51
|
Morevati M, Fang EF, Mace ML, Kanbay M, Gravesen E, Nordholm A, Egstrand S, Hornum M. Roles of NAD + in Acute and Chronic Kidney Diseases. Int J Mol Sci 2022; 24:ijms24010137. [PMID: 36613582 PMCID: PMC9820289 DOI: 10.3390/ijms24010137] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 12/08/2022] [Accepted: 12/13/2022] [Indexed: 12/24/2022] Open
Abstract
Nicotinamide adenine dinucleotide (oxidized form, NAD+) is a critical coenzyme, with functions ranging from redox reactions and energy metabolism in mitochondrial respiration and oxidative phosphorylation to being a central player in multiple cellular signaling pathways, organ resilience, health, and longevity. Many of its cellular functions are executed via serving as a co-substrate for sirtuins (SIRTs), poly (ADP-ribose) polymerases (PARPs), and CD38. Kidney damage and diseases are common in the general population, especially in elderly persons and diabetic patients. While NAD+ is reduced in acute kidney injury (AKI) and chronic kidney disease (CKD), mounting evidence indicates that NAD+ augmentation is beneficial to AKI, although conflicting results exist for cases of CKD. Here, we review recent progress in the field of NAD+, mainly focusing on compromised NAD+ levels in AKI and its effect on essential cellular pathways, such as mitochondrial dysfunction, compromised autophagy, and low expression of the aging biomarker αKlotho (Klotho) in the kidney. We also review the compromised NAD+ levels in renal fibrosis and senescence cells in the case of CKD. As there is an urgent need for more effective treatments for patients with injured kidneys, further studies on NAD+ in relation to AKI/CKD may shed light on novel therapeutics.
Collapse
Affiliation(s)
- Marya Morevati
- Department of Nephrology, Rigshospitalet, University of Copenhagen, 2100 Copenhagen, Denmark
- Correspondence:
| | - Evandro Fei Fang
- Department of Clinical Molecular Biology, University of Oslo and Akershus University Hospital, 1478 Lørenskog, Norway
| | - Maria L. Mace
- Department of Nephrology, Rigshospitalet, University of Copenhagen, 2100 Copenhagen, Denmark
| | - Mehmet Kanbay
- Division of Nephrology, Department of Medicine, Koç University School of Medicine, Istanbul 34010, Turkey
| | - Eva Gravesen
- Department of Pathology, Herlev Hospital, University of Copenhagen, 2730 Copenhagen, Denmark
| | - Anders Nordholm
- Department of Nephrology, Rigshospitalet, University of Copenhagen, 2100 Copenhagen, Denmark
| | - Søren Egstrand
- Department of Nephrology, Rigshospitalet, University of Copenhagen, 2100 Copenhagen, Denmark
| | - Mads Hornum
- Department of Nephrology, Rigshospitalet, University of Copenhagen, 2100 Copenhagen, Denmark
| |
Collapse
|
52
|
An Untargeted Metabolomics Approach on Carfilzomib-Induced Nephrotoxicity. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27227929. [PMID: 36432029 PMCID: PMC9697636 DOI: 10.3390/molecules27227929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 11/14/2022] [Accepted: 11/15/2022] [Indexed: 11/19/2022]
Abstract
BACKGROUND Carfilzomib (Cfz) is an anti-cancer drug related to cardiorenal adverse events, with cardiovascular and renal complications limiting its clinical use. Despite the important progress concerning the discovery of the underlying causes of Cfz-induced nephrotoxicity, the molecular/biochemical background is still not well clarified. Furthermore, the number of metabolomics-based studies concerning Cfz-induced nephrotoxicity is limited. METHODS A metabolomics UPLC-HRMS-DIA methodology was applied to three bio-sample types i.e., plasma, kidney, and urine, obtained from two groups of mice, namely (i) Cfz (8 mg Cfz/ kg) and (ii) Control (0.9% NaCl) (n = 6 per group). Statistical analysis, involving univariate and multivariate tools, was applied for biomarker detection. Furthermore, a sub-study was developed, aiming to estimate metabolites' correlation among bio-samples, and to enlighten potential mechanisms. RESULTS Cfz mostly affects the kidneys and urine metabolome. Fifty-four statistically important metabolites were discovered, and some of them have already been related to renal diseases. Furthermore, the correlations between bio-samples revealed patterns of metabolome alterations due to Cfz. CONCLUSIONS Cfz causes metabolite retention in kidney and dysregulates (up and down) several metabolites associated with the occurrence of inflammation and oxidative stress.
Collapse
|
53
|
Ou-Yang YN, Yuan MD, Yang ZM, Min Z, Jin YX, Tian ZM. Revealing the Pathogenesis of Salt-Sensitive Hypertension in Dahl Salt-Sensitive Rats through Integrated Multi-Omics Analysis. Metabolites 2022; 12:1076. [PMID: 36355159 PMCID: PMC9694938 DOI: 10.3390/metabo12111076] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 11/03/2022] [Accepted: 11/04/2022] [Indexed: 10/18/2023] Open
Abstract
Salt-induced renal metabolism dysfunction is an important mechanism of salt-sensitive hypertension. Given that the gut-liver axis is the first hit of a high-salt diet (HSD), we aimed to identify the extra-renal mechanism from hepatic metabolism and gut microbiota, and attempted to relieve the salt-induced metabolic dysfunctions by curcumin. Untargeted metabolomics analysis was performed to identify the changes in hepatic metabolic pathways, and integrated analysis was employed to reveal the relationship between hepatic metabolic dysfunction and gut microbial composition. HSD induced significant increase in fumaric acid, l-lactic acid, creatinine, l-alanine, glycine, and l-cysteine levels, and amino acids metabolism pathways associated with glycolysis were significantly altered, including alanine, aspartate, and glutamate metabolism; glycine, serine, and threonine metabolism, which were involved in the regulation of blood pressure. Integrated multi-omics analysis revealed that changes in Paraprevotella, Erysipelotrichaceae, and genera from Clostridiales are associated with metabolic disorders. Gene functional predication analysis based on 16S Ribosomal RNA sequences showed that the dysfunction in hepatic metabolism were correlated with enhanced lipopolysaccharide (LPS) biosynthesis and apoptosis in gut microbes. Curcumin (50 mg/kg/d) might reduce gut microbes-associated LPS biosynthesis and apoptosis, partially reverse metabolic dysfunction, ameliorate renal oxidative stress, and protect against salt-sensitive hypertension.
Collapse
Affiliation(s)
- Ya-nan Ou-Yang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an 710049, China
| | - Meng-di Yuan
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an 710049, China
| | | | - Zhuo Min
- Department of Brewing Engineering, Moutai University, Renhuai 564500, China
| | - Yue-xin Jin
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an 710049, China
| | - Zhong-min Tian
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an 710049, China
| |
Collapse
|
54
|
Doke T, Susztak K. The multifaceted role of kidney tubule mitochondrial dysfunction in kidney disease development. Trends Cell Biol 2022; 32:841-853. [PMID: 35473814 PMCID: PMC9464682 DOI: 10.1016/j.tcb.2022.03.012] [Citation(s) in RCA: 52] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 03/27/2022] [Accepted: 03/30/2022] [Indexed: 12/24/2022]
Abstract
More than 800 million people suffer from kidney disease. Genetic studies and follow-up animal models and cell biological experiments indicate the key role of proximal tubule metabolism. Kidneys have one of the highest mitochondrial densities. Mitochondrial biogenesis, mitochondrial fusion and fission, and mitochondrial recycling, such as mitophagy are critical for proper mitochondrial function. Mitochondrial dysfunction can lead to an energetic crisis, orchestrate different types of cell death (apoptosis, necroptosis, pyroptosis, and ferroptosis), and influence cellular calcium levels and redox status. Collectively, mitochondrial defects in renal tubules contribute to epithelial atrophy, inflammation, or cell death, orchestrating kidney disease development.
