1
|
Hu Y, Ni X, Chen Q, Qu Y, Chen K, Zhu G, Zhang M, Xu N, Bai X, Wang J, Ma Y, Luo Q, Cai K. Predicting diabetic kidney disease with serum metabolomics and gut microbiota. Sci Rep 2025; 15:12179. [PMID: 40204798 PMCID: PMC11982385 DOI: 10.1038/s41598-025-91281-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Accepted: 02/19/2025] [Indexed: 04/11/2025] Open
Abstract
This study aims to identify biomarkers for reliably predicting diabetic kidney disease (DKD), systematically characterize serum metabolites and gut microbiota in DKD patients, and investigate the correlation between differential serum metabolites and gut microbiota. From September 2021 to January 2023, 90 subjects were recruited: 30 with DKD, 30 with type 2 diabetes mellitus (T2DM), and 30 normal controls (NCs). Serum metabolites, including 180 different metabolites, were analyzed using untargeted metabolomics UPLC-MS/MS, and gut microbiota were assessed via 16S rRNA sequencing. Differential metabolites were identified through univariate (t-test or Mann-Whitney U-test, P < 0.05) and multivariate analyses (OPLS-DA model, VIP > 1, P < 0.05), followed by selection using the least absolute shrinkage and selection operator (LASSO). The selected overlapping serum metabolites, along with DKD-associated differential gut microbiota, were used to develop a logistic regression prediction model for DKD based on six markers. In the DKD group compared to the DM and NC groups, 39 and 60 differential serum metabolites were identified, respectively (VIP > 1, P < 0.01). Among these, 36 serum metabolites, including alpha-Hydroxyisobutyric acid, were significantly elevated in DKD patients compared to those with DM. Of these, 28 metabolites showed a negative correlation with estimated glomerular filtration rate (eGFR), while 29 showed a positive correlation with urine albumin creatinine ratio (UACR). Patients with DKD were further categorized into subgroups (DKD middle and DKD early) based on eGFR (eGFR < 90 ml/min/1.73m2, eGFR ≥ 90 ml/min/1.73m2), revealing 23 differential metabolites. Dysbiosis of the gut microbiota was evident in DKD patients, with lower relative abundances of g_Prevotella and g_Faecalibacterium compared to the DM and NC groups. Subgroup analysis indicated decreased relative abundances of g_Prevotella and g_Faecalibacterium in the DKD middle group, along with a decrease in g_Klebsiella compared to the DKD early group, which correlated positively with DKD patients' eGFR. There were 11 common metabolites among the three groups of differential metabolites. Among these, three serum metabolites-imidazolepropionic acid, adipoylcarnitine, and 1-methylhistidine-were identified as predictive serum metabolic markers. Disease prediction models (logistic regression models) were constructed based on these three metabolites combined with three genera of bacteria. These models demonstrated strong discriminatory power for diagnosing patients with DKD compared to patients with DM (area under the receiver operating characteristic curve (AUROC) = 0.939 and precision-recall curve (AUPR) = 0.940). The models also effectively discriminated between patients with DKD and NCs (0.976, 0.973). This study revealed distinctive serum metabolites and gut microbiota in patients with DKD. It demonstrated the potential utility of three specific serum metabolites and three genera of bacteria in diagnosing patients with DKD and assessing their renal dysfunction.
Collapse
Affiliation(s)
- Yuyun Hu
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou, 310000, Zhejiang, China
- Department of Nephrology, Ningbo No.2 Hospital, Ningbo, 315010, China
| | - Xue Ni
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou, 310000, Zhejiang, China
- Department of Nephrology, Ningbo No.2 Hospital, Ningbo, 315010, China
| | - Qinghuo Chen
- Department of Nephrology, Ningbo No.2 Hospital, Ningbo, 315010, China
| | - Yihui Qu
- Department of Nephrology, Ningbo No.2 Hospital, Ningbo, 315010, China
| | - Kanan Chen
- Department of Nephrology, Ningbo No.2 Hospital, Ningbo, 315010, China
| | - Gaohui Zhu
- Department of Endocrinology, Ningbo Zhenhai Hospital of Traditional Chinese Medicine, Ningbo, Zhejiang, China
| | - Minqiao Zhang
- Department of Nephrology, The First People's Hospital of Xiangshan, Ningbo, 315700, China
| | - Ningjie Xu
- Department of Nephrology, Ningbo No.2 Hospital, Ningbo, 315010, China
| | - Xu Bai
- Department of Nephrology, Ningbo No.2 Hospital, Ningbo, 315010, China
| | - Jing Wang
- Department of Nephrology, Ningbo No.2 Hospital, Ningbo, 315010, China
| | - Yanhong Ma
- Department of Oncology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qun Luo
- Department of Nephrology, Ningbo No.2 Hospital, Ningbo, 315010, China
| | - Kedan Cai
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou, 310000, Zhejiang, China.
- Department of Nephrology, Ningbo No.2 Hospital, Ningbo, 315010, China.
| |
Collapse
|
2
|
Vanholder R, Glorieux G, Argiles A, Burtey S, Cohen G, Duranton F, Koppe L, Massy ZA, Ortiz A, Masereeuw R, Stamatialis D, Jankowski J. Metabolomics to Identify Unclassified Uremic Toxins: A Comprehensive Literature Review. Kidney Med 2025; 7:100955. [PMID: 39980938 PMCID: PMC11841090 DOI: 10.1016/j.xkme.2024.100955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/22/2025] Open
Abstract
A comprehensive review of known uremic retention molecules goes back to more than 10 years ago and did not consider metabolomic analyses. The present analysis searches for as of yet unclassified solutes retained in chronic kidney disease (CKD) by analyzing metabolites associated with relevant outcomes of CKD. This untargeted metabolomics-based approach is compared with a conventional targeted literature search. For the selected molecules, the literature was screened for arguments regarding toxic (harmful), beneficial, or neutral effects in experimental or clinical studies. Findings were independently crosschecked. In total, 103 molecules were selected. No literature on any effect was found for 55 substances, 3 molecules had no significant effect, and 13 others showed beneficial effects. For the remaining 32 compounds, we found at least one report of a toxic effect. Whereas 62.5% of the compounds with at least one study on a toxic effect was retrieved via the bottom-up approach, 69.2% of the substances originating from metabolomics-based approaches showed a beneficial effect. Our results suggest that untargeted metabolomics offer a more balanced view of uremic retention than the targeted approaches, with higher chances of revealing the beneficial potential of some of the metabolites.
Collapse
Affiliation(s)
- Raymond Vanholder
- Nephrology Section, Department of Internal Medicine and Pediatrics, Ghent University Hospital, Ghent, Belgium
| | - Griet Glorieux
- Nephrology Section, Department of Internal Medicine and Pediatrics, Ghent University Hospital, Ghent, Belgium
| | - Angel Argiles
- RD Néphrologie, Montpellier, France
- Néphrologie Dialyse Saint Guilhem, Sète, France
| | - Stéphane Burtey
- C2VN, Aix-Marseille Université, INSERM, INRAE, Marseille, France
| | - Gerald Cohen
- Department of Nephrology and Dialysis, Medical University of Vienna, Vienna, Austria
| | | | - Laetitia Koppe
- Department of Nephrology, Hospices Civils de Lyon, Centre Hospitalier Lyon Sud, Université de Lyon, Lyon, France
- CarMeN lab, INSERM U1060, Université Claude Bernard Lyon 1, France
| | - Ziad A. Massy
- Inserm Unit 1018, Team 5, CESP, Hôpital Paul Brousse, Paris-Sud University (UPS), Villejuif, France
- Versailles Saint-Quentin-en-Yvelines University (Paris-Ile-de-France-Ouest University, UVSQ), Villejuif, France
- Department of Nephrology, Ambroise Paré University Hospital, APHP, Boulogne-Billancourt/Paris, France
| | - Alberto Ortiz
- Department of Nephrology and Hypertension, IIS-Fundacion Jimenez Diaz UAM, Madrid, Spain
- RICORS2040, Madrid, Spain
| | - Rosalinde Masereeuw
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Dimitrios Stamatialis
- Advanced Organ Bioengineering and Therapeutics, Technical Medical Centre, Faculty of Science and Technology, University of Twente, Enschede, The Netherlands
| | - Joachim Jankowski
- Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, Aachen, Germany
- Aachen-Maastricht Institute for CardioRenal Disease (AMICARE), RWTH Aachen University, Aachen, Germany
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| |
Collapse
|
3
|
Pandey S. Metabolomics Characterization of Disease Markers in Diabetes and Its Associated Pathologies. Metab Syndr Relat Disord 2024; 22:499-509. [PMID: 38778629 DOI: 10.1089/met.2024.0038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/25/2024] Open
Abstract
With the change in lifestyle of people, there has been a considerable increase in diabetes, which brings with it certain follow-up pathological conditions, which lead to a substantial medical burden. Identifying biomarkers that aid in screening, diagnosis, and prognosis of diabetes and its associated pathologies would help better patient management and facilitate a personalized treatment approach for prevention and treatment. With the advancement in techniques and technologies, metabolomics has emerged as an omics approach capable of large-scale high throughput data analysis and identifying and quantifying metabolites that provide an insight into the underlying mechanism of the disease and its progression. Diabetes and metabolomics keywords were searched in correspondence with the assigned keywords, including kidney, cardiovascular diseases and critical illness from PubMed and Scopus, from its inception to Dec 2023. The relevant studies from this search were extracted and included in the study. This review is focused on the biomarkers identified in diabetes, diabetic kidney disease, diabetes-related development of CVD, and its role in critical illness.
Collapse
Affiliation(s)
- Swarnima Pandey
- School of Pharmacy, Department of Pharmaceutical Sciences, University of Maryland, Baltimore, Maryland, USA
| |
Collapse
|
4
|
Luo Y, Zhang W, Qin G. Metabolomics in diabetic nephropathy: Unveiling novel biomarkers for diagnosis (Review). Mol Med Rep 2024; 30:156. [PMID: 38963028 PMCID: PMC11258608 DOI: 10.3892/mmr.2024.13280] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 06/21/2024] [Indexed: 07/05/2024] Open
Abstract
Diabetic nephropathy (DN) also known as diabetic kidney disease, is a major microvascular complication of diabetes and a leading cause of end‑stage renal disease (ESRD), which affects the morbidity and mortality of patients with diabetes. Despite advancements in diabetes care, current diagnostic methods, such as the determination of albuminuria and the estimated glomerular filtration rate, are limited in sensitivity and specificity, often only identifying kidney damage after considerable morphological changes. The present review discusses the potential of metabolomics as an approach for the early detection and management of DN. Metabolomics is the study of metabolites, the small molecules produced by cellular processes, and may provide a more sensitive and specific diagnostic tool compared with traditional methods. For the purposes of this review, a systematic search was conducted on PubMed and Google Scholar for recent human studies published between 2011 and 2023 that used metabolomics in the diagnosis of DN. Metabolomics has demonstrated potential in identifying metabolic biomarkers specific to DN. The ability to detect a broad spectrum of metabolites with high sensitivity and specificity may allow for earlier diagnosis and better management of patients with DN, potentially reducing the progression to ESRD. Furthermore, metabolomics pathway analysis assesses the pathophysiological mechanisms underlying DN. On the whole, metabolomics is a potential tool in the diagnosis and management of DN. By providing a more in‑depth understanding of metabolic alterations associated with DN, metabolomics could significantly improve early detection, enable timely interventions and reduce the healthcare burdens associated with this condition.
Collapse
Affiliation(s)
- Yuanyuan Luo
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Wei Zhang
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Guijun Qin
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| |
Collapse
|
5
|
Tabata S, Yamashita Y, Inai Y, Morita S, Kosako H, Takagi T, Shide K, Manabe S, Matsuoka TA, Shimoda K, Sonoki T, Ihara Y, Tamura S. C-Mannosyl tryptophan is a novel biomarker for thrombocytosis of myeloproliferative neoplasms. Sci Rep 2024; 14:18858. [PMID: 39143127 PMCID: PMC11324734 DOI: 10.1038/s41598-024-69496-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 08/05/2024] [Indexed: 08/16/2024] Open
Abstract
C-Mannosyl tryptophan (CMW), a unique glycosylated amino acid, is considered to be produced by degradation of C-mannosylated proteins in living organism. Although protein C-mannosylation is involved in the folding and secretion of substrate proteins, the pathophysiological function in the hematological system is still unclear. This study aimed to assess CMW in the human hematological disorders. The serum CMW levels of 94 healthy Japanese workers were quantified using hydrophilic interaction liquid chromatography. Platelet count was positively correlated with serum CMW levels. The clinical significance of CMW in thrombocytosis of myeloproliferative neoplasms (T-MPN) including essential thrombocythemia (ET) were investigated. The serum CMW levels of the 34 patients with T-MPN who presented with thrombocytosis were significantly higher than those of the 52 patients with control who had other hematological disorders. In patients with T-MPN, serum CMW levels were inversely correlated with anemia, which was related to myelofibrosis (MF). Bone marrow biopsy samples were obtained from 18 patients with ET, and serum CMW levels were simultaneously measured. Twelve patients with bone marrow fibrosis had significantly higher CMW levels than 6 patients without bone marrow fibrosis. Collectively, these results suggested that CMW could be a novel biomarker to predict MF progression in T-MPN.
Collapse
Affiliation(s)
- Shotaro Tabata
- Department of Hematology/Oncology, Wakayama Medical University, Wakayama, Japan
| | - Yusuke Yamashita
- Department of Hematology/Oncology, Wakayama Medical University, Wakayama, Japan
| | - Yoko Inai
- Department of Biochemistry, Wakayama Medical University, Wakayama, Japan
| | - Shuhei Morita
- The First Department of Internal Medicine, Wakayama Medical University, Wakayama, Japan.
| | - Hideki Kosako
- Department of Hematology/Oncology, Wakayama Medical University, Wakayama, Japan
| | - Tomoyuki Takagi
- The First Department of Internal Medicine, Wakayama Medical University, Wakayama, Japan
- Wakayama City Medical Association Seijinbyo Center, Wakayama, Japan
| | - Kotaro Shide
- Division of Hematology, Diabetes, and Endocrinology, Department of Internal Medicine, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
| | - Shino Manabe
- School of Pharmacy and Pharmaceutical Sciences, Hoshi University, Tokyo, Japan
- Research Center for Pharmaceutical Development, Graduate School of Pharmaceutical Science & Faculty of Pharmaceutical Sciences, Tohoku University, Miyagi, Japan
| | - Taka-Aki Matsuoka
- The First Department of Internal Medicine, Wakayama Medical University, Wakayama, Japan
| | - Kazuya Shimoda
- Division of Hematology, Diabetes, and Endocrinology, Department of Internal Medicine, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
| | - Takashi Sonoki
- Department of Hematology/Oncology, Wakayama Medical University, Wakayama, Japan
| | - Yoshito Ihara
- Department of Biochemistry, Wakayama Medical University, Wakayama, Japan.
| | - Shinobu Tamura
- Department of Hematology/Oncology, Wakayama Medical University, Wakayama, Japan.
- Department of Emergency and Critical Care Medicine, Wakayama Medical University, Wakayama, Japan.
| |
Collapse
|
6
|
Wong PL, Zolkeflee NKZ, Ramli NS, Tan CP, Azlan A, Abas F. Acute toxicity profiling of medicinal herb Ardisia elliptica leaf extract by conventional evaluations and proton nuclear magnetic resonance (NMR) metabolomics. J Tradit Complement Med 2024; 14:456-466. [PMID: 39035686 PMCID: PMC11259702 DOI: 10.1016/j.jtcme.2024.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 02/08/2024] [Accepted: 02/20/2024] [Indexed: 07/23/2024] Open
Abstract
Background and aim Interest in the safety of herbal medicine is growing rapidly regarding knowledge and challenges in natural products. Hence, this study aimed to reveal the toxicological profile of Ardisia elliptica, a traditional medicinal plant used in the treatment of various illnesses. Experimental procedure Acute toxicity study was performed on female and male Sprague Dawley rats with a single oral administration of 2000 mg/kg BW of 70% ethanolic A. elliptica leaf extract, using a combination of conventional investigations and 1H-NMR-based metabolomics approaches. Results Physical, hematological, biochemical, and histopathological assessments demonstrated the usual rat profile, with no mortality and delayed toxicity 14 days after administration. 1H NMR serum metabolomics depicted similar metabolites between normal and treated groups. Nevertheless, 1H NMR of urinary metabolomics revealed perturbation in carbohydrate, amino acid, and energy metabolism within 24h after extract administration, while no accumulation of toxic biomarkers in the collected biological fluids on Day 14. A minor gender-based difference revealed the influence of sex hormones and different energy expenditure on response to extract treatment. Conclusion This study suggested that 2000 mg/kg BW of 70% ethanolic A. elliptica leaf extract is considered as safe for consumption and offered a comprehensive overview of the response of physiological and metabolic aspects applicable to food and herbal product development.
Collapse
Affiliation(s)
- Pei Lou Wong
- Department of Food Science, Faculty of Food Science and Technology, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia
| | - Nur Khaleeda Zulaikha Zolkeflee
- Natural Medicines and Products Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia
| | - Nurul Shazini Ramli
- Department of Food Science, Faculty of Food Science and Technology, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia
| | - Chin Ping Tan
- Department of Food Technology, Faculty of Food Science and Technology, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia
| | - Azrina Azlan
- Department of Nutrition and Dietetics, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | - Faridah Abas
- Department of Food Science, Faculty of Food Science and Technology, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia
- Natural Medicines and Products Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia
| |
Collapse
|
7
|
Jalan A, Jayasree PJ, Karemore P, Narayan KP, Khandelia P. Decoding the 'Fifth' Nucleotide: Impact of RNA Pseudouridylation on Gene Expression and Human Disease. Mol Biotechnol 2024; 66:1581-1598. [PMID: 37341888 DOI: 10.1007/s12033-023-00792-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Accepted: 06/08/2023] [Indexed: 06/22/2023]
Abstract
Cellular RNAs, both coding and noncoding are adorned by > 100 chemical modifications, which impact various facets of RNA metabolism and gene expression. Very often derailments in these modifications are associated with a plethora of human diseases. One of the most oldest of such modification is pseudouridylation of RNA, wherein uridine is converted to a pseudouridine (Ψ) via an isomerization reaction. When discovered, Ψ was referred to as the 'fifth nucleotide' and is chemically distinct from uridine and any other known nucleotides. Experimental evidence accumulated over the past six decades, coupled together with the recent technological advances in pseudouridine detection, suggest the presence of pseudouridine on messenger RNA, as well as on diverse classes of non-coding RNA in human cells. RNA pseudouridylation has widespread effects on cellular RNA metabolism and gene expression, primarily via stabilizing RNA conformations and destabilizing interactions with RNA-binding proteins. However, much remains to be understood about the RNA targets and their recognition by the pseudouridylation machinery, the regulation of RNA pseudouridylation, and its crosstalk with other RNA modifications and gene regulatory processes. In this review, we summarize the mechanism and molecular machinery involved in depositing pseudouridine on target RNAs, molecular functions of RNA pseudouridylation, tools to detect pseudouridines, the role of RNA pseudouridylation in human diseases like cancer, and finally, the potential of pseudouridine to serve as a biomarker and as an attractive therapeutic target.
