1
|
Chen L, Xu S, Wang S, Chen H, Han Y. Xiaohuafuning tang intervenes liver-depression-and-spleen-deficiency syndrome chronic-atrophic-gastritis by reshaping amino acid metabolism through gut Microbiota. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 136:156346. [PMID: 39740378 DOI: 10.1016/j.phymed.2024.156346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 11/22/2024] [Accepted: 12/23/2024] [Indexed: 01/02/2025]
Abstract
BACKGROUND Xiaohua Funing Tang (XHFND) is a decoction formula of traditional Chinese medicine (TCM) and possesses the potential to manage chronic atrophic gastritis (CAG) with liver depression and spleen deficiency (LDSD), but the mechanisms were still unclear. PURPOSE Our aim is to reveal the overall synergistic mechanisms of XHFND against CAG with LDSD. METHODS Based on a CAG rat model with LDSD, this study combined metabolomics, gut microbiota, and network pharmacology techniques to demonstrate the XHFND mechanisms with multiple components and targets. RESULTS Through the integration analysis of gut microbiome and metabolome using metorigin, we found that XHFND regulates arginine metabolism levels in the urea cycle by regulating the gut ecological environment and the host. The XHFND mainly promotes aspartate 1 metabolism by regulating the abundance of odoribacter, bacteroides, phocaeicola, lachnospire, and intestinimonas, intervened in the imbalance of arginine metabolism in CAG rats with LDSD, suppresses the pathogenic Th17 cell differentiation, and inhibits the gastric mucosa damage in rats. Through Cytoscape analysis of network pharmacology and metabolomics integration, we found that XHFND might regulate host phospholipid metabolism through PTEN and PIK3CA to inhibit the PI3K-AKT-TSC axis and then inhibit mTORC1 to control arginine metabolism in the urea cycle and produce polyamines, thereby inhibiting the pathogenic Th17 cell differentiation and preventing rat gastric mucosa damage. Intervention in arginine metabolism in the urea cycle is the primary pathway in which XHFND exerts its therapeutic effects. XHFND may mainly control the pathogenic Th17 cell differentiation in the gastric mucosa of model rats through the pathway. CONCLUSION This study indicated that XHFND might intervene in treating CAG with LDSD from multiple levels and perspectives to suppress the pathogenic Th17 cell differentiation, which aligns with the characteristics of TCM treatment. This study presents experimental evidence for the clinical use of XHFND and promotes the development of drugs for the therapy of CAG with LDSD.
Collapse
Affiliation(s)
- Li Chen
- Institue of Pharmaceutical Department, The First Affiliated Hospital of Anhui University of Traditional Chinese Medicine, No 117, Meishan Road, Hefei 230031, Anhui, PR China
| | - ShaoYu Xu
- Institue of School of Pharmacy, Anhui University of Chinese Medicine, No 350, Longzihu Road, Hefei 230012, Anhui, PR China
| | - Sheng Wang
- Institue of Pharmaceutical Department, The First Affiliated Hospital of Anhui University of Traditional Chinese Medicine, No 117, Meishan Road, Hefei 230031, Anhui, PR China
| | - Hao Chen
- Institue of Pharmaceutical Department, The First Affiliated Hospital of Anhui University of Traditional Chinese Medicine, No 117, Meishan Road, Hefei 230031, Anhui, PR China.
| | - Yanquan Han
- Institue of Pharmaceutical Department, The First Affiliated Hospital of Anhui University of Traditional Chinese Medicine, No 117, Meishan Road, Hefei 230031, Anhui, PR China.
| |
Collapse
|
2
|
Li H, Cheng Z, Wu D, Hu Q. Nitric oxide and mitochondrial function in cardiovascular diseases. Nitric Oxide 2024; 154:42-50. [PMID: 39577487 DOI: 10.1016/j.niox.2024.11.007] [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: 07/02/2024] [Revised: 09/11/2024] [Accepted: 11/19/2024] [Indexed: 11/24/2024]
Abstract
Nitric oxide (NO) has been highlighted as an important factor in cardiovascular system. As a signaling molecule in the cardiovascular system, NO can relax blood vessels, lower blood pressure, and prevent platelet aggregation. Mitochondria serve as a central hub for cellular metabolism and intracellular signaling, and their dysfunction can lead to a variety of diseases. Accumulating evidence suggests that NO can act as a regulator of mitochondria, affecting mitochondrial function and cellular activity, which in turn mediates the onset and progression of disease. However, there is a lack of comprehensive understanding of how NO regulates mitochondrial function in the cardiovascular system. This review aims to summarize the regulation of mitochondrial function by nitric oxide in cardiovascular related diseases, as well as the multifaceted and complex roles of NO in the cardiovascular system. Understanding the mechanism of NO mediated mitochondrial function can provide new insights for the prevention and treatment of cardiovascular diseases.
Collapse
Affiliation(s)
- Haoqi Li
- Department of Pharmacy, School of Medicine, Shanghai University, Shanghai, 200444, China
| | - Zijie Cheng
- Department of Pharmacy, School of Medicine, Shanghai University, Shanghai, 200444, China
| | - Dan Wu
- Department of Pharmacy, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China.
| | - Qingxun Hu
- Department of Pharmacy, School of Medicine, Shanghai University, Shanghai, 200444, China.
| |
Collapse
|
3
|
Qin QR, Chen J, Hu WL, Liu JJ, Liu MY, Huang F, Hu MJ. Association of tricarboxylic acid cycle related-metabolites with hypertension in older adults: a community-based cross-sectional study. J Hum Hypertens 2024:10.1038/s41371-024-00976-5. [PMID: 39528733 DOI: 10.1038/s41371-024-00976-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 10/29/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024]
Abstract
Hypertension is still a common chronic disease worldwide and seriously affects human health. Aim of this study was to investigate the association between tricarboxylic acid (TCA) cycle-related metabolites and hypertension in older adults. A total of 1127 community-dwelling older adults were included in this cross-sectional analysis, of whom 609 were assigned to the hypertension group and 518 to the no-hypertension group. Plasma concentrations of 8 TCA cycle-related metabolites (citrate, cis-aconitate, isocitrate, 2-oxoglutarate, succinate, maleate, fumarate, and malate) were determined by gas chromatography-triple quadrupole mass spectrometry. Multivariate logistic regression was used to assess the association between these metabolites and hypertension risk. After adjustment for covariates, we found that the increased plasma concentrations of 2-oxoglutarate and malate were significantly associated with hypertension. These two associations remained unchanged after using the false discovery rate (FDR)-adjustment method (both FDR-adjusted P-trend <0.05). In stratified analysis, these two associations were not modified by overweight, physical activity, and current drinking (all P-interaction >0.05). In the multivariate diagnostic model, the inclusion of these two metabolites modestly and significantly improved the diagnosis of hypertension (AUC = 0.64, 95% CI: 0.60-0.67). These results indicated that plasma 2-oxoglutarate and malate might be the candidate metabolic markers of hypertension among older adults. However, further longitudinal and experimental studies are necessary to confirm this finding.
Collapse
Affiliation(s)
- Qi-Rong Qin
- Ma'anshan Center for Disease Control and Prevention, Ma'anshan, China
- The Affiliated Ma'anshan Center for Disease Control and Prevention of Anhui Medical University, Ma'anshan, China
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, China
| | - Jian Chen
- Ma'anshan Center for Disease Control and Prevention, Ma'anshan, China
- The Affiliated Ma'anshan Center for Disease Control and Prevention of Anhui Medical University, Ma'anshan, China
| | - Wen-Lei Hu
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, China
| | - Jian-Jun Liu
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, China
| | - Meng-Yu Liu
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, China
| | - Fen Huang
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, China
| | - Ming-Jun Hu
- Department of Health Management Center, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
| |
Collapse
|
4
|
Xu B, Dissanayake LV, Levchenko V, Zietara A, Kravtsova O, Staruschenko A. Deletion of Kcnj16 altered transcriptomic and metabolomic profiles of Dahl salt-sensitive rats. iScience 2024; 27:110901. [PMID: 39328933 PMCID: PMC11424968 DOI: 10.1016/j.isci.2024.110901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 04/06/2024] [Accepted: 09/04/2024] [Indexed: 09/28/2024] Open
Abstract
The inwardly rectifying K+ channel Kir5.1 (Kcnj16) is essential in renal salt handling and blood pressure control. However, the underlying mechanisms are not fully understood. Here, we integrated transcriptomics and metabolomics to comprehensively profile the changes in genes and metabolites in the Dahl salt-sensitive (SS) rat lacking Kcnj16 to identify potential mechanisms. Consistent with the phenotype of knockout (KO) rats, the transcriptomic profile predicted reduced blood pressure, kidney damage, and increased ion transport. Canonical pathway analysis suggested activation of metabolic-related pathways while suppression of immune response-related pathways in KO rats. Untargeted metabolomic analysis revealed different metabolic profiles between wild-type (WT) and KO rats. Integration of transcriptomic and metabolomic profiles suggested altered tricarboxylic acid (TCA) cycle, amino acid metabolism, and reactive oxygen species (ROS) metabolism that are related to SS hypertension. In conclusion, besides increased ion transport, our data suggest suppressed immune response-related and altered metabolic-related pathways of SS rats lacking Kir5.1.
Collapse
Affiliation(s)
- Biyang Xu
- Department of Molecular Pharmacology & Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Lashodya V Dissanayake
- Department of Molecular Pharmacology & Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Vladislav Levchenko
- Department of Molecular Pharmacology & Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Adrian Zietara
- Department of Molecular Pharmacology & Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Olha Kravtsova
- Department of Molecular Pharmacology & Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Alexander Staruschenko
- Department of Molecular Pharmacology & Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
- Hypertension and Kidney Research Center, University of South Florida, Tampa, FL, USA
- James A. Haley Veteran's Hospital, Tampa, FL, USA
| |
Collapse
|
5
|
Silva-Lance F, Montejano-Montelongo I, Bautista E, Nielsen LK, Johansson PI, Marin de Mas I. Integrating Genome-Scale Metabolic Models with Patient Plasma Metabolome to Study Endothelial Metabolism In Situ. Int J Mol Sci 2024; 25:5406. [PMID: 38791446 PMCID: PMC11121795 DOI: 10.3390/ijms25105406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 04/30/2024] [Accepted: 05/08/2024] [Indexed: 05/26/2024] Open
Abstract
Patient blood samples are invaluable in clinical omics databases, yet current methodologies often fail to fully uncover the molecular mechanisms driving patient pathology. While genome-scale metabolic models (GEMs) show promise in systems medicine by integrating various omics data, having only exometabolomic data remains a limiting factor. To address this gap, we introduce a comprehensive pipeline integrating GEMs with patient plasma metabolome. This pipeline constructs case-specific GEMs using literature-based and patient-specific metabolomic data. Novel computational methods, including adaptive sampling and an in-house developed algorithm for the rational exploration of the sampled space of solutions, enhance integration accuracy while improving computational performance. Model characterization involves task analysis in combination with clustering methods to identify critical cellular functions. The new pipeline was applied to a cohort of trauma patients to investigate shock-induced endotheliopathy using patient plasma metabolome data. By analyzing endothelial cell metabolism comprehensively, the pipeline identified critical therapeutic targets and biomarkers that can potentially contribute to the development of therapeutic strategies. Our study demonstrates the efficacy of integrating patient plasma metabolome data into computational models to analyze endothelial cell metabolism in disease contexts. This approach offers a deeper understanding of metabolic dysregulations and provides insights into diseases with metabolic components and potential treatments.
Collapse
Affiliation(s)
- Fernando Silva-Lance
- Novo Nordisk Foundation Center for Biosustainability, Danish Technical University, 2800 Lyngby, Denmark
| | | | - Eric Bautista
- Novo Nordisk Foundation Center for Biosustainability, Danish Technical University, 2800 Lyngby, Denmark
| | - Lars K. Nielsen
- Novo Nordisk Foundation Center for Biosustainability, Danish Technical University, 2800 Lyngby, Denmark
- CAG Center for Endotheliomics, Copenhagen University Hospital, Rigshospitalet, 2100 Copenhagen, Denmark
| | - Pär I. Johansson
- CAG Center for Endotheliomics, Copenhagen University Hospital, Rigshospitalet, 2100 Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Igor Marin de Mas
- Novo Nordisk Foundation Center for Biosustainability, Danish Technical University, 2800 Lyngby, Denmark
- CAG Center for Endotheliomics, Copenhagen University Hospital, Rigshospitalet, 2100 Copenhagen, Denmark
| |
Collapse
|
6
|
Lassé M, Rinschen MM. High Salt Remodels Kidney Metabolism: Metabolite Fuel, Fate, and Signals. FUNCTION 2024; 5:zqae006. [PMID: 38486978 PMCID: PMC10935453 DOI: 10.1093/function/zqae006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 01/25/2024] [Accepted: 01/26/2024] [Indexed: 03/17/2024] Open
Affiliation(s)
- Moritz Lassé
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf (UKE), Hamburg 20246, Germany
- Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany
| | - Markus M Rinschen
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf (UKE), Hamburg 20246, Germany
- Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany
- Department of Biomedicine, Aarhus University, Aarhus 8000, Denmark
- Aarhus Institute of Advanced Studies (AIAS), Aarhus 8000, Denmark
| |
Collapse
|
7
|
Stadler K, Ilatovskaya DV. Renal Epithelial Mitochondria: Implications for Hypertensive Kidney Disease. Compr Physiol 2023; 14:5225-5242. [PMID: 38158371 PMCID: PMC11194858 DOI: 10.1002/cphy.c220033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2024]
Abstract
According to the Centers for Disease Control and Prevention, 1 in 2 U.S. adults have hypertension, and more than 1 in 7 chronic kidney disease. In fact, hypertension is the second leading cause of kidney failure in the United States; it is a complex disease characterized by, leading to, and caused by renal dysfunction. It is well-established that hypertensive renal damage is accompanied by mitochondrial damage and oxidative stress, which are differentially regulated and manifested along the nephron due to the diverse structure and functions of renal cells. This article provides a summary of the relevant knowledge of mitochondrial bioenergetics and metabolism, focuses on renal mitochondrial function, and discusses the evidence that has been accumulated regarding the role of epithelial mitochondrial bioenergetics in the development of renal tissue dysfunction in hypertension. © 2024 American Physiological Society. Compr Physiol 14:5225-5242, 2024.