Collapse
Affiliation(s)
- Tomohito Doke
- Department of Medicine, Renal Electrolyte and Hypertension Division, University of Pennsylvania, Philadelphia, PA, USA; Department of Genetics, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA.
| | - Katalin Susztak
- Department of Medicine, Renal Electrolyte and Hypertension Division, University of Pennsylvania, Philadelphia, PA, USA; Department of Genetics, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA.
| |
Collapse
|
55
|
Zeng L, Chen M, Ahmad H, Zheng X, Ouyang Y, Yang P, Yang Z, Gao D, Tian Z. Momordica charantia Extract Confers Protection Against Hypertension in Dahl Salt-Sensitive Rats. PLANT FOODS FOR HUMAN NUTRITION (DORDRECHT, NETHERLANDS) 2022; 77:373-382. [PMID: 35705768 DOI: 10.1007/s11130-022-00971-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 05/09/2022] [Indexed: 06/15/2023]
Abstract
Hypertension is one of the main factors of cardiovascular disease worldwide and is strongly related to the overall mortality. High salt intake is a major risk factors for hypertension. Identifying functional foods that can help prevent mechanistic abnormalities mediating salt-induced hypertension is an issue of considerable nutraceutical and scientific interest. Dietary Momordica charantia may be an alternative approach to avoid salt-induced hypertension. Dahl salt-sensitive (DSS) rats were used to determine whether Momordica charantia water extracts (ME) exerts anti-hypertensive effects in the present study. ME gavage could significantly prevented the increase of blood pressure, blood urea nitrogen, creatinine, and urine protein-to-creatinine ratio of DSS rats. Metabolomics analysis indicated that high-salt diet induced abnormal amino acid metabolism was related to nitric oxide (NO) deficiency, but ME gavage could upregulate the activities of nitric oxide synthase, aspartate aminotransferase, argininosuccinate lyase, argininosuccinate synthase and restore endogenous synthesis of arginine and NO. Meanwhile, renal function was improved after ME gavage. Citrulline, as one of the important component in ME, could attenuate salt-induced hypertension by increasing endogenous synthesis of arginine and NO. Antioxidants in ME, such as phenolic compound, may avoid high-salt induced oxidative stress in DSS rats, which may be another mechanism by which ME prevented blood pressure increase. Thus, the present study indicated that feeding Momordica charantia could avoid high-salt-induced hypertension in DSS rats.
Collapse
Affiliation(s)
- Li Zeng
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China
| | - Meng Chen
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China
| | - Hussain Ahmad
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China
| | - Xuewei Zheng
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China
| | - Yanan Ouyang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China
| | - Pengfei Yang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China
| | - Zhe Yang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China
| | - Di Gao
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China
| | - Zhongmin Tian
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China.
| |
Collapse
|
56
|
Yu Z, Zhou M, Liu J, Zhao W. Underlying antihypertensive mechanism of egg white-derived peptide QIGLF using renal metabolomics analysis. Food Res Int 2022; 157:111457. [PMID: 35761693 DOI: 10.1016/j.foodres.2022.111457] [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: 03/17/2022] [Revised: 05/26/2022] [Accepted: 06/01/2022] [Indexed: 11/24/2022]
Abstract
The kidney is an important target organ in the treatment of hypertension, but the effect of peptide QIGLF with antihypertensive activity on kidneys remains unknown. In the work, we aimed to further understand the hypotensive effects of QIGLF in spontaneously hypertensive rats (SHRs) using widely targeted metabolomics technology to investigate the kidney metabolic profiling variations. After four weeks of oral administration, the results showed different renal metabolomics profiles between QIGLF and model groups. Besides, a total of 10 potential biomarkers were identified, that is, 3-hydroxybutanoate, 20-hydroxyeicosatetraenoic acid, 19(S)-hydroxyeicosatetraenoic acid, 15-oxoETE, L-ornithine, malonate, uridine, uridine 5'-monophosphate, argininosuccinic acid, and N-carbamoyl-L-aspartate. These metabolites might exhibit antihypertensive activity of QIGLF by regulating synthesis and degradation of ketone bodies, arachidonic acid metabolism, pyrimidine metabolism, and arginine biosynthesis. These findings suggest that QIGLF might alleviate hypertension by inhibiting renal inflammation, promoting natriuresis, and regulating renal nitric oxide production.
Collapse
Affiliation(s)
- Zhipeng Yu
- College of Food Science and Engineering, Bohai University, Jinzhou 121013, PR China; School of Food Science and Engineering, Hainan University, Haikou 570228, PR China
| | - Mingjie Zhou
- College of Food Science and Engineering, Bohai University, Jinzhou 121013, PR China
| | - Jingbo Liu
- Lab of Nutrition and Functional Food, Jilin University, Changchun 130062, PR China
| | - Wenzhu Zhao
- School of Food Science and Engineering, Hainan University, Haikou 570228, PR China.
| |
Collapse
|
57
|
Gao Z, Chen X. Fatty Acid β-Oxidation in Kidney Diseases: Perspectives on Pathophysiological Mechanisms and Therapeutic Opportunities. Front Pharmacol 2022; 13:805281. [PMID: 35517820 PMCID: PMC9065343 DOI: 10.3389/fphar.2022.805281] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Accepted: 03/31/2022] [Indexed: 11/13/2022] Open
Abstract
The kidney is a highly metabolic organ and requires a large amount of ATP to maintain its filtration-reabsorption function, and mitochondrial fatty acid β-oxidation serves as the main source of energy to meet its functional needs. Reduced and inefficient fatty acid β-oxidation is thought to be a major mechanism contributing to kidney diseases, including acute kidney injury, chronic kidney disease and diabetic nephropathy. PPARα, AMPK, sirtuins, HIF-1, and TGF-β/SMAD3 activation have all been shown to play key roles in the regulation of fatty acid β-oxidation in kidney diseases, and restoration of fatty acid β-oxidation by modulation of these molecules can ameliorate the development of such diseases. Here, we disentangle the lipid metabolism regulation properties and potential mechanisms of mesenchymal stem cells and their extracellular vesicles, and emphasize the role of mesenchymal stem cells on lipid metabolism. This review aims to highlight the important role of fatty acid β-oxidation in the progression of kidney diseases, and to explore the fatty acid β-oxidation effects and therapeutic potential of mesenchymal stem cells for kidney diseases.
Collapse
Affiliation(s)
- Zhumei Gao
- Department of Nephrology, The Second Hospital of Jilin University, Jilin, China
| | - Xiangmei Chen
- Department of Nephrology, The Second Hospital of Jilin University, Jilin, China.,Department of Nephrology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
58
|
Zeng L, Liu Z, Zhou L, Chen M, Zheng X, Yang P, Zhao X, Tian Z. Effects of almonds on ameliorating salt-induced hypertension in dahl salt-sensitive rats. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2022; 102:2710-2722. [PMID: 34708426 DOI: 10.1002/jsfa.11611] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 10/23/2021] [Accepted: 10/28/2021] [Indexed: 06/13/2023]
Abstract
BACKGROUND Excessive dietary salt intake is related to an increased risk of hypertension. Dietary functional foods probably could help to improve salt-induced hypertension. In this study, Dahl salt-sensitive (DSS) rats were used to investigate their metabolic differences from those of salt-resistant SS.13BN rats and determine whether dietary protein-rich almonds could ameliorate salt-induced elevation of blood pressure in DSS rats. RESULTS After high-salt intake, the systolic blood pressure and mean arterial pressure of the DSS rats increased dramatically. Metabolomics analysis indicated abnormal amino acid metabolism in their kidneys. Their renal nitric oxide (NO) content and nitric oxide synthase activity decreased significantly after high-salt diet. Oxidative stress also occurred in DSS rats. After the DSS rats received almond supplementation, the levels of various amino acids in their kidney increased, and renal arginine and NO contents were upregulated. Their renal hydrogen peroxide and malonaldehyde levels decreased, whereas renal catalase, superoxide dismutase and glutathione peroxidase activities and glutathione levels increased. CONCLUSION The renal abnormal amino acid metabolism of DSS rats contributed to the impaired NO production in response to high-salt intake. Together with salt-induced oxidative stress, high-salt diet intake ultimately led to an increase in the blood pressure of DSS rats. Protein-rich almond supplementation might prevent the development of salt-induced hypertension by restoring arginine and NO regeneration and alleviating salt-induced oxidative stress. © 2021 Society of Chemical Industry.
