1
|
Zhang P, Huang C, Liu H, Zhang M, Liu L, Zhai Y, Zhang J, Yang J, Yang J. The mechanism of the NFAT transcription factor family involved in oxidative stress response. J Cardiol 2024; 83:30-36. [PMID: 37149283 DOI: 10.1016/j.jjcc.2023.04.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 04/20/2023] [Accepted: 04/28/2023] [Indexed: 05/08/2023]
Abstract
As a transcriptional activator widely expressed in various tissues, nuclear factor of activated T cells (NFAT) is involved in the regulation of the immune system, the development of the heart and brain systems, and classically mediating pathological processes such as cardiac hypertrophy. Oxidative stress is an imbalance of intracellular redox status, characterized by excessive generation of reactive oxygen species, accompanied by mitochondrial dysfunction, calcium overload, and subsequent lipid peroxidation, inflammation, and apoptosis. Oxidative stress occurs during various pathological processes, such as chronic hypoxia, vascular smooth muscle cell phenotype switching, ischemia-reperfusion, and cardiac remodeling. Calcium overload leads to an increase in intracellular calcium concentration, while NFAT can be activated through calcium-calcineurin, which is also the main regulatory mode of NFAT factors. This review focuses on the effects of NFAT transcription factors on reactive oxygen species production, calcium overload, mitochondrial dysfunction, redox reactions, lipid peroxidation, inflammation, and apoptosis in response to oxidative stress. We hope to provide a reference for the functions and characteristics of NFAT involved in various stages of oxidative stress as well as related potential targets.
Collapse
Affiliation(s)
- Peiyue Zhang
- Department of Cardiology, The First College of Clinical Medical Science, China Three Gorges University, Yichang, China; Institute of Cardiovascular Diseases, China Three Gorges University, Yichang, China; HuBei Clinical Research Center for Ischemic Cardiovascular Disease, Yichang, China
| | - Cuiyuan Huang
- HuBei Clinical Research Center for Ischemic Cardiovascular Disease, Yichang, China
| | - Haiyin Liu
- Department of Cardiology, The First College of Clinical Medical Science, China Three Gorges University, Yichang, China; Institute of Cardiovascular Diseases, China Three Gorges University, Yichang, China; HuBei Clinical Research Center for Ischemic Cardiovascular Disease, Yichang, China
| | - Mengting Zhang
- Department of Cardiology, The First College of Clinical Medical Science, China Three Gorges University, Yichang, China; Institute of Cardiovascular Diseases, China Three Gorges University, Yichang, China; HuBei Clinical Research Center for Ischemic Cardiovascular Disease, Yichang, China
| | - Li Liu
- Institute of Cardiovascular Diseases, China Three Gorges University, Yichang, China; HuBei Clinical Research Center for Ischemic Cardiovascular Disease, Yichang, China
| | - Yuhong Zhai
- Department of Cardiology, The First College of Clinical Medical Science, China Three Gorges University, Yichang, China; Institute of Cardiovascular Diseases, China Three Gorges University, Yichang, China; HuBei Clinical Research Center for Ischemic Cardiovascular Disease, Yichang, China
| | - Jing Zhang
- Institute of Cardiovascular Diseases, China Three Gorges University, Yichang, China; HuBei Clinical Research Center for Ischemic Cardiovascular Disease, Yichang, China
| | - Jian Yang
- Department of Cardiology, The First College of Clinical Medical Science, China Three Gorges University, Yichang, China; Institute of Cardiovascular Diseases, China Three Gorges University, Yichang, China.
| | - Jun Yang
- Department of Cardiology, The First College of Clinical Medical Science, China Three Gorges University, Yichang, China; Institute of Cardiovascular Diseases, China Three Gorges University, Yichang, China.