Collapse
Affiliation(s)
- Krisztian Stadler
- Oxidative Stress and Disease Laboratory, Pennington Biomedical Research Center, Baton Rouge, Louisiana, USA
| | - Daria V. Ilatovskaya
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| |
Collapse
|
8
|
Edosuyi O, Igbe I, Oyekan A. Fumarate and its downstream signalling pathways in the cardiorenal system: Recent insights and novel expositions in the etiology of hypertension. Eur J Pharmacol 2023; 961:176186. [PMID: 37944846 PMCID: PMC10843741 DOI: 10.1016/j.ejphar.2023.176186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 11/02/2023] [Accepted: 11/03/2023] [Indexed: 11/12/2023]
Abstract
Hypertension, a risk factor for cardiorenal disease has a huge global health impact. Hence, there is a continuous search for new therapeutic targets and putative antihypertensive ligands. This search has transcended into the realm of mitochondrial metabolism which has been reported to underline the etiology of certain diseases, including hypertension. Recently, genetic alterations in the tricarboxylic acid (TCA) cycle enzyme, fumarase, which converts fumarate to malate, reportedly worsened salt-sensitive hypertension. These novel expositions shifted focus into the activity of TCA in the pathogenesis of hypertension. There is now evidence to show that a mechanistic link exists between blood pressure regulation and intermediaries in the TCA cycle involving fumarate metabolism. Fumarate has been reported to mediate the actions of endogenous ligands such as nitric oxide (NO), and hypoxia inducible factor (HIF)-1α. Similarly, there has been upregulation of protective genes such as nuclear erythroid factor 2 (Nrf2) and reduction in the expression of certain markers like kidney injury molecule 1 (KIM-1). There are reports of interactions with endogenous enzymes such as catalase (CAT) and renin via the activation of GPR91. Fumarate has also been shown to modulate the actions of renal ion channels and by extension, natriuresis. These actions of fumarate have conferred a reno- and cardio-protective effect in hypertension. This review evaluates the role of the TCA cycle, its mechanistic links, and significant contribution to blood pressure regulation with a view to understanding the possibility of a new pathological axis which may be involved in the pathogenesis of hypertension.
Collapse
Affiliation(s)
- Osaze Edosuyi
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, University of Benin, PMB 1154, Benin City, Nigeria; Center for Cardiovascular Diseases, Gray Hall Suites, Rm 256, College of Pharmacy & Health Sciences, Texas Southern University, 3100, Cleburne Street, Houston, TX, USA.
| | - Ighodaro Igbe
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, University of Benin, PMB 1154, Benin City, Nigeria
| | - Adebayo Oyekan
- Center for Cardiovascular Diseases, Gray Hall Suites, Rm 256, College of Pharmacy & Health Sciences, Texas Southern University, 3100, Cleburne Street, Houston, TX, USA
| |
Collapse
|
9
|
Zhu Y, Wang J, Yao L, Huang Y, Yang H, Yu X, Chen X, Chen Y. Electroacupuncture at BL15 attenuates chronic fatigue syndrome by downregulating iNOS/NO signaling in C57BL/6 mice. Anat Rec (Hoboken) 2023; 306:3073-3084. [PMID: 35608198 DOI: 10.1002/ar.24953] [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: 12/02/2021] [Revised: 03/19/2022] [Accepted: 04/22/2022] [Indexed: 11/11/2022]
Abstract
Chronic fatigue syndrome (CFS) has a high incidence due to the increased pressure of daily life and work in modern society. Our previous clinical studies have found the effects of electroacupuncture (EA) on CFS patients, however, the mechanism of EA on CFS is still unknown. In this study, we investigated the effects of EA on cardiac function in a CFS mouse model to explore its underlying mechanism. The mice were randomly divided into three groups: control, CFS, and CFS mice receiving EA (CFS + EA). After behavioral assessments and echocardiographic measurement, blood and heart tissue of the mice were collected for biochemical tests, and then we evaluated the effects of EA on the CFS mouse model when nitric oxide (NO) levels were enhanced by l-arginine. The results showed that EA ameliorated the injured motor and cardiac function. Meanwhile, EA also inhibited increased expression of inducible nitric oxide synthase (iNOS) at heart tissue and the serum NO levels in mice subjected to sustained forced swimming stress. Furthermore, the NO level in serum increased with l-arginine administration, which blocked the effects of EA on CFS mice. This study suggested that EA could improve the motor function and cardiac function in CFS mice and its effects may be associated with the down-regulation of iNOS/NO signaling.
Collapse
Affiliation(s)
- Yang Zhu
- The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jingya Wang
- South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Lin Yao
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yunxuan Huang
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Haitao Yang
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiaojiang Yu
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xinghua Chen
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yongjun Chen
- South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
- Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangzhou, China
| |
Collapse
|
10
|
Trejo-Moreno C, Alvarado-Ojeda ZA, Méndez-Martínez M, Cruz-Muñoz ME, Castro-Martínez G, Arrellín-Rosas G, Zamilpa A, Jimenez-Ferrer JE, Baez Reyes JC, Fragoso G, Salgado GR. Aqueous Fraction from Cucumis sativus Aerial Parts Attenuates Angiotensin II-Induced Endothelial Dysfunction In Vivo by Activating Akt. Nutrients 2023; 15:4680. [PMID: 37960332 PMCID: PMC10649625 DOI: 10.3390/nu15214680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 10/27/2023] [Accepted: 11/02/2023] [Indexed: 11/15/2023] Open
Abstract
BACKGROUND Endothelial dysfunction (ED) is a marker of vascular damage and a precursor of cardiovascular diseases such as hypertension, which involve inflammation and organ damage. Nitric oxide (NO), produced by eNOS, which is induced by pAKT, plays a crucial role in the function of a healthy endothelium. METHODS A combination of subfractions SF1 and SF3 (C4) of the aqueous fraction from Cucumis sativus (Cs-Aq) was evaluated to control endothelial dysfunction in vivo and on HMEC-1 cells to assess the involvement of pAkt in vitro. C57BL/6J mice were injected daily with angiotensin II (Ang-II) for 10 weeks. Once hypertension was established, either Cs-AqC4 or losartan was orally administered along with Ang-II for a further 10 weeks. Blood pressure (BP) was measured at weeks 0, 5, 10, 15, and 20. In addition, serum creatinine, inflammatory status (in the kidney), tissue damage, and vascular remodeling (in the liver and aorta) were evaluated. Cs-AqC4 was also tested in vitro on HMEC-1 cells stimulated by Ang-II to assess the involvement of Akt phosphorylation. RESULTS Cs-AqC4 decreased systolic and diastolic BP, reversed vascular remodeling, decreased IL-1β and TGF-β, increased IL-10, and decreased kidney and liver damage. In HMEC-1 cells, AKT phosphorylation and NO production were increased. CONCLUSIONS Cs-AqC4 controlled inflammation and vascular remodeling, alleviating hypertension; it also improved tissue damage associated with ED, probably via Akt activation.
Collapse
Affiliation(s)
- Celeste Trejo-Moreno
- Facultad de Medicina, Universidad Autónoma del Estado de Morelos, Cuernavaca 62350, Morelos, Mexico; (C.T.-M.); (Z.A.A.-O.); (M.E.C.-M.); (G.A.-R.)
| | - Zimri Aziel Alvarado-Ojeda
- Facultad de Medicina, Universidad Autónoma del Estado de Morelos, Cuernavaca 62350, Morelos, Mexico; (C.T.-M.); (Z.A.A.-O.); (M.E.C.-M.); (G.A.-R.)
| | - Marisol Méndez-Martínez
- Departamento de Sistemas Biológicos, División de Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana-Xochimilco, Ciudad de México 04960, Mexico;
| | - Mario Ernesto Cruz-Muñoz
- Facultad de Medicina, Universidad Autónoma del Estado de Morelos, Cuernavaca 62350, Morelos, Mexico; (C.T.-M.); (Z.A.A.-O.); (M.E.C.-M.); (G.A.-R.)
| | - Gabriela Castro-Martínez
- Doctorado en Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana, Ciudad de México 04960, Mexico;
| | - Gerardo Arrellín-Rosas
- Facultad de Medicina, Universidad Autónoma del Estado de Morelos, Cuernavaca 62350, Morelos, Mexico; (C.T.-M.); (Z.A.A.-O.); (M.E.C.-M.); (G.A.-R.)
- Facultad de Ciencias de la Salud, Universidad Panamericana, Ciudad de México 03920, Mexico
| | - Alejandro Zamilpa
- Centro de Investigación Biomédica del Sur, Instituto Mexicano del Seguro Social, Xochitepec 62790, Morelos, Mexico; (A.Z.); (J.E.J.-F.)
| | - Jesús Enrique Jimenez-Ferrer
- Centro de Investigación Biomédica del Sur, Instituto Mexicano del Seguro Social, Xochitepec 62790, Morelos, Mexico; (A.Z.); (J.E.J.-F.)
| | - Juan Carlos Baez Reyes
- Escuela Nacional Preparatoria No. 1, Universidad Nacional Autónoma de México, Ciudad de México 16030, Mexico;
| | - Gladis Fragoso
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico
| | - Gabriela Rosas Salgado
- Facultad de Medicina, Universidad Autónoma del Estado de Morelos, Cuernavaca 62350, Morelos, Mexico; (C.T.-M.); (Z.A.A.-O.); (M.E.C.-M.); (G.A.-R.)
| |
Collapse
|
11
|
Holeček M. Aspartic Acid in Health and Disease. Nutrients 2023; 15:4023. [PMID: 37764806 PMCID: PMC10536334 DOI: 10.3390/nu15184023] [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: 08/28/2023] [Revised: 09/12/2023] [Accepted: 09/15/2023] [Indexed: 09/29/2023] Open
Abstract
Aspartic acid exists in L- and D-isoforms (L-Asp and D-Asp). Most L-Asp is synthesized by mitochondrial aspartate aminotransferase from oxaloacetate and glutamate acquired by glutamine deamidation, particularly in the liver and tumor cells, and transamination of branched-chain amino acids (BCAAs), particularly in muscles. The main source of D-Asp is the racemization of L-Asp. L-Asp transported via aspartate-glutamate carrier to the cytosol is used in protein and nucleotide synthesis, gluconeogenesis, urea, and purine-nucleotide cycles, and neurotransmission and via the malate-aspartate shuttle maintains NADH delivery to mitochondria and redox balance. L-Asp released from neurons connects with the glutamate-glutamine cycle and ensures glycolysis and ammonia detoxification in astrocytes. D-Asp has a role in brain development and hypothalamus regulation. The hereditary disorders in L-Asp metabolism include citrullinemia, asparagine synthetase deficiency, Canavan disease, and dicarboxylic aminoaciduria. L-Asp plays a role in the pathogenesis of psychiatric and neurologic disorders and alterations in BCAA levels in diabetes and hyperammonemia. Further research is needed to examine the targeting of L-Asp metabolism as a strategy to fight cancer, the use of L-Asp as a dietary supplement, and the risks of increased L-Asp consumption. The role of D-Asp in the brain warrants studies on its therapeutic potential in psychiatric and neurologic disorders.
Collapse
Affiliation(s)
- Milan Holeček
- Department of Physiology, Faculty of Medicine in Hradec Králové, Charles University, Šimkova 870, 500 03 Hradec Králové, Czech Republic
| |
Collapse
|
12
|
Du TT, Liu XC, He Y, Gao X, Liu ZZ, Wang ZL, Li LQ. Changes of gut microbiota and tricarboxylic acid metabolites may be helpful in early diagnosis of necrotizing enterocolitis: A pilot study. Front Microbiol 2023; 14:1119981. [PMID: 37007499 PMCID: PMC10050441 DOI: 10.3389/fmicb.2023.1119981] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 02/23/2023] [Indexed: 03/17/2023] Open
Abstract
PurposeWe aimed to explore the value of gut microbiota and tricarboxylic acid (TCA) metabolites in early diagnosis of necrotizing enterocolitis (NEC) among infants with abdominal manifestations.MethodsThirty-two preterm infants with abdominal manifestations at gestational age ≤ 34 weeks were included in the study and were divided into non-NEC (n = 16) and NEC (n = 16) groups. Faecal samples were collected when the infants were enrolled. The gut microbiota was analysed with high-throughput sequencing, and TCA metabolites were measured with multiple reaction monitoring (MRM) targeted metabolomics. Receiver operating characteristic (ROC) curves were generated to explore the predictive value of the obtained data.ResultsThere was no significant difference in alpha diversity or beta diversity between the two groups (p > 0.05). At the phylum level, Proteobacteria increased, and Actinomycetota decreased in the NEC group (p < 0.05). At the genus level, Bifidobacterium and Lactobacillaceae decreased significantly, and at the species level, unclassified Staphylococcus, Lactobacillaceae and Bifidobacterium animalis subsp. lactis decreased in the NEC group (p < 0.05). Further Linear discriminant analysis effect sizes (LEfSe) analysis showed that the change in Proteobacteria at the phylum level and Lactobacillaceae and Bifidobacterium at the genus level scored higher than 4. The concentrations of succinate, L-malic acid and oxaloacetate in the NEC group significantly increased (p < 0.05), and the areas under the ROC curve for these metabolites were 0.6641, 0.7617, and 0.7344, respectively.ConclusionDecreased unclassified Staphylococcus, Lactobacillaceae and Bifidobacterium animalis subsp. lactis at the species level as well as the increase in the contents of some TCA metabolites, including succinate, L-malic acid and oxaloacetate, have potential value for the early diagnosis of NEC.
Collapse
Affiliation(s)
- Ting-Ting Du
- Neonatal Diagnosis and Treatment Centre of Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Xiao-Chen Liu
- Neonatal Diagnosis and Treatment Centre of Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Yu He
- Neonatal Diagnosis and Treatment Centre of Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Xiong Gao
- Neonatal Diagnosis and Treatment Centre of Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Zhen-Zhen Liu
- Neonatal Diagnosis and Treatment Centre of Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Zheng-Li Wang
- Neonatal Diagnosis and Treatment Centre of Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Lu-Quan Li
- Neonatal Diagnosis and Treatment Centre of Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China
- Jiangxi Hospital Affiliated Children’s Hospital of Chongqing Medical University, Nanchang, China
- *Correspondence: Lu-Quan Li,
| |
Collapse
|
13
|
Das RP, Singh BG, Aishwarya J, Kumbhare LB, Kunwar A. 3,3'-Diselenodipropionic acid immobilised gelatin gel: a biomimic catalytic nitric oxide generating material for topical wound healing application. Biomater Sci 2023; 11:1437-1450. [PMID: 36602012 DOI: 10.1039/d2bm01964g] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Nitric oxide (NO) plays a pivotal role in the wound healing process and promotes the generation of healthy endothelium. In this work, a simple method has been developed for fabricating a diselenide grafted gelatin gel, which reduces NO donors such as S-nitroso-N-acetylpenicillamine (SNAP) by glutathione peroxidase-like mechanism to produce NO. Briefly, the process involved covalently conjugating 3,3'-diselenodipropionic acid (DSePA) with gelatin via carbodiimide coupling. The resulting gelatin-DSePA conjugate (G-Se-Se-G) demonstrated NO production upon incubation with SNAP and glutathione (GSH) with the flux of 4.8 ± 0.6 nmol cm-2 min-1 and 1.6 ± 0.1 nmol cm-2 min-1 at 10 min and 40 min, respectively. The G-Se-Se-G recovered even after 5 days of incubation with the reaction mixture retaining catalytic activity up to 74%. Subsequently, G-Se-Se-G was suspended (5% w/v) in water with lecithin (6% w/w of gelatin) and F127 (3% w/w of gelatin) to prepare gel through temperature dependant gelation method. The fabricated G-Se-Se-G gel exhibited desirable rheological characteristics and excellent mechanical stability under storage conditions and did not cause any significant toxicity in normal human keratinocytes (HaCaT) and fibroblast cells (WI38) up to 50 μg ml-1 of selenium equivalent. Finally, mice studies confirmed that topically applied G-Se-Se-G gel and SNAP promoted faster epithelization and collagen deposition at the wound site. In conclusion, the development of a biomimetic NO generating gel with sustained activity and biocompatibility was achieved.