Collapse
Affiliation(s)
- Li Zeng
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Zerong Liu
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Luxin Zhou
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Meng Chen
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Xuewei Zheng
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Pengfei Yang
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Xinrui Zhao
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Zhongmin Tian
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
59
|
Tang C, Jiang H, Zhao B, Lin Y, Lin S, Chen T, Su Y, Zhang Y, Zhou L, Li L, Lin J, Lu Z, Wang Y, Zhao Z, Liu L, Wang Y, Zhang J, Shen H. The association between bilirubin and hypertension among a Chinese ageing cohort: a prospective follow-up study. J Transl Med 2022; 20:108. [PMID: 35246141 PMCID: PMC8895624 DOI: 10.1186/s12967-022-03309-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 02/14/2022] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Hypertension is highly prevalent and associated with the elevated risks of cardiovascular diseases, dementia, and physical disabilities among adults. Although the correlation between bilirubin and hypertension has been reported, the observation in quinquagenarian population is scarce. We aimed to examine bilirubin-hypertension association in Guankou Ageing Cohort Study. METHODS Participants ≥ 55 years were recruited and their questionnaires and physical examination data were collected. Kaplan-Meier survival analysis and Cox proportional hazards regression were implemented to assess the hypertension risk. The non-liner dose-response relationships of bilirubin-hypertension were determined by restricted cubic spline (RCS) models. Receiver operating characteristic (ROC) curves and multiple factors analysis (MFA) were performed to evaluate the predictive abilities. RESULTS 1881 eligible participants (male 43.75%, female 56.25%) with the median age of 61.00 (59.00-66.00) were included. The hazard ratio (HR, 95% CI) of serum total bilirubin (STB) and unconjugated bilirubin (UCB) were 1.03 (1.01-1.05) and 1.05 (1.03-1.07), while conjugated bilirubin (CB) showed a weak protective effect with the HR of 0.96 (0.92-0.99), and the associations remained significant in all models. RCS analyses further indicated the similar bidirectional effects of STB and UCB with the cut-off of 12.17 μmol/L and 8.59 μmol/L, while CB exhibited inverse bidirectional dose-response relationship with a cut-off of 3.47 μmol/L. ROC curves and MFA showed baseline STB combined with age, BMI, and waist circumference could well discriminate the low and high of hypertension risk. CONCLUSIONS Our findings suggested the higher levels of total and unconjugated bilirubin were hazardous factors of hypertension, while an inverse effect presented when more bilirubin was conjugated.
Collapse
Affiliation(s)
- Chen Tang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnotics, School of Public Health, Xiamen University, No. 4221-117 South Xiang'an Road, Xiang'an District, Xiamen, 361102, Fujian, People's Republic of China
| | - Hanxiang Jiang
- Xiamen Guankou Hospital, No. 270 Jingshan Road, Jimei District, Xiamen, 361023, Fujian, People's Republic of China
| | - Bin Zhao
- Clinical Medical Laboratory, Xiang'an Hospital of Xiamen University, No. 2000, East Xiang'an Road, Xiamen, 361102, Fujian, People's Republic of China
| | - Yi Lin
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnotics, School of Public Health, Xiamen University, No. 4221-117 South Xiang'an Road, Xiang'an District, Xiamen, 361102, Fujian, People's Republic of China
| | - Shengnan Lin
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnotics, School of Public Health, Xiamen University, No. 4221-117 South Xiang'an Road, Xiang'an District, Xiamen, 361102, Fujian, People's Republic of China
| | - Tianmu Chen
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnotics, School of Public Health, Xiamen University, No. 4221-117 South Xiang'an Road, Xiang'an District, Xiamen, 361102, Fujian, People's Republic of China
| | - Yanhua Su
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnotics, School of Public Health, Xiamen University, No. 4221-117 South Xiang'an Road, Xiang'an District, Xiamen, 361102, Fujian, People's Republic of China
| | - Yiqin Zhang
- Department of Nephrology, The Second Affiliated Hospital of Xiamen Medical College, No. 566 Shengguang Road, Jimei District, Xiamen, 361021, Fujian, People's Republic of China
| | - Lina Zhou
- Department of Nephrology, The Second Affiliated Hospital of Xiamen Medical College, No. 566 Shengguang Road, Jimei District, Xiamen, 361021, Fujian, People's Republic of China
| | - Lianmeng Li
- Xiamen Guankou Hospital, No. 270 Jingshan Road, Jimei District, Xiamen, 361023, Fujian, People's Republic of China
| | - Jincheng Lin
- Xiamen Guankou Hospital, No. 270 Jingshan Road, Jimei District, Xiamen, 361023, Fujian, People's Republic of China
| | - Zhonghua Lu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnotics, School of Public Health, Xiamen University, No. 4221-117 South Xiang'an Road, Xiang'an District, Xiamen, 361102, Fujian, People's Republic of China
| | - Yao Wang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnotics, School of Public Health, Xiamen University, No. 4221-117 South Xiang'an Road, Xiang'an District, Xiamen, 361102, Fujian, People's Republic of China
| | - Zeyu Zhao
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnotics, School of Public Health, Xiamen University, No. 4221-117 South Xiang'an Road, Xiang'an District, Xiamen, 361102, Fujian, People's Republic of China
| | - Longjian Liu
- Department of Epidemiology and Biostatistics, Dornsife School of Public Health, Drexel University, 3141 Chestnut Street, Philadelphia, PA, USA
| | - Yuxin Wang
- Department of Nephrology, The Second Affiliated Hospital of Xiamen Medical College, No. 566 Shengguang Road, Jimei District, Xiamen, 361021, Fujian, People's Republic of China.
| | - Jie Zhang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnotics, School of Public Health, Xiamen University, No. 4221-117 South Xiang'an Road, Xiang'an District, Xiamen, 361102, Fujian, People's Republic of China.
| | - Heqing Shen
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnotics, School of Public Health, Xiamen University, No. 4221-117 South Xiang'an Road, Xiang'an District, Xiamen, 361102, Fujian, People's Republic of China.
| |
Collapse
|
60
|
Correia MJ, Pimpão AB, Fernandes DGF, Morello J, Sequeira CO, Calado J, Antunes AMM, Almeida MS, Branco P, Monteiro EC, Vicente JB, Serpa J, Pereira SA. Cysteine as a Multifaceted Player in Kidney, the Cysteine-Related Thiolome and Its Implications for Precision Medicine. Molecules 2022; 27:1416. [PMID: 35209204 PMCID: PMC8874463 DOI: 10.3390/molecules27041416] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 02/15/2022] [Accepted: 02/17/2022] [Indexed: 11/16/2022] Open
Abstract
In this review encouraged by original data, we first provided in vivo evidence that the kidney, comparative to the liver or brain, is an organ particularly rich in cysteine. In the kidney, the total availability of cysteine was higher in cortex tissue than in the medulla and distributed in free reduced, free oxidized and protein-bound fractions (in descending order). Next, we provided a comprehensive integrated review on the evidence that supports the reliance on cysteine of the kidney beyond cysteine antioxidant properties, highlighting the relevance of cysteine and its renal metabolism in the control of cysteine excess in the body as a pivotal source of metabolites to kidney biomass and bioenergetics and a promoter of adaptive responses to stressors. This view might translate into novel perspectives on the mechanisms of kidney function and blood pressure regulation and on clinical implications of the cysteine-related thiolome as a tool in precision medicine.
Collapse
Affiliation(s)
- Maria João Correia
- CEDOC, NOVA Medical School, Universidade Nova de Lisboa, 1169-056 Lisboa, Portugal; (M.J.C.); (A.B.P.); (J.M.); (C.O.S.); (M.S.A.); (P.B.); (E.C.M.); (J.S.)
| | - António B. Pimpão
- CEDOC, NOVA Medical School, Universidade Nova de Lisboa, 1169-056 Lisboa, Portugal; (M.J.C.); (A.B.P.); (J.M.); (C.O.S.); (M.S.A.); (P.B.); (E.C.M.); (J.S.)
| | - Dalila G. F. Fernandes
- Instituto de Tecnologia Química e Biológica António Xavier (ITQB NOVA), 2780-157 Oeiras, Portugal; (D.G.F.F.); (J.B.V.)
| | - Judit Morello
- CEDOC, NOVA Medical School, Universidade Nova de Lisboa, 1169-056 Lisboa, Portugal; (M.J.C.); (A.B.P.); (J.M.); (C.O.S.); (M.S.A.); (P.B.); (E.C.M.); (J.S.)
| | - Catarina O. Sequeira
- CEDOC, NOVA Medical School, Universidade Nova de Lisboa, 1169-056 Lisboa, Portugal; (M.J.C.); (A.B.P.); (J.M.); (C.O.S.); (M.S.A.); (P.B.); (E.C.M.); (J.S.)
| | - Joaquim Calado
- Centre for Toxicogenomics and Human Health (ToxOmics), Genetics, Oncology and Human Toxicology, Nova Medical School/Faculdade de Ciências Médicas, Universidade Nova de Lisboa, 1169-056 Lisboa, Portugal;
- Nephrology Department, Centro Hospitalar Universitário de Lisboa Central, 1069-166 Lisboa, Portugal
| | - Alexandra M. M. Antunes
- Centro de Química Estrutural, Institute of Molecular Sciences, Instituto Superior Técnico, 1049-001 Lisboa, Portugal;
| | - Manuel S. Almeida
- CEDOC, NOVA Medical School, Universidade Nova de Lisboa, 1169-056 Lisboa, Portugal; (M.J.C.); (A.B.P.); (J.M.); (C.O.S.); (M.S.A.); (P.B.); (E.C.M.); (J.S.)
