designed ankyrin repeat proteins. Anal. Bioanal. Chem. 409, 1827–1836 (2017).

    CAS  PubMed  Google Scholar 

  66. Florinskaya, A. et al. SPR biosensors in direct molecular fishing: implications for protein interactomics. Sensors 18, 1616 (2018).

    PubMed Central  Google Scholar 

  67. Kleifeld, O. et al. Isotopic labeling of terminal amines in complex samples identifies protein N-termini and protease cleavage products. Nat. Biotechnol. 28, 281–288 (2010).

    CAS  PubMed  Google Scholar 

  68. Choudhary, C., Weinert, B. T., Nishida, Y., Verdin, E. & Mann, M. The growing landscape of lysine acetylation links metabolism and cell signalling. Nat. Rev. Mol. Cell Biol. 15, 536–550 (2014).

    CAS  PubMed  Google Scholar 

  69. Macek, B., Mann, M. & Olsen, J. V. Global and site-specific quantitative phosphoproteomics: principles and applications. Annu. Rev. Pharmacol. Toxicol. 49, 199–221 (2009).

    CAS  PubMed  Google Scholar 

  70. Lienhard, G. E. Non-functional phosphorylations? Trends Biochem. Sci. 33, 351–352 (2008).

    CAS  PubMed  Google Scholar 

  71. Landry, C. R., Levy, E. D. & Michnick, S. W. Weak functional constraints on phosphoproteomes. Trends Genet. 25, 193–197 (2009).

    CAS  PubMed  Google Scholar 

  72. Ochoa, D. et al. The functional landscape of the human phosphoproteome. Nat. Biotechnol. 38, 365–373 (2020).

    CAS  PubMed  Google Scholar 

  73. Beltrao, P. et al. Systematic functional prioritization of protein posttranslational modifications. Cell 150, 413–425 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  74. Tonna, S. J. et al. NPHS2 variation in focal and segmental glomerulosclerosis. BMC Nephrol. 9, 13 (2008).

    PubMed  PubMed Central  Google Scholar 

  75. Rinschen, M. M. et al. Phosphoproteomic analysis reveals regulatory mechanisms at the kidney filtration barrier. J. Am. Soc. Nephrol. 25, 1509–1522 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  76. Rinschen, M. M. et al. Comparative phosphoproteomic analysis of mammalian glomeruli reveals conserved podocin C-terminal phosphorylation as a determinant of slit diaphragm complex architecture. Proteomics 15, 1326–1331 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  77. Buvall, L. et al. Synaptopodin is a coincidence detector of tyrosine versus serine/threonine phosphorylation for the modulation of Rho protein crosstalk in podocytes. J. Am. Soc. Nephrol. 28, 837–851 (2017).

    CAS  PubMed  Google Scholar 

  78. Miller, M. L. et al. Linear motif atlas for phosphorylation-dependent signaling. Sci. Signal. 1, ra2 (2008).

    PubMed  PubMed Central  Google Scholar 

  79. Garg, P., Verma, R., Nihalani, D., Johnstone, D. B. & Holzman, L. B. Neph1 cooperates with nephrin to transduce a signal that induces actin polymerization. Mol. Cell. Biol. 27, 8698–8712 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  80. Schroeter, C. B. et al. Protein half-life determines expression of proteostatic networks in podocyte differentiation. FASEB J. 32, 4696–4713 (2018).

    CAS  PubMed  Google Scholar 

  81. Yaddanapudi, S. et al. CD2AP in mouse and human podocytes controls a proteolytic program that regulates cytoskeletal structure and cellular survival. J. Clin. Invest. 121, 3965–3980 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  82. Yamamoto-Nonaka, K. et al. Cathepsin D in podocytes is important in the pathogenesis of proteinuria and CKD. J. Am. Soc. Nephrol. 27, 2685–2700 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  83. Garsen, M. et al. Cathepsin L is crucial for the development of early experimental diabetic nephropathy. Kidney Int. 90, 1012–1022 (2016).

