1
|
Liu J, Li Z, Zhang Z, Shen Z. Uncovering the mechanism of Huangkui capsule in the treatment of diabetic kidney disease based on network pharmacology and experimental validation. Sci Rep 2025; 15:6503. [PMID: 39987179 PMCID: PMC11846948 DOI: 10.1038/s41598-025-91264-w] [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: 04/22/2024] [Accepted: 02/19/2025] [Indexed: 02/24/2025] Open
Abstract
Diabetic kidney disease (DKD) is a main complication of diabetes mellitus. experimental in vitro validation and Network pharmacology were used in this study to explore the potential mechanism of Huangkui capsules (HKC) in treating DKD. First, we used CCK8 to analyze the optimal drug concentration of HKC. Next, we used flow cytometry, ELISA, Scratch test, and immunofluorescence to examine the apoptosis, oxidative stress, inflammatory factors, and fibrotic factors (FN and α-SMA) expression in HK-2 cells. Thereafter, in order to determine the potential molecular mechanisms underlying the therapeutic effect of HKC in DKD. Compounds contained in HKC were explored by UPLC-Q-TOF-MS/MS. SwissTargetPrediction was utilized for predicting potential gene targets of these compounds. OMIM, DisGeNet and GeneCards databases were employed to identify DKD-related genes. Meanwhile, the association of compounds with DKD genes was examined by protein-protein interaction, GO and KEGG analysis. Finally, molecular docking and molecular dynamics simulation were adopted for further validation. The results showed that HKC had 40 active ingredients, 1051 possible gene targets, and 133 DKD-HKC intersection genes. IL6, TNF, GAPDH, AKT1, PPARG, and TP53 were candidate hub genes by which HKC exerted its anti-DKD function based on molecular docking, molecular dynamics simulation and experimental results. To conclude, this study sheds more lights on the possible pharmacological activities of HKC in DKD and a foundation for further clinical application.
Collapse
Affiliation(s)
- Junhong Liu
- Department of Gastroenterology, The Fuyang Affiliated Hospital of Anhui Medical University, Fuyang, 236000, Anhui, China
| | - Ziwei Li
- Department of Clinical Nutrition, The Fuyang Affiliated Hospital of Anhui Medical University, Fuyang, 236000, Anhui, China
| | - ZongYao Zhang
- Department of General Surgery, The First Affiliated Hospital of Anhui University of Science and Technology, Huainan, 232007, China
| | - Zhongyuan Shen
- Department of Radiology, The Fuyang Affiliated Hospital of Anhui Medical University, Fuyang, 236000, Anhui, China
| |
Collapse
|
2
|
Wang ZH, Dong Q, Yan Q, Yu WR, Zhang DD, Yi R. Constructing the biomolecular networks associated with diabetic nephropathy and dissecting the effects of biomolecule variation underlying pathogenesis. Endocr J 2024; 71:1031-1043. [PMID: 39069497 PMCID: PMC11778351 DOI: 10.1507/endocrj.ej24-0170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 06/05/2024] [Indexed: 07/30/2024] Open
Abstract
Diabetic nephropathy (DN) is a common and serious complication of diabetes, contributing significantly to patient mortality. Complication of DN (CDN) ranks as the second leading cause of end-stage renal disease globally. To address this, understanding the genetic regulation underlying DN is crucial for personalized treatment strategies. In this study, we identified genes and lncRNAs associated with diabetes and diabetic nephropathy constructing a DN-related lncRNA-mRNA network (DNLMN). This network, characterized by scale-free biomolecular properties, generated through the study of topological properties, elucidates key regulatory interactions. Enrichment analysis of important network modules revealed critical biological processes and pathways involved in DN pathogenesis. In the second step, we investigated the differential expression and co-expression of hub nodes in diseased and normal individuals, identifying lncRNA-mRNA relationships implicated in disease regulation. Finally, we gathered DN-related single nucleotide polymorphisms (SNPs) and lncRNAs from the LincSNP 3.0 database. The DNLMN encompasses SNP-associated lncRNAs, and transcription factors (TFs) linked to differentially expressed lncRNAs between diseased and normal samples. These results underscore the significance of biomolecular networks in disease progression and highlighting the role of biomolecular variability contributes to personalized disease phenotyping and treatment.
Collapse
Affiliation(s)
- Zi-Han Wang
- Department of Endocrine, the First Affiliated Hospital of Harbin Medical University, Harbin 150001, People’s Republic of China
| | - Qi Dong
- Department of Neurology, the First Affiliated Hospital of Harbin Medical University, Harbin 150001, People’s Republic of China
| | - Qian Yan
- Department of Endocrine, the First Affiliated Hospital of Harbin Medical University, Harbin 150001, People’s Republic of China
| | - Wan-Rong Yu
- Department of Endocrine, the First Affiliated Hospital of Harbin Medical University, Harbin 150001, People’s Republic of China
| | - Dan-Dan Zhang
- Department of Endocrine, the First Affiliated Hospital of Harbin Medical University, Harbin 150001, People’s Republic of China
| | - Ran Yi
- Department of Endocrine, the First Affiliated Hospital of Harbin Medical University, Harbin 150001, People’s Republic of China
| |
Collapse
|
3
|
Garmaa G, Manzéger A, Haghighi S, Kökény G. HK-2 cell response to TGF-β highly depends on cell culture medium formulations. Histochem Cell Biol 2024; 161:69-79. [PMID: 37752256 PMCID: PMC10794419 DOI: 10.1007/s00418-023-02237-x] [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] [Accepted: 08/31/2023] [Indexed: 09/28/2023]
Abstract
The immortalized human renal proximal tubular epithelial cell line HK-2 is most commonly used to study renal cell physiology and human kidney diseases with tubulointerstitial fibrosis such as diabetic nephropathy, obstructive uropathy or allograft fibrosis. Epithelial-to-mesenchymal transition (EMT) is the main pathological process of tubulointerstitial fibrosis in vitro. Transforming growth factor-beta (TGF-β) is a key inducer of EMT. Several pro-fibrotic gene expression differences have been observed in a TGF-β-induced EMT model of HK-2 cells. However, growth conditions and medium formulations might greatly impact these differences. We investigated gene and protein expression of HK-2 cells cultured in six medium formulations. TGF-β1 increased the expression of ACTA2, TGFB1, COL4A1, EGR2, VIM and CTGF genes while reducing PPARG in all medium formulations. Interestingly, TGF-β1 treatment either increased or decreased EGR1, FN, IL6 and C3 gene expression, depending on medium formulations. The cell morphology was slightly affected, but immunoblots revealed TGFB1 and vimentin protein overexpression in all media. However, fibronectin expression as well as the nuclear translocation of EGR1 was medium dependent. In conclusion, our study demonstrates that, using the HK-2 in vitro model of EMT, the meticulous selection of appropriate cell culture medium formulation is essential to achieve reliable scientific results.
Collapse
Affiliation(s)
- Gantsetseg Garmaa
- Institute of Translational Medicine, Semmelweis University, Nagyvárad tér 4, Budapest, 1089, Hungary
| | - Anna Manzéger
- Institute of Translational Medicine, Semmelweis University, Nagyvárad tér 4, Budapest, 1089, Hungary
- International Nephrology Research and Training Center, Semmelweis University, Nagyvárad tér 4, Budapest, 1089, Hungary
| | - Samaneh Haghighi
- Institute of Translational Medicine, Semmelweis University, Nagyvárad tér 4, Budapest, 1089, Hungary
| | - Gábor Kökény
- Institute of Translational Medicine, Semmelweis University, Nagyvárad tér 4, Budapest, 1089, Hungary.