Collapse
Affiliation(s)
- Ram P Das
- Radiation & Photochemistry Division, Bhabha Atomic Research Centre, Trombay, Mumbai-400085, India. .,Homi Bhabha National Institute, Anushaktinagar, Mumbai-400094, India
| | - Beena G Singh
- Radiation & Photochemistry Division, Bhabha Atomic Research Centre, Trombay, Mumbai-400085, India. .,Homi Bhabha National Institute, Anushaktinagar, Mumbai-400094, India
| | - J Aishwarya
- Radiation & Photochemistry Division, Bhabha Atomic Research Centre, Trombay, Mumbai-400085, India. .,Homi Bhabha National Institute, Anushaktinagar, Mumbai-400094, India.,Advanced Centre for Treatment, Research and Education in Cancer, Mumbai-410210, India
| | - Liladhar B Kumbhare
- Chemistry Division, Bhabha Atomic Research Centre, Trombay, Mumbai-400085, India
| | - Amit Kunwar
- Radiation & Photochemistry Division, Bhabha Atomic Research Centre, Trombay, Mumbai-400085, India. .,Homi Bhabha National Institute, Anushaktinagar, Mumbai-400094, India
| |
Collapse
|
14
|
Yang P, Deng F, Yuan M, Chen M, Zeng L, Ouyang Y, Chen X, Zhao B, Yang Z, Tian Z. Metabolomics reveals the defense mechanism of histidine supplementation on high-salt exposure-induced hepatic oxidative stress. Life Sci 2023; 314:121355. [PMID: 36596407 DOI: 10.1016/j.lfs.2022.121355] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 12/21/2022] [Accepted: 12/28/2022] [Indexed: 01/02/2023]
Abstract
AIMS This study mainly evaluated the protective mechanism of histidine against the hepatic oxidative stress after high-salt exposure (HSE) through combined analysis of non-targeted metabolomics and biological metabolic networks. MATERIALS AND METHODS Dahl salt-sensitive (SS) rats were fed with normal-salt diet or HSE ± histidine in addition to drinking water for 14 days. Gas chromatography-mass spectrometry was used to analyze the hepatic metabolites. The metabolic profile was analyzed by SIMCA-14.1, the metabolic correlation network was performed using Gephi-0.9.2, and pathway enrichment was analyzed using MetaboAnalyst 5.0 online website. KEY FINDINGS Results indicated that HSE disturbed the hepatic metabolic profile, generated abnormal liver metabolism and exacerbated oxidative stress. Histidine supplementation significantly reversed the hepatic metabolic profile. Of note, 14 differential metabolic pathways were enriched after histidine supplementation, most of which played an important role in ameliorating redox and nitric oxide (NO) metabolism. Histidine administration decreased the levels of hydroperoxide and malondialdehyde, and increased the activities of antioxidant enzymes (Catalase, Superoxide Dismutase, Glutathione S-transferase and Glutathione reductases). Histidine effectively enhanced the endogenous synthesis of glutathione by increasing the levels of glutamate and cysteine, thereby enhancing the antioxidant capacity of the glutathione system. After histidine administration, lysine, glutamate, and hypotaurine owned a higher metabolic centrality in the correlation network. In addition, histidine could also effectively increase the endogenous synthesis of NO by enhancing the L-arginine/NO pathway. SIGNIFICANCE This study offers new insights into the metabolic mechanisms underlying the antioxidant protective effect of histidine on the liver.
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
| | - Fenfen Deng
- 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
| | - Mengdi 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
| | - 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
| | - 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
| | - Bin Zhao
- 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
|
15
|
Edosuyi O, Adesuyi A, Choi M, Igbe I, Oyekan A. Malate reduced blood pressure and exerted differential effects on renal hemodynamics; role of the nitric oxide system and renal epithelial sodium channels (E NaC). Eur J Pharmacol 2023; 938:175441. [PMID: 36463945 PMCID: PMC9772084 DOI: 10.1016/j.ejphar.2022.175441] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 11/10/2022] [Accepted: 11/29/2022] [Indexed: 12/05/2022]
Abstract
Malate regulates blood pressure via nitric oxide production in salt-sensitive rats, a genetic model of hypertension. This study investigated the possible contributions of malate to blood pressure regulation and renal haemodynamics in normotensive rats. Malate (0.1, 0.3 and 1 μg/kg, iv) was injected into rats or L-nitro-arginine methyl ester (L-NAME)-treated rats and mean arterial blood pressure (MABP), cortical blood flow (CBF), and medullary blood flow (MBF), was measured. The clearance study involved infusion of malate at 0.1 μg/kg/h into rats, and MABP, CBF, MBF, glomerular filtration rate (GFR), urine volume (UV) and sodium output (UNaV) were determined. Mechanistic studies to evaluate the role of renal sodium channels involved the treatment with malate (600 mg/kg, po), amiloride (2.5 mg/kg, po) or hydrochlorothiazide (HCTZ) (10 mg/kg, po), and UV and UNaV were determined. Malate elicited significant peak reductions in MABP (124 ± 6.5 vs 105 ± 3.1 mmHg) at 0.1 μg/kg), CBF (231 ± 18.5 vs 205 ± 10.9 PU). L-NAME did not reverse the effect of malate on MABP but tended to blunt the effect on CBF (40%) and MBF (87%) at 0.3 μg/kg. Infusion of malate reduced MABP, CBF, and MBF in a time-dependent manner (p<0.05). Malate exerted a three-fold decrease in GFR in a time-related fashion (p<0.05) as well as increased UV. UNaV increased by 86% in malate-treated-amiloride rats (p<0.05). These data indicate that malate modulates blood pressure and exerts vascular and tubular effects on renal function that may involve epithelial sodium channels (ENaC).
Collapse
Affiliation(s)
- Osaze Edosuyi
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, University of Benin, PMB 1154, Benin City, Nigeria; Center for Cardiovascular Diseases, Gray Hall Suites, Rm 256, College of Pharmacy & Health Sciences, Texas Southern University, 3100, Cleburne Street, Houston, TX, USA.
| | - Ayobami Adesuyi
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, University of Benin, PMB 1154, Benin City, Nigeria; Community Medicine Department, Babcock University Teaching Hospital, IIishan-Remo, Ogun state, Nigeria
| | - Myung Choi
- Center for Cardiovascular Diseases, Gray Hall Suites, Rm 256, College of Pharmacy & Health Sciences, Texas Southern University, 3100, Cleburne Street, Houston, TX, USA
| | - Ighodaro Igbe
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, University of Benin, PMB 1154, Benin City, Nigeria
| | - Adebayo Oyekan
- Center for Cardiovascular Diseases, Gray Hall Suites, Rm 256, College of Pharmacy & Health Sciences, Texas Southern University, 3100, Cleburne Street, Houston, TX, USA
| |
Collapse
|
16
|
Vigers T, Vinovskis C, Li LP, Prasad P, Heerspink H, D'Alessandro A, Reisz JA, Piani F, Cherney DZ, van Raalte DH, Nadeau KJ, Pavkov ME, Nelson RG, Pyle L, Bjornstad P. Plasma levels of carboxylic acids are markers of early kidney dysfunction in young people with type 1 diabetes. Pediatr Nephrol 2023; 38:193-202. [PMID: 35507146 PMCID: PMC10182875 DOI: 10.1007/s00467-022-05531-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 02/25/2022] [Accepted: 03/07/2022] [Indexed: 01/10/2023]
Abstract
BACKGROUND We compared plasma metabolites of amino acid oxidation and the tricarboxylic acid (TCA) cycle in youth with and without type 1 diabetes mellitus (T1DM) and related the metabolites to glomerular filtration rate (GFR), renal plasma flow (RPF), and albuminuria. Metabolites associated with impaired kidney function may warrant future study as potential biomarkers or even future interventions to improve kidney bioenergetics. METHODS Metabolomic profiling of fasting plasma samples using a targeted panel of 644 metabolites and an untargeted panel of 19,777 metabolites was performed in 50 youth with T1DM ≤ 10 years and 20 controls. GFR and RPF were ascertained by iohexol and p-aminohippurate clearance, and albuminuria calculated as urine albumin to creatinine ratio. Sparse partial least squares discriminant analysis and moderated t tests were used to identify metabolites associated with GFR and RPF. RESULTS Adolescents with and without T1DM were similar in age (16.1 ± 3.0 vs. 16.1 ± 2.9 years) and BMI (23.4 ± 5.1 vs. 22.7 ± 3.7 kg/m2), but those with T1DM had higher GFR (189 ± 40 vs. 136 ± 22 ml/min) and RPF (820 ± 125 vs. 615 ± 65 ml/min). Metabolites of amino acid oxidation and the TCA cycle were significantly lower in adolescents with T1DM vs. controls, and the measured metabolites were able to discriminate diabetes status with an AUC of 0.82 (95% CI: 0.71, 0.93) and error rate of 0.21. Lower glycine (r:-0.33, q = 0.01), histidine (r:-0.45, q < 0.001), methionine (r: -0.29, q = 0.02), phenylalanine (r: -0.29, q = 0.01), serine (r: -0.42, q < 0.001), threonine (r: -0.28, q = 0.02), citrate (r: -0.35, q = 0.003), fumarate (r: -0.24, q = 0.04), and malate (r: -0.29, q = 0.02) correlated with higher GFR. Lower glycine (r: -0.28, q = 0.04), phenylalanine (r:-0.3, q = 0.03), fumarate (r: -0.29, q = 0.04), and malate (r: -0.5, q < 0.001) correlated with higher RPF. Lower histidine (r: -0.28, q = 0.02) was correlated with higher mean ACR. CONCLUSIONS In conclusion, adolescents with relatively short T1DM duration exhibited lower plasma levels of carboxylic acids that associated with hyperfiltration and hyperperfusion. TRIAL REGISTRATION ClinicalTrials.gov NCT03618420 and NCT03584217 A higher resolution version of the Graphical abstract is available as Supplementary information.
Collapse
Affiliation(s)
- Timothy Vigers
- Department of Pediatrics, Section of Endocrinology, University of Colorado School of Medicine, Aurora, CO, USA.
- Department of Biostatistics and Informatics, Colorado School of Public Health, 13123 E 16th Ave, A036-B265, Aurora, CO, 80045, USA.
| | - Carissa Vinovskis
- Department of Pediatrics, Section of Endocrinology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Lu-Ping Li
- Department of Radiology, NorthShore University HealthSystem, Evanston, IL, USA
| | - Pottumarthi Prasad
- Department of Radiology, NorthShore University HealthSystem, Evanston, IL, USA
| | - Hiddo Heerspink
- Department Clinical Pharmacy and Pharmacology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Angelo D'Alessandro
- Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora, CO, USA
| | - Julie A Reisz
- Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora, CO, USA
| | - Federica Piani
- Department of Pediatrics, Section of Endocrinology, University of Colorado School of Medicine, Aurora, CO, USA
| | - David Z Cherney
- Department of Medicine, Division of Nephrology, University of Toronto School of Medicine, Toronto, Ontario, Canada
| | - Daniel H van Raalte
- Diabetes Center, Department of Internal Medicine, Amsterdam University Medical Centers, location VUmc, Amsterdam, the Netherlands
| | - Kristen J Nadeau
- Department of Pediatrics, Section of Endocrinology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Meda E Pavkov
- Division of Diabetes Translation, Center for Disease Control and Prevention, Atlanta, GA, USA
| | - Robert G Nelson
- Chronic Kidney Disease Section, Phoenix Epidemiology and Clinical Research Branch, NIDDK, Phoenix, AZ, USA
| | - Laura Pyle
- Department of Pediatrics, Section of Endocrinology, University of Colorado School of Medicine, Aurora, CO, USA
- Department of Biostatistics and Informatics, Colorado School of Public Health, 13123 E 16th Ave, A036-B265, Aurora, CO, 80045, USA
| | - Petter Bjornstad
- Department of Pediatrics, Section of Endocrinology, University of Colorado School of Medicine, Aurora, CO, USA
- Department of Medicine, Division of Nephrology, University of Colorado School of Medicine, Aurora, CO, USA
| |
Collapse
|
17
|
Wild Watermelon-Extracted Juice Ingestion Reduces Peripheral Arterial Stiffness with an Increase in Nitric Oxide Production: A Randomized Crossover Pilot Study. Nutrients 2022; 14:nu14245199. [PMID: 36558358 PMCID: PMC9780996 DOI: 10.3390/nu14245199] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 11/29/2022] [Accepted: 12/05/2022] [Indexed: 12/12/2022] Open
Abstract
Wild watermelon contains various nutrients, but the effect of its acute ingestion on arterial stiffness is unclear. This study aimed to investigate whether a single bout of acute ingestion of wild watermelon-extracted juice decreased arterial stiffness concomitant with an increase in nitric oxide (NO) production. Twelve healthy young female participants were tested under two conditions in a randomized, double-blind crossover study: (1) a beverage containing 90 g of wild watermelon extract and (2) a control beverage: a placebo. Pulse wave velocity (PWV), an index of arterial stiffness, blood flow, and plasma nitrate/nitrite (NOx) levels were measured in the supine position at 30, 60, and 90 min after the intake of each beverage. The changes in femoral-ankle PWV were significantly reduced after wild watermelon-extracted juice intake compared to those in the placebo group. Additionally, the changes in blood flow in the posterior tibial artery and plasma NOx levels after intake of wild watermelon-extracted juice were significantly increased compared to those in the placebo group. These data show that acute ingestion of wild watermelon-extracted juice reduces peripheral (lower limb) arterial stiffness and increases NO bioavailability. To confirm these associations, more detailed investigations of the nutrients that influence these effects should be conducted.
