Abstract

Metabolomics is a field of systems biology that draws on the scientific methods of other groups to qualitatively or quantitatively characterize small molecule metabolites in organisms, revealing their interconnections with the state of the organism at an overall relative macroscopic level. Diabetic kidney disease (DKD) is well known as a chronic metabolic disease, and metabolomics provides an excellent platform for its clinical study. A growing number of metabolomic analyses have revealed that individuals with DKD have metabolic disturbances of multiple substances in their bodies. With the continuous development and improvement of metabolomic analysis technology, the application of metabolomics in the clinical research of DKD is also expanding. This review discusses the recent progress of metabolomics in the early diagnosis, disease prognosis, and pathogenesis of DKD at the level of small molecule metabolites in vivo.

Diabetes mellitus (DM) is one of the chronic noncommunicable diseases that seriously endangers human life and health, affects the quality of life of patients, and increases the economic burden of countries. According to the latest data from the International Diabetes Federation, there were about 463 million people with DM worldwide in 2019, and it was expected to reach 700 million by 2045.1 Diabetic kidney disease (DKD) is not only one of the most common chronic microvascular complications of diabetes but is also an important cause of end-stage renal disease (ESRD). Some research shows that about 30% to 50% of ESRD worldwide is due to DKD.2 Alicic et al3 revealed that the prevalence of DKD in diabetic patients to be 40%. Current clinical indicators used to diagnose DKD include estimation of glomerular filtration rate (eGFR) and urine albumin creatinine ratio (UACR), although they have limited high sensitivity and specificity. The eGFR decreases significantly only when severe renal damage occurs in patients with DKD because the blood creatinine, which is used to calculate the eGFR, drops significantly only when the eGFR is below 50% of normal. Increased UACR is considered a characteristic manifestation of renal damage, but some studies have shown that a significant proportion of DKD patients do not have albuminuria.4,5 In terms of treatment, despite the fact that the number of patients with DM and DKD is increasing annually, there are very limited therapies available to slow down or reverse the progression. The main therapeutic strategies for DKD include strict blood pressure and glycemic control, antidiabetic drugs, etc; however, these measures only slow down the progression of DKD, and there is no way to reverse DKD so far. This requires us to examine DKD from a new perspective, deepen the mechanism research, discover more biomarkers that can help early diagnosis, and develop new treatment strategies.

DKD is a chronic metabolic disease that often causes metabolic disorders of water, electrolytes, proteins, and lipids in the body. The dynamics of small molecule metabolites (relative molecular mass <1000) in an organism reflect the physiological or pathological state of the organism in real time. Metabolomics gives us a new perspective on DKD, which would help us to deepen our mechanistic studies, discover more biological markers for early diagnosis, and develop new therapeutic strategies.

Metabolomics

Metabolomics originated in the 1970s,6 when Professor Nicholson7 proposed the concept of “metabolomics” and drew up a metabolic fingerprint by scanning body fluids through nuclear magnetic resonance (NMR) techniques. With the completion of the sequencing of the Human Genome Project marking the advent of the postgenomic era, more and more research is focusing on the functional analysis of genomes. The emergence of metabolomics bridges the gap between genes and disease phenotypes and provides a possibility for the functional analysis of genomes. Metabolomics mainly focuses on the quantitative analysis of small molecule metabolites in organisms by following the research ideas of genomics to find the relationship between metabolites and physiological/pathological changes in the organism. Currently, metabolomics, genomics, transcriptomics, and proteomics have become important components of systems biology. The core concept of metabolomics is to use the metabolic state of the organism to reflect its overall functional status. It allows a comprehensive analysis of the dynamics of endogenous and exogenous compounds in the body and systematically reflects the expression levels of genes and proteins in the body in response to various stimuli. Many metabolic substances in the body have been maintained in a relatively balanced and stable state for a long time, which is regulated and influenced by genes, diet, environmental factors, and intestinal microorganisms; once this balance is disrupted, it would herald the onset or development of disease.8 Metabolomics, as an emerging omics discipline, has a wide range of applications in the fields of plants, food, disease diagnosis, and microbiology.9 Meanwhile, it has a multidisciplinary intersection and is closely related to organic chemistry, analytical chemistry, chemometrics, informatics, and biology.10,11 Metabolomics is positioned downstream of proteomics and belongs to the continuation of the central dogma, which allows the mapping of body fluids (plasma, urine, tissue fluids, etc) and metabolic profiles to dynamically reflect the function and state of the organism. Compared with transcriptomics and proteomics, metabolomics has its own unique advantages: (1) as a continuation of the central dogma, it can amplify small changes at the gene level; (2) as the end product of gene expression and body metabolism, the number of metabolites in the body is much smaller than the number of genes and proteins; (3) as a relatively macroscopic representation of the body’s state, metabolomics is closer to the phenotype of disease and allows us to visualize the current physiological or pathological state of the organism; (4) the chemical composition of metabolites is similar across species, and the metabolomic analysis techniques used are more generalizable.12

In general, the main processes of metabolomics research include sample collection and preparation, sample testing, metabolic data collection, and data analysis. Currently, the samples used for metabolomics are mainly from blood, urine, and tissue fluids, and a sufficient number of representative samples are usually required to reduce individual differences. Different analysis platforms require different sample preparation processes; for example, nuclear magnetic resonance (NMR) does not require much processing of the sample and is noninvasive and nondestructive,13 while liquid chromatography/gas chromatography (LC/GC) coupled with mass spectrometry (MS) may require derivatization to increase the volatility of the sample.14 Data analysis is the most critical part of performing metabolomics studies. The sample testing process would generate a large amount of fragmented data. In order to extract potentially valuable information from these data, it is necessary to reduce the dimensionality of the data with the help of some multivariate mathematical statistical analysis methods. Partial least squares-discriminant analysis (PLS-DA), principal component analysis (PCA), neural networks, cluster analysis, and support vector machines are the most commonly used data analysis methods in metabolomics.12,15 The appropriate data analysis method is selected according to the specific experimental design scheme as well as the experimental results. For example, PCA can be chosen when the results are more different between groups, while PLS-DA is preferred when the results are less different between groups and the sample size is more different between groups.15 On the basis of PLS-DA, characteristic variables of diseases is then screened out by the variable importance of projection (VIP), with the screening criterion of VIP >1.16 In turn, this would then be used to construct machine learning models and regression models for early identification and diagnosis of diseases. In addition, metabolic pathway enrichment analysis based on characteristic variables can find the specific metabolic pathways associated with diseases, which would contribute to the study of pathogenesis. MetaboAnalyst (4.0), the R-code-based online analysis tool for metabolomics, provides a convenient one-stop shop for metabolomics data analysis (screening of characteristic markers, cluster analysis, and enrichment of metabolic pathways), interpretation, and integration with other omics data.17 The basic flow chart of metabolomics research is shown (FIGURE 1).

Basic flow chart of metabolomics research. PLS-DA, partial least squares-discriminant analysis.
FigURE 1.

Basic flow chart of metabolomics research. PLS-DA, partial least squares-discriminant analysis.

Metabolomics Analysis Platform

Given that metabolomics requires the detection of large samples, high-throughput analytical platforms play a crucial role in this. Currently, metabolomics typically employs NMR, MS, LC/GC, and coupling of various analytical platforms to analyze and map metabolites.18 The coupling of these techniques can improve the resolution, sensitivity, and selectivity during sample analysis and help to map more metabolic profiles. Different metabolomics analysis platforms have their own unique features14,19–22; the characteristics of the main metabolomics analysis platforms are shown in TABLE 1.

