Background: Chronic kidney disease (CKD) presents a persistent global health challenge, characterized by complex pathophysiology and diverse progression patterns. Metabolomics has emerged as a valuable tool in unraveling the intricate molecular mechanisms driving CKD progression. Summary: This comprehensive review provides a summary of recent progress in the field of metabolomics in kidney disease with a focus on spatial metabolomics to shed important insights to enhancing our understanding of CKD progression, emphasizing its transformative potential in early disease detection, refined risk assessment, and the development of targeted interventions to improve patient outcomes. Key Message: Through an extensive analysis of metabolic pathways and small-molecule fluctuations, bulk and spatial metabolomics offers unique insights spanning the entire spectrum of CKD, from early stages to advanced disease states. Recent advances in metabolomics technology have enabled spatial identification of biomarkers to provide breakthrough discoveries in predicting CKD trajectory and enabling personalized risk assessment. Furthermore, metabolomics can help decipher the complex molecular intricacies associated with kidney diseases for exciting novel therapeutic approaches. A recent example is the identification of adenine as a key marker of kidney fibrosis for diabetic kidney disease using both untargeted and targeted bulk and spatial metabolomics. The metabolomics studies were critical to identify a new biomarker for kidney failure and to guide new therapeutics for diabetic kidney disease. Similar approaches are being pursued for acute kidney injury and other kidney diseases to enhance precision medicine decision-making.

A significant burden on healthcare systems worldwide is imposed by kidney diseases, which affect millions of individuals [1, 2]. These diseases encompass a wide spectrum of conditions, ranging from chronic kidney disease (CKD) [3], acute kidney injury (AKI) [4], and diabetic kidney disease (DKD) [5] as well as various glomerular and tubular disorders. Early diagnosis of these diseases and managing them effectively can significantly improve patient outcomes and reduce the socioeconomic impact of kidney disease. According to data from the National Kidney Foundation (NKF) in 2018, a total of 785,883 Americans were diagnosed with kidney failure, necessitating either dialysis or a kidney transplant for survival [6]. Among this group, 554,038 patients underwent dialysis treatment, while an additional 229,887 patients received kidney transplants. Unfortunately, individuals with kidney disease face a sobering reality as they are five to ten times more likely to succumb prematurely rather than experience dialysis or transplantation [7, 8]. In 2018, more than 100,000 people lost their lives to kidney disease despite ongoing efforts to address this critical health issue [3, 9, 10]. CKD has become a prominent and pressing issue in the 21st century, emerging as a leading cause of both mortality and suffering. This alarming trend can be attributed, at least in part, to the increase in risk factors such as obesity, hypertension, and diabetes mellitus (DM) [11].

While advancements have led to a decline in mortality rates for individuals with end-stage kidney disease (ESKD), the Global Burden of Disease (GBD) studies have revealed that CKD has now taken center stage as a major contributor to worldwide mortality [12, 13]. Consequently, it is crucial to identify, monitor, and treat CKD effectively. Identifying the disease in the early stages in transition from AKI to CKD, known as acute kidney disease (AKD), is critical as survivors of post-AKI incidents face an increased lifetime risk of mortality [14‒16]. One of the most common types of CKD is DKD, which results from hyperglycemia and leads to gradual changes in the function of the kidneys. The initial stages of DKD typically exhibit no noticeable clinical symptoms. It is often only when albuminuria is detected, often 10 years after the onset of diabetes, that the kidney damage is already advanced, resulting in a swift decline in kidney function toward ESKD [17]. However, if DKD is identified and addressed in its early stages, its progression can be halted or significantly delayed. A significant challenge with current diagnostic methods is their inability to detect ongoing disease during the silent phase of diabetic nephropathy (DN) as well as in overt disease which is not characterized by albuminuria. Developing innovative biomarkers capable of predicting progression and reflecting biologically significant pathways in DKD has the potential to improve the management of individuals with diabetes [5, 18]. A recent example is the identification of adenine as a key marker of kidney fibrosis for DKD [19].

Adenine, a pivotal purine nucleobase, plays a crucial role in cellular functions, including DNA/RNA synthesis, ATP-mediated energy processes, extracellular signaling, and enzyme cofactors. Under normal physiological concentrations, adenine undergoes absorption and metabolism via adenine phosphoribosyl transferase (APRT), resulting in the formation of AMP and various derivatives. The resultant metabolite xanthine is then converted to uric acid, which is excreted by the kidneys. However, when adenine accumulates with APRT deficiency, adenine follows an alternative pathway catalyzed by xanthine oxidase, leading to the formation of 2,8-dihydroxyadenine. Due to its limited solubility under normal urine pH conditions, 2,8-dihydroxyadenine may precipitate as crystals, presenting a potential risk of renal injury [20]. The kidneys play a pivotal role in maintaining purine homeostasis, and disruptions in this process may contribute to renal dysfunction [21]. Emerging evidence indicates that exposure to adenine also stimulates oxidative stress in the kidneys and may be responsible for cellular toxicity [22]. Exploring specific kidney diseases associated with adenine metabolism or its derivatives is of particular interest. Studies focused on adenine and its associated metabolic pathways may provide insights into conditions where adenine plays a role in the development of nephrolithiasis, glomerular disorders, or tubulointerstitial diseases [23]. Recent studies using untargeted metabolomics, targeted metabolomics, and spatial metabolomics have indicated that endogenous adenine plays a critical role in common kidney diseases, such as DKD. Apart from DN, adenine is also involved in familial kidney diseases such as APRT deficiency, leading to nephrolithiasis and kidney failure, tubulointerstitial diseases, and glomerular disorders. Plasma levels and RBC levels of adenine have been found to be increased with ESKD. Exogenous adenine is also one of the most widely accepted models of experimental CKD. According to our experimental results, the best reservoir for adenine is urine apart from the tissues, with low levels in blood [24]. Adenine itself does not exert a direct influence on the acid-base status of a patient. Nonetheless, the metabolism of adenine and associated compounds can impact the acid-base balance through its involvement in diverse metabolic processes such as purine metabolism, uric acid production, diabetic ketoacidosis, acid-base regulation, etc. [20]. Such investigations contribute to a more nuanced understanding of the intricate relationship between metabolites and kidney health.

This review offers a thorough analysis of a dynamic and swiftly evolving discipline that bridges metabolomics and nephrology. It encompasses discussions on the analytical methodologies employed in metabolomics, the spectrum of kidney diseases under scrutiny, the clinical utility of metabolomics in kidney disease contexts, the mechanistic insights gleaned through metabolomic approaches, as well as the challenges faced and potential future avenues in this exciting and fast-evolving field.

Different Types and Prevalence of Kidney Diseases

Kidney diseases encompass a spectrum of conditions impacting kidney structure and function [25]. CKD, a global health concern, exhibits varying stages based on estimated glomerular filtration rate (eGFR), ultimately progressing to ESKD [3, 12, 13]. Lack of awareness, as early CKD stages often remain asymptomatic, contributes to high morbidity and mortality. CKD also amplifies the mortality associated with other conditions like cardiovascular diseases, type 2 diabetes (T2DM), hypertension, and infections such as HIV, malaria, and COVID-19 [3, 11, 25, 26]. AKI, common in hospitalized patients, involves sudden kidney function decline due to infections, medications, or dehydration. DN stems from uncontrolled diabetes, leading to proteinuria and kidney function decline [4, 27, 28]. Polycystic kidney disease is a rare genetic condition forming fluid-filled cysts in the kidneys [29]. Nephrotic syndrome indicates kidney damage and features excessive protein in urine, swelling, and high cholesterol [30]. Hypertensive nephropathy results from uncontrolled high blood pressure, gradually impairing kidney function [31]. Awareness, early detection through health checkups, and lifestyle modifications play crucial roles in managing kidney diseases and improving overall well-being [11, 32‒34]. In many cases of kidney disease, there is often advanced disease which is difficult to arrest, and therefore, it is critical to identify the disease in its early, silent stages.

Current Diagnostic Tools, Markers, and Challenges in Treatments

The diagnosis of kidney diseases relies on a range of diagnostic tools and markers, including serum creatinine and blood urea nitrogen measurements, as well as urinalysis, to assess kidney health and detect abnormalities [35, 36]. The glomerular filtration rate serves as a precise measurement of kidney function, aiding in accurate diagnosis and staging [18, 37‒39]. Various imaging techniques such as ultrasound, CT scan, MRI, and matrix-assisted laser desorption/ionization MS imaging (MALDI-MSI) are instrumental in visualizing kidney structure and identifying structural irregularities [18, 40, 41]. Treatment challenges for kidney diseases are diverse and depend on the underlying causes. Common treatment approaches involve managing blood pressure and blood sugar levels, dietary adjustments, and medications. In severe cases, dialysis or kidney transplantation may be necessary. Emerging therapies, including gene therapies and regenerative medicine, show promise but will require identifying patients at early stages of the disease. Early detection and preventive measures remain pivotal in effectively managing kidney diseases, emphasizing the importance of new biomarkers indicating relevant risk factors for precision intervention to improve outcomes [42].

Role of Metabolomics in Understanding the Biochemical Pathways Associated with Kidney Diseases

Metabolomics plays a pivotal role in unraveling the intricate biochemical pathways associated with kidney diseases. Metabolomics can provide a comprehensive analysis of small molecules or metabolites within biological samples, such as urine and blood, to gain insights into the metabolic changes that occur during the development and progression of kidney diseases [25‒30, 43] and serve as biomarkers that are associated with different etiologies of kidney diseases. By analyzing the pattern of metabolites, researchers can gain insights into the altered biochemical pathways in the kidneys with insights often not possible from reliance on transcriptomics and proteomics alone. Metabolomics helps in understanding how metabolic processes are disrupted in kidney diseases, shedding light on the underlying metabolic pathways. Metabolomics allows for a personalized approach to treatment by identifying patient-specific metabolic profiles, which can be used to tailor treatment strategies and monitor responses to therapy, [28]. It can reveal how potential medications affect metabolic pathways and help in the design of more targeted and effective therapies. This information can guide treatment decisions and interventions. Metabolomics aids in understanding the impact of dietary choices on kidney health by informing dietary recommendations and interventions for individuals with kidney diseases. Metabolomics can also assist in assessing an individual’s risk of developing kidney diseases based on their metabolic profile and genetic predisposition using approaches such as metabolite-based genome-wide association study [44]. Early identification of at-risk individuals can lead to preventive measures [19]. Overall, metabolomics offers a comprehensive and powerful approach to unravel the biochemical and genetic basis of kidney diseases. As our understanding of metabolomics continues to grow, so does our ability to combat kidney diseases effectively.