- International Nephrology Research and Training Center, Semmelweis University, Nagyvárad tér 4, Budapest, 1089, Hungary.
| |
Collapse
|
4
|
Zhao L, Hao Y, Tang S, Han X, Li R, Zhou X. Energy metabolic reprogramming regulates programmed cell death of renal tubular epithelial cells and might serve as a new therapeutic target for acute kidney injury. Front Cell Dev Biol 2023; 11:1276217. [PMID: 38054182 PMCID: PMC10694365 DOI: 10.3389/fcell.2023.1276217] [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: 08/11/2023] [Accepted: 11/08/2023] [Indexed: 12/07/2023] Open
Abstract
Acute kidney injury (AKI) induces significant energy metabolic reprogramming in renal tubular epithelial cells (TECs), thereby altering lipid, glucose, and amino acid metabolism. The changes in lipid metabolism encompass not only the downregulation of fatty acid oxidation (FAO) but also changes in cell membrane lipids and triglycerides metabolism. Regarding glucose metabolism, AKI leads to increased glycolysis, activation of the pentose phosphate pathway (PPP), inhibition of gluconeogenesis, and upregulation of the polyol pathway. Research indicates that inhibiting glycolysis, promoting the PPP, and blocking the polyol pathway exhibit a protective effect on AKI-affected kidneys. Additionally, changes in amino acid metabolism, including branched-chain amino acids, glutamine, arginine, and tryptophan, play an important role in AKI progression. These metabolic changes are closely related to the programmed cell death of renal TECs, involving autophagy, apoptosis, necroptosis, pyroptosis, and ferroptosis. Notably, abnormal intracellular lipid accumulation can impede autophagic clearance, further exacerbating lipid accumulation and compromising autophagic function, forming a vicious cycle. Recent studies have demonstrated the potential of ameliorating AKI-induced kidney damage through calorie and dietary restriction. Consequently, modifying the energy metabolism of renal TECs and dietary patterns may be an effective strategy for AKI treatment.
Collapse
Affiliation(s)
- Limei Zhao
- The Fifth Clinical Medical College of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yajie Hao
- The Fifth Clinical Medical College of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Shuqin Tang
- The Fifth Clinical Medical College of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Xiutao Han
- The Third Clinical College, Shanxi University of Chinese Medicine, Jinzhong, Shanxi, China
| | - Rongshan Li
- Department of Nephrology, Shanxi Provincial People’s Hospital, The Fifth Clinical Medical College of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Xiaoshuang Zhou
- Department of Nephrology, Shanxi Provincial People’s Hospital, The Fifth Clinical Medical College of Shanxi Medical University, Taiyuan, Shanxi, China
| |
Collapse
|
5
|
Braga PC, Bernardino RL, Guerra-Carvalho B, Carrageta DF, Oliveira PF, Rodrigues AS, Alves MG. The progression from mild to severe hyperglycemia coupled with insulin resistance causes mitochondrial dysfunction and alters the metabolic secretome of epithelial kidney cells. Exp Cell Res 2023; 431:113744. [PMID: 37648074 DOI: 10.1016/j.yexcr.2023.113744] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 08/08/2023] [Accepted: 08/11/2023] [Indexed: 09/01/2023]
Abstract
Diabetic nephropathy (DN) and insulin resistance (IR) in kidney cells are considered main causes for end-stage renal failure. However, it is unclear how IR affects early stages of the disease. Here, we investigate the impact of mild (11 mM) and severe (22 mM) hyperglycemia, with and without induced IR, on cellular metabolism and mitochondrial bioenergetics in a human kidney cell line (HK-2). IR in HK-2 cells was induced with palmitic acid and cellular cytotoxicity was studied. We evaluated the impact of mild and severe hyperglycemia with and without IR on the metabolic secretome of the cells, their live-cell mitochondria function, mitochondrial membrane potential, and mitochondrial complex activities. Furthermore, we measured fatty acid oxidation and lipid accumulation. Cells cultured under mild hyperglycemic conditions exhibited increased mitochondrial bioenergetic parameters, such as basal respiration, ATP-linked production, maximal respiration capacity, and spare respiration capacity. However, these parameters decreased when cells were cultured under higher glucose concentrations when IR was induced. Our data suggests that progression from mild to severe hyperglycemia induces a metabolic shift, where gluconeogenic amino acids play a crucial role in supplying the energy requirements of HK-2. To our knowledge, this is the first study to evaluate the progression from mild to severe hyperglycemia allied to IR in human kidney cells. This work highlights that this progression leads to mitochondrial dysfunction and alters the metabolic profile of kidney cells. These results identify possible targets for early intervention in DN.
Collapse
Affiliation(s)
- Patrícia C Braga
- Unit for Multidisciplinary Research in Biomedicine (UMIB), Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Porto, Portugal; ITR- Laboratory for Integrative and Translational Research in Population Health, Porto, Portugal; Laboratory of Physiology, Department of Imuno-physiology and Pharmacology, Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Porto, Portugal.
| | - Raquel L Bernardino
- Unit for Multidisciplinary Research in Biomedicine (UMIB), Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Porto, Portugal; ITR- Laboratory for Integrative and Translational Research in Population Health, Porto, Portugal.
| | - Bárbara Guerra-Carvalho
- Unit for Multidisciplinary Research in Biomedicine (UMIB), Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Porto, Portugal; ITR- Laboratory for Integrative and Translational Research in Population Health, Porto, Portugal; Laboratory of Physiology, Department of Imuno-physiology and Pharmacology, Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Porto, Portugal; LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, Portugal.
| | - David F Carrageta
- Unit for Multidisciplinary Research in Biomedicine (UMIB), Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Porto, Portugal; ITR- Laboratory for Integrative and Translational Research in Population Health, Porto, Portugal; Laboratory of Physiology, Department of Imuno-physiology and Pharmacology, Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Porto, Portugal.
| | - Pedro F Oliveira
- LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, Portugal.
| | - Anabela S Rodrigues
- Unit for Multidisciplinary Research in Biomedicine (UMIB), Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Porto, Portugal; ITR- Laboratory for Integrative and Translational Research in Population Health, Porto, Portugal; Department of Nephrology, Santo António Hospital, CHUdSA, Porto, Portugal.
| | - Marco G Alves
- Institute of Biomedicine - iBiMED and Department of Medical Sciences, University of Aveiro, 3810-193, Aveiro, Portugal.
| |
Collapse
|
6
|
Ortiz SR, Field MS. C2C12 Muscle Myotubes, but not Kidney Proximal Tubule HK-2 Cells, Elevate Erythritol Synthesis in Response to Oxidative Stress. Curr Dev Nutr 2023; 7:102012. [PMID: 37877034 PMCID: PMC10590721 DOI: 10.1016/j.cdnut.2023.102012] [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: 07/20/2023] [Revised: 09/19/2023] [Accepted: 09/25/2023] [Indexed: 10/26/2023] Open
Abstract
Background As a biomarker, elevated serum erythritol concentrations predict type 2 diabetes and cardiovascular disease onset. Erythritol was recently shown to be a product of human glucose metabolism through the pentose phosphate pathway. The regulation of erythritol synthesis from glucose has been explored in cancer cells but not in nontransformed cells. Objective The kidneys and skeletal muscle have increased erythritol content in response to dietary sucrose, which suggests that they may significantly contribute to circulating erythritol concentrations. In the present study, we evaluated if conditions that promote erythritol synthesis in cancer cells are consistent in skeletal muscle and kidney cells. Methods C2C12 myotubules were used as a model for skeletal muscle, and human kidney (HK)-2 human proximal tubule cells were used to model kidney. C2C12 cells were exposed to high- or low-glucose conditions. Both C2C12 and HK-2 cells were exposed to the free radical generator menadione, then intracellular reactive oxygen species (ROS) and erythritol concentrations were measured. Intracellular sorbitol concentrations were also measured because increased polyol flux was also observed after exposure to excess glucose and oxidative stress. Results Intracellular erythritol concentrations were significantly elevated in C2C12 cells following both high-glucose and menadione treatment. In contrast, HK-2 cells did not increase erythritol synthesis in response to oxidative stress. Generation of ROS through hydrogen peroxide exposure elevated sorbitol concentrations in both C2C12 and HK-2 cells, whereas generation of radicals with menadione treatment did not affect sorbitol production in either cell type. Conclusions These findings highlight that the factors contributing to elevated erythritol synthesis vary between cell types. More specifically, these studies demonstrate that muscle cells increase erythritol synthesis in response to both high glucose in culture medium and oxidative stress, whereas kidney cells increase erythritol synthesis only in response to high glucose.