| |
Collapse
|
2
|
Rong Z, Li F, Zhang R, Niu S, Di X, Ni L, Liu C. Ant-Neointimal Formation Effects of SLC6A6 in Preventing Vascular Smooth Muscle Cell Proliferation and Migration via Wnt/β-Catenin Signaling. Int J Mol Sci 2023; 24:ijms24033018. [PMID: 36769341 PMCID: PMC9917619 DOI: 10.3390/ijms24033018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 01/07/2023] [Accepted: 01/09/2023] [Indexed: 02/09/2023] Open
Abstract
Vascular smooth muscle cells (VSMCs) play an important role in the pathogenesis of vascular remolding, such as atherosclerosis and restenosis. Solute carrier family 6 member 6 (SLC6A6) is a transmembrane transporter that maintains a variety of physiological functions and is highly expressed in VSMCs. However, its role on VSMCs during neointimal formation remains unknown. In this study, mRNA and protein levels of SLC6A6 were examined using models of VSMC phenotype switching in vivo and in vitro and human artery samples with or without atherosclerosis. SLC6A6 gain- and loss-of-function approaches were performed by adenovirus infection or small interfering RNA (siRNA) transfection, respectively. Reactive oxygen species (ROS), proliferation, migration, and phenotype-related proteins of VSMCs were measured. Vascular stenosis rate and related genes were assessed in a rat vascular balloon injury model overexpressing SLC6A6. SLC6A6 was downregulated in dedifferentiated VSMCs, atherosclerotic vascular tissues, and injured vascular tissues. SLC6A6 suppressed VSMC proliferation and migration, while increasing contractile VSMC proteins. Mechanistically, SLC6A6 overexpression reduced ROS production and inhibited the Wnt/β-catenin pathway. Furthermore, SLC6A6 overexpression suppressed neointimal formation in vivo. Collectively, overexpression of SLC6A6 suppresses neointimal formation by inhibiting VSMC proliferation and migration via Wnt/β-catenin signaling and maintaining the VSMC contractile phenotype.
Collapse
Affiliation(s)
| | | | | | | | | | - Leng Ni
- Correspondence: (L.N.); (C.L.); Tel.: +86-010-69152501 (L.N.); +86-010-69152500 (C.L.)
| | - Changwei Liu
- Correspondence: (L.N.); (C.L.); Tel.: +86-010-69152501 (L.N.); +86-010-69152500 (C.L.)
| |
Collapse
|
3
|
Yang L, Li Z, Ouyang Y. Taurine attenuates ER stress‑associated apoptosis and catabolism in nucleus pulposus cells. Mol Med Rep 2022; 25:172. [PMID: 35315493 PMCID: PMC8971911 DOI: 10.3892/mmr.2022.12688] [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: 11/29/2021] [Accepted: 02/10/2022] [Indexed: 11/20/2022] Open
Abstract
Nucleus pulposus (NP) apoptosis and subsequent excessive degradation of the extracellular matrix (ECM) are key pathological characteristics of intervertebral disc degeneration (IDD). The present study aims to examine the signaling processes underlying the effects of taurine on IDD, with specific focus on endoplasmic reticulum (ER) stress-mediated apoptosis and ECM degradation, in NP cells. To clarify the role of taurine in IDD, NP cells were treated with various concentrations of taurine and IL-1β or thapsigargin (TG). Cell Counting Kit-8, western blotting, TUNEL, immunofluorescence assays and reverse transcription-quantitative PCR were applied to measure cell viability, the expression of ER stress-associated proteins (GRP78, CHOP and caspase-12), apoptosis and the levels of metabolic factors associated with ECM (MMP-1, 3, 9, ADAMTS-4, 5 and collagen II), respectively. Taurine was found to attenuate ER stress and prevent apoptosis in NP cells induced by IL-1β treatment. Additionally, taurine significantly decreased the expression of ER stress-activated glucose regulatory protein 78, C/EBP homologous protein and caspase-12. TUNEL results revealed that taurine decreased the number of apoptotic TG-treated NP cells. TG-treated NP cells also exhibited characteristics of increased ECM degradation, supported by observations of increased MMP-1, MMP-3, MMP-9 and A disintegrin and metalloproteinase with thrombospondin motifs (ADAMTS)-4 and ADAMTS-5 expression in addition to decreased collagen-II expression. However, taurine treatment significantly reversed all indicators of excessive ECM catabolism aforementioned. These data suggest that taurine can mediate protection against apoptosis and ECM degradation in NP cells by inhibiting ER stress, implicating therapeutic potential for the treatment of IDD.