Analytical Techniques Used in Metabolomics

Analytical techniques in metabolomics include nuclear magnetic resonance (NMR), GC, high-performance liquid chromatography, inductively coupled plasma atomic emission spectroscopy, inductively coupled plasma mass spectrometry, LC-MS, and GC-MS. Both NMR (1H and 13C) and mass spectrometry (MS) (LC-MS and GC-MS) are essential techniques in metabolomics. However, NMR faces a significant drawback in sensitivity compared to MS, making it challenging to detect or gather information about low-abundance metabolites in biological fluids [45]. MS serves as a pivotal tool in metabolomics research, providing heightened sensitivity through both GC-MS and LC-MS. However, GC-MS has drawbacks, including the necessity for readily volatile samples and occasional chemical derivatization, which can take at least 1–2 days [46]. Additionally, it is limited to analyzing relatively small molecular masses (m/z, <800) and is unsuitable for medium polar compounds. In contrast, LC-MS offers distinct advantages, requiring minimal sample preparation and enabling analysis of diverse metabolite classes that cannot be separated or fragmented by GC-MS due to their higher molecular mass or polarity. Successful metabolomics often relies on LC-MS combined with soft ionization techniques like MALDI [47, 48] and electrospray ionization (ESI) [49, 50]. These interfaces coupled to mass analyzers play a vital role in MS, sorting ions by their mass-to-charge ratios (m/z). Types of mass spec separation and detection approaches include time of flight, triple quadrupole, ion trap, and orbitrap. Time of flight utilizes an electric field to accelerate ions from the source to the detector based on travel time, distinguishing ions by size. Orbitraps are widely used in proteomics and metabolomics across diverse fields. It uses electrostatic trapping and frequency detection to determine ion m/z. Ions from a sample are trapped in an electrostatic field, undergo harmonic oscillations, and their frequency directly correlates with m/z. This enables accurate mass determination. Orbitrap instruments offer high mass resolution, crucial for separating and identifying metabolites, reducing false positives in metabolomics. They are highly sensitive, detecting metabolites at low concentrations, crucial for studying diverse metabolites in biological samples. With a wide dynamic range, orbitraps simultaneously detect abundant and trace-level metabolites. Their rapid data acquisition makes them suitable for efficient high-throughput metabolomics studies [51]. LC-MS with multiple-reaction monitoring (MRM) is a key MS/MS technique for precise measurement of low-concentration metabolites in metabolomics [43, 45, 50, 52]. The typical workflow for the targeted and untargeted metabolomics approaches is shown in Figure 1 [53].

Fig. 1.

This figure illustrates the sequential steps involved in both untargeted and targeted metabolomics approaches for investigating disease biomarkers. The figure was drawn using the BioRender software application [53].

Fig. 1.

This figure illustrates the sequential steps involved in both untargeted and targeted metabolomics approaches for investigating disease biomarkers. The figure was drawn using the BioRender software application [53].

Close modal

The metabolomics approach is an emerging and highly prominent technological frontier. This methodology entails meticulous scrutiny of metabolites, encompassing both endogenous and exogenous varieties (with a molecular mass <1,500 Da), derived from specific biological fluids and has emerged as a powerful approach to unraveling the complex metabolic alterations associated with kidney diseases [54]. Metabolomics can be categorized into two primary approaches: targeted and untargeted metabolomics. Targeted metabolomics focuses on the identification and quantification of a limited set of metabolites, using standards and calibration curves, associated with specific pathways or biochemical processes [52]. In contrast, untargeted metabolomics aims to capture as many diverse metabolite species as possible, followed by candidate annotation using various databases such as ChemSpider, METLIN, Human Metabolome Database (HMDB), MassBank, mzCloud, GNPS, Lipid Blast, and others. This approach proves invaluable for the relative quantification of metabolites and enables the prediction of metabolic changes within the studied groups [45]. Several analytical platforms are accessible for the study of metabolomics, with NMR spectroscopy (1H and 13C NMR) and MS (LC-MS and GC-MS) emerging as the primary techniques for such investigations. Each of these techniques offers distinct advantages, particularly in terms of sensitivity, selectivity, and throughput, when addressing the complexities of various sample types such as urine, blood plasma/serum, tissue, cell extracts, or media [47].

In recent years, spatial metabolomics has received much recognition as a transformative approach for understanding complex diseases and interpreting heterogenous pathology in tissue section. MALDI-MSI technology has gained prominence as a cutting-edge analytical tool in the field of spatial metabolomics, enabling researchers to dissect the intricate molecular mechanisms involved in development of specific pathologic features in diseased kidneys [55]. This field is dedicated to the detection and interpretation of metabolites within their precise spatial context. Notably, the adoption of MALDI-MSI has played a pivotal and widely embraced role in transforming this approach [43]. The schematic approach of the MALDI-MSI is shown in Figure 2. Other platforms of spatial metabolomics include desorption ESI-MSI (DESI-MSI), nano-droplet extraction surface analysis (Nano-DESI); DESI-MSI enables direct surface analysis without extensive sample preparation. It utilizes charged solvent droplets to desorb and ionize molecules on the surface, with subsequent MS providing spatially resolved information on compound distribution. Nano-DESI, operating at micro and nanoscales, utilizes tiny droplets for high-resolution analysis of biological samples. Valuable in metabolomics, pharmacology, and environmental science, Nano-DESI allows detailed mapping of molecules in complex biological systems at the microscale [56].

Fig. 2.

A comprehensive workflow for MALDI-MSI spatial metabolomics is outlined as follows: a tissue embedding: tissue samples undergo cryoembedding to ensure the preservation of both structural integrity and biochemical composition. b Cryosectioning and mounting: Thin sections (7–10 µm) are precisely cut from cryoembedded tissue and placed onto microscopy slides coated with indium tin oxide (ITO). c Autofluorescent imaging: Autofluorescent imaging of tissue sections is performed for quality control and data co-registration. d MALDI matrix application: A MALDI ionization matrix (e.g., DHB, DAN, 9-AA, NEDC) is applied to the tissue sections, facilitating ionization for subsequent MSI data. e High-resolution MS imaging (MSI): Metabolites are measured with high spatial resolution (pixel size <20 μm) through MSI techniques, yielding detailed spatial information. f Histological analysis: Histological staining (e.g., H&E, PAS, and IHC) is performed on either post-MALDI tissue section or serial adjacent sections to link spatial metabolomic data to functional regions of the organ. g Data integration – MALDI-MSI: MALDI-MSI spatial metabolomic raw data are uploaded to annotation platforms like METASPACE, enabling peak-to-molecular ID annotation. h Data integration – optical imaging: Optical imaging data are combined with MALDI-MSI for co-localization of metabolites, enhancing the interpretation of spatial information. i Additional histological analysis: Further histological analyses may be performed on the same or adjacent tissue sections for comprehensive correlation with metabolomic data. j Final data integration: Comprehensive data integration is conducted, merging MALDI-MSI spatial metabolomic findings with optical imaging and histological information. This combined workflow enables a holistic exploration of spatial metabolomics, linking molecular information to the anatomical and functional context of the tissue under investigation.

Fig. 2.

A comprehensive workflow for MALDI-MSI spatial metabolomics is outlined as follows: a tissue embedding: tissue samples undergo cryoembedding to ensure the preservation of both structural integrity and biochemical composition. b Cryosectioning and mounting: Thin sections (7–10 µm) are precisely cut from cryoembedded tissue and placed onto microscopy slides coated with indium tin oxide (ITO). c Autofluorescent imaging: Autofluorescent imaging of tissue sections is performed for quality control and data co-registration. d MALDI matrix application: A MALDI ionization matrix (e.g., DHB, DAN, 9-AA, NEDC) is applied to the tissue sections, facilitating ionization for subsequent MSI data. e High-resolution MS imaging (MSI): Metabolites are measured with high spatial resolution (pixel size <20 μm) through MSI techniques, yielding detailed spatial information. f Histological analysis: Histological staining (e.g., H&E, PAS, and IHC) is performed on either post-MALDI tissue section or serial adjacent sections to link spatial metabolomic data to functional regions of the organ. g Data integration – MALDI-MSI: MALDI-MSI spatial metabolomic raw data are uploaded to annotation platforms like METASPACE, enabling peak-to-molecular ID annotation. h Data integration – optical imaging: Optical imaging data are combined with MALDI-MSI for co-localization of metabolites, enhancing the interpretation of spatial information. i Additional histological analysis: Further histological analyses may be performed on the same or adjacent tissue sections for comprehensive correlation with metabolomic data. j Final data integration: Comprehensive data integration is conducted, merging MALDI-MSI spatial metabolomic findings with optical imaging and histological information. This combined workflow enables a holistic exploration of spatial metabolomics, linking molecular information to the anatomical and functional context of the tissue under investigation.

Close modal

Recent research endeavors have aimed to identify differentially expressed metabolites in AKD through an array of metabolomic methodologies and bio samples. A decade ago, a study utilized the LC-MS technique with Sprague-Dawley rats, subjecting them to bilateral renal artery clamping for 45 min, with or without L-carnitine pretreatment. Postischemic animals showed peaking serum concentrations of creatinine and blood urea nitrogen 24 h after reperfusion, which was reduced by L-carnitine. The authors attributed lipid metabolism changes as critical factors in ischemia/reperfusion kidney damage [57]. Another study by Wei et al. [58] employed GC-MS and LC-MS for a murine ischemic AKI metabolomics study, identifying 404 substances from tissue samples and 293 metabolites from plasma. Renal ischemia notably decreased plasma levels of several substances, including betaine, tyrosine, glutamine, and others, revealing significant alterations in glucose, lipid, and purine metabolism, as well as osmotic regulation and prostaglandin modulation. These findings highlighted the potential connection between hypoperfusion/ischemia and the key elements of ischemic AKI, including tubulopathy, inflammation, and (micro) vasculopathy [58]. Elmariah et al. [59] conducted metabolomic profiling using LC-MS on a cohort of 44 patients, including 22 with a history of CKD. Their study was cross-referenced with the Framingham Heart Study, encompassing 2,164 individuals. This investigation revealed alterations in plasma concentrations of certain metabolites, ultimately identifying S-5-adenosylhomocysteine as strongly correlated with serum creatinine and serving as a reliable marker for AKI. A metabolomics investigation utilizing LC-MS/MS was carried out on 42 kidney transplant patients, of whom 30 developed AKI [60]. Findings from this study indicated an adverse impact on tryptophan and arginine metabolism, with the concentration of these two amino acids being lower in transplant patients compared to that in healthy volunteers. Furthermore, in the comparison between AKI patients and patients without AKI, the levels of tryptophan and arginine were lower in AKI patients than in non-AKI patients. In 2021, a study utilized LC-MS/MS to investigate targeted metabolic profiling in cisplatin-induced AKI, focusing on tryptophan metabolism alterations [61]. Targeting 29 metabolites, the study found that indoxyl sulfate accumulated in a dose-dependent manner. Additional experiments with chlormethiazole, a CYP2E1 inhibitor, demonstrated the functional significance of indoxyl sulfate, showing that reducing hepatic indoxyl sulfate synthesis attenuated cisplatin-induced AKI.

Another recent study by Chen et al. [62] focused on 30 AKI patients and 20 healthy controls, proposing that 2-S-glutathionyl glutathione acetate, 5-l-glutamyl-taurine, and l-phosphoarginine were involved in altered cytochrome P450, arginine, and proline metabolism, serving as potential diagnostic markers for AKI. A separate urine metabolomic study conducted by Tian et al. [63] of in post-cardiac surgery subjects (n = 159) found that 55 patients experienced AKI, while the kidney functions of the remaining subjects were unaffected. The authors suggested that tyrosyl-gamma-glutamate, deoxycholic acid glycine conjugate, 5-acetylamino-6-amino-3-methyluracil, arginyl-arginine, and L-methionine could act as predictive factors for AKI in cardiac surgery patients [63]. To gain a deeper understanding of AKI, various experimental models have been tested, including rats, mice, murine, and pigs. In addition to these models, diverse metabolic analyses conducted on AKI patients revealed a metabolomic differentiation between patients with and without AKI. Notably, symmetric dimethylarginine (SDMA) and tryptophan, or the 5-HIAA/5-HT ratio, were identified as potential AKI risk predictors (AUC >0.9). These findings, along with various other studies [64‒69] exploring the role of metabolomics in AKI diagnosis and risk prediction, are summarized in Table 1.

Table 1.