Collapse
|
18
|
Ahmad H, Zhao X, Ahmad N, Khan A, Jin Y, Du J, Zheng X, Zeng L, Ouyang Y, Yang P, Chen M, Li X, Yang Z, Tian Z. Benincasa hispida extracts positively regulated high salt-induced hypertension in Dahl salt-sensitive rats: Impact on biochemical profile and metabolic patterns. J Food Biochem 2022; 46:e14497. [PMID: 36314446 DOI: 10.1111/jfbc.14497] [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: 07/05/2022] [Revised: 10/12/2022] [Accepted: 10/20/2022] [Indexed: 12/29/2022]
Abstract
Salt-induced hypertension is one of the major issues worldwide and one of the main factors involved in heart and kidney failure. The objective of this study was to investigate the potential role of Benincasa hispida extracts on high salt-induced hypertension in Dahl-salt sensitive (D-SS) rats and to find out the metabolic and biochemical pattern involved in the reduction of hypertension. Twenty-six Dahl salt-sensitive (D-SS) rats were selected and divided into four groups. The metabolic strategy was applied to test the extracts on salt-sensitive hypertension in kidney. Gas Chromatography-Mass spectrometry (GC-MS) was used to identify the potent biochemical profile in renal medulla and cortex of rat kidneys. The differential metabolites of cortex and medulla, enrichment analysis and pathway analysis were performed using metabolomics data. The GC-MS data revealed that 24 different antihypertensive metabolites was detected in renal cortex, while 16 were detected in renal medulla between different groups. The significantly metabolic pathways namely citrate cycle, glutathione metabolism, glycine, serine, and threonine metabolism, glyoxylate and dicarboxylate metabolism, glycerolipid metabolism, alanine, aspartate and glutamate metabolism in renal cortex and glycerolipid metabolism, pentose phosphate pathway, citrate cycle, glycolysis, glycerophospholipid metabolism, phenylalanine, tyrosine and tryptophan biosynthesis in renal medulla were involved in the process of Hypertension. The results suggest that the extract mainly alter the metabolic pathways of amino acid in Dahl salt-sensitive rats and its antioxidant potential reduced the hypertension patterns of Salt-sensitive rat. The antihypertensive components malic acid, aspartic acid, and glycine of extract can be used as therapeutic drugs to protect kidneys from salt-induced hypertension. PRACTICAL APPLICATIONS: Hypertension is a multifactorial disease and one of the risk factors for heart and kidney failure. Benincasa hispida is a widely used vegetable in China, which belongs to the Cucurbitaceae family. Benincasa hispida (wax gourd) has been used in traditional Chinese medicine for the treatment of inflammation and hypertension. The Benincasa hispida contains many compounds such as amino acids, carbohydrates, volatile compounds, vitamins, and minerals. The amino acid present in the pulp of Benincasa hispida are ornithine, threonine, aspartate, glutamate, serine, glycine, proline, alanine, valine, cysteine, isoleucine, tyrosine, leucine, lysine, phenylalanine, histidine, arginine, and γ-aminobutyric acid. Our results showed that Benincasa hispida is one of the potent natural antioxidants and can maintain normal blood pressure in Dahl salt-sensitive rats (D-SS). In conclusion, the current results provide good theoretical basis for the development and research using Benincasa hispida as an effective natural antioxidant for hypertension.
Collapse
Affiliation(s)
- Hussain Ahmad
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Sciences and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Xinrui Zhao
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Sciences and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Nisar Ahmad
- Center for Biotechnology and Microbiology, University of Swat, Swat, Pakistan
| | - Abbas Khan
- Department of Bioinformatics and Biological Statistics, School of Life Sciences and Biotechnology, Shanghai Jiaotong University, Shanghai, China
| | - Yuexin Jin
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Sciences and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Jie Du
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Sciences and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Xuewei Zheng
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Sciences and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Li Zeng
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Sciences and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Yanan Ouyang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Sciences and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Pengfei Yang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Sciences and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Meng Chen
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Sciences and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Xiaoxue Li
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Sciences and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Zhe Yang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Sciences and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Zhongmin Tian
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Sciences and Technology, Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
19
|
Hou E, Yan J, Zhu X, Qiao J. High-salt diet contributes to excess oxidative stress and abnormal metabolism in mouse ovaries. Biomed Chromatogr 2022; 36:e5500. [PMID: 36068010 DOI: 10.1002/bmc.5500] [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: 05/20/2022] [Revised: 08/11/2022] [Accepted: 09/03/2022] [Indexed: 11/05/2022]
Abstract
High-salt diets (HSDs) are associated with elevated levels of reactive oxygen species (ROS), which play a key role in ovarian disorders. However, it is not yet clear whether HSDs impact ovarian redox balance and metabolism. Accordingly, in this study, we analyzed the effect of HSDs on ovarian redox balance by biochemical analysis and further dissected its possible mechanism by metabolic analysis combined with correlation network method. We found that ROS and H2 O2 levels were significantly increased in the ovarian tissue of mice receiving an HSD for 4 weeks. The enhanced activity of NADPH oxidase may contribute to an increase in ROS in ovarian tissue after excessive salt consumption. Meanwhile, the activities of key antioxidant enzymes, including superoxide dismutase 2, glutathione peroxidase, glucose-6-phosphate dehydrogenase, and 6-phosphogluconate dehydrogenase increased significantly. The apparent activation of antioxidant defense appeared insufficient as the glutathione, GSH/GSSG ratio, and NADPH/NADP+ ratio decreased significantly. In addition, HSDs significantly altered the metabolic patterns of ovarian tissue in mice, and pathways were mainly enriched in fatty acid metabolism. Arachidonic acid was an altered hub metabolite according to Pearson correlation network analysis. Mechanistically, an HSD increased the concentration of arachidonic acid in ovarian tissue, inducing high NADPH oxidase activity, which increased the levels of ROS and H2 O2 . Our results indicate that HSDs can lead to increased oxidative stress and dramatically alter the metabolic patterns in mouse ovarian tissues.
Collapse
Affiliation(s)
- Entai Hou
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China.,National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, China.,Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, China.,Beijing key Laboratory of Reproductive Endocrinology and Assisted Reproduction (Peking University Third Hospital), Beijing, China
| | - Jie Yan
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China.,National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, China.,Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, China.,Beijing key Laboratory of Reproductive Endocrinology and Assisted Reproduction (Peking University Third Hospital), Beijing, China
| | - Xiaohui Zhu
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China.,National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, China.,Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, China.,Beijing key Laboratory of Reproductive Endocrinology and Assisted Reproduction (Peking University Third Hospital), Beijing, China
| | - Jie Qiao
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China.,National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, China.,Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, China.,Beijing key Laboratory of Reproductive Endocrinology and Assisted Reproduction (Peking University Third Hospital), Beijing, China
| |
Collapse
|
20
|
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: 0.7] [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
|
21
|
Chatre L, Ducat A, Spradley FT, Palei AC, Chéreau C, Couderc B, Thomas KC, Wilson AR, Amaral LM, Gaillard I, Méhats C, Lagoutte I, Jacques S, Miralles F, Batteux F, Granger JP, Ricchetti M, Vaiman D. Increased NOS coupling by the metabolite tetrahydrobiopterin (BH4) reduces preeclampsia/IUGR consequences. Redox Biol 2022; 55:102406. [PMID: 35964341 PMCID: PMC9389306 DOI: 10.1016/j.redox.2022.102406] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 07/04/2022] [Accepted: 07/11/2022] [Indexed: 11/24/2022] Open
Abstract
Preeclampsia (PE) is a high-prevalence pregnancy disease characterized by placental insufficiency, gestational hypertension, and proteinuria. Overexpression of the A isoform of the STOX1 transcription factor (STOX1A) recapitulates PE in mice, and STOX1A overexpressing trophoblasts recapitulate PE patients hallmarks in terms of gene expression and pathophysiology. STOX1 overexpression induces nitroso-redox imbalance and mitochondrial hyper-activation. Here, by a thorough analysis on cell models, we show that STOX1 overexpression in trophoblasts alters inducible nitric oxide synthase (iNOS), nitric oxide (NO) content, the nitroso-redox balance, the antioxidant defense, and mitochondrial function. This is accompanied by specific alterations of the Krebs cycle leading to reduced l-malate content. By increasing NOS coupling using the metabolite tetrahydrobiopterin (BH4) we restore this multi-step pathway in vitro. Moving in vivo on two different rodent models (STOX1 mice and RUPP rats, alike early onset and late onset preeclampsia, respectively), we show by transcriptomics that BH4 directly reverts STOX1-deregulated gene expression including glutathione metabolism, oxidative phosphorylation, cholesterol metabolism, inflammation, lipoprotein metabolism and platelet activation, successfully treating placental hypotrophy, gestational hypertension, proteinuria and heart hypertrophy. In the RUPP rats we show that the major fetal issue of preeclampsia, Intra Uterine Growth Restriction (IUGR), is efficiently corrected. Our work posits on solid bases BH4 as a novel potential therapy for preeclampsia.
Collapse
Affiliation(s)
- Laurent Chatre
- Institut Pasteur, Department of Developmental & Stem Cell Biology, Stem Cell & Development, 25-28 Rue du Dr. Roux, Paris, France; UMR 3738 CNRS, 25 Rue du Dr. Roux, Paris, 75015, France
| | - Aurélien Ducat
- Institut Cochin U1016, INSERM UMR8104 CNRS, 24, rue du Fg St Jacques, Paris, France
| | - Frank T Spradley
- Department of Surgery, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA
| | - Ana C Palei
- Department of Surgery, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA
| | - Christiane Chéreau
- Institut Cochin U1016, INSERM UMR8104 CNRS, 24, rue du Fg St Jacques, Paris, France
| | - Betty Couderc
- Institut Cochin U1016, INSERM UMR8104 CNRS, 24, rue du Fg St Jacques, Paris, France
| | - Kamryn C Thomas
- Department of Surgery, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA
| | - Anna R Wilson
- Department of Surgery, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA
| | - Lorena M Amaral
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA
| | - Irène Gaillard
- Institut Cochin U1016, INSERM UMR8104 CNRS, 24, rue du Fg St Jacques, Paris, France
| | - Céline Méhats
- Institut Cochin U1016, INSERM UMR8104 CNRS, 24, rue du Fg St Jacques, Paris, France
| | - Isabelle Lagoutte
- Institut Cochin U1016, INSERM UMR8104 CNRS, 24, rue du Fg St Jacques, Paris, France
| | - Sébastien Jacques
- Institut Cochin U1016, INSERM UMR8104 CNRS, 24, rue du Fg St Jacques, Paris, France
| | - Francisco Miralles
- Institut Cochin U1016, INSERM UMR8104 CNRS, 24, rue du Fg St Jacques, Paris, France
| | - Frédéric Batteux
- Institut Cochin U1016, INSERM UMR8104 CNRS, 24, rue du Fg St Jacques, Paris, France
| | - Joey P Granger
- Department of Physiology & Biophysics, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA
| | - Miria Ricchetti
- Institut Pasteur, Department of Developmental & Stem Cell Biology, Stem Cell & Development, 25-28 Rue du Dr. Roux, Paris, France; UMR 3738 CNRS, 25 Rue du Dr. Roux, Paris, 75015, France; Institut Pasteur, Molecular Mechanisms of Pathological and Physiological Ageing, 25-28 Rue du Dr. Roux, Paris, France
| | - Daniel Vaiman
- Institut Cochin U1016, INSERM UMR8104 CNRS, 24, rue du Fg St Jacques, Paris, France.
| |
Collapse
|
22
|
Edosuyi O, Choi M, Igbe I, Oyekan A. Effects of fumarate on renal vascular reactivity and the modulation of blood pressure in normotensive rats: Possible contribution of the nitric oxide synthase-nitric oxide system. RESEARCH RESULTS IN PHARMACOLOGY 2022. [DOI: 10.3897/rrpharmacology.8.79765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Introduction: Fumarate, the tricarboxylic acid (TCA) cycle intermediary, has been linked to nitric oxide (NO) production. NO plays a prominent role in the physiological regulation of blood pressure and renal hemodynamics. This study is aimed to investigate any contribution of fumarate to blood pressure and renal hemodynamics in normotensive rats with a possible link to the nitrergic system.
Materials and methods: Fumarate (1, 3 and 10 µmol) was injected into isolated perfused kidneys, pre-constricted with epinephrine (30 µM). The fumarase inhibitor, pyromellitic acid (PMA) (1, 3 and 10 µM), was used to perfuse the isolated kidney and perfusate was collected for nitric oxide and fumarate assays. An acute blood pressure study involved the injection of bolus doses of fumarate (0.1, 0.3 and 1 µg/kg, iv) or PMA (1, 3 and 10 µg/kg, iv) to normotensive rats in the presence of N(ω)-nitro-L-arginine methyl ester (L-NAME) (10 mg/kg, iv) or PMA (1, 3 and 10 µg/kg).
Results and discussion: Fumarate reduced perfusion pressure and elicited a peak reduction at the highest dose. Perfusing the kidney with PMA caused a paradoxical increase in perfusion pressure (70%, p<0.05), compared to baseline. Bolus doses of fumarate reduced blood pressure (-29.3±6.2 mmHg, p<0.05), cortical blood flow (CBF) and increased medullary blood flow (MBF). L-NAME did not abolish the vasodilatory effect of fumarate, but reduced the magnitude of response (50%, p<0.05). PMA did not significantly affect the vasodilatory effect of fumarate (p>0.05).
Conclusion: These data suggest that fumarate exerts a vasodilatory effect on renal and systemic hemodynamics that may partly involve the nitric oxide signaling.