    CAS  PubMed  Google Scholar 

  84. Rinschen, M. M. et al. N-degradomic analysis reveals a proteolytic network processing the podocyte cytoskeleton. J. Am. Soc. Nephrol. 28, 2867–2878 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  85. Rinschen, M. M., Huesgen, P. F. & Koch, R. E. The podocyte protease web: uncovering the gatekeepers of glomerular disease. Am. J. Physiol. Ren. Physiol. 315, F1812–F1816 (2018).

    CAS  Google Scholar 

  86. Späth, M. R. et al. The proteome microenvironment determines the protective effect of preconditioning in cisplatin-induced acute kidney injury. Kidney Int. 95, 333–349 (2019).

    PubMed  Google Scholar 

  87. Bensimon, A., Heck, A. J. R. & Aebersold, R. Mass spectrometry-based proteomics and network biology. Annu. Rev. Biochem. 81, 379–405 (2012).

    CAS  PubMed  Google Scholar 

  88. Creixell, P. et al. Pathway and network analysis of cancer genomes. Nat. Methods 12, 615–621 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  89. Türei, D., Korcsmáros, T. & Saez-Rodriguez, J. OmniPath: guidelines and gateway for literature-curated signaling pathway resources. Nat. Methods 13, 966–967 (2016).

    PubMed  Google Scholar 

  90. Rodchenkov, I. et al. Pathway commons 2019 update: integration, analysis and exploration of pathway data. Nucleic Acids Res. 48, D489–D497 (2020).

    CAS  PubMed  Google Scholar 

  91. Huang, J. K. et al. Systematic evaluation of molecular networks for discovery of disease genes. Cell Syst. 6, 484–495.e5 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  92. Gonzalez-Vicente, A., Hopfer, U. & Garvin, J. L. Developing tools for analysis of renal genomic data: an invitation to participate. J. Am. Soc. Nephrol. 28, 3438–3440 (2017).

    PubMed  PubMed Central  Google Scholar 

  93. Terfve, C. et al. System-wide quantitative proteomics of the metabolic syndrome in mice: genotypic and dietary effects. J. Proteome Res. 16, 831–841 (2017).

    CAS  PubMed  Google Scholar 

  94. Goh, W. W. B. & Wong, L. Networks in proteomics analysis of cancer. Curr. Opin. Biotechnol. 24, 1122–1128 (2013).

    CAS  PubMed  Google Scholar 

  95. Schoeberl, B. et al. Therapeutically targeting ErbB3: a key node in ligand-induced activation of the ErbB receptor-PI3K axis. Sci. Signal. 2, ra31 (2009).

    PubMed  Google Scholar 

  96. Schoeberl, B. et al. Systems biology driving drug development: from design to the clinical testing of the anti-ErbB3 antibody seribantumab (MM-121). NPJ Syst. Biol. Appl. 3, 16034 (2017).

    PubMed  PubMed Central  Google Scholar 

  97. Kholodenko, B., Yaffe, M. B. & Kolch, W. Computational approaches for analyzing information flow in biological networks. Sci. Signal. 5, re1 (2012).

    PubMed  Google Scholar 

  98. Le Novère, N. Quantitative and logic modelling of molecular and gene networks. Nat. Rev. Genet. 16, 146–158 (2015).

    PubMed  PubMed Central  Google Scholar 

  99. Lamb, J. et al. The connectivity map: using gene-expression signatures to connect small molecules, genes, and disease. Science 313, 1929–1935 (2006).

    CAS  PubMed  Google Scholar 

  100. Subramanian, A. et al. A next generation connectivity map: L1000 platform and the first 1,000,000 profiles. Cell 171, 1437–1452.e17 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  101. Williams, V. R. et al. Connectivity mapping of a chronic kidney disease progression signature identified lysine deacetylases as novel therapeutic targets. Kidney Int. 98, 116–132 (2020).

    CAS  PubMed  Google Scholar 

  102. Tajti, F. et al. A functional landscape of CKD entities from public transcriptomic data. Kidney Int. Rep. 5, 211–224 (2020).