Summary of metabolites affected in AKI identified/quantified by targeted/untargeted metabolomics approach

ReferenceAnalysis platformSample typeStudy designFindings
Liu et al. [57LC-MS Kidney tissue and serum Bilateral renal ischemia (45 min) in Sprague-Dawley rats, L-carnitine pretreatment. Increase of serum lysophospholipids and free fatty acids, decreased activity of serum phospholipase A2/ischemia-associated dysregulation of lipid metabolism. 
Elmariah et al. [59LC-MS Plasma Prospective, observational; TAVR patients (n = 44) and participants of the Framingham Heart Study (n = 2,164). 5-Adenosylhomocysteine AKI-predictive, even after adjustment for baseline creatinine. 
Zhang et al. [60LC-MS Plasma Kidney transplant recipients, living donors; n = 42 (AKI in n = 30), 24 healthy subjects as controls Tryptophan lower in transplant patients than in controls and lower in AKI than in non-AKI patients; AKI prediction through tryptophan and symmetric SDMA in combination. 
Tan et al. [61LC-MS Kidney cortex and medulla Cisplatin-induced AKI in Sprague-Dawley rats, targeted analysis of the tryptophan metabolism Identification of indoxyl sulfate as the key regulator of tissue damage in cisplatin-induced AKI. 
Chen et al. [62LC-MS Serum and urine 30 AKI subjects and 20 healthy controls, gender- and age-matched Glutathione acetate, 5-L-glutamyl-taurine, and l-phosphoarginine higher in AKI, positive correlation with serum creatinine. 
Tian et al. [63LC-MS Urine Patients that receive (on-pump) coronary artery bypass surgery; post-surgery AKI in n = 55; stable kidney function in n = 104. Identification of 5 AKI-predictive urine metabolites. 
Franiek et al. [65GC-MS and DI-MS Urine Urine samples from children with either pre-AKI (n = 15), established AKI (n = 22), and controls (n = 30) 20 metabolites discriminated between pre-AKI and established AKI; 13 metabolites predicted AKI up to 3 days in advance 
ReferenceAnalysis platformSample typeStudy designFindings
Liu et al. [57LC-MS Kidney tissue and serum Bilateral renal ischemia (45 min) in Sprague-Dawley rats, L-carnitine pretreatment. Increase of serum lysophospholipids and free fatty acids, decreased activity of serum phospholipase A2/ischemia-associated dysregulation of lipid metabolism. 
Elmariah et al. [59LC-MS Plasma Prospective, observational; TAVR patients (n = 44) and participants of the Framingham Heart Study (n = 2,164). 5-Adenosylhomocysteine AKI-predictive, even after adjustment for baseline creatinine. 
Zhang et al. [60LC-MS Plasma Kidney transplant recipients, living donors; n = 42 (AKI in n = 30), 24 healthy subjects as controls Tryptophan lower in transplant patients than in controls and lower in AKI than in non-AKI patients; AKI prediction through tryptophan and symmetric SDMA in combination. 
Tan et al. [61LC-MS Kidney cortex and medulla Cisplatin-induced AKI in Sprague-Dawley rats, targeted analysis of the tryptophan metabolism Identification of indoxyl sulfate as the key regulator of tissue damage in cisplatin-induced AKI. 
Chen et al. [62LC-MS Serum and urine 30 AKI subjects and 20 healthy controls, gender- and age-matched Glutathione acetate, 5-L-glutamyl-taurine, and l-phosphoarginine higher in AKI, positive correlation with serum creatinine. 
Tian et al. [63LC-MS Urine Patients that receive (on-pump) coronary artery bypass surgery; post-surgery AKI in n = 55; stable kidney function in n = 104. Identification of 5 AKI-predictive urine metabolites. 
Franiek et al. [65GC-MS and DI-MS Urine Urine samples from children with either pre-AKI (n = 15), established AKI (n = 22), and controls (n = 30) 20 metabolites discriminated between pre-AKI and established AKI; 13 metabolites predicted AKI up to 3 days in advance 

TAVR, transcatheter aortic valve replacement.

DKD is a specific type of kidney disease that occurs in individuals with diabetes and is a leading cause of CKD. Managing diabetes effectively is essential in preventing or slowing the progression of both DKD and CKD. Metabolomics has proven to be an invaluable tool in comprehending the pathophysiology of CKD, which significantly contributes to morbidity and mortality [5, 19, 27, 47]. From a metabolomics perspective, DM is considered a quintessential metabolic disease due to the numerous metabolic variations linked with it. Recent studies have focused on identifying differentially expressed metabolites in DKD through various metabolomic approaches and biological samples, revealing several metabolites as potential biomarkers for predicting the onset of diabetes. Notably, separate studies by Wang et al. [70], Newgard et al. [71], and Guasch‐Ferre et al. [72] have indicated the involvement of isoleucine, leucine, and valine in the development of T2DM. Additionally, a comprehensive study by Merino et al. [73] involving 1,150 individuals with normal fasting glucose in a 20‐year follow‐up highlighted the correlation between increased plasma taurine concentration and decreased glycine with the onset of T2DM. A MS metabolomics investigation conducted a decade ago by Hirayama et al. [74] on 78 subjects at various stages of DKD revealed increased levels of aspartic acid, citrulline, SDMA, and kynurenine in the DKD group. Conversely, Zhang et al. [75] reported reduced plasma concentrations of several amino acids, including isoleucine, leucine, valine, and phenylalanine, in individuals with T2DM. The observed contradictions in amino acid metabolism across studies might be attributed to variations in drug treatments and interventions among individuals with T2DM.

Our group quantified 94 urine metabolites in cohorts of patients with DM and CKD (DM with DKD; n = 61), in patients with type 1 DM without CKD (n = 32), type 2 DM without CKD (n = 41), and in healthy controls (n = 23) by using a targeted GC-MS technique [27]. The study revealed that 13 metabolites which included 3-hydroxy isovalerate, aconitic acid, citric acid, 2-ethyl 3-OH propionate, glycolic acid, homovanillic acid, 3-hydroxy isobutyrate, 2-methyl acetoacetate, 3-methyl adipic acid, 3-methyl crotonyl glycine, 3-hydroxy propionate, tiglylglycine, and uracil were statistically related to eGFR or albuminuria (albumin-to-creatinine ratio), and these metabolites levels were consistently reduced in the urine of DM with CKD. These metabolites play key roles in processes such as the Krebs cycle and lipid synthesis [76, 77]. Notably, 12 of these metabolites remained significant in DM with CKD compared to DM without CKD. Additional studies by van der Kloet et al. [78], a FinnDiane study, corroborated these observations, indicating a decrease in hippuric acid levels in the cohort with progressive disease. Additionally, there was an elevation in S-(3-oxododecanoyl) cysteamine and acylcarnitines in this group.

The urine metabolomic analysis also suggested a decline in mitochondrial function, confirmed by urine exosome analysis and immunohistochemistry [27], indicating a reduction in mitochondrial content. This decrease in content could be associated with lower levels of the coactivator PGC1a, a crucial component in mitochondrial biogenesis. Corresponding studies corroborated these observations, highlighting alterations in specific metabolites during the progression of DKD [79, 80]. Overall, this research emphasizes the potential of urine metabolomics as a noninvasive technique for detecting kidney biomarkers and quantitating kidney health. These findings not only shed light on the diminished mitochondrial activity in DKD but also offer the possibility of discovering new therapeutic targets and innovative biomarkers for evaluating kidney function in diabetes. A recent investigation by Parmar et al. [47] explored the ADMA/SDMA ratio (ASR) in urine across various stages of diabetes, including normal glucose tolerance, newly diagnosed diabetes, macroalbuminuria (MAC), and diabetic microalbuminuria. The results indicated a decrease in the ASR in the microalbuminuria and MAC groups compared to the newly diagnosed diabetes and normal glucose tolerance subjects, suggesting its potential diagnostic value for the early detection of DN among Asian Indians. In conjunction with these studies, several other research works [81‒91] have identified different biomarkers using urine, plasma, and tissue, aiding in the early detection of DKD through diverse metabolic pathways using the metabolomics approach tabulated in Table 2.

Table 2.

Summary of metabolites affected in DKD identified/quantified by the targeted/untargeted metabolomics approach

ReferenceAnalysis platformSample typeStudy designFindings
Wang et al. [70LC-MS Blood 2,422 eligible, nondiabetic attendees to a routine examination between 1991 and 1995, 201 individuals developed new-onset diabetes during a 12-year follow-up period. Branched chain and aromatic amino acids emerged as predictors of the future development of diabetes. 
Zhang et al. [81LC-MS/MS Plasma 1-year prospective study on healthy subjects (n = 132); T2DM without DKD (n = 132); T2DM with DKD (n = 132). Chinese patients with T2D, elevated tyrosine was associated with increased risk of DN. 
Tavares et al. [82GC-MS Plasma Patients with DKD (n = 56); mean 2.5 years of follow-up. 1,5-Anhydroglucitol is associated with incident CKD. 
Sharma et al. [27GC-MS Urine Healthy subjects (n = 23); diabetes with DKD (n = 61); type 1 diabetes without DKD (n = 32); T2DM without DKD (n = 41). Renal organic ion transport and mitochondrial function are dysregulated in DKD. 
Parmar et al. [47MALDI-TOF and LC/MS Urine Individuals with (a) NGT (n = 95), (b) IGT (n = 80), (c) NDD (n = 120), (d) T2DM with MIC (n = 140), and (e) T2DM with MAC (n = 120). ASR as a potential early diagnostic biomarker for DN among the Asian Indians. 
Sharma et al. [19MALDI-MSI and Zip-Chip Urine and kidney tissue Patients with T2DM and impaired eGFR from the Pima cohort (n = 54), CRIC cohort (n = 904), SMART2D cohort (n = 309) Endogenous adenine may be a causative factor in DKD. 
Saulnier et al. [83GCMS/MS Urine T2DM with early DKD (n = 62) Urine aconitic and glycolic acids correlated positively with glomerular filtration surface density, and these may associate with early glomerular lesions in DKD. 
Niewczas et al. [84LC-MS Plasma T2DM that developed ESRD (n = 40); T2DM without ESRD (n = 40). Abnormal plasma concentrations of putative uremic solutes and essential amino acids either contribute to progression to ESKD or are a manifestation of an early stage(s) of the disease process that leads to ESKD in T2D. 
Ng et al. et al. [85LC‐MS and GC‐MS Urine T2DM with normal eGFR (n = 46); T2DM with low eGFR (n = 44). Octanol, oxalic acid, phosphoric acid, benzamide, creatinine, 3,5-dimethoxymandelic amide, and N-acetyl glutamine were selected as the best subset for prediction and allowed excellent classification of a low eGFR (AUC = 0.996). 
ReferenceAnalysis platformSample typeStudy designFindings
Wang et al. [70LC-MS Blood 2,422 eligible, nondiabetic attendees to a routine examination between 1991 and 1995, 201 individuals developed new-onset diabetes during a 12-year follow-up period. Branched chain and aromatic amino acids emerged as predictors of the future development of diabetes. 
Zhang et al. [81LC-MS/MS Plasma 1-year prospective study on healthy subjects (n = 132); T2DM without DKD (n = 132); T2DM with DKD (n = 132). Chinese patients with T2D, elevated tyrosine was associated with increased risk of DN. 
Tavares et al. [82GC-MS Plasma Patients with DKD (n = 56); mean 2.5 years of follow-up. 1,5-Anhydroglucitol is associated with incident CKD. 
Sharma et al. [27GC-MS Urine Healthy subjects (n = 23); diabetes with DKD (n = 61); type 1 diabetes without DKD (n = 32); T2DM without DKD (n = 41). Renal organic ion transport and mitochondrial function are dysregulated in DKD. 
Parmar et al. [47MALDI-TOF and LC/MS Urine Individuals with (a) NGT (n = 95), (b) IGT (n = 80), (c) NDD (n = 120), (d) T2DM with MIC (n = 140), and (e) T2DM with MAC (n = 120). ASR as a potential early diagnostic biomarker for DN among the Asian Indians. 
Sharma et al. [19MALDI-MSI and Zip-Chip Urine and kidney tissue Patients with T2DM and impaired eGFR from the Pima cohort (n = 54), CRIC cohort (n = 904), SMART2D cohort (n = 309) Endogenous adenine may be a causative factor in DKD. 
Saulnier et al. [83GCMS/MS Urine T2DM with early DKD (n = 62) Urine aconitic and glycolic acids correlated positively with glomerular filtration surface density, and these may associate with early glomerular lesions in DKD. 
Niewczas et al. [84LC-MS Plasma T2DM that developed ESRD (n = 40); T2DM without ESRD (n = 40). Abnormal plasma concentrations of putative uremic solutes and essential amino acids either contribute to progression to ESKD or are a manifestation of an early stage(s) of the disease process that leads to ESKD in T2D. 
Ng et al. et al. [85LC‐MS and GC‐MS Urine T2DM with normal eGFR (n = 46); T2DM with low eGFR (n = 44). Octanol, oxalic acid, phosphoric acid, benzamide, creatinine, 3,5-dimethoxymandelic amide, and N-acetyl glutamine were selected as the best subset for prediction and allowed excellent classification of a low eGFR (AUC = 0.996). 