- Hospital de Santa Cruz, Centro Hospitalar de Lisboa Ocidental, 2790-134 Carnaxide, Portugal
| | - Patrícia Branco
- CEDOC, NOVA Medical School, Universidade Nova de Lisboa, 1169-056 Lisboa, Portugal; (M.J.C.); (A.B.P.); (J.M.); (C.O.S.); (M.S.A.); (P.B.); (E.C.M.); (J.S.)
- Hospital de Santa Cruz, Centro Hospitalar de Lisboa Ocidental, 2790-134 Carnaxide, Portugal
| | - Emília C. Monteiro
- CEDOC, NOVA Medical School, Universidade Nova de Lisboa, 1169-056 Lisboa, Portugal; (M.J.C.); (A.B.P.); (J.M.); (C.O.S.); (M.S.A.); (P.B.); (E.C.M.); (J.S.)
| | - João B. Vicente
- Instituto de Tecnologia Química e Biológica António Xavier (ITQB NOVA), 2780-157 Oeiras, Portugal; (D.G.F.F.); (J.B.V.)
| | - Jacinta Serpa
- CEDOC, NOVA Medical School, Universidade Nova de Lisboa, 1169-056 Lisboa, Portugal; (M.J.C.); (A.B.P.); (J.M.); (C.O.S.); (M.S.A.); (P.B.); (E.C.M.); (J.S.)
- Instituto Português de Oncologia de Lisboa Francisco Gentil (IPOLFG), 1099-023 Lisboa, Portugal
| | - Sofia A. Pereira
- CEDOC, NOVA Medical School, Universidade Nova de Lisboa, 1169-056 Lisboa, Portugal; (M.J.C.); (A.B.P.); (J.M.); (C.O.S.); (M.S.A.); (P.B.); (E.C.M.); (J.S.)
| |
Collapse
|
61
|
Jung J, Park WY, Kim YJ, Kim M, Choe M, Jin K, Seo JH, Ha E. 3-Hydroxybutyrate Ameliorates the Progression of Diabetic Nephropathy. Antioxidants (Basel) 2022; 11:antiox11020381. [PMID: 35204263 PMCID: PMC8868458 DOI: 10.3390/antiox11020381] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 02/07/2022] [Accepted: 02/10/2022] [Indexed: 02/04/2023] Open
Abstract
Studies report beneficial effects of 3-hydroxybutyrate (3-OHB) on the treatment of type 2 diabetes and obesity, but the effects of 3-OHB on diabetic nephropathy have not been elucidated. This study was designed to investigate the efficacy and mechanism of 3-OHB against progression of diabetic nephropathy (DN). Mice (db/db) were fed normal chow, high-fat, or ketogenic diets (KD) containing precursors of 3-OHB. Hyperglycemia was determined based on random glucose level (≥250 mg/dL). Fasting blood glucose and body weights were measured once a week. Twenty four-hour urine albumin to creatinine ratio was determined 5 weeks after the differential diet. Energy expenditure was measured 9 weeks after the differential diet. Body weights were significantly lower in the KD group than those in other groups, but no significant differences in fasting blood glucose levels among three groups were observed. Urine albumin to creatinine ratio and serum blood urea nitrogen (BUN) to creatinine ratio in the KD group were significantly lower than in other groups. Histologic and quantitative analysis of mesangial area suggested that KD delayed the progression of DN phenotype in db/db mice. Metabolic cage analysis also revealed that KD increased energy expenditure in db/db mice. In vitro studies with proximal tubular cells revealed that 3-OHB stimulated autophagic flux. 3-OHB increased LC3 I to LC3 II ratio, phosphorylation of AMPK, beclin, p62 degradation, and NRF2 expression. Moreover, we found that 3-OHB attenuated high glucose-induced reactive oxygen species (ROS) levels in proximal tubular cells. In vivo study also confirmed increased LC3 and decreased ROS levels in the kidney of KD mice. In summary, this study shows in both in vivo and in vitro models that 3-OHB delays the progression of DN by augmenting autophagy and inhibiting oxidative stress.
Collapse
Affiliation(s)
- Jeeyoun Jung
- Clinical Medicine Division, Korea Institute of Oriental Medicine, Daejeon 34054, Korea;
| | - Woo Yeong Park
- Department of Internal Medicine, Keimyung University School of Medicine, Daegu 42601, Korea;
| | - Yun Jin Kim
- Department of Biochemistry, Keimyung University School of Medicine, Daegu 42601, Korea; (Y.J.K.); (M.K.)
| | - Mikyung Kim
- Department of Biochemistry, Keimyung University School of Medicine, Daegu 42601, Korea; (Y.J.K.); (M.K.)
| | - Misun Choe
- Department of Pathology, Keimyung University School of Medicine, Daegu 42601, Korea;
| | - Kyubok Jin
- Department of Internal Medicine, Keimyung University School of Medicine, Daegu 42601, Korea;
- Correspondence: (K.J.); (J.H.S.); (E.H.)
| | - Ji Hae Seo
- Department of Biochemistry, Keimyung University School of Medicine, Daegu 42601, Korea; (Y.J.K.); (M.K.)
- Correspondence: (K.J.); (J.H.S.); (E.H.)
| | - Eunyoung Ha
- Department of Biochemistry, Keimyung University School of Medicine, Daegu 42601, Korea; (Y.J.K.); (M.K.)
- Correspondence: (K.J.); (J.H.S.); (E.H.)
| |
Collapse
|
62
|
Tomar N, Zhang X, Kandel SM, Sadri S, Yang C, Liang M, Audi SH, Cowley AW, Dash RK. Substrate-dependent differential regulation of mitochondrial bioenergetics in the heart and kidney cortex and outer medulla. BIOCHIMICA ET BIOPHYSICA ACTA. BIOENERGETICS 2022; 1863:148518. [PMID: 34864090 PMCID: PMC8957717 DOI: 10.1016/j.bbabio.2021.148518] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 10/29/2021] [Accepted: 11/20/2021] [Indexed: 05/05/2023]
Abstract
The kinetics and efficiency of mitochondrial oxidative phosphorylation (OxPhos) can depend on the choice of respiratory substrates. Furthermore, potential differences in this substrate dependency among different tissues are not well-understood. Here, we determined the effects of different substrates on the kinetics and efficiency of OxPhos in isolated mitochondria from the heart and kidney cortex and outer medulla (OM) of Sprague-Dawley rats. The substrates were pyruvate+malate, glutamate+malate, palmitoyl-carnitine+malate, alpha-ketoglutarate+malate, and succinate±rotenone at saturating concentrations. The kinetics of OxPhos were interrogated by measuring mitochondrial bioenergetics under different ADP perturbations. Results show that the kinetics and efficiency of OxPhos are highly dependent on the substrates used, and this dependency is distinctly different between heart and kidney. Heart mitochondria showed higher respiratory rates and OxPhos efficiencies for all substrates in comparison to kidney mitochondria. Cortex mitochondria respiratory rates were higher than OM mitochondria, but OM mitochondria OxPhos efficiencies were higher than cortex mitochondria. State 3 respiration was low in heart mitochondria with succinate but increased significantly in the presence of rotenone, unlike kidney mitochondria. Similar differences were observed in mitochondrial membrane potential. Differences in H2O2 emission in the presence of succinate±rotenone were observed in heart mitochondria and to a lesser extent in OM mitochondria, but not in cortex mitochondria. Bioenergetics and H2O2 emission data with succinate±rotenone indicate that oxaloacetate accumulation and reverse electron transfer may play a more prominent regulatory role in heart mitochondria than kidney mitochondria. These studies provide novel quantitative data demonstrating that the choice of respiratory substrates affects mitochondrial responses in a tissue-specific manner.