Collapse
Affiliation(s)
- Abhishek Jalan
- Department of Biological Sciences, Birla Institute of Technology and Science, Pilani - Hyderabad Campus, Jawahar Nagar, Kapra Mandal, Medchal-Malkajgiri District, Telangana, 500078, India
| | - P J Jayasree
- Department of Biological Sciences, Birla Institute of Technology and Science, Pilani - Hyderabad Campus, Jawahar Nagar, Kapra Mandal, Medchal-Malkajgiri District, Telangana, 500078, India
| | - Pragati Karemore
- Department of Biological Sciences, Birla Institute of Technology and Science, Pilani - Hyderabad Campus, Jawahar Nagar, Kapra Mandal, Medchal-Malkajgiri District, Telangana, 500078, India
| | - Kumar Pranav Narayan
- Department of Biological Sciences, Birla Institute of Technology and Science, Pilani - Hyderabad Campus, Jawahar Nagar, Kapra Mandal, Medchal-Malkajgiri District, Telangana, 500078, India
| | - Piyush Khandelia
- Department of Biological Sciences, Birla Institute of Technology and Science, Pilani - Hyderabad Campus, Jawahar Nagar, Kapra Mandal, Medchal-Malkajgiri District, Telangana, 500078, India.
| |
Collapse
|
8
|
Schlosser P, Surapaneni AL, Borisov O, Schmidt IM, Zhou L, Anderson A, Deo R, Dubin R, Ganz P, He J, Kimmel PL, Li H, Nelson RG, Porter AC, Rahman M, Rincon-Choles H, Shah V, Unruh ML, Vasan RS, Zheng Z, Feldman HI, Waikar SS, Köttgen A, Rhee EP, Coresh J, Grams ME. Association of Integrated Proteomic and Metabolomic Modules with Risk of Kidney Disease Progression. J Am Soc Nephrol 2024; 35:923-935. [PMID: 38640019 PMCID: PMC11230725 DOI: 10.1681/asn.0000000000000343] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 04/01/2024] [Indexed: 04/21/2024] Open
Abstract
Key Points Integrated analysis of proteome and metabolome identifies modules associated with CKD progression and kidney failure. Ephrin transmembrane proteins and podocyte-expressed CRIM1 and NPNT emerged as central components and warrant experimental and clinical investigation. Background Proteins and metabolites play crucial roles in various biological functions and are frequently interconnected through enzymatic or transport processes. Methods We present an integrated analysis of 4091 proteins and 630 metabolites in the Chronic Renal Insufficiency Cohort study (N =1708; average follow-up for kidney failure, 9.5 years, with 537 events). Proteins and metabolites were integrated using an unsupervised clustering method, and we assessed associations between clusters and CKD progression and kidney failure using Cox proportional hazards models. Analyses were adjusted for demographics and risk factors, including the eGFR and urine protein–creatinine ratio. Associations were identified in a discovery sample (random two thirds, n =1139) and then evaluated in a replication sample (one third, n =569). Results We identified 139 modules of correlated proteins and metabolites, which were represented by their principal components. Modules and principal component loadings were projected onto the replication sample, which demonstrated a consistent network structure. Two modules, representing a total of 236 proteins and 82 metabolites, were robustly associated with both CKD progression and kidney failure in both discovery and validation samples. Using gene set enrichment, several transmembrane-related terms were identified as overrepresented in these modules. Transmembrane–ephrin receptor activity displayed the largest odds (odds ratio=13.2, P value = 5.5×10−5). A module containing CRIM1 and NPNT expressed in podocytes demonstrated particularly strong associations with kidney failure (P value = 2.6×10−5). Conclusions This study demonstrates that integration of the proteome and metabolome can identify functions of pathophysiologic importance in kidney disease.
Collapse
Affiliation(s)
- Pascal Schlosser
- Department of Epidemiology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland
- Institute of Genetic Epidemiology, Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Centre for Integrative Biological Signalling Studies (CIBSS), University of Freiburg, Freiburg, Germany
| | - Aditya L. Surapaneni
- Department of Epidemiology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland
- Division of Precision Medicine, Department of Medicine, NYU Langone Health, New York, New York
| | - Oleg Borisov
- Institute of Genetic Epidemiology, Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Insa M. Schmidt
- Section of Nephrology, Department of Medicine, Boston Medical Center and Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts
| | - Linda Zhou
- Department of Epidemiology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland
| | - Amanda Anderson
- Department of Epidemiology, Tulane University, New Orleans, Louisiana
| | - Rajat Deo
- Division of Cardiovascular Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Ruth Dubin
- Division of Nephrology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Peter Ganz
- Division of Cardiology, University of California, San Francisco, San Francisco, California
| | - Jiang He
- Department of Epidemiology, Tulane University, New Orleans, Louisiana
| | - Paul L. Kimmel
- Division of Kidney, Urologic, and Hematologic Diseases, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland
| | - Hongzhe Li
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Robert G. Nelson
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Phoenix, Arizona
- Research Division, Joslin Diabetes Center, Boston, Massachusetts
| | - Anna C. Porter
- Renal Service, Wellington Regional Hospital, Wellington, New Zealand
| | - Mahboob Rahman
- Department of Kidney Medicine, Cleveland Clinic Foundation, Cleveland, Ohio
| | | | - Vallabh Shah
- Department of Internal Medicine, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Mark L. Unruh
- Department of Internal Medicine, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Ramachandran S. Vasan
- University of Texas Health Sciences Center, San Antonio, Texas
- Section of Preventive Medicine and Epidemiology, Department of Medicine, Boston Medical Center and Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts
| | - Zihe Zheng
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Harold I. Feldman
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Sushrut S. Waikar
- Section of Nephrology, Department of Medicine, Boston Medical Center and Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts
| | - Anna Köttgen
- Department of Epidemiology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland
- Institute of Genetic Epidemiology, Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Eugene P. Rhee
- Nephrology Division and Endocrine Unit, Massachusetts General Hospital, Boston, Massachusetts
| | - Josef Coresh
- Department of Epidemiology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland
- Optimal Aging Institute, Departments of Population Health and Medicine, NYU Grossman School of Medicine, New York, New York
| | - Morgan E. Grams
- Department of Epidemiology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland
- Division of Precision Medicine, Department of Medicine, NYU Langone Health, New York, New York
| |
Collapse
|
9
|
Sorokoumova AA, Seryapina AA, Polityko YK, Yanshole LV, Tsentalovich YP, Gilinsky МА, Markel АL. Urine metabolic profile in rats with arterial hypertension of different genesis. Vavilovskii Zhurnal Genet Selektsii 2024; 28:299-307. [PMID: 38952704 PMCID: PMC11214897 DOI: 10.18699/vjgb-24-34] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 12/29/2023] [Accepted: 01/30/2024] [Indexed: 07/03/2024] Open
Abstract
The diversity of pathogenetic mechanisms underlying arterial hypertension leads to the necessity to devise a personalized approach to the diagnosis and treatment of the disease. Metabolomics is one of the promising methods for personalized medicine, as it provides a comprehensive understanding of the physiological processes occurring in the body. The metabolome is a set of low-molecular substances available for detection in a sample and representing intermediate and final products of cell metabolism. Changes in the content and ratio of metabolites in the sample mark the corresponding pathogenetic mechanisms by highlighting them, which is especially important for such a multifactorial disease as arterial hypertension. To identify metabolomic markers for hypertensive conditions of different origins, three forms of arterial hypertension (AH) were studied: rats with hereditary AH (ISIAH rat strain); rats with AH induced by L-NAME administration (a model of endothelial dysfunction with impaired NO production); rats with AH caused by the administration of deoxycorticosterone in combination with salt loading (hormone-dependent form - DOCA-salt AH). WAG rats were used as normotensive controls. 24-hour urine samples were collected from all animals and analyzed by quantitative NMR spectroscopy for metabolic profiling. Then, potential metabolomic markers for the studied forms of hypertensive conditions were identified using multivariate statistics. Analysis of the data obtained showed that hereditary stress-induced arterial hypertension in ISIAH rats was characterized by a decrease in the following urine metabolites: nicotinamide and 1-methylnicotinamide (markers of inflammatory processes), N- acetylglutamate (nitric oxide cycle), isobutyrate and methyl acetoacetate (gut microbiota). Pharmacologically induced forms of hypertension (the L-NAME and DOCA+NaCl groups) do not share metabolomic markers with hereditary AH. They are differentiated by N,N-dimethylglycine (both groups), choline (the L-NAME group) and 1-methylnicotinamide (the group of rats with DOCA-salt hypertension).
Collapse
Affiliation(s)
- A A Sorokoumova
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - A A Seryapina
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Yu K Polityko
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia Scientific Research Institute of Neurosciences and Medicine, Novosibirsk, Russia
| | - L V Yanshole
- International Tomography Center of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Yu P Tsentalovich
- International Tomography Center of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - М А Gilinsky
- Scientific Research Institute of Neurosciences and Medicine, Novosibirsk, Russia
| | - А L Markel
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia Novosibirsk State University, Novosibirsk, Russia
| |
Collapse
|
10
|
Mathew AV, Kayampilly P, Byun J, Nair V, Afshinnia F, Chai B, Brosius FC, Kretzler M, Pennathur S. Tubular dysfunction impairs renal excretion of pseudouridine in diabetic kidney disease. Am J Physiol Renal Physiol 2024; 326:F30-F38. [PMID: 37916286 PMCID: PMC11194048 DOI: 10.1152/ajprenal.00252.2022] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 10/02/2023] [Accepted: 10/26/2023] [Indexed: 11/03/2023] Open
Abstract
Plasma nucleosides-pseudouridine (PU) and N2N2-dimethyl guanosine (DMG) predict the progression of type 2 diabetic kidney disease (DKD) to end-stage renal disease, but the mechanisms underlying this relationship are not well understood. We used a well-characterized model of type 2 diabetes (db/db mice) and control nondiabetic mice (db/m mice) to characterize the production and excretion of PU and DMG levels using liquid chromatography-mass spectrometry. The fractional excretion of PU and DMG was decreased in db/db mice compared with control mice at 24 wk before any changes to renal function. We then examined the dynamic changes in nucleoside metabolism using in vivo metabolic flux analysis with the injection of labeled nucleoside precursors. Metabolic flux analysis revealed significant decreases in the ratio of urine-to-plasma labeling of PU and DMG in db/db mice compared with db/m mice, indicating significant tubular dysfunction in diabetic kidney disease. We observed that the gene and protein expression of the renal tubular transporters involved with nucleoside transport in diabetic kidneys in mice and humans was reduced. In conclusion, this study strongly suggests that tubular handling of nucleosides is altered in early DKD, in part explaining the association of PU and DMG with human DKD progression observed in previous studies.NEW & NOTEWORTHY Tubular dysfunction explains the association between the nucleosides pseudouridine and N2N2-dimethyl guanosine and diabetic kidney disease.
Collapse
Affiliation(s)
- Anna V Mathew
- Division of Nephrology, Department of Medicine, University of Michigan, Ann Arbor, Michigan, United States
| | - Pradeep Kayampilly
- Division of Nephrology, Department of Medicine, University of Michigan, Ann Arbor, Michigan, United States
| | - Jaeman Byun
- Division of Nephrology, Department of Medicine, University of Michigan, Ann Arbor, Michigan, United States
| | - Viji Nair
- Division of Nephrology, Department of Medicine, University of Michigan, Ann Arbor, Michigan, United States
| | - Farsad Afshinnia
- Division of Nephrology, Department of Medicine, University of Michigan, Ann Arbor, Michigan, United States
| | - Biaoxin Chai
- Division of Nephrology, Department of Medicine, University of Michigan, Ann Arbor, Michigan, United States
| | - Frank C Brosius
- Division of Nephrology, Department of Medicine, University of Michigan, Ann Arbor, Michigan, United States
- Department of Medicine, University of Arizona, Tucson, Arizona, United States
| | - Matthias Kretzler
- Division of Nephrology, Department of Medicine, University of Michigan, Ann Arbor, Michigan, United States
| | - Subramaniam Pennathur
- Division of Nephrology, Department of Medicine, University of Michigan, Ann Arbor, Michigan, United States
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, United States
| |
Collapse
|
11
|
Qian F, Zhao L, Zhang D, Yu M, Zhou W, Jin J. Serum metabolomics detected by LDI-TOF-MS can be used to distinguish between diabetic patients with and without diabetic kidney disease. FEBS Open Bio 2023; 13:1844-1858. [PMID: 37525631 PMCID: PMC10549217 DOI: 10.1002/2211-5463.13683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Revised: 06/21/2023] [Accepted: 07/31/2023] [Indexed: 08/02/2023] Open
Abstract
Diabetic kidney disease (DKD) is an important cause of end-stage renal disease with changes in metabolic characteristics. The objective of this study was to study changes in serum metabolic characteristics in patients with DKD and to examine metabolite panels to distinguish DKD from diabetes with matrix-assisted laser desorption/ionization time-of-flight mass spectrometry (MALDI-TOF-MS). We recruited 40 type II diabetes mellitus (T2DM) patients with or without DKD from a single center for a cross-sectional study. Serum metabolic profiling was performed with MALDI-TOF-MS using a vertical silicon nanowire array. Differential metabolites between DKD and diabetes patients were selected, and their relevance to the clinical parameters of DKD was analyzed. We applied machine learning methods to the differential metabolite panels to distinguish DKD patients from diabetes patients. Twenty-four differential serum metabolites between DKD patients and diabetes patients were identified, which were mainly enriched in butyrate metabolism, TCA cycle, and alanine, aspartate, and glutamate metabolism. Among the metabolites, l-kynurenine was positively correlated with urinary microalbumin, urinary microalbumin/creatinine ratio (UACR), creatinine, and urea nitrogen content. l-Serine, pimelic acid, 5-methylfuran-2-carboxylic acid, 4-methylbenzaldehyde, and dihydrouracil were associated with the estimated glomerular filtration rate (eGFR). The panel of differential metabolites could be used to distinguish between DKD and diabetes patients with an AUC value reaching 0.9899-0.9949. Among the differential metabolites, l-kynurenine was related to the progression of DKD. The differential metabolites exhibited excellent performance at distinguishing between DKD and diabetes. This study provides a novel direction for metabolomics-based clinical detection of DKD.
Collapse
Affiliation(s)
- Fengmei Qian
- The Second School of Clinical MedicineZhejiang Chinese Medical UniversityHangzhouChina
| | - Li Zhao
- Department of Nephrology, Urology & Nephrology CenterZhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College)China
| | - Di Zhang
- Department of Nephrology, Urology & Nephrology CenterZhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College)China
| | - Mengjie Yu
- Department of Nephrology, Urology & Nephrology CenterZhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College)China
| | - Wei Zhou
- Department of NephrologyThe First People's Hospital of Hangzhou Lin'an District, Affiliated Lin'an People's Hospital, Hangzhou Medical CollegeChina
| | - Juan Jin
- Department of NephrologyThe First People's Hospital of Hangzhou Lin'an District, Affiliated Lin'an People's Hospital, Hangzhou Medical CollegeChina
| |
Collapse
|
12
|
Li C, Gao L, Lv C, Li Z, Fan S, Liu X, Rong X, Huang Y, Liu J. Active role of amino acid metabolism in early diagnosis and treatment of diabetic kidney disease. Front Nutr 2023; 10:1239838. [PMID: 37781128 PMCID: PMC10539689 DOI: 10.3389/fnut.2023.1239838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 08/23/2023] [Indexed: 10/03/2023] Open
Abstract
Diabetic Kidney Disease (DKD) is one of the significant microvascular consequences of type 2 diabetes mellitus with a complex etiology and protracted course. In the early stages of DKD, the majority of patients experience an insidious onset and few overt clinical symptoms and indicators, but they are prone to develop end-stage renal disease in the later stage, which is life-threatening. The abnormal amino acid metabolism is tightly associated with the development of DKD, which involves several pathological processes such as oxidative stress, inflammatory response, and immune response and is also closely related to autophagy, mitochondrial dysfunction, and iron death. With a focus on taurine, branched-chain amino acids (BCAAs) and glutamine, we explored the biological effects of various amino acid mechanisms linked to DKD, the impact of amino acid metabolism in the early diagnosis of DKD, and the role of amino acid metabolism in treating DKD, to offer fresh objectives and guidelines for later early detection and DKD therapy.
Collapse
Affiliation(s)
- Chenming Li
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Clinical Pharmacology Department, Second Affiliated Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Lidong Gao
- College of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin Key Laboratory of Modern Chinese Medicine Theory of Innovation and Application, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Chunxiao Lv
- Clinical Pharmacology Department, Second Affiliated Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Ziqiang Li
- Clinical Pharmacology Department, Second Affiliated Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Shanshan Fan
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Clinical Pharmacology Department, Second Affiliated Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xinyue Liu
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Clinical Pharmacology Department, Second Affiliated Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xinyi Rong
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Clinical Pharmacology Department, Second Affiliated Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yuhong Huang
- Clinical Pharmacology Department, Second Affiliated Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Jia Liu
- Clinical Pharmacology Department, Second Affiliated Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
13
|
Shi C, Wan Y, He A, Wu X, Shen X, Zhu X, Yang J, Zhou Y. Urinary metabolites associate with the presence of diabetic kidney disease in type 2 diabetes and mediate the effect of inflammation on kidney complication. Acta Diabetol 2023; 60:1199-1207. [PMID: 37184672 PMCID: PMC10359369 DOI: 10.1007/s00592-023-02094-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 04/10/2023] [Indexed: 05/16/2023]
Abstract
AIMS Diabetic kidney disease (DKD) is the one of the leading causes of end-stage kidney disease. Unraveling novel biomarker signatures capable to identify patients with DKD is favorable for tackle the burden. Here, we investigated the possible association between urinary metabolites and the presence of DKD in type 2 diabetes (T2D), and further, whether the associated metabolites improve discrimination of DKD and mediate the effect of inflammation on kidney involvement was evaluated. METHODS Two independent cohorts comprising 192 individuals (92 DKD) were analyzed. Urinary metabolites were analyzed by targeted metabolome profiling and inflammatory cytokine IL-18 were measured by ELISA. Differentially expressed metabolites were selected and mediation analysis was carried out. RESULTS Seven potential metabolite biomarkers (i.e., S-Adenosyl-L-homocysteine, propionic acid, oxoadipic acid, leucine, isovaleric acid, isobutyric acid, and indole-3-carboxylic acid) were identified using the discovery and validation design. In the pooled analysis, propionic acid, oxoadipic acid, leucine, isovaleric acid, isobutyric acid, and indole-3-carboxylic acid were markedly and independently associated with DKD. The composite index of 7 potential metabolite biomarkers (CMI) mediated 32.99% of the significant association between the inflammatory IL-18 and DKD. Adding the metabolite biomarkers improved the discrimination of DKD. CONCLUSIONS In T2D, several associated urinary metabolites were identified to improve the prediction of DKD. Whether interventions aimed at reducing CMI also reduce the risk of DKD especially in patients with high IL-18 needs further investigations.
Collapse
Affiliation(s)
- Caifeng Shi
- Center for Kidney Disease, Second Affiliated Hospital of Nanjing Medical University, No. 262 N Zhongshan Road, Nanjing, 210003, Jiangsu, China
| | - Yemeng Wan
- Center for Kidney Disease, Second Affiliated Hospital of Nanjing Medical University, No. 262 N Zhongshan Road, Nanjing, 210003, Jiangsu, China
| | - Aiqin He
- Center for Kidney Disease, Second Affiliated Hospital of Nanjing Medical University, No. 262 N Zhongshan Road, Nanjing, 210003, Jiangsu, China
| | - Xiaomei Wu
- Center for Kidney Disease, Second Affiliated Hospital of Nanjing Medical University, No. 262 N Zhongshan Road, Nanjing, 210003, Jiangsu, China
| | - Xinjia Shen
- Center for Kidney Disease, Second Affiliated Hospital of Nanjing Medical University, No. 262 N Zhongshan Road, Nanjing, 210003, Jiangsu, China
| | - Xueting Zhu
- Center for Kidney Disease, Second Affiliated Hospital of Nanjing Medical University, No. 262 N Zhongshan Road, Nanjing, 210003, Jiangsu, China
| | - Junwei Yang
- Center for Kidney Disease, Second Affiliated Hospital of Nanjing Medical University, No. 262 N Zhongshan Road, Nanjing, 210003, Jiangsu, China.
| | - Yang Zhou
- Center for Kidney Disease, Second Affiliated Hospital of Nanjing Medical University, No. 262 N Zhongshan Road, Nanjing, 210003, Jiangsu, China.
| |
Collapse
|
14
|
Xu J, Chen Q, Cai M, Han X, Lu H. Ultra-high performance liquid chromatography coupled to tandem mass spectrometry-based metabolomics study of diabetic distal symmetric polyneuropathy. J Diabetes Investig 2023; 14:1110-1120. [PMID: 37347226 PMCID: PMC10445193 DOI: 10.1111/jdi.14041] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 05/17/2023] [Accepted: 05/29/2023] [Indexed: 06/23/2023] Open
Abstract
AIMS/INTRODUCTION Distal symmetric polyneuropathy (DSPN) is a common complication of type 2 diabetes mellitus, but the underlining mechanisms have not yet been elucidated. The current study was designed to screen the feature metabolites classified as potential biomarkers, and to provide deeper insights into the underlying distinctive metabolic changes during disease progression. MATERIALS AND METHODS Plasma metabolite profiles were obtained by the ultra-high liquid chromatography coupled to tandem mass spectrometry method from healthy control participants, patients with type 2 diabetes mellitus and patients with DSPN. Potential biomarkers were selected through comprehensive analysis of statistically significant differences between groups. RESULTS Overall, 938 metabolites were identified. Among them, 12 metabolites (dimethylarginine, N6-acetyllysine, N-acetylhistidine, N,N,N-trimethyl-alanylproline betaine, cysteine, 7-methylguanine, N6-carbamoylthreonyladenosine, pseudouridine, 5-methylthioadenosine, N2,N2-dimethylguanosine, aconitate and C-glycosyl tryptophan) were identified as the specific biomarkers. The content of 12 metabolites were significantly higher in the DSPN group compared with the other two groups. Additionally, they showed good performance to discriminate the DSPN state. Correlation analyses showed that the levels of 12 metabolites might be more closely related to the glucose metabolic changes, followed by the levels of lipid metabolism. CONCLUSIONS The finding of the 12 signature metabolites might provide a novel perspective for the pathogenesis of DSPN. Future studies are required to test this observation further.