Table 1.

The Characteristics of the Main Metabolomics Analysis Platforms

Analysis PlatformAdvantagesDisadvantages
NMRSimple handling of samples;
noninvasive and unbiased detection and analysis;
lower cost
High investment in equipment;
relatively low detection sensitivity;
narrow dynamic detection range;
difficult to detect metabolic small molecules with relatively large differences in concentration simultaneously
MSExcellent repeatability and objectivity;
high sensitivity and wide detection range;
rapid analysis and identification of multiple chemicals simultaneously
Preparation of large quantities of samples;
relatively high daily costs;
requires some knowledge of the metabolic small molecules being tested
LC-MSHigh sensitivity;
no need for high temperature;
no derivatization of the sample is required;
relatively simple sample preparation compared to GC-MS
Poor repeatability;
database not yet robust
GC-MSHigh precision, sensitivity and durability;
excellent performance in the analysis of volatile substances;
library of standard spectra available
Sample derivatization;
more cumbersome sample preparation;
difficult to use for difficult volatiles
Analysis PlatformAdvantagesDisadvantages
NMRSimple handling of samples;
noninvasive and unbiased detection and analysis;
lower cost
High investment in equipment;
relatively low detection sensitivity;
narrow dynamic detection range;
difficult to detect metabolic small molecules with relatively large differences in concentration simultaneously
MSExcellent repeatability and objectivity;
high sensitivity and wide detection range;
rapid analysis and identification of multiple chemicals simultaneously
Preparation of large quantities of samples;
relatively high daily costs;
requires some knowledge of the metabolic small molecules being tested
LC-MSHigh sensitivity;
no need for high temperature;
no derivatization of the sample is required;
relatively simple sample preparation compared to GC-MS
Poor repeatability;
database not yet robust
GC-MSHigh precision, sensitivity and durability;
excellent performance in the analysis of volatile substances;
library of standard spectra available
Sample derivatization;
more cumbersome sample preparation;
difficult to use for difficult volatiles

GC-MS, gas chromatography-mass spectrometry; LC-MS, liquid chromatography–mass spectrometry; MS, mass spectrometry; NMR, nuclear magnetic resonance.

Table 1.

The Characteristics of the Main Metabolomics Analysis Platforms

Analysis PlatformAdvantagesDisadvantages
NMRSimple handling of samples;
noninvasive and unbiased detection and analysis;
lower cost
High investment in equipment;
relatively low detection sensitivity;
narrow dynamic detection range;
difficult to detect metabolic small molecules with relatively large differences in concentration simultaneously
MSExcellent repeatability and objectivity;
high sensitivity and wide detection range;
rapid analysis and identification of multiple chemicals simultaneously
Preparation of large quantities of samples;
relatively high daily costs;
requires some knowledge of the metabolic small molecules being tested
LC-MSHigh sensitivity;
no need for high temperature;
no derivatization of the sample is required;
relatively simple sample preparation compared to GC-MS
Poor repeatability;
database not yet robust
GC-MSHigh precision, sensitivity and durability;
excellent performance in the analysis of volatile substances;
library of standard spectra available
Sample derivatization;
more cumbersome sample preparation;
difficult to use for difficult volatiles
Analysis PlatformAdvantagesDisadvantages
NMRSimple handling of samples;
noninvasive and unbiased detection and analysis;
lower cost
High investment in equipment;
relatively low detection sensitivity;
narrow dynamic detection range;
difficult to detect metabolic small molecules with relatively large differences in concentration simultaneously
MSExcellent repeatability and objectivity;
high sensitivity and wide detection range;
rapid analysis and identification of multiple chemicals simultaneously
Preparation of large quantities of samples;
relatively high daily costs;
requires some knowledge of the metabolic small molecules being tested
LC-MSHigh sensitivity;
no need for high temperature;
no derivatization of the sample is required;
relatively simple sample preparation compared to GC-MS
Poor repeatability;
database not yet robust
GC-MSHigh precision, sensitivity and durability;
excellent performance in the analysis of volatile substances;
library of standard spectra available
Sample derivatization;
more cumbersome sample preparation;
difficult to use for difficult volatiles

GC-MS, gas chromatography-mass spectrometry; LC-MS, liquid chromatography–mass spectrometry; MS, mass spectrometry; NMR, nuclear magnetic resonance.

Early Diagnosis of DKD Based on Metabolomics

Although DKD still faces great challenges in terms of treatment, early diagnosis of DKD has become a promising research direction. The gold standard for the diagnosis of DKD is renal puncture biopsy, but this invasive test is difficult for patients to accept in the clinical setting. At present, DKD is mostly diagnosed clinically, and the main diagnostic criteria are eGFR and UACR. As mentioned above, eGFR and UACR still have some limitations as diagnostic criteria for DKD, which forces us to search for biological markers with higher sensitivity and specificity. Metabolomics, with its high-throughput detection technology and unique data processing methods, is uniquely positioned to find specific biological markers (TABLE 2).23–36

Table 2.

Biological Markers in the Early Diagnosis of Diabetic Kidney Disease (DKD)