Graphical abstract:
Collapse
|
23
|
Gu X, Li S, Lu M, Li Y, Wang Q, Chen L, Jia Y, Cao S, Zhang T, Zhou M, Gou X. Investigation of Gynura segetum root extract (GSrE) induced hepatotoxicity based on metabolomic signatures and microbial community profiling in rats. Front Microbiol 2022; 13:947757. [PMID: 36016788 PMCID: PMC9396145 DOI: 10.3389/fmicb.2022.947757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 07/14/2022] [Indexed: 11/26/2022] Open
Abstract
In recent years, many reports focus on the hepatotoxicity of Gynura segetum root extract (GSrE), but the interaction between GSrE and the gut microbiota is still unclear. This study investigated the mechanism of GSrE-induced hepatotoxicity of different doses and exposure durations by combining metabolomics and gut microbiota analysis. SD rats were divided into 3 groups: blank, low-dose (7.5 g/kg), and high-dose (15 g/kg) groups. Urine and feces samples were collected on day 0, day 10, and day 21. Metabolomics based on gas chromatography-mass spectrometry (GC-MS) was carried out to identify metabolites and metabolic pathways. 16S rDNA gene sequencing was applied to investigate the composition of gut microbiota before and after GSrE-induced hepatotoxicity. Finally, a correlation analysis of metabolites and gut microbiota was performed. Differential metabolites in urine and feces involved amino acids, carbohydrates, lipids, organic acids, and short chain fatty acids. Among them, L-valine, L-proline, DL-arabinose, pentanoic acid, D-allose, and D-glucose in urine and D-lactic acid and glycerol in fecal metabolites depended on the exposure of time and dose. In addition, 16S rDNA sequencing analysis revealed that GSrE-induced hepatotoxicity significantly altered the composition of gut microbiota, namely, f_Muribaculaceae_Unclassified, Lactobacillus, Bacteroides, Lachnospiraceae_NK4A136_group, f_Ruminococcaceae_Unclassified, Prevotellaceae_Ga6A1_group, and Escherichia-Shigella. The correlation analysis between gut microbiota and differential metabolites showed the crosstalk between the gut microbiota and metabolism in host involving energy, lipid, and amino acid metabolisms. In summary, our findings revealed that peripheral metabolism and gut microbiota disorders were time- and dose-related and the correlation between gut microbiota and metabolites in GSrE-induced hepatotoxicity.
Collapse
Affiliation(s)
- Xinyi Gu
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Shuwei Li
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Mengna Lu
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ying Li
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Qixue Wang
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Long Chen
- Experiment Center of Science and Technology, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yiqun Jia
- Experiment Center of Science and Technology, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Shan Cao
- Central Laboratory, Baoshan District Hospital of Integrated Traditional Chinese and Western Medicine of Shanghai, Shanghai, China
| | - Ting Zhang
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Mingmei Zhou
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- *Correspondence: Mingmei Zhou
| | - Xiaojun Gou
- Central Laboratory, Baoshan District Hospital of Integrated Traditional Chinese and Western Medicine of Shanghai, Shanghai, China
- Xiaojun Gou
| |
Collapse
|
24
|
Cardiac disruption of SDHAF4-mediated mitochondrial complex II assembly promotes dilated cardiomyopathy. Nat Commun 2022; 13:3947. [PMID: 35803927 PMCID: PMC9270418 DOI: 10.1038/s41467-022-31548-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 06/15/2022] [Indexed: 12/30/2022] Open
Abstract
Succinate dehydrogenase, which is known as mitochondrial complex II, has proven to be a fascinating machinery, attracting renewed and increased interest in its involvement in human diseases. Herein, we find that succinate dehydrogenase assembly factor 4 (SDHAF4) is downregulated in cardiac muscle in response to pathological stresses and in diseased hearts from human patients. Cardiac loss of Sdhaf4 suppresses complex II assembly and results in subunit degradation and complex II deficiency in fetal mice. These defects are exacerbated in young adults with globally impaired metabolic capacity and activation of dynamin-related protein 1, which induces excess mitochondrial fission and mitophagy, thereby causing progressive dilated cardiomyopathy and lethal heart failure in animals. Targeting mitochondria via supplementation with fumarate or inhibiting mitochondrial fission improves mitochondrial dynamics, partially restores cardiac function and prolongs the lifespan of mutant mice. Moreover, the addition of fumarate is found to dramatically improve cardiac function in myocardial infarction mice. These findings reveal a vital role for complex II assembly in the development of dilated cardiomyopathy and provide additional insights into therapeutic interventions for heart diseases. Functional succinate dehydrogenase (SDH) complex is vital to mitochondrial homeostasis. Here the authors show that disruption of SDH assembly in the heart causes dilated cardiomyopathy via impairing the mitochondrial integrity and metabolism and that mitochondrial interventions can be an effective approach to ameliorate the disease progression.
Collapse
|
25
|
Janus hydrogel with dual antibacterial and angiogenesis functions for enhanced diabetic wound healing. CHINESE CHEM LETT 2022. [DOI: 10.1016/j.cclet.2022.07.048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
26
|
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: 0.3] [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
|
27
|
Amaral AG, Moretto IA, Zandonadi FDS, Zamora-Obando HR, Rocha I, Sussulini A, Thomaz AAD, Oliveira RV, Santos AMD, Simionato AVC. Comprehending Cardiac Dysfunction by Oxidative Stress: Untargeted Metabolomics of In Vitro Samples. Front Chem 2022; 10:836478. [PMID: 35464220 PMCID: PMC9023746 DOI: 10.3389/fchem.2022.836478] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 02/09/2022] [Indexed: 12/16/2022] Open
Abstract
Cardiovascular diseases (CVDs) are noncommunicable diseases known for their complex etiology and high mortality rate. Oxidative stress (OS), a condition in which the release of free radical exceeds endogenous antioxidant capacity, is pivotal in CVC, such as myocardial infarction, ischemia/reperfusion, and heart failure. Due to the lack of information about the implications of OS on cardiovascular conditions, several methodologies have been applied to investigate the causes and consequences, and to find new ways of diagnosis and treatment as well. In the present study, cardiac dysfunction was evaluated by analyzing cells’ alterations with untargeted metabolomics, after simulation of an oxidative stress condition using hydrogen peroxide (H2O2) in H9c2 myocytes. Optimizations of H2O2 concentration, cell exposure, and cell recovery times were performed through MTT assays. Intracellular metabolites were analyzed right after the oxidative stress (oxidative stress group) and after 48 h of cell recovery (recovery group) by ultra-high-performance liquid chromatography coupled to mass spectrometry (UHPLC-MS) in positive and negative ESI ionization mode. Significant alterations were found in pathways such as “alanine, aspartate and glutamate metabolism”, “glycolysis”, and “glutathione metabolism”, mostly with increased metabolites (upregulated). Furthermore, our results indicated that the LC-MS method is effective for studying metabolism in cardiomyocytes and generated excellent fit (R2Y > 0.987) and predictability (Q2 > 0.84) values.
Collapse
|
28
|
Wang X, Lv W, Xu J, Zheng A, Zeng M, Cao K, Wang X, Cui Y, Li H, Yang M, Shao Y, Zhang F, Zou X, Long J, Feng Z, Liu J. Hepatic Suppression of Mitochondrial Complex II Assembly Drives Systemic Metabolic Benefits. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2105587. [PMID: 35037426 PMCID: PMC8948583 DOI: 10.1002/advs.202105587] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Indexed: 05/05/2023]
Abstract
Alternate day fasting (ADF), the most popular form of caloric restriction, has shown to improve metabolic health in preclinical subjects, while intrinsic network underpinning the process remains unclear. Here, it is found that liver undergoes dramatic metabolic reprogramming during ADF, accompanied surprisingly with unique complex II dysfunction attributing to suspended complex II assembly via suppressing SDHAF4, a recently identified assembly factor. Despite moderate mitochondrial complex II dysfunction, hepatic Sdhaf4 knockout mice present intriguingly improved glucose tolerance and systemic insulin sensitivity, consistent with mice after ADF intervention. Mechanistically, it is found that hepatocytes activate arginine-nitric oxide (NO) biosynthesis axle in response to complex II and citric acid cycle dysfunction, the release of NO from liver can target muscle and adipocytes in addition to its autocrine action for enhanced insulin sensitivity. These results highlight the pivotal role of liver in ADF-associated systemic benefits, and suggest that targeting hepatic complex II assembly can be an intriguing strategy against metabolic disorders.
Collapse
Affiliation(s)
- Xueqiang Wang
- Center for Mitochondrial Biology and MedicineThe Key Laboratory of Biomedical Information Engineering of Ministry of EducationSchool of Life Science and TechnologyXi'an Jiaotong UniversityXi'anShaanxi710049China
| | - Weiqiang Lv
- Center for Mitochondrial Biology and MedicineThe Key Laboratory of Biomedical Information Engineering of Ministry of EducationSchool of Life Science and TechnologyXi'an Jiaotong UniversityXi'anShaanxi710049China
| | - Jie Xu
- Center for Mitochondrial Biology and MedicineThe Key Laboratory of Biomedical Information Engineering of Ministry of EducationSchool of Life Science and TechnologyXi'an Jiaotong UniversityXi'anShaanxi710049China
| | - Adi Zheng
- Center for Mitochondrial Biology and MedicineThe Key Laboratory of Biomedical Information Engineering of Ministry of EducationSchool of Life Science and TechnologyXi'an Jiaotong UniversityXi'anShaanxi710049China
| | - Mengqi Zeng
- Center for Mitochondrial Biology and MedicineThe Key Laboratory of Biomedical Information Engineering of Ministry of EducationSchool of Life Science and TechnologyXi'an Jiaotong UniversityXi'anShaanxi710049China
| | - Ke Cao
- Center for Mitochondrial Biology and MedicineThe Key Laboratory of Biomedical Information Engineering of Ministry of EducationSchool of Life Science and TechnologyXi'an Jiaotong UniversityXi'anShaanxi710049China
| | - Xun Wang
- Center for Mitochondrial Biology and MedicineThe Key Laboratory of Biomedical Information Engineering of Ministry of EducationSchool of Life Science and TechnologyXi'an Jiaotong UniversityXi'anShaanxi710049China
| | - Yuting Cui
- Center for Mitochondrial Biology and MedicineThe Key Laboratory of Biomedical Information Engineering of Ministry of EducationSchool of Life Science and TechnologyXi'an Jiaotong UniversityXi'anShaanxi710049China
| | - Hao Li
- Center for Mitochondrial Biology and MedicineThe Key Laboratory of Biomedical Information Engineering of Ministry of EducationSchool of Life Science and TechnologyXi'an Jiaotong UniversityXi'anShaanxi710049China
| | - Meng Yang
- Center for Mitochondrial Biology and MedicineThe Key Laboratory of Biomedical Information Engineering of Ministry of EducationSchool of Life Science and TechnologyXi'an Jiaotong UniversityXi'anShaanxi710049China
| | - Yongping Shao
- Frontier Institute of Science and TechnologyXi'an Jiaotong UniversityXi'anShaanxi710049China
| | - Fang Zhang
- Department of OphthalmologyShanghai General HospitalShanghai Jiao Tong University School of MedicineXi'anShanghai200240China
- National Clinical Research Center for Eye DiseasesShanghai200240China
| | - Xuan Zou
- National & Local Joint Engineering Research Center of Biodiagnosis and BiotherapyThe Second Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShannxi710004China
- Shaanxi Provincial Clinical Research Center for Hepatic & Splenic DiseasesThe Second Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShannxi710004China
| | - Jiangang Long
- Center for Mitochondrial Biology and MedicineThe Key Laboratory of Biomedical Information Engineering of Ministry of EducationSchool of Life Science and TechnologyXi'an Jiaotong UniversityXi'anShaanxi710049China
| | - Zhihui Feng
- Frontier Institute of Science and TechnologyXi'an Jiaotong UniversityXi'anShaanxi710049China
- National & Local Joint Engineering Research Center of Biodiagnosis and BiotherapyThe Second Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShannxi710004China
- University of Health and Rehabilitation SciencesQingdaoShandong266071China
| | - Jiankang Liu
- Center for Mitochondrial Biology and MedicineThe Key Laboratory of Biomedical Information Engineering of Ministry of EducationSchool of Life Science and TechnologyXi'an Jiaotong UniversityXi'anShaanxi710049China
- University of Health and Rehabilitation SciencesQingdaoShandong266071China
| |
Collapse
|
29
|
Zhang X, Chen M, Yan E, Wang Y, Ma C, Zhang P, Yin J. Dietary Malic Acid Supplementation Induces Skeletal Muscle Fiber-Type Transition of Weaned Piglets and Further Improves Meat Quality of Finishing Pigs. Front Nutr 2022; 8:825495. [PMID: 35145985 PMCID: PMC8821922 DOI: 10.3389/fnut.2021.825495] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 12/27/2021] [Indexed: 12/21/2022] Open
Abstract
The aim of this study was to investigate effects of dietary malic acid supplementation on skeletal muscle fiber-type transition during nursery period and the subsequent meat quality of finishing pigs. Results showed that malic acid supplementation for 28 days increased oxidative fiber percentage of weaned piglets, accompanied by the increased aerobic oxidation in serum and longissimus thoracis (LT) muscle. Additionally, activities of total antioxidant capacity and glutathione peroxidase in serum were increased. Moreover, dietary malic acid supplementation during nursery period tended to increase pH24h and significantly decreased drip loss in LT muscle of finishing pigs. The content of total saturated fatty acid (SFA) and total monounsaturated fatty acid in LT muscle was significantly decreased, whereas the ratio of polyunsaturated fatty acid to SFA tended to increase. Together, dietary malic acid supplementation during nursery period can effectively increase antioxidant capacity and oxidative fibers percentage of weaned piglets, and further improve water holding capacity and nutritional values of pork in finishing pigs.
Collapse
Affiliation(s)
- Xin Zhang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Meixia Chen
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing, China
| | - Enfa Yan
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Yubo Wang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Chenghong Ma
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Pengguang Zhang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Jingdong Yin
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
- *Correspondence: Jingdong Yin
| |
Collapse
|
30
|
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: 0.3] [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
|
31
|
Huang CC, Huang YL, Lin CH, Chen JW. Plasma metabolomic profiles associated with hypertension and blood pressure in response to thiazide diuretics. Hypertens Res 2021; 45:464-473. [PMID: 34952949 DOI: 10.1038/s41440-021-00825-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 11/03/2021] [Accepted: 11/08/2021] [Indexed: 11/09/2022]
Abstract
This study aimed to identify the metabolomic alterations associated with hypertension (HTN) and the response of blood pressure (BP) to thiazide diuretics. A total of 50 participants previously untreated for HTN were prospectively recruited. After a 2-week lifestyle adjustment, 30 participants with systolic BP ≥ 140 mmHg and/or diastolic BP ≥ 90 mmHg were classified into the HTN group and prescribed hydrochlorothiazide (HCTZ) at 50 mg per day for 2 weeks. The remaining 20 participants, who had relatively normal BP, were assigned to the normotension group. Metabolomic profiles related to the response of BP to thiazide diuretics were analyzed. A total of 73 differential metabolites were found to be associated with HTN, and 27 metabolites were significantly changed upon HCTZ treatment (HCTZ-sensitive metabolites). Among the identified metabolites, 7 (aspartate, histidine, C5-DC, C5-M-DC, C14:1, phosphatidylcholine ae C34:1, and phosphatidylcholine ae C34:3) were positively associated with HTN and decreased in abundance upon HCTZ treatment (HCTZ-reduced/HTN-associated metabolites). Moreover, multivariate analysis of 20 metabolites whose baseline levels were associated with the response of BP revealed that aspartate, glutamate, lysophosphatidylcholine C16:0, lysophosphatidylcholine C20:3, and sphingomyelin C24:1 were independently related to systolic BP reduction, and lysophosphatidylcholine C20:3 was independently associated with diastolic BP reduction. In conclusion, we identified 5 metabolites independently related to BP changes with HCTZ treatment. An advanced biomarker profile of thiazide-induced metabolomic changes may provide a clue with which to further explore the complex and mixed effects of thiazide treatment in a clinical setting.