Collapse
Affiliation(s)
- Jiahui Xu
- Department of EndocrinologyShuguang Hospital Affiliated to Shanghai University of Traditional Chinese MedicineShanghaiChina
| | - Qingguang Chen
- Department of EndocrinologyShuguang Hospital Affiliated to Shanghai University of Traditional Chinese MedicineShanghaiChina
| | - Mengjie Cai
- Department of EndocrinologyShuguang Hospital Affiliated to Shanghai University of Traditional Chinese MedicineShanghaiChina
| | - Xu Han
- Department of EndocrinologyShuguang Hospital Affiliated to Shanghai University of Traditional Chinese MedicineShanghaiChina
| | - Hao Lu
- Department of EndocrinologyShuguang Hospital Affiliated to Shanghai University of Traditional Chinese MedicineShanghaiChina
| |
Collapse
|
15
|
Aleidi SM, Al Fahmawi H, Masoud A, Rahman AA. Metabolomics in diabetes mellitus: clinical insight. Expert Rev Proteomics 2023; 20:451-467. [PMID: 38108261 DOI: 10.1080/14789450.2023.2295866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 12/13/2023] [Indexed: 12/19/2023]
Abstract
INTRODUCTION Diabetes Mellitus (DM) is a chronic heterogeneous metabolic disorder characterized by hyperglycemia due to the destruction of insulin-producing pancreatic β cells and/or insulin resistance. It is now considered a global epidemic disease associated with serious threats to a patient's life. Understanding the metabolic pathways involved in disease pathogenesis and progression is important and would improve prevention and management strategies. Metabolomics is an emerging field of research that offers valuable insights into the metabolic perturbation associated with metabolic diseases, including DM. AREA COVERED Herein, we discussed the metabolomics in type 1 and 2 DM research, including its contribution to understanding disease pathogenesis and identifying potential novel biomarkers clinically useful for disease screening, monitoring, and prognosis. In addition, we highlighted the metabolic changes associated with treatment effects, including insulin and different anti-diabetic medications. EXPERT OPINION By analyzing the metabolome, the metabolic disturbances involved in T1DM and T2DM can be explored, enhancing our understanding of the disease progression and potentially leading to novel clinical diagnostic and effective new therapeutic approaches. In addition, identifying specific metabolites would be potential clinical biomarkers for predicting the disease and thus preventing and managing hyperglycemia and its complications.
Collapse
Affiliation(s)
- Shereen M Aleidi
- Department of Biopharmaceutics and Clinical Pharmacy, School of Pharmacy, The University of Jordan, Amman, Jordan
| | - Hiba Al Fahmawi
- Department of Biopharmaceutics and Clinical Pharmacy, School of Pharmacy, The University of Jordan, Amman, Jordan
| | - Afshan Masoud
- Proteomics Resource Unit, Obesity Research Center, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Anas Abdel Rahman
- Department of Biochemistry and Molecular Medicine, College of Medicine, Al Faisal University, Riyadh, Saudi Arabia
- Metabolomics Section, Department of Clinical Genomics, Center for Genomics Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
- Department of Chemistry, Memorial University of Newfoundland, St. John's, Newfoundland and Labrador, Canada
| |
Collapse
|
16
|
Abe T. [Therapy for CKD and DKD]. Nihon Yakurigaku Zasshi 2023; 158:319-325. [PMID: 37394553 DOI: 10.1254/fpj.22133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Diabetic kidney disease is a major cause of renal failure that urgently necessitates a breakthrough in disease management. Specific remedies are needed for preventing Type 2 diabetes which causes significant changes in an array of plasma metabolites. By untargeted metabolome analysis, phenyl sulfate (PS) increased with the progression of diabetes. In experimental diabetes models, PS administration induces albuminuria and podocyte damage due to the mitochondrial dysfunction. By clinical diabetic kidney disease (DKD) cohort analysis, it was also confirmed that the PS levels significantly correlate with basal and predicted 2-year progression of albuminuria. Phenol is synthesized from dietary tyrosine by gut bacterial-specific tyrosine phenol-lyase (TPL), and absorbed phenol is metabolized into PS in the liver. Inhibition of TPL reduces not only the circulating PS level but also albuminuria in diabetic mice. TPL inhibitor did not significantly alter the major composition, showing the non-lethal inhibition of microbial-specific enzymes has a therapeutic advantage, with lower selective pressure for the development of drug resistance. Clinically, 362 patients in a multi-center clinical study in diabetic nephropathy cohort (U-CARE) were analyzed with full data. The basal plasma PS level significantly correlated with ACR, eGFR, age, duration, HbA1c and uric acid, but not with suPAR. Multiple regression analysis revealed that ACR was the only factor that significantly correlated with PS. By stratified logistic regression analysis, in the microalbuminuria group, PS was the only factor related to the amount of change in the 2-year ACR in all models. PS is not only an early diagnosis marker, but also a modifiable cause and therefore a target for the treatment of DKD. Reduction of microbiota-derived phenol by the inhibitor should represent another aspect for developing drugs of DKD prevention.
Collapse
Affiliation(s)
- Takaaki Abe
- Division of Medical Science, Tohoku University Graduate School of Biomedical Engineering
- Department of Clinical Biology and Hormonal Regulation, Tohoku University Graduate School of Medicine
- AMED Moon Program Manager
| |
Collapse
|
17
|
Liu J, Nair V, Zhao YY, Chang DY, Limonte C, Bansal N, Fermin D, Eichinger F, Tanner EC, Bellovich KA, Steigerwalt S, Bhat Z, Hawkins JJ, Subramanian L, Rosas SE, Sedor JR, Vasquez MA, Waikar SS, Bitzer M, Pennathur S, Brosius FC, De Boer I, Chen M, Kretzler M, Ju W, for the Kidney Precision Medicine Project and Michigan Translational Core C-PROBE Investigator Group. Multi-Scalar Data Integration Links Glomerular Angiopoietin-Tie Signaling Pathway Activation With Progression of Diabetic Kidney Disease. Diabetes 2022; 71:2664-2676. [PMID: 36331122 PMCID: PMC9750948 DOI: 10.2337/db22-0169] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 08/17/2022] [Indexed: 11/06/2022]
Abstract
Diabetic kidney disease (DKD) is the leading cause of end-stage kidney disease (ESKD). Prognostic biomarkers reflective of underlying molecular mechanisms are critically needed for effective management of DKD. A three-marker panel was derived from a proteomics analysis of plasma samples by an unbiased machine learning approach from participants (N = 58) in the Clinical Phenotyping and Resource Biobank study. In combination with standard clinical parameters, this panel improved prediction of the composite outcome of ESKD or a 40% decline in glomerular filtration rate. The panel was validated in an independent group (N = 68), who also had kidney transcriptomic profiles. One marker, plasma angiopoietin 2 (ANGPT2), was significantly associated with outcomes in cohorts from the Cardiovascular Health Study (N = 3,183) and the Chinese Cohort Study of Chronic Kidney Disease (N = 210). Glomerular transcriptional angiopoietin/Tie (ANG-TIE) pathway scores, derived from the expression of 154 ANG-TIE signaling mediators, correlated positively with plasma ANGPT2 levels and kidney outcomes. Higher receptor expression in glomeruli and higher ANG-TIE pathway scores in endothelial cells corroborated potential functional effects in the kidney from elevated plasma ANGPT2 levels. Our work suggests that ANGPT2 is a promising prognostic endothelial biomarker with likely functional impact on glomerular pathogenesis in DKD.
Collapse
Affiliation(s)
- Jiahao Liu
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Viji Nair
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI
| | - Yi-yang Zhao
- Renal Division, Department of Medicine, Peking University First Hospital, Beijing, China
| | - Dong-yuan Chang
- Renal Division, Department of Medicine, Peking University First Hospital, Beijing, China
| | | | - Nisha Bansal
- Division of Nephrology, University of Washington, Seattle, WA
| | - Damian Fermin
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI
| | - Felix Eichinger
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI
| | - Emily C. Tanner
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI
| | | | - Susan Steigerwalt
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, MI
| | - Zeenat Bhat
- Department of Nephrology and Hypertension, Department of Medicine, Wayne State University, Detroit, MI
| | - Jennifer J. Hawkins
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI
| | - Lalita Subramanian
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI
| | - Sylvia E. Rosas
- Kidney and Hypertension Unit, Joslin Diabetes Center and Harvard Medical School, Boston, MA
| | - John R. Sedor
- Department of Medicine, Cleveland Clinic, Cleveland, OH
| | - Miguel A. Vasquez
- Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX
| | - Sushrut S. Waikar
- Section of Nephrology, Department of Medicine, Boston University School of Medicine and Boston Medical Center, Brookline, MA
| | - Markus Bitzer
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI
| | - Subramaniam Pennathur
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI
| | - Frank C. Brosius
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI
- Division of Nephrology, Department of Medicine, University of Arizona, Tucson, AZ
| | - Ian De Boer
- Division of Nephrology, University of Washington, Seattle, WA
| | - Min Chen
- Renal Division, Department of Medicine, Peking University First Hospital, Beijing, China
| | - Matthias Kretzler
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI
| | - Wenjun Ju
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI
| | | |
Collapse
|
18
|
Discovering a trans-omics biomarker signature that predisposes high risk diabetic patients to diabetic kidney disease. NPJ Digit Med 2022; 5:166. [PMID: 36323795 PMCID: PMC9630270 DOI: 10.1038/s41746-022-00713-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 10/18/2022] [Indexed: 11/27/2022] Open
Abstract
Diabetic kidney disease is the leading cause of end-stage kidney disease worldwide; however, the integration of high-dimensional trans-omics data to predict this diabetic complication is rare. We develop artificial intelligence (AI)-assisted models using machine learning algorithms to identify a biomarker signature that predisposes high risk patients with diabetes mellitus (DM) to diabetic kidney disease based on clinical information, untargeted metabolomics, targeted lipidomics and genome-wide single nucleotide polymorphism (SNP) datasets. This involves 618 individuals who are split into training and testing cohorts of 557 and 61 subjects, respectively. Three models are developed. In model 1, the top 20 features selected by AI give an accuracy rate of 0.83 and an area under curve (AUC) of 0.89 when differentiating DM and non-DM individuals. In model 2, among DM patients, a biomarker signature of 10 AI-selected features gives an accuracy rate of 0.70 and an AUC of 0.76 when identifying subjects at high risk of renal impairment. In model 3, among non-DM patients, a biomarker signature of 25 AI-selected features gives an accuracy rate of 0.82 and an AUC of 0.76 when pinpointing subjects at high risk of chronic kidney disease. In addition, the performance of the three models is rigorously verified using an independent validation cohort. Intriguingly, analysis of the protein-protein interaction network of the genes containing the identified SNPs (RPTOR, CLPTM1L, ALDH1L1, LY6D, PCDH9, B3GNTL1, CDS1, ADCYAP and FAM53A) reveals that, at the molecular level, there seems to be interconnected factors that have an effect on the progression of renal impairment among DM patients. In conclusion, our findings reveal the potential of employing machine learning algorithms to augment traditional methods and our findings suggest what molecular mechanisms may underlie the complex interaction between DM and chronic kidney disease. Moreover, the development of our AI-assisted models will improve precision when diagnosing renal impairment in predisposed patients, both DM and non-DM. Finally, a large prospective cohort study is needed to validate the clinical utility and mechanistic implications of these biomarker signatures.
Collapse
|
19
|
Zhu H, Wang M, Xiong X, Du Y, Li D, Wang Z, Ge W, Zhu Y. Plasma metabolomic profiling reveals factors associated with dose-adjusted trough concentration of tacrolimus in liver transplant recipients. Front Pharmacol 2022; 13:1045843. [PMID: 36386159 PMCID: PMC9659571 DOI: 10.3389/fphar.2022.1045843] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 10/13/2022] [Indexed: 07/30/2023] Open
Abstract
Inter- and intrapatient variability of tacrolimus exposure is a vital prognostic risk factor for the clinical outcome of liver transplantation. New factors or biomarkers characterizing tacrolimus disposition is essential for optimal dose prediction in recipients of liver transplant. The aim of the study was to identify potential plasma metabolites associated with the dose-adjusted trough concentration of tacrolimus in liver transplant recipients by using a global metabolomic approach. A total of 693 plasma samples were collected from 137 liver transplant recipients receiving tacrolimus and regular therapeutic drug monitoring. Untargeted metabolomic analysis was performed by ultraperformance liquid chromatography-quadrupole time-of-flight mass spectrometry. Univariate and multivariate analyses with a mixed linear model were conducted, and the results showed that the dose-adjusted tacrolimus trough concentration was associated with 31 endogenous metabolites, including medium- and long-chain acylcarnitines such as stearoylcarnitine (β = 0.222, p = 0.001), microbiota-derived uremic retention solutes such as indolelactic acid (β = 0.194, p = 0.007), bile acids such as taurohyodeoxycholic acid (β = -0.056, p = 0.002), and steroid hormones such as testosterone (β = 0.099, p = 0.001). A multiple linear mixed model including 11 metabolites and clinical information was established with a suitable predictive performance (correlation coefficient based on fixed effects = 0.64 and correlation coefficient based on fixed and random effects = 0.78). These data demonstrated that microbiota-derived uremic retention solutes, bile acids, steroid hormones, and medium- and long-chain acylcarnitines were the main metabolites associated with the dose-adjusted trough concentration of tacrolimus in liver transplant recipients.
Collapse
Affiliation(s)
- Huaijun Zhu
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
- Department of Pharmacy, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
- Nanjing Medical Center for Clinical Pharmacy, Nanjing, China
| | - Min Wang
- Department of Pharmacy, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
- Nanjing Medical Center for Clinical Pharmacy, Nanjing, China
| | - Xiaofu Xiong
- Department of Pharmacy, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yao Du
- Department of Pharmacy, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
- Nanjing Medical Center for Clinical Pharmacy, Nanjing, China
| | - Danying Li
- Department of Pharmacy, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
- Nanjing Medical Center for Clinical Pharmacy, Nanjing, China
| | - Zhou Wang
- State Key Laboratory of Quality Research in Chinese Medicine and School of Pharmacy, Macau University of Science and Technology, Macau, China
| | - Weihong Ge
- Department of Pharmacy, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
- Nanjing Medical Center for Clinical Pharmacy, Nanjing, China
| | - Yizhun Zhu
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
- State Key Laboratory of Quality Research in Chinese Medicine and School of Pharmacy, Macau University of Science and Technology, Macau, China
| |
Collapse
|
20
|
Wen D, Zheng Z, Surapaneni A, Yu B, Zhou L, Zhou W, Xie D, Shou H, Avila-Pacheco J, Kalim S, He J, Hsu CY, Parsa A, Rao P, Sondheimer J, Townsend R, Waikar SS, Rebholz CM, Denburg MR, Kimmel PL, Vasan RS, Clish CB, Coresh J, Feldman HI, Grams ME, Rhee EP, the CKD Biomarkers Consortium and CRIC Study Investigators. Metabolite profiling of CKD progression in the chronic renal insufficiency cohort study. JCI Insight 2022; 7:e161696. [PMID: 36048534 PMCID: PMC9714776 DOI: 10.1172/jci.insight.161696] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 08/31/2022] [Indexed: 11/17/2022] Open
Abstract
BACKGROUNDMetabolomic profiling in individuals with chronic kidney disease (CKD) has the potential to identify novel biomarkers and provide insight into disease pathogenesis.METHODSWe examined the association between blood metabolites and CKD progression, defined as the subsequent development of end-stage renal disease (ESRD) or estimated glomerular filtrate rate (eGFR) halving, in 1,773 participants of the Chronic Renal Insufficiency Cohort (CRIC) study, 962 participants of the African-American Study of Kidney Disease and Hypertension (AASK), and 5,305 participants of the Atherosclerosis Risk in Communities (ARIC) study.RESULTSIn CRIC, more than half of the measured metabolites were associated with CKD progression in minimally adjusted Cox proportional hazards models, but the number and strength of associations were markedly attenuated by serial adjustment for covariates, particularly eGFR. Ten metabolites were significantly associated with CKD progression in fully adjusted models in CRIC; 3 of these metabolites were also significant in fully adjusted models in AASK and ARIC, highlighting potential markers of glomerular filtration (pseudouridine), histamine metabolism (methylimidazoleacetate), and azotemia (homocitrulline). Our findings also highlight N-acetylserine as a potential marker of kidney tubular function, with significant associations with CKD progression observed in CRIC and ARIC.CONCLUSIONOur findings demonstrate the application of metabolomics to identify potential biomarkers and causal pathways in CKD progression.FUNDINGThis study was supported by the NIH (U01 DK106981, U01 DK106982, U01 DK085689, R01 DK108803, and R01 DK124399).
