1
|
Chen T, Xiang L, Zhang W, Xia Z, Chen W. AGXT2 Suppresses the Proliferation and Dissemination of Hepatocellular Carcinoma Cells by Modulating Intracellular Lipid Metabolism. J Hepatocell Carcinoma 2024; 11:1623-1639. [PMID: 39206420 PMCID: PMC11353308 DOI: 10.2147/jhc.s470250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Accepted: 08/18/2024] [Indexed: 09/04/2024] Open
Abstract
Purpose Alanine glyoxylate aminotransferase (AGXT) family members are crucial in cancer processes, but their role in hepatocellular carcinoma (HCC) metabolism is unclear. This study investigates AGXT2's function in HCC. Patients and Methods AGTX2 expression was studied using bioinformatics, real-time reverse transcriptase-polymerase chain reaction (RT-qPCR), Western blot, and Enzyme-linked immunosorbent assay (ELISA). A lentivirus-induced AGTX2 overexpression cell model was analyzed with RNA sequencing (RNA-seq) and liquid chromatography-mass spectrometry (LC-MS). Cholesterol levels were confirmed by Oil Red O staining. AGTX2 effects were evaluated through cell cycle analysis, wound healing, and transwell migration assays.Tumorigenic effects were observed in NOD-SCID IL2Rγnull (NTG) mice in subcutaneous experiments. Protein interaction was examined through co-immunoprecipitation methods. Results We observed a significant reduction in AGXT2 mRNA and protein levels in both HCC tumor tissues and serum samples from patients with liver cancer, which was associated with a worse prognosis. The activation of AGXT2 has been shown to effectively decrease cholesterol levels in liver cancer cells, serving as an antagonist in the cholesterol metabolism pathway. An increase in low density lipoprotein receptor (LDLR) mRNA was noted in cells overexpressing AGXT2, accompanied by a decrease in LDLR protein and an elevation in proprotein convertase subtilisin/kexin type 9 (PCSK9) mRNA and protein levels. Molecular docking and co-immunoprecipitation experiments further elucidated the interaction between AGXT2 and LDLR proteins. AGXT2 was observed to suppress the migratory and invasive capabilities of HCC cells, inducing cell cycle arrest in the G2/M phase. AGXT2 activation inhibited subcutaneous liver cancer tumor growth in NTG mice. Conclusion AGXT2 was found to lower cholesterol levels in liver cancer cells, possibly through interactions with the LDLR protein and modulation of PCSK9-mediated LDLR degradation. This mechanism may impede cholesterol transport to liver cancer cells, thereby suppressing their growth and metastasis.
Collapse
Affiliation(s)
- Tian Chen
- Department of Laboratory Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, People’s Republic of China
| | - Lunjian Xiang
- Hepatobiliary Surgery, Chongqing University Three Gorges Hospital, Chongqing, People’s Republic of China
| | - Wenjin Zhang
- Chongqing Municipality Clinical Research Center for Endocrinology and Metabolic Diseases, Chongqing University Three Gorges Hospital, Chongqing, People’s Republic of China
| | - Zhenyi Xia
- Thoracic surgery, Chongqing University Three Gorges Hospital, Chongqing, People’s Republic of China
| | - Weixian Chen
- Department of Laboratory Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, People’s Republic of China
| |
Collapse
|
2
|
Alshahrani A, Aljada A, Masood A, Mujammami M, Alfadda AA, Musambil M, Alanazi IO, Al Dubayee M, Abdel Rahman AM, Benabdelkamel H. Proteomic Profiling Identifies Distinct Regulation of Proteins in Obese Diabetic Patients Treated with Metformin. Pharmaceuticals (Basel) 2023; 16:1345. [PMID: 37895816 PMCID: PMC10609691 DOI: 10.3390/ph16101345] [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: 07/20/2023] [Revised: 09/12/2023] [Accepted: 09/20/2023] [Indexed: 10/29/2023] Open
Abstract
Background: Obesity and type 2 diabetes mellitus (T2DM) are characterized by underlying low-grade chronic inflammation. Metformin has been used as the first line of therapy in T2DM as it decreases hepatic glucose production and glucose intestinal absorption, enhances insulin sensitivity and weight loss, and is known to ameliorate inflammation. The mechanisms through which metformin exerts its effect remain unclear. Proteomics has emerged as a unique approach to explore the biological changes associated with diseases, including T2DM. It provides insight into the circulating biomarkers/mediators which could be utilized for disease screening, diagnosis, and prognosis. Methods: This study evaluated the proteomic changes in obese (Ob), obese diabetics (OD), and obese diabetic patients on metformin (ODM) using a 2D DIGE MALDI-TOF mass spectrometric approach. Results: Significant changes in sixteen plasma proteins (15 up and 1 down, ANOVA, p ≤ 0.05; fold change ≥ 1.5) were observed in the ODM group when compared to the Ob and OD groups. Bioinformatic network pathway analysis revealed that the majority of these altered plasma proteins are involved in distinct pathways involving acute-phase response, inflammation, and oxidative response and were centered around HNF4A, ERK, JNK, and insulin signaling pathways. Conclusions: Our study provides important information about the possible biomarkers altered by metformin treatment in obese patients with and without T2DM. These altered plasma proteins are involved in distinct pathways involving acute-phase response, inflammation, and oxidative response and were centered around HNF4A, ERK, JNK, and insulin signaling pathways. The presented proteomic profiling approach may help in identifying potential biomarkers/mediators affected by metformin treatment in T2DM and inform the understanding of metformin's mechanisms of action.