Collapse
Affiliation(s)
- Chin-Chou Huang
- Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan. .,School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan. .,Institute of Pharmacology, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan. .,Cardiovascular Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan.
| | - Yi-Long Huang
- Department of Life Sciences and Institute of Genome Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan.,Aging and Health Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Chao-Hsiung Lin
- Department of Life Sciences and Institute of Genome Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan. .,Aging and Health Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan.
| | - Jaw-Wen Chen
- Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan.,Institute of Pharmacology, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.,Cardiovascular Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan.,Healthcare and Services Center, Taipei Veterans General Hospital, Taipei, Taiwan
| |
Collapse
|
32
|
Zheng X, Zhou L, Jin Y, Zhao X, Ahmad H, OuYang Y, Chen S, Du J, Chen X, Chen L, Gao D, Yang Z, Tian Z. β-Aminoisobutyric acid supplementation attenuated salt-sensitive hypertension in Dahl salt-sensitive rats through prevention of insufficient fumarase. Amino Acids 2021; 54:169-180. [PMID: 34837556 DOI: 10.1007/s00726-021-03092-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 10/13/2021] [Indexed: 10/19/2022]
Abstract
The human Dietary Approaches to Stop Hypertension-Sodium Trial has shown that β-aminoisobutyric acid (BAIBA) may prevent the development of salt-sensitive hypertension (SSHT). However, the specific antihypertensive mechanism remains unclear in the renal tissues of salt-sensitive (SS) rats. In this study, BAIBA (100 mg/kg/day) significantly attenuated SSHT via increased nitric oxide (NO) content in the renal medulla, and it induced a significant increase in NO synthesis substrates (L-arginine and malic acid) in the renal medulla. BAIBA enhanced the activity levels of total NO synthase (NOS), inducible NOS, and constitutive NOS. BAIBA resulted in increased fumarase activity and decreased fumaric acid content in the renal medulla. The high-salt diet (HSD) decreased fumarase expression in the renal cortex, and BAIBA increased fumarase expression in the renal medulla and renal cortex. Furthermore, in the renal medulla, BAIBA increased the levels of ATP, ADP, AMP, and ADP/ATP ratio, thus further activating AMPK phosphorylation. BAIBA prevented the decrease in renal medullary antioxidative defenses induced by the HSD. In conclusion, BAIBA's antihypertensive effect was underlined by the phosphorylation of AMPK, the prevention of fumarase's activity reduction caused by the HSD, and the enhancement of NO content, which in concert attenuated SSHT in SS rats.
Collapse
Affiliation(s)
- 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
| | - 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
| | - Yuexin 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
| | - Xinrui Zhao
- 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
| | - 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
| | - Sa 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
| | - Jie Du
- 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
| | - Lan 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
| | - 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
| | - 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
|
33
|
Involvement of Tricarboxylic Acid Cycle Metabolites in Kidney Diseases. Biomolecules 2021; 11:biom11091259. [PMID: 34572472 PMCID: PMC8465464 DOI: 10.3390/biom11091259] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 08/17/2021] [Accepted: 08/23/2021] [Indexed: 02/08/2023] Open
Abstract
Mitochondria are complex organelles that orchestrate several functions in the cell. The primary function recognized is energy production; however, other functions involve the communication with the rest of the cell through reactive oxygen species (ROS), calcium influx, mitochondrial DNA (mtDNA), adenosine triphosphate (ATP) levels, cytochrome c release, and also through tricarboxylic acid (TCA) metabolites. Kidney function highly depends on mitochondria; hence mitochondrial dysfunction is associated with kidney diseases. In addition to oxidative phosphorylation impairment, other mitochondrial abnormalities have been described in kidney diseases, such as induction of mitophagy, intrinsic pathway of apoptosis, and releasing molecules to communicate to the rest of the cell. The TCA cycle is a metabolic pathway whose primary function is to generate electrons to feed the electron transport system (ETS) to drives energy production. However, TCA cycle metabolites can also release from mitochondria or produced in the cytosol to exert different functions and modify cell behavior. Here we review the involvement of some of the functions of TCA metabolites in kidney diseases.
Collapse
|
34
|
Mussap M, Fanos V. Could metabolomics drive the fate of COVID-19 pandemic? A narrative review on lights and shadows. Clin Chem Lab Med 2021; 59:1891-1905. [PMID: 34332518 DOI: 10.1515/cclm-2021-0414] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 07/19/2021] [Indexed: 02/07/2023]
Abstract
Human Severe Acute Respiratory Syndrome CoronaVirus 2 (SARS-CoV-2) infection activates a complex interaction host/virus, leading to the reprogramming of the host metabolism aimed at the energy supply for viral replication. Alterations of the host metabolic homeostasis strongly influence the immune response to SARS-CoV-2, forming the basis of a wide range of outcomes, from the asymptomatic infection to the onset of COVID-19 and up to life-threatening acute respiratory distress syndrome, vascular dysfunction, multiple organ failure, and death. Deciphering the molecular mechanisms associated with the individual susceptibility to SARS-CoV-2 infection calls for a system biology approach; this strategy can address multiple goals, including which patients will respond effectively to the therapeutic treatment. The power of metabolomics lies in the ability to recognize endogenous and exogenous metabolites within a biological sample, measuring their concentration, and identifying perturbations of biochemical pathways associated with qualitative and quantitative metabolic changes. Over the last year, a limited number of metabolomics- and lipidomics-based clinical studies in COVID-19 patients have been published and are discussed in this review. Remarkable alterations in the lipid and amino acid metabolism depict the molecular phenotype of subjects infected by SARS-CoV-2; notably, structural and functional data on the lipids-virus interaction may open new perspectives on targeted therapeutic interventions. Several limitations affect most metabolomics-based studies, slowing the routine application of metabolomics. However, moving metabolomics from bench to bedside cannot imply the mere determination of a given metabolite panel; rather, slotting metabolomics into clinical practice requires the conversion of metabolic patient-specific data into actionable clinical applications.
Collapse
Affiliation(s)
- Michele Mussap
- Laboratory Medicine, Department of Surgical Sciences, School of Medicine, University of Cagliari, Monserrato, Italy
| | - Vassilios Fanos
- Neonatal Intensive Care Unit, Department of Surgical Sciences, School of Medicine, University of Cagliari, Monserrato, Italy
| |
Collapse
|
35
|
Künstner A, Aherrahrou R, Hirose M, Bruse P, Ibrahim SM, Busch H, Erdmann J, Aherrahrou Z. Effect of Differences in the Microbiome of Cyp17a1-Deficient Mice on Atherosclerotic Background. Cells 2021; 10:1292. [PMID: 34070975 PMCID: PMC8224745 DOI: 10.3390/cells10061292] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 05/12/2021] [Accepted: 05/19/2021] [Indexed: 12/11/2022] Open
Abstract
CYP17A1 is a cytochrome P450 enzyme that has 17-alpha-hydroxylase and C17,20-lyase activities. Cyp17a11 deficiency is associated with high body mass and visceral fat deposition in atherosclerotic female ApoE knockout (KO, d/d or -/-) mice. In the present study, we aimed to investigate the effects of diet and Cyp17a1 genotype on the gut microbiome. Female Cyp17a1 (d/d) × ApoE (d/d) (DKO) and ApoE (d/d) (controls) were fed either standard chow or a Western-type diet (WTD), and we demonstrated the effects of genetics and diet on the body mass of the mice and composition of their gut microbiome. We found a significantly lower alpha diversity after accounting for the ecological network structure in DKO mice and WTD-fed mice compared with chow-fed ApoE(d/d). Furthermore, we found a strong significant positive association of the Firmicutes vs. Bacteroidota ratio with body mass and the circulating total cholesterol and triglyceride concentrations of the mice when feeding the WTD, independent of the Cyp17a1 genotype. Further pathway enrichment and network analyses revealed a substantial effect of Cyp17a1 genotype on associated cardiovascular and obesity-related pathways involving aspartate and L-arginine. Future studies are required to validate these findings and further investigate the role of aspartate/L-arginine pathways in the obesity and body fat distribution in our mouse model.
Collapse
Affiliation(s)
- Axel Künstner
- Medical Systems Biology Group, Lübeck Institute for Experimental Dermatology, University of Lübeck, 23562 Lübeck, Germany; (A.K.); (H.B.)
- Institute for Cardiogenetics, University of Lübeck, 23562 Lübeck, Germany; (R.A.); (P.B.); (J.E.)
| | - Redouane Aherrahrou
- Institute for Cardiogenetics, University of Lübeck, 23562 Lübeck, Germany; (R.A.); (P.B.); (J.E.)
- Centre for Public Health Genomics, Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908-0717, USA
| | - Misa Hirose
- Lübeck Institute for Experimental Dermatology, University of Lübeck, 23562 Lübeck, Germany; (M.H.); (S.M.I.)
| | - Petra Bruse
- Institute for Cardiogenetics, University of Lübeck, 23562 Lübeck, Germany; (R.A.); (P.B.); (J.E.)
| | - Saleh Mohamed Ibrahim
- Lübeck Institute for Experimental Dermatology, University of Lübeck, 23562 Lübeck, Germany; (M.H.); (S.M.I.)
- College of Medicine and Sharjah Institute for Medical Research, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Hauke Busch
- Medical Systems Biology Group, Lübeck Institute for Experimental Dermatology, University of Lübeck, 23562 Lübeck, Germany; (A.K.); (H.B.)
- Institute for Cardiogenetics, University of Lübeck, 23562 Lübeck, Germany; (R.A.); (P.B.); (J.E.)
| | - Jeanette Erdmann
- Institute for Cardiogenetics, University of Lübeck, 23562 Lübeck, Germany; (R.A.); (P.B.); (J.E.)
- DZHK (German Centre for Cardiovascular Research), University Heart Centre Lübeck, 23562 Lübeck, Germany
| | - Zouhair Aherrahrou
- Institute for Cardiogenetics, University of Lübeck, 23562 Lübeck, Germany; (R.A.); (P.B.); (J.E.)
- DZHK (German Centre for Cardiovascular Research), University Heart Centre Lübeck, 23562 Lübeck, Germany
| |
Collapse
|
36
|
Edosuyi O, Choi M, Igbe I, Oyekan A. Fumarate exerted an antihypertensive effect and reduced kidney injury molecule (KIM)-1 expression in deoxycorticosterone acetate-salt hypertension. Clin Exp Hypertens 2021; 43:555-564. [PMID: 33879002 DOI: 10.1080/10641963.2021.1916943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Background: The tricarboxylic (TCA) acid cycle provides the energy needed for regulatory functions in the cardio-renal system. Recently, a genetic defect in the TCA cycle enzyme, fumarase hydratase, altered L-arginine metabolism and exacerbated hypertension in salt-sensitive rats. This study evaluated the effect of fumarate and its possible link to L-arginine metabolism in deoxycorticosterone (DOCA)-salt hypertension, a non-genetic model of hypertension.Method: Hypertension was induced with DOCA (25 mg/kg s.c, twice weekly) + 1% NaCL in uninephrectomised rats placed on fumarate (1 g/L, ad libitum). Blood pressure was measured in conscious rats via carotid cannulation. Biochemical and western blot analyses were carried out on kidney fractions.Results: Fumarate reduced mean blood pressure (198 ± 5 vs 167 ± 7 mmHg, p < .01), increased nitric oxide levels in the renal cortex (36.1 ± 2 vs 61.3 ± 4 nM/µg) and medulla (27.4 ± 1 vs 54.1 ± 2 nM/µg) of DOCA-salt rats (p < .01). Consistent with this, arginase activity was reduced (threefold) in the renal medulla but not cortex of DOCA-salt rats. Fumarate increased superoxide dismutase activity in the medulla (p < .001) of DOCA-hypertensive rats. However, catalase activity was exacerbated by fumarate in both renal cortex (4.5 ± 1 vs 11.2 ± 1) and medulla (3.7 ± 1 vs 16.3 ± 1 units/mg) of DOCA-salt rats (p < .001). Proteinuria (64.6%), kidney injury molecule-1 expression and kidney weight were reduced in DOCA-hypertensive rats treated with fumarate (p< .05). However, there was a paradoxical increase in TGF-β expression in fumarate-treated DOCA-salt rats. Conclusion: These data show that fumarate attenuated hypertension, renal injury and improved the redox state of the kidney in DOCA/salt hypertension by mechanisms involving selective reduction of L-arginine metabolism.
Collapse
Affiliation(s)
- Osaze Edosuyi
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, University of Benin, Benin, Nigeria.,Center for Cardiovascular Diseases, Department of Pharmaceutical and Environmental Health Sciences, College of Pharmacy & Health Sciences, Texas Southern University, Houston, TX, USA
| | - Myung Choi
- Center for Cardiovascular Diseases, Department of Pharmaceutical and Environmental Health Sciences, College of Pharmacy & Health Sciences, Texas Southern University, Houston, TX, USA
| | - Ighodaro Igbe
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, University of Benin, Benin, Nigeria
| | - Adebayo Oyekan
- Center for Cardiovascular Diseases, Department of Pharmaceutical and Environmental Health Sciences, College of Pharmacy & Health Sciences, Texas Southern University, Houston, TX, USA
| |
Collapse
|
37
|
Wang Z, Cheng C, Yang X, Zhang C. L-phenylalanine attenuates high salt-induced hypertension in Dahl SS rats through activation of GCH1-BH4. PLoS One 2021; 16:e0250126. [PMID: 33857222 PMCID: PMC8049246 DOI: 10.1371/journal.pone.0250126] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 03/31/2021] [Indexed: 02/07/2023] Open
Abstract
Amino acid metabolism plays an important role in controlling blood pressure by regulating the production of NO and ROS. The present study examined amino acid levels in the serum of Dahl SS rats and SS.13BN rats fed a low or high salt diet. We observed that 8 of 27 amino acids responded to a high salt diet in SS rats. Thus, we hypothesized that a defect in amino acids may contribute to the development of salt-induced hypertension. L-phenylalanine was used to treat SS rats with a low or high salt diet. The results demonstrated that L-phenylalanine supplementation significantly enhanced the serum nitrite levels and attenuated the high salt-induced hypertension in SS rats. Low levels of BH4 and nitrite and the impaired vascular response to acetylcholine were rescued by L-phenylalanine supplementation. Moreover, increased GTP cyclohydrolase (GCH1) mRNA, levels of BH4 and nitrite, and reduced superoxide production were observed in the kidneys of hypertensive SS rats with L-phenylalanine. The antihypertensive effects of L-phenylalanine might be mediated by enhancing BH4 biosynthesis and decreasing superoxide production from NO synthase, thereby protecting vascular and kidney function with reduced ROS and elevated NO levels. The present study demonstrated that L-phenylalanine supplementation restored vascular function, suggesting L-phenylalanine represented a potential target to attenuate high salt-sensitive hypertension through GCH1-BH4.