    PubMed  Google Scholar 

  103. Schanstra, J. P. et al. Systems biology identifies cytosolic PLA2 as a target in vascular calcification treatment. JCI Insight 4, e125638 (2019).

    PubMed Central  Google Scholar 

  104. Litichevskiy, L. et al. A library of phosphoproteomic and chromatin signatures for characterizing cellular responses to drug perturbations. Cell Syst. 6, 424–443.e7 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  105. Calizo, R. C. et al. Disruption of podocyte cytoskeletal biomechanics by dasatinib leads to nephrotoxicity. Nat. Commun. 10, 2061 (2019).

    PubMed  PubMed Central  Google Scholar 

  106. Azeloglu, E. U. et al. Interconnected network motifs control podocyte morphology and kidney function. Sci. Signal. 7, ra12 (2014).

    PubMed  PubMed Central  Google Scholar 

  107. Moraru, I. I. et al. The virtual cell modeling and simulation software environment. IET Syst. Biol. 2, 352–362 (2008).

    CAS  PubMed  Google Scholar 

  108. Falkenberg, C. V. et al. Fragility of foot process morphology in kidney podocytes arises from chaotic spatial propagation of cytoskeletal instability. PLoS Comput. Biol. 13, e1005433 (2017).

    PubMed  PubMed Central  Google Scholar 

  109. Rinschen, M. M. et al. YAP-mediated mechanotransduction determines the podocyte’s response to damage. Sci. Signal. 10, eaaf8165 (2017).

    PubMed  Google Scholar 

  110. Wong, J. S., Meliambro, K., Ray, J. & Campbell, K. N. Hippo signaling in the kidney: the good and the bad. Am. J. Physiol. Ren. Physiol. 311, F241–F248 (2016).

    CAS  Google Scholar 

  111. Klein, J. et al. The KUPKB: a novel Web application to access multiomics data on kidney disease. FASEB J. 26, 2145–2153 (2012).

    CAS  PubMed  Google Scholar 

  112. Rhodes, D. R. et al. Oncomine 3.0: genes, pathways, and networks in a collection of 18,000 cancer gene expression profiles. Neoplasia 9, 166–180 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  113. Vizcaíno, J. A. et al. ProteomeXchange provides globally coordinated proteomics data submission and dissemination. Nat. Biotechnol. 32, 223–226 (2014).

    PubMed  PubMed Central  Google Scholar 

  114. Perez-Riverol, Y. et al. The PRIDE database and related tools and resources in 2019: improving support for quantification data. Nucleic Acids Res. 47, D442–D450 (2019).

    CAS  PubMed  Google Scholar 

  115. Sullivan, K. M. & Susztak, K. Unravelling the complex genetics of common kidney diseases: from variants to mechanisms. Nat. Rev. Nephrol. https://doi.org/10.1038/s41581-020-0298-1 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  116. Gillies, C. E. et al. An eQTL landscape of kidney tissue in human nephrotic syndrome. Am. J. Hum. Genet. 103, 232–244 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  117. Qiu, C. et al. Renal compartment-specific genetic variation analyses identify new pathways in chronic kidney disease. Nat. Med. 24, 1721–1731 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  118. Rinschen, M. M., Ivanisevic, J., Giera, M. & Siuzdak, G. Identification of bioactive metabolites using activity metabolomics. Nat. Rev. Mol. Cell Biol. 20, 353–367 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  119. Narita, T., Weinert, B. T. & Choudhary, C. Functions and mechanisms of non-histone protein acetylation. Nat. Rev. Mol. Cell Biol. 20, 156–174 (2019).

    CAS  PubMed  Google Scholar 

  120. James, A. M. et al. The causes and consequences of nonenzymatic protein acylation. Trends Biochem. Sci. 43, 921–932 (2018).