Collapse
Affiliation(s)
- Awad Alshahrani
- Department of Medicine, Ministry of National Guard Health Affairs, Riyadh 11426, Saudi Arabia; (A.A.); (M.A.D.)
- King Abdullah International Medical Research Center, College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Riyadh 11481, Saudi Arabia
| | - Ahmad Aljada
- Department of Biochemistry and Molecular Medicine, College of Medicine, Al Faisal University, Riyadh 11533, Saudi Arabia;
| | - Afshan Masood
- Proteomics Resource Unit, Obesity Research Center, College of Medicine, King Saud University, P.O. Box 2925 (98), Riyadh 11461, Saudi Arabia; (A.M.); (A.A.A.); (M.M.)
| | - Muhammad Mujammami
- Endocrinology and Diabetes Unit, Department of Medicine, College of Medicine, King Saud University, Riyadh 11461, Saudi Arabia;
- University Diabetes Center, King Saud University Medical City, King Saud University, Riyadh 11461, Saudi Arabia
| | - Assim A. Alfadda
- Proteomics Resource Unit, Obesity Research Center, College of Medicine, King Saud University, P.O. Box 2925 (98), Riyadh 11461, Saudi Arabia; (A.M.); (A.A.A.); (M.M.)
- Department of Medicine, College of Medicine and King Saud Medical City, King Saud University, P.O. Box 2925 (98), Riyadh 11461, Saudi Arabia
| | - Mohthash Musambil
- Proteomics Resource Unit, Obesity Research Center, College of Medicine, King Saud University, P.O. Box 2925 (98), Riyadh 11461, Saudi Arabia; (A.M.); (A.A.A.); (M.M.)
| | - Ibrahim O. Alanazi
- Healthy Aging Research Institute, Health Sector, King Abdulaziz City for Science and Technology (KACST), P.O. Box 6086, Riyadh 11442, Saudi Arabia;
| | - Mohammed Al Dubayee
- Department of Medicine, Ministry of National Guard Health Affairs, Riyadh 11426, Saudi Arabia; (A.A.); (M.A.D.)
- King Abdullah International Medical Research Center, College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Riyadh 11481, Saudi Arabia
| | - Anas M. Abdel Rahman
- Department of Biochemistry and Molecular Medicine, College of Medicine, Al Faisal University, Riyadh 11533, Saudi Arabia;
- Metabolomics Section, Department of Clinical Genomics, Center for Genomics Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh 11564, Saudi Arabia
| | - Hicham Benabdelkamel
- Proteomics Resource Unit, Obesity Research Center, College of Medicine, King Saud University, P.O. Box 2925 (98), Riyadh 11461, Saudi Arabia; (A.M.); (A.A.A.); (M.M.)