Collapse
Affiliation(s)
- Zhengjun Wang
- School of Psychology, Shaanxi Normal University and Key Laboratory for Behavior and Cognitive Neuroscience of Shaanxi Province, Xi’an, China
- * E-mail: (ZW); (CZ)
| | - Chen Cheng
- School of Psychology, Shaanxi Normal University and Key Laboratory for Behavior and Cognitive Neuroscience of Shaanxi Province, Xi’an, China
| | - Xiaoyu Yang
- School of Life Science, Shaanxi Normal University, Xi’an, China
| | - Chen Zhang
- College of Animal Sciences, Zhejiang University, Hangzhou, China
- School of Life Sciences, Northwestern Polytechnical University, Xi’an, China
- * E-mail: (ZW); (CZ)
| |
Collapse
|
38
|
Moiz B, Garcia J, Basehore S, Sun A, Li A, Padmanabhan S, Albus K, Jang C, Sriram G, Clyne AM. 13C Metabolic Flux Analysis Indicates Endothelial Cells Attenuate Metabolic Perturbations by Modulating TCA Activity. Metabolites 2021; 11:metabo11040226. [PMID: 33917224 PMCID: PMC8068087 DOI: 10.3390/metabo11040226] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 03/30/2021] [Accepted: 04/01/2021] [Indexed: 11/16/2022] Open
Abstract
Disrupted endothelial metabolism is linked to endothelial dysfunction and cardiovascular disease. Targeted metabolic inhibitors are potential therapeutics; however, their systemic impact on endothelial metabolism remains unknown. In this study, we combined stable isotope labeling with 13C metabolic flux analysis (13C MFA) to determine how targeted inhibition of the polyol (fidarestat), pentose phosphate (DHEA), and hexosamine biosynthetic (azaserine) pathways alters endothelial metabolism. Glucose, glutamine, and a four-carbon input to the malate shuttle were important carbon sources in the baseline human umbilical vein endothelial cell (HUVEC) 13C MFA model. We observed two to three times higher glutamine uptake in fidarestat and azaserine-treated cells. Fidarestat and DHEA-treated HUVEC showed decreased 13C enrichment of glycolytic and TCA metabolites and amino acids. Azaserine-treated HUVEC primarily showed 13C enrichment differences in UDP-GlcNAc. 13C MFA estimated decreased pentose phosphate pathway flux and increased TCA activity with reversed malate shuttle direction in fidarestat and DHEA-treated HUVEC. In contrast, 13C MFA estimated increases in both pentose phosphate pathway and TCA activity in azaserine-treated cells. These data show the potential importance of endothelial malate shuttle activity and suggest that inhibiting glycolytic side branch pathways can change the metabolic network, highlighting the need to study systemic metabolic therapeutic effects.
Collapse
Affiliation(s)
- Bilal Moiz
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA; (B.M.); (A.S.); (A.L.); (S.P.); (K.A.)
| | - Jonathan Garcia
- School of Bioengineering, Science, and Heath Systems, Drexel University, Philadelphia, PA 19104, USA; (J.G.); (S.B.)
| | - Sarah Basehore
- School of Bioengineering, Science, and Heath Systems, Drexel University, Philadelphia, PA 19104, USA; (J.G.); (S.B.)
| | - Angela Sun
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA; (B.M.); (A.S.); (A.L.); (S.P.); (K.A.)
| | - Andrew Li
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA; (B.M.); (A.S.); (A.L.); (S.P.); (K.A.)
| | - Surya Padmanabhan
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA; (B.M.); (A.S.); (A.L.); (S.P.); (K.A.)
| | - Kaitlyn Albus
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA; (B.M.); (A.S.); (A.L.); (S.P.); (K.A.)
| | - Cholsoon Jang
- Department of Biological Chemistry, Chao Family Comprehensive Cancer Center, University of California Irvine, Irvine, CA 92697, USA;
| | - Ganesh Sriram
- Department of Chemical and Biomolecular Engineering, University of Maryland, College Park, MD 20742, USA;
| | - Alisa Morss Clyne
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA; (B.M.); (A.S.); (A.L.); (S.P.); (K.A.)
- Correspondence: ; Tel.: +1-301-405-9806
| |
Collapse
|
39
|
Basso PJ, Andrade-Oliveira V, Câmara NOS. Targeting immune cell metabolism in kidney diseases. Nat Rev Nephrol 2021; 17:465-480. [PMID: 33828286 DOI: 10.1038/s41581-021-00413-7] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/25/2021] [Indexed: 02/06/2023]
Abstract
Insights into the relationship between immunometabolism and inflammation have enabled the targeting of several immunity-mediated inflammatory processes that underlie infectious diseases and cancer or drive transplant rejection, but this field remains largely unexplored in kidney diseases. The kidneys comprise heterogeneous cell populations, contain distinct microenvironments such as areas of hypoxia and hypersalinity, and are responsible for a functional triad of filtration, reabsorption and secretion. These distinctive features create myriad potential metabolic therapeutic targets in the kidney. Immune cells have crucial roles in the maintenance of kidney homeostasis and in the response to kidney injury, and their function is intricately connected to their metabolic properties. Changes in nutrient availability and biomolecules, such as cytokines, growth factors and hormones, initiate cellular signalling events that involve energy-sensing molecules and other metabolism-related proteins to coordinate immune cell differentiation, activation and function. Disruption of homeostasis promptly triggers the metabolic reorganization of kidney immune and non-immune cells, which can promote inflammation and tissue damage. The metabolic differences between kidney and immune cells offer an opportunity to specifically target immunometabolism in the kidney.
Collapse
Affiliation(s)
- Paulo José Basso
- Laboratory of Immunobiology of Transplantation, Department of Immunology, Universidade de São Paulo, São Paulo, São Paulo, Brazil
| | | | - Niels Olsen Saraiva Câmara
- Laboratory of Immunobiology of Transplantation, Department of Immunology, Universidade de São Paulo, São Paulo, São Paulo, Brazil. .,Laboratory of Clinical and Experimental Immunology, Division of Nephrology, Universidade Federal de São Paulo, São Paulo, São Paulo, Brazil.
| |
Collapse
|
40
|
Yang T, Chakraborty S, Mandal J, Mei X, Joe B. Microbiota and Metabolites as Factors Influencing Blood Pressure Regulation. Compr Physiol 2021; 11:1731-1757. [PMID: 33792901 DOI: 10.1002/cphy.c200009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The study of microbes has rapidly expanded in recent years due to a surge in our understanding that humans host a plethora of commensal microbes, which reside in their bodies and depending upon their composition, contribute to either normal physiology or pathophysiology. This article provides a general foundation for learning about host-commensal microbial interactions as an emerging area of research. The article is divided into two sections. The first section is dedicated to introducing commensal microbiota and its known effects on the host. The second section is on metabolites, which are biochemicals that the host and the microbes use for bi-directional communication with each other. Together, the sections review what is known about how microbes interact with the host to impact cardiovascular physiology, especially blood pressure regulation. © 2021 American Physiological Society. Compr Physiol 11:1731-1757, 2021.
Collapse
Affiliation(s)
- Tao Yang
- Center for Hypertension and Precision Medicine and Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio, USA
| | - Saroj Chakraborty
- Center for Hypertension and Precision Medicine and Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio, USA
| | - Juthika Mandal
- Center for Hypertension and Precision Medicine and Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio, USA
| | - Xue Mei
- Center for Hypertension and Precision Medicine and Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio, USA
| | - Bina Joe
- Center for Hypertension and Precision Medicine and Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio, USA
| |
Collapse
|
41
|
Abstract
Hypertension is a leading risk factor for disease burden worldwide. The kidneys, which have a high specific metabolic rate, play an essential role in the long-term regulation of arterial blood pressure. In this review, we discuss the emerging role of renal metabolism in the development of hypertension. Renal energy and substrate metabolism is characterized by several important and, in some cases, unique features. Recent advances suggest that alterations of renal metabolism may result from genetic abnormalities or serve initially as a physiological response to environmental stressors to support tubular transport, which may ultimately affect regulatory pathways and lead to unfavorable cellular and pathophysiological consequences that contribute to the development of hypertension.
Collapse
Affiliation(s)
- Zhongmin Tian
- grid.43169.390000 0001 0599 1243The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an, Shaanxi China
| | - Mingyu Liang
- grid.30760.320000 0001 2111 8460Center of Systems Molecular Medicine, Department of Physiology, Medical College of Wisconsin, Milwaukee, WI USA
| |
Collapse
|
42
|
Deng Y, Huang C, Su J, Pan CW, Ke C. Identification of biomarkers for essential hypertension based on metabolomics. Nutr Metab Cardiovasc Dis 2021; 31:382-395. [PMID: 33495028 DOI: 10.1016/j.numecd.2020.11.023] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 11/10/2020] [Accepted: 11/20/2020] [Indexed: 01/09/2023]
Abstract
AIM Essential hypertension (EH) is one of the most important public health problems worldwide. However, the pathogenesis of EH is unclear and early diagnostic methods are lacking. Metabolomics demonstrates great potential for biomarker discovery and the mechanistic exploration of metabolic diseases. DATA SYNTHESIS This review included human and animal metabolomics studies related to EH in the PubMed and Web of Science databases between February 1996 and May 2020. The study designs, EH standards, and reported metabolic biomarkers were systematically examined and compared. The pathway analysis was conducted through the online software MetaboAnalyst 4.0. Twenty-two human studies and fifteen animal studies were included in this systematic review. There were many frequently reported biomarkers with consistent trends (e.g., pyruvate, lactic acid, valine, and tryptophan) in human and animal studies, and thus had potential as biomarkers of EH. In addition, several shared metabolic pathways, including alanine, aspartate, and glutamate metabolism, aminoacyl-tRNA biosynthesis, and arginine biosynthesis, were identified in human and animal metabolomics studies. These biomarkers and pathways, closely related to insulin resistance, the inflammatory state, and impaired nitric oxide production, were demonstrated to contribute to EH development. CONCLUSIONS This study summarized valuable metabolic biomarkers and pathways that could offer opportunities for the early diagnosis or prediction of EH and the discovery of the metabolic mechanisms of EH.
Collapse
Affiliation(s)
- Yueting Deng
- Medical College of Soochow University, Suzhou, 215123, PR China
| | - Chen Huang
- Medical College of Soochow University, Suzhou, 215123, PR China
| | - Jingyue Su
- Medical College of Soochow University, Suzhou, 215123, PR China
| | - Chen-Wei Pan
- School of Public Health, Medical College of Soochow University, 199 Renai Road, Suzhou, 215123, PR China.
| | - Chaofu Ke
- Department of Epidemiology and Biostatistics, School of Public Health, Medical College of Soochow University, 199 Renai Road, Suzhou, 215123, PR China.
| |
Collapse
|
43
|
Lim Y, Tang KD, Karpe AV, Beale DJ, Totsika M, Kenny L, Morrison M, Punyadeera C. Chemoradiation therapy changes oral microbiome and metabolomic profiles in patients with oral cavity cancer and oropharyngeal cancer. Head Neck 2021; 43:1521-1534. [PMID: 33527579 DOI: 10.1002/hed.26619] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 01/06/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Patients with oral cavity cancer (OCC) and oropharyngeal cancer (OPC) are often seen with locoregionally advanced disease requiring complex multimodality treatments. These treatments may have detrimental effects on the oral microbiome, which is critical to maintaining physiological balance and health. METHODS The effects of different OCC and OPC treatment types on the oral microbiome and metabolomic profiles for 24-month post-treatment in patients with OCC and OPC were investigated using 16S rRNA gene amplicon next-generation sequencing and gas chromatography-mass spectrometry (GC-MS), respectively. RESULTS Chemoradiation resulted in oral dysbiosis with specific depletion of genera which regulate the enterosalivary nitrate-nitrite-nitric oxide pathway. These data also correlate with the oral metabolomic profiles with nitric oxide-related precursor, modulator, or catalyst significantly downregulated in saliva samples from patients' postchemoradiation. CONCLUSIONS Together, we have shown that oral dysbiosis due to the effects of chemoradiation could potentially have an impact on OCC and OPC patient's quality of life post-treatment.