    CAS  PubMed  Google Scholar 

  121. Hart, G. W., Slawson, C., Ramirez-Correa, G. & Lagerlof, O. Cross talk between O-GlcNAcylation and phosphorylation: roles in signaling, transcription, and chronic disease. Annu. Rev. Biochem. 80, 825–858 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  122. Rinschen, M. M. et al. Metabolic rewiring of the hypertensive kidney. Sci. Signal. 12, eaax9760 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  123. Terfve, C. D. A., Wilkes, E. H., Casado, P., Cutillas, P. R. & Saez-Rodriguez, J. Large-scale models of signal propagation in human cells derived from discovery phosphoproteomic data. Nat. Commun. 6, 1–11 (2015).

    Google Scholar 

  124. Kang, H. M. et al. Defective fatty acid oxidation in renal tubular epithelial cells has a key role in kidney fibrosis development. Nat. Med. 21, 37–46 (2015).

    CAS  PubMed  Google Scholar 

  125. Gordin, D. et al. Characterization of glycolytic enzymes and pyruvate kinase M2 in type 1 and 2 diabetic nephropathy. Diabetes Care 42, 1263–1273 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  126. Tape, C. J. Systems biology analysis of heterocellular signaling. Trends Biotechnol. 34, 627–637 (2016).

    CAS  PubMed  Google Scholar 

  127. Malone, A. F., Wu, H. & Humphreys, B. D. Bringing renal biopsy interpretation into the molecular age with single-cell RNA sequencing. Semin. Nephrol. 38, 31–39 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  128. Höhne, M. et al. Single-nephron proteomes connect morphology and function in proteinuric kidney disease. Kidney Int. 93, 1308–1319 (2018).

    PubMed  Google Scholar 

  129. Fribourg, M. et al. T-cell exhaustion correlates with improved outcomes in kidney transplant recipients. Kidney Int. 96, 436–449 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  130. Brähler, S. et al. Opposing roles of dendritic cell subsets in experimental GN. J. Am. Soc. Nephrol. 29, 138–154 (2018).

    PubMed  Google Scholar 

  131. Singh, N. et al. Development of a 2-dimensional atlas of the human kidney with imaging mass cytometry. JCI Insight 4, e129477 (2019).

    PubMed Central  Google Scholar 

  132. Newman, A. M. et al. Robust enumeration of cell subsets from tissue expression profiles. Nat. Methods 12, 453–457 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  133. Drewes, G. & Knapp, S. Chemoproteomics and chemical probes for target discovery. Trends Biotechnol. 36, 1275–1286 (2018).

    CAS  PubMed  Google Scholar 

  134. Klaeger, S. et al. The target landscape of clinical kinase drugs. Science 358, eaan4368 (2017).

    PubMed  PubMed Central  Google Scholar 

  135. Huber, K. V. M. et al. Stereospecific targeting of MTH1 by (S)-crizotinib as an anticancer strategy. Nature 508, 222–227 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  136. Becher, I. et al. Thermal profiling reveals phenylalanine hydroxylase as an off-target of panobinostat. Nat. Chem. Biol. 12, 908–910 (2016).

    CAS  PubMed  Google Scholar 

  137. Bartram, M. P. et al. Three-layered proteomic characterization of a novel ACTN4 mutation unravels its pathogenic potential in FSGS. Hum. Mol. Genet. 25, 1152–1164 (2016).

    CAS  PubMed  Google Scholar 

  138. Gillet, L. C. et al. Targeted data extraction of the MS/MS spectra generated by data-independent acquisition: a new concept for consistent and accurate proteome analysis. Mol. Cell Proteomics 11, O111.016717 (2012).

    PubMed  PubMed Central  Google Scholar 

  139. Ong, S.-E. et al. Stable isotope labeling by amino acids in cell culture, SILAC, as a simple and accurate approach to expression proteomics. Mol. Cell Proteom. 1, 376–386 (2002).