Collapse
Affiliation(s)
- Liuxie Yang
- Department of Orthopedics, Shanghai Jing'an District Zhabei Central Hospital, Shanghai 200040, P.R. China
| | - Zhenhuan Li
- Department of Orthopedics, Shanghai Jing'an District Zhabei Central Hospital, Shanghai 200040, P.R. China
| | - Yueping Ouyang
- Department of Orthopedics, Shanghai Changzheng Hospital, Second Military Medical University, Shanghai 200003, P.R. China
| |
Collapse
|
4
|
Drug resistance of targeted therapy for advanced non-small cell lung cancer harbored EGFR mutation: from mechanism analysis to clinical strategy. J Cancer Res Clin Oncol 2021; 147:3653-3664. [PMID: 34661758 DOI: 10.1007/s00432-021-03828-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 10/05/2021] [Indexed: 12/19/2022]
Abstract
PURPOSE Non-small cell lung cancer (NSCLC) accounts for about 85% in all cases of lung cancer. In recent years, molecular targeting drugs for NSCLC have been developed rapidly. The epidermal growth factor receptor-tyrosine kinase inhibitors (EGFR-TKIs) have changed the paradigm of cancer therapy from empirical cytotoxic chemotherapy to molecular-targeted cancer therapy. Currently, there are three generations of EGFR-TKIs, all of which have achieved good efficacy in clinical therapy. However, most patients developed drug resistance after 6-13 months EGFR-TKIs treatment. Therefore, a comprehensive understanding of EGFR-TKIs resistance mechanisms is of vital importance for clinical management of NSCLC. METHODS Relevant data and information about the topic were obtained by searching PubMed (Medline), Web of Science and Google Scholar using the subject headings, such as "NSCLC", "EGFR-TKIs resistance", "EGFR mutations", "human epidermal growth factor receptor-2 (HER2/erbB-2)", "hepatocyte growth factor (HGF)", "vascular endothelial growth factor (VEGF)", "insulin-like growth factor 1 (IGF-1)", "epithelial-mesenchymal transition (EMT)", "phosphatase and tensin homolog (PTEN)", "RAS mutation", "BRAF mutation", "signal transducer and activator of transcription 3 (STAT3)", and "tumor microenvironment", etc. RESULTS: The mechanisms for EGFR-TKIs resistance include EGFR mutations, upregulation of HER2, HGF/c-MET, VEGF IGF1, EMT and STAT3 pathways, mutations of PTEN, RAS and BRAF genes, and activation of other by-pass pathways. These mechanisms are interconnected and can be potential targets for the treatment of NSCLC. CONCLUSION In this review, we discuss the mechanisms of EGFR-TKIs drug resistance and the clinical strategies to overcome drug resistance from the perspective of EGFR-TKIs combined treatment.
Collapse
|
5
|
NFAT5-Mediated Signalling Pathways in Viral Infection and Cardiovascular Dysfunction. Int J Mol Sci 2021; 22:ijms22094872. [PMID: 34064510 PMCID: PMC8124654 DOI: 10.3390/ijms22094872] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 04/30/2021] [Accepted: 05/02/2021] [Indexed: 02/06/2023] Open
Abstract
The nuclear factor of activated T cells 5 (NFAT5) is well known for its sensitivity to cellular osmolarity changes, such as in the kidney medulla. Accumulated evidence indicates that NFAT5 is also a sensitive factor to stress signals caused by non-hypertonic stimuli such as heat shock, biomechanical stretch stress, ischaemia, infection, etc. These osmolality-related and -unrelated stimuli can induce NFAT5 upregulation, activation and nuclear accumulation, leading to its protective role against various detrimental effects. However, dysregulation of NFAT5 expression may cause pathological conditions in different tissues, leading to a variety of diseases. These protective or pathogenic effects of NFAT5 are dictated by the regulation of its target gene expression and activation of its signalling pathways. Recent studies have found a number of kinases that participate in the phosphorylation/activation of NFAT5 and related signal proteins. Thus, this review will focus on the NFAT5-mediated signal transduction pathways. As for the stimuli that upregulate NFAT5, in addition to the stresses caused by hyperosmotic and non-hyperosmotic environments, other factors such as miRNA, long non-coding RNA, epigenetic modification and viral infection also play an important role in regulating NFAT5 expression; thus, the discussion in this regard is another focus of this review. As the heart, unlike the kidneys, is not normally exposed to hypertonic environments, studies on NFAT5-mediated cardiovascular diseases are just emerging and rapidly progressing. Therefore, we have also added a review on the progress made in this field of research.