ResearchersCountryPlatformSpeciesSampleMajor Differential MetabolitesMetabolic Pathways
Peng et al23ChinaLC-MSHumanPlasma5-HET, LTB4, 5,6-DHET, 14,15-DHET, and 9,10-diHOMELipoxygenase metabolites and cytochrome P450s metabolic pathway
Devi et al24IndiaHPLC-MSHumanPlasmaAcyl ethanolamides, acetylcholine, monoacylglycerols, and cortisol
Gordin et al25USALC-MSHumanPlasmaSorbitol, aconitate, and fumarateGlycolytic, polyol, and tricarboxylic acid cycle pathways
Liu et al26ChinaHPLC-MSHumanUrine5-Hydroxyindoleacetic acid, deoxycholic acid, nutriacholic acidTryptophan metabolism, bile acid metabolism, and glycine metabolism pathway
Ng et al27SingaporeGC-MSHumanUrineOctanol, oxalic acid, phosphoric acid, benzamide, creatinine, and N-acetylglutamine
Tan et al28Singapore,LC-MSHumanPlasmaGlutamine, phenylacetylglutamine, 3-indoxyl sulfate, xanthine, and dimethyluric acid
Zhu et al29ChinaNPLC-TOF/MSHumanPlasmaLPC (C18:2), PE (C16:0/18:1), PE (pC18:0/20:4), PI (C18:0/22:6), PS (C18:0/18:0), SM (dC18:0/20:2)
Hirayama et al30JapanCE-TOF/MSHumanSerumCreatinine, aspartic acid, γ-butyrobetaine, citrulline, and symmetric dimethylarginine
Zhang et al31ChinaHPLC-MSHumanSerumL-Tryptophan, 5-hydroxyindoleacetic acid, indole-3-acetamideTryptophan metabolism pathway
Shao et al32ChinaGC-TOF/MSHumanSerum and urineSerum: benzoic acid, fumaric acid, erythrose, fructose 6-phosphate, taurine, and L-glutamine.
Urine: D-glucose, L-valine, L-histidine, sucrose, gluconic acid, glycine, and oxalic acid
Serum: 9 metabolic pathways
Urine: 12 metabolic pathways
Zhang et al33ChinaUPLC-MSHumanSerumHexadecanoic acid (C16:0), linolelaidic acid (C18:2N6T), linoleic acid (C18:2N6C), piperidine, and azoxystrobin acidlinoleic acid metabolism, aminoacyl-tRNA biosynthesis, and arginine metabolism
Du et al34ChinaGC/LC–MSRatPlasmaOleic acid, glutamate, and guanosine
Ma et al35ChinaUPLC-MSHumanUrineDihydrouracil, ureidopropionic acid, and pantothenic acidPantothenate and coenzyme A biosynthesis pathway
Dai et al36ChinaUPLC-TOF/MSRatSerum and urineSerum: guanosine triphosphate, lysoPC(18:1), retinyl ester, etc
Urine: aminoadipic acid, adenine, and 2-oxo-4-methylthiobutanoic acid, etc
Serum: purinem, alanine, aspartate, and glutamate metabolism
Urine: purine, lysine degradation, and sphingolipid metabolism
ResearchersCountryPlatformSpeciesSampleMajor Differential MetabolitesMetabolic Pathways
Peng et al23ChinaLC-MSHumanPlasma5-HET, LTB4, 5,6-DHET, 14,15-DHET, and 9,10-diHOMELipoxygenase metabolites and cytochrome P450s metabolic pathway
Devi et al24IndiaHPLC-MSHumanPlasmaAcyl ethanolamides, acetylcholine, monoacylglycerols, and cortisol
Gordin et al25USALC-MSHumanPlasmaSorbitol, aconitate, and fumarateGlycolytic, polyol, and tricarboxylic acid cycle pathways
Liu et al26ChinaHPLC-MSHumanUrine5-Hydroxyindoleacetic acid, deoxycholic acid, nutriacholic acidTryptophan metabolism, bile acid metabolism, and glycine metabolism pathway
Ng et al27SingaporeGC-MSHumanUrineOctanol, oxalic acid, phosphoric acid, benzamide, creatinine, and N-acetylglutamine
Tan et al28Singapore,LC-MSHumanPlasmaGlutamine, phenylacetylglutamine, 3-indoxyl sulfate, xanthine, and dimethyluric acid
Zhu et al29ChinaNPLC-TOF/MSHumanPlasmaLPC (C18:2), PE (C16:0/18:1), PE (pC18:0/20:4), PI (C18:0/22:6), PS (C18:0/18:0), SM (dC18:0/20:2)
Hirayama et al30JapanCE-TOF/MSHumanSerumCreatinine, aspartic acid, γ-butyrobetaine, citrulline, and symmetric dimethylarginine
Zhang et al31ChinaHPLC-MSHumanSerumL-Tryptophan, 5-hydroxyindoleacetic acid, indole-3-acetamideTryptophan metabolism pathway
Shao et al32ChinaGC-TOF/MSHumanSerum and urineSerum: benzoic acid, fumaric acid, erythrose, fructose 6-phosphate, taurine, and L-glutamine.
Urine: D-glucose, L-valine, L-histidine, sucrose, gluconic acid, glycine, and oxalic acid
Serum: 9 metabolic pathways
Urine: 12 metabolic pathways
Zhang et al33ChinaUPLC-MSHumanSerumHexadecanoic acid (C16:0), linolelaidic acid (C18:2N6T), linoleic acid (C18:2N6C), piperidine, and azoxystrobin acidlinoleic acid metabolism, aminoacyl-tRNA biosynthesis, and arginine metabolism
Du et al34ChinaGC/LC–MSRatPlasmaOleic acid, glutamate, and guanosine
Ma et al35ChinaUPLC-MSHumanUrineDihydrouracil, ureidopropionic acid, and pantothenic acidPantothenate and coenzyme A biosynthesis pathway
Dai et al36ChinaUPLC-TOF/MSRatSerum and urineSerum: guanosine triphosphate, lysoPC(18:1), retinyl ester, etc
Urine: aminoadipic acid, adenine, and 2-oxo-4-methylthiobutanoic acid, etc
Serum: purinem, alanine, aspartate, and glutamate metabolism
Urine: purine, lysine degradation, and sphingolipid metabolism

GC-MS, gas chromatography-mass spectrometry; HPLC-MS, high performance liquid chromatography-mass spectrometry; LC-MS, liquid chromatography–mass spectrometry.

Table 2.

Biological Markers in the Early Diagnosis of Diabetic Kidney Disease (DKD)

ResearchersCountryPlatformSpeciesSampleMajor Differential MetabolitesMetabolic Pathways
Peng et al23ChinaLC-MSHumanPlasma5-HET, LTB4, 5,6-DHET, 14,15-DHET, and 9,10-diHOMELipoxygenase metabolites and cytochrome P450s metabolic pathway
Devi et al24IndiaHPLC-MSHumanPlasmaAcyl ethanolamides, acetylcholine, monoacylglycerols, and cortisol
Gordin et al25USALC-MSHumanPlasmaSorbitol, aconitate, and fumarateGlycolytic, polyol, and tricarboxylic acid cycle pathways
Liu et al26ChinaHPLC-MSHumanUrine5-Hydroxyindoleacetic acid, deoxycholic acid, nutriacholic acidTryptophan metabolism, bile acid metabolism, and glycine metabolism pathway
Ng et al27SingaporeGC-MSHumanUrineOctanol, oxalic acid, phosphoric acid, benzamide, creatinine, and N-acetylglutamine
Tan et al28Singapore,LC-MSHumanPlasmaGlutamine, phenylacetylglutamine, 3-indoxyl sulfate, xanthine, and dimethyluric acid
Zhu et al29ChinaNPLC-TOF/MSHumanPlasmaLPC (C18:2), PE (C16:0/18:1), PE (pC18:0/20:4), PI (C18:0/22:6), PS (C18:0/18:0), SM (dC18:0/20:2)
Hirayama et al30JapanCE-TOF/MSHumanSerumCreatinine, aspartic acid, γ-butyrobetaine, citrulline, and symmetric dimethylarginine
Zhang et al31ChinaHPLC-MSHumanSerumL-Tryptophan, 5-hydroxyindoleacetic acid, indole-3-acetamideTryptophan metabolism pathway
Shao et al32ChinaGC-TOF/MSHumanSerum and urineSerum: benzoic acid, fumaric acid, erythrose, fructose 6-phosphate, taurine, and L-glutamine.
Urine: D-glucose, L-valine, L-histidine, sucrose, gluconic acid, glycine, and oxalic acid
Serum: 9 metabolic pathways
Urine: 12 metabolic pathways
Zhang et al33ChinaUPLC-MSHumanSerumHexadecanoic acid (C16:0), linolelaidic acid (C18:2N6T), linoleic acid (C18:2N6C), piperidine, and azoxystrobin acidlinoleic acid metabolism, aminoacyl-tRNA biosynthesis, and arginine metabolism
Du et al34ChinaGC/LC–MSRatPlasmaOleic acid, glutamate, and guanosine
Ma et al35ChinaUPLC-MSHumanUrineDihydrouracil, ureidopropionic acid, and pantothenic acidPantothenate and coenzyme A biosynthesis pathway
Dai et al36ChinaUPLC-TOF/MSRatSerum and urineSerum: guanosine triphosphate, lysoPC(18:1), retinyl ester, etc
Urine: aminoadipic acid, adenine, and 2-oxo-4-methylthiobutanoic acid, etc
Serum: purinem, alanine, aspartate, and glutamate metabolism
Urine: purine, lysine degradation, and sphingolipid metabolism
ResearchersCountryPlatformSpeciesSampleMajor Differential MetabolitesMetabolic Pathways
Peng et al23ChinaLC-MSHumanPlasma5-HET, LTB4, 5,6-DHET, 14,15-DHET, and 9,10-diHOMELipoxygenase metabolites and cytochrome P450s metabolic pathway
Devi et al24IndiaHPLC-MSHumanPlasmaAcyl ethanolamides, acetylcholine, monoacylglycerols, and cortisol
Gordin et al25USALC-MSHumanPlasmaSorbitol, aconitate, and fumarateGlycolytic, polyol, and tricarboxylic acid cycle pathways
Liu et al26ChinaHPLC-MSHumanUrine5-Hydroxyindoleacetic acid, deoxycholic acid, nutriacholic acidTryptophan metabolism, bile acid metabolism, and glycine metabolism pathway
Ng et al27SingaporeGC-MSHumanUrineOctanol, oxalic acid, phosphoric acid, benzamide, creatinine, and N-acetylglutamine
Tan et al28Singapore,LC-MSHumanPlasmaGlutamine, phenylacetylglutamine, 3-indoxyl sulfate, xanthine, and dimethyluric acid
Zhu et al29ChinaNPLC-TOF/MSHumanPlasmaLPC (C18:2), PE (C16:0/18:1), PE (pC18:0/20:4), PI (C18:0/22:6), PS (C18:0/18:0), SM (dC18:0/20:2)
Hirayama et al30JapanCE-TOF/MSHumanSerumCreatinine, aspartic acid, γ-butyrobetaine, citrulline, and symmetric dimethylarginine
Zhang et al31ChinaHPLC-MSHumanSerumL-Tryptophan, 5-hydroxyindoleacetic acid, indole-3-acetamideTryptophan metabolism pathway
Shao et al32ChinaGC-TOF/MSHumanSerum and urineSerum: benzoic acid, fumaric acid, erythrose, fructose 6-phosphate, taurine, and L-glutamine.
Urine: D-glucose, L-valine, L-histidine, sucrose, gluconic acid, glycine, and oxalic acid
Serum: 9 metabolic pathways
Urine: 12 metabolic pathways
Zhang et al33ChinaUPLC-MSHumanSerumHexadecanoic acid (C16:0), linolelaidic acid (C18:2N6T), linoleic acid (C18:2N6C), piperidine, and azoxystrobin acidlinoleic acid metabolism, aminoacyl-tRNA biosynthesis, and arginine metabolism
Du et al34ChinaGC/LC–MSRatPlasmaOleic acid, glutamate, and guanosine
Ma et al35ChinaUPLC-MSHumanUrineDihydrouracil, ureidopropionic acid, and pantothenic acidPantothenate and coenzyme A biosynthesis pathway
Dai et al36ChinaUPLC-TOF/MSRatSerum and urineSerum: guanosine triphosphate, lysoPC(18:1), retinyl ester, etc
Urine: aminoadipic acid, adenine, and 2-oxo-4-methylthiobutanoic acid, etc
Serum: purinem, alanine, aspartate, and glutamate metabolism
Urine: purine, lysine degradation, and sphingolipid metabolism