Collapse
Affiliation(s)
- Namrata Tomar
- Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee WI-53226, United States of America
| | - Xiao Zhang
- Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee WI-53226, United States of America
| | - Sunil M Kandel
- Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee WI-53226, United States of America
| | - Shima Sadri
- Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee WI-53226, United States of America
| | - Chun Yang
- Department of Physiology, Medical College of Wisconsin, Milwaukee WI-53226, United States of America
| | - Mingyu Liang
- Department of Physiology, Medical College of Wisconsin, Milwaukee WI-53226, United States of America; Center of Systems Molecular Medicine, Medical College of Wisconsin, Milwaukee WI-53226, United States of America
| | - Said H Audi
- Department of Biomedical Engineering, Marquette University, Milwaukee WI-53223, United States of America
| | - Allen W Cowley
- Department of Physiology, Medical College of Wisconsin, Milwaukee WI-53226, United States of America; Center of Systems Molecular Medicine, Medical College of Wisconsin, Milwaukee WI-53226, United States of America.
| | - Ranjan K Dash
- Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee WI-53226, United States of America; Department of Physiology, Medical College of Wisconsin, Milwaukee WI-53226, United States of America; Center of Systems Molecular Medicine, Medical College of Wisconsin, Milwaukee WI-53226, United States of America.
| |
Collapse
|
63
|
Liu L, Bai F, Song H, Xiao R, Wang Y, Yang H, Ren X, Li S, Gao L, Ma C, Yang X, Liang X. Upregulation of TIPE1 in tubular epithelial cell aggravates diabetic nephropathy by disrupting PHB2 mediated mitophagy. Redox Biol 2022; 50:102260. [PMID: 35152003 PMCID: PMC8844679 DOI: 10.1016/j.redox.2022.102260] [Citation(s) in RCA: 52] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 02/01/2022] [Accepted: 02/04/2022] [Indexed: 12/24/2022] Open
Abstract
Renal tubular epithelial cells (RTECs) are one of the most mitochondria-rich cell types, and are thus vulnerable to mitochondrial dysregulation, which is defined as a pivotal event in tubular damage in diabetic nephropathy (DN). However, the underlying mechanisms remain largely unknown. Here, we investigated the role and mechanisms of tumor necrosis factor alpha-induced protein 8-like 1 (TNFAIP8L1/TIPE1) in high glucose (HG)-induced mitochondrial dysfunction in RTECs and DN progression. TIPE1 expression was predominantly upregulated in RTECs in patients with DN and mice with streptozotocin (STZ)-induced DN. Conditional knockout of Tipe1 in RTECs significantly decreased the urine protein creatinine ratio, renal tubular damage, epithelial-mesenchymal transition, and interstitial fibrosis in STZ-induced mice. RNA sequencing revealed that citrate cycle-related genes were positively enriched in the renal tissues of RTEC-specific Tipe1 knockout mice. Tipe1 deficiency upregulated ATP levels, mitochondrial membrane potential, and respiration rate, but downregulated mitochondrial ROS levels in RTECs. Furthermore, Tipe1 ablation led to enhanced mitophagy in RTECs, indicative of increased LC3II, PINK1, and Parkin expression, but decreased p62 expression in mitochondria. Mechanistically, mass spectrometry screening and co-immunoprecipitation assays revealed the interaction of TIPE1 with prohibitin 2 (PHB2), a crucial mitophagy receptor. Intriguingly, TIPE1 promoted the ubiquitination and proteasomal degradation of PHB2. Subsequently, PHB2 knockdown almost abrogated the improvement of Tipe1 loss on HG-induced tubular cell mitophagy and damage. Thus, TIPE1 disrupts mitochondrial homeostasis in RTECs and promotes tubular damage by destabilizing PHB2 under HG conditions. Hence, TIPE1 may act as a potential therapeutic target to prevent DN progression. TIPE1 is highly expressed in RTECs of DN. TIPE1 aggravates diabetic renal injury and fibrosis. TIPE1 disrupts the mitochondria homeostasis of RTECs by inhibiting mitophagy. TIPE1 interacts with and promotes the proteasomal degradation of PHB2.
Collapse
Affiliation(s)
- Lei Liu
- Key Laboratory for Experimental Teratology of Ministry of Education and Department of Immunology, School of Basic Medical Sciences, Cheeloo Medical College, Shandong University, Jinan, Shandong, China; Department of Nephropathy, Qilu Hospital of Shandong University, Cheeloo Medical College, Shandong University, Jinan, Shandong, Jinan, Shandong, PR China
| | - Fang Bai
- Department of Nephropathy, Qilu Hospital of Shandong University, Cheeloo Medical College, Shandong University, Jinan, Shandong, Jinan, Shandong, PR China
| | - Hui Song
- Key Laboratory for Experimental Teratology of Ministry of Education and Department of Immunology, School of Basic Medical Sciences, Cheeloo Medical College, Shandong University, Jinan, Shandong, China
| | - Rong Xiao
- Key Laboratory for Experimental Teratology of Ministry of Education and Department of Immunology, School of Basic Medical Sciences, Cheeloo Medical College, Shandong University, Jinan, Shandong, China
| | - Yuzhen Wang
- Key Laboratory for Experimental Teratology of Ministry of Education and Department of Immunology, School of Basic Medical Sciences, Cheeloo Medical College, Shandong University, Jinan, Shandong, China
| | - Huimin Yang
- Department of Nephropathy, Qilu Hospital of Shandong University, Cheeloo Medical College, Shandong University, Jinan, Shandong, Jinan, Shandong, PR China
| | - Xiaolei Ren
- Key Laboratory for Experimental Teratology of Ministry of Education and Department of Immunology, School of Basic Medical Sciences, Cheeloo Medical College, Shandong University, Jinan, Shandong, China
| | - Shuangjie Li
- Key Laboratory for Experimental Teratology of Ministry of Education and Department of Immunology, School of Basic Medical Sciences, Cheeloo Medical College, Shandong University, Jinan, Shandong, China
| | - Lifen Gao
- Key Laboratory for Experimental Teratology of Ministry of Education and Department of Immunology, School of Basic Medical Sciences, Cheeloo Medical College, Shandong University, Jinan, Shandong, China; Shandong Collaborative Innovation Center of Technology and Equipment for Biological Diagnosis and Therapy, Jinan, Shandong, China
| | - Chunhong Ma
- Key Laboratory for Experimental Teratology of Ministry of Education and Department of Immunology, School of Basic Medical Sciences, Cheeloo Medical College, Shandong University, Jinan, Shandong, China; Shandong Collaborative Innovation Center of Technology and Equipment for Biological Diagnosis and Therapy, Jinan, Shandong, China
| | - Xiangdong Yang
- Department of Nephropathy, Qilu Hospital of Shandong University, Cheeloo Medical College, Shandong University, Jinan, Shandong, Jinan, Shandong, PR China
| | - Xiaohong Liang
- Key Laboratory for Experimental Teratology of Ministry of Education and Department of Immunology, School of Basic Medical Sciences, Cheeloo Medical College, Shandong University, Jinan, Shandong, China; Shandong Collaborative Innovation Center of Technology and Equipment for Biological Diagnosis and Therapy, Jinan, Shandong, China.
| |
Collapse
|
64
|
Yang P, Zhou L, Chen M, Zeng L, Ouyang Y, Zheng X, Chen X, Yang Z, Tian Z. Supplementation of amino acids and organic acids prevents the increase in blood pressure induced by high salt in Dahl salt-sensitive rats. Food Funct 2022; 13:891-903. [PMID: 34994761 DOI: 10.1039/d1fo03577k] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
A high-salt (HS) diet leads to metabolic disorders in Dahl salt-sensitive (SS) rats, and promotes the development of hypertension. According to the changes in the metabolites of SS rats, a set of combined dietary supplements containing amino acids and organic acids (AO) were designed. The purpose of the present study was to evaluate the effect of AO supplementation on the blood pressure of SS rats after the HS diet and clarify the mechanism of AO by metabolomics and biochemical analyses. The results showed that AO supplementation avoided the elevation of blood pressure induced by the HS diet in SS rats, increased the renal antioxidant enzyme activities (catalase, superoxide dismutase, glutathione reductase, and glutathione S-transferase), reduced the H2O2 and MDA levels, and restored the normal antioxidant status of the serum and kidneys. AO also reversed the decrease in the nitric oxide (NO) levels and NO synthase activity induced by the HS feed, which involved the L-arginine/NO pathway. Metabolomics analysis showed that AO administration increased the levels of amino acids such as cysteine, glycine, hypotaurine, and lysine in the renal medulla and the levels of leucine, isoleucine, and serine in the renal cortex. Of note, lysine, hypotaurine and glycine had higher metabolic centrality in the metabolic correlation network of the renal medulla after AO administration. In conclusion, AO intervention could prevent HS diet-induced hypertension in SS rats by restoring the metabolic homeostasis of the kidneys. Hence, AO has the potential to become a functional food additive to improve salt-sensitive hypertension.