| |
Collapse
|
3
|
Kozlova AA, Rubets E, Vareltzoglou MR, Jarzebska N, Ragavan VN, Chen Y, Martens-Lobenhoffer J, Bode-Böger SM, Gainetdinov RR, Rodionov RN, Bernhardt N. Knock-out of the critical nitric oxide synthase regulator DDAH1 in mice impacts amphetamine sensitivity and dopamine metabolism. J Neural Transm (Vienna) 2023; 130:1097-1112. [PMID: 36792833 PMCID: PMC10460711 DOI: 10.1007/s00702-023-02597-7] [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: 11/09/2022] [Accepted: 01/28/2023] [Indexed: 02/17/2023]
Abstract
The enzyme dimethylarginine dimethylaminohydrolase 1 (DDAH1) plays a pivotal role in the regulation of nitric oxide levels by degrading the main endogenous nitric oxide synthase inhibitor asymmetric dimethylarginine (ADMA). Growing evidence highlight the potential implication of DDAH/ADMA axis in the etiopathogenesis of several neuropsychiatric and neurological disorders, yet the underlying molecular mechanisms remain elusive. In this study, we sought to investigate the role of DDAH1 in behavioral endophenotypes with neuropsychiatric relevance. To achieve this, a global DDAH1 knock-out (DDAH1-ko) mouse strain was employed. Behavioral testing and brain region-specific neurotransmitter profiling have been conducted to assess the effect of both genotype and sex. DDAH1-ko mice exhibited increased exploratory behavior toward novel objects, altered amphetamine response kinetics and decreased dopamine metabolite 3,4-dihydroxyphenylacetic acid (DOPAC) level in the piriform cortex and striatum. Females of both genotypes showed the most robust amphetamine response. These results support the potential implication of the DDAH/ADMA pathway in central nervous system processes shaping the behavioral outcome. Yet, further experiments are required to complement the picture and define the specific brain-regions and mechanisms involved.
Collapse
Affiliation(s)
- Alena A Kozlova
- Department of Psychiatry and Psychotherapy, University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307, Dresden, Germany
| | - Elena Rubets
- Division of Angiology, Department of Internal Medicine III, University Center for Vascular Medicine, Technische Universität Dresden, 01307, Dresden, Germany
| | - Magdalini R Vareltzoglou
- Department of Psychiatry and Psychotherapy, University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307, Dresden, Germany
| | - Natalia Jarzebska
- Division of Angiology, Department of Internal Medicine III, University Center for Vascular Medicine, Technische Universität Dresden, 01307, Dresden, Germany
| | - Vinitha N Ragavan
- Division of Angiology, Department of Internal Medicine III, University Center for Vascular Medicine, Technische Universität Dresden, 01307, Dresden, Germany
| | - Yingjie Chen
- Department of Physiology & Biophysics, University of Mississippi Medical Center, Jackson, MS, 39216, USA
| | | | - Stefanie M Bode-Böger
- Institute of Clinical Pharmacology, Otto-Von-Guericke University, Magdeburg, Germany
| | - Raul R Gainetdinov
- Institute of Translational Biomedicine and Saint-Petersburg University Hospital, Saint-Petersburg State University, 199034, Saint-Petersburg, Russia
| | - Roman N Rodionov
- Division of Angiology, Department of Internal Medicine III, University Center for Vascular Medicine, Technische Universität Dresden, 01307, Dresden, Germany
| | - Nadine Bernhardt
- Department of Psychiatry and Psychotherapy, University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307, Dresden, Germany.
| |
Collapse
|
4
|
Ragavan VN, Nair PC, Jarzebska N, Angom RS, Ruta L, Bianconi E, Grottelli S, Tararova ND, Ryazanskiy D, Lentz SR, Tommasi S, Martens-Lobenhoffer J, Suzuki-Yamamoto T, Kimoto M, Rubets E, Chau S, Chen Y, Hu X, Bernhardt N, Spieth PM, Weiss N, Bornstein SR, Mukhopadhyay D, Bode-Böger SM, Maas R, Wang Y, Macchiarulo A, Mangoni AA, Cellini B, Rodionov RN. A multicentric consortium study demonstrates that dimethylarginine dimethylaminohydrolase 2 is not a dimethylarginine dimethylaminohydrolase. Nat Commun 2023; 14:3392. [PMID: 37296100 PMCID: PMC10256801 DOI: 10.1038/s41467-023-38467-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Accepted: 04/27/2023] [Indexed: 06/12/2023] Open
Abstract
Dimethylarginine dimethylaminohydrolase 1 (DDAH1) protects against cardiovascular disease by metabolising the risk factor asymmetric dimethylarginine (ADMA). However, the question whether the second DDAH isoform, DDAH2, directly metabolises ADMA has remained unanswered. Consequently, it is still unclear if DDAH2 may be a potential target for ADMA-lowering therapies or if drug development efforts should focus on DDAH2's known physiological functions in mitochondrial fission, angiogenesis, vascular remodelling, insulin secretion, and immune responses. Here, an international consortium of research groups set out to address this question using in silico, in vitro, cell culture, and murine models. The findings uniformly demonstrate that DDAH2 is incapable of metabolising ADMA, thus resolving a 20-year controversy and providing a starting point for the investigation of alternative, ADMA-independent functions of DDAH2.