Collapse
|
6
|
The PKA-p38MAPK-NFAT5-Organic Osmolytes Pathway in Duchenne Muscular Dystrophy: From Essential Player in Osmotic Homeostasis, Inflammation and Skeletal Muscle Regeneration to Therapeutic Target. Biomedicines 2021; 9:biomedicines9040350. [PMID: 33808305 PMCID: PMC8066813 DOI: 10.3390/biomedicines9040350] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 03/19/2021] [Accepted: 03/24/2021] [Indexed: 11/30/2022] Open
Abstract
In Duchenne muscular dystrophy (DMD), the absence of dystrophin from the dystrophin-associated protein complex (DAPC) causes muscle membrane instability, which leads to myofiber necrosis, hampered regeneration, and chronic inflammation. The resulting disabled DAPC-associated cellular pathways have been described both at the molecular and the therapeutical level, with the Toll-like receptor nuclear factor kappa-light-chain-enhancer of activated B cells pathway (NF-ƘB), Janus kinase/signal transducer and activator of transcription proteins, and the transforming growth factor-β pathways receiving the most attention. In this review, we specifically focus on the protein kinase A/ mitogen-activated protein kinase/nuclear factor of activated T-cells 5/organic osmolytes (PKA-p38MAPK-NFAT5-organic osmolytes) pathway. This pathway plays an important role in osmotic homeostasis essential to normal cell physiology via its regulation of the influx/efflux of organic osmolytes. Besides, NFAT5 plays an essential role in cell survival under hyperosmolar conditions, in skeletal muscle regeneration, and in tissue inflammation, closely interacting with the master regulator of inflammation NF-ƘB. We describe the involvement of the PKA-p38MAPK-NFAT5-organic osmolytes pathway in DMD pathophysiology and provide a clear overview of which therapeutic molecules could be of potential benefit to DMD patients. We conclude that modulation of the PKA-p38MAPK-NFAT5-organic osmolytes pathway could be developed as supportive treatment for DMD in conjunction with genetic therapy.
Collapse
|
7
|
Alaimo A, Di Santo MC, Domínguez Rubio AP, Chaufan G, García Liñares G, Pérez OE. Toxic effects of A2E in human ARPE-19 cells were prevented by resveratrol: a potential nutritional bioactive for age-related macular degeneration treatment. Arch Toxicol 2019; 94:553-572. [PMID: 31792590 DOI: 10.1007/s00204-019-02637-w] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 11/26/2019] [Indexed: 01/23/2023]
Abstract
Age-related macular degeneration (AMD) is a late-onset retinal disease and the leading cause of central vision loss in the elderly. Degeneration of retinal pigment epithelial cells (RPE) is a crucial contributing factor responsible for the onset and progression of AMD. The toxic fluorophore N-retinyl-N-retinylidene ethanolamine (A2E), a major lipofuscin component, accumulates in RPE cells with age. Phytochemicals with antioxidant properties may have a potential role in both the prevention and treatment of this age-related ocular disease. Particularly, there is an increased interest in the therapeutic effects of resveratrol (RSV), a naturally occurring polyphenol (3,4',5-trihydroxystilbene). However, the underlying mechanism of the RSV antioxidative effect in ocular diseases has not been well explored. We hypothesized that this bioactive compound may have beneficial effects for AMD. To this end, to investigate the potential profits of RSV against A2E-provoked oxidative damage, we used human RPE cell line (ARPE-19). RSV (25 µM) attenuates the cytotoxicity and the typical morphological characteristics of apoptosis observed in 25 µM A2E-laden cells. RSV pretreatment strengthened cell monolayer integrity through the preservation of the transepithelial electrical resistance and reduced the fluorescein isothiocyanate (FITC)-dextran diffusion rate as well as cytoskeleton architecture. In addition, RSV exhorts protective effects against A2E-induced modifications in the intracellular redox balance. Finally, RSV also prevented A2E-induced mitochondrial network fragmentation. These findings reinforce the idea that RSV represents an attractive bioactive for therapeutic intervention against ocular diseases associated with oxidative stress such as AMD.