Collapse
Affiliation(s)
- Semira R. Ortiz
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, United States
| | - Martha S. Field
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, United States
| |
Collapse
|
7
|
Balint L, Socaciu C, Socaciu AI, Vlad A, Gadalean F, Bob F, Milas O, Cretu OM, Suteanu-Simulescu A, Glavan M, Ienciu S, Mogos M, Jianu DC, Ursoniu S, Dumitrascu V, Vlad D, Popescu R, Petrica L. Metabolites Potentially Derived from Gut Microbiota Associated with Podocyte, Proximal Tubule, and Renal and Cerebrovascular Endothelial Damage in Early Diabetic Kidney Disease in T2DM Patients. Metabolites 2023; 13:893. [PMID: 37623837 PMCID: PMC10456401 DOI: 10.3390/metabo13080893] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 07/26/2023] [Accepted: 07/26/2023] [Indexed: 08/26/2023] Open
Abstract
Complications due to type 2 diabetes mellitus (T2DM) such as diabetic kidney disease (DKD) and cerebral small vessel disease (CSVD) have a powerful impact on mortality and morbidity. Our current diagnostic markers have become outdated as T2DM-related complications continue to develop. The aim of the investigation was to point out the relationship between previously selected metabolites which are potentially derived from gut microbiota and indicators of endothelial, proximal tubule (PT), and podocyte dysfunction, and neurosonological indices. The study participants were 20 healthy controls and 90 T2DM patients divided into three stages: normoalbuminuria, microalbuminuria, and macroalbuminuria. Serum and urine metabolites were determined by untargeted and targeted metabolomic techniques. The markers of endothelial, PT and podocyte dysfunction were assessed by ELISA technique, and the neurosonological indices were provided by an ultrasound device with high resolution (MYLAB 8-ESAOTE Italy). The descriptive statistical analysis was followed by univariable and multivariable linear regression analyses. In conclusion, in serum, arginine (sArg), butenoylcarnitine (sBCA), and indoxyl sulfate (sIS) expressed a biomarker potential in terms of renal endothelial dysfunction and carotid atherosclerosis, whereas sorbitol (sSorb) may be a potential biomarker of blood-brain barrier (BBB) dysfunction. In urine, BCA and IS were associated with markers of podocyte damage, whereas PCS correlated with markers of PT dysfunction.
Collapse
Affiliation(s)
- Lavinia Balint
- Department of Internal Medicine II—Division of Nephrology, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Sq. No. 2, County Emergency Hospital Timisoara, 300041 Timisoara, Romania; (L.B.); (F.B.); (O.M.); (A.S.-S.); (M.G.); (S.I.); (M.M.); (L.P.)
- Centre for Molecular Research in Nephrology and Vascular Disease, Faculty of Medicine, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Sq. No. 2, 300041 Timisoara, Romania; (C.S.); (A.V.); (D.C.J.); (S.U.); (V.D.); (D.V.); (R.P.)
| | - Carmen Socaciu
- Centre for Molecular Research in Nephrology and Vascular Disease, Faculty of Medicine, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Sq. No. 2, 300041 Timisoara, Romania; (C.S.); (A.V.); (D.C.J.); (S.U.); (V.D.); (D.V.); (R.P.)
- Research Center for Applied Biotechnology and Molecular Therapy Biodiatech, SC Proplanta, Trifoiului 12G, 400478 Cluj-Napoca, Romania
| | - Andreea Iulia Socaciu
- Department of Occupational Health, University of Medicine and Pharmacy “Iuliu Haţieganu”, Victor Babes 8, 400347 Cluj-Napoca, Romania;
| | - Adrian Vlad
- Centre for Molecular Research in Nephrology and Vascular Disease, Faculty of Medicine, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Sq. No. 2, 300041 Timisoara, Romania; (C.S.); (A.V.); (D.C.J.); (S.U.); (V.D.); (D.V.); (R.P.)
- Department of Internal Medicine II—Division of Diabetes and Metabolic Diseases, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Sq. No. 2, County Emergency Hospital Timisoara, 300041 Timisoara, Romania
| | - Florica Gadalean
- Department of Internal Medicine II—Division of Nephrology, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Sq. No. 2, County Emergency Hospital Timisoara, 300041 Timisoara, Romania; (L.B.); (F.B.); (O.M.); (A.S.-S.); (M.G.); (S.I.); (M.M.); (L.P.)
- Centre for Molecular Research in Nephrology and Vascular Disease, Faculty of Medicine, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Sq. No. 2, 300041 Timisoara, Romania; (C.S.); (A.V.); (D.C.J.); (S.U.); (V.D.); (D.V.); (R.P.)
| | - Flaviu Bob
- Department of Internal Medicine II—Division of Nephrology, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Sq. No. 2, County Emergency Hospital Timisoara, 300041 Timisoara, Romania; (L.B.); (F.B.); (O.M.); (A.S.-S.); (M.G.); (S.I.); (M.M.); (L.P.)
- Centre for Molecular Research in Nephrology and Vascular Disease, Faculty of Medicine, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Sq. No. 2, 300041 Timisoara, Romania; (C.S.); (A.V.); (D.C.J.); (S.U.); (V.D.); (D.V.); (R.P.)
| | - Oana Milas
- Department of Internal Medicine II—Division of Nephrology, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Sq. No. 2, County Emergency Hospital Timisoara, 300041 Timisoara, Romania; (L.B.); (F.B.); (O.M.); (A.S.-S.); (M.G.); (S.I.); (M.M.); (L.P.)
- Centre for Molecular Research in Nephrology and Vascular Disease, Faculty of Medicine, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Sq. No. 2, 300041 Timisoara, Romania; (C.S.); (A.V.); (D.C.J.); (S.U.); (V.D.); (D.V.); (R.P.)
| | - Octavian Marius Cretu
- Department of Surgery I—Division of Surgical Semiology I, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Sq. No. 2, Emergency Clinical Municipal Hospital Timisoara, 300041 Timisoara, Romania;
| | - Anca Suteanu-Simulescu
- Department of Internal Medicine II—Division of Nephrology, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Sq. No. 2, County Emergency Hospital Timisoara, 300041 Timisoara, Romania; (L.B.); (F.B.); (O.M.); (A.S.-S.); (M.G.); (S.I.); (M.M.); (L.P.)
- Centre for Molecular Research in Nephrology and Vascular Disease, Faculty of Medicine, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Sq. No. 2, 300041 Timisoara, Romania; (C.S.); (A.V.); (D.C.J.); (S.U.); (V.D.); (D.V.); (R.P.)
| | - Mihaela Glavan
- Department of Internal Medicine II—Division of Nephrology, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Sq. No. 2, County Emergency Hospital Timisoara, 300041 Timisoara, Romania; (L.B.); (F.B.); (O.M.); (A.S.-S.); (M.G.); (S.I.); (M.M.); (L.P.)
- Centre for Molecular Research in Nephrology and Vascular Disease, Faculty of Medicine, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Sq. No. 2, 300041 Timisoara, Romania; (C.S.); (A.V.); (D.C.J.); (S.U.); (V.D.); (D.V.); (R.P.)
| | - Silvia Ienciu
- Department of Internal Medicine II—Division of Nephrology, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Sq. No. 2, County Emergency Hospital Timisoara, 300041 Timisoara, Romania; (L.B.); (F.B.); (O.M.); (A.S.-S.); (M.G.); (S.I.); (M.M.); (L.P.)
- Centre for Molecular Research in Nephrology and Vascular Disease, Faculty of Medicine, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Sq. No. 2, 300041 Timisoara, Romania; (C.S.); (A.V.); (D.C.J.); (S.U.); (V.D.); (D.V.); (R.P.)
| | - Maria Mogos
- Department of Internal Medicine II—Division of Nephrology, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Sq. No. 2, County Emergency Hospital Timisoara, 300041 Timisoara, Romania; (L.B.); (F.B.); (O.M.); (A.S.-S.); (M.G.); (S.I.); (M.M.); (L.P.)
- Centre for Molecular Research in Nephrology and Vascular Disease, Faculty of Medicine, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Sq. No. 2, 300041 Timisoara, Romania; (C.S.); (A.V.); (D.C.J.); (S.U.); (V.D.); (D.V.); (R.P.)
| | - Dragos Catalin Jianu
- Centre for Molecular Research in Nephrology and Vascular Disease, Faculty of Medicine, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Sq. No. 2, 300041 Timisoara, Romania; (C.S.); (A.V.); (D.C.J.); (S.U.); (V.D.); (D.V.); (R.P.)