ASR, ADMA/SDMA ratio; NGT, normal glucose tolerance; NDD, newly diagnosed diabetes; MIC, microalbuminuria; IGT, impaired glucose tolerance; TOF, time of flight.

Apart from DKD, a comprehensive analysis of CKD progression remains incomplete. Several studies have utilized metabolomics to uncover new insights for ESKD and advanced CKD. A recent study by Dahabiyeh et al. [92] aimed to gain fresh insights into perturbed biochemical reactions and identify distinct biomarkers between ESKD and CKD. The study, employing an isotope-labeled LC-MS approach on ESKD patients (n = 13) and CKD patients (n = 13), revealed significant alterations in 193 metabolites between ESRD and CKD. These metabolites were primarily involved in aminoacyl-tRNA biosynthesis, branched-chain amino acid biosynthesis, taurine metabolism, kynurenine, and tryptophan metabolism. Notably, the study highlighted the upregulation of three kynurenine derivatives, 2-aminobenzoic acid, xanthurenic acid, and hydroxy picolinic acid, in ESRD compared to CKD, owing to the significant decrease in the glomerular filtration rate with CKD progression to ESRD. Mitrofanova et al. [93] reviewed the lipid pathogenesis in CKD conditions, discussing the clinical and experimental evidence of pathogenic lipid droplet accumulation in kidney parenchyma and the molecular mechanisms by which cholesterol, fatty acids, triglycerides, and lipid droplet accumulation contribute to CKD progression. The authors also focused on current and emerging therapeutic strategies for treating and preventing lipid-induced nephrotoxicity. Numerous studies have been conducted employing metabolomics in models of CKD, identifying promising metabolites that could serve as biomarkers [94‒96]. Mor et al. [97] recently conducted a review on the kynurenine pathway in CKD. Primarily, the aromatic amino acid tryptophan undergoes extensive metabolism through various kynurenine-related pathways, resulting in the generation of numerous biologically active compounds such as 3-hydroxyanthranilic acid, 3-hydroxykynurenine, anthranilic acid, kynurenine, KYNA, kynurenic acid, quinolinic acid, tryptophan, and xanthurenic acid. These metabolites may contribute to the development of systemic disorders that accompany the progression of CKD [97]. A recent study led by Hu et al. [98] highlighted trimethylamine N-oxide (TMAO) as a promising marker for potentially decelerating the progression of CKD. The study suggested that controlling TMAO production through inhibition using 3,3-dimethyl-1-butanol or disruption of gut microbiota function with an antibiotic cocktail reduced renal injury in a murine CKD model. This study underscores the potential of metabolomics, suggesting TMAO production as a novel strategy to mitigate the progression of renal injury in CKD.

Patients with advanced kidney disease like CKD often require dialysis or kidney transplantation. Hemodialysis (HD) plays a pivotal role in renal replacement therapy for ESKD patients, and understanding the HD process, efficacy, and patterns through plasma metabolomics has significant potential. In a study by Ragi et al. [49], the behavior of uremic toxins, including indoxyl sulfate, p-cresol sulfate, phenyl sulfate, catechol sulfate, and guaiacol sulfate, along with creatinine and urea, was evaluated in the pre-dialysis (n = 90), post-dialysis (n = 90), and healthy control (n = 74) groups. The study revealed the dominance of p-cresol sulfate and indoxyl sulfate in the pre-dialysis group. Moreover, the study demonstrated that the concentrations of phenyl sulfate, catechol sulfate, and guaiacol sulfate were approximately 50% of that of indoxyl sulfate. The dialytic clearance of indoxyl sulfate and p-cresol sulfate was found to be less than 35%, while that of the other three sulfates was 50–58%. The concentration levels of these targeted uremic toxins could be used to assess kidney function and the efficacy of HD [49]. A 2023 study led by Pallerla et al. [50] evaluated the levels of amino acids and other related metabolites in ESKD patients using LC-MS/MS and GC-MS techniques. The study developed analytical methods for 29 targeted metabolites, revealing dialytic losses ranging from 52% to 45% for arginine, lysine, and histidine and 38–26% for glycine, cysteine, proline, alanine, threonine, glutamine, valine, and methionine. In contrast, the dialytic loss was low (≤12%) for aspartic acid, glutamic acid, asparagine, leucine, tyrosine, tryptophan, and isoleucine (Table 3) [50]. A recent untargeted metabolomics investigation focused on plasma samples obtained from individuals with a single kidney, non-dialysis CKD, ESKD, and a control group of healthy individuals [99]. The study revealed that more than 400 features in the plasma of non-dialysis CKD and ESKD patients were significantly different from those in the control group. In contrast, individuals with a single kidney exhibited alterations in less than 35 features. The metabolites N,N,N-trimethyl-L-alanyl-L-proline betaine (TMAP, AUROC = 0.815) and pyrocatechol sulfate (AUROC = 0.888) demonstrated superior accuracy compared to creatinine (AUROC = 0.745) in identifying individuals with a single kidney. Furthermore, when comparing pre- and post-dialysis samples, TMAP emerged as the most robust biomarker for dialytic clearance (AUROC = 0.993). The key finding of this study is the identification of TMAP as a potential novel plasma biomarker, indicating reduced kidney function in early CKD, ESKD, and hemodialytic clearance [99]. Moreover, the study highlighted pronounced metabolic differences between transplantation and dialysis therapy, emphasizing the differences of nocturnal intermittent peritoneal dialysis in dialytic metabolite clearance compared to conventional HD as nocturnal intermittent peritoneal dialysis relies predominantly on the passive diffusion of solutes, which may lead to reduced metabolite clearance. Consequently, employing multiple metabolites could offer a more comprehensive evaluation of dialysis, assisting clinicians in selecting an optimal modality for their patients.

Table 3.

Summary of metabolites affected in CKD and ESKD identified/quantified by the targeted/untargeted metabolomics approach

ReferenceAnalysis platformSample typeStudy designFindings
Dahabiyeh et al. [92LC-MS Serum Two groups of patients; ESRD (n = 13) and CKD (n = 19). In ESRD, 8 patients were on HD and 4 on peritoneal dialysis. Patients in CKD group had different stages of CKD with a mean eGFR of 46.74 mL/min. A total of 193 metabolites were significantly altered in ESKD compared to CKD. Mainly, 2-aminobenzoic acid, xanthurenic acid, and hydroxy picolinic acid were upregulated in ESKD compared to CKD due to the significant decrease in GFR with the progression of CKD to ESKD. 
Ragi et al. [49LC-MS Plasma Blood samples were collected from 90 HD patients (both PRE-HD and POST-HD) and 74 HCs. The plasma samples were subjected to direct ESI-HRMS and LC/HRMS for untargeted metabolomics and LC-MS/MS for quantitative analysis. In the PRE-HD cohort, heightened concentrations of p-cresol sulfate and indoxyl sulfate were noted, while phenyl sulfate, catechol sulfate, and guaiacol sulfate exhibited levels approximately 50% of those observed for indoxyl sulfate. The dialytic clearance for indoxyl sulfate and p-cresol sulfate was determined to be below 35%, while the clearance rates for the remaining three sulfates ranged from 50% to 58%. 
Hu et al. [98LC-MS Plasma Plasma samples were collected from the patients with uremia and from healthy controls Reduced production of TMAO could serve as a novel marker to mitigate the progression of renal injury in CKD. 
Pallerla et al. [50GC-MS and LC-MS Plasma Blood samples of 60 HD patients (both PRE-HD and POST-HD), who were on regular maintenance HD twice/thrice a week, and 60 HCs were collected for GC-MS and LC-MS analysis. Phenylacetylglutamine displayed elevated levels in the HD groups when compared to the HC group. In contrast, glutamic acid exhibited lower concentrations in the HD groups in comparison to the HC group. The dialytic loss was found to range from 52% to 45% for arginine, lysine, and histidine, while glycine, cysteine, proline, alanine, threonine, glutamine, valine, and methionine showed dialytic losses ranging from 38% to 26%. Notably, aspartic acid, glutamic acid, asparagine, leucine, tyrosine, tryptophan, and isoleucine demonstrated minimal dialytic loss (≤12%). 
ReferenceAnalysis platformSample typeStudy designFindings
Dahabiyeh et al. [92LC-MS Serum Two groups of patients; ESRD (n = 13) and CKD (n = 19). In ESRD, 8 patients were on HD and 4 on peritoneal dialysis. Patients in CKD group had different stages of CKD with a mean eGFR of 46.74 mL/min. A total of 193 metabolites were significantly altered in ESKD compared to CKD. Mainly, 2-aminobenzoic acid, xanthurenic acid, and hydroxy picolinic acid were upregulated in ESKD compared to CKD due to the significant decrease in GFR with the progression of CKD to ESKD. 
Ragi et al. [49LC-MS Plasma Blood samples were collected from 90 HD patients (both PRE-HD and POST-HD) and 74 HCs. The plasma samples were subjected to direct ESI-HRMS and LC/HRMS for untargeted metabolomics and LC-MS/MS for quantitative analysis. In the PRE-HD cohort, heightened concentrations of p-cresol sulfate and indoxyl sulfate were noted, while phenyl sulfate, catechol sulfate, and guaiacol sulfate exhibited levels approximately 50% of those observed for indoxyl sulfate. The dialytic clearance for indoxyl sulfate and p-cresol sulfate was determined to be below 35%, while the clearance rates for the remaining three sulfates ranged from 50% to 58%. 
Hu et al. [98LC-MS Plasma Plasma samples were collected from the patients with uremia and from healthy controls Reduced production of TMAO could serve as a novel marker to mitigate the progression of renal injury in CKD. 
Pallerla et al. [50GC-MS and LC-MS Plasma Blood samples of 60 HD patients (both PRE-HD and POST-HD), who were on regular maintenance HD twice/thrice a week, and 60 HCs were collected for GC-MS and LC-MS analysis. Phenylacetylglutamine displayed elevated levels in the HD groups when compared to the HC group. In contrast, glutamic acid exhibited lower concentrations in the HD groups in comparison to the HC group. The dialytic loss was found to range from 52% to 45% for arginine, lysine, and histidine, while glycine, cysteine, proline, alanine, threonine, glutamine, valine, and methionine showed dialytic losses ranging from 38% to 26%. Notably, aspartic acid, glutamic acid, asparagine, leucine, tyrosine, tryptophan, and isoleucine demonstrated minimal dialytic loss (≤12%). 

