Matrix-Assisted Laser Desorption/Ionization Mass Spectrometry Imaging: Diagnostic Pathways and Metabolites for Renal Tumor Entities

Background: Correct tumor subtyping of primary renal tumors is essential for treatment decision in daily routine. Most of the tumors can be classified based on morphology alone. Nevertheless, some diagnoses are difficult, and further investigations are needed for correct tumor subtyping. Besides histochemical investigations, high-mass-resolution matrix-assisted laser desorption/ionization mass spectrometry imaging (MALDI-MSI) can detect new diagnostic biomarkers and hence improve the diagnostic. Patients and Methods: Formalin-fixed paraffin embedded tissue specimens from clear cell renal cell carcinoma (ccRCC, n = 552), papillary renal cell carcinoma (pRCC, n = 122), chromophobe renal cell carcinoma (chRCC, n = 108), and renal oncocytoma (rO, n = 71) were analyzed by high-mass-resolution MALDI fourier-transform ion cyclotron resonance (FT-ICR) MSI. The SPACiAL pipeline was executed for automated co-registration of histological and molecular features. Pathway enrichment and pathway topology analysis were performed to determine significant differences between RCC subtypes. Results: We discriminated the four histological subtypes (ccRCC, pRCC, chRCC, and rO) and established the subtype-specific pathways and metabolic profiles. rO showed an enrichment of pentose phosphate, taurine and hypotaurine, glycerophospholipid, amino sugar and nucleotide sugar, fructose and mannose, glycine, serine, and threonine pathways. ChRCC is defined by enriched pathways including the amino sugar and nucleotide sugar, fructose and mannose, glycerophospholipid, taurine and hypotaurine, glycine, serine, and threonine pathways. Pyrimidine, amino sugar and nucleotide sugar, glycerophospholipids, and glutathione pathways are enriched in ccRCC. Furthermore, we detected enriched phosphatidylinositol and glycerophospholipid pathways in pRCC. Conclusion: In summary, we performed a classification system with a mean accuracy in tumor discrimination of 85.13%. Furthermore, we detected tumor-specific biomarkers for the four most common primary renal tumors by MALDI-MSI. This method is a useful tool in differential diagnosis and biomarker detection.

[1]  V. Copié,et al.  Nuclear magnetic resonance and surface-assisted laser desorption/ionization mass spectrometry-based metabolome profiling of urine samples from kidney cancer patients. , 2020, Journal of pharmaceutical and biomedical analysis.

[2]  Jialing Zhang,et al.  Mass Spectrometry Imaging enables Discrimination of Renal Oncocytoma from Renal Cell Cancer Subtypes and Normal Kidney Tissues. , 2019, Cancer research.

[3]  M. Attimonelli,et al.  Papillary Renal Cell Carcinomas Rewire Glutathione Metabolism and Are Deficient in Both Anabolic Glucose Synthesis and Oxidative Phosphorylation , 2019, Cancers.

[4]  K. Kinoshita,et al.  Value of global metabolomics in association with diagnosis and clinicopathological factors of renal cell carcinoma , 2019, International journal of cancer.

[5]  Eden L. Romm,et al.  Recognition of early and late stages of bladder cancer using metabolites and machine learning , 2019, Metabolomics.

[6]  M. Attimonelli,et al.  Metabolic reprogramming and elevation of glutathione in chromophobe renal cell carcinomas , 2019, bioRxiv.

[7]  Lorenzo Marconi,et al.  European Association of Urology Guidelines on Renal Cell Carcinoma: The 2019 Update. , 2019, European urology.

[8]  V. Mootha,et al.  Early loss of mitochondrial complex I and rewiring of glutathione metabolism in renal oncocytoma , 2018, Proceedings of the National Academy of Sciences.

[9]  E. Reznik,et al.  Impairment of gamma-glutamyl transferase 1 activity in the metabolic pathogenesis of chromophobe renal cell carcinoma , 2018, Proceedings of the National Academy of Sciences.