Collapse
Affiliation(s)
- Pengfei Yang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China.
| | - Luxin Zhou
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China.
| | - Meng Chen
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China.
| | - Li Zeng
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China.
| | - Yanan Ouyang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China.
| | - Xuewei Zheng
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China.
| | - Xiangbo Chen
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China.
| | - Zhe Yang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China.
| | - Zhongmin Tian
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China.
| |
Collapse
|
65
|
Chen HQ, Gong JY, Xing K, Liu MZ, Ren H, Luo JQ. Pharmacomicrobiomics: Exploiting the Drug-Microbiota Interactions in Antihypertensive Treatment. Front Med (Lausanne) 2022; 8:742394. [PMID: 35127738 PMCID: PMC8808336 DOI: 10.3389/fmed.2021.742394] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 12/29/2021] [Indexed: 12/15/2022] Open
Abstract
Hypertension is a leading risk factor for cardiovascular diseases and can reduce life expectancy. Owing to the widespread use of antihypertensive drugs, patients with hypertension have improved blood pressure control over the past few decades. However, for a considerable part of the population, these drugs still cannot significantly improve their symptoms. In order to explore the reasons behind, pharmacomicrobiomics provide unique insights into the drug treatment of hypertension by investigating the effect of bidirectional interaction between gut microbiota and antihypertensive drugs. This review discusses the relationship between antihypertensive drugs and the gut microbiome, including changes in drug pharmacokinetics and gut microbiota composition. In addition, we highlight how our current knowledge of antihypertensive drug-microbiota interactions to develop gut microbiota-based personalized ways for disease management, including antihypertensive response biomarker, microbial-targeted therapies, probiotics therapy. Ultimately, a better understanding of the impact of pharmacomicrobiomics in the treatment of hypertension will provide important information for guiding rational clinical use and individualized use.
Collapse
Affiliation(s)
- Hui-Qing Chen
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Jin-Yu Gong
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Kai Xing
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Mou-Ze Liu
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Institute of Clinical Pharmacy, Central South University, Changsha, China
| | - Huan Ren
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China
| | - Jian-Quan Luo
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Institute of Clinical Pharmacy, Central South University, Changsha, China
- *Correspondence: Jian-Quan Luo
| |
Collapse
|
66
|
Zhang X, Tomar N, Kandel SM, Audi SH, Cowley AW, Dash RK. Substrate- and Calcium-Dependent Differential Regulation of Mitochondrial Oxidative Phosphorylation and Energy Production in the Heart and Kidney. Cells 2021; 11:131. [PMID: 35011693 PMCID: PMC8750792 DOI: 10.3390/cells11010131] [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] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 12/26/2021] [Accepted: 12/28/2021] [Indexed: 11/16/2022] Open
Abstract
Mitochondrial dehydrogenases are differentially stimulated by Ca2+. Ca2+ has also diverse regulatory effects on mitochondrial transporters and other enzymes. However, the consequences of these regulatory effects on mitochondrial oxidative phosphorylation (OxPhos) and ATP production, and the dependencies of these consequences on respiratory substrates, have not been investigated between the kidney and heart despite the fact that kidney energy requirements are second only to those of the heart. Our objective was, therefore, to elucidate these relationships in isolated mitochondria from the kidney outer medulla (OM) and heart. ADP-induced mitochondrial respiration was measured at different CaCl2 concentrations in the presence of various respiratory substrates, including pyruvate + malate (PM), glutamate + malate (GM), alpha-ketoglutarate + malate (AM), palmitoyl-carnitine + malate (PCM), and succinate + rotenone (SUC + ROT). The results showed that, in both heart and OM mitochondria, and for most complex I substrates, Ca2+ effects are biphasic: small increases in Ca2+ concentration stimulated, while large increases inhibited mitochondrial respiration. Furthermore, significant differences in substrate- and Ca2+-dependent O2 utilization towards ATP production between heart and OM mitochondria were observed. With PM and PCM substrates, Ca2+ showed more prominent stimulatory effects in OM than in heart mitochondria, while with GM and AM substrates, Ca2+ had similar biphasic regulatory effects in both OM and heart mitochondria. In contrast, with complex II substrate SUC + ROT, only inhibitory effects on mitochondrial respiration was observed in both the heart and the OM. We conclude that the regulatory effects of Ca2+ on mitochondrial OxPhos and ATP synthesis are biphasic, substrate-dependent, and tissue-specific.
Collapse
Affiliation(s)
- Xiao Zhang
- Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee, WI 53226, USA; (X.Z.); (N.T.); (S.M.K.)
| | - Namrata Tomar
- Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee, WI 53226, USA; (X.Z.); (N.T.); (S.M.K.)
| | - Sunil M. Kandel
- Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee, WI 53226, USA; (X.Z.); (N.T.); (S.M.K.)
| | - Said H. Audi
- Department of Biomedical Engineering, Marquette University, Milwaukee, WI 53223, USA;
| | - Allen W. Cowley
- Center of Systems Molecular Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Ranjan K. Dash
- Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee, WI 53226, USA; (X.Z.); (N.T.); (S.M.K.)
- Center of Systems Molecular Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| |
Collapse
|
67
|
Faivre A, Verissimo T, Auwerx H, Legouis D, de Seigneux S. Tubular Cell Glucose Metabolism Shift During Acute and Chronic Injuries. Front Med (Lausanne) 2021; 8:742072. [PMID: 34778303 PMCID: PMC8585753 DOI: 10.3389/fmed.2021.742072] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 10/11/2021] [Indexed: 12/28/2022] Open
Abstract
Acute and chronic kidney disease are responsible for large healthcare costs worldwide. During injury, kidney metabolism undergoes profound modifications in order to adapt to oxygen and nutrient shortage. Several studies highlighted recently the importance of these metabolic adaptations in acute as well as in chronic phases of renal disease, with a potential deleterious effect on fibrosis progression. Until recently, glucose metabolism in the kidney has been poorly studied, even though the kidney has the capacity to use and produce glucose, depending on the segment of the nephron. During physiology, renal proximal tubular cells use the beta-oxidation of fatty acid to generate large amounts of energy, and can also produce glucose through gluconeogenesis. In acute kidney injury, proximal tubular cells metabolism undergo a metabolic shift, shifting away from beta-oxidation of fatty acids and gluconeogenesis toward glycolysis. In chronic kidney disease, the loss of fatty acid oxidation is also well-described, and data about glucose metabolism are emerging. We here review the modifications of proximal tubular cells glucose metabolism during acute and chronic kidney disease and their potential consequences, as well as the potential therapeutic implications.