Collapse
Affiliation(s)
- Vinitha N Ragavan
- Department of Internal Medicine III, Technische Universität Dresden, Dresden, Germany
- Department of Clinical Pharmacology, College of Medicine and Public Health, Flinders University and Flinders Medical Centre, Bedford Park, Adelaide, SA, Australia
| | - Pramod C Nair
- Department of Clinical Pharmacology, College of Medicine and Public Health, Flinders University and Flinders Medical Centre, Bedford Park, Adelaide, SA, Australia
- Flinders Health and Medical Research Institute (FHMRI), College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
- Cancer Program, South Australian Health and Medical Research Institute (SAHMRI), University of Adelaide, Adelaide, SA, Australia
- Discipline of Medicine, Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| | - Natalia Jarzebska
- Department of Internal Medicine III, Technische Universität Dresden, Dresden, Germany
| | - Ramcharan Singh Angom
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, Jacksonville, FL, USA
| | - Luana Ruta
- Department of Pharmaceutical Sciences, University of Perugia, via del Liceo 1, Perugia, Italy
| | - Elisa Bianconi
- Department of Pharmaceutical Sciences, University of Perugia, via del Liceo 1, Perugia, Italy
| | - Silvia Grottelli
- Department of Medicine and Surgery, University of Perugia, P.le L. Sevari 1, Perugia, Italy
| | | | | | - Steven R Lentz
- Department of Internal Medicine, The University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Sara Tommasi
- Department of Clinical Pharmacology, College of Medicine and Public Health, Flinders University and Flinders Medical Centre, Bedford Park, Adelaide, SA, Australia
| | | | - Toshiko Suzuki-Yamamoto
- Department of Nutritional Science, Faculty of Health and Welfare Science, Okayama Prefectural University, Okayama, Japan
| | - Masumi Kimoto
- Department of Nutritional Science, Faculty of Health and Welfare Science, Okayama Prefectural University, Okayama, Japan
| | - Elena Rubets
- Department of Internal Medicine III, Technische Universität Dresden, Dresden, Germany
| | - Sarah Chau
- Department of Cardiovascular Medicine, Mayo Clinic College of Medicine and Science, Rochester, NY, USA
| | - Yingjie Chen
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS, USA
| | - Xinli Hu
- Institute of Molecular Medicine, Beijing University, Beijing, China
| | - Nadine Bernhardt
- Department of Psychiatry and Psychotherapy, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Peter M Spieth
- Department of Anesthesiology and Critical Care Medicine, University Hospital Dresden, Technische Universität Dresden, Dresden, Germany
| | - Norbert Weiss
- Department of Internal Medicine III, Technische Universität Dresden, Dresden, Germany
| | - Stefan R Bornstein
- Department of Internal Medicine III, Technische Universität Dresden, Dresden, Germany
- School of Cardiovascular and Metabolic Medicine and Sciences, Faculty of Life Sciences & Medicine, King's College London, London, UK
| | - Debabrata Mukhopadhyay
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, Jacksonville, FL, USA
| | - Stefanie M Bode-Böger
- Institute of Clinical Pharmacology, Otto von Guericke University, Magdeburg, Germany
| | - Renke Maas
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- FAU New - Research Center for New Bioactive Compounds, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Ying Wang
- Department of Cardiovascular Medicine, Mayo Clinic College of Medicine and Science, Rochester, NY, USA
| | - Antonio Macchiarulo
- Department of Pharmaceutical Sciences, University of Perugia, via del Liceo 1, Perugia, Italy
| | - Arduino A Mangoni
- Department of Clinical Pharmacology, College of Medicine and Public Health, Flinders University and Flinders Medical Centre, Bedford Park, Adelaide, SA, Australia
| | - Barbara Cellini
- Department of Medicine and Surgery, University of Perugia, P.le L. Sevari 1, Perugia, Italy
| | - Roman N Rodionov
- Department of Internal Medicine III, Technische Universität Dresden, Dresden, Germany.