[10]  David S. Wishart,et al.  MetaboAnalyst 4.0: towards more transparent and integrative metabolomics analysis , 2018, Nucleic Acids Res..

[11]  David S. Wishart,et al.  HMDB 4.0: the human metabolome database for 2018 , 2017, Nucleic Acids Res..

[12]  M. Attimonelli,et al.  Renal oncocytoma characterized by the defective complex I of the respiratory chain boosts the synthesis of the ROS scavenger glutathione , 2017, OncoTarget.

[13]  A. Holmgren,et al.  Redox Signaling Mediated by Thioredoxin and Glutathione Systems in the Central Nervous System. , 2017, Antioxidants & redox signaling.

[14]  S. Williamson,et al.  Modern Pathologic Diagnosis of Renal Oncocytoma , 2017, Journal of kidney cancer and VHL.

[15]  B. Delahunt,et al.  Diagnostic criteria for oncocytic renal neoplasms: a survey of urologic pathologists. , 2017, Human pathology.

[16]  Michael Becker,et al.  FDR-controlled metabolite annotation for high-resolution imaging mass spectrometry , 2016, Nature Methods.

[17]  W. Weichert,et al.  PD-1/PD-L1 expression in chromophobe renal cell carcinoma: An immunological exception? , 2016, Medical Oncology.

[18]  Na Sun,et al.  High-mass-resolution MALDI mass spectrometry imaging of metabolites from formalin-fixed paraffin-embedded tissue , 2016, Nature Protocols.

[19]  P. Humphrey,et al.  The 2016 WHO Classification of Tumours of the Urinary System and Male Genital Organs-Part A: Renal, Penile, and Testicular Tumours. , 2016, European urology.

[20]  Chris Sander,et al.  An Integrated Metabolic Atlas of Clear Cell Renal Cell Carcinoma. , 2016, Cancer cell.

[21]  D. Ribatti,et al.  Metabolomic profile of glycolysis and the pentose phosphate pathway identifies the central role of glucose-6-phosphate dehydrogenase in clear cell-renal cell carcinoma , 2015, Oncotarget.

[22]  H. Schlüter,et al.  MALDI imaging on tissue microarrays identifies molecular features associated with renal cell cancer phenotype. , 2014, Anticancer research.

[23]  E. Gazzano,et al.  The pentose phosphate pathway: an antioxidant defense and a crossroad in tumor cell fate. , 2012, Free radical biology & medicine.

[24]  E. Mikros,et al.  (1)H NMR metabonomic analysis in renal cell carcinoma: a possible diagnostic tool. , 2010, Journal of proteome research.

[25]  H. Moch,et al.  Chromophobe Renal Cell Carcinoma: Histomorphologic Characteristics and Evaluation of Conventional Pathologic Prognostic Parameters in 145 Cases , 2008, The American journal of surgical pathology.

[26]  Ximing J. Yang,et al.  Renal oncocytoma with and without intravascular extension into the branches of renal vein have the same morphological, immunohistochemical and genetic features , 2008, Virchows Archiv.

[27]  K. Sipos,et al.  Maturation of Cytosolic Iron-Sulfur Proteins Requires Glutathione* , 2002, The Journal of Biological Chemistry.

[28]  S. Tickoo,et al.  Renal oncocytoma: a reappraisal of morphologic features with clinicopathologic findings in 80 cases. , 1999, The American journal of surgical pathology.

[29]  A. Meister,et al.  On the discovery of glutathione. , 1988, Trends in biochemical sciences.

[30]  S. Srivastava,et al.  Detoxification of xenobiotics by glutathione S‐transferases in erythrocytes: the transport of the conjugate of glutathione and 1‐chloro‐2,4‐dinitrobenzene , 1983, British journal of haematology.

[31]  H. Krebs,et al.  Regulation of the pentose phosphate cycle. , 1974, The Biochemical journal.

[32]  Hiroyuki Ogata,et al.  KEGG: Kyoto Encyclopedia of Genes and Genomes , 1999, Nucleic Acids Res..