Collapse
Affiliation(s)
- Agustina Alaimo
- Departamento de Química Biológica, Laboratorio Interdisciplinario de Dinámica Celular y Nanoherramientas, CONICET-Universidad de Buenos Aires, Instituto de Química Biológica Ciencias Exactas y Naturales (IQUIBICEN), Facultad de Ciencias Exactas y Naturales, Pabellón 2, Ciudad Universitaria, 1428, Buenos Aires, Argentina.
| | - Mariana Carolina Di Santo
- Departamento de Química Biológica, Laboratorio Interdisciplinario de Dinámica Celular y Nanoherramientas, CONICET-Universidad de Buenos Aires, Instituto de Química Biológica Ciencias Exactas y Naturales (IQUIBICEN), Facultad de Ciencias Exactas y Naturales, Pabellón 2, Ciudad Universitaria, 1428, Buenos Aires, Argentina
| | - Ana Paula Domínguez Rubio
- Departamento de Química Biológica, Laboratorio Interdisciplinario de Dinámica Celular y Nanoherramientas, CONICET-Universidad de Buenos Aires, Instituto de Química Biológica Ciencias Exactas y Naturales (IQUIBICEN), Facultad de Ciencias Exactas y Naturales, Pabellón 2, Ciudad Universitaria, 1428, Buenos Aires, Argentina
| | - Gabriela Chaufan
- Departamento de Química Biológica, Laboratorio de Enzimología, Estrés Oxidativo y Metabolismo, CONICET-Universidad de Buenos Aires, Instituto de Química Biológica Ciencias Exactas y Naturales (IQUIBICEN), Facultad de Ciencias Exactas y Naturales, Pabellón 2, Ciudad Universitaria, 1428, Buenos Aires, Argentina
| | - Guadalupe García Liñares
- Departamento de Química Orgánica, Laboratorio de Biocatálisis, CONICET-Universidad de Buenos Aires, Unidad de Microanálisis y Métodos Físicos en Química Orgánica (UMYMFOR), Facultad de Ciencias Exactas y Naturales, Pabellón 2, Ciudad Universitaria, 1428, Buenos Aires, Argentina
| | - Oscar Edgardo Pérez
- Departamento de Química Biológica, Laboratorio Interdisciplinario de Dinámica Celular y Nanoherramientas, CONICET-Universidad de Buenos Aires, Instituto de Química Biológica Ciencias Exactas y Naturales (IQUIBICEN), Facultad de Ciencias Exactas y Naturales, Pabellón 2, Ciudad Universitaria, 1428, Buenos Aires, Argentina.
| |
Collapse
|
8
|
Roles of taurine in cognitive function of physiology, pathologies and toxication. Life Sci 2019; 231:116584. [DOI: 10.1016/j.lfs.2019.116584] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 06/09/2019] [Accepted: 06/17/2019] [Indexed: 11/23/2022]
|
9
|
Abstract
Malnutrition is a common feature of chronic and acute diseases, often associated with a poor prognosis, including worsening of clinical outcome, owing, among other factors, to dysfunction of the most internal organs and systems affecting the absorption, metabolism and elimination of drugs and nutrients. Taurine is involved in numerous biological processes and is required in increased amounts in response to pathological conditions. The aim of this study was to describe the behaviour of taurine in well-nourished (WN) rats and to analyse the influence of protein-energy undernutrition on the pharmacokinetic (PK) parameters of taurine, using a PK model. Wistar rats were randomly distributed into two groups, WN and undernourished (UN), and taurine was administered intravenously or orally at different doses: 1, 10 and 100 mg. Population pharmacokinetic modelling of plasma levels was performed using the NONMEM 7.2 program. Several distribution and absorption models were explored in combination with dose and/or time covariate effects. Covariates such as nutritional status, serum albumin, body weight and score of undernutrition were used. A two-compartment population pharmacokinetic model with zero-order endogenous formation, passive absorption, first-order kinetics distribution and non-linear elimination with parallel Michaelis-Menten excretion and reabsorption processes best described taurine pharmacokinetics. Undernutrition acted as a covariate reducing the V max of the active elimination process. Data analysis showed linear absorption and distribution, and non-linear elimination processes for taurine. Elimination of taurine was reduced in UN animals, suggesting that the reabsorption process via the secretion transporter was modified in that group.