GFR, glomerular filtration rate; ESI, electrospray ionization; PRE-HD, pre-dialysis; POST-HD, post-dialysis; HC, healthy control.

Metabolomics has significantly contributed to our understanding of the dynamic changes in renal metabolism associated with kidney diseases, revealing shifts in metabolite profiles that can act as biomarkers for the early detection and monitoring of these conditions. However, the spatial resolution and capability of traditional metabolomic techniques to capture metabolite distribution within specific kidney regions and cell types are not possible. By identifying which structures or cell types within the kidney experience disturbances in metabolic pathways, we can target therapeutics more effectively and reduce off-target effects. To create a detailed spatial map of kidney metabolites, MALDI-MSI has emerged as a powerful tool, enabling researchers to uncover precise alterations in metabolites at a spatial level, identify potential therapeutic targets, and advance our comprehension of the pathophysiological processes involved in kidney diseases [100]. In recent studies [19] involving the Chronic Renal Insufficiency Cohort (CRIC; including n = 558 with normal albuminuria or microalbuminuria and n = 341 with MAC) [101], the Singapore Study of Macro-Angiopathy and Reactivity in Type 2 Diabetes (SMART2D; n = 309), and an American Indian cohort (n = 54), advanced Zip-Chip-MS and MALDI-MSI (spatial metabolomics) techniques were employed to analyze kidney tissues and urine samples. The study demonstrated that the urine adenine/creatinine ratio (UAdCR) measurement was closely linked to the progression of DKD in the American Indian, CRIC, and SMART2D cohorts with T2DM without MAC. Notably, the UAdCR emerged as an independent predictor of ESKD and all-cause mortality. The distribution (localization) of adenine in kidney tissue from healthy control subjects (shown in Fig. 3a) and diabetic kidney tissue (shown in Fig. 3b) was performed using MALDI-MSI, and the comparative analyses between these two study groups are shown in Figure 3c. These findings demonstrated that adenine is co-localized with regions of tubulointerstitial and vascular pathology. The study also demonstrated that inhibition of adenine is beneficial in the mouse model of DKD. Overall, the data indicate the promising clinical utility of the UAdCR as a potential precision biomarker for identifying high-risk patients, despite lack of proteinuria and potential targeted therapeutics for these high risk patients. Adenine has now been demonstrated to be increased in DKD in both patients with T1D and T2D, thus establishing itself as a major cause of kidney disease. The measurement of adenine in the urine appears to reflect kidney production of adenine and is therefore clinically relevant. The authors have found that high levels of urine adenine identify patients at high risk of kidney failure across many ethnicities and geographic regions and detection of adenine in kidney tissue is meaningful during clinical examinations, especially with the spatial metabolomics as it provides valuable insights into the localization and concentration of adenine within the kidney tissue. Investigating into the involvement of adenine in kidney diseases holds the potential to enhance our comprehension of renal pathophysiology. This exploration offers the opportunity to uncover molecular mechanisms, identify biomarkers, and introduce novel paths for therapeutic interventions. The outcomes of the research into adenine metabolism carry the promise of refining diagnostic precision, enhancing treatment effectiveness and ultimately improving overall patient outcomes within the field of kidney diseases.

Fig. 3.

Spatial metabolomics identifies adenine in regions of pathology in non-macroalbuminuric patients with diabetes. a Adenine was localized to regions of normal glomeruli and vessels in the normal kidney. b In a diabetic kidney, adenine is diffusely increased across the tissue section and prominent in regions of sclerotic blood vessels, glomeruli with mild sclerosis, and regions of atrophic tubules and interstitial inflammation. c Two-tailed Student’s t test was used for the comparison. Data represent mean ± SEM. This figure was reprinted from a recent JCI article [19].

Fig. 3.

Spatial metabolomics identifies adenine in regions of pathology in non-macroalbuminuric patients with diabetes. a Adenine was localized to regions of normal glomeruli and vessels in the normal kidney. b In a diabetic kidney, adenine is diffusely increased across the tissue section and prominent in regions of sclerotic blood vessels, glomeruli with mild sclerosis, and regions of atrophic tubules and interstitial inflammation. c Two-tailed Student’s t test was used for the comparison. Data represent mean ± SEM. This figure was reprinted from a recent JCI article [19].

Close modal

A recent (spatial metabolomics) study using apolipoprotein E knockout mice, diabetes was induced through streptozotocin injections, while the control group received citrate injections, and both were subjected to a cholesterol-enriched diet. This model consistently shows elevated plasma cholesterol levels compared to wild-type mice (∼400 mg/dL vs. ∼80 mg/dL), regardless of diet. Interestingly, the cholesterol-enriched diet is not expected to significantly impact mouse cholesterol levels in their setup. However, authors studied whether elevated cholesterol may influence phosphatidylinositol (PI) metabolic changes in the proximal tubular PT_S3 segment, given the known interaction between PI lipids and cholesterol homeostasis. The crosstalk between PI lipid and cholesterol pathways, linked through acetyl-CoA, suggests potential effects on both cholesterol and PI lipid species due to high glucose levels in the diabetic setting. Metabolic histology analysis using MALDI-MSI highlighted the sensitivity of the PT_S3 segment to diabetes-induced changes, suggesting its role as an accelerating factor in DKD [102].

An important study by Wang et al. [103] pioneered an elegant application of MALDI-MSI for high-spatial resolution metabolomics coupled with isotope tracing, to address the intricate challenge of cell-to-cell heterogeneity during ischemia in complex tissues like the kidney. This innovative approach facilitates the precise mapping of cell-type-specific dynamic changes in central carbon metabolism, even within heterogeneous tissue architectures. Through strategic integration with multiplexed immunofluorescence staining, the authors successfully discerned metabolic changes in endothelial and proximal tubular cell types within a renal ischemia-reperfusion injury model. Notably, this methodology revealed region-specific metabolic perturbations during both the injury and recovery phases, uncovering unexpected anomalies in ostensibly normal cells. The authors aptly demonstrated the power of this approach in unraveling the complexities of kidney homeostasis, illustrating its broader potential for investigating tissue homeostasis across diverse organs [103].

While metabolomics plays a crucial role in understanding biological processes, its capacity to provide a comprehensive description of these processes is limited. The evolving field generates a wealth of data that must be integrated and analyzed alongside other omics data for complete interpretation. Combining metabolomic analysis with bioinformatics yields crucial insights into complex diseases. Common statistical analyses of metabolomics data include univariate methods like fold change analysis, t tests, and volcano plots for two-group data, one-way ANOVA, post hoc analysis, and correlation analysis for multigroup data. Multivariate approaches, including multiple variance analysis, multiple regression analysis, principal component analysis, partial least squares discriminant analysis, cluster analysis, and machine learning methods such as random forest and Support Vector Machine, allow the evaluation of input metabolomic datasets and identification of metabolites undergoing abnormal changes. Subsequent data mining methods help distinguish functionally relevant metabolites [104]. The selection of a suitable p value for ranking significantly expressed metabolites is crucial for determining a reliable threshold [105]. Metabolomics analyses rely on powerful software tools such as MetaboAnalyst 5.0, MS-DIAL 4.0, MetFlow, METLIN, MetaSpace, GraphPad, and Lilikoi 2.0, among others, which aid in establishing an effective metabolomics analysis pipeline for researchers [106].

Over the past decade, significant progress has been made in metabolomics, broadening the detection of various metabolites. However, when compared to transcriptomics and proteomics, which offer near-complete coverage, metabolomics currently captures only a small portion of these numerous metabolites. The chemical diversity of metabolites and their wide-ranging levels within cells add complexity to metabolite measurements. The current literature does not support the possibility of extracting and measuring all metabolites through a single extraction and analytical method. Therefore, researchers often employ various extraction techniques and combinations of analytical approaches to enhance metabolite coverage. Given the diversity in the goals and methodologies of metabolite analyses, it is crucial to establish guidelines for acquiring and reporting metabolite data as there are numerous potential sources of error and misinterpretation. Hence, there is a pressing need to develop guidelines to ensure the robustness of obtained and reported metabolite data. These guidelines should encompass sampling, extraction, storage, metabolite identification, handling large sample numbers, and recommendations for reporting metabolite identification methods and the levels of certainty in quantification. Notably, Fernie et al. [107] have made recommendations for reporting metabolite data, which they suggest should be a standard practice for future manuscript submissions. These recommendations cover both LC-MS and GC-MS data, enhancing the quality of metabolite data reporting and contributing to community efforts to improve metabolite annotation, ultimately benefiting the field of metabolomics.

Metabolomics stands at the forefront of precision medicine, merging omics and clinical data with epidemiological and environmental factors to enable precise diagnoses and personalized interventions tailored to each patient. In the context of kidney disease management, biomarkers are indispensable for improving survival rates and preventing the onset of kidney disease. Several large consortiums, including the Kidney Precision Medicine Project (KPMP) and the Human Biomolecular Atlas Program (HubMap), integrate spatial and bulk metabolomics as critical platforms in interrogating human tissue samples [108, 109]. However, a major hurdle for metabolite identification persists in the scarcity of high-quality spectral data across diverse instruments and collision energies. Identifying unidentified mass spectral features remains a major challenge in the field of metabolomics. Recent progress in instrument technology, experimental methodologies, and data analysis softwares such as PubChem, ChemSpider, the HMDB, BioCyc, and KEGG has alleviated many of the challenges associated with MS-based metabolomics. To enhance the confidence of compound identification from MS-based datasets, future strategies should focus on techniques that eliminate unwanted MS features, expand MS databases and data curation, automate metabolite identification, and standardize analytical methods across various platforms. Development in terms of instrumentation and data analysis programs is essential to tackle the intricate biological complexities. Anticipated advancements are expected in single-cell metabolomics and MSI to gain insights into the pathophysiology of kidney diseases. Recognizing the heterogeneous nature of the kidney tissue and the diverse subcellular processes within it, MS imaging has gained prominence for revealing the spatial distribution of specific metabolites in kidney tissues. Various imaging techniques, including MALDI, nanostructure imaging MS, DESI-MS, and secondary-ion MS, contribute to this field. Notably, MALDI-MSI has emerged as a powerful and sophisticated method with high sensitivity and throughput, enabling the localization of a broad spectrum of metabolites within specific regions of tissue samples [19, 55]. Recent groundbreaking discoveries using the MALDI-MSI approach [19, 110, 111] have accelerated the clinical application of metabolomics. The recent study to demonstrate that the UAdCR measurement closely correlates with DKD progression has provided compelling evidence to add the UAdCR to be a new independent predictor of ESKD and mortality [112], adding to conventional markers of albuminuria and baseline eGFR [19]. While MALDI-MSI has been instrumental in enhancing our understanding of various diseases, there remains a need to confirm the spatial confirmation of metabolites through further MALDI-MS/MS analysis. These evolving perspectives are poised to significantly contribute to the understanding and management of various diseases, including different types of kidney diseases.