- Department of Neurosciences—Division of Neurology, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Sq. No. 2, County Emergency Hospital Timisoara, 300041 Timisoara, Romania
- Centre for Cognitive Research in Neuropsychiatric Pathology (Neuropsy-Cog), Faculty of Medicine, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Sq. No. 2, 300041 Timisoara, Romania
| | - Sorin Ursoniu
- Centre for Molecular Research in Nephrology and Vascular Disease, Faculty of Medicine, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Sq. No. 2, 300041 Timisoara, Romania; (C.S.); (A.V.); (D.C.J.); (S.U.); (V.D.); (D.V.); (R.P.)
- Centre for Cognitive Research in Neuropsychiatric Pathology (Neuropsy-Cog), Faculty of Medicine, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Sq. No. 2, 300041 Timisoara, Romania
- Department of Functional Sciences III, Division of Public Health and History of Medicine, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Sq. No. 2, 300041 Timisoara, Romania
- Centre for Translational Research and Systems Medicine, Faculty of Medicine, “Victor Babes” University of Medicine and Pharmacy, Eftimie, Murgu Sq. No. 2, 300041 Timisoara, Romania
| | - Victor Dumitrascu
- Centre for Molecular Research in Nephrology and Vascular Disease, Faculty of Medicine, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Sq. No. 2, 300041 Timisoara, Romania; (C.S.); (A.V.); (D.C.J.); (S.U.); (V.D.); (D.V.); (R.P.)
- Department of Biochemistry and Pharmacology IV, Division of Pharmacology, “Victor Babes” University of Medicine and Pharmacy, No. 2, Eftimie Murgu Sq., 300041 Timisoara, Romania
| | - Daliborca Vlad
- Centre for Molecular Research in Nephrology and Vascular Disease, Faculty of Medicine, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Sq. No. 2, 300041 Timisoara, Romania; (C.S.); (A.V.); (D.C.J.); (S.U.); (V.D.); (D.V.); (R.P.)
- Department of Biochemistry and Pharmacology IV, Division of Pharmacology, “Victor Babes” University of Medicine and Pharmacy, No. 2, Eftimie Murgu Sq., 300041 Timisoara, Romania
| | - Roxana Popescu
- Centre for Molecular Research in Nephrology and Vascular Disease, Faculty of Medicine, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Sq. No. 2, 300041 Timisoara, Romania; (C.S.); (A.V.); (D.C.J.); (S.U.); (V.D.); (D.V.); (R.P.)
- Department of Microscopic Morphology II, Division of Cell and Molecular Biology II, “Victor Babes” University of Medicine and Pharmacy, No. 2, Eftimie Murgu Sq., 300041 Timisoara, Romania
| | - Ligia Petrica
- Department of Internal Medicine II—Division of Nephrology, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Sq. No. 2, County Emergency Hospital Timisoara, 300041 Timisoara, Romania; (L.B.); (F.B.); (O.M.); (A.S.-S.); (M.G.); (S.I.); (M.M.); (L.P.)
- Centre for Molecular Research in Nephrology and Vascular Disease, Faculty of Medicine, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Sq. No. 2, 300041 Timisoara, Romania; (C.S.); (A.V.); (D.C.J.); (S.U.); (V.D.); (D.V.); (R.P.)
- Centre for Cognitive Research in Neuropsychiatric Pathology (Neuropsy-Cog), Faculty of Medicine, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Sq. No. 2, 300041 Timisoara, Romania
- Centre for Translational Research and Systems Medicine, Faculty of Medicine, “Victor Babes” University of Medicine and Pharmacy, Eftimie, Murgu Sq. No. 2, 300041 Timisoara, Romania
| |
Collapse
|
8
|
Balint L, Socaciu C, Socaciu AI, Vlad A, Gadalean F, Bob F, Milas O, Cretu OM, Suteanu-Simulescu A, Glavan M, Ienciu S, Mogos M, Jianu DC, Petrica L. Quantitative, Targeted Analysis of Gut Microbiota Derived Metabolites Provides Novel Biomarkers of Early Diabetic Kidney Disease in Type 2 Diabetes Mellitus Patients. Biomolecules 2023; 13:1086. [PMID: 37509122 PMCID: PMC10377254 DOI: 10.3390/biom13071086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 06/29/2023] [Accepted: 07/04/2023] [Indexed: 07/30/2023] Open
Abstract
Diabetic kidney disease (DKD) is one of the most debilitating complications of type 2 diabetes mellitus (T2DM), as it progresses silently to end-stage renal disease (ESRD). The discovery of novel biomarkers of early DKD becomes acute, as its incidence is reaching catastrophic proportions. Our study aimed to quantify previously identified metabolites from serum and urine through untargeted ultra-high-performance liquid chromatography coupled with electrospray ionization-quadrupole-time of flight-mass spectrometry (UHPLC-QTOF-ESI+-MS) techniques, such as the following: arginine, dimethylarginine, hippuric acid, indoxyl sulfate, p-cresyl sulfate, L-acetylcarnitine, butenoylcarnitine and sorbitol. The study concept was based on the targeted analysis of selected metabolites, using the serum and urine of 20 healthy subjects and 90 T2DM patients with DKD in different stages (normoalbuminuria-uACR < 30 mg/g; microalbuminuria-uACR 30-300 mg/g; macroalbuminuria-uACR > 300 mg/g). The quantitative evaluation of metabolites was performed with pure standards, followed by the validation methods such as the limit of detection (LOD) and the limit of quantification (LOQ). The following metabolites from this study resulted as possible biomarkers of early DKD: in serum-arginine, dimethylarginine, hippuric acid, indoxyl sulfate, butenoylcarnitine and sorbitol and in urine-p-cresyl sulfate.
Collapse
Affiliation(s)
- Lavinia Balint
- Department of Internal Medicine II-Division of Nephrology, "Victor Babes" University of Medicine and Pharmacy, Eftimie Murgu Sq. No. 2, County Emergency Hospital, 300041 Timisoara, Romania
- Center for Molecular Research in Nephrology and Vascular Disease, Faculty of Medicine, "Victor Babes" University of Medicine and Pharmacy, Eftimie Murgu Sq. No. 2, 300041 Timisoara, Romania
| | - Carmen Socaciu
- Center for Molecular Research in Nephrology and Vascular Disease, Faculty of Medicine, "Victor Babes" University of Medicine and Pharmacy, Eftimie Murgu Sq. No. 2, 300041 Timisoara, Romania
- Research Center for Applied Biotechnology and Molecular Therapy Biodiatech, SC Proplanta, Trifoiului 12G, 400478 Cluj-Napoca, Romania
| | - Andreea Iulia Socaciu
- Department of Occupational Health, University of Medicine and Pharmacy "Iuliu Haţieganu", Victor Babes 8, 400347 Cluj-Napoca, Romania
| | - Adrian Vlad
- Center for Molecular Research in Nephrology and Vascular Disease, Faculty of Medicine, "Victor Babes" University of Medicine and Pharmacy, Eftimie Murgu Sq. No. 2, 300041 Timisoara, Romania
- Department of Internal Medicine II-Division of Diabetes and Metabolic Diseases, "Victor Babes" University of Medicine and Pharmacy, Eftimie Murgu Sq. No. 2, County Emergency Hospital, 300041 Timisoara, Romania
| | - Florica Gadalean
- Department of Internal Medicine II-Division of Nephrology, "Victor Babes" University of Medicine and Pharmacy, Eftimie Murgu Sq. No. 2, County Emergency Hospital, 300041 Timisoara, Romania
- Center for Molecular Research in Nephrology and Vascular Disease, Faculty of Medicine, "Victor Babes" University of Medicine and Pharmacy, Eftimie Murgu Sq. No. 2, 300041 Timisoara, Romania
| | - Flaviu Bob
- Department of Internal Medicine II-Division of Nephrology, "Victor Babes" University of Medicine and Pharmacy, Eftimie Murgu Sq. No. 2, County Emergency Hospital, 300041 Timisoara, Romania
- Center for Molecular Research in Nephrology and Vascular Disease, Faculty of Medicine, "Victor Babes" University of Medicine and Pharmacy, Eftimie Murgu Sq. No. 2, 300041 Timisoara, Romania
| | - Oana Milas
- Department of Internal Medicine II-Division of Nephrology, "Victor Babes" University of Medicine and Pharmacy, Eftimie Murgu Sq. No. 2, County Emergency Hospital, 300041 Timisoara, Romania
- Center for Molecular Research in Nephrology and Vascular Disease, Faculty of Medicine, "Victor Babes" University of Medicine and Pharmacy, Eftimie Murgu Sq. No. 2, 300041 Timisoara, Romania