- College of Medicine and Public Health, Flinders University and Flinders Medical Center, Adelaide, SA, Australia.
| |
Collapse
|
5
|
Kumon H, Miyake Y, Yoshino Y, Iga JI, Tanaka K, Senba H, Kimura E, Higaki T, Matsuura B, Kawamoto R, Ueno SI. Functional AGXT2 SNP rs37369 Variant Is a Risk Factor for Diabetes Mellitus: Baseline Data From the Aidai Cohort Study in Japan. Can J Diabetes 2022; 46:829-834. [PMID: 35961823 DOI: 10.1016/j.jcjd.2022.06.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2022] [Revised: 05/16/2022] [Accepted: 06/13/2022] [Indexed: 01/06/2023]
Abstract
OBJECTIVES The relationship between alanine-glyoxylate aminotransferase 2 (AGXT2) single-nucleotide polymorphisms (SNPs) and diabetes mellitus (DM) has not been investigated. Therefore, we performed a case-control study to examine this relationship. METHODS The study subjects included 2,390 Japanese men and women aged 34 to 88 years. In total, 190 cases were defined as having a fasting plasma glucose level ≥126 mg/dL, having a glycated hemoglobin ≥6.5% or currently using diabetic medication. The 2,200 remaining participants served as control subjects. RESULTS Compared with study subjects with the CC genotype of AGXT2 SNP rs37369, those with the TT, but not CT, genotype had a significantly increased risk of DM: the adjusted odds ratio (OR) for the TT genotype was 1.83 (95% confidence interval [CI], 1.04 to 3.47). AGXT2 SNPs rs37370 and rs180749 were not significantly associated with the risk of DM. The CTA haplotype of rs37370, rs37369 and rs180749 was significantly positively associated with the risk of DM (crude OR, 1.25; 95% CI, 1.01 to 1.56), whereas the CCA haplotype was significantly inversely related to DM (crude OR, 0.53; 95% CI, 0.27 to 0.95). The multiplicative interaction between AGXT2 SNP rs37369 and smoking status with regard to the risk of DM was not significant (p=0.32 for interaction). CONCLUSIONS This is the first study to show significant associations between AGXT2 SNP rs37369, the CTA haplotype, and the CCA haplotype and DM. No interaction with regard to the risk of DM was observed between rs37369 and smoking.
Collapse
Affiliation(s)
- Hiroshi Kumon
- Department of Neuropsychiatry, Molecules and Function, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Yoshihiro Miyake
- Department of Epidemiology and Public Health, Ehime University Graduate School of Medicine, Ehime, Japan; Integrated Medical and Agricultural School of Public Health, Ehime University, Ehime, Japan; Research Promotion Unit, Translation Research Center, Ehime University Hospital, Ehime, Japan; Center for Data Science, Ehime University, Ehime, Japan
| | - Yuta Yoshino
- Department of Neuropsychiatry, Molecules and Function, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Jun-Ichi Iga
- Department of Neuropsychiatry, Molecules and Function, Ehime University Graduate School of Medicine, Ehime, Japan.