The review article covers several of metabolomics and the accelerated advances in the field, especially in the last 2–3 years. Spatial metabolomics has provided dramatic insights by linking metabolites to disease features, and the kidney biopsy has been an excellent example of how to incorporate several omics datasets for clinical implications. The uAdCR shows promise as a new disease causing diagnostic marker. The identification and examination of adenine and related metabolites is expected to enhance our understanding of kidney disease pathophysiology and offers encouraging prospects for better prognosis and early diagnosis. Current limitations of bulk and spatial metabolomics include incomplete and nonoverlapping metabolite coverage across various methods, the necessity for structural confirmation, and the introduction of variations during sample preparation and data analysis. Advancements in high-resolution imaging and atmospheric pressure imaging are on the horizon. The anticipated use of MSI holds the promise of a deeper understanding of kidney pathology as it enables the visualization and localization of metabolites within specific regions of tissue samples.

K.S. is a member of the DSMB for Cara Therapeutics and has had research support from Boehringer-Ingelheim. He holds equity in SygnaMap.

This work was supported by NIDDK UO1, VA Merit, and Department of Defense grants.

N.R. contributed to the design, prepared the initial draft, and provided final approval for the published version. K.S. contributed to the conception, conducted critical revisions, and gave the final approval for the published version.

1.
Camara
NO
,
Iseki
K
,
Kramer
H
,
Liu
ZH
,
Sharma
K
.
Kidney disease and obesity: epidemiology, mechanisms and treatment
.
Nat Rev Nephrol
.
2017
;
13
(
3
):
181
90
.
2.
Hoste
EAJ
,
Kellum
JA
,
Selby
NM
,
Zarbock
A
,
Palevsky
PM
,
Bagshaw
SM
, et al
.
Global epidemiology and outcomes of acute kidney injury
.
Nat Rev Nephrol
.
2018
;
14
(
10
):
607
25
.
3.
Charles
C
,
Ferris
AH
.
Chronic kidney disease
.
Prim Care
.
2020
;
47
(
4
):
585
95
.
4.
Levey
AS
,
James
MT
.
Acute kidney injury
.
Ann Intern Med
.
2017
;
167
(
9
):
ITC66
80
.
5.
Rheinberger
M
,
Boger
CA
.
[Diabetic nephropathy: new insights into diagnosis, prevention and treatment]
.
Dtsch Med Wochenschr
.
2014
;
139
(
14
):
704
6
.
6.
Johansen
KL
,
Chertow
GM
,
Foley
RN
,
Gilbertson
DT
,
Herzog
CA
,
Ishani
A
, et al
.
US renal data system 2020 annual data report: epidemiology of kidney disease in the United States
.
Am J Kidney Dis
.
2021
;
77
(
4 Suppl 1
):
A7
A8
.
7.
Ramakrishnan
K
,
Scheid
DC
.
Diagnosis and management of acute pyelonephritis in adults
.
Am Fam Physician
.
2005
;
71
(
5
):
933
42
.
8.
The OPTN Website offers a wealth of information about transplantation
. Available from: https://optn.transplant.hrsa.gov/.
9.
Thurlow
JS
,
Joshi
M
,
Yan
G
,
Norris
KC
,
Agodoa
LY
,
Yuan
CM
, et al
.
Global epidemiology of end-stage kidney disease and disparities in kidney replacement therapy
.
Am J Nephrol
.
2021
;
52
(
2
):
98
107
.
10.
Erfanpoor
S
,
Etemad
K
,
Kazempour
S
,
Hadaegh
F
,
Hasani
J
,
Azizi
F
, et al
.
Diabetes, hypertension, and incidence of chronic kidney disease: is there any multiplicative or additive interaction
.
Int J Endocrinol Metab
.
2021
;
19
(
1
):
e101061
.
11.
Kovesdy
CP
.
Epidemiology of chronic kidney disease: an update 2022
.
Kidney Int Suppl
.
2022
;
12
(
1
):
7
11
.
12.
Luyckx
VA
,
Al-Aly
Z
,
Bello
AK
,
Bellorin-Font
E
,
Carlini
RG
,
Fabian
J
, et al
.
Sustainable Development Goals relevant to kidney health: an update on progress
.
Nat Rev Nephrol
.
2021
;
17
(
1
):
15
32
.
13.
Susantitaphong
P
,
Cruz
DN
,
Cerda
J
,
Abulfaraj
M
,
Alqahtani
F
,
Koulouridis
I
, et al
.
World incidence of AKI: a meta-analysis
.
Clin J Am Soc Nephrol
.
2013
;
8
(
9
):
1482
93
.
14.
Chawla
LS
,
Bellomo
R
,
Bihorac
A
,
Goldstein
SL
,
Siew
ED
,
Bagshaw
SM
, et al
.
Acute kidney disease and renal recovery: consensus report of the Acute Disease Quality Initiative (ADQI) 16 Workgroup
.
Nat Rev Nephrol
.
2017
;
13
(
4
):
241
57
.
15.
Rewa
O
,
Bagshaw
SM
.
Acute kidney injury-epidemiology, outcomes and economics
.
Nat Rev Nephrol
.
2014
;
10
(
4
):
193
207
.
16.
Chang
CH
,
Chen
SW
,
Chen
JJ
,
Chan
YH
,
Yen
CL
,
Lee
TH
, et al
.
Incidence and transition of acute kidney injury, acute kidney disease to chronic kidney disease after acute type A aortic dissection surgery
.
J Clin Med
.
2021
;
10
(
20
):
4769
.
17.
Gross
JL
,
de Azevedo
MJ
,
Silveiro
SP
,
Canani
LH
,
Caramori
ML
,
Zelmanovitz
T
.
Diabetic nephropathy: diagnosis, prevention, and treatment
.
Diabetes Care
.
2005
;
28
(
1
):
164
76
.
18.
Ferenbach
DA
,
Bonventre
JV
.
Acute kidney injury and chronic kidney disease: from the laboratory to the clinic
.
Nephrol Ther
.
2016
;
12
(
Suppl 1
):
S41
8
.
19.
Sharma
K
,
Zhang
G
,
Hansen
J
,
Bjornstad
P
,
Lee
HJ
,
Menon
R
, et al
.
Endogenous adenine mediates kidney injury in diabetic models and predicts diabetic kidney disease in patients
.
J Clin Invest
.
2023
;
133
(
20
):
e170341
.
20.
Dos Santos
IF
,
Sheriff
S
,
Amlal
S
,
Ahmed
RPH
,
Thakar
CV
,
Amlal
H
.
Adenine acts in the kidney as a signaling factor and causes salt- and water-losing nephropathy: early mechanism of adenine-induced renal injury
.
Am J Physiol Ren Physiol
.
2019
;
316
(
4
):
F743
57
.
21.
Qiu
YL
,
Zhu
H
,
Ma
SG
,
Liu
H
,
Li
Q
,
Ji
CM
.
Evaluation of inflammatory and oxidative biomarkers in children with well-controlled congenital hypothyroidism
.
J Pediatr Endocrinol Metab
.
2015
;
28
(
7–8
):
761
5
.
22.
Johnson
RJ
,
Lanaspa
MA
,
Gaucher
EA
.
Uric acid: a danger signal from the RNA world that may have a role in the epidemic of obesity, metabolic syndrome, and cardiorenal disease: evolutionary considerations
.
Semin Nephrol
.
2011
;
31
(
5
):
394
9
.
23.
Klahr
S
,
Levey
AS
,
Beck
GJ
,
Caggiula
AW
,
Hunsicker
L
,
Kusek
JW
, et al
.
The effects of dietary protein restriction and blood-pressure control on the progression of chronic renal disease. Modification of Diet in Renal Disease Study Group
.
N Engl J Med
.
1994
;
330
(
13
):
877
84
.
24.
Slominska
EM
,
Szolkiewicz
M
,
Smolenski
RT
,
Rutkowski
B
,
Swierczynski
J
.
High plasma adenine concentration in chronic renal failure and its relation to erythrocyte ATP
.
Nephron
.
2002
;
91
(
2
):
286
91
.
25.
Jdiaa
SS
,
Mansour
R
,
El Alayli
A
,
Gautam
A
,
Thomas
P
,
Mustafa
RA
.
COVID-19 and chronic kidney disease: an updated overview of reviews
.
J Nephrol
.
2022
;
35
(
1
):
69
85
.
26.
Patschan
D
,
Patschan
S
,
Matyukhin
I
,
Hoffmeister
M
,
Lauxmann
M
,
Ritter
O
, et al
.
Metabolomics in acute kidney injury: the experimental perspective
.
J Clin Med Res
.
2023
;
15
(
6
):
283
91
.
27.
Sharma
K
,
Karl
B
,
Mathew
AV
,
Gangoiti
JA
,
Wassel
CL
,
Saito
R
, et al
.
Metabolomics reveals signature of mitochondrial dysfunction in diabetic kidney disease
.
J Am Soc Nephrol
.
2013
;
24
(
11
):
1901
12
.
28.
Darshi
M
,
Van Espen
B
,
Sharma
K
.
Metabolomics in diabetic kidney disease: unraveling the biochemistry of a silent killer
.
Am J Nephrol
.
2016
;
44
(
2
):
92
103
.
29.
Bergmann
C
,
Guay-Woodford
LM
,
Harris
PC
,
Horie
S
,
Peters
DJM
,
Torres
VE
.
Polycystic kidney disease
.
Nat Rev Dis Primers
.
2018
;
4
(
1
):
50
.
30.
Song
Z
,
Wu
H
,
Cao
H
,
Tang
M
,
Yang
S
,
Qin
L
.
Nephrotic syndrome with acute pulmonary embolism in young adults: two case reports
.
Medicine
.
2018
;
97
(
29
):
e11495
.
31.
Ameer
OZ
.
Hypertension in chronic kidney disease: what lies behind the scene
.
Front Pharmacol
.
2022
;
13
:
949260
.
32.
Li
PK
,
Garcia-Garcia
G
,
Lui
SF
,
Andreoli
S
,
Fung
WW
,
Hradsky
A
, et al
.
Kidney health for everyone everywhere - from prevention to detection and equitable access to care
.
Indian J Nephrol
.
2020
;
30
(
2
):
63
71
.
33.
Whaley-Connell
A
,
Nistala
R
,
Chaudhary
K
.
The importance of early identification of chronic kidney disease
.
Mo Med
.
108
(
1
):
25
8
.
34.
Zhang
WR
,
Parikh
CR
.
Biomarkers of acute and chronic kidney disease
.
Annu Rev Physiol
.
2019
;
81
:
309
33
.
35.
Obert
LA
,
Elmore
SA
,
Ennulat
D
,
Frazier
KS
.
A review of specific biomarkers of chronic renal injury and their potential application in nonclinical safety assessment studies
.
Toxicol Pathol
.
2021
;
49
(
5
):
996
1023
.
36.
King
H
,
Aubert
RE
,
Herman
WH
.
Global burden of diabetes, 1995-2025: prevalence, numerical estimates, and projections
.
Diabetes Care
.
1998
;