GC-MS, gas chromatography-mass spectrometry; HPLC-MS, high performance liquid chromatography-mass spectrometry; LC-MS, liquid chromatography–mass spectrometry.

Currently, most of the biological markers identified based on metabolomics that contribute to the early diagnosis of DKD are focused on fatty acids, amino acids, lipids, and nucleotide metabolites. Fatty acids have an important impact on the metabolism of the body, and fat mobilization can increase the level of nitric oxide (NO) accumulation in the matrix, ultimately leading to increased glomerulosclerosis.37 Some studies have shown that pathologically elevated saturated fatty acids have a strong toxic effect on cells,34 which in turn promotes the development and progression of diabetes.28 The role of unsaturated fatty acids in DKD is still a controversial topic. Some studies have shown that oleic acid ameliorates palmitic acid-induced endoplasmic reticulum stress, inflammation, and insulin resistance38,39; however, other researchers suggested that high levels of oleic acid cause endoplasmic reticulum stress and apoptosis, causing proliferation or damage to glomerular thylakoid cells.40,41 Phospholipids are key components of the phospholipid bilayer of biological membranes, which includes glycerophospholipids and sphingolipids. Peng et al42 showed that the expression levels of phosphatidylethanolamine (m/z = 750), phosphatidylglycerol (m/z = 747), and phosphatidylcholine (m/z = 802) were lower in DKD patients than in healthy controls. In addition, Zhu et al29 identified plasma phospholipids in T2DM and DKD patients based on an HPLC-ESI/MS platform and screened 18 phospholipids (VIP >1, P < .05) as potential biomarkers by PLS-DA data analysis, including 2 novel biomarkers, phosphatidylinositol (m/z = 909) and sphingomyelin (m/z = 801). Several amino acid metabolic abnormalities were also observed in DKD individuals. Glutamate has an important role in regulating insulin secretion and maintaining glucose homeostasis in the body,43 which has been suggested as a potential biological marker for early diagnosis of DKD.34 In addition, plasma tryptophan levels were found to be positively correlated with estimated glomerular filtration rate in studies,31 while its metabolite, kynurenine, was negatively correlated with eGFR.30

Prognostic Risk Assessment for DKD Based on Metabolomics

Metabolites in the organism are constantly changing dynamically in response to internal and external factors, and fluctuations in the metabolic profile suggest changes in the physiological/pathological state of the organism. By mapping and analyzing the metabolic profile of the organism, the disease progression and prognosis would be better assessed. Tavares et al44 investigated the role of plasma metabolites in the assessment of DKD prognosis by an untargeted metabolomics approach. The results of the research showed that an adverse event (death, doubling of blood creatinine, or dialysis treatment) occurred in 30.3% of DKD patients during 2.5 years of follow-up; univariate Cox regression analysis showed that 1,5-anhydroglucitol (hazard ratio (HR): 0.10, 95% CI: 0.02–0.63, P =.01), norvaline (HR: 0.01, 95% CI: 0.001–0.4, P =.01), and aspartic acid (HR: 0.12, 95% CI: 0.02–0.64, P =.01) were negatively associated with the occurrence of DKD adverse events. Niewczas et al45 conducted a prospective study with 158 patients with type 1 diabetic kidney disease followed up for a long period of time (median follow-up, 11.5 years). During the follow-up period, 99 patients (63%) progressed to ESRD; for the shorter the duration of diabetes, the younger the age, and the poorer the glycemic control, the faster the decline in renal function.45 To further find the independent risk factors for the progression of DKD, they examined the serum of patients by high-throughput metabolomics technology, and the results revealed that 7 modified metabolites, including C-glycosyltryptophan, pseudouridine, O-sulfotyrosine, N-acetylthreonine, N-acetylserine, N6-carbamoylthreonyladenosine, and N6-acetyllysine, were independent risk factors for the progression of DKD to ERSD.45 Another comprehensive study of plasma metabolic profiles in patients with type 2 diabetes mellitus showed that certain metabolites (amino acids and their derivatives) predicted the onset of ESRD approximately 10 years earlier and independent of baseline albuminuria and renal function.46 Tofte et al47 illustrated that alterations in plasma metabolites (particularly polyols and branched-chain amino acids) were associated with future renal impairment in the type 1 diabetes mellitus (T1DM) population; ribonucleic acid was associated with a higher risk, while the amino acids isoleucine, leucine, and valine were associated with a lower risk of combined renal endpoint (≥30% decline in eGFR, ESRD, and all-cause mortality). Acylcarnitine is essential for intracellular energy metabolism and plays an essential role in the β-oxidation metabolism of fatty acids. Research has shown impaired fatty acid oxidation in patients with DKD; the abnormal plasma acylcarnitine levels could be a reflection of the degree of disease progression.48,49 It has also been shown that metabolites such as uremic toxin and carnitine were strongly associated with the progression of microproteinuria in patients with T1DM.50 Looker et al51 showed that 14 markers, such as symmetrical dimethylarginine, uracil, acylcarnitine, and hydroxyproline, were associated with a rapid decline in renal function in patients with T2DM, suggesting that their combination might greatly improve the prediction of renal function in the future (TABLE 3).44–47,49–51

Table 3.