| | - Keiko Tanaka
- Department of Epidemiology and Public Health, Ehime University Graduate School of Medicine, Ehime, Japan; Integrated Medical and Agricultural School of Public Health, Ehime University, Ehime, Japan; Research Promotion Unit, Translation Research Center, Ehime University Hospital, Ehime, Japan; Center for Data Science, Ehime University, Ehime, Japan
| | - Hidenori Senba
- Department of Epidemiology and Public Health, Ehime University Graduate School of Medicine, Ehime, Japan; Department of Internal Medicine, Matsuyama Shimin Hospital, Ehime, Japan
| | - Eizen Kimura
- Integrated Medical and Agricultural School of Public Health, Ehime University, Ehime, Japan; Center for Data Science, Ehime University, Ehime, Japan; Department of Medical Informatics, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Takashi Higaki
- Department of Regional Pediatrics and Perinatology, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Bunzo Matsuura
- Department of Lifestyle-Related Medicine and Endocrinology, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Ryuichi Kawamoto
- Integrated Medical and Agricultural School of Public Health, Ehime University, Ehime, Japan; Department of Community Medicine, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Shu-Ichi Ueno
- Department of Neuropsychiatry, Molecules and Function, Ehime University Graduate School of Medicine, Ehime, Japan; Integrated Medical and Agricultural School of Public Health, Ehime University, Ehime, Japan
| |
Collapse
|
6
|
Yuan X, Cai L, Hu F, Xie L, Chen X, Wu J, Li Q. Evaluation of the predictive values of elevated serum L-homoarginine and dimethylarginines in preeclampsia. Amino Acids 2022; 54:1215-1227. [PMID: 35752997 PMCID: PMC9365731 DOI: 10.1007/s00726-022-03177-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 05/28/2022] [Indexed: 01/14/2023]
Abstract
L-homoarginine (hARG) is involved in nitric oxide biosynthesis, but its role and concentration in preeclampsia (PE) have not been fully revealed. The purpose of this study was to develop and validate a feasible clinical assay to quantify serum hARG, arginine (ARG), asymmetric (ADMA) and symmetric dimethylarginines (SDMA) levels by LC-MS/MS and investigate their differences at different stages of pregnancy with or without preeclampsia. Serum samples were collected from 84 pregnant women without complications (controls), 84 with mild preeclampsia (MPE), and 81 with severe preeclampsia (SPE) at various gestation stages (before the 20th week, during the 20th-28th week or after the 28th week of gestation). No significant difference in ARG levels was observed between PE and controls at any stage (P > 0.05). The serum hARG levels and hARG/ADMA ratios of MPE before the 20th week were higher than those of controls (P < 0.001). ADMA levels of MPE were higher than those of controls during the 20th-28th week (P < 0.01). SDMA levels of SPE were higher than those of MPE (P < 0.01) and controls (P < 0.05) after the 28th week. Elevated serum hARG before the 20th week was identified as an independent predictor for PE (OR = 1.478, 95% CI 1.120-1.950). ROC curve analysis showed serum hARG before the 20th week had a good potential to predict MPE (AUC = 0.875, 95% CI 0.759-0.948). In conclusion, our study indicated that elevated serum hARG and dimethylarginine levels detected by LC-MS/MS might serve as potential biomarkers for the early prediction of PE.
Collapse
Affiliation(s)
- Xiangmei Yuan
- Department of Laboratory Medicine, Wusong Branch, Zhongshan Hospital, Fudan University, Shanghai, 200940, China
| | - Leiming Cai
- Department of Laboratory Medicine, Wusong Branch, Zhongshan Hospital, Fudan University, Shanghai, 200940, China
| | - Fengmei Hu
- Shanghai AB Sciex Analytical Instrument Trading Co., Ltd., Shanghai, 200050, China
| | - Li Xie
- Department of Laboratory Medicine, Wusong Branch, Zhongshan Hospital, Fudan University, Shanghai, 200940, China
| | - Xiong Chen
- Department of Gynecology and Obstetrics, Wusong Branch, Zhongshan Hospital, Fudan University, Shanghai, 200940, China
| | - Jingjing Wu
- Department of Gynecology and Obstetrics, Wusong Branch, Zhongshan Hospital, Fudan University, Shanghai, 200940, China
| | - Qian Li
- Department of Laboratory Medicine, Wusong Branch, Zhongshan Hospital, Fudan University, Shanghai, 200940, China.
| |
Collapse
|
7
|
Szeliga M, Albrecht J. Roles of nitric oxide and polyamines in brain tumor growth. Adv Med Sci 2021; 66:199-205. [PMID: 33711670 DOI: 10.1016/j.advms.2021.02.006] [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: 11/27/2020] [Revised: 02/08/2021] [Accepted: 02/26/2021] [Indexed: 12/27/2022]
Abstract
Nitric oxide (NO) and polyamines: putrescine, spermidine and spermine, are key arginine metabolites in mammalian tissues that play critical roles i.a. in regulation of vascular tone (NO), and cell cycle regulation (polyamines). In the brain, both classes of molecules additionally have neuromodulatory and neuroprotective potential, and NO also a neurotoxic potential. Here we review evidence that brain tumors use the NO- and polyamine-synthesizing machineries to the