21
(
9
):
1414
31
.
37.
Zelnick
LR
,
Leca
N
,
Young
B
,
Bansal
N
.
Association of the estimated glomerular filtration rate with vs without a coefficient for race with time to eligibility for kidney transplant
.
JAMA Netw Open
.
2021
;
4
(
1
):
e2034004
.
38.
Zsom
L
,
Zsom
M
,
Salim
SA
,
Fulop
T
.
Estimated glomerular filtration rate in chronic kidney disease: a critical review of estimate-based predictions of individual outcomes in kidney disease
.
Toxins
.
2022
;
14
(
2
):
127
.
39.
Vemuri
P
,
Knopman
DS
,
Jack
CR
Jr
,
Lundt
ES
,
Weigand
SD
,
Zuk
SM
, et al
.
Association of kidney function biomarkers with brain MRI findings: the BRINK study
.
J Alzheimers Dis
.
2017
;
55
(
3
):
1069
82
.
40.
Zhao
Y
,
Wang
Y
,
Xu
Y
,
Zhao
Y
,
Qu
Y
,
Zhang
H
, et al
.
Comparison of diagnostic value for chronic kidney disease between 640-slice computed tomography kidney scan and conventional computed tomography scan
.
Contrast Media Mol Imaging
.
2022
;
2022
:
6587617
.
41.
Blackmond
N
,
Loh
T
,
Kanke
JD
,
Khilanani
U
,
Weitzman
R
.
Challenges in management of kidney failure in a free clinic setting: a case report
.
Cureus
.
2022
;
14
(
6
):
e26352
.
42.
Go
AS
,
Chertow
GM
,
Fan
D
,
McCulloch
CE
,
Hsu
CY
.
Chronic kidney disease and the risks of death, cardiovascular events, and hospitalization
.
N Engl J Med
.
2004
;
351
(
13
):
1296
305
.
43.
de Boer
IH
,
Khunti
K
,
Sadusky
T
,
Tuttle
KR
,
Neumiller
JJ
,
Rhee
CM
, et al
.
Diabetes management in chronic kidney disease: a consensus report by the American diabetes association (ADA) and kidney disease: improving global outcomes (KDIGO)
.
Diabetes Care
.
2022
;
45
(
12
):
3075
90
.
44.
Montemayor
D
,
Sharma
K
.
mGWAS: next generation genetic prediction in kidney disease
.
Nat Rev Nephrol
.
2020
;
16
(
5
):
255
6
.
45.
Beckonert
O
,
Keun
HC
,
Ebbels
TM
,
Bundy
J
,
Holmes
E
,
Lindon
JC
, et al
.
Metabolic profiling, metabolomic and metabonomic procedures for NMR spectroscopy of urine, plasma, serum and tissue extracts
.
Nat Protoc
.
2007
;
2
(
11
):
2692
703
.
46.
Pan
Z
,
Raftery
D
.
Comparing and combining NMR spectroscopy and mass spectrometry in metabolomics
.
Anal Bioanal Chem
.
2007
;
387
(
2
):
525
7
.
47.
Parmar
D
,
Bhattacharya
N
,
Kannan
S
,
Vadivel
S
,
Pandey
GK
,
Ghanate
A
, et al
.
Plausible diagnostic value of urinary isomeric dimethylarginine ratio for diabetic nephropathy
.
Sci Rep
.
2020
;
10
(
1
):
2970
.
48.
Goodacre
R
,
Vaidyanathan
S
,
Dunn
WB
,
Harrigan
GG
,
Kell
DB
.
Metabolomics by numbers: acquiring and understanding global metabolite data
.
Trends Biotechnol
.
2004
;
22
(
5
):
245
52
.
49.
Ragi
N
,
Pallerla
P
,
Babi Reddy Gari
AR
,
Lingampelly
SS
,
Ketavarapu
V
,
Addipilli
R
, et al
.
Assessment of uremic toxins in advanced chronic kidney disease patients on maintenance hemodialysis by LC-ESI-MS/MS
.
Metabolomics
.
2023
;
19
(
3
):
14
.
50.
Pallerla
P
,
Ragi
N
,
Gari
A
,
Bhumireddy
SR
,
Addipilli
R
,
Rodda
R
, et al
.
Evaluation of amino acids and other related metabolites levels in end-stage renal disease (ESRD) patients on hemodialysis by LC/MS/MS and GC/MS
.
Anal Bioanal Chem
.
2023
;
415
(
26
):
6491
509
.
51.
Meister
I
,
Zhang
P
,
Sinha
A
,
Skold
CM
,
Wheelock
AM
,
Izumi
T
, et al
.
High-precision automated workflow for urinary untargeted metabolomic epidemiology
.
Anal Chem
.
2021
;
93
(
12
):
5248
58
.
52.
Benito
S
,
Sanchez-Ortega
A
,
Unceta
N
,
Andrade
F
,
Aldamiz-Echevarria
L
,
Goicolea
MA
, et al
.
Untargeted metabolomics for plasma biomarker discovery for early chronic kidney disease diagnosis in pediatric patients using LC-QTOF-MS
.
Analyst
.
2018
;
143
(
18
):
4448
58
.
53.
Ivanisevic
J
,
Thomas
A
.
Metabolomics as a tool to understand pathophysiological processes
.
Methods Mol Biol
.
2018
;
1730
:
3
28
.
54.
Benito
S
,
Sanchez
A
,
Unceta
N
,
Andrade
F
,
Aldamiz-Echevarria
L
,
Goicolea
MA
, et al
.
LC-QTOF-MS-based targeted metabolomics of arginine-creatine metabolic pathway-related compounds in plasma: application to identify potential biomarkers in pediatric chronic kidney disease
.
Anal Bioanal Chem
.
2016
;
408
(
3
):
747
60
.
55.
Miyamoto
S
,
Hsu
CC
,
Hamm
G
,
Darshi
M
,
Diamond-Stanic
M
,
Decleves
AE
, et al
.
Mass spectrometry imaging reveals elevated glomerular ATP/AMP in diabetes/obesity and identifies sphingomyelin as a possible mediator
.
EBioMedicine
.
2016
;
7
:
121
34
.
56.
Yang
M
,
Unsihuay
D
,
Hu
H
,
Nguele Meke
F
,
Qu
Z
,
Zhang
ZY
, et al
.
Nano-DESI mass spectrometry imaging of proteoforms in biological tissues with high spatial resolution
.
Anal Chem
.
2023
;
95
(
12
):
5214
22
.
57.
Liu
Y
,
Yan
S
,
Ji
C
,
Dai
W
,
Hu
W
,
Zhang
W
, et al
.
Metabolomic changes and protective effect of (L)-carnitine in rat kidney ischemia/reperfusion injury
.
Kidney Blood Press Res
.
2012
;
35
(
5
):
373
81
.
58.
Wei
Q
,
Xiao
X
,
Fogle
P
,
Dong
Z
.
Changes in metabolic profiles during acute kidney injury and recovery following ischemia/reperfusion
.
PLoS One
.
2014
;
9
(
9
):
e106647
.
59.
Elmariah
S
,
Farrell
LA
,
Daher
M
,
Shi
X
,
Keyes
MJ
,
Cain
CH
, et al
.
Metabolite profiles predict acute kidney injury and mortality in patients undergoing transcatheter aortic valve replacement
.
J Am Heart Assoc
.
2016
;
5
(
3
):
e002712
.
60.
Zhang
F
,
Wang
Q
,
Xia
T
,
Fu
S
,
Tao
X
,
Wen
Y
, et al
.
Diagnostic value of plasma tryptophan and symmetric dimethylarginine levels for acute kidney injury among tacrolimus-treated kidney transplant patients by targeted metabolomics analysis
.
Sci Rep
.
2018
;
8
(
1
):
14688
.
61.
Tan
B
,
Chen
J
,
Qin
S
,
Liao
C
,
Zhang
Y
,
Wang
D
, et al
.
Tryptophan pathway-targeted metabolomics study on the mechanism and intervention of cisplatin-induced acute kidney injury in rats
.
Chem Res Toxicol
.
2021
;
34
(
7
):
1759
68
.
62.
Chen
C
,
Zhang
P
,
Bao
G
,
Fang
Y
,
Chen
W
.
Discovery of potential biomarkers in acute kidney injury by ultra-high-performance liquid chromatography-tandem quadrupole time-of-flight mass spectrometry (UPLC-Q/TOF-MS)
.
Int Urol Nephrol
.
2021
;
53
(
12
):
2635
43
.
63.
Tian
M
,
Liu
X
,
Chen
L
,
Hu
S
,
Zheng
Z
,
Wang
L
, et al
.
Urine metabolites for preoperative prediction of acute kidney injury after coronary artery bypass graft surgery
.
J Thorac Cardiovasc Surg
.
2023
;
165
(
3
):
1165
75.e3
.
64.
Lee
HS
,
Kim
SM
,
Jang
JH
,
Park
HD
,
Lee
SY
.
Serum 5-hydroxyindoleacetic acid and ratio of 5-hydroxyindoleacetic acid to serotonin as metabolomics indicators for acute oxidative stress and inflammation in vancomycin-associated acute kidney injury
.
Antioxidants
.
2021
;
10
(
6
):
895
.
65.
Franiek
A
,
Sharma
A
,
Cockovski
V
,
Wishart
DS
,
Zappitelli
M
,
Blydt-Hansen
TD
.
Urinary metabolomics to develop predictors for pediatric acute kidney injury
.
Pediatr Nephrol
.
2022
;
37
(
9
):
2079
90
.
66.
Gisewhite
S
,
Stewart
IJ
,
Beilman
G
,
Lusczek
E
.
Urinary metabolites predict mortality or need for renal replacement therapy after combat injury
.
Crit Care
.
2021
;
25
(
1
):
119
.
67.
Sun
J
,
Cao
Z
,
Schnackenberg
L
,
Pence
L
,
Yu
LR
,
Choudhury
D
, et al
.
Serum metabolite profiles predict outcomes in critically ill patients receiving renal replacement therapy
.
J Chromatogr B Analyt Technol Biomed Life Sci
.
2021
;
1187
:
123024
.
68.
Ion
V
,
Nys
G
,
Cobraiville
G
,
Cavalier
E
,
Crommen
J
,
Servais
AC
, et al
.
Ultra-high-performance liquid chromatography-mass spectrometry method for neutrophil gelatinase-associated lipocalin as a predictive biomarker in acute kidney injury
.
Talanta
.
2019
;
195
:
668
75
.
69.
Lim
YJ
,
Xiu
SG
,
Kuruvilla
MS
,
Winquist
E
,
Welch
S
,
Black
M
, et al
.
Metabolomic identification of predictive and early biomarkers of cisplatin-induced acute kidney injury in adult head and neck cancer patients
.
Br J Clin Pharmacol
.
2023
.
70.
Wang
TJ
,
Larson
MG
,
Vasan
RS
,
Cheng
S
,
Rhee
EP
,
McCabe
E
, et al
.
Metabolite profiles and the risk of developing diabetes
.
Nat Med
.
2011
;
17
(
4
):
448
53
.
71.
Newgard
CB
,
An
J
,
Bain
JR
,
Muehlbauer
MJ
,
Stevens
RD
,
Lien
LF
, et al
.
A branched-chain amino acid-related metabolic signature that differentiates obese and lean humans and contributes to insulin resistance
.
Cell Metab
.
2009
;
9
(
4
):
311
26
.
72.
Guasch-Ferre
M
,
Hruby
A
,
Toledo
E
,
Clish
CB
,
Martinez-Gonzalez
MA
,
Salas-Salvado
J
, et al
.
Metabolomics in prediabetes and diabetes: a systematic review and meta-analysis
.
Diabetes Care
.
2016
;
39
(
5
):
833
46
.
73.
Merino
J
,
Leong
A
,
Liu
CT
,
Porneala