Prognostic Markers in Diabetic Kidney Disease

ResearchersCountryPlatformSpeciesSampleMajor Differential Metabolites
Tavares et al44BrazilGC-MSHumanPlasma1,5-anhydroglucitol, norvaline, and aspartic acid
Niewczas et al45USAGC/LC-MSHumanSerumC-glycosyltryptophan, pseudouridine, O-sulfotyrosine, N-acetylthreonine, N-acetylserine, N6-carbamoylthreonyladenosine and N6-acetyllysine
Niewczas et al46USAMetabolonHumanPlasmaEssential amino acids and their derivatives, acylcarnitines
Tofte et al47DenmarkGC-TOF/MSHumanSerumRibonic acid, myo-inositol, hydroxy butyrate 3,4-dihydroxybutanoic acid, and branched chain amino acids
Afshinni et al49USAMSHumanSerumPolyunsaturated triacylglycerols and C16-C20 acylcarnitines
Haukka et al50FinlandUPLC-MSHumanSerumErythritol, 3-phenylpropionate, and N-trimethyl-5-aminovalerate
Looker et al51UKLC-MSHumanSerumSymmetrical dimethylarginine, uracil, acylcarnitine, and hydroxyproline
ResearchersCountryPlatformSpeciesSampleMajor Differential Metabolites
Tavares et al44BrazilGC-MSHumanPlasma1,5-anhydroglucitol, norvaline, and aspartic acid
Niewczas et al45USAGC/LC-MSHumanSerumC-glycosyltryptophan, pseudouridine, O-sulfotyrosine, N-acetylthreonine, N-acetylserine, N6-carbamoylthreonyladenosine and N6-acetyllysine
Niewczas et al46USAMetabolonHumanPlasmaEssential amino acids and their derivatives, acylcarnitines
Tofte et al47DenmarkGC-TOF/MSHumanSerumRibonic acid, myo-inositol, hydroxy butyrate 3,4-dihydroxybutanoic acid, and branched chain amino acids
Afshinni et al49USAMSHumanSerumPolyunsaturated triacylglycerols and C16-C20 acylcarnitines
Haukka et al50FinlandUPLC-MSHumanSerumErythritol, 3-phenylpropionate, and N-trimethyl-5-aminovalerate
Looker et al51UKLC-MSHumanSerumSymmetrical dimethylarginine, uracil, acylcarnitine, and hydroxyproline

GC-MS, gas chromatography-mass spectrometry; GC/LC-MS, gas chromatography/liquid chromatography-mass spectrometry; UPLC-MS, ultra-high performance liquid chromatography-mass spectrometry.

Table 3.

Prognostic Markers in Diabetic Kidney Disease

ResearchersCountryPlatformSpeciesSampleMajor Differential Metabolites
Tavares et al44BrazilGC-MSHumanPlasma1,5-anhydroglucitol, norvaline, and aspartic acid
Niewczas et al45USAGC/LC-MSHumanSerumC-glycosyltryptophan, pseudouridine, O-sulfotyrosine, N-acetylthreonine, N-acetylserine, N6-carbamoylthreonyladenosine and N6-acetyllysine
Niewczas et al46USAMetabolonHumanPlasmaEssential amino acids and their derivatives, acylcarnitines
Tofte et al47DenmarkGC-TOF/MSHumanSerumRibonic acid, myo-inositol, hydroxy butyrate 3,4-dihydroxybutanoic acid, and branched chain amino acids
Afshinni et al49USAMSHumanSerumPolyunsaturated triacylglycerols and C16-C20 acylcarnitines
Haukka et al50FinlandUPLC-MSHumanSerumErythritol, 3-phenylpropionate, and N-trimethyl-5-aminovalerate
Looker et al51UKLC-MSHumanSerumSymmetrical dimethylarginine, uracil, acylcarnitine, and hydroxyproline
ResearchersCountryPlatformSpeciesSampleMajor Differential Metabolites
Tavares et al44BrazilGC-MSHumanPlasma1,5-anhydroglucitol, norvaline, and aspartic acid
Niewczas et al45USAGC/LC-MSHumanSerumC-glycosyltryptophan, pseudouridine, O-sulfotyrosine, N-acetylthreonine, N-acetylserine, N6-carbamoylthreonyladenosine and N6-acetyllysine
Niewczas et al46USAMetabolonHumanPlasmaEssential amino acids and their derivatives, acylcarnitines
Tofte et al47DenmarkGC-TOF/MSHumanSerumRibonic acid, myo-inositol, hydroxy butyrate 3,4-dihydroxybutanoic acid, and branched chain amino acids
Afshinni et al49USAMSHumanSerumPolyunsaturated triacylglycerols and C16-C20 acylcarnitines
Haukka et al50FinlandUPLC-MSHumanSerumErythritol, 3-phenylpropionate, and N-trimethyl-5-aminovalerate
Looker et al51UKLC-MSHumanSerumSymmetrical dimethylarginine, uracil, acylcarnitine, and hydroxyproline

GC-MS, gas chromatography-mass spectrometry; GC/LC-MS, gas chromatography/liquid chromatography-mass spectrometry; UPLC-MS, ultra-high performance liquid chromatography-mass spectrometry.

Study of the Pathogenesis of DKD on Metabolomics

Metabolic profiles represent the complete collection of metabolites in an organism, and perturbations in metabolic profiles may help to reveal the pathogenesis of the disease early. The convergence of multiple omics plays an important role in the study of DKD pathogenesis. Recent studies have shown that a novel oral hypoglycemic agent (SGLT-1) could act as a renoprotective agent by inhibiting glucose reabsorption and reducing renal tubular energy expenditure,52–54 which laterally suggested an energy imbalance in the renal tissues of DKD patients. Sharma et al,55 through urinary metabolomics studies in DKD patients, identified 13 metabolites whose expression was significantly reduced, and the reduction in organic anion expression was associated with the downregulation of the expression of the organic anion transporter (OAT) gene in DKD patients. Bioinformatics analysis revealed that 12 of the 13 differentially expressed metabolites were associated with mitochondrial metabolism and that the expression of PGC1α, a major regulator of mitochondrial activity, was significantly lower in the renal tissue of DKD patients (overall inhibition of mitochondrial activity).55 Multiple metabolic pathways (pentose phosphate, purine and pyrimidine metabolism, hexosamine biosynthesis, and tricarboxylic acid cycle) were overactivated, and mitochondrial complex 1 activity was enhanced in DKD mouse models (streptozotocin-induced), while specific mitochondrial complex 1 inhibitors were effective in reducing glomerular and tubular injury through modulation of the body’s metabolic state and oxidative stress.56 Some scholars have reported abnormalities in fatty acid metabolism in DKD.33 Saulnier et al57 showed a correlation between urinary metabolites and renal structure in patients with DKD, where short-chain fatty acids in urine were associated with structural destruction of endothelial cells in the early stages of DKD. In addition, other studies have shown that defects in fatty acid oxidation play a key role in the development of renal fibrosis.48 Multi-omics studies in DKD have revealed that accumulation of extracellular matrix, abnormal activation of the inflammatory microenvironment, and metabolic disturbances contributed to the development of glomerulosclerosis and tubulointerstitial fibrosis. Further integrative analysis revealed that linoleic acid metabolism and fatty acid β-oxidation are significantly inhibited in the pathogenesis and progression of DKD.58 Carnitine is an important substance in the oxidative metabolism of fatty acids and protects against cellular oxidative stress from various sources59; indeed, Devi et al24 showed abnormal carnitine metabolism in patients with DKD. The role of NADPH oxidase 4 in the development and progression of DKD is poorly understood. You et al60 demonstrated the presence of abnormal tricarboxylic acid cycle-related metabolites in DKD mice and that NADPH oxidase 4 could cause an increase in ferredoxin levels. This in turn led to endoplasmic reticulum oxidative stress and increased expression of HIF-1α and TGF-β. Chinese medicines are increasingly used in the treatment of DKD, but the mechanism of action is not well understood. Studies in network pharmacology combined with metabolomics have shown that herbal medicines (shenyankangfu tablets) may improve insulin resistance by regulating the biosynthesis of unsaturated fatty acids and the metabolism of starch and sucrose.61 Epigenetics has become more widely accepted in the pathogenesis of DKD.62 High glucose conditions may alter the histone amino acid modification status (methylation, acetylation), causing changes in chromatin structure, facilitating the binding of transcription factors, and ultimately leading to the expression of genes associated with inflammation and fibrosis.63 Epigenetic markers could not only lead to abnormal expression of metabolism-related genes64 but may also predispose offspring to metabolic abnormalities and shortened lifespan.65 A prospective cohort study by Niewczas et al45 demonstrated that circulating levels of 7 modified metabolites (C-glycosyltryptophan, pseudouridine, O-sulfotyrosine, N-acetylthreonine, N-acetylserine, N6-carbamoylthreonyladenosine, and N6-acetyllysine) were associated with the rate of decline in renal function and the risk of ESRD. It has also been observed that elevated levels of serum modified amino acid metabolites (O-sulfotyrosine) were highly correlated with chronic decompensation of renal function.66

Conclusion

Metabolomics, as an important component of systems biology, is expanding its application in DKD clinical research. Numerous small molecule metabolites (such as amino acids and lipids) in the body have been screened and are expected to become biological markers for the early diagnosis of DKD. The evolution of the metabolic profile with the disease course gives us enough opportunities to evaluate the prognosis of DKD, enabling us to make early interventions. Metabolomics as a new tool gives us a new perspective through which to look at the pathogenesis of DKD. Powered by information biology techniques, we have understood that there are disorders of mitochondrial energy metabolism within individuals with DKD, which severely compromise the structure and function of the kidney. Epigenetics plays an important role in the pathogenesis of DKD, and an increasing number of researches show the presence of abnormal modified metabolites in DKD patients. In addition, metabolomics also has given herbal medicine an arena to demonstrate its efficacy. In the future, with the innovation of high-throughput analysis platforms, cross-fertilization of multi-omics, and comprehensive infiltration of information biotechnology, we will be further exposed to the nature of DKD, which is necessary if we are to overcome it.

Abbreviations

    Abbreviations
     
  • DKD

    diabetic kidney disease

  •  
  • DM

    diabetes mellitus

  •  
  • ESRD

    end-stage renal disease

  •  
  • eGFR

    estimation of glomerular filtration rate

  •  
  • UACR

    urine albumin creatinine ratio

  •  
  • NMR

    nuclear magnetic resonance

  •  
  • LC/GC

    liquid chromatography/gas chromatography

  •  
  • MS

    mass spectrometry

  •  
  • PLS-DA

    partial least squares-discriminant analysis

  •  
  • PCA

    principal component analysis

  •  
  • VIP

    variable importance of projection

  •  
  • NO

    nitric oxide

  •  
  • T1DM

    type 1 diabetes mellitus

  •  
  • OAT

    organic anion transporter

  •  
  • HR

    hazard ratio

Funding

Grant of Zhejiang Medicine and Health Technology Project, China (No. 2018ZH029, 2020KY871), the Major Project for Science & Technology Innovation 2025 in Ningbo, China (No. 2019B10035), and Grant of Ningbo Social Development, China (No. 2019C50080).

REFERENCES

1.

Saeedi
P
,
Petersohn
I
,
Salpea
P
, et al.
Global and regional diabetes prevalence estimates for 2019 and projections for 2030 and 2045: results from the International Diabetes Federation Diabetes Atlas, 9th edition
.
Diabetes Res Clin Pract.
2019
;
157
:
107843
. doi:10.1016/j.diabres.2019.107843.

2.

Ruiz-Ortega
M
,
Rodrigues-Diez
RR
,
Lavoz
C
,
Rayego-Mateos
S
.
Special issue “Diabetic nephropathy: diagnosis, prevention, and treatment.”
Clin Med.
2020
;
9
(
3):8–13.

3.

Alicic
RZ
,
Rooney
MT
,
Tuttle
KR
.
Diabetic kidney disease: challenges, progress, and possibilities
.
Clin J Am Soc Nephrol.
2017
;
12
(
12
):
2032
2045
.

4.

Parving
H-H
,
Chaturvedi
N
,
Viberti
G
,
Mogensen
CE
.
Does microalbuminuria predict diabetic nephropathy?
Diabetes Care.
2002
;
25
(
2
):
406
407
. doi:10.2337/diacare.25.2.406.

5.

Vistisen
D
,
Andersen
GS
,
Hulman
A
,
Persson
F
,
Rossing
P
,
Jørgensen
ME
.
Progressive decline in estimated glomerular filtration rate in patients with diabetes after moderate loss in kidney function—even without albuminuria
.
Diabetes Care.
2019
;
42
(
10
):
1886
1894
. doi:10.2337/dc19-0349.

6.

Gates
SC
,
Sweeley
CC
.
Quantitative metabolic profiling based on gas chromatography
.
Clin Chem.
1978
;
24
(
10
):
1663
1673
.

7.

Nicholson
JK
,
Lindon
JC
,
Holmes
E
.
Metabonomics: understanding the metabolic responses of living systems to pathophysiological stimuli via multivariate statistical analysis of biological NMR spectroscopic data
.
Xenobiotica.
1999
;
29
(
11
):
1181
1189
.

8.

Beger
RD
,
Dunn
W
,
Schmidt
MA
, et al.
Metabolomics enables precision medicine: “a white paper, community perspective.”
Metabolomics.
2016
;
12
(
10
):
149
.

9.

Costa Dos Santos
G
,
Renovato-Martins
M
,
de Brito
NM
.
The remodel of the “central dogma”: a metabolomics interaction perspective
.
Metabolomics.
2021
;
17
(
5
):
48
.

10.

Pinu
FR
,
Villas-Boas
SG
.
Extracellular microbial metabolomics: the state of the art
.
Metabolites.
2017
;
7
(
3):43.

11.

Ott
K-H
,
Araníbar
N
,
Singh
B
,
Stockton
GW
.
Metabonomics classifies pathways affected by bioactive compounds: artificial neural network classification of NMR spectra of plant extracts
.
Phytochem.
2003
;
62
(
6
):
971
985
.

12.

Taylor
J
,
King
RD
,
Altmann
T
,
Fiehn
O
.
Application of metabolomics to plant genotype discrimination using statistics and machine learning
.
Bioinformatics.
2002
;
18
(
Suppl 2
):
S241
S248
.

13.

Beckonert
O
,
Keun
HC
,
Ebbels
TMD
, et al.
Metabolic profiling, metabolomic and metabonomic procedures for NMR spectroscopy of urine, plasma, serum and tissue extracts
.
Nat Protocols.
2007
;
2
(
11
):
2692
2703
.

14.

Zhang
A
,
Sun
H
,
Wang
P
,
Han
Y
,
Wang
X
.
Modern analytical techniques in metabolomics analysis
.
Analyst.
2012
;
137
(
2
):
293
300
.

15.

Liu
R
,
Yang
Z
.
Single cell metabolomics using mass spectrometry: techniques and data analysis
.
Anal Chim Acta.
2021
;
1143
:
124
134
.

16.

Olsson
M
,
Hellman
U
,
Wixner
J
,
Anan
I
.
Metabolomics analysis for diagnosis and biomarker discovery of transthyretin amyloidosis
.
Amyloid.
2021
;
28
(
4
):
234
242
.

17.

Chong
J
,
Soufan
O
,
Li
C
, et al.
MetaboAnalyst 4.0: towards more transparent and integrative metabolomics analysis
.
Nucleic Acids Res.
2018
;
46
(
W1
):
W486
W494
.

18.

Li
B
,
He
X
,
Jia
W
,
Li
H
.
Novel applications of metabolomics in personalized medicine: a mini-review
.
Molecules.
2017
;
22
(
7):1173.

19.

Wishart
DS
.
Emerging applications of metabolomics in drug discovery and precision medicine
.
Nat Rev Drug Discov.
2016
;
15
(
7
):
473
484
.

20.

Beale
DJ
,
Pinu
FR
,
Kouremenos
KA
, et al.
Review of recent developments in GC-MS approaches to metabolomics-based research
.
Metabolomics.
2018
;
14
(
11
):
152
.

21.

Pasikanti
KK
,
Ho
PC
,
Chan
ECY
.
Gas chromatography/mass spectrometry in metabolic profiling of biological fluids
.
J Chromatogr B Analyt Technol Biomed Life Sci.
2008
;
871
(
2
):
202
211
.

22.

Gika
HG
,
Theodoridis
GA
,
Plumb
RS
,
Wilson
ID
.
Current practice of liquid chromatography-mass spectrometry in metabolomics and metabonomics
.
J Pharm Biomed Anal.
2014
;
87
:
12
25
.

23.

Peng
L
,
Sun
B
,
Liu
Y
, et al.
Increased lipoxygenase and decreased cytochrome P450s metabolites correlated with the incidence of diabetic nephropathy: potential role of eicosanoids from metabolomics in type 2 diabetic patients
.
Clin Exp Pharmacol Physiol.
2021
;
48
(
5
):
679
685
.

24.

Devi
S
,
Nongkhlaw
B
,
Limesh
M
, et al.
Acyl ethanolamides in diabetes and diabetic nephropathy: novel targets from untargeted plasma metabolomic profiles of South Asian Indian men
.
Sci Rep.
2019
;
9
(
1
):
18117
.

25.

Gordin
D
,
Shah
H
,
Shinjo
T
, et al.
Characterization of glycolytic enzymes and pyruvate kinase M2 in type 1 and 2 diabetic nephropathy
.
Diabetes Care.
2019
;
42
(
7
):
1263
1273
.

26.

Liu
Y
,
Chen
X
,
Liu
Y
, et al.
Metabolomic study of the protective effect of Gandi capsule for diabetic nephropathy
.
Chem Biol Interact.
2019
;
314
:
108815
.

27.

Ng
DPK
,
Salim
A
,
Liu
Y
, et al.
A metabolomic study of low estimated GFR in non-proteinuric type 2 diabetes mellitus
.
Diabetologia.
2012
;
55
(
2
):
499
508
.

28.

Tan
YM
,
Gao
Y
,
Teo
G
, et al.
Plasma metabolome and lipidome associations with type 2 diabetes and diabetic nephropathy
.
Metabolites
2021
;
11
(
4):228.

29.

Zhu
C
,
Liang
Q-l
,
Hu
P
,
Wang
Y-m
,
Luo
G-a
.
Phospholipidomic identification of potential plasma biomarkers associated with type 2 diabetes mellitus and diabetic nephropathy
.
Talanta
2011
;
85
(
4
):
1711
1720
.

30.

Hirayama
A
,
Nakashima
E
,
Sugimoto
M
, et al.
Metabolic profiling reveals new serum biomarkers for differentiating diabetic nephropathy
.
Anal Bioanal Chem.
2012
;
404
(
10
):
3101
3109
.

31.

Zhang
F
,
Guo
R
,
Cui
W
, et al.
Untargeted serum metabolomics and tryptophan metabolism profiling in type 2 diabetic patients with diabetic glomerulopathy
.
Ren Fail.
2021
;
43
(
1
):
980
992
.

32.

Shao
M
,
Lu
H
,
Yang
M
, et al.
Serum and urine metabolomics reveal potential biomarkers of T2DM patients with nephropathy
.
Ann. Transl. Med.
2020
;
8
(
5
):
199
.

33.

Zhang
H
,
Zuo
J-J
,
Dong
S-S
, et al.
Identification of potential serum metabolic biomarkers of diabetic kidney disease: a widely targeted metabolomics study
.
J Diabetes Res.
2020
;
2020
:
3049098
.

34.

Du
Y
,
Xu
B-J
,
Deng
X
, et al.
Predictive metabolic signatures for the occurrence and development of diabetic nephropathy and the intervention of Ginkgo biloba leaves extract based on gas or liquid chromatography with mass spectrometry
.
J Pharm Biomed Anal.
2019
;
166
:
30
39
.

35.

Ma
T
,
Liu
T
,
Xie
P
, et al.
UPLC-MS-based urine nontargeted metabolic profiling identifies dysregulation of pantothenate and CoA biosynthesis pathway in diabetic kidney disease
.
Life Sci.
2020
;
258
:
118160
.

36.

Dai
X
,
Su
S
,
Cai
H
, et al.
Protective effects of total glycoside from leaves on diabetic nephropathy rats via regulating the metabolic profiling and modulating the TGF-β1 and Wnt/β-catenin signaling pathway
.
Front Pharmacol.
2018
;
9
:
1012
.

37.

Toyama
T
,
Shimizu
M
,
Furuichi
K
,
Kaneko
S
,
Wada
T
.
Treatment and impact of dyslipidemia in diabetic nephropathy
.
Clin Exp Nephrol.
2014
;
18
(
2
):
201
205
.

38.

Sargsyan
E
,
Artemenko
K
,
Manukyan
L
,
Bergquist
J
,
Bergsten
P
.
Oleate protects beta-cells from the toxic effect of palmitate by activating pro-survival pathways of the ER stress response
.
Biochim Biophys Acta.
2016
;
1861
(
9 Pt A
):
1151
1160
.

39.

Sommerweiss
D
,
Gorski
T
,
Richter
S
,
Garten
A
,
Kiess
W
.
Oleate rescues INS-1E β-cells from palmitate-induced apoptosis by preventing activation of the unfolded protein response
.
Biochem. Biophys. Res.
2013
;
441
(
4
):
770
776
.

40.

Yao
F
,
Li
Z
,
Ehara
T
, et al.
Fatty acid-binding protein 4 mediates apoptosis via endoplasmic reticulum stress in mesangial cells of diabetic nephropathy
.
Mol Cell Endocrinol.
2015
;
411
:
232
242
.

41.

Benito-Vicente
A
,
Jebari-Benslaiman
S
,
Galicia-Garcia
U
,
Larrea-Sebal
A
,
Uribe
KB
,
Martin
C
.
Molecular mechanisms of lipotoxicity-induced pancreatic β-cell dysfunction
.
Int Rev Cell Mol Biol
2021
;
359
:
357
402
.

42.

Pang
L-Q
,
Liang
Q-L
,
Wang
Y-M
,
Ping
L
,
Luo
G-A
.
Simultaneous determination and quantification of seven major phospholipid classes in human blood using normal-phase liquid chromatography coupled with electrospray mass spectrometry and the application in diabetes nephropathy
.
J Chromatogr B Analyt Technol Biomed Life Sci.
2008
;
869
(
1-2
):
118
125
.

43.

Rhee
SY
,
Jung
ES
,
Park
HM
, et al.
Plasma glutamine and glutamic acid are potential biomarkers for predicting diabetic retinopathy
.
Metabolomics.
2018
;
14
(
7
):
89
.

44.

Tavares
G
,
Venturini
G
,
Padilha
K
, et al.
1,5-Anhydroglucitol predicts CKD progression in macroalbuminuric diabetic kidney disease: results from non-targeted metabolomics
.
Metabolomics.
2018
;
14
(
4
):
39
.

45.

Niewczas
MA
,
Mathew
AV
,
Croall
S
, et al.
Circulating modified metabolites and a risk of ESRD in patients with type 1 diabetes and chronic kidney disease
.
Diabetes Care.
2017
;
40
(
3
):
383
390
.

46.

Niewczas
MA
,
Sirich
TL
,
Mathew
AV
, et al.
Uremic solutes and risk of end-stage renal disease in type 2 diabetes: metabolomic study
.
Kidney Int.
2014
;
85
(
5
):
1214
1224
.

47.

Tofte
N
,
Suvitaival
T
,
Trost
K
, et al.
Metabolomic assessment reveals alteration in polyols and branched chain amino acids associated with present and future renal impairment in a discovery cohort of 637 persons with type 1 diabetes
.
Front Endocrinol.
2019
;
10
:
818
.

48.

Kang
HM
,
Ahn
SH
,
Choi
P
, et al.
Defective fatty acid oxidation in renal tubular epithelial cells has a key role in kidney fibrosis development
.
Nat Med.
2015
;
21
(
1
):
37
46
.

49.

Afshinnia
F
,
Nair
V
,
Lin
J
, et al.
Increased lipogenesis and impaired β-oxidation predict type 2 diabetic kidney disease progression in American Indians
.
JCI insight
2019
;
4
(
21):e130317
.

50.

Haukka
JK
,
Sandholm
N
,
Forsblom
C
,
Cobb
JE
,
Groop
P-H
,
Ferrannini
E
.
Metabolomic profile predicts development of microalbuminuria in individuals with type 1 diabetes
.
Sci Rep.
2018
;
8
(
1
):
13853
.

51.

Looker
HC
,
Colombo
M
,
Hess
S
, et al.
Biomarkers of rapid chronic kidney disease progression in type 2 diabetes
.
Kidney Int.
2015
;
88
(
4
):
888
896
.

52.

Heerspink
HJL
,
Stefánsson
BV
,
Correa-Rotter
R
, et al.
Dapagliflozin in patients with chronic kidney disease
.
N Engl J Med.
2020
;
383
(
15
):
1436
1446
.

53.

Perkovic
V
,
Jardine
MJ
,
Neal
B
, et al.
Canagliflozin and renal outcomes in type 2 diabetes and nephropathy
.
N Engl J Med.
2019
;
380
(
24
):
2295
2306
.

54.

Guthrie
R
.
Canagliflozin and cardiovascular and renal events in type 2 diabetes
.
Postgrad Med.
2018
;
130
(
2
):
149
153
.

55.

Sharma
K
,
Karl
B
,
Mathew
AV
, et al.
Metabolomics reveals signature of mitochondrial dysfunction in diabetic kidney disease
. J Am Soc Nephrol.
2013
;
24
(
11
):
1901
1912
.

56.

Wu
M
,
Li
S
,
Yu
X
, et al.
Mitochondrial activity contributes to impaired renal metabolic homeostasis and renal pathology in STZ-induced diabetic mice
.
Am J Physiol Renal Physiol.
2019
;
317
(
3
):
F593
F605
.

57.

Saulnier
P-J
,
Darshi
M
,
Wheelock
KM
, et al.
Urine metabolites are associated with glomerular lesions in type 2 diabetes
.
Metabolomics.
2018
;
14
(
6
):
84
.

58.

Sha
Q
,
Lyu
J
,
Zhao
M
,
Li
H
,
Guo
M
,
Sun
Q
.
Multi-omics analysis of diabetic nephropathy reveals potential new mechanisms and drug targets
.
Front Genet.
2020
;
11
:
616435
.

59.

Hoppel
C
.
The role of carnitine in normal and altered fatty acid metabolism
.
Am J Kidney Dis.
2003
;
41
(
4 suppl 4
):
S4
S12
.

60.

You
Y-H
,
Quach
T
,
Saito
R
,
Pham
J
,
Sharma
K
.
Metabolomics reveals a key role for fumarate in mediating the effects of NADPH oxidase 4 in diabetic kidney disease
.
J Am Soc Nephrol.
2016
;
27
(
2
):
466
481
.

61.

Wang
X
,
He
Q
,
Chen
Q
, et al.
Network pharmacology combined with metabolomics to study the mechanism of Shenyan Kangfu tablets in the treatment of diabetic nephropathy
.
J Ethnopharmacol.
2021
;
270
:
113817
.

62.

Reddy
MA
,
Tak Park
J
,
Natarajan
R
.
Epigenetic modifications in the pathogenesis of diabetic nephropathy
.
Semin Nephrol.
2013
;
33
(
4
):
341
353
.

63.

Kato
M
,
Natarajan
R
.
Diabetic nephropathy—emerging epigenetic mechanisms
.
Nat Rev Nephrol.
2014
;
10
(
9
):
517
530
.

64.

Villeneuve
LM
,
Natarajan
R
.
The role of epigenetics in the pathology of diabetic complications
.
Am J Physiol Renal Physiol.
2010
;
299
(
1
):
F14
F25
.

65.

Wang
J
,
Wu
Z
,
Li
D
, et al.
Nutrition, epigenetics, and metabolic syndrome
.
Antioxid Redox Signal.
2012
;
17
(
2
):
282
301
.

66.

Chen
S
,
Liu
Y-H
,
Dai
D-P
, et al.
Using circulating O-sulfotyrosine in the differential diagnosis of acute kidney injury and chronic kidney disease
.
BMC Nephrol.
2021
;
22
(
1
):
66
.

Author notes

Yushan Mao and Yan Li contributed equally to this work.

This article is published and distributed under the terms of the Oxford University Press, Standard Journals Publication Model (https://dbpia.nl.go.kr/pages/standard-publication-reuse-rights)