| | - Octavian Marius Cretu
- Department of Surgery I-Division of Surgical Semiology I, "Victor Babes" University of Medicine and Pharmacy, Eftimie Murgu Sq. No. 2, Emergency Clinical Municipal Hospital, 300041 Timisoara, Romania
| | - Anca Suteanu-Simulescu
- Department of Internal Medicine II-Division of Nephrology, "Victor Babes" University of Medicine and Pharmacy, Eftimie Murgu Sq. No. 2, County Emergency Hospital, 300041 Timisoara, Romania
- Center for Molecular Research in Nephrology and Vascular Disease, Faculty of Medicine, "Victor Babes" University of Medicine and Pharmacy, Eftimie Murgu Sq. No. 2, 300041 Timisoara, Romania
| | - Mihaela Glavan
- Department of Internal Medicine II-Division of Nephrology, "Victor Babes" University of Medicine and Pharmacy, Eftimie Murgu Sq. No. 2, County Emergency Hospital, 300041 Timisoara, Romania
- Center for Molecular Research in Nephrology and Vascular Disease, Faculty of Medicine, "Victor Babes" University of Medicine and Pharmacy, Eftimie Murgu Sq. No. 2, 300041 Timisoara, Romania
| | - Silvia Ienciu
- Department of Internal Medicine II-Division of Nephrology, "Victor Babes" University of Medicine and Pharmacy, Eftimie Murgu Sq. No. 2, County Emergency Hospital, 300041 Timisoara, Romania
- Center for Molecular Research in Nephrology and Vascular Disease, Faculty of Medicine, "Victor Babes" University of Medicine and Pharmacy, Eftimie Murgu Sq. No. 2, 300041 Timisoara, Romania
| | - Maria Mogos
- Department of Internal Medicine II-Division of Nephrology, "Victor Babes" University of Medicine and Pharmacy, Eftimie Murgu Sq. No. 2, County Emergency Hospital, 300041 Timisoara, Romania
- Center for Molecular Research in Nephrology and Vascular Disease, Faculty of Medicine, "Victor Babes" University of Medicine and Pharmacy, Eftimie Murgu Sq. No. 2, 300041 Timisoara, Romania
| | - Dragos Catalin Jianu
- Center for Molecular Research in Nephrology and Vascular Disease, Faculty of Medicine, "Victor Babes" University of Medicine and Pharmacy, Eftimie Murgu Sq. No. 2, 300041 Timisoara, Romania
- Department of Neurosciences-Division of Neurology, "Victor Babes" University of Medicine and Pharmacy, Eftimie Murgu Sq. No. 2, County Emergency Hospital, 300041 Timisoara, Romania
- Center for Cognitive Research in Neuropsychiatric Pathology (Neuropsy-Cog), Faculty of Medicine, "Victor Babes" University of Medicine and Pharmacy, Eftimie Murgu Sq. No. 2, 300041 Timisoara, Romania
| | - Ligia Petrica
- Department of Internal Medicine II-Division of Nephrology, "Victor Babes" University of Medicine and Pharmacy, Eftimie Murgu Sq. No. 2, County Emergency Hospital, 300041 Timisoara, Romania
- Center for Molecular Research in Nephrology and Vascular Disease, Faculty of Medicine, "Victor Babes" University of Medicine and Pharmacy, Eftimie Murgu Sq. No. 2, 300041 Timisoara, Romania
- Center for Cognitive Research in Neuropsychiatric Pathology (Neuropsy-Cog), Faculty of Medicine, "Victor Babes" University of Medicine and Pharmacy, Eftimie Murgu Sq. No. 2, 300041 Timisoara, Romania
- Center for Translational Research and Systems Medicine, Faculty of Medicine, "Victor Babes" University of Medicine and Pharmacy, Eftimie, Murgu Sq. No. 2, 300041 Timisoara, Romania
| |
Collapse
|
9
|
Ortiz SR, Field MS. Sucrose Intake Elevates Erythritol in Plasma and Urine in Male Mice. J Nutr 2023; 153:1889-1902. [PMID: 37245661 DOI: 10.1016/j.tjnut.2023.05.022] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 05/15/2023] [Accepted: 05/23/2023] [Indexed: 05/30/2023] Open
Abstract
BACKGROUND Elevated serum erythritol concentration is a predictive biomarker of diabetes and cardiovascular incidence and complications. Erythritol is synthesized endogenously from glucose, but little is known regarding the origin of elevated circulating erythritol in vivo. OBJECTIVES In vitro evidence indicates that intracellular erythritol is elevated by high-glucose cell culture conditions and that final step of erythritol synthesis is catalyzed by the enzymes sorbitol dehydrogenase (SORD) and alcohol dehydrogenase (ADH) 1. The purpose of this study was to determine whether dietary intake and/or diet-induced obesity affect erythritol synthesis in mice and whether this relationship is modified by the loss of the enzymes SORD or ADH1. METHODS First, 8-wk-old male Sord+/+, Sord-/-, Adh1+/+, and Adh1-/- mice were fed either low-fat diet (LFD) with 10% fat-derived calories or diet-induced obesity high-fat diet (HFD) with 60% fat-derived calories for 8 wk. Plasma and tissue erythritol concentrations were measured using gas chromatography-mass spectrometry. Second, male wild-type 8-wk-old C57BL/6J mice were fed LFD or HFD with plain drinking water or 30% sucrose water for 8 wk. Blood glucose and plasma and urinary erythritol concentrations were measured in nonfasted and fasted samples. Tissue erythritol was measured after killing. Finally, male Sord+/+ and Sord-/- mice were fed LFD with 30% sucrose water for 2 wk; then, nonfasted plasma, urine, and tissue erythritol concentrations were quantified. RESULTS Plasma and tissue erythritol concentrations were not affected by loss of Sord or Adh1 in mice fed LFD or HFD. In wild-type mice, consumption of 30% sucrose water significantly elevated plasma and urinary erythritol concentrations on both LFD-fed and HFD-fed mice compared with that of plain water. Sord genotype did not affect plasma or urinary erythritol concentration in response to sucrose feeding, but Sord-/- mice had reduced kidney erythritol content compared with wild-type littermates in response to sucrose. CONCLUSIONS Sucrose intake, not HFD, elevates erythritol synthesis and excretion in mice. Loss of ADH1 or SORD does not significantly affect erythritol concentration in mice.
Collapse
Affiliation(s)
- Semira R Ortiz
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, USA
| | - Martha S Field
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, USA.
| |
Collapse
|
10
|
Aihara S, Torisu K, Hirashima Y, Kitazono T, Nakano T. Acrolein produced during acute kidney injury promotes tubular cell death. Biochem Biophys Res Commun 2023; 666:137-145. [PMID: 37187091 DOI: 10.1016/j.bbrc.2023.05.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 04/28/2023] [Accepted: 05/08/2023] [Indexed: 05/17/2023]
Abstract
Acute kidney injury is an important global health concern as it is associated with high morbidity and mortality. Polyamines, essential for cell growth and proliferation, are known to inhibit cardiovascular disease. However, under conditions of cellular damage, toxic acrolein is produced from polyamines by the enzyme spermine oxidase (SMOX). We used a mouse renal ischemia-reperfusion model and human proximal tubule cells (HK-2) to investigate whether acrolein exacerbates acute kidney injury by renal tubular cell death. Acrolein visualized by acroleinRED was increased in ischemia-reperfusion kidneys, particularly in tubular cells. When HK-2 cells were cultured under 1% oxygen for 24 h, then switched to 21% oxygen for 24 h (hypoxia-reoxygenation), acrolein accumulated and SMOX mRNA and protein levels were increased. Acrolein induced cell death and fibrosis-related TGFB1 mRNA in HK-2 cells. Administration of the acrolein scavenger cysteamine suppressed the acrolein-induced upregulation of TGFB1 mRNA. Cysteamine also inhibited a decrease in the mitochondrial membrane potential observed by MitoTrackerCMXRos, and cell death induced by hypoxia-reoxygenation. The siRNA-mediated knockdown of SMOX also suppressed hypoxia-reoxygenation-induced acrolein accumulation and cell death. Our study suggests that acrolein exacerbates acute kidney injury by promoting tubular cell death during ischemia-reperfusion injury. Treatment to control the accumulation of acrolein might be an effective therapeutic option for renal ischemia-reperfusion injury.