Collapse
Affiliation(s)
- Yenkai Lim
- The Saliva and Liquid Biopsy Translational Research Team, School of Biomedical Sciences, Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia.,The Translational Research Institute, Brisbane, Queensland, Australia
| | - Kai Dun Tang
- The Saliva and Liquid Biopsy Translational Research Team, School of Biomedical Sciences, Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia.,The Translational Research Institute, Brisbane, Queensland, Australia
| | - Avinash V Karpe
- The Land and Water, Commonwealth Scientific and Industrial Research Organisation, Ecosciences Precinct Dutton Park, Brisbane, Queensland, Australia
| | - David J Beale
- The Land and Water, Commonwealth Scientific and Industrial Research Organisation, Ecosciences Precinct Dutton Park, Brisbane, Queensland, Australia
| | - Makrina Totsika
- School of Biomedical Sciences, Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Liz Kenny
- The School of Medicine, University of Queensland, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia
| | - Mark Morrison
- The University of Queensland Diamantina Institute, University of Queensland, Translational Research Institute, Brisbane, Queensland, Australia
| | - Chamindie Punyadeera
- The Saliva and Liquid Biopsy Translational Research Team, School of Biomedical Sciences, Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia.,The Translational Research Institute, Brisbane, Queensland, Australia
| |
Collapse
|
44
|
Yang P, Zhao X, Zhou L, Jin Y, Zheng X, Ouyang Y, Chen M, Zeng L, Chen S, Chen X, Tian Z. Protective effect of oral histidine on hypertension in Dahl salt-sensitive rats induced by high-salt diet. Life Sci 2021; 270:119134. [PMID: 33513395 DOI: 10.1016/j.lfs.2021.119134] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 01/15/2021] [Accepted: 01/15/2021] [Indexed: 01/11/2023]
Abstract
AIMS Salt-sensitive hypertension is a risk factor for cardiovascular disease. Previous studies have shown that insufficient arginine in the kidney caused by metabolic imbalance is an important factor in salt-sensitive hypertension. Whether the high nitrogen content of histidine can affect the balance of nitrogen metabolism in Dahl salt-sensitive (SS) rats. This article aimed to study the effects of oral histidine on salt-sensitive hypertension, kidney damage and metabolic patterns of high-salt diet in SS rats. MAIN METHODS Adult rats were divided into four groups, and blood pressure was measured using a non-invasive tail-cuff system. Gas chromatography-mass spectrometry analyzed metabolites in serum and kidney tissues. KEY FINDINGS High-salt diet significantly increased the blood pressure of rats and aggravated kidney damage. Of note, histidine can attenuate salt-sensitive hypertension and kidney damage by improving metabolic pattern, reducing Reactive Oxygen Species (ROS) and increasing nitric oxide levels in SS rats. SIGNIFICANCE These results suggest that histidine could be a potential adjuvant to prevent and control 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
| | - Xinrui Zhao
- 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
| | - Yuexin 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
| | - 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
| | - 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
| | - Sa 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
| | - 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
| | - 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
|
45
|
Infante T, Costa D, Napoli C. Novel Insights Regarding Nitric Oxide and Cardiovascular Diseases. Angiology 2021; 72:411-425. [PMID: 33478246 DOI: 10.1177/0003319720979243] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Nitric oxide (NO) is a powerful mediator with biological activities such as vasodilation and prevention of vascular smooth muscle cell proliferation as well as functional regulation of cardiac cells. Thus, impaired production or reduced bioavailability of NO predisposes to the onset of different cardiovascular (CV) diseases. Alterations in the redox balance associated with excitation-contraction coupling have been identified in heart failure (HF), thus contributing to contractile abnormalities and arrhythmias. For its ability to influence cell proliferation and angiogenesis, NO may be considered a therapeutic option for the management of several CV diseases. Several clinical studies and trials investigated therapeutic NO strategies for systemic hypertension, atherosclerosis, and/or prevention of in stent restenosis, coronary heart disease (CHD), pulmonary arterial hypertension (PAH), and HF, although with mixed results in long-term treatment and effective dose administered in selected groups of patients. Tadalafil, sildenafil, and cinaguat were evaluated for the treatment of PAH, whereas vericiguat was investigated in the treatment of HF patients with reduced ejection fraction. Furthermore, supplementation with hydrogen sulfide, tetrahydrobiopterin, and nitrite/nitrate has shown beneficial effects at the vascular level.
Collapse
Affiliation(s)
- Teresa Infante
- Department of Advanced Clinical and Surgical Sciences, 18994University of Campania "Luigi Vanvitelli," Naples, Italy
| | - Dario Costa
- U.O.C. Division of Clinical Immunology, Immunohematology, Transfusion Medicine and Transplant Immunology, Clinical Department of Internal Medicine and Specialistics, 18994University of Campania "L. Vanvitelli," Naples, Italy
| | - Claudio Napoli
- Department of Advanced Clinical and Surgical Sciences, 18994University of Campania "Luigi Vanvitelli," Naples, Italy.,IRCCS SDN, Naples, Italy
| |
Collapse
|
46
|
Rao Y, Kuang Z, Li C, Guo S, Xu Y, Zhao D, Hu Y, Song B, Jiang Z, Ge Z, Liu X, Li C, Chen S, Ye J, Huang Z, Lu Y. Gut Akkermansia muciniphila ameliorates metabolic dysfunction-associated fatty liver disease by regulating the metabolism of L-aspartate via gut-liver axis. Gut Microbes 2021; 13:1-19. [PMID: 34030573 PMCID: PMC8158032 DOI: 10.1080/19490976.2021.1927633] [Citation(s) in RCA: 125] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2020] [Revised: 04/27/2021] [Accepted: 04/30/2021] [Indexed: 02/04/2023] Open
Abstract
The gut bacterium Akkermansia muciniphila has been increasingly recognized for its therapeutic potential in treating metabolic disorders, including obesity, diabetes, and metabolicdysfunction-associated fatty liver disease (MAFLD). However, its underlying mechanism involved in its well-known metabolic actions needs further evaluation. The present study explored the therapeutic effect and mechanism of A. muciniphila in intervening MAFLD by using a high-fat and high-cholesterol (HFC) diet induced obese mice model. Mice treated with A. muciniphila efficiently reversed MAFLD in the liver, such as hepatic steatosis, inflammatory, and liver injury. These therapeutic effects persisted after long-term drug withdrawal and were slightly weakened in the antibiotics-treated obese mice. A. muciniphila treatment efficiently increased mitochondrial oxidation and bile acid metabolism in the gut-liver axis, ameliorated oxidative stress-induced cell apoptosis in gut, leading to the reshaping of the gut microbiota composition. These metabolic improvements occurred with increased L-aspartate levels in the liver that transported from the gut. The administration of L-aspartate in vitro or in mice displayed the similar beneficial metabolic effects mentioned above and efficiently ameliorated MAFLD. Together, these data indicate that the anti-MAFLD activity of A. muciniphila correlated with lipid oxidation and improved gut-liver interactions through regulating the metabolism of L-aspartate. A. muciniphila could be a potential agent for clinical intervention in MAFLD.
Collapse
Affiliation(s)
- Yong Rao
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Sun Yat-sen University, Guangzhou, China
| | - Zhiqi Kuang
- Run Ze Laboratory for Gastrointestinal Microbiome Study, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
- Biomedical Center of Sun Yat-sen University, Guangzhou, China
| | - Chan Li
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Sun Yat-sen University, Guangzhou, China
| | - Shiyao Guo
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Sun Yat-sen University, Guangzhou, China
| | - Yaohao Xu
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Sun Yat-sen University, Guangzhou, China
| | - Dandan Zhao
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Sun Yat-sen University, Guangzhou, China
| | - Yutao Hu
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Sun Yat-sen University, Guangzhou, China
| | - Bingbing Song
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Sun Yat-sen University, Guangzhou, China
| | - Zhi Jiang
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Sun Yat-sen University, Guangzhou, China
| | - Zhenhuang Ge
- Run Ze Laboratory for Gastrointestinal Microbiome Study, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
- Biomedical Center of Sun Yat-sen University, Guangzhou, China
| | - Xiyuan Liu
- Run Ze Laboratory for Gastrointestinal Microbiome Study, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
- Biomedical Center of Sun Yat-sen University, Guangzhou, China
| | - Chengdao Li
- Run Ze Laboratory for Gastrointestinal Microbiome Study, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
- Biomedical Center of Sun Yat-sen University, Guangzhou, China
| | - Shuobin Chen
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Sun Yat-sen University, Guangzhou, China
| | - Jiming Ye
- Lipid Biology and Metabolic Disease Research Group, School of Health and Biomedical Sciences, RMIT University, Melbourne, Australia
| | - Zhishu Huang
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Sun Yat-sen University, Guangzhou, China
| | - Yongjun Lu
- Run Ze Laboratory for Gastrointestinal Microbiome Study, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
- Biomedical Center of Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
47
|
Zheng X, Zhao X, Jin Y, Zhou L, Yang P, Ahmad H, Tian Z. High salt diet contributes to hypertension by weakening the medullary tricarboxylic acid cycle and antioxidant system in Dahl salt-sensitive rats. Biochimie 2020; 181:154-161. [PMID: 33347925 DOI: 10.1016/j.biochi.2020.12.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Revised: 12/04/2020] [Accepted: 12/14/2020] [Indexed: 01/11/2023]
Abstract
High salt diet (HSD, 8% NaCl) contributes to salt-sensitive hypertension, this study aimed to determine the effect of HSD on salt-sensitive hypertension by combining proteomic with metabolomics methods. Salt-sensitive rats were fed on HSD and normal salt diet (NSD, 0.4% NaCl) for two weeks before further analysis. Proteomic analysis showed the differential expression proteins (DEPs) were primarily mapped in the tricarboxylic acid (TCA)-cycle, glycolysis/gluconeogenesis, and other pathways associated with multiple amino acids. HSD decreased the medullary activities and protein expression level of two key enzymes of TCA-cycle, MDH and NADP+-IDH. Metabolomics showed three serous TCA-cycle-associated compounds, including decreased malic acid, decreased citric acid, and increased fumaric acid were differentially detected, which resulted in a decrease in NO content and an increase in H2O2 content in serum. The content of GSH, GSH/GSSG ratio, and synthesis substrates of GSH-cysteine and glycine, were significantly decreased by HSD, thus attenuated the antioxidant system in the renal medulla. HSD enhanced the medullary pentose phosphate pathway, which finally increased the concentration of NADPH and NADP+, NADPH/NADP+, and the activity of NADPH oxidase in the renal medulla. Additionally, HSD enhanced the glycolysis pathway in the renal medulla. In summary, HSD significantly weakened the TCA cycle, and attenuated the antioxidant system in the renal medulla, which finally contributed to salt-sensitive hypertension.
Collapse
Affiliation(s)
- 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
| | - Xinrui Zhao
- 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
| | - Yuexin 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
| | - 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
| | - 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
| | - 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
| | - 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
|
48
|
Lim HJ, Jang YJ, Lee KH, Kim DM. Translational Detection of Nonproteinogenic Amino Acids Using an Engineered Complementary Cell-Free Protein Synthesis Assay. Anal Chem 2020; 92:11505-11510. [PMID: 32794704 DOI: 10.1021/acs.analchem.0c01978] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
We developed a simple and rapid method for analyzing nonproteinogenic amino acids that does not require conventional chromatographic equipment. In this technique, nonproteinogenic amino acids were first converted to a proteinogenic amino acid through in vitro metabolism in a cell extract. The proteinogenic amino acid generated from the nonproteinogenic precursors were then incorporated into a reporter protein using a cell-free protein synthesis system. The titers of the nonproteinogenic amino acids could be readily quantified by measuring the activity of reporter proteins. This method, which combines the enzymatic conversion of target amino acids with translational analysis, makes amino acid analysis more accessible while minimizing the cost and time requirements. We anticipate that the same strategy could be extended to the detection of diverse biochemical molecules with clinical and industrial implications.
Collapse
Affiliation(s)
- Hye Jin Lim
- Department of Chemical Engineering and Applied Chemistry, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, Korea
| | - Yeon Jae Jang
- Department of Chemical Engineering and Applied Chemistry, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, Korea
| | - Kyung-Ho Lee
- Department of Chemical Engineering and Applied Chemistry, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, Korea
| | - Dong-Myung Kim
- Department of Chemical Engineering and Applied Chemistry, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, Korea
| |
Collapse
|
49
|
Wang Q, Yue J, Zhou X, Zheng M, Cao B, Li J. Ouabain regulates kidney metabolic profiling in rat offspring of intrauterine growth restriction induced by low-protein diet. Life Sci 2020; 259:118281. [PMID: 32798554 DOI: 10.1016/j.lfs.2020.118281] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 08/10/2020] [Accepted: 08/11/2020] [Indexed: 11/18/2022]
Abstract
AIMS Intrauterine growth restriction (IUGR) can increase the risk of hypertension and kidney disease at adulthood due to fetal programming. In our previous study, we found that supplementation with low concentration of ouabain during pregnancy could restore glomerulus numbers at birth, rescuing kidney development. However, the metabolic pattern of kidney in IUGR offspring and the effect of ouabain have not been evaluated. MAIN METHODS In this study, based on GC-MS and LC-MS platforms, we used the protein restriction rat model to explore the molecular mechanisms of kidney damage induced by IUGR and the protective effect of ouabain. KEY FINDINGS The results showed that malnutrition could induce IUGR in rat offspring at the 20th gestational day but ouabain treatment could partially reverse the body and kidney weight loss. Ouabain treatment could upregulate arginine, N-acetylornithine and carbamoyl phosphate as well as adenine nucleotide and guanine nucleotide downregulated by low-protein diet. Moreover, six metabolites were identified to be significantly correlated with fetal kidney weight, with 3 metabolites involved in arginine metabolism (arginine, N-acetylornithine, urea) and UDP-glucuronate correlated positively, while lysine and anthranilate correlated negatively. SIGNIFICANCE The results suggested that the underlying mechanism of ouabain against renal maldevelopment involved the metabolic regulation, particularly the arginine metabolism, which played an important role in the development of fetal kidney.
Collapse
Affiliation(s)
- Qien Wang
- Phase I Clinical Trials Unit, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Jing Yue
- Department of Obstetrics and Gynecology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Xuan Zhou
- Phase I Clinical Trials Unit, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Meihong Zheng
- Phase I Clinical Trials Unit, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Bei Cao
- Phase I Clinical Trials Unit, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China.
| | - Juan Li
- Phase I Clinical Trials Unit, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China.
| |
Collapse
|
50
|
Qingda granules attenuate hypertensive cardiac remodeling and inflammation in spontaneously hypertensive rats. Biomed Pharmacother 2020; 129:110367. [PMID: 32559624 DOI: 10.1016/j.biopha.2020.110367] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 05/22/2020] [Accepted: 06/02/2020] [Indexed: 01/08/2023] Open
Abstract
Qingda granules (QDG) are derived from QingXuanJiangYa Decoction (QXJYD) a traditional Chinese medication that has been used to treat hypertension for more than 60 years. QXJYD has been shown to be effective in rat models of hypertension. However, the effects of QDG on hypertension remain largely unknown. In the current study, baicalin was identified as one of the main components of QDG using Ultra Performance Liquid Chromatography (UPLC) analysis. We investigated the effects of QDG on blood pressure, cardiac remodeling, and cardiac inflammation. QDG (0.8 g/kg/day) treatment attenuated the elevated blood pressure in spontaneously hypertensive rats (SHRs). Moreover, QDG treatment reduced the degree of myocardial fiber disarray, degeneration and necrosis of myocardial cells, expression of ANP and BNP, as well as collagen content of SHRs. Moreover, we further assessed the effect of QDG treatment on cardiac inflammation and found that QDG treatment reduced CD68 protein expression, decreased levels of IL-6 and TNF-α in both serum and cardiac tissues, as well as suppressed activation of NF-κB pathway in cardiac tissues of SHRs. Differential expressed metabolites (DEMs) analysis identified 41 increased and 51 decreased metabolites in the cardiac tissues of SHRs after QDG treatment. In summary, QDG treatment of SHRs attenuated the elevated blood pressure and ameliorated cardiac remodeling and inflammation, in part, through suppression of NF-κB pathway and DEMs, which provide a basis for other therapeutic uses of this TCM.
Collapse
|