Collapse
Affiliation(s)
- Anna Faivre
- Laboratory of Nephrology, Geneva University Hospitals, Geneva, Switzerland.,Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland
| | - Thomas Verissimo
- Laboratory of Nephrology, Geneva University Hospitals, Geneva, Switzerland.,Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland
| | - Hannah Auwerx
- Laboratory of Nephrology, Geneva University Hospitals, Geneva, Switzerland.,Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland
| | - David Legouis
- Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland.,Intensive Care Unit, Department of Acute Medicine, Geneva University Hospitals, Geneva, Switzerland
| | - Sophie de Seigneux
- Laboratory of Nephrology, Geneva University Hospitals, Geneva, Switzerland.,Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland
| |
Collapse
|
68
|
Hwang S, Chung KW. Targeting fatty acid metabolism for fibrotic disorders. Arch Pharm Res 2021; 44:839-856. [PMID: 34664210 DOI: 10.1007/s12272-021-01352-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 10/11/2021] [Indexed: 02/06/2023]
Abstract
Fibrosis is defined by abnormal accumulation of extracellular matrix, which can affect virtually every organ system under diseased conditions. Fibrotic tissue remodeling often leads to organ dysfunction and is highly associated with increased morbidity and mortality. The disease burden caused by fibrosis is substantial, and the medical need for effective antifibrotic therapies is essential. Significant progress has been made in understanding the molecular mechanism and pathobiology of fibrosis, such as transforming growth factor-β (TGF-β)-mediated signaling pathways. However, owing to the complex and dynamic properties of fibrotic disorders, there are currently no therapeutic options that can prevent or reverse fibrosis. Recent studies have revealed that alterations in fatty acid metabolic processes are common mechanisms and core pathways that play a central role in different fibrotic disorders. Excessive lipid accumulation or defective fatty acid oxidation is associated with increased lipotoxicity, which directly contributes to the development of fibrosis. Genetic alterations or pharmacologic targeting of fatty acid metabolic processes have great potential for the inhibition of fibrosis development. Furthermore, mechanistic studies have revealed active interactions between altered metabolic processes and fibrosis development. Several well-known fibrotic factors change the lipid metabolic processes, while altered metabolic processes actively participate in fibrosis development. This review summarizes the recent evidence linking fatty acid metabolism and fibrosis, and provides new insights into the pathogenesis of fibrotic diseases for the development of drugs for fibrosis prevention and treatment.
Collapse
Affiliation(s)
- Seonghwan Hwang
- College of Pharmacy, Pusan National University, Busan, 46214, Republic of Korea
| | - Ki Wung Chung
- College of Pharmacy, Pusan National University, Busan, 46214, Republic of Korea.
| |
Collapse
|
69
|
Correia MJ, Pimpão AB, Lopes-Coelho F, Sequeira CO, Coelho NR, Gonçalves-Dias C, Barouki R, Coumoul X, Serpa J, Morello J, Monteiro EC, Pereira SA. Aryl Hydrocarbon Receptor and Cysteine Redox Dynamics Underlie (Mal)adaptive Mechanisms to Chronic Intermittent Hypoxia in Kidney Cortex. Antioxidants (Basel) 2021; 10:antiox10091484. [PMID: 34573115 PMCID: PMC8469308 DOI: 10.3390/antiox10091484] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 09/14/2021] [Accepted: 09/15/2021] [Indexed: 12/31/2022] Open
Abstract
We hypothesized that an interplay between aryl hydrocarbon receptor (AhR) and cysteine-related thiolome at the kidney cortex underlies the mechanisms of (mal)adaptation to chronic intermittent hypoxia (CIH), promoting arterial hypertension (HTN). Using a rat model of CIH-HTN, we investigated the impact of short-term (1 and 7 days), mid-term (14 and 21 days, pre-HTN), and long-term intermittent hypoxia (IH) (up to 60 days, established HTN) on CYP1A1 protein level (a sensitive hallmark of AhR activation) and cysteine-related thiol pools. We found that acute and chronic IH had opposite effects on CYP1A1 and the thiolome. While short-term IH decreased CYP1A1 and increased protein-S-thiolation, long-term IH increased CYP1A1 and free oxidized cysteine. In addition, an in vitro administration of cystine, but not cysteine, to human endothelial cells increased Cyp1a1 expression, supporting cystine as a putative AhR activator. This study supports CYP1A1 as a biomarker of obstructive sleep apnea (OSA) severity and oxidized pools of cysteine as risk indicator of OSA-HTN. This work contributes to a better understanding of the mechanisms underlying the phenotype of OSA-HTN, mimicked by this model, which is in line with precision medicine challenges in OSA.
Collapse
Affiliation(s)
- Maria João Correia
- CEDOC, NOVA Medical School, Universidade Nova de Lisboa, 1169-056 Lisboa, Portugal; (M.J.C.); (A.B.P.); (F.L.-C.); (C.O.S.); (N.R.C.); (C.G.-D.); (J.S.); (J.M.); (E.C.M.)
| | - António B. Pimpão
- CEDOC, NOVA Medical School, Universidade Nova de Lisboa, 1169-056 Lisboa, Portugal; (M.J.C.); (A.B.P.); (F.L.-C.); (C.O.S.); (N.R.C.); (C.G.-D.); (J.S.); (J.M.); (E.C.M.)
| | - Filipa Lopes-Coelho
- CEDOC, NOVA Medical School, Universidade Nova de Lisboa, 1169-056 Lisboa, Portugal; (M.J.C.); (A.B.P.); (F.L.-C.); (C.O.S.); (N.R.C.); (C.G.-D.); (J.S.); (J.M.); (E.C.M.)
- Instituto Português de Oncologia de Lisboa Francisco Gentil (IPOLFG), Rua Prof Lima Basto, 1099-023 Lisboa, Portugal
| | - Catarina O. Sequeira
- CEDOC, NOVA Medical School, Universidade Nova de Lisboa, 1169-056 Lisboa, Portugal; (M.J.C.); (A.B.P.); (F.L.-C.); (C.O.S.); (N.R.C.); (C.G.-D.); (J.S.); (J.M.); (E.C.M.)
| | - Nuno R. Coelho
- CEDOC, NOVA Medical School, Universidade Nova de Lisboa, 1169-056 Lisboa, Portugal; (M.J.C.); (A.B.P.); (F.L.-C.); (C.O.S.); (N.R.C.); (C.G.-D.); (J.S.); (J.M.); (E.C.M.)
| | - Clara Gonçalves-Dias
- CEDOC, NOVA Medical School, Universidade Nova de Lisboa, 1169-056 Lisboa, Portugal; (M.J.C.); (A.B.P.); (F.L.-C.); (C.O.S.); (N.R.C.); (C.G.-D.); (J.S.); (J.M.); (E.C.M.)
| | - Robert Barouki
- INSERM UMR-S 1124, 3TS, Environmental Toxicity, Therapeutic Targets, Cellular Signaling and Biomarkers, Université de Paris, 45 rue des Saints-Pères, 75006 Paris, France; (R.B.); (X.C.)
| | - Xavier Coumoul
- INSERM UMR-S 1124, 3TS, Environmental Toxicity, Therapeutic Targets, Cellular Signaling and Biomarkers, Université de Paris, 45 rue des Saints-Pères, 75006 Paris, France; (R.B.); (X.C.)
| | - Jacinta Serpa
- CEDOC, NOVA Medical School, Universidade Nova de Lisboa, 1169-056 Lisboa, Portugal; (M.J.C.); (A.B.P.); (F.L.-C.); (C.O.S.); (N.R.C.); (C.G.-D.); (J.S.); (J.M.); (E.C.M.)
- Instituto Português de Oncologia de Lisboa Francisco Gentil (IPOLFG), Rua Prof Lima Basto, 1099-023 Lisboa, Portugal
| | - Judit Morello
- CEDOC, NOVA Medical School, Universidade Nova de Lisboa, 1169-056 Lisboa, Portugal; (M.J.C.); (A.B.P.); (F.L.-C.); (C.O.S.); (N.R.C.); (C.G.-D.); (J.S.); (J.M.); (E.C.M.)
| | - Emília C. Monteiro
- CEDOC, NOVA Medical School, Universidade Nova de Lisboa, 1169-056 Lisboa, Portugal; (M.J.C.); (A.B.P.); (F.L.-C.); (C.O.S.); (N.R.C.); (C.G.-D.); (J.S.); (J.M.); (E.C.M.)
| | - Sofia A. Pereira
- CEDOC, NOVA Medical School, Universidade Nova de Lisboa, 1169-056 Lisboa, Portugal; (M.J.C.); (A.B.P.); (F.L.-C.); (C.O.S.); (N.R.C.); (C.G.-D.); (J.S.); (J.M.); (E.C.M.)