Collapse
|
10
|
Wen C, Li F, Zhang L, Duan Y, Guo Q, Wang W, He S, Li J, Yin Y. Taurine is Involved in Energy Metabolism in Muscles, Adipose Tissue, and the Liver. Mol Nutr Food Res 2018; 63:e1800536. [DOI: 10.1002/mnfr.201800536] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Revised: 09/13/2018] [Indexed: 01/05/2023]
Affiliation(s)
- Chaoyue Wen
- Laboratory of Animal Nutrition and Human HealthHunan international joint laboratory of Animal Intestinal Ecology and HealthCollege of Life ScienceHunan Normal University Changsha Hunan 410081 China
| | - Fengna Li
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic ProcessKey Laboratory of Agro‐ecological Processes in Subtropical RegionInstitute of Subtropical AgricultureChinese Academy of SciencesHunan Provincial Engineering Research Center for Healthy Livestock and Poultry ProductionScientific Observing and Experimental Station of Animal Nutrition and Feed Science in South‐CentralMinistry of Agriculture Changsha 410125 China
- Hunan Co‐Innovation Center of Animal Production SafetyCICAPSHunan Collaborative Innovation Center for Utilization of Botanical Functional Ingredients Changsha 410128 China
| | - Lingyu Zhang
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic ProcessKey Laboratory of Agro‐ecological Processes in Subtropical RegionInstitute of Subtropical AgricultureChinese Academy of SciencesHunan Provincial Engineering Research Center for Healthy Livestock and Poultry ProductionScientific Observing and Experimental Station of Animal Nutrition and Feed Science in South‐CentralMinistry of Agriculture Changsha 410125 China
- University of Chinese Academy of Sciences Beijing 100039 China
| | - Yehui Duan
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic ProcessKey Laboratory of Agro‐ecological Processes in Subtropical RegionInstitute of Subtropical AgricultureChinese Academy of SciencesHunan Provincial Engineering Research Center for Healthy Livestock and Poultry ProductionScientific Observing and Experimental Station of Animal Nutrition and Feed Science in South‐CentralMinistry of Agriculture Changsha 410125 China
| | - Qiuping Guo
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic ProcessKey Laboratory of Agro‐ecological Processes in Subtropical RegionInstitute of Subtropical AgricultureChinese Academy of SciencesHunan Provincial Engineering Research Center for Healthy Livestock and Poultry ProductionScientific Observing and Experimental Station of Animal Nutrition and Feed Science in South‐CentralMinistry of Agriculture Changsha 410125 China
- University of Chinese Academy of Sciences Beijing 100039 China
| | - Wenlong Wang
- Laboratory of Animal Nutrition and Human HealthHunan international joint laboratory of Animal Intestinal Ecology and HealthCollege of Life ScienceHunan Normal University Changsha Hunan 410081 China
| | - Shanping He
- Laboratory of Animal Nutrition and Human HealthHunan international joint laboratory of Animal Intestinal Ecology and HealthCollege of Life ScienceHunan Normal University Changsha Hunan 410081 China
| | - Jianzhong Li
- Laboratory of Animal Nutrition and Human HealthHunan international joint laboratory of Animal Intestinal Ecology and HealthCollege of Life ScienceHunan Normal University Changsha Hunan 410081 China
| | - Yulong Yin
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic ProcessKey Laboratory of Agro‐ecological Processes in Subtropical RegionInstitute of Subtropical AgricultureChinese Academy of SciencesHunan Provincial Engineering Research Center for Healthy Livestock and Poultry ProductionScientific Observing and Experimental Station of Animal Nutrition and Feed Science in South‐CentralMinistry of Agriculture Changsha 410125 China
- Hunan Co‐Innovation Center of Animal Production SafetyCICAPSHunan Collaborative Innovation Center for Utilization of Botanical Functional Ingredients Changsha 410128 China
| |
Collapse
|
11
|
Yu M, Sun S, Yu J, Du F, Zhang S, Yang W, Xiao J, Xie B. Discovery and Validation of Potential Serum Biomarkers for Pediatric Patients with Congenital Heart Diseases by Metabolomics. J Proteome Res 2018; 17:3517-3525. [PMID: 30207476 DOI: 10.1021/acs.jproteome.8b00466] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
To identify and screen serum biomarkers to determine pediatric patients with congenital heart diseases (PCH) from healthy control children (NC), a total of 614 clinically diagnosed subjects from three hospitals, including 491 PCH and 234 NC, were enrolled for nontargeted proton nuclear magnetic resonance spectroscopy (1H NMR)-based and targeted ultra-high-performance liquid chromatography-tandem mass spectroscopy (UPLC-MS/MS)-based metabolomics studies. Nineteen serum metabolites distinguishing PCH from NC were identified by 1H NMR-based metabolomic analysis. The amino acid and choline metabolic pathways were considered to be closely related to PCH. The serum levels of 13 metabolites in these two pathways were further determined by UPLC-MS/MS and observed to be altered significantly in PCH. Taurine, glutamine, and glutamate presented considerable diagnostic value for the diagnosis of PCH (AUROC > 0.80). Logistic regression analysis showed that a combination of four variables, namely, betaine, taurine, glutamine, and phenylalanine, yields a high diagnostic value (AUROC = 0.949) and prediction accuracy (89.1%) for differentiating PCH from the NC, and the sensitivity and specificity were 93.9 and 95.2%, respectively. Further double-blind sample prediction showed that the accuracy of the model was 83.8% for 80 unknown samples. Our results showed that the serum amino acid and choline metabolite levels in PCH were changed considerably. The combination of four metabolites, namely, betaine, taurine, glutamine, and phenylalanine, can be used as potential serum biomarkers in PCH diagnosis, which contributes to the early PCH screening.