Collapse
Affiliation(s)
- Seishi Aihara
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.
| | - Kumiko Torisu
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan; Department of Integrated Therapy for Chronic Kidney Disease, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.
| | - Yutaro Hirashima
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.
| | - Takanari Kitazono
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.
| | - Toshiaki Nakano
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan; Center for Cohort Studies, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, Japan.
| |
Collapse
|
11
|
Ortiz SR, Heinz A, Hiller K, Field MS. Erythritol synthesis is elevated in response to oxidative stress and regulated by the non-oxidative pentose phosphate pathway in A549 cells. Front Nutr 2022; 9:953056. [PMID: 36276829 PMCID: PMC9582529 DOI: 10.3389/fnut.2022.953056] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 09/20/2022] [Indexed: 11/30/2022] Open
Abstract
Background Erythritol is a predictive biomarker of cardiometabolic diseases and is produced from glucose metabolism through the pentose phosphate pathway (PPP). Little is known regarding the regulation of endogenous erythritol synthesis in humans. Objective In the present study, we investigated the stimuli that promote erythritol synthesis in human lung carcinoma cells and characterized potential points of regulation along the PPP. Methods Human A549 lung carcinoma cells were chosen for their known ability to synthesize erythritol. A549 cells were treated with potential substrates for erythritol production, including glucose, fructose, and glycerol. Using siRNA knockdown, we assessed the necessity of enzymes G6PD, TKT, TALDO, and SORD for erythritol synthesis. We also used position-specific 13C-glucose tracers to determine whether the carbons for erythritol synthesis are derived directly from glycolysis or through the oxidative PPP. Finally, we assessed if erythritol synthesis responds to oxidative stress using chemical and genetic models. Results Intracellular erythritol was directly associated with media glucose concentration. In addition, siRNA knockdown of TKT or SORD inhibited erythritol synthesis, whereas siG6PD did not. Both chemically induced oxidative stress and constitutive activation of the antioxidant response transcription factor NRF2 elevated intracellular erythritol. Conclusion Our findings indicate that in A549 cells, erythritol synthesis is proportional to flux through the PPP and is regulated by non-oxidative PPP enzymes.
Collapse
Affiliation(s)
- Semira R. Ortiz
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, United States
| | - Alexander Heinz
- Department of Bioinformatics and Biochemistry, Braunschweig Integrated Centre of Systems Biology (BRICS), Technische Universität Braunschweig, Braunschweig, Germany
| | - Karsten Hiller
- Department of Bioinformatics and Biochemistry, Braunschweig Integrated Centre of Systems Biology (BRICS), Technische Universität Braunschweig, Braunschweig, Germany
| | - Martha S. Field
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, United States,*Correspondence: Martha S. Field,
| |
Collapse
|
12
|
Chen J, Peng H, Chen C, Wang Y, Sang T, Cai Z, Zhao Q, Chen S, Lin X, Eling T, Wang X. NAG-1/GDF15 inhibits diabetic nephropathy via inhibiting AGE/RAGE-mediated inflammation signaling pathways in C57BL/6 mice and HK-2 cells. Life Sci 2022; 311:121142. [DOI: 10.1016/j.lfs.2022.121142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 10/16/2022] [Accepted: 10/27/2022] [Indexed: 11/07/2022]
|
13
|
Metabolic Adaptation as Potential Target in Papillary Renal Cell Carcinomas Based on Their In Situ Metabolic Characteristics. Int J Mol Sci 2022; 23:ijms231810587. [PMID: 36142502 PMCID: PMC9503093 DOI: 10.3390/ijms231810587] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 09/07/2022] [Accepted: 09/09/2022] [Indexed: 11/24/2022] Open
Abstract
Metabolic characteristics of kidney cancers have mainly been obtained from the most frequent clear cell renal cell carcinoma (CCRCC) studies. Moreover, the bioenergetic perturbances that affect metabolic adaptation possibilities of papillary renal cell carcinoma (PRCC) have not yet been detailed. Therefore, our study aimed to analyze the in situ metabolic features of PRCC vs. CCRCC tissues and compared the metabolic characteristics of PRCC, CCRCC, and normal tubular epithelial cell lines. The protein and mRNA expressions of the molecular elements in mammalian target of rapamycin (mTOR) and additional metabolic pathways were analyzed in human PRCC cases compared to CCRCC. The metabolic protein expression pattern, metabolite content, mTOR, and metabolic inhibitor sensitivity of renal carcinoma cell lines were also studied and compared with tubular epithelial cells, as “normal” control. We observed higher protein expressions of the “alternative bioenergetic pathway” elements, in correlation with the possible higher glutamine and acetate consumption in PRCC cells instead of higher glycolytic and mTOR activity in CCRCCs. Increased expression of certain metabolic pathway markers correlates with the detected differences in metabolite ratios, as well. The lower lactate/pyruvate, lactate/malate, and higher pyruvate/citrate intracellular metabolite ratios in PRCC compared to CCRCC cell lines suggest that ACHN (PRCC) have lower Warburg glycolytic capacity, less pronounced pyruvate to lactate producing activity and shifted OXPHOS phenotype. However, both studied renal carcinoma cell lines showed higher mTOR activity than tubular epithelial cells cultured in vitro, the metabolite ratio, the enzyme expression profiles, and the higher mitochondrial content also suggest increased importance of mitochondrial functions, including mitochondrial OXPHOS in PRCCs. Additionally, PRCC cells showed significant mTOR inhibitor sensitivity and the used metabolic inhibitors increased the effect of rapamycin in combined treatments. Our study revealed in situ metabolic differences in mTOR and metabolic protein expression patterns of human PRCC and CCRCC tissues as well as in cell lines. These underline the importance in the development of specific new treatment strategies, new mTOR inhibitors, and other anti-metabolic drug combinations in PRCC therapy.
Collapse
|
14
|
Wang M, Pang Y, Guo Y, Tian L, Liu Y, Shen C, Liu M, Meng Y, Cai Z, Wang Y, Zhao W. Metabolic reprogramming: A novel therapeutic target in diabetic kidney disease. Front Pharmacol 2022; 13:970601. [PMID: 36120335 PMCID: PMC9479190 DOI: 10.3389/fphar.2022.970601] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 08/01/2022] [Indexed: 11/13/2022] Open
Abstract
Diabetic kidney disease (DKD) is one of the most common microvascular complications of diabetes mellitus. However, the pathological mechanisms contributing to DKD are multifactorial and poorly understood. Diabetes is characterized by metabolic disorders that can bring about a series of changes in energy metabolism. As the most energy-consuming organs secondary only to the heart, the kidneys must maintain energy homeostasis. Aberrations in energy metabolism can lead to cellular dysfunction or even death. Metabolic reprogramming, a shift from mitochondrial oxidative phosphorylation to glycolysis and its side branches, is thought to play a critical role in the development and progression of DKD. This review focuses on the current knowledge about metabolic reprogramming and the role it plays in DKD development. The underlying etiologies, pathological damages in the involved cells, and potential molecular regulators of metabolic alterations are also discussed. Understanding the role of metabolic reprogramming in DKD may provide novel therapeutic approaches to delay its progression to end-stage renal disease.
Collapse
|
15
|
Oxidative Stress and Lipid Dysregulation in Lipid Droplets: A Connection to Chronic Kidney Disease Revealed in Human Kidney Cells. Antioxidants (Basel) 2022; 11:antiox11071387. [PMID: 35883878 PMCID: PMC9312214 DOI: 10.3390/antiox11071387] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 07/10/2022] [Accepted: 07/14/2022] [Indexed: 02/04/2023] Open
Abstract
Chronic kidney disease (CKD), which is defined as a condition causing the gradual loss of kidney function, shows renal lipid droplet (LD) accumulation that is associated with oxidative damage. There is a possibility that an LD abnormality in quality plays a role in CKD development. This study aimed to explore the chemical composition of LDs that are induced in human kidney cells during exposure to free fatty acids as an LD source and oxidized lipoproteins as oxidative stress. The LDs were aspirated directly from cells using nanotips, followed by in-tip microextraction, and the LD lipidomic profiling was conducted using nanoelectrospray mass spectrometry. As a result, the free fatty acids increased the LD lipid content and, at the same time, changed their composition significantly. The oxidized lipoproteins caused distorted proportions of intact lipids, such as triacylglycerols (TG), phosphatidylcholines (PC), phosphatidylethanolamines (PE), and cholesteryl esters (CE). Notably, the oxidized lipids, including the hydroperoxides of TG, PC, and PE, exhibited significant elevations in dose-dependent manners. Furthermore, the dysregulation of intact lipids was paralleled with the accumulation of lipid hydroperoxides. The present study has revealed that the oxidation of lipids and the dysregulation of the lipid metabolism coexisted in LDs in the kidney cells, which has provided a potential new target for diagnosis and new insights into CKD.
Collapse
|
16
|
Abstract
Altered lipid metabolism is a characteristic feature and potential driving factor of acute kidney injury (AKI). Of the lipids that accumulate in injured renal tissues, ceramides are potent regulators of metabolism and cell fate. Up-regulation of ceramide synthesis is a common feature shared across several AKI etiologies in vitro and in vivo. Furthermore, ceramide accumulation is an early event in the natural history of AKI that precedes cell death and organ dysfunction. Emerging evidence suggests that inhibition of ceramide accumulation may improve renal outcomes in several models of AKI. This review examines the landscape of ceramide metabolism and regulation in the healthy and injured kidney. Furthermore, we discuss the body of literature regarding ceramides as therapeutic targets for AKI and consider potential mechanisms by which ceramides drive kidney pathogenesis.
Collapse
Affiliation(s)
- Rebekah J Nicholson
- Department of Nutrition and Integrative Physiology, Diabetes and Metabolism Research Center, University of Utah, Salt Lake City, UT
| | - William L Holland
- Department of Nutrition and Integrative Physiology, Diabetes and Metabolism Research Center, University of Utah, Salt Lake City, UT
| | - Scott A Summers
- Department of Nutrition and Integrative Physiology, Diabetes and Metabolism Research Center, University of Utah, Salt Lake City, UT.
| |
Collapse
|
17
|
Li H, Zhang H, Yan F, He Y, Ji A, Liu Z, Li M, Ji X, Li C. Kidney and plasma metabolomics provide insights into the molecular mechanisms of urate nephropathy in a mouse model of hyperuricemia. Biochim Biophys Acta Mol Basis Dis 2022; 1868:166374. [PMID: 35276331 DOI: 10.1016/j.bbadis.2022.166374] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 02/04/2022] [Accepted: 03/04/2022] [Indexed: 02/07/2023]
Abstract
Hyperuricemia (HUA) is closely associated with kidney damage and kidney diseases in humans; however, the underlying mechanisms of HUA-induced kidney diseases remain unknown. In the present study, we examined the kidney and plasma metabolic profiles in a HUA mouse model constructed by knocking out (Ko) the urate oxidase (Uox) gene. The Uox-Ko mice were characterized by an increase in uric acid, glycine, 3'-adenosine monophosphate, citrate, N-acetyl-l-glutamate, l-kynurenine, 5-hydroxyindoleacetate, xanthurenic acid, cortisol, and (-)-prostaglandin e2 together with a decrease of inosine in the kidneys. These altered metabolites confirmed disturbances of purine metabolism, amino acid biosynthesis, tryptophan metabolism, and neuroactive ligand-receptor interaction in Uox-Ko mice. Betaine and biotin were related to kidney function and identified as the potential plasma metabolic biomarker for predicting urate nephropathy (UN). Taken together, these results revealed the underlying pathogenic mechanisms of UN. Investigating these pathways might provide novel targets for the therapeutic intervention of UN and can potentially lead to new treatment strategies.
Collapse
Affiliation(s)
- Hailong Li
- Institute of Metabolic Diseases, Qingdao University, Qingdao 266003, China; Shandong Provincial Key Laboratory of Metabolic Disease and Qingdao Key Laboratory of Gout, The Affiliated Hospital of Qingdao University, Qingdao 266003, China
| | - Hui Zhang
- Institute of Metabolic Diseases, Qingdao University, Qingdao 266003, China; Shandong Provincial Key Laboratory of Metabolic Disease and Qingdao Key Laboratory of Gout, The Affiliated Hospital of Qingdao University, Qingdao 266003, China
| | - Fei Yan
- Shandong Provincial Key Laboratory of Metabolic Disease and Qingdao Key Laboratory of Gout, The Affiliated Hospital of Qingdao University, Qingdao 266003, China
| | - Yuwei He
- Shandong Provincial Key Laboratory of Metabolic Disease and Qingdao Key Laboratory of Gout, The Affiliated Hospital of Qingdao University, Qingdao 266003, China
| | - Aichang Ji
- Shandong Provincial Key Laboratory of Metabolic Disease and Qingdao Key Laboratory of Gout, The Affiliated Hospital of Qingdao University, Qingdao 266003, China
| | - Zhen Liu
- Shandong Provincial Key Laboratory of Metabolic Disease and Qingdao Key Laboratory of Gout, The Affiliated Hospital of Qingdao University, Qingdao 266003, China
| | - Maichao Li
- Shandong Provincial Key Laboratory of Metabolic Disease and Qingdao Key Laboratory of Gout, The Affiliated Hospital of Qingdao University, Qingdao 266003, China
| | - Xiaopeng Ji
- Shandong Provincial Key Laboratory of Metabolic Disease and Qingdao Key Laboratory of Gout, The Affiliated Hospital of Qingdao University, Qingdao 266003, China
| | - Changgui Li
- Institute of Metabolic Diseases, Qingdao University, Qingdao 266003, China; Shandong Provincial Key Laboratory of Metabolic Disease and Qingdao Key Laboratory of Gout, The Affiliated Hospital of Qingdao University, Qingdao 266003, China.
| |
Collapse
|
18
|
Untargeted Metabolomics Analysis of the Serum Metabolic Signature of Childhood Obesity. Nutrients 2022; 14:nu14010214. [PMID: 35011090 PMCID: PMC8747180 DOI: 10.3390/nu14010214] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 12/29/2021] [Accepted: 12/31/2021] [Indexed: 02/04/2023] Open
Abstract
Obesity rates among children are growing rapidly worldwide, placing massive pressure on healthcare systems. Untargeted metabolomics can expand our understanding of the pathogenesis of obesity and elucidate mechanisms related to its symptoms. However, the metabolic signatures of obesity in children have not been thoroughly investigated. Herein, we explored metabolites associated with obesity development in childhood. Untargeted metabolomic profiling was performed on fasting serum samples from 27 obese Caucasian children and adolescents and 15 sex- and age-matched normal-weight children. Three metabolomic assays were combined and yielded 726 unique identified metabolites: gas chromatography–mass spectrometry (GC–MS), hydrophilic interaction liquid chromatography coupled to mass spectrometry (HILIC LC–MS/MS), and lipidomics. Univariate and multivariate analyses showed clear discrimination between the untargeted metabolomes of obese and normal-weight children, with 162 significantly differentially expressed metabolites between groups. Children with obesity had higher concentrations of branch-chained amino acids and various lipid metabolites, including phosphatidylcholines, cholesteryl esters, triglycerides. Thus, an early manifestation of obesity pathogenesis and its metabolic consequences in the serum metabolome are correlated with altered lipid metabolism. Obesity metabolite patterns in the adult population were very similar to the metabolic signature of childhood obesity. Identified metabolites could be potential biomarkers and used to study obesity pathomechanisms.
Collapse
|
19
|
Study of the potential neuroprotective effect of Dunaliella salina extract in SH-SY5Y cell model. Anal Bioanal Chem 2021; 414:5357-5371. [PMID: 34923590 PMCID: PMC9242911 DOI: 10.1007/s00216-021-03819-1] [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] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 11/03/2021] [Accepted: 12/01/2021] [Indexed: 01/13/2023]
Abstract
Alzheimer’s disease (AD) is the most common form of dementia caused by a progressive loss of neurons from different regions of the brain. This multifactorial pathophysiology has been widely characterized by neuroinflammation, extensive oxidative damage, synaptic loss, and neuronal cell death. In this sense, the design of multi-target strategies to prevent or delay its progression is a challenging goal. In the present work, different in vitro assays including antioxidant, anti-inflammatory, and anti-cholinergic activities of a carotenoid-enriched extract from Dunaliella salina microalgae obtained by supercritical fluid extraction are studied. Moreover, its potential neuroprotective effect in the human neuron-like SH-SY5Y cell model against remarkable hallmarks of AD was also evaluated. In parallel, a comprehensive metabolomics study based on the use of charged-surface hybrid chromatography (CSH) and hydrophilic interaction liquid chromatography (HILIC) coupled to high-resolution tandem mass spectrometry (Q-TOF MS/MS) was applied to evaluate the effects of the extract on the metabolism of the treated cells. The use of advanced bioinformatics and statistical tools allowed the identification of more than 314 metabolites in SH-SY5Y cells, of which a great number of phosphatidylcholines, triacylglycerols, and fatty acids were significantly increased, while several phosphatidylglycerols were decreased, compared to controls. These lipidomic changes in cells along with the possible role exerted by carotenoids and other minor compounds on the cell membrane might explain the observed neuroprotective effect of the D. salina extract. However, future experiments using in vivo models to corroborate this hypothesis must be carried out.
Collapse
|
20
|
Yun J, Ren J, Liu Y, Dai L, Song L, Ma X, Luo S, Song Y. MicroRNA (miR)-590-3p alleviates high-glucose induced renal tubular epithelial cell damage by targeting C-X3-C motif chemokine ligand 1 (CX3CL1) in diabetic nephropathy. Bioengineered 2021; 13:634-644. [PMID: 34898373 PMCID: PMC8805927 DOI: 10.1080/21655979.2021.2012548] [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] [Indexed: 11/11/2022] Open
Abstract
We attempted to analyze the clinical value of microRNA (miR)-590-3p in diabetic nephropathy (DN) patients and its role in high glucose (HG)-induced renal tubular epithelial cell (HK-2) injury. Serum levels of miR-590-3p were detected by quantitative real-time polymerase chain reaction (qRT-PCR). Spearman correlation coefficient analysis of the correlation between miR-590-3p and clinical indicators. The diagnostic value of miR-590-3p was analyzed by the receiver operating characteristic (ROC) curve. Then, the DN cell model induced by HG in HK-2 cells was established. Enzyme-linked immunosorbent assay (ELISA), flow cytometry, and CCK-8 assay were employed to assess cell inflammation, oxidative stress, apoptosis, and proliferation. Dual-luciferase reporter assay confirmed the target of miR-590-3p. Serum miR-590-3p was reduced in patients of DN, which was positively correlated with eGFR and negatively associated with albuminuria. Furthermore, miR-590-3p also can diagnose patients of DN from healthy subjects or patients of T2DM. Furthermore, miR-590-3p was decreased in a concentration- and time-dependent manner during HG-induction. miR-590-3p overexpression bated HG-induced inhibition effect on cell proliferation and promotion effects on apoptosis, oxidative stress, and inflammation. C-X3-C motif chemokine ligand1 (CX3CL1) is the target of miR-590-3p, whose levels were enhanced in DN patients and are negatively regulated by miR-590-3p. Our discoveries offered new insights that reduced miR-590-3p as a potential biomarker for the diagnosis of DN, and elevated miR-590-3p can alleviate renal tubular injury by HG-induced through targeting CX3XL1, which may be a novel target for improving the development of DN.
Collapse
Affiliation(s)
- Jie Yun
- Department of Nephrology, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Heilongjiang, China
| | - Jinyu Ren
- Department of Encephalopathy, Second Hospital Affiliated to Heilongjiang University of Chinese Medicine, Heilongjiang, China
| | - Yufei Liu
- Department of Blood Purification, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Heilongjiang, China
| | - Lijuan Dai
- Department of Nephrology, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Heilongjiang, China
| | - Liqun Song
- Department of Nephrology, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Heilongjiang, China
| | - Xiaopeng Ma
- Department of Nephrology, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Heilongjiang, China
| | - Shan Luo
- Department of Nephrology, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Heilongjiang, China
| | - Yexu Song
- Department of Science and Technology, Heilongjiang University of Chinese Medicine, Heilongjiang, China
| |
Collapse
|
21
|
Gallego R, Suárez-Montenegro ZJ, Ibáñez E, Herrero M, Valdés A, Cifuentes A. In vitro Neuroprotective Potential and Lipidomics Study of Olive Leaves Extracts Enriched in Triterpenoids. Front Nutr 2021; 8:769218. [PMID: 34708068 PMCID: PMC8542692 DOI: 10.3389/fnut.2021.769218] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 09/15/2021] [Indexed: 11/13/2022] Open
Abstract
Alzheimer's Disease (AD) is the most common form of dementia that is associated with extracellular amyloid beta (Aβ) plaque formation. Genetic, environmental, and nutrition factors have been suggested as contributors to oxidative stress and neuroinflammation events that are connected to AD etiology, and secondary metabolites, such as triterpenes, have shown promising results in AD prevention. In this work, the neuroprotective and anti-inflammatory potential of an olive leaves fraction enriched in triterpenoid compounds obtained using supercritical fluid extraction (SFE) and dynamic adsorption/desorption using sea sand as adsorbent has been performed. In addition, a comprehensive lipidomics study of the response of SH-SY5Y neuroblastoma cell line to this fraction was carried out using advanced analytical methodologies, namely, charged-surface hybrid chromatography-quadrupole-time of flight mass spectrometry (CSH-Q-TOF MS/MS). The use of freely available lipidomic annotation tools and databases, and stringent cut-off filters allowed the annotation of more than 250 intracellular lipids. Advanced bioinformatics and statistical tools showed a number of phosphatidylcholines and phosphatidylethanolamines significantly increased, which could explain the protection against the cell death caused by Aβ1-42. Moreover, several triacylglycerols were found decreased. These results suggest triterpenoids from olive leaves as good neuroprotective candidates, and open a new gate for future experiments using in vivo models to corroborate this hypothesis.
Collapse
Affiliation(s)
- Rocío Gallego
- Foodomics Laboratory, Institute of Food Science Research (CIAL, CSIC), Madrid, Spain
| | - Zully J. Suárez-Montenegro
- Foodomics Laboratory, Institute of Food Science Research (CIAL, CSIC), Madrid, Spain
- Departamento de Procesos Industriales, Facultad de Ingeniería Agroindustrial, Universidad de Nariño, Pasto, Colombia
| | - Elena Ibáñez
- Foodomics Laboratory, Institute of Food Science Research (CIAL, CSIC), Madrid, Spain
| | - Miguel Herrero
- Foodomics Laboratory, Institute of Food Science Research (CIAL, CSIC), Madrid, Spain
| | - Alberto Valdés
- Foodomics Laboratory, Institute of Food Science Research (CIAL, CSIC), Madrid, Spain
| | - Alejandro Cifuentes
- Foodomics Laboratory, Institute of Food Science Research (CIAL, CSIC), Madrid, Spain
| |
Collapse
|
22
|
Exploratory Metabolomic Analysis Based on Reversed-Phase Liquid Chromatography-Mass Spectrometry to Study an In Vitro Model of Hypoxia-Induced Metabolic Alterations in HK-2 Cells. Int J Mol Sci 2021; 22:ijms22147399. [PMID: 34299017 PMCID: PMC8304667 DOI: 10.3390/ijms22147399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 06/29/2021] [Accepted: 07/07/2021] [Indexed: 11/17/2022] Open
Abstract
Oxygen deficiency in cells, tissues, and organs can not only prevent the proper development of biological functions but it can also lead to several diseases and disorders. In this sense, the kidney deserves special attention since hypoxia can be considered an important factor in the pathophysiology of both acute kidney injury and chronic kidney disease. To provide better knowledge to unveil the molecular mechanisms involved, new studies are necessary. In this sense, this work aims to study, for the first time, an in vitro model of hypoxia-induced metabolic alterations in human proximal tubular HK-2 cells because renal proximal tubules are particularly susceptible to hypoxia. Different groups of cells, cultivated under control and hypoxia conditions at 0.5, 5, 24, and 48 h, were investigated using untargeted metabolomic approaches based on reversed-phase liquid chromatography–mass spectrometry. Both intracellular and extracellular fluids were studied to obtain a large metabolite coverage. On the other hand, multivariate and univariate analyses were carried out to find the differences among the cell groups and to select the most relevant variables. The molecular features identified as affected metabolites were mainly amino acids and Amadori compounds. Insights about their biological relevance are also provided.
Collapse
|