Collapse
Affiliation(s)
- Donghai Wen
- Nephrology Division and
- Endocrine Unit, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Zihe Zheng
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Aditya Surapaneni
- Department of Epidemiology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland, USA
- Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins University, Baltimore, Maryland, USA
| | - Bing Yu
- Department of Epidemiology, Human Genetics & Environmental Sciences, University of Texas Health Sciences Center at Houston School of Public Health, Houston, Texas, USA
| | - Linda Zhou
- Department of Epidemiology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland, USA
- Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins University, Baltimore, Maryland, USA
| | - Wen Zhou
- Nephrology Division and
- Endocrine Unit, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Dawei Xie
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Haochang Shou
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | | | | | - Jiang He
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, Louisiana, USA
| | - Chi-Yuan Hsu
- Division of Nephrology, University of California San Francisco School of Medicine, San Francisco, California, USA
- Division of Research, Kaiser Permanente Northern California, Oakland, California, USA
| | - Afshin Parsa
- Division of Kidney, Urologic, and Hematologic Diseases, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), Bethesda, Maryland, USA
| | - Panduranga Rao
- Division of Nephrology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - James Sondheimer
- Division of Nephrology and Hypertension, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Raymond Townsend
- Renal-Electrolyte and Hypertension Division, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Sushrut S. Waikar
- Section of Nephrology, Boston University School of Medicine, Boston Medical Center, Boston, Massachusetts, USA
| | - Casey M. Rebholz
- Department of Epidemiology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland, USA
- Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins University, Baltimore, Maryland, USA
| | - Michelle R. Denburg
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Division of Pediatric Nephrology, Children’s Hospital of Philadelphia, and
- Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Paul L. Kimmel
- Division of Kidney, Urologic, and Hematologic Diseases, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), Bethesda, Maryland, USA
| | - Ramachandran S. Vasan
- Department of Epidemiology, Boston University School of Public Health, Boston, Massachusetts, USA
- Sections of Preventive Medicine and Epidemiology and Cardiology, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Clary B. Clish
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Josef Coresh
- Department of Epidemiology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland, USA
- Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins University, Baltimore, Maryland, USA
| | - Harold I. Feldman
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Morgan E. Grams
- Department of Epidemiology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland, USA
- Department of Medicine, New York University, New York, New York, USA
| | - Eugene P. Rhee
- Nephrology Division and
- Endocrine Unit, Massachusetts General Hospital, Boston, Massachusetts, USA
| | | |
Collapse
|
21
|
Bernard L, Zhou L, Surapaneni A, Chen J, Rebholz CM, Coresh J, Yu B, Boerwinkle E, Schlosser P, Grams ME. Serum Metabolites and Kidney Outcomes: The Atherosclerosis Risk in Communities Study. Kidney Med 2022; 4:100522. [PMID: 36046612 PMCID: PMC9420957 DOI: 10.1016/j.xkme.2022.100522] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
Rationale & Objective Novel metabolite biomarkers of kidney failure with replacement therapy (KFRT) may help identify people at high risk for adverse kidney outcomes and implicated pathways may aid in developing targeted therapeutics. Study Design Prospective cohort. Setting & Participants The cohort included 3,799 Atherosclerosis Risk in Communities study participants with serum samples available for measurement at visit 1 (1987-1989). Exposure Baseline serum levels of 318 metabolites. Outcomes Incident KFRT, kidney failure (KFRT, estimated glomerular filtration rate <15 mL/min/1.73 m2, or death from kidney disease). Analytical Approach Because metabolites are often intercorrelated and represent shared pathways, we used a high dimension reduction technique called Netboost to cluster metabolites. Longitudinal associations between clusters of metabolites and KFRT and kidney failure were estimated using a Cox proportional hazards model. Results Mean age of study participants was 53 years, 61% were African American, and 13% had diabetes. There were 160 KFRT cases and 357 kidney failure cases over a mean of 23 years. The 314 metabolites were grouped in 43 clusters. Four clusters were significantly associated with risk of KFRT and 6 were associated with kidney failure (including 3 shared clusters). The 3 shared clusters suggested potential pathways perturbed early in kidney disease: cluster 5 (15 metabolites involved in alanine, aspartate, and glutamate metabolism as well as 5-oxoproline and several gamma-glutamyl amino acids), cluster 26 (6 metabolites involved in sugar and inositol phosphate metabolism), and cluster 34 (21 metabolites involved in glycerophospholipid metabolism). Several individual metabolites were also significantly associated with both KFRT and kidney failure, including glucose and mannose, which were associated with higher risk of both outcomes, and 5-oxoproline, gamma-glutamyl amino acids, linoleoylglycerophosphocholine, 1,5-anhydroglucitol, which were associated with lower risk of both outcomes. Limitations Inability to determine if the metabolites cause or are a consequence of changes in kidney function. Conclusions We identified several clusters of metabolites reproducibly associated with development of KFRT. Future experimental studies are needed to validate our findings as well as continue unraveling metabolic pathways involved in kidney function decline.
Collapse
|
22
|
Wang CH, Lu WL, Chiang SL, Tsai TH, Liu SC, Hsieh CH, Su PH, Huang CY, Tsai FJ, Lin YJ, Huang YN. T Cells Mediate Kidney Tubular Injury via Impaired PDHA1 and Autophagy in Type 1 Diabetes. J Clin Endocrinol Metab 2022; 107:2556-2570. [PMID: 35731579 DOI: 10.1210/clinem/dgac378] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Indexed: 11/19/2022]
Abstract
CONTEXT Nephropathy is a severe complication of type 1 diabetes (T1DM). However, the interaction between the PDHA1-regulated mechanism and CD4+ T cells in the early stage of kidney tubular injury remains unknown. OBJECTIVE To evaluate the role of PDHA1 in the regulation of tubular cells and CD4+ T cells and further to study its interaction in tubular cell injury in T1DM. METHODS Plasma and total RNA were collected from T cells of T1DM patients (n = 35) and healthy donors (n = 33) and evaluated for neutrophil gelatinase-associated lipocalin (NGAL), kidney injury molecule-1, PDHA1, and biomarkers of CD4+ T cells including T helper 1 cells (Th1) and regulatory T cells (Treg) markers. HK-2 cells cocultured with CD4+ T cells from T1DM patients or healthy donors (HDs) to evaluate the interaction with CD4+ T cells. RESULTS Increased PDHA1 gene expression levels in CD4+ T cells were positively associated with the plasma level of NGAL in T1DM patients and HDs. Our data demonstrated that the Th1/Treg subsets skewed Th1 in T1DM. Knockdown of PDHA1 in kidney tubular cells decreased ATP/ROS production, NAD/NADH ratio, mitochondrial respiration, and cell apoptosis. Furthermore, PDHA1 depletion induced impaired autophagic flux. Coculture of tubular cells and T1DM T cells showed impaired CPT1A, upregulated FASN, and induced kidney injury. CONCLUSION Our findings indicate that Th1 cells induced tubular cell injury through dysregulated metabolic reprogramming and autophagy, thereby indicating a new therapeutic approach for kidney tubular injury in T1DM.
Collapse
Affiliation(s)
- Chung-Hsing Wang
- Division of Genetics and Metabolism, Children's Hospital of China Medical University, Taichung 40402, Taiwan
- School of Medicine, China Medical University, Taichung 40402, Taiwan
| | - Wen-Li Lu
- Division of Genetics and Metabolism, Children's Hospital of China Medical University, Taichung 40402, Taiwan
| | - Shang-Lun Chiang
- Department of Medical Laboratory Science, College of Medical Science and Technology, I-Shou University, Kaohsiung 82445, Taiwan
| | - Tsung-Hsun Tsai
- Division of Urology, Department of Surgery, Taichung Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Taichung 42743, Taiwan
| | - Su-Ching Liu
- Department of Medical Research, Children's Hospital of China Medical University, Taichung 40402, Taiwan
| | - Chia-Hung Hsieh
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 40402
- Department of Medical Research, China Medical University Hospital, Taichung 40402, Taiwan
| | - Pen-Hua Su
- Department of Pediatrics, Chung Shan Medical University Hospital, Taichung 40242, Taiwan
- School of Medicine, Chung Shan Medical University; Taichung 40242, Taiwan
| | - Chih-Yang Huang
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 40402
- Department of Medical Research, China Medical University Hospital, Taichung 40402, Taiwan
- Cardiovascular and Mitochondrial Related Disease Research Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 97002, Taiwan
- Center of General Education, Buddhist Tzu Chi Medical Foundation, Tzu Chi University of Science and Technology, Hualien 97002, Taiwan
- Department of Medical Laboratory Science and Biotechnology, Asia University, Taichung 41354, Taiwan
| | - Fuu-Jen Tsai
- Genetic Center, Department of Medical Research, China Medical University Hospital, Taichung 40402, Taiwan
| | - Yu-Jung Lin
- Cardiovascular and Mitochondrial Related Disease Research Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 97002, Taiwan
| | - Yu-Nan Huang
- Division of Genetics and Metabolism, Children's Hospital of China Medical University, Taichung 40402, Taiwan
- Department of Life Sciences, National Chung Hsing University, Taichung 40227, Taiwan
| |
Collapse
|
23
|
Shin HE, Won CW, Kim M. Metabolomic profiles to explore biomarkers of severe sarcopenia in older men: A pilot study. Exp Gerontol 2022; 167:111924. [PMID: 35963453 DOI: 10.1016/j.exger.2022.111924] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 07/19/2022] [Accepted: 08/07/2022] [Indexed: 11/04/2022]
Abstract
BACKGROUND The pathophysiology of sarcopenia is complex and multifactorial; however, it has not yet been fully elucidated. Identifying metabolomic profiles may help clarify the mechanisms underlying sarcopenia. OBJECTIVE This pilot study explored potential noninvasive biomarkers of severe sarcopenia through metabolomic analysis in community-dwelling older men. METHODS Twenty older men (mean age: 81.9 ± 2.8 years) were selected from the Korean Frailty and Aging Cohort Study. Participants with severe sarcopenia (n = 10) were compared with non-sarcopenic, age- and body mass index-matched controls (n = 10). Severe sarcopenia was defined as low muscle mass, low muscle strength, and low physical performance using the Asian Working Group for Sarcopenia 2019 criteria. Non-targeted metabolomic profiling of plasma metabolites was performed using capillary electrophoresis time-of-flight mass spectrometry and absolute quantification was performed in target metabolites. RESULTS Among 191 plasma metabolic peaks, the concentrations of 10 metabolites significantly differed between severe sarcopenia group and non-sarcopenic controls. The plasma concentrations of L-alanine, homocitrulline, N-acetylserine, gluconic acid, N-acetylalanine, proline, and sulfotyrosine were higher, while those of 4-methyl-2-oxovaleric acid, 3-methyl-2-oxovaleric acid, and tryptophan were lower in participants with severe sarcopenia than in non-sarcopenic controls (all, p < 0.05). Among the 53 metabolites quantified as target metabolites, L-alanine (area under the receiver operating characteristic curve [AUC] = 0.760; p = 0.049), gluconic acid (AUC = 0.800; p = 0.023), proline (AUC = 0.785; p = 0.031), and tryptophan (AUC = 0.800; p = 0.023) determined the presence of severe sarcopenia. CONCLUSIONS Plasma metabolomic analysis demonstrated that L-alanine, gluconic acid, proline, and tryptophan may be potential biomarkers of severe sarcopenia. The identified metabolites can provide new insights into the underlying pathophysiology of severe sarcopenia and serve as the basis for preventive interventions.
Collapse
Affiliation(s)
- Hyung Eun Shin
- Department of Biomedical Science and Technology, Graduate School, Kyung Hee University, Seoul 02447, South Korea
| | - Chang Won Won
- Elderly Frailty Research Center, Department of Family Medicine, College of Medicine, Kyung Hee University, Kyung Hee University Medical Center, Seoul 02447, South Korea.
| | - Miji Kim
- Department of Biomedical Science and Technology, College of Medicine, East-West Medical Research Institute, Kyung Hee University, Seoul 02447, South Korea.
| |
Collapse
|
24
|
Shah HS, Moreno LO, Morieri ML, Tang Y, Mendonca C, Jobe JM, Thacker JB, Mitri J, Monti S, Niewczas MA, Pennathur S, Doria A. Serum Orotidine: A Novel Biomarker of Increased CVD Risk in Type 2 Diabetes Discovered Through Metabolomics Studies. Diabetes Care 2022; 45:1882-1892. [PMID: 35696261 PMCID: PMC9346986 DOI: 10.2337/dc21-1789] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 04/26/2022] [Indexed: 02/03/2023]
Abstract
OBJECTIVE To identify novel biomarkers of cardiovascular disease (CVD) risk in type 2 diabetes (T2D) via a hypothesis-free global metabolomics study, while taking into account renal function, an important confounder often overlooked in previous metabolomics studies of CVD. RESEARCH DESIGN AND METHODS We conducted a global serum metabolomics analysis using the Metabolon platform in a discovery set from the Joslin Kidney Study having a nested case-control design comprising 409 individuals with T2D. Logistic regression was applied to evaluate the association between incident CVD events and each of the 671 metabolites detected by the Metabolon platform, before and after adjustment for renal function and other CVD risk factors. Significant metabolites were followed up with absolute quantification assays in a validation set from the Joslin Heart Study including 599 individuals with T2D with and without clinical evidence of significant coronary heart disease (CHD). RESULTS In the discovery set, serum orotidine and 2-piperidinone were significantly associated with increased odds of incident CVD after adjustment for glomerular filtration rate (GFR) (odds ratio [OR] per SD increment 1.94 [95% CI 1.39-2.72], P = 0.0001, and 1.62 [1.26-2.08], P = 0.0001, respectively). Orotidine was also associated with increased odds of CHD in the validation set (OR 1.39 [1.11-1.75]), while 2-piperidinone did not replicate. Furthermore, orotidine, being inversely associated with GFR, mediated 60% of the effects of declining renal function on CVD risk. Addition of orotidine to established clinical predictors improved (P < 0.05) C statistics and discrimination indices for CVD risk (ΔAUC 0.053, rIDI 0.48, NRI 0.42) compared with the clinical predictors alone. CONCLUSIONS Through a robust metabolomics approach, with independent validation, we have discovered serum orotidine as a novel biomarker of increased odds of CVD in T2D, independent of renal function. Additionally, orotidine may be a biological mediator of the increased CVD risk associated with poor kidney function and may help improve CVD risk prediction in T2D.
Collapse
Affiliation(s)
- Hetal S. Shah
- Section on Genetics and Epidemiology, Research Division, Joslin Diabetes Center, Boston, MA
- Department of Medicine, Harvard Medical School, Boston, MA
| | - Lorena Ortega Moreno
- Department of Basic Health Sciences, Universidad Rey Juan Carlos, Alcorcón, Spain
- High Performance Research Group in Physiopathology and Pharmacology of the Digestive System (NeuGut), Universidad Rey Juan Carlos, Alcorcón, Spain
| | | | - Yaling Tang
- Section on Genetics and Epidemiology, Research Division, Joslin Diabetes Center, Boston, MA
- Department of Medicine, Harvard Medical School, Boston, MA
| | - Christine Mendonca
- Section on Genetics and Epidemiology, Research Division, Joslin Diabetes Center, Boston, MA
| | - Jenny Marie Jobe
- Section on Genetics and Epidemiology, Research Division, Joslin Diabetes Center, Boston, MA
| | - Jonathan B. Thacker
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI
| | - Joanna Mitri
- Department of Medicine, Harvard Medical School, Boston, MA
| | - Stefano Monti
- Computational Biomedicine, Department of Medicine, Boston University, Boston, MA
| | - Monika A. Niewczas
- Section on Genetics and Epidemiology, Research Division, Joslin Diabetes Center, Boston, MA
- Department of Medicine, Harvard Medical School, Boston, MA
| | - Subramaniam Pennathur
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI
| | - Alessandro Doria
- Section on Genetics and Epidemiology, Research Division, Joslin Diabetes Center, Boston, MA
- Department of Medicine, Harvard Medical School, Boston, MA
| |
Collapse
|
25
|
Banimfreg BH, Alshraideh H, Shamayleh A, Guella A, Semreen MH, Al Bataineh MT, Soares NC. Untargeted Metabolomic Plasma Profiling of Emirati Dialysis Patients with Diabetes versus Non-Diabetic: A Pilot Study. Biomolecules 2022; 12:962. [PMID: 35883517 PMCID: PMC9313445 DOI: 10.3390/biom12070962] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 06/29/2022] [Accepted: 07/04/2022] [Indexed: 12/14/2022] Open
Abstract
Diabetic kidney disease (DKD) is a severe irreversible complication of diabetes mellitus that further disturbs glucose metabolism. Identifying metabolic changes in the blood may provide early insight into DKD pathogenesis. This study aims to determine blood biomarkers differentiating DKD from non-diabetic kidney disease in the Emirati population utilizing the LC-MS/MS platform. Blood samples were collected from hemodialysis subjects with and without diabetes to detect indicators of pathological changes using an untargeted metabolomics approach. Metabolic profiles were analyzed based on clinically confirmed diabetic status and current HbA1c values. Five differentially significant metabolites were identified based on the clinically confirmed diabetic status, including hydroxyprogesterone and 3,4-Dihydroxymandelic acid. Similarly, we identified seven metabolites with apparent differences between Dialysis Diabetic (DD) and Dialysis non-Diabetic (DND) groups, including isovalerylglycine based on HbA1c values. Likewise, the top three metabolic pathways, including Tyrosine metabolism, were identified following the clinically confirmed diabetic status. As a result, nine different metabolites were enriched in the identified metabolic pathways, such as 3,4-Dihydroxymandelic acid. As a result, eleven different metabolites were enriched, including Glycerol. This study provides an insight into blood metabolic changes related to DKD that may lead to more effective management strategies.
Collapse
Affiliation(s)
- Bayan Hassan Banimfreg
- Department of Industrial Engineering, College of Engineering, American University of Sharjah, Sharjah P.O. Box 26666, United Arab Emirates; (B.H.B.); (H.A.)
| | - Hussam Alshraideh
- Department of Industrial Engineering, College of Engineering, American University of Sharjah, Sharjah P.O. Box 26666, United Arab Emirates; (B.H.B.); (H.A.)
| | - Abdulrahim Shamayleh
- Department of Industrial Engineering, College of Engineering, American University of Sharjah, Sharjah P.O. Box 26666, United Arab Emirates; (B.H.B.); (H.A.)
| | - Adnane Guella
- Nephrology Department, University Hospital Sharjah, Sharjah P.O. Box 72772, United Arab Emirates;
| | - Mohammad Harb Semreen
- Department of Medicinal Chemistry, College of Pharmacy, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates; (M.H.S.); (N.C.S.)
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates
| | - Mohammad Tahseen Al Bataineh
- College of Medicine, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates
- College of Medicine and Health Sciences, Department of Genetics and Molecular Biology, Khalifa University of Science and Technology, Abu Dhabi P.O. Box 127788, United Arab Emirates
- Center for Biotechnology, Khalifa University of Science and Technology, Abu Dhabi P.O. Box 127788, United Arab Emirates
| | - Nelson C. Soares
- Department of Medicinal Chemistry, College of Pharmacy, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates; (M.H.S.); (N.C.S.)
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates
| |
Collapse
|
26
|
Characteristics of Normalization Methods in Quantitative Urinary Metabolomics—Implications for Epidemiological Applications and Interpretations. Biomolecules 2022; 12:biom12070903. [PMID: 35883459 PMCID: PMC9313036 DOI: 10.3390/biom12070903] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Revised: 06/16/2022] [Accepted: 06/21/2022] [Indexed: 01/25/2023] Open
Abstract
A systematic comparison is presented for the effects of seven different normalization schemes in quantitative urinary metabolomics. Morning spot urine samples were analyzed with nuclear magnetic resonance (NMR) spectroscopy from a population-based group of 994 individuals. Forty-four metabolites were quantified and the metabolite–metabolite associations and the associations of metabolite concentrations with two representative clinical measures, body mass index and mean arterial pressure, were analyzed. Distinct differences were observed when comparing the effects of normalization for the intra-urine metabolite associations with those for the clinical associations. The metabolite–metabolite associations show quite complex patterns of similarities and dissimilarities between the different normalization methods, while the epidemiological association patterns are consistent, leading to the same overall biological interpretations. The results indicate that, in general, the normalization method appears to have only minor influences on standard epidemiological regression analyses with clinical/physiological measures. Multimetabolite normalization schemes showed consistent results with the customary creatinine reference. Nevertheless, interpretations of intra-urine metabolite associations and nuanced understanding of the epidemiological associations call for comparisons with different normalizations and accounting for the physiology, metabolism and kidney function related to the normalization schemes.
Collapse
|
27
|
Geng X, Li Z, Yang Y. Emerging Role of Epitranscriptomics in Diabetes Mellitus and Its Complications. Front Endocrinol (Lausanne) 2022; 13:907060. [PMID: 35692393 PMCID: PMC9184717 DOI: 10.3389/fendo.2022.907060] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 04/14/2022] [Indexed: 01/13/2023] Open
Abstract
Diabetes mellitus (DM) and its related complications are among the leading causes of disability and mortality worldwide. Substantial studies have explored epigenetic regulation that is involved in the modifications of DNA and proteins, but RNA modifications in diabetes are still poorly investigated. In recent years, posttranscriptional epigenetic modification of RNA (the so-called 'epitranscriptome') has emerged as an interesting field of research. Numerous modifications, mainly N6 -methyladenosine (m6A), have been identified in nearly all types of RNAs and have been demonstrated to have an indispensable effect in a variety of human diseases, such as cancer, obesity, and diabetes. Therefore, it is particularly important to understand the molecular basis of RNA modifications, which might provide a new perspective for the pathogenesis of diabetes mellitus and the discovery of new therapeutic targets. In this review, we aim to summarize the recent progress in the epitranscriptomics involved in diabetes and diabetes-related complications. We hope to provide some insights for enriching the understanding of the epitranscriptomic regulatory mechanisms of this disease as well as the development of novel therapeutic targets for future clinical benefit.
Collapse
Affiliation(s)
- Xinqian Geng
- Department of Endocrinology, The Affiliated Hospital of Yunnan University and the Second People’s Hospital of Yunnan Province, Kunming, China
| | - Zheng Li
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, China
| | - Ying Yang
- Department of Endocrinology, The Affiliated Hospital of Yunnan University and the Second People’s Hospital of Yunnan Province, Kunming, China
| |
Collapse
|
28
|
Balasubramanian R, Hu J, Guasch-Ferre M, Li J, Sorond F, Zhao Y, Shutta KH, Salas-Salvado J, Hu F, Clish CB, Rexrode KM. Metabolomic Profiles Associated With Incident Ischemic Stroke. Neurology 2022; 98:e483-e492. [PMID: 34853177 PMCID: PMC8826464 DOI: 10.1212/wnl.0000000000013129] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 11/16/2021] [Indexed: 02/03/2023] Open
Abstract
BACKGROUND AND OBJECTIVES Women have higher lifetime risk of stroke than men, and metabolic factors seem more strongly associated with stroke for women than men. However, few studies in either men or women have evaluated metabolomic profiles and incident stroke. METHODS We applied liquid chromatography-tandem mass spectrometry to measure 519 plasma metabolites in a discovery set of women in the Nurses' Health Study (NHS; 454 incident ischemic stroke cases, 454 controls) with validation in 2 independent, prospective cohorts: Prevención con Dieta Mediterránea (PREDIMED; 118 stroke cases, 791 controls) and Nurses' Health Study 2 (NHS2; 49 ischemic stroke cases, 49 controls). We applied logistic regression models with stroke as the outcome to adjust for multiple risk factors; the false discovery rate was controlled through the q value method. RESULTS Twenty-three metabolites were significantly associated with incident stroke in NHS after adjustment for traditional risk factors (q < 0.05). Of these, 14 metabolites were available in PREDIMED and 3 were significantly associated with incident stroke: methionine sulfoxide, N6-acetyllysine, and sucrose (q < 0.05). In NHS2, one of the 23 metabolites (glucuronate) was significantly associated with incident stroke (q < 0.05). For all 4 metabolites, higher levels were associated with increased risk. These 4 metabolites were used to create a stroke metabolite score (SMS) in the NHS and tested in PREDIMED. Per unit of standard deviation of SMS, the odds ratio for incident stroke was 4.12 (95% confidence interval [CI] 2.26-7.51) in PREDIMED, after adjustment for risk factors. In PREDIMED, the area under the receiver operating characteristic curve (AUC) for the model including SMS and traditional risk factors was 0.70 (95% CI 0.75-0.79) vs the AUC for the model including the traditional risk factors only of 0.65 (95% CI 0.70-0.75), corresponding to a 5% improvement in risk prediction with SMS (p < 0.005). DISCUSSION Metabolites associated with stroke included 2 amino acids, a carboxylic acid, and sucrose. A composite SMS including these metabolites was associated with ischemic stroke and showed improvement in risk prediction beyond traditional risk factors. CLASSIFICATION OF EVIDENCE This study provides Class II evidence that a SMS accurately predicts incident ischemic stroke risk.
Collapse
Affiliation(s)
- Raji Balasubramanian
- From the Department of Biostatistics and Epidemiology (R.B., Y.Z., K.H.S.), University of Massachusetts-Amherst; Division of Women's Health (J.H., K.M.R.) and Channing Division of Network Medicine, Department of Medicine (M.G.-F., F.H.), Brigham and Women's Hospital, Harvard Medical School; Departments of Nutrition (M.G.-F., J.L., F.H.) and Epidemiology (J.L., F.H.), Harvard T.H. Chan School of Public Health, Boston, MA; Davee Department of Neurology, Division of Stroke and Neurocritical Care (F.S.), Northwestern Feinberg School of Medicine, Chicago, IL; Departament de Bioquímica i Biotecnologia, Unitat de Nutrició (J.S.S.), Universitat Rovira i Virgili, Reus; Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN) (J.S.-S.), Institute of Health Carlos III, Madrid; Nutrition Unit, Pere Virgili Research Institute (IISPV) (J.S.-S.), University Hospital of Sant Joan de Reus, Spain; and Broad Institute of the Massachusetts Institute of Technology and Harvard University (C.B.C.), Cambridge.
| | - Jie Hu
- From the Department of Biostatistics and Epidemiology (R.B., Y.Z., K.H.S.), University of Massachusetts-Amherst; Division of Women's Health (J.H., K.M.R.) and Channing Division of Network Medicine, Department of Medicine (M.G.-F., F.H.), Brigham and Women's Hospital, Harvard Medical School; Departments of Nutrition (M.G.-F., J.L., F.H.) and Epidemiology (J.L., F.H.), Harvard T.H. Chan School of Public Health, Boston, MA; Davee Department of Neurology, Division of Stroke and Neurocritical Care (F.S.), Northwestern Feinberg School of Medicine, Chicago, IL; Departament de Bioquímica i Biotecnologia, Unitat de Nutrició (J.S.S.), Universitat Rovira i Virgili, Reus; Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN) (J.S.-S.), Institute of Health Carlos III, Madrid; Nutrition Unit, Pere Virgili Research Institute (IISPV) (J.S.-S.), University Hospital of Sant Joan de Reus, Spain; and Broad Institute of the Massachusetts Institute of Technology and Harvard University (C.B.C.), Cambridge
| | - Marta Guasch-Ferre
- From the Department of Biostatistics and Epidemiology (R.B., Y.Z., K.H.S.), University of Massachusetts-Amherst; Division of Women's Health (J.H., K.M.R.) and Channing Division of Network Medicine, Department of Medicine (M.G.-F., F.H.), Brigham and Women's Hospital, Harvard Medical School; Departments of Nutrition (M.G.-F., J.L., F.H.) and Epidemiology (J.L., F.H.), Harvard T.H. Chan School of Public Health, Boston, MA; Davee Department of Neurology, Division of Stroke and Neurocritical Care (F.S.), Northwestern Feinberg School of Medicine, Chicago, IL; Departament de Bioquímica i Biotecnologia, Unitat de Nutrició (J.S.S.), Universitat Rovira i Virgili, Reus; Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN) (J.S.-S.), Institute of Health Carlos III, Madrid; Nutrition Unit, Pere Virgili Research Institute (IISPV) (J.S.-S.), University Hospital of Sant Joan de Reus, Spain; and Broad Institute of the Massachusetts Institute of Technology and Harvard University (C.B.C.), Cambridge
| | - Jun Li
- From the Department of Biostatistics and Epidemiology (R.B., Y.Z., K.H.S.), University of Massachusetts-Amherst; Division of Women's Health (J.H., K.M.R.) and Channing Division of Network Medicine, Department of Medicine (M.G.-F., F.H.), Brigham and Women's Hospital, Harvard Medical School; Departments of Nutrition (M.G.-F., J.L., F.H.) and Epidemiology (J.L., F.H.), Harvard T.H. Chan School of Public Health, Boston, MA; Davee Department of Neurology, Division of Stroke and Neurocritical Care (F.S.), Northwestern Feinberg School of Medicine, Chicago, IL; Departament de Bioquímica i Biotecnologia, Unitat de Nutrició (J.S.S.), Universitat Rovira i Virgili, Reus; Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN) (J.S.-S.), Institute of Health Carlos III, Madrid; Nutrition Unit, Pere Virgili Research Institute (IISPV) (J.S.-S.), University Hospital of Sant Joan de Reus, Spain; and Broad Institute of the Massachusetts Institute of Technology and Harvard University (C.B.C.), Cambridge
| | - Farzaneh Sorond
- From the Department of Biostatistics and Epidemiology (R.B., Y.Z., K.H.S.), University of Massachusetts-Amherst; Division of Women's Health (J.H., K.M.R.) and Channing Division of Network Medicine, Department of Medicine (M.G.-F., F.H.), Brigham and Women's Hospital, Harvard Medical School; Departments of Nutrition (M.G.-F., J.L., F.H.) and Epidemiology (J.L., F.H.), Harvard T.H. Chan School of Public Health, Boston, MA; Davee Department of Neurology, Division of Stroke and Neurocritical Care (F.S.), Northwestern Feinberg School of Medicine, Chicago, IL; Departament de Bioquímica i Biotecnologia, Unitat de Nutrició (J.S.S.), Universitat Rovira i Virgili, Reus; Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN) (J.S.-S.), Institute of Health Carlos III, Madrid; Nutrition Unit, Pere Virgili Research Institute (IISPV) (J.S.-S.), University Hospital of Sant Joan de Reus, Spain; and Broad Institute of the Massachusetts Institute of Technology and Harvard University (C.B.C.), Cambridge
| | - Yibai Zhao
- From the Department of Biostatistics and Epidemiology (R.B., Y.Z., K.H.S.), University of Massachusetts-Amherst; Division of Women's Health (J.H., K.M.R.) and Channing Division of Network Medicine, Department of Medicine (M.G.-F., F.H.), Brigham and Women's Hospital, Harvard Medical School; Departments of Nutrition (M.G.-F., J.L., F.H.) and Epidemiology (J.L., F.H.), Harvard T.H. Chan School of Public Health, Boston, MA; Davee Department of Neurology, Division of Stroke and Neurocritical Care (F.S.), Northwestern Feinberg School of Medicine, Chicago, IL; Departament de Bioquímica i Biotecnologia, Unitat de Nutrició (J.S.S.), Universitat Rovira i Virgili, Reus; Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN) (J.S.-S.), Institute of Health Carlos III, Madrid; Nutrition Unit, Pere Virgili Research Institute (IISPV) (J.S.-S.), University Hospital of Sant Joan de Reus, Spain; and Broad Institute of the Massachusetts Institute of Technology and Harvard University (C.B.C.), Cambridge
| | - Katherine H Shutta
- From the Department of Biostatistics and Epidemiology (R.B., Y.Z., K.H.S.), University of Massachusetts-Amherst; Division of Women's Health (J.H., K.M.R.) and Channing Division of Network Medicine, Department of Medicine (M.G.-F., F.H.), Brigham and Women's Hospital, Harvard Medical School; Departments of Nutrition (M.G.-F., J.L., F.H.) and Epidemiology (J.L., F.H.), Harvard T.H. Chan School of Public Health, Boston, MA; Davee Department of Neurology, Division of Stroke and Neurocritical Care (F.S.), Northwestern Feinberg School of Medicine, Chicago, IL; Departament de Bioquímica i Biotecnologia, Unitat de Nutrició (J.S.S.), Universitat Rovira i Virgili, Reus; Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN) (J.S.-S.), Institute of Health Carlos III, Madrid; Nutrition Unit, Pere Virgili Research Institute (IISPV) (J.S.-S.), University Hospital of Sant Joan de Reus, Spain; and Broad Institute of the Massachusetts Institute of Technology and Harvard University (C.B.C.), Cambridge
| | - Jordi Salas-Salvado
- From the Department of Biostatistics and Epidemiology (R.B., Y.Z., K.H.S.), University of Massachusetts-Amherst; Division of Women's Health (J.H., K.M.R.) and Channing Division of Network Medicine, Department of Medicine (M.G.-F., F.H.), Brigham and Women's Hospital, Harvard Medical School; Departments of Nutrition (M.G.-F., J.L., F.H.) and Epidemiology (J.L., F.H.), Harvard T.H. Chan School of Public Health, Boston, MA; Davee Department of Neurology, Division of Stroke and Neurocritical Care (F.S.), Northwestern Feinberg School of Medicine, Chicago, IL; Departament de Bioquímica i Biotecnologia, Unitat de Nutrició (J.S.S.), Universitat Rovira i Virgili, Reus; Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN) (J.S.-S.), Institute of Health Carlos III, Madrid; Nutrition Unit, Pere Virgili Research Institute (IISPV) (J.S.-S.), University Hospital of Sant Joan de Reus, Spain; and Broad Institute of the Massachusetts Institute of Technology and Harvard University (C.B.C.), Cambridge
| | - Frank Hu
- From the Department of Biostatistics and Epidemiology (R.B., Y.Z., K.H.S.), University of Massachusetts-Amherst; Division of Women's Health (J.H., K.M.R.) and Channing Division of Network Medicine, Department of Medicine (M.G.-F., F.H.), Brigham and Women's Hospital, Harvard Medical School; Departments of Nutrition (M.G.-F., J.L., F.H.) and Epidemiology (J.L., F.H.), Harvard T.H. Chan School of Public Health, Boston, MA; Davee Department of Neurology, Division of Stroke and Neurocritical Care (F.S.), Northwestern Feinberg School of Medicine, Chicago, IL; Departament de Bioquímica i Biotecnologia, Unitat de Nutrició (J.S.S.), Universitat Rovira i Virgili, Reus; Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN) (J.S.-S.), Institute of Health Carlos III, Madrid; Nutrition Unit, Pere Virgili Research Institute (IISPV) (J.S.-S.), University Hospital of Sant Joan de Reus, Spain; and Broad Institute of the Massachusetts Institute of Technology and Harvard University (C.B.C.), Cambridge
| | - Clary B Clish
- From the Department of Biostatistics and Epidemiology (R.B., Y.Z., K.H.S.), University of Massachusetts-Amherst; Division of Women's Health (J.H., K.M.R.) and Channing Division of Network Medicine, Department of Medicine (M.G.-F., F.H.), Brigham and Women's Hospital, Harvard Medical School; Departments of Nutrition (M.G.-F., J.L., F.H.) and Epidemiology (J.L., F.H.), Harvard T.H. Chan School of Public Health, Boston, MA; Davee Department of Neurology, Division of Stroke and Neurocritical Care (F.S.), Northwestern Feinberg School of Medicine, Chicago, IL; Departament de Bioquímica i Biotecnologia, Unitat de Nutrició (J.S.S.), Universitat Rovira i Virgili, Reus; Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN) (J.S.-S.), Institute of Health Carlos III, Madrid; Nutrition Unit, Pere Virgili Research Institute (IISPV) (J.S.-S.), University Hospital of Sant Joan de Reus, Spain; and Broad Institute of the Massachusetts Institute of Technology and Harvard University (C.B.C.), Cambridge
| | - Kathryn M Rexrode
- From the Department of Biostatistics and Epidemiology (R.B., Y.Z., K.H.S.), University of Massachusetts-Amherst; Division of Women's Health (J.H., K.M.R.) and Channing Division of Network Medicine, Department of Medicine (M.G.-F., F.H.), Brigham and Women's Hospital, Harvard Medical School; Departments of Nutrition (M.G.-F., J.L., F.H.) and Epidemiology (J.L., F.H.), Harvard T.H. Chan School of Public Health, Boston, MA; Davee Department of Neurology, Division of Stroke and Neurocritical Care (F.S.), Northwestern Feinberg School of Medicine, Chicago, IL; Departament de Bioquímica i Biotecnologia, Unitat de Nutrició (J.S.S.), Universitat Rovira i Virgili, Reus; Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN) (J.S.-S.), Institute of Health Carlos III, Madrid; Nutrition Unit, Pere Virgili Research Institute (IISPV) (J.S.-S.), University Hospital of Sant Joan de Reus, Spain; and Broad Institute of the Massachusetts Institute of Technology and Harvard University (C.B.C.), Cambridge
| |
Collapse
|
29
|
Mu X, Yang M, Ling P, Wu A, Zhou H, Jiang J. Acylcarnitines: Can They Be Biomarkers of Diabetic Nephropathy? Diabetes Metab Syndr Obes 2022; 15:247-256. [PMID: 35125878 PMCID: PMC8811266 DOI: 10.2147/dmso.s350233] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 01/13/2022] [Indexed: 12/22/2022] Open
Abstract
Diabetic nephropathy (DN), one of the most serious microvascular complications of diabetes mellitus (DM), may progress to end-stage renal disease (ESRD). Current biochemical biomarkers, such as urinary albumin excretion rate (UAER), have limitations for early screening and monitoring of DN. Recent studies have identified some metabolites as candidate biomarkers for early detection of DN. In this review, we summarize the role of dysregulated acylcarnitines (AcylCNs) in DN pathophysiology. Lower abundance of short- and medium-chain AcylCNs and higher long-chain AcylCNs often occurred in DM with normal albuminuria and microalbuminuria, compared with advanced stages of DN. The increase of long-chain AcylCNs was supposed to be an adaptive compensation in fat acids (FAs) oxidation in the early stage of DN. Conversely, the decrease of long-chain AcylCNs was due to incomplete oxidation of FAs in advanced stage of DN. Thus, AcylCNs may serve as sensitive biomarkers in predicting the risk of DN.
Collapse
Affiliation(s)
- Xiaodie Mu
- Department of Nephrology, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, People’s Republic of China
| | - Min Yang
- Department of Nephrology, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, People’s Republic of China
| | - Peiyao Ling
- Department of Nephrology, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, People’s Republic of China
| | - Aihua Wu
- Department of Nephrology, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, People’s Republic of China
| | - Hua Zhou
- Department of Nephrology, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, People’s Republic of China
| | - Jingting Jiang
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, People’s Republic of China
| |
Collapse
|
30
|
Hansen CS, Suvitaival T, Theilade S, Mattila I, Lajer M, Trošt K, Ahonen L, Hansen TW, Legido-Quigley C, Rossing P, Ahluwalia TS. Cardiovascular Autonomic Neuropathy in Type 1 Diabetes Is Associated With Disturbances in TCA, Lipid, and Glucose Metabolism. Front Endocrinol (Lausanne) 2022; 13:831793. [PMID: 35498422 PMCID: PMC9046722 DOI: 10.3389/fendo.2022.831793] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 03/11/2022] [Indexed: 11/17/2022] Open
Abstract
INTRODUCTION Diabetic cardiovascular autonomic neuropathy (CAN) is associated with increased mortality and morbidity. To explore metabolic mechanisms associated with CAN we investigated associations between serum metabolites and CAN in persons with type 1 diabetes (T1D). MATERIALS AND METHODS Cardiovascular reflex tests (CARTs) (heart rate response to: deep breathing; lying-to-standing test; and the Valsalva maneuver) were used to diagnose CAN in 302 persons with T1D. More than one pathological CARTs defined the CAN diagnosis. Serum metabolomics and lipidomic profiles were analyzed with two complementary non-targeted mass-spectrometry methods. Cross-sectional associations between metabolites and CAN were assessed by linear regression models adjusted for relevant confounders. RESULTS Participants were median (IQR) aged 55(49, 63) years, 48% males with diabetes duration 39(32, 47) years, HbA1c 63(55,69) mmol/mol and 34% had CAN. A total of 75 metabolites and 106 lipids were analyzed. In crude models, the CAN diagnosis was associated with higher levels of hydroxy fatty acids (2,4- and 3,4-dihydroxybutanoic acids, 4-deoxytetronic acid), creatinine, sugar derivates (ribitol, ribonic acid, myo-inositol), citric acid, glycerol, phenols, phosphatidylcholines and lower levels of free fatty acids and the amino acid methionine (p<0.05). Upon adjustment, positive associations with the CAN diagnoses were retained for hydroxy fatty acids, tricarboxylic acid (TCA) cycle-based sugar derivates, citric acid, and phenols (P<0.05). CONCLUSION Metabolic pathways, including the TCA cycle, hydroxy fatty acids, phosphatidylcholines and sugar derivatives are associated with the CAN diagnosis in T1D. These pathway may be part of the pathogeneses leading to CAN and may be modifiable risk factors for the complication.
Collapse
Affiliation(s)
- Christian S. Hansen
- Complications Research, Steno Diabetes Center Copenhagen, Herlev, Denmark
- *Correspondence: Christian S. Hansen,
| | - Tommi Suvitaival
- Complications Research, Steno Diabetes Center Copenhagen, Herlev, Denmark
| | - Simone Theilade
- Complications Research, Steno Diabetes Center Copenhagen, Herlev, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
- The Department of Medicine, Herlev-Gentofte Hospital, Copenhagen, Denmark
| | - Ismo Mattila
- Complications Research, Steno Diabetes Center Copenhagen, Herlev, Denmark
| | - Maria Lajer
- Complications Research, Steno Diabetes Center Copenhagen, Herlev, Denmark
| | - Kajetan Trošt
- Complications Research, Steno Diabetes Center Copenhagen, Herlev, Denmark
| | - Linda Ahonen
- Complications Research, Steno Diabetes Center Copenhagen, Herlev, Denmark
| | - Tine W. Hansen
- Complications Research, Steno Diabetes Center Copenhagen, Herlev, Denmark
| | | | - Peter Rossing
- Complications Research, Steno Diabetes Center Copenhagen, Herlev, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Tarunveer S. Ahluwalia
- Complications Research, Steno Diabetes Center Copenhagen, Herlev, Denmark
- The Bioinformatics Center, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
31
|
Huang G, Li M, Li Y, Mao Y. OUP accepted manuscript. Lab Med 2022; 53:545-551. [PMID: 35748329 DOI: 10.1093/labmed/lmac041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Affiliation(s)
- Guoqing Huang
- Department of Endocrinology, The Affiliated Hospital of Medical School, Ningbo University, Ningbo, China
- School of Medicine, Ningbo University, Ningbo, China
| | - Mingcai Li
- School of Medicine, Ningbo University, Ningbo, China
| | - Yan Li
- Department of Endocrinology, The Affiliated Hospital of Medical School, Ningbo University, Ningbo, China
- School of Medicine, Ningbo University, Ningbo, China
| | - Yushan Mao
- Department of Endocrinology, The Affiliated Hospital of Medical School, Ningbo University, Ningbo, China
| |
Collapse
|
32
|
Wang X, Liu J, Hui X, Song Y. Metabolomics Applied to Cord Serum in Preeclampsia Newborns: Implications for Neonatal Outcomes. Front Pediatr 2022; 10:869381. [PMID: 35547553 PMCID: PMC9082809 DOI: 10.3389/fped.2022.869381] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 03/11/2022] [Indexed: 11/17/2022] Open
Abstract
Preeclampsia (PE) is one of the leading causes of maternal and perinatal morbidity and mortality. However, it is still uncertain how PE affects neonate metabolism. We conducted an untargeted metabolomics analysis of cord blood to explore the metabolic changes in PE neonates. Umbilical cord serum samples from neonates with preeclampsia (n = 29) and non-preeclampsia (non-PE) (n = 32) pregnancies were analyzed using the UHPLC-QE-MS metabolomic platform. Different metabolites were screened, and pathway analysis was conducted. A subgroup analysis was performed among PE neonates to compare the metabolome between appropriate-for-gestational-age infants (n = 21) and small-for-gestational-age (SGA) infants (n = 8). A total of 159 different metabolites were detected in PE and non-PE neonates. Creatinine, N4-acetylcytidine, sphingomyelin (D18:1/16:0), pseudouridine, uric acid, and indolelactic acid were the most significant differential metabolites in the cord serum of PE neonates. Differential metabolite levels were elevated in PE neonates and were involved in the following metabolic pathways: glycine, serine, and threonine metabolism; sphingolipid, glyoxylate, and dicarboxylate metabolism; and arginine biosynthesis. In PE neonates, SGA neonates showed increased levels of hexacosanoyl carnitine and decreased abundance of 3-hydroxybutyric acid and 3-sulfinoalanine. Taurine-related metabolism and ketone body-related pathways were mainly affected. Based on the UHPLC-QE-MS metabolomics analysis, we identified the metabolic profiles of PE and SGA neonates. The abundance of metabolites related to certain amino acid, sphingolipid, and energy metabolism increased in the umbilical cord serum of PE neonates.
Collapse
Affiliation(s)
- Xiaoxu Wang
- Department of Obstetrics and Gynecology, National Clinical Research Centre for Obstetric and Gynecologic Diseases, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jieying Liu
- State Key Laboratory of Complex Severe and Rare Diseases, Medical Research Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiangyi Hui
- State Key Laboratory of Complex Severe and Rare Diseases, Medical Research Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yingna Song
- Department of Obstetrics and Gynecology, National Clinical Research Centre for Obstetric and Gynecologic Diseases, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
33
|
Mutter S, Valo E, Aittomäki V, Nybo K, Raivonen L, Thorn LM, Forsblom C, Sandholm N, Würtz P, Groop PH. Urinary metabolite profiling and risk of progression of diabetic nephropathy in 2670 individuals with type 1 diabetes. Diabetologia 2022; 65:140-149. [PMID: 34686904 PMCID: PMC8660744 DOI: 10.1007/s00125-021-05584-3] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 08/11/2021] [Indexed: 12/25/2022]
Abstract
AIMS/HYPOTHESIS This prospective, observational study examines associations between 51 urinary metabolites and risk of progression of diabetic nephropathy in individuals with type 1 diabetes by employing an automated NMR metabolomics technique suitable for large-scale urine sample collections. METHODS We collected 24-h urine samples for 2670 individuals with type 1 diabetes from the Finnish Diabetic Nephropathy study and measured metabolite concentrations by NMR. Individuals were followed up for 9.0 ± 5.0 years until their first sign of progression of diabetic nephropathy, end-stage kidney disease or study end. Cox regressions were performed on the entire study population (overall progression), on 1999 individuals with normoalbuminuria and 347 individuals with macroalbuminuria at baseline. RESULTS Seven urinary metabolites were associated with overall progression after adjustment for baseline albuminuria and chronic kidney disease stage (p < 8 × 10-4): leucine (HR 1.47 [95% CI 1.30, 1.66] per 1-SD creatinine-scaled metabolite concentration), valine (1.38 [1.22, 1.56]), isoleucine (1.33 [1.18, 1.50]), pseudouridine (1.25 [1.11, 1.42]), threonine (1.27 [1.11, 1.46]) and citrate (0.84 [0.75, 0.93]). 2-Hydroxyisobutyrate was associated with overall progression (1.30 [1.16, 1.45]) and also progression from normoalbuminuria (1.56 [1.25, 1.95]). Six amino acids and pyroglutamate were associated with progression from macroalbuminuria. CONCLUSIONS/INTERPRETATION Branched-chain amino acids and other urinary metabolites were associated with the progression of diabetic nephropathy on top of baseline albuminuria and chronic kidney disease. We found differences in associations for overall progression and progression from normo- and macroalbuminuria. These novel discoveries illustrate the utility of analysing urinary metabolites in entire population cohorts.
Collapse
Affiliation(s)
- Stefan Mutter
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Helsinki, Finland
- Department of Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Erkka Valo
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Helsinki, Finland
- Department of Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | | | | | | | - Lena M Thorn
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Helsinki, Finland
- Department of Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Department of General Practice and Primary Health Care, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Carol Forsblom
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Helsinki, Finland
- Department of Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Niina Sandholm
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Helsinki, Finland
- Department of Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | | | - Per-Henrik Groop
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Helsinki, Finland.
- Department of Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland.
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
- Department of Diabetes, Central Clinical School, Monash University, Melbourne, VIC, Australia.
| |
Collapse
|
34
|
Green R, Lord J, Xu J, Maddock J, Kim M, Dobson R, Legido-Quigley C, Wong A, Richards M, Proitsi P. Metabolic correlates of late midlife cognitive outcomes: findings from the 1946 British Birth Cohort. Brain Commun 2021; 4:fcab291. [PMID: 35187482 PMCID: PMC8853724 DOI: 10.1093/braincomms/fcab291] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 09/17/2021] [Accepted: 12/10/2021] [Indexed: 11/14/2022] Open
Abstract
Investigating associations between metabolites and late midlife cognitive function could reveal potential markers and mechanisms relevant to early dementia. Here, we systematically explored the metabolic correlates of cognitive outcomes measured across the seventh decade of life, while untangling influencing life course factors. Using levels of 1019 metabolites profiled by liquid chromatography-mass spectrometry (age 60-64), we evaluated relationships between metabolites and cognitive outcomes in the British 1946 Birth Cohort (N = 1740). We additionally conducted pathway and network analyses to allow for greater insight into potential mechanisms, and sequentially adjusted for life course factors across four models, including sex and blood collection (Model 1), Model 1 + body mass index and lipid medication (Model 2), Model 2 + social factors and childhood cognition (Model 3) and Model 3 + lifestyle influences (Model 4). After adjusting for multiple tests, 155 metabolites, 10 pathways and 5 network modules were associated with cognitive outcomes. Of the 155, 35 metabolites were highly connected in their network module (termed 'hub' metabolites), presenting as promising marker candidates. Notably, we report relationships between a module comprised of acylcarnitines and processing speed which remained robust to life course adjustment, revealing palmitoylcarnitine (C16) as a hub (Model 4: β = -0.10, 95% confidence interval = -0.15 to -0.052, P = 5.99 × 10-5). Most associations were sensitive to adjustment for social factors and childhood cognition; in the final model, four metabolites remained after multiple testing correction, and 80 at P < 0.05. Two modules demonstrated associations that were partly or largely attenuated by life course factors: one enriched in modified nucleosides and amino acids (overall attenuation = 39.2-55.5%), and another in vitamin A and C metabolites (overall attenuation = 68.6-92.6%). Our other findings, including a module enriched in sphingolipid pathways, were entirely explained by life course factors, particularly childhood cognition and education. Using a large birth cohort study with information across the life course, we highlighted potential metabolic mechanisms associated with cognitive function in late midlife, suggesting marker candidates and life course relationships for further study.
Collapse
Affiliation(s)
- Rebecca Green
- Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, UK
- UK National Institute for Health Research (NIHR) Maudsley Biomedical Research Centre, South London and Maudsley Trust, London, UK
| | - Jodie Lord
- Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, UK
| | - Jin Xu
- Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, UK
- Institute of Pharmaceutical Science, King’s College London, London, UK
| | - Jane Maddock
- MRC Unit for Lifelong Health & Ageing at UCL, University College London, London, UK
| | - Min Kim
- Steno Diabetes Center Copenhagen, Gentofte, Denmark
| | - Richard Dobson
- Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, UK
- UK National Institute for Health Research (NIHR) Maudsley Biomedical Research Centre, South London and Maudsley Trust, London, UK
- Health Data Research UK London, University College London, London, UK
- NIHR Biomedical Research Centre at University College London, Hospitals NHS Foundation Trust, London, UK
| | - Cristina Legido-Quigley
- Institute of Pharmaceutical Science, King’s College London, London, UK
- Steno Diabetes Center Copenhagen, Gentofte, Denmark
| | - Andrew Wong
- MRC Unit for Lifelong Health & Ageing at UCL, University College London, London, UK
| | - Marcus Richards
- MRC Unit for Lifelong Health & Ageing at UCL, University College London, London, UK
| | - Petroula Proitsi
- Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, UK
| |
Collapse
|
35
|
Cui H, Shu S, Li Y, Yan X, Chen X, Chen Z, Hu Y, Chang Y, Hu Z, Wang X, Song J. Plasma Metabolites-Based Prediction in Cardiac Surgery-Associated Acute Kidney Injury. J Am Heart Assoc 2021; 10:e021825. [PMID: 34719239 PMCID: PMC8751958 DOI: 10.1161/jaha.121.021825] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Background Cardiac surgery–associated acute kidney injury (CSA‐AKI) is a common postoperative complication following cardiac surgery. Currently, there are no reliable methods for the early prediction of CSA‐AKI in hospitalized patients. This study developed and evaluated the diagnostic use of metabolomics‐based biomarkers in patients with CSA‐AKI. Methods and Results A total of 214 individuals (122 patients with acute kidney injury [AKI], 92 patients without AKI as controls) were enrolled in this study. Plasma samples were analyzed by liquid chromatography tandem mass spectrometry using untargeted and targeted metabolomic approaches. Time‐dependent effects of selected metabolites were investigated in an AKI swine model. Multiple machine learning algorithms were used to identify plasma metabolites positively associated with CSA‐AKI. Metabolomic analyses from plasma samples taken within 24 hours following cardiac surgery were useful for distinguishing patients with AKI from controls without AKI. Gluconic acid, fumaric acid, and pseudouridine were significantly upregulated in patients with AKI. A random forest model constructed with selected clinical parameters and metabolites exhibited excellent discriminative ability (area under curve, 0.939; 95% CI, 0.879–0.998). In the AKI swine model, plasma levels of the 3 discriminating metabolites increased in a time‐dependent manner (R2, 0.480–0.945). Use of this AKI predictive model was then confirmed in the validation cohort (area under curve, 0.972; 95% CI, 0.947–0.996). The predictive model remained robust when tested in a subset of patients with early‐stage AKI in the validation cohort (area under curve, 0.943; 95% CI, 0.883–1.000). Conclusions High‐resolution metabolomics is sufficiently powerful for developing novel biomarkers. Plasma levels of 3 metabolites were useful for the early identification of CSA‐AKI.
Collapse
Affiliation(s)
- Hao Cui
- The Cardiomyopathy Research Group State Key Laboratory of Cardiovascular Disease Fuwai HospitalNational Center for Cardiovascular DiseasesChinese Academy of Medical Sciences and Peking Union Medical College Beijing China
| | - Songren Shu
- The Cardiomyopathy Research Group State Key Laboratory of Cardiovascular Disease Fuwai HospitalNational Center for Cardiovascular DiseasesChinese Academy of Medical Sciences and Peking Union Medical College Beijing China
| | - Yuan Li
- Department of Cardiovascular Surgery Fuwai HospitalNational Center for Cardiovascular DiseasesChinese Academy of Medical Sciences and Peking Union Medical College Beijing China
| | - Xin Yan
- The Cardiomyopathy Research Group State Key Laboratory of Cardiovascular Disease Fuwai HospitalNational Center for Cardiovascular DiseasesChinese Academy of Medical Sciences and Peking Union Medical College Beijing China
| | - Xiao Chen
- The Cardiomyopathy Research Group State Key Laboratory of Cardiovascular Disease Fuwai HospitalNational Center for Cardiovascular DiseasesChinese Academy of Medical Sciences and Peking Union Medical College Beijing China
| | - Zujun Chen
- Surgical Intensive Care Unit Fuwai HospitalNational Center for Cardiovascular DiseasesChinese Academy of Medical Sciences and Peking Union Medical College Beijing China
| | - Yuxuan Hu
- Capital Normal University High School Beijing China
| | - Yuan Chang
- The Cardiomyopathy Research Group State Key Laboratory of Cardiovascular Disease Fuwai HospitalNational Center for Cardiovascular DiseasesChinese Academy of Medical Sciences and Peking Union Medical College Beijing China
| | - Zhenliang Hu
- The Cardiomyopathy Research Group State Key Laboratory of Cardiovascular Disease Fuwai HospitalNational Center for Cardiovascular DiseasesChinese Academy of Medical Sciences and Peking Union Medical College Beijing China
| | - Xin Wang
- Department of Cardiovascular Surgery Fuwai HospitalNational Center for Cardiovascular DiseasesChinese Academy of Medical Sciences and Peking Union Medical College Beijing China.,Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials Center for Cardiovascular Experimental Study and Evaluation Fuwai HospitalNational Center for Cardiovascular DiseasesChinese Academy of Medical Sciences and Peking Union Medical College Beijing China
| | - Jiangping Song
- The Cardiomyopathy Research Group State Key Laboratory of Cardiovascular Disease Fuwai HospitalNational Center for Cardiovascular DiseasesChinese Academy of Medical Sciences and Peking Union Medical College Beijing China
| |
Collapse
|
36
|
Bajaj JS, Garcia-Tsao G, Reddy KR, O’Leary JG, Vargas HE, Lai JC, Kamath PS, Tandon P, Subramanian RM, Thuluvath P, Fagan A, Sehrawat T, de la Rosa Rodriguez R, Thacker LR, Wong F. Admission Urinary and Serum Metabolites Predict Renal Outcomes in Hospitalized Patients With Cirrhosis. Hepatology 2021; 74:2699-2713. [PMID: 34002868 PMCID: PMC9338693 DOI: 10.1002/hep.31907] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Revised: 04/30/2021] [Accepted: 05/04/2021] [Indexed: 12/12/2022]
Abstract
BACKGROUND AND AIMS Acute kidney injury (AKI) has a poor prognosis in cirrhosis. Given the variability of creatinine, the prediction of AKI and dialysis by other markers is needed. The aim of this study is to determine the role of serum and urine metabolomics in the prediction of AKI and dialysis in an inpatient cirrhosis cohort. APPROACH AND RESULTS Inpatients with cirrhosis from 11 North American Consortium of End-stage Liver Disease centers who provided admission serum/urine when they were AKI and dialysis-free were included. Analysis of covariance adjusted for demographics, infection, and cirrhosis severity was performed to identify metabolites that differed among patients (1) who developed AKI or not; (2) required dialysis or not; and/pr (3) within AKI subgroups who needed dialysis or not. We performed random forest and AUC analyses to identify specific metabolite(s) associated with outcomes. Logistic regression with clinical variables with/without metabolites was performed. A total of 602 patients gave serum (218 developed AKI, 80 needed dialysis) and 435 gave urine (164 developed AKI, 61 needed dialysis). For AKI prediction, clinical factor-adjusted AUC was 0.91 for serum and 0.88 for urine. Major metabolites such as uremic toxins (2,3-dihydroxy-5-methylthio-4-pentenoic acid [DMTPA], N2N2dimethylguanosine, uridine/pseudouridine) and tryptophan/tyrosine metabolites (kynunerate, 8-methoxykyunerate, quinolinate) were higher in patients who developed AKI. For dialysis prediction, clinical factor-adjusted AUC was 0.93 for serum and 0.91 for urine. Similar metabolites as AKI were altered here. For dialysis prediction in those with AKI, the AUC was 0.81 and 0.79 for serum/urine. Lower branched-chain amino-acid (BCAA) metabolites but higher cysteine, tryptophan, glutamate, and DMTPA were seen in patients with AKI needing dialysis. Serum/urine metabolites were additive to clinical variables for all outcomes. CONCLUSIONS Specific admission urinary and serum metabolites were significantly additive to clinical variables to predict AKI development and dialysis initiation in inpatients with cirrhosis. These observations can potentially facilitate earlier initiation of renoprotective measures.
Collapse
Affiliation(s)
- Jasmohan S. Bajaj
- Virginia Commonwealth University and Central Virginia Veterans Healthcare System, Richmond, VA
| | | | | | | | | | | | | | | | | | | | - Andrew Fagan
- Virginia Commonwealth University and Central Virginia Veterans Healthcare System, Richmond, VA
| | | | | | - Leroy R. Thacker
- Virginia Commonwealth University and Central Virginia Veterans Healthcare System, Richmond, VA
| | | |
Collapse
|
37
|
Jin Q, Ma RCW. Metabolomics in Diabetes and Diabetic Complications: Insights from Epidemiological Studies. Cells 2021; 10:cells10112832. [PMID: 34831057 PMCID: PMC8616415 DOI: 10.3390/cells10112832] [Citation(s) in RCA: 113] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Revised: 10/11/2021] [Accepted: 10/13/2021] [Indexed: 12/18/2022] Open
Abstract
The increasing prevalence of diabetes and its complications, such as cardiovascular and kidney disease, remains a huge burden globally. Identification of biomarkers for the screening, diagnosis, and prognosis of diabetes and its complications and better understanding of the molecular pathways involved in the development and progression of diabetes can facilitate individualized prevention and treatment. With the advancement of analytical techniques, metabolomics can identify and quantify multiple biomarkers simultaneously in a high-throughput manner. Providing information on underlying metabolic pathways, metabolomics can further identify mechanisms of diabetes and its progression. The application of metabolomics in epidemiological studies have identified novel biomarkers for type 2 diabetes (T2D) and its complications, such as branched-chain amino acids, metabolites of phenylalanine, metabolites involved in energy metabolism, and lipid metabolism. Metabolomics have also been applied to explore the potential pathways modulated by medications. Investigating diabetes using a systems biology approach by integrating metabolomics with other omics data, such as genetics, transcriptomics, proteomics, and clinical data can present a comprehensive metabolic network and facilitate causal inference. In this regard, metabolomics can deepen the molecular understanding, help identify potential therapeutic targets, and improve the prevention and management of T2D and its complications. The current review focused on metabolomic biomarkers for kidney and cardiovascular disease in T2D identified from epidemiological studies, and will also provide a brief overview on metabolomic investigations for T2D.
Collapse
Affiliation(s)
- Qiao Jin
- Department of Medicine and Therapeutics, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, China;
| | - Ronald Ching Wan Ma
- Department of Medicine and Therapeutics, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, China;
- Laboratory for Molecular Epidemiology in Diabetes, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
- Hong Kong Institute of Diabetes and Obesity, The Chinese University of Hong Kong, Hong Kong, China
- Chinese University of Hong Kong-Shanghai Jiao Tong University Joint Research Centre in Diabetes Genomics and Precision Medicine, The Chinese University of Hong Kong, Hong Kong, China
- Correspondence: ; Fax: +852-26373852
| |
Collapse
|
38
|
Human age-declined saliva metabolic markers determined by LC-MS. Sci Rep 2021; 11:18135. [PMID: 34518599 PMCID: PMC8437986 DOI: 10.1038/s41598-021-97623-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Accepted: 08/26/2021] [Indexed: 01/20/2023] Open
Abstract
Metabolites in human biofluids reflect individual physiological states influenced by various factors. Using liquid chromatography-mass spectrometry (LC–MS), we conducted non-targeted, non-invasive metabolomics using saliva of 27 healthy volunteers in Okinawa, comprising 13 young (30 ± 3 year) and 14 elderly (76 ± 4 year) subjects. Few studies have comprehensively identified age-dependent changes in salivary metabolites. Among 99 salivary metabolites, 21 were statistically age-related. All of the latter decline in abundance with advancing age, except ATP, which increased 1.96-fold in the elderly, possibly due to reduced ATP consumption. Fourteen age-linked and highly correlated compounds function in a metabolic network involving the pentose-phosphate pathway, glycolysis/gluconeogenesis, amino acids, and purines/pyrimidines nucleobases. The remaining seven less strongly correlated metabolites, include ATP, anti-oxidation-related glutathione disulfide, muscle-related acetyl-carnosine, N-methyl-histidine, creatinine, RNA-related dimethyl-xanthine and N-methyl-adenosine. In addition, glutamate and N-methyl-histidine are related to taste, so their decline suggests that the elderly lose some ability to taste. Reduced redox metabolism and muscle activity are suggested by changes in glutathione and acetyl-carnosine. These age-linked salivary metabolites together illuminate a metabolic network that reflects a decline of oral functions during human aging.
Collapse
|
39
|
Minakata S, Manabe S, Inai Y, Ikezaki M, Nishitsuji K, Ito Y, Ihara Y. Protein C-Mannosylation and C-Mannosyl Tryptophan in Chemical Biology and Medicine. Molecules 2021; 26:molecules26175258. [PMID: 34500691 PMCID: PMC8433626 DOI: 10.3390/molecules26175258] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 08/25/2021] [Accepted: 08/26/2021] [Indexed: 12/25/2022] Open
Abstract
C-Mannosylation is a post-translational modification of proteins in the endoplasmic reticulum. Monomeric α-mannose is attached to specific Trp residues at the first Trp in the Trp-x-x-Trp/Cys (W-x-x-W/C) motif of substrate proteins, by the action of C-mannosyltransferases, DPY19-related gene products. The acceptor substrate proteins are included in the thrombospondin type I repeat (TSR) superfamily, cytokine receptor type I family, and others. Previous studies demonstrated that C-mannosylation plays critical roles in the folding, sorting, and/or secretion of substrate proteins. A C-mannosylation-defective gene mutation was identified in humans as the disease-associated variant affecting a C-mannosylation motif of W-x-x-W of ADAMTSL1, which suggests the involvement of defects in protein C-mannosylation in human diseases such as developmental glaucoma, myopia, and/or retinal defects. On the other hand, monomeric C-mannosyl Trp (C-Man-Trp), a deduced degradation product of C-mannosylated proteins, occurs in cells and extracellular fluids. Several studies showed that the level of C-Man-Trp is upregulated in blood of patients with renal dysfunction, suggesting that the metabolism of C-Man-Trp may be involved in human kidney diseases. Together, protein C-mannosylation is considered to play important roles in the biosynthesis and functions of substrate proteins, and the altered regulation of protein C-manosylation may be involved in the pathophysiology of human diseases. In this review, we consider the biochemical and biomedical knowledge of protein C-mannosylation and C-Man-Trp, and introduce recent studies concerning their significance in biology and medicine.
Collapse
Affiliation(s)
- Shiho Minakata
- Department of Biochemistry, Wakayama Medical University, 811-1 Kimiidera, Wakayama, Wakayama 641-0012, Japan; (S.M.); (Y.I.); (M.I.); (K.N.)
| | - Shino Manabe
- Pharmaceutical Department, The Institute of Medicinal Chemistry, Hoshi University, 2-4-41 Ebara, Shinagawa, Tokyo 142-8501, Japan;
- Research Center for Pharmaceutical Development, Graduate School of Pharmaceutical Science & Faculty of Pharmaceutical Sciences, Tohoku University, 6-3 Aoba, Sendai, Miyagi 980-8578, Japan
| | - Yoko Inai
- Department of Biochemistry, Wakayama Medical University, 811-1 Kimiidera, Wakayama, Wakayama 641-0012, Japan; (S.M.); (Y.I.); (M.I.); (K.N.)
| | - Midori Ikezaki
- Department of Biochemistry, Wakayama Medical University, 811-1 Kimiidera, Wakayama, Wakayama 641-0012, Japan; (S.M.); (Y.I.); (M.I.); (K.N.)
| | - Kazuchika Nishitsuji
- Department of Biochemistry, Wakayama Medical University, 811-1 Kimiidera, Wakayama, Wakayama 641-0012, Japan; (S.M.); (Y.I.); (M.I.); (K.N.)
| | - Yukishige Ito
- Department of Chemistry, Graduate School of Science, Osaka University, 1-1 Machikaneyama, Toyonaka, Osaka 560-0043, Japan;
- RIKEN Cluster for Pioneering Research, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Yoshito Ihara
- Department of Biochemistry, Wakayama Medical University, 811-1 Kimiidera, Wakayama, Wakayama 641-0012, Japan; (S.M.); (Y.I.); (M.I.); (K.N.)
- Correspondence: ; Tel.: +81-73-441-0628
| |
Collapse
|
40
|
Denburg MR, Xu Y, Abraham AG, Coresh J, Chen J, Grams ME, Feldman HI, Kimmel PL, Rebholz CM, Rhee EP, Vasan RS, Warady BA, Furth SL. Metabolite Biomarkers of CKD Progression in Children. Clin J Am Soc Nephrol 2021; 16:1178-1189. [PMID: 34362785 PMCID: PMC8455058 DOI: 10.2215/cjn.00220121] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 06/17/2021] [Indexed: 02/04/2023]
Abstract
BACKGROUND AND OBJECTIVES Metabolomics facilitates the discovery of biomarkers and potential therapeutic targets for CKD progression. DESIGN, SETTING, PARTICIPANTS, & MEASUREMENTS We evaluated an untargeted metabolomics quantification of stored plasma samples from 645 Chronic Kidney Disease in Children (CKiD) participants. Metabolites were standardized and logarithmically transformed. Cox proportional hazards regression examined the association between 825 nondrug metabolites and progression to the composite outcome of KRT or 50% reduction of eGFR, adjusting for age, sex, race, body mass index, hypertension, glomerular versus nonglomerular diagnosis, proteinuria, and baseline eGFR. Stratified analyses were performed within subgroups of glomerular/nonglomerular diagnosis and baseline eGFR. RESULTS Baseline characteristics were 391 (61%) male; median age 12 years; median eGFR 54 ml/min per 1.73 m2; 448 (69%) nonglomerular diagnosis. Over a median follow-up of 4.8 years, 209 (32%) participants developed the composite outcome. Unique association signals were identified in subgroups of baseline eGFR. Among participants with baseline eGFR ≥60 ml/min per 1.73 m2, two-fold higher levels of seven metabolites were significantly associated with higher hazards of KRT/halving of eGFR events: three involved in purine and pyrimidine metabolism (N6-carbamoylthreonyladenosine, hazard ratio, 16; 95% confidence interval, 4 to 60; 5,6-dihydrouridine, hazard ratio, 17; 95% confidence interval, 5 to 55; pseudouridine, hazard ratio, 39; 95% confidence interval, 8 to 200); two amino acids, C-glycosyltryptophan, hazard ratio, 24; 95% confidence interval 6 to 95 and lanthionine, hazard ratio, 3; 95% confidence interval, 2 to 5; the tricarboxylic acid cycle intermediate 2-methylcitrate/homocitrate, hazard ratio, 4; 95% confidence interval, 2 to 7; and gulonate, hazard ratio, 10; 95% confidence interval, 3 to 29. Among those with baseline eGFR <60 ml/min per 1.73 m2, a higher level of tetrahydrocortisol sulfate was associated with lower risk of progression (hazard ratio, 0.8; 95% confidence interval, 0.7 to 0.9). CONCLUSIONS Untargeted plasma metabolomic profiling facilitated discovery of novel metabolite associations with CKD progression in children that were independent of established clinical predictors and highlight the role of select biologic pathways.
Collapse
Affiliation(s)
- Michelle R. Denburg
- Division of Nephrology, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania,Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania,Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania,Center for Pediatric Clinical Effectiveness, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Yunwen Xu
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Alison G. Abraham
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Josef Coresh
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Jingsha Chen
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Morgan E. Grams
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Harold I. Feldman
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania,Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Paul L. Kimmel
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland
| | - Casey M. Rebholz
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Eugene P. Rhee
- Department of Medicine, Massachusetts General Hospital, Department of Medicine, Harvard University, Boston, Massachusetts
| | - Ramachandran S. Vasan
- Department of Medicine, Boston University School of Medicine, Boston University School of Public Health, and Boston University Center for Computing and Data Science, Boston, Massachusetts
| | - Bradley A. Warady
- Children’s Mercy Kansas City, Department of Pediatrics, University of Missouri-Kansas City School of Medicine, Kansas City, Missouri
| | - Susan L. Furth
- Division of Nephrology, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania,Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania,Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | | |
Collapse
|
41
|
Zheng S, Zhang X, Li Z, Hoene M, Fritsche L, Zheng F, Li Q, Fritsche A, Peter A, Lehmann R, Zhao X, Xu G. Systematic, Modifying Group-Assisted Strategy Expanding Coverage of Metabolite Annotation in Liquid Chromatography-Mass Spectrometry-Based Nontargeted Metabolomics Studies. Anal Chem 2021; 93:10916-10924. [PMID: 34328315 DOI: 10.1021/acs.analchem.1c01715] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
From microbes to human beings, nontargeted metabolic profiling by liquid chromatography (LC)-mass spectrometry (MS) has been commonly used to investigate metabolic alterations. Still, a major challenge is the annotation of metabolites from thousands of detected features. The aim of our research was to go beyond coverage of metabolite annotation in common nontargeted metabolomics studies by an integrated multistep strategy applying data-dependent acquisition (DDA)-based ultrahigh-performance liquid chromatography (UHPLC)-high-resolution mass spectrometry (HRMS) analysis followed by comprehensive neutral loss matches for characteristic metabolite modifications and database searches in a successive manner. Using pooled human urine as a model sample for method establishment, we found 22% of the detected compounds having modifying structures. Major types of metabolite modifications in urine were glucuronidation (33%), sulfation (20%), and acetylation (6%). Among the 383 annotated metabolites, 100 were confirmed by standard compounds and 50 modified metabolites not present in common databases such as human metabolite database (HMDB) and Kyoto Encyclopedia of Genes and Genomes (KEGG) were structurally elucidated. Practicability was tested by the investigation of urines from pregnant women diagnosed with gestational diabetes mellitus vs healthy controls. Overall, 83 differential metabolites were annotated and 67% of them were modified metabolites including five previously unreported compounds. To conclude, the systematic modifying group-assisted strategy can be taken as a useful tool to extend the number of annotated metabolites in biological and biomedical nontargeted studies.
Collapse
Affiliation(s)
- Sijia Zheng
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiuqiong Zhang
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zaifang Li
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Miriam Hoene
- Institute for Clinical Chemistry and Pathobiochemistry, University Hospital Tuebingen, Hoppe-Seyler-Str. 3, Tuebingen 72076, Germany
| | - Louise Fritsche
- German Center for Diabetes Research (DZD), Tuebingen 72076, Germany.,Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Zentrum Muenchen at the University of Tuebingen, Tuebingen 72076, Germany.,Internal Medicine 4, University Hospital Tuebingen, Otfried-Mueller-Str. 10, Tuebingen 72076, Germany
| | - Fujian Zheng
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qi Li
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
| | - Andreas Fritsche
- German Center for Diabetes Research (DZD), Tuebingen 72076, Germany.,Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Zentrum Muenchen at the University of Tuebingen, Tuebingen 72076, Germany.,Internal Medicine 4, University Hospital Tuebingen, Otfried-Mueller-Str. 10, Tuebingen 72076, Germany
| | - Andreas Peter
- Institute for Clinical Chemistry and Pathobiochemistry, University Hospital Tuebingen, Hoppe-Seyler-Str. 3, Tuebingen 72076, Germany.,German Center for Diabetes Research (DZD), Tuebingen 72076, Germany.,Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Zentrum Muenchen at the University of Tuebingen, Tuebingen 72076, Germany
| | - Rainer Lehmann
- Institute for Clinical Chemistry and Pathobiochemistry, University Hospital Tuebingen, Hoppe-Seyler-Str. 3, Tuebingen 72076, Germany.,German Center for Diabetes Research (DZD), Tuebingen 72076, Germany.,Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Zentrum Muenchen at the University of Tuebingen, Tuebingen 72076, Germany
| | - Xinjie Zhao
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
| | - Guowang Xu
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
42
|
Macioszek S, Wawrzyniak R, Kranz A, Kordalewska M, Struck-Lewicka W, Dudzik D, Biesemans M, Maternik M, Żurowska AM, Markuszewski MJ. Comprehensive Metabolic Signature of Renal Dysplasia in Children. A Multiplatform Metabolomics Concept. Front Mol Biosci 2021; 8:665661. [PMID: 34395519 PMCID: PMC8358436 DOI: 10.3389/fmolb.2021.665661] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 07/19/2021] [Indexed: 11/16/2022] Open
Abstract
Renal dysplasia is a severe congenital abnormality of the kidney parenchyma, which is an important cause of end-stage renal failure in childhood and early adulthood. The diagnosis of renal dysplasia relies on prenatal or postnatal ultrasounds as children show no specific clinical symptoms before chronic kidney disease develops. Prompt diagnosis is important in terms of early introduction of nephroprotection therapy and improved long-term prognosis. Metabolomics was applied to study children with renal dysplasia to provide insight into the changes in biochemical pathways underlying its pathology and in search of early indicators for facilitated diagnosis. The studied cohort consisted of 72 children, 39 with dysplastic kidneys and 33 healthy controls. All subjects underwent comprehensive urine metabolic profiling with the use of gas chromatography and liquid chromatography coupled to mass spectrometry, with two complementary separation modes of the latter. Univariate and multivariate statistical calculations identified a total of nineteen metabolites, differentiating the compared cohorts, independent of their estimated glomerular filtration rate. Seven acylcarnitines, xanthine, and glutamine were downregulated in the urine of renal dysplasia patients. Conversely, renal dysplasia was associated with higher urinary levels of dimethylguanosine, threonic acid or glyceric acid. This is the first metabolomic study of subjects with renal dysplasia. The authors define a characteristic urine metabolic signature in children with dysplastic kidneys, irrespective of renal function, linking the condition with altered fatty acid oxidation, amino acid and purine metabolisms.
Collapse
Affiliation(s)
- Szymon Macioszek
- Department of Biopharmaceutics and Pharmacodynamics, Medical University of Gdańsk, Gdańsk, Poland
| | - Renata Wawrzyniak
- Department of Biopharmaceutics and Pharmacodynamics, Medical University of Gdańsk, Gdańsk, Poland
| | - Anna Kranz
- Department of Pediatrics, Nephrology and Hypertension, Medical University of Gdańsk, Gdańsk, Poland
| | - Marta Kordalewska
- Department of Biopharmaceutics and Pharmacodynamics, Medical University of Gdańsk, Gdańsk, Poland
| | - Wiktoria Struck-Lewicka
- Department of Biopharmaceutics and Pharmacodynamics, Medical University of Gdańsk, Gdańsk, Poland
| | - Danuta Dudzik
- Department of Biopharmaceutics and Pharmacodynamics, Medical University of Gdańsk, Gdańsk, Poland
| | - Margot Biesemans
- Department of Biopharmaceutics and Pharmacodynamics, Medical University of Gdańsk, Gdańsk, Poland
| | - Michał Maternik
- Department of Pediatrics, Nephrology and Hypertension, Medical University of Gdańsk, Gdańsk, Poland
| | | | - Michał J Markuszewski
- Department of Biopharmaceutics and Pharmacodynamics, Medical University of Gdańsk, Gdańsk, Poland
| |
Collapse
|
43
|
Barutta F, Bellini S, Canepa S, Durazzo M, Gruden G. Novel biomarkers of diabetic kidney disease: current status and potential clinical application. Acta Diabetol 2021; 58:819-830. [PMID: 33528734 DOI: 10.1007/s00592-020-01656-9] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 12/09/2020] [Indexed: 12/12/2022]
Abstract
Diabetic kidney disease (DKD) is a leading cause of end-stage renal disease (ESRD). Although both albuminuria and glomerular filtration rate (GFR) are well-established diagnostic/prognostic biomarkers of DKD, they have important limitations. There is, thus, increasing quest to find novel biomarkers to identify the disease in an early stage and to improve risk stratification. In this review, we will outline the major pitfalls of currently available markers, describe promising novel biomarkers, and discuss their potential clinical relevance. In particular, we will focus on the importance of recent advancements in multi-omic technologies in the discovery of new DKD biomarkers. In addition, we will provide an update on new emerging approaches to explore renal function and structure, using functional tests and imaging.
Collapse
Affiliation(s)
- Federica Barutta
- Department of Medical Sciences, University of Turin, Turin, Italy.
| | - Stefania Bellini
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Silvia Canepa
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Marilena Durazzo
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Gabriella Gruden
- Department of Medical Sciences, University of Turin, Turin, Italy
| |
Collapse
|
44
|
Hasegawa S, Inagi R. Harnessing Metabolomics to Describe the Pathophysiology Underlying Progression in Diabetic Kidney Disease. Curr Diab Rep 2021; 21:21. [PMID: 33974145 PMCID: PMC8113300 DOI: 10.1007/s11892-021-01390-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/27/2021] [Indexed: 02/06/2023]
Abstract
PURPOSE OF REVIEW Diabetic kidney disease (DKD), a leading cause of end-stage kidney disease, is the result of metabolic network alterations in the kidney. Therefore, metabolomics is an effective tool for understanding its pathophysiology, finding key biomarkers, and developing a new treatment strategy. In this review, we summarize the application of metabolomics to DKD research. RECENT FINDINGS Alterations in renal energy metabolism including the accumulation of tricarboxylic acid cycle and glucose metabolites are observed in the early stage of DKD, and they finally lead to mitochondrial dysfunction in advanced DKD. Mitochondrial fission-fusion imbalance and dysregulated organelle crosstalk might contribute to this process. Moreover, metabolomics has identified several uremic toxins including phenyl sulfate and tryptophan derivatives as promising biomarkers that mediate DKD progression. Recent advances in metabolomics have clarified the role of dysregulated energy metabolism and uremic toxins in DKD pathophysiology. Integration of multi-omics data will provide additional information for identifying critical drivers of DKD.
Collapse
Affiliation(s)
- Sho Hasegawa
- Division of Chronic Kidney Disease Pathophysiology, The University of Tokyo Graduate School of Medicine, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655 Japan
- Division of Nephrology and Endocrinology, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Reiko Inagi
- Division of Chronic Kidney Disease Pathophysiology, The University of Tokyo Graduate School of Medicine, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655 Japan
| |
Collapse
|
45
|
Moon S, Tsay JJ, Lampert H, Md Dom ZI, Kostic AD, Smiles A, Niewczas MA. Circulating short and medium chain fatty acids are associated with normoalbuminuria in type 1 diabetes of long duration. Sci Rep 2021; 11:8592. [PMID: 33883567 PMCID: PMC8060327 DOI: 10.1038/s41598-021-87585-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 03/30/2021] [Indexed: 11/08/2022] Open
Abstract
A substantial number of subjects with Type 1 Diabetes (T1D) of long duration never develop albuminuria or renal function impairment, yet the underlying protective mechanisms remain unknown. Therefore, our study included 308 Joslin Kidney Study subjects who had T1D of long duration (median: 24 years), maintained normal renal function and had either normoalbuminuria or a broad range of albuminuria within the 2 years preceding the metabolomic determinations. Serum samples were subjected to global metabolomic profiling. 352 metabolites were detected in at least 80% of the study population. In the logistic analyses adjusted for multiple testing (Bonferroni corrected α = 0.000028), we identified 38 metabolites associated with persistent normoalbuminuria independently from clinical covariates. Protective metabolites were enriched in Medium Chain Fatty Acids (MCFAs) and in Short Chain Fatty Acids (SCFAs) and particularly involved odd-numbered and dicarboxylate Fatty Acids. One quartile change of nonanoate, the top protective MCFA, was associated with high odds of having persistent normoalbuminuria (OR (95% CI) 0.14 (0.09, 0.23); p < 10-12). Multivariable Random Forest analysis concordantly indicated to MCFAs as effective classifiers. Associations of the relevant Fatty Acids with albuminuria seemed to parallel associations with tubular biomarkers. Our findings suggest that MCFAs and SCFAs contribute to the metabolic processes underlying protection against albuminuria development in T1D that are independent from mechanisms associated with changes in renal function.
Collapse
Affiliation(s)
- Salina Moon
- Research Division, Joslin Diabetes Center, One Joslin Place, Boston, MA, 02215, USA
| | - John J Tsay
- Research Division, Joslin Diabetes Center, One Joslin Place, Boston, MA, 02215, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Medicine, Veterans Affairs Boston Healthcare System, Boston, MA, USA
| | - Heather Lampert
- Research Division, Joslin Diabetes Center, One Joslin Place, Boston, MA, 02215, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Department of Family Medicine, Brown University, Providence, RI, USA
| | - Zaipul I Md Dom
- Research Division, Joslin Diabetes Center, One Joslin Place, Boston, MA, 02215, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Aleksandar D Kostic
- Research Division, Joslin Diabetes Center, One Joslin Place, Boston, MA, 02215, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Department of Microbiology, Harvard Medical School, Boston, MA, USA
| | - Adam Smiles
- Research Division, Joslin Diabetes Center, One Joslin Place, Boston, MA, 02215, USA
| | - Monika A Niewczas
- Research Division, Joslin Diabetes Center, One Joslin Place, Boston, MA, 02215, USA.
- Department of Medicine, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
46
|
Steinbrenner I, Schultheiss UT, Kotsis F, Schlosser P, Stockmann H, Mohney RP, Schmid M, Oefner PJ, Eckardt KU, Köttgen A, Sekula P. Urine Metabolite Levels, Adverse Kidney Outcomes, and Mortality in CKD Patients: A Metabolome-wide Association Study. Am J Kidney Dis 2021; 78:669-677.e1. [PMID: 33839201 DOI: 10.1053/j.ajkd.2021.01.018] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 01/22/2021] [Indexed: 01/01/2023]
Abstract
RATIONALE & OBJECTIVE Mechanisms underlying the variable course of disease progression in patients with chronic kidney disease (CKD) are incompletely understood. The aim of this study was to identify novel biomarkers of adverse kidney outcomes and overall mortality, which may offer insights into pathophysiologic mechanisms. STUDY DESIGN Metabolome-wide association study. SETTING & PARTICIPANTS 5,087 patients with CKD enrolled in the observational German Chronic Kidney Disease Study. EXPOSURES Measurements of 1,487 metabolites in urine. OUTCOMES End points of interest were time to kidney failure (KF), a combined end point of KF and acute kidney injury (KF+AKI), and overall mortality. ANALYTICAL APPROACH Statistical analysis was based on a discovery-replication design (ratio 2:1) and multivariable-adjusted Cox regression models. RESULTS After a median follow-up of 4 years, 362 patients died, 241 experienced KF, and 382 experienced KF+AKI. Overall, we identified 55 urine metabolites whose levels were significantly associated with adverse kidney outcomes and/or mortality. Higher levels of C-glycosyltryptophan were consistently associated with all 3 main end points (hazard ratios of 1.43 [95% CI, 1.27-1.61] for KF, 1.40 [95% CI, 1.27-1.55] for KF+AKI, and 1.47 [95% CI, 1.33-1.63] for death). Metabolites belonging to the phosphatidylcholine pathway showed significant enrichment. Members of this pathway contributed to the improvement of the prediction performance for KF observed when multiple metabolites were added to the well-established Kidney Failure Risk Equation. LIMITATIONS Findings among patients of European ancestry with CKD may not be generalizable to the general population. CONCLUSIONS Our comprehensive screen of the association between urine metabolite levels and adverse kidney outcomes and mortality identifies metabolites that predict KF and represents a valuable resource for future studies of biomarkers of CKD progression.
Collapse
Affiliation(s)
- Inga Steinbrenner
- Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg
| | - Ulla T Schultheiss
- Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg; Department of Medicine IV-Nephrology and Primary Care, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg
| | - Fruzsina Kotsis
- Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg; Department of Medicine IV-Nephrology and Primary Care, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg
| | - Pascal Schlosser
- Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg
| | - Helena Stockmann
- Department of Nephrology and Medical Intensive Care, Charité-Universitätsmedizin Berlin, Berlin
| | | | - Matthias Schmid
- Department of Medical Biometry, Informatics and Epidemiology, University Hospital Bonn, Bonn
| | - Peter J Oefner
- Institute of Functional Genomics, University of Regensburg, Regensburg, Germany
| | - Kai-Uwe Eckardt
- Department of Nephrology and Medical Intensive Care, Charité-Universitätsmedizin Berlin, Berlin; Department of Nephrology and Hypertension, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen; Institute of Functional Genomics, University of Regensburg, Regensburg, Germany
| | - Anna Köttgen
- Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg.
| | - Peggy Sekula
- Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg.
| | | |
Collapse
|
47
|
Chen S, Liu YH, Dai DP, Zhu ZB, Dai Y, Wu ZM, Zhang LP, Duan ZF, Lu L, Ding FH, Zhu JZ, Zhang RY. Using circulating O-sulfotyrosine in the differential diagnosis of acute kidney injury and chronic kidney disease. BMC Nephrol 2021; 22:66. [PMID: 33622294 PMCID: PMC7903698 DOI: 10.1186/s12882-021-02268-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Accepted: 02/11/2021] [Indexed: 11/23/2022] Open
Abstract
Background Sulfation of tyrosine, yielding O-sulfotyrosine, is a common but fixed post-translational modification in eukaryotes. Patients with increased circulating O-sulfotyrosine levels experience a faster decline in renal function with progression to end-stage renal disease (ESRD). In the present study, we measured serum O-sulfotyrosine levels in individuals with chronic kidney disease (CKD) and acute kidney injury (AKI) to explore its ability to differentiate AKI from CKD. Methods A total of 135 patients (20 with AKI and 115 with CKD) were recruited prospectively for liquid chromatography-mass spectrometry assessment of circulating O-sulfotyrosine. We also studied C57BL/6 mice with CKD after 5/6 nephrectomy (Nx). Blood samples were drawn from the tail vein on Day 1, 3, 5, 7, 14, 30, 60, and 90 after CKD. Serum separation and characterization of creatinine, blood urea nitrogen (BUN), and O-sulfotyrosine was performed. Thus, the time-concentration curves of the O-sulfotyrosine level demonstrate the variation of kidney dysfunction. Results The serum levels of O-sulfotyrosine were markedly increased in patients with CKD compared with AKI. Median O-sulfotyrosine levels in CKD patients versus AKI, respectively, were as follows:243.61 ng/mL(interquartile range [IQR] = 171.90–553.86) versus 126.55 ng/mL (IQR = 48.19–185.03, P = 0.004). In patients with CKD, O-sulfotyrosine levels were positively correlated with creatinine, BUN, and Cystatin C (r = 0.63, P < 0.001; r = 0.49, P < 0.001; r = 0.61, P < 0.001, respectively) by the multivariate linear regression analysis (β = 0.71, P < 0.001; β = 0.40, P = 0.002; β = 0.73, P < 0.001, respectively). However, this association was not statistically significant in patients with AKI (r = − 0.17, P = 0.472; r = 0.11, P = 0.655; r = 0.09, P = 0.716, respectively). The receiver operating characteristic (ROC) analysis illustrated that the area under the curve was 0.80 (95% confidence interval [CI] 0.71–0.89; P < 0.001) and the optimal cut-off value of serum O-sulfotyrosine suggesting AKI was < 147.40 ng/mL with a sensitivity and specificity of 80.90 and 70.00% respectively. In animal experiments, serum levels of O-sulfotyrosine in mice were elevated on Day 7 after 5/6 nephrectomy (14.89 ± 1.05 vs. 8.88 ± 2.62 ng/mL, P < 0.001) until Day 90 (32.65 ± 5.59 vs. 8.88 ± 2.62 ng/mL, P < 0.001). Conclusion Serum O-sulfotyrosine levels were observed correlated with degrading renal function and in CKD patients substantially higher than those in AKI patients. Thus serum O-sulfotyrosine facilitated the differential diagnosis of AKI from CKD.
Collapse
Affiliation(s)
- Shuai Chen
- Department of Vascular & Cardiology, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yong-Hua Liu
- Department of Cardiology, Bao Shan People's Hospital, Baoshan, Yunnan Province, China
| | - Dao-Peng Dai
- Department of Vascular & Cardiology, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zheng-Bin Zhu
- Department of Vascular & Cardiology, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yang Dai
- Department of Vascular & Cardiology, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhi-Ming Wu
- Department of Vascular & Cardiology, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Li-Ping Zhang
- Department of Cardiology, Bao Shan People's Hospital, Baoshan, Yunnan Province, China
| | - Zhi-Feng Duan
- Department of Cardiology, Bao Shan People's Hospital, Baoshan, Yunnan Province, China
| | - Lin Lu
- Department of Vascular & Cardiology, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Feng-Hua Ding
- Department of Vascular & Cardiology, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Jin-Zhou Zhu
- Department of Vascular & Cardiology, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Rui-Yan Zhang
- Department of Vascular & Cardiology, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
48
|
Morita S, Inai Y, Minakata S, Kishimoto S, Manabe S, Iwahashi N, Ino K, Ito Y, Akamizu T, Ihara Y. Quantification of serum C-mannosyl tryptophan by novel assay to evaluate renal function and vascular complications in patients with type 2 diabetes. Sci Rep 2021; 11:1946. [PMID: 33479412 PMCID: PMC7820242 DOI: 10.1038/s41598-021-81479-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 01/06/2021] [Indexed: 11/23/2022] Open
Abstract
C-Mannosyl tryptophan (CMW) is a unique glycosylated amino acid, and a candidate novel biomarker of renal function. In type 2 diabetes (T2D), a combination of metabolites including CMW has recently been the focus of novel biomarkers for the evaluation of renal function and prediction of its decline. However, previous quantification methods for serum CMW have several limitations. We recently established a novel assay for quantifying serum CMW. Serum CMW from 99 Japanese patients with T2D was quantified by this assay using hydrophilic interaction liquid chromatography. The serum CMW levels were cross-sectionally characterized in relation to clinical features, including renal function and vascular complications. Serum CMW level was more strongly correlated with serum creatinine and cystatin C levels and with eGFR than with albumin urea level. The ROC curve to detect eGFR < 60 ml/min/1.73 m2 revealed that the cutoff serum CMW level was 337.5 nM (AUC 0.883). Serum CMW levels were higher in patients with a history of macroangiopathy than in those without history. They correlated with ankle-brachial pressure index, whereas cystatin C did not. Serum CMW levels quantified by the novel assay could be useful in evaluation of glomerular filtration of renal function and peripheral arterial disease in T2D.
Collapse
Affiliation(s)
- Shuhei Morita
- First Department of Medicine, Wakayama Medical University, 811-1 Kimiidera, Wakayama, Wakayama, 641-0012, Japan.
| | - Yoko Inai
- Department of Biochemistry, Wakayama Medical University, 811-1 Kimiidera, Wakayama, Wakayama, 641-0012, Japan
| | - Shiho Minakata
- Department of Biochemistry, Wakayama Medical University, 811-1 Kimiidera, Wakayama, Wakayama, 641-0012, Japan
| | - Shohei Kishimoto
- First Department of Medicine, Wakayama Medical University, 811-1 Kimiidera, Wakayama, Wakayama, 641-0012, Japan
| | - Shino Manabe
- Pharmaceutical Department & The Institute of Medicinal Chemistry, Hoshi University, 2-4-41 Ebara, Shinagawa, Tokyo, 142-8501, Japan
- Research Center for Pharmaceutical Development, Graduate School of Pharmaceutical Sciences & Faculty of Pharmaceutical Sciences, Tohoku University, 6-3 Aoba, Sendai, Miyagi, 980-8578, Japan
| | - Naoyuki Iwahashi
- Department of Obstetrics and Gynecology, Wakayama Medical University, 811-1 Kimiidera, Wakayama, Wakayama, 641-0012, Japan
| | - Kazuhiko Ino
- Department of Obstetrics and Gynecology, Wakayama Medical University, 811-1 Kimiidera, Wakayama, Wakayama, 641-0012, Japan
| | - Yukishige Ito
- RIKEN Cluster for Pioneering Research, 2-1 Hirosawa, Wako, Saitama, 351-0198, Japan
- Department of Chemistry, Graduate School of Science, Osaka University, 1-1 Machikaneyama, Toyonaka, Osaka, 560-0043, Japan
| | - Takashi Akamizu
- First Department of Medicine, Wakayama Medical University, 811-1 Kimiidera, Wakayama, Wakayama, 641-0012, Japan
| | - Yoshito Ihara
- Department of Biochemistry, Wakayama Medical University, 811-1 Kimiidera, Wakayama, Wakayama, 641-0012, Japan.
| |
Collapse
|
49
|
Yin L, Zhu X, Novák P, Zhou L, Gao L, Yang M, Zhao G, Yin K. The epitranscriptome of long noncoding RNAs in metabolic diseases. Clin Chim Acta 2021; 515:80-89. [PMID: 33422492 DOI: 10.1016/j.cca.2021.01.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Revised: 12/30/2020] [Accepted: 01/04/2021] [Indexed: 02/06/2023]
Abstract
Long noncoding RNAs (lncRNAs) have abundant content and extensive functions that regulate the expression of genes at multiple levels. Recently, transcriptome-wide analysis confirmed that RNA can undergo various chemical modifications in response to stimulation by the environment that further determine the action mechanisms of RNAs and expand the diversity of the transcriptome. Modifications that occur in lncRNAs can affect their expression and the regulation of downstream molecules by changing the secondary structure, splicing, degradation or molecular stability of lncRNAs. During the development of metabolic diseases, reversible RNA modifications show a complex transcriptional landscape. Although a wide quantity and variety of lncRNA modifications have been identified, the knowledge regarding their underlying actions in alcohol use disorders (AUDs), osteoporosis, obesity, and cardiovascular disease (CVD) is still in its infancy. Herein, we will focus on the epitranscriptomic modifications that occur on lncRNAs and the crosstalk between them that affect metabolic diseases.
Collapse
Affiliation(s)
- Linjie Yin
- Research Lab for Clinical & Translational Medicine, Medical School, University of South China, Hengyang 421001, China; The Second Affiliated Hospital of Guilin Medical University, Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin, Guangxi 541100, China
| | - Xiao Zhu
- The Second Affiliated Hospital of Guilin Medical University, Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin, Guangxi 541100, China
| | - Petr Novák
- The Second Affiliated Hospital of Guilin Medical University, Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin, Guangxi 541100, China
| | - Le Zhou
- The Second Affiliated Hospital of Guilin Medical University, Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin, Guangxi 541100, China
| | - Ling Gao
- Research Lab for Clinical & Translational Medicine, Medical School, University of South China, Hengyang 421001, China
| | - Min Yang
- Research Lab for Clinical & Translational Medicine, Medical School, University of South China, Hengyang 421001, China; The Second Affiliated Hospital of Guilin Medical University, Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin, Guangxi 541100, China
| | - GuoJun Zhao
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan City People's Hospital, Qingyuan 511518, China.
| | - Kai Yin
- The Second Affiliated Hospital of Guilin Medical University, Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin, Guangxi 541100, China.
| |
Collapse
|
50
|
Li Q, Liu X, Yang J, Erlund I, Lernmark Å, Hagopian W, Rewers M, She JX, Toppari J, Ziegler AG, Akolkar B, Krischer JP. Plasma Metabolome and Circulating Vitamins Stratified Onset Age of an Initial Islet Autoantibody and Progression to Type 1 Diabetes: The TEDDY Study. Diabetes 2021; 70:282-292. [PMID: 33106256 PMCID: PMC7876562 DOI: 10.2337/db20-0696] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 10/20/2020] [Indexed: 12/11/2022]
Abstract
Children's plasma metabolome, especially lipidome, reflects gene regulation and dietary exposures, heralding the development of islet autoantibodies (IA) and type 1 diabetes (T1D). The Environmental Determinants of Diabetes in the Young (TEDDY) study enrolled 8,676 newborns by screening of HLA-DR-DQ genotypes at six clinical centers in four countries, profiled metabolome, and measured concentrations of ascorbic acid, 25-hydroxyvitamin D [25(OH)D], and erythrocyte membrane fatty acids following birth until IA seroconversion under a nested case-control design. We grouped children having an initial autoantibody only against insulin (IAA-first) or GAD (GADA-first) by unsupervised clustering of temporal lipidome, identifying a subgroup of children having early onset of each initial autoantibody, i.e., IAA-first by 12 months and GADA-first by 21 months, consistent with population-wide early seroconversion age. Differential analysis showed that infants having reduced plasma ascorbic acid and cholesterol experienced IAA-first earlier, while early onset of GADA-first was preceded by reduced sphingomyelins at infancy. Plasma 25(OH)D prior to either autoantibody was lower in T1D progressors compared with nonprogressors, with simultaneous lower diglycerides, lysophosphatidylcholines, triglycerides, and alanine before GADA-first. Plasma ascorbic acid and 25(OH)D at infancy were lower in HLA-DR3/DR4 children among IA case subjects but not in matched control subjects, implying gene expression dysregulation of circulating vitamins as latent signals for IA or T1D progression.
Collapse
Affiliation(s)
- Qian Li
- Health Informatics Institute, University of South Florida, Tampa, FL
| | - Xiang Liu
- Health Informatics Institute, University of South Florida, Tampa, FL
| | - Jimin Yang
- Health Informatics Institute, University of South Florida, Tampa, FL
| | - Iris Erlund
- Department of Government Services, Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Åke Lernmark
- Department of Clinical Sciences, Clinical Research Centre, Skåne University Hospital, Lund University, Malmö, Sweden
| | | | - Marian Rewers
- Barbara Davis Center for Childhood Diabetes, University of Colorado Denver, Aurora, CO
| | - Jin-Xiong She
- Center for Biotechnology and Genomic Medicine, Medical College of Georgia, Augusta University, Augusta, GA
| | - Jorma Toppari
- Department of Pediatrics, Turku University Hospital, Turku, Finland
- Department of Physiology, University of Turku, Turku, Finland
| | - Anette-G Ziegler
- Institute of Diabetes Research, Helmholtz Zentrum München, Munich, Germany
- Forschergruppe Diabetes, Technical University of Munich, Klinikum Rechts der Isar, Munich, Germany
- Forschergruppe Diabetes e.V. at Helmholtz Zentrum München, Munich, Germany
| | - Beena Akolkar
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD
| | | |
Collapse
|