Collapse
Affiliation(s)
- Mengjie Yu
- Department of Infectious Diseases, the Second Affiliated Hospital of Nanchang University, School of Pharmaceutical Science , Nanchang University , Nanchang 330006 , P. R. China
| | - Shuilin Sun
- Department of Infectious Diseases, the Second Affiliated Hospital of Nanchang University, School of Pharmaceutical Science , Nanchang University , Nanchang 330006 , P. R. China
| | - Jiangqing Yu
- Medical College of Nanchang University , Nanchang University , Nanchang 330006 , P. R. China.,Respiratory Medicine , The First People's Hospital of Jingdezheng City , Jingdezheng 333000 , P. R. China
| | - Fen Du
- Department of Infectious Diseases, the Second Affiliated Hospital of Nanchang University, School of Pharmaceutical Science , Nanchang University , Nanchang 330006 , P. R. China
| | - Shouhua Zhang
- Department of General Surgery , Jiangxi Children's Hospital , Nanchang 330006 , P. R. China
| | - Wenlong Yang
- Department of Infectious Diseases, the Second Affiliated Hospital of Nanchang University, School of Pharmaceutical Science , Nanchang University , Nanchang 330006 , P. R. China
| | - Juhua Xiao
- Department of Ultrasound , Jiangxi Provincial Maternal and Child Health Hospital , Nanchang 330006 , P. R. China
| | - Baogang Xie
- Department of Infectious Diseases, the Second Affiliated Hospital of Nanchang University, School of Pharmaceutical Science , Nanchang University , Nanchang 330006 , P. R. China
| |
Collapse
|
12
|
High salt intake causes leptin resistance and obesity in mice by stimulating endogenous fructose production and metabolism. Proc Natl Acad Sci U S A 2018; 115:3138-3143. [PMID: 29507217 DOI: 10.1073/pnas.1713837115] [Citation(s) in RCA: 182] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Dietary guidelines for obesity typically focus on three food groups (carbohydrates, fat, and protein) and caloric restriction. Intake of noncaloric nutrients, such as salt, are rarely discussed. However, recently high salt intake has been reported to predict the development of obesity and insulin resistance. The mechanism for this effect is unknown. Here we show that high intake of salt activates the aldose reductase-fructokinase pathway in the liver and hypothalamus, leading to endogenous fructose production with the development of leptin resistance and hyperphagia that cause obesity, insulin resistance, and fatty liver. A high-salt diet was also found to predict the development of diabetes and nonalcoholic fatty liver disease in a healthy population. These studies provide insights into the pathogenesis of obesity and diabetes and raise the potential for reduction in salt intake as an additional interventional approach for reducing the risk for developing obesity and metabolic syndrome.
Collapse
|
13
|
Zhang Z, Zhao L, Zhou Y, Lu X, Wang Z, Wang J, Li W. Taurine ameliorated homocysteine-induced H9C2 cardiomyocyte apoptosis by modulating endoplasmic reticulum stress. Apoptosis 2017; 22:647-661. [DOI: 10.1007/s10495-017-1351-9] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
14
|
Han Y, Gao P, Qiu S, Zhang L, Yang L, Zuo J, Zhong C, Zhu S, Liu W. MTERF2 contributes to MPP+-induced mitochondrial dysfunction and cell damage. Biochem Biophys Res Commun 2016; 471:177-83. [DOI: 10.1016/j.bbrc.2016.01.156] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Accepted: 01/24/2016] [Indexed: 02/05/2023]
|