    CAS  Google Scholar 

  140. McAlister, G. C. et al. Increasing the multiplexing capacity of TMTs using reporter ion isotopologues with isobaric masses. Anal. Chem. 84, 7469–7478 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  141. Ngo, D. et al. Aptamer-based proteomic profiling reveals novel candidate biomarkers and pathways in cardiovascular disease. Circulation 134, 270–285 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  142. Knepper, M. A. Proteomics and the kidney. J. Am. Soc. Nephrol. 13, 1398–1408 (2002).

    CAS  PubMed  Google Scholar 

  143. Hoppe-Seyler, F. & Butz, K. Peptide aptamers: powerful new tools for molecular medicine. J. Mol. Med. 78, 426–430 (2000).

    CAS  PubMed  Google Scholar 

  144. Geyer, C. R., Colman-Lerner, A. & Brent, R. “Mutagenesis” by peptide aptamers identifies genetic network members and pathway connections. Proc. Natl Acad. Sci. USA 96, 8567–8572 (1999).

    CAS  PubMed  PubMed Central  Google Scholar 

  145. Anderson, N. L. et al. Mass spectrometric quantitation of peptides and proteins using stable isotope standards and capture by anti-peptide antibodies (SISCAPA). J. Proteome Res. 3, 235–244 (2004).

    CAS  PubMed  Google Scholar 

  146. Neubert, H. et al. 2018 white paper on recent issues in bioanalysis: focus on immunogenicity assays by hybrid LBA/LCMS and regulatory feedback (Part 2 — PK, PD & ADA assays by hybrid LBA/LCMS & regulatory agencies’ inputs on bioanalysis, biomarkers and immunogenicity). Bioanalysis 10, 1897–1917 (2018).

    CAS  PubMed  Google Scholar 

  147. Bodenmiller, B. Multiplexed epitope-based tissue imaging for discovery and healthcare applications. Cell Syst. 2, 225–238 (2016).

    CAS  PubMed  Google Scholar 

  148. Casadonte, R. et al. Imaging mass spectrometry analysis of renal amyloidosis biopsies reveals protein co-localization with amyloid deposits. Anal. Bioanal. Chem. 407, 5323–5331 (2015).

    CAS  PubMed  Google Scholar 

  149. Rhee, H.-W. et al. Proteomic mapping of mitochondria in living cells via spatially restricted enzymatic tagging. Science 339, 1328–1331 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

The authors apologize to all researchers whose work could not be cited due to space limitations. M.M.R. was supported by the DFG (RI2811/1 and RI2811/2), as well as the Young Investigator Award from the Novo Nordisk Foundation, grant number NNF19OC0056043. The authors thank Nicolas Palacio-Escat (Heidelberg University) for help with the initial figure panels and Aurelien Dugourd (Aachen University Hospital) for critically reading the manuscript before submission.

Author information

Authors and Affiliations

Authors

Contributions

All authors researched data for the article, made substantial contributions to discussions of the content, wrote the article, and reviewed or edited the manuscript before submission.

Corresponding author

Correspondence to Markus M. Rinschen.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information

Nature Reviews Nephrology thanks the anonymous reviewers for their contribution to the peer review of this work.

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Related links

Connectivity map: https://clue.io/cmap

Proteome central: http://proteomecentral.proteomexchange.org/cgi/GetDataset

Renal genomics portal: https://www.wikipathways.org/index.php/Portal:RenalGenomics

Glossary

Proteoforms

Distinct forms of a protein molecule that arise from the same transcript or gene.

Intensity-based absolute quantification

An algorithm used for protein copy number estimation in which total protein intensity (that is, the sum of all peptide intensities) is divided by the number of peptides.

Surface plasmon resonance

A biophysical method used to probe protein–protein (and molecule–molecule) interactions.

PhoNEMES modelling

A computational tool used to build logic models of signalling networks from discovery mass-spectrometry-based phosphoproteomic data.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Rinschen, M.M., Saez-Rodriguez, J. The tissue proteome in the multi-omic landscape of kidney disease. Nat Rev Nephrol 17, 205–219 (2021). https://doi.org/10.1038/s41581-020-00348-5

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41581-020-00348-5

This article is cited by

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research