- Correspondence:
| |
Collapse
|
70
|
Sainsily X, Coquerel D, Giguère H, Dumont L, Tran K, Noll C, Ionescu AL, Côté J, Longpré JM, Carpentier A, Marsault É, Lesur O, Sarret P, Auger-Messier M. Elabela Protects Spontaneously Hypertensive Rats From Hypertension and Cardiorenal Dysfunctions Exacerbated by Dietary High-Salt Intake. Front Pharmacol 2021; 12:709467. [PMID: 34385922 PMCID: PMC8353398 DOI: 10.3389/fphar.2021.709467] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 07/14/2021] [Indexed: 12/11/2022] Open
Abstract
Objectives: Arterial hypertension, when exacerbated by excessive dietary salt intake, worsens the morbidity and mortality rates associated with cardiovascular and renal diseases. Stimulation of the apelinergic system appears to protect against several circulatory system diseases, but it remains unknown if such beneficial effects are conserved in severe hypertension. Therefore, we aimed at determining whether continuous infusion of apelinergic ligands (i.e., Apelin-13 and Elabela) exerted cardiorenal protective effects in spontaneously hypertensive (SHR) rats receiving high-salt diet. Methods: A combination of echocardiography, binding assay, histology, and biochemical approaches were used to investigate the cardiovascular and renal effects of Apelin-13 or Elabela infusion over 6 weeks in SHR fed with normal-salt or high-salt chow. Results: High-salt intake upregulated the cardiac and renal expression of APJ receptor in SHR. Importantly, Elabela was more effective than Apelin-13 in reducing high blood pressure, cardiovascular and renal dysfunctions, fibrosis and hypertrophy in high-salt fed SHR. Unlike Apelin-13, the beneficial effects of Elabela were associated with a counter-regulatory role of the ACE/ACE2/neprilysin axis of the renin-angiotensin-aldosterone system (RAAS) in heart and kidneys of salt-loaded SHR. Interestingly, Elabela also displayed higher affinity for APJ in the presence of high salt concentration and better resistance to RAAS enzymes known to cleave Apelin-13. Conclusion: These findings highlight the protective action of the apelinergic system against salt-induced severe hypertension and cardiorenal failure. As compared with Apelin-13, Elabela displays superior pharmacodynamic and pharmacokinetic properties that warrant further investigation of its therapeutic use in cardiovascular and kidney diseases.
Collapse
Affiliation(s)
- Xavier Sainsily
- Département de Médecine, Centre de Recherche du CHUS, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC, Canada.,Département de Pharmacologie et Physiologie, Centre de Recherche du CHUS, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC, Canada.,Institut de Pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - David Coquerel
- Département de Médecine, Centre de Recherche du CHUS, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC, Canada.,Institut de Pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Hugo Giguère
- Département de Médecine, Centre de Recherche du CHUS, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC, Canada.,Institut de Pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Lauralyne Dumont
- Département de Médecine, Centre de Recherche du CHUS, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC, Canada.,Institut de Pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Kien Tran
- Département de Pharmacologie et Physiologie, Centre de Recherche du CHUS, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC, Canada.,Institut de Pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Christophe Noll
- Département de Médecine, Centre de Recherche du CHUS, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Andrei L Ionescu
- Département de Médecine, Centre de Recherche du CHUS, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC, Canada.,Institut de Pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Jérôme Côté
- Département de Pharmacologie et Physiologie, Centre de Recherche du CHUS, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC, Canada.,Institut de Pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Jean-Michel Longpré
- Département de Pharmacologie et Physiologie, Centre de Recherche du CHUS, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC, Canada.,Institut de Pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - André Carpentier
- Département de Médecine, Centre de Recherche du CHUS, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Éric Marsault
- Département de Pharmacologie et Physiologie, Centre de Recherche du CHUS, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC, Canada.,Institut de Pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Olivier Lesur
- Département de Médecine, Centre de Recherche du CHUS, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC, Canada.,Institut de Pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Philippe Sarret
- Département de Pharmacologie et Physiologie, Centre de Recherche du CHUS, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC, Canada.,Institut de Pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Mannix Auger-Messier
- Département de Médecine, Centre de Recherche du CHUS, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC, Canada.,Institut de Pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada
| |
Collapse
|
71
|
Liu J, Liu Y, Wang F, Liang M. miR-204: Molecular Regulation and Role in Cardiovascular and Renal Diseases. Hypertension 2021; 78:270-281. [PMID: 34176282 DOI: 10.1161/hypertensionaha.121.14536] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The field of microRNA research has evolved from studies aiming to gauge the importance of microRNAs to those focusing on understanding a subset of specific microRNAs that have emerged as potent regulators of molecular systems and pathophysiological conditions. In this article, we review the molecular features and regulation of miR-204 and the growing body of evidence for an important role of miR-204 in the regulation of cardiovascular and renal physiology and pathophysiological processes. miR-204 exhibits a highly tissue-specific expression pattern, and miR-204 abundance is regulated by several transcriptional and posttranscriptional mechanisms. Strong evidence supports a role for miR-204 in attenuating pulmonary arterial hypertension and hypertensive and diabetic renal injury while promoting hypertension and endothelial dysfunction in a wide range of model systems. miR-204 may influence these disease processes by targeting several biological pathways in a tissue-specific manner. miR-204 is dysregulated in patients with cardiovascular and renal diseases. The unequivocal functional roles and clear clinical relevance indicate that miR-204 is a high-value microRNA in cardiovascular and renal diseases.
Collapse
Affiliation(s)
- Jing Liu
- Department of Physiology, Center of Systems Molecular Medicine, Medical College of Wisconsin, Milwaukee
| | - Yong Liu
- Department of Physiology, Center of Systems Molecular Medicine, Medical College of Wisconsin, Milwaukee
| | - Feng Wang
- Department of Physiology, Center of Systems Molecular Medicine, Medical College of Wisconsin, Milwaukee
| | - Mingyu Liang
- Department of Physiology, Center of Systems Molecular Medicine, Medical College of Wisconsin, Milwaukee
| |
Collapse
|
72
|
Pan X, Geng X, Liu Y, Yu M, Mishra MK, Xu X, Ding X, Liu P, Liang M. Transfer RNA Fragments in the Kidney in Hypertension. Hypertension 2021; 77:1627-1637. [PMID: 33775129 DOI: 10.1161/hypertensionaha.121.16994] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Xiaoqing Pan
- Department of Mathematics, Shanghai Normal University, China (X.P.).,Center of Systems Molecular Medicine, Department of Physiology, Medical College of Wisconsin, Milwaukee (X.P., X.G., Y.L., M.K.M., P.L., M.L.)
| | - Xuemei Geng
- Center of Systems Molecular Medicine, Department of Physiology, Medical College of Wisconsin, Milwaukee (X.P., X.G., Y.L., M.K.M., P.L., M.L.).,Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai Institute of Kidney and Dialysis, Shanghai Key Laboratory of Kidney and Blood Purification, Shanghai Medical Center of Kidney Disease, China (X.G., X.X., X.D.)
| | - Yong Liu
- Center of Systems Molecular Medicine, Department of Physiology, Medical College of Wisconsin, Milwaukee (X.P., X.G., Y.L., M.K.M., P.L., M.L.)
| | - Mengqian Yu
- Department of Respiratory Medicine, Sir Run Run Shaw Hospital and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China (M.Y., P.L.)
| | - Manoj K Mishra
- Center of Systems Molecular Medicine, Department of Physiology, Medical College of Wisconsin, Milwaukee (X.P., X.G., Y.L., M.K.M., P.L., M.L.)
| | - Xialian Xu
- Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai Institute of Kidney and Dialysis, Shanghai Key Laboratory of Kidney and Blood Purification, Shanghai Medical Center of Kidney Disease, China (X.G., X.X., X.D.)
| | - Xiaoqiang Ding
- Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai Institute of Kidney and Dialysis, Shanghai Key Laboratory of Kidney and Blood Purification, Shanghai Medical Center of Kidney Disease, China (X.G., X.X., X.D.)
| | - Pengyuan Liu
- Center of Systems Molecular Medicine, Department of Physiology, Medical College of Wisconsin, Milwaukee (X.P., X.G., Y.L., M.K.M., P.L., M.L.).,Department of Respiratory Medicine, Sir Run Run Shaw Hospital and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China (M.Y., P.L.)
| | - Mingyu Liang
- Center of Systems Molecular Medicine, Department of Physiology, Medical College of Wisconsin, Milwaukee (X.P., X.G., Y.L., M.K.M., P.L., M.L.)
| |
Collapse
|