B
,
Walford
GA
,
von Grotthuss
M
, et al
.
Metabolomics insights into early type 2 diabetes pathogenesis and detection in individuals with normal fasting glucose
.
Diabetologia
.
2018
;
61
(
6
):
1315
24
.
74.
Hirayama
A
,
Nakashima
E
,
Sugimoto
M
,
Akiyama
S
,
Sato
W
,
Maruyama
S
, et al
.
Metabolic profiling reveals new serum biomarkers for differentiating diabetic nephropathy
.
Anal Bioanal Chem
.
2012
;
404
(
10
):
3101
9
.
75.
Zhang
X
,
Wang
Y
,
Hao
F
,
Zhou
X
,
Han
X
,
Tang
H
, et al
.
Human serum metabonomic analysis reveals progression axes for glucose intolerance and insulin resistance statuses
.
J Proteome Res
.
2009
;
8
(
11
):
5188
95
.
76.
Rathod
R
,
Gajera
B
,
Nazir
K
,
Wallenius
J
,
Velagapudi
V
.
Simultaneous measurement of tricarboxylic acid cycle intermediates in different biological matrices using liquid chromatography-tandem mass spectrometry; quantitation and comparison of TCA cycle intermediates in human serum, plasma, kasumi-1 cell and murine liver tissue
.
Metabolites
.
2020
;
10
(
3
):
103
.
77.
Peng
W
,
Tan
S
,
Xu
Y
,
Wang
L
,
Qiu
D
,
Cheng
C
, et al
.
LC-MS/MS metabolome analysis detects the changes in the lipid metabolic profiles of dMMR and pMMR cells
.
Oncol Rep
.
2018
;
40
(
2
):
1026
34
.
78.
van der Kloet FM
.
Discovery of early-stage biomarkers for diabetic kidney disease using ms-based metabolomics (FinnDiane study)
.
Metabolomics
.
2012
.
79.
Spiegelman
BM
.
Transcriptional control of mitochondrial energy metabolism through the PGC1 coactivators
.
Novartis Found Symp
.
2007
;
287
:
60
3
; discussion 63-9.
80.
Barres
R
,
Osler
ME
,
Yan
J
,
Rune
A
,
Fritz
T
,
Caidahl
K
, et al
.
Non-CpG methylation of the PGC-1alpha promoter through DNMT3B controls mitochondrial density
.
Cel Metab
.
2009
;
10
(
3
):
189
98
.
81.
Zhang
S
,
Li
X
,
Luo
H
,
Fang
ZZ
,
Ai
H
.
Role of aromatic amino acids in pathogeneses of diabetic nephropathy in Chinese patients with type 2 diabetes
.
J Diabetes Complications
.
2020
;
34
(
10
):
107667
.
82.
Tavares
G
,
Venturini
G
,
Padilha
K
,
Zatz
R
,
Pereira
AC
,
Thadhani
RI
, et al
.
1,5-Anhydroglucitol predicts CKD progression in macroalbuminuric diabetic kidney disease: results from non-targeted metabolomics
.
Metabolomics
.
2018
;
14
(
4
):
39
.
83.
Saulnier
PJ
,
Darshi
M
,
Wheelock
KM
,
Looker
HC
,
Fufaa
GD
,
Knowler
WC
, et al
.
Urine metabolites are associated with glomerular lesions in type 2 diabetes
.
Metabolomics
.
2018
;
14
(
6
):
84
.
84.
Niewczas
MA
,
Sirich
TL
,
Mathew
AV
,
Skupien
J
,
Mohney
RP
,
Warram
JH
, et al
.
Uremic solutes and risk of end-stage renal disease in type 2 diabetes: metabolomic study
.
Kidney Int
.
2014
;
85
(
5
):
1214
24
.
85.
Ng
DP
,
Salim
A
,
Liu
Y
,
Zou
L
,
Xu
FG
,
Huang
S
, et al
.
A metabolomic study of low estimated GFR in non-proteinuric type 2 diabetes mellitus
.
Diabetologia
.
2012
;
55
(
2
):
499
508
.
86.
Zhang
H
,
Zuo
JJ
,
Dong
SS
,
Lan
Y
,
Wu
CW
,
Mao
GY
, et al
.
Identification of potential serum metabolic biomarkers of diabetic kidney disease: a widely targeted metabolomics study
.
J Diabetes Res
.
2020
;
2020
:
3049098
.
87.
Pena
MJ
,
Lambers Heerspink
HJ
,
Hellemons
ME
,
Friedrich
T
,
Dallmann
G
,
Lajer
M
, et al
.
Urine and plasma metabolites predict the development of diabetic nephropathy in individuals with Type 2 diabetes mellitus
.
Diabet Med
.
2014
;
31
(
9
):
1138
47
.
88.
Chen
F
,
Kantagowit
P
,
Nopsopon
T
,
Chuklin
A
,
Pongpirul
K
.
Prediction and diagnosis of chronic kidney disease development and progression using machine-learning: protocol for a systematic review and meta-analysis of reporting standards and model performance
.
PLoS One
.
2023
;
18
(
2
):
e0278729
.
89.
Davies
R
.
The metabolomic quest for a biomarker in chronic kidney disease
.
Clin Kidney J
.
2018
;
11
(
5
):
694
703
.
90.
Kalim
S
,
Rhee
EP
.
An overview of renal metabolomics
.
Kidney Int
.
2017
;
91
(
1
):
61
9
. .
91.
Noh
SA
,
Kim
SM
,
Park
SH
,
Kim
DJ
,
Lee
JW
,
Kim
YG
, et al
.
Alterations in lipid profile of the aging kidney identified by MALDI imaging mass spectrometry
.
J Proteome Res
.
2019
;
18
(
7
):
2803
12
.
92.
Dahabiyeh
LA
,
Nimer
RM
,
Sumaily
KM
,
Alabdaljabar
MS
,
Jacob
M
,
Sabi
EM
, et al
.
Metabolomics profiling distinctively identified end-stage renal disease patients from chronic kidney disease patients
.
Sci Rep
.
2023
;
13
(
1
):
6161
.
93.
Mitrofanova
A
,
Merscher
S
,
Fornoni
A
.
Kidney lipid dysmetabolism and lipid droplet accumulation in chronic kidney disease
.
Nat Rev Nephrol
.
2023
;
19
(
10
):
629
45
.
94.
Wishart
DS
,
Guo
A
,
Oler
E
,
Wang
F
,
Anjum
A
,
Peters
H
, et al
.
HMDB 5.0: the human metabolome database for 2022
.
Nucleic Acids Res
.
2022
;
50
(
D1
):
D622
31
.
95.
Whiley
L
,
Nye
LC
,
Grant
I
,
Andreas
N
,
Chappell
KE
,
Sarafian
MH
, et al
.
Ultrahigh-Performance liquid chromatography tandem mass spectrometry with electrospray ionization quantification of tryptophan metabolites and markers of gut health in serum and plasma-application to clinical and epidemiology cohorts
.
Anal Chem
.
2019
;
91
(
8
):
5207
16
.
96.
Smith
KW
,
Flinders
B
,
Thompson
PD
,
Cruickshank
FL
,
Mackay
CL
,
Heeren
RMA
, et al
.
Spatial localization of vitamin D metabolites in mouse kidney by mass spectrometry imaging
.
ACS Omega
.
2020
;
5
(
22
):
13430
7
.
97.
Mor
A
,
Kalaska
B
,
Pawlak
D
.
Kynurenine pathway in chronic kidney disease: what’s old, what’s new, and what’s next
.
Int J Tryptophan Res
.
2020
;
13
:
1178646920954882
.
98.
Hu
DY
,
Wu
MY
,
Chen
GQ
,
Deng
BQ
,
Yu
HB
,
Huang
J
, et al
.
Metabolomics analysis of human plasma reveals decreased production of trimethylamine N-oxide retards the progression of chronic kidney disease
.
Br J Pharmacol
.
2022
;
179
(
17
):
4344
59
.
99.
Velenosi
TJ
,
Thomson
BKA
,
Tonial
NC
,
RaoPeters
AAE
,
Mio
MA
,
Lajoie
GA
, et al
.
Untargeted metabolomics reveals N, N, N-trimethyl-L-alanyl-L-proline betaine (TMAP) as a novel biomarker of kidney function
.
Sci Rep
.
2019
;
9
(
1
):
6831
.
100.
Shimma
S
.
Mass spectrometry imaging
.
Mass Spectrom
.
2022
;
11
(
1
):
A0102
.
101.
Zhang
J
,
Fuhrer
T
,
Ye
H
,
Kwan
B
,
Montemayor
D
,
Tumova
J
, et al
.
High-throughput metabolomics and diabetic kidney disease progression: evidence from the chronic renal insufficiency (CRIC) study
.
Am J Nephrol
.
2022
;
53
(
2–3
):
215
25
.
102.
Rietjens
RGJ
,
Wang
G
,
van der Velden
AIM
,
Koudijs
A
,
Avramut
MC
,
Kooijman
S
, et al
.
Phosphatidylinositol metabolism of the renal proximal tubule S3 segment is disturbed in response to diabetes
.
Sci Rep
.
2023
;
13
(
1
):
6261
.
103.
Wang
G
,
Heijs
B
,
Kostidis
S
,
Mahfouz
A
,
Rietjens
RGJ
,
Bijkerk
R
, et al
.
Analyzing cell-type-specific dynamics of metabolism in kidney repair
.
Nat Metab
.
2022
;
4
(
9
):
1109
18
.
104.
Sugimoto
M
,
Kawakami
M
,
Robert
M
,
Soga
T
,
Tomita
M
.
Bioinformatics tools for mass spectroscopy-based metabolomic data processing and analysis
.
Curr Bioinform
.
2012
;
7
(
1
):
96
108
.
105.
Xia
J
,
Wishart
DS
.
MSEA: a web-based tool to identify biologically meaningful patterns in quantitative metabolomic data
.
Nucleic Acids Res
.
2010
;
38
(
Web Server issue
):
W71
7
.
106.
Chen
Y
,
Li
EM
,
Xu
LY
.
Guide to metabolomics analysis: a bioinformatics workflow
.
Metabolites
.
2022
;
12
(
4
):
357
.
107.
Fernie
AR
,
Aharoni
A
,
Willmitzer
L
,
Stitt
M
,
Tohge
T
,
Kopka
J
, et al
.
Recommendations for reporting metabolite data
.
Plant Cell
.
2011
;
23
(
7
):
2477
82
.
108.
Jain
S
,
Pei
L
,
Spraggins
JM
,
Angelo
M
,
Carson
JP
,
Gehlenborg
N
, et al
.
Author correction: advances and prospects for the human BioMolecular atlas program (HuBMAP)
.
Nat Cel Biol
.
2024
;
25
(
8
):
1089
100
.
109.
Hansen
J
,
Sealfon
R
,
Menon
R
,
Eadon
MT
,
Lake
BB
,
Steck
B
, et al
.
A reference tissue atlas for the human kidney
.
Sci Adv
.
2022
;
8
(
23
):
eabn4965
.
110.
Vicari
M
,
Mirzazadeh
R
,
Nilsson
A
,
Shariatgorji
R
,
Bjarterot
P
,
Larsson
L
, et al
.
Spatial multimodal analysis of transcriptomes and metabolomes in tissues
.
Nat Biotechnol
.
2023
.
111.
Bourceau
P
,
Geier
B
,
Suerdieck
V
,
Bien
T
,
Soltwisch
J
,
Dreisewerd
K
, et al
.
Visualization of metabolites and microbes at high spatial resolution using MALDI mass spectrometry imaging and in situ fluorescence labeling
.
Nat Protoc
.
2023
;
18
(
10
):
3050
79
.
112.
Drexler
Y
,
Fornoni
A
.
Adenine crosses the biomarker bridge: from’omics to treatment in diabetic kidney disease
.
J Clin Invest
.
2023
;
133
(
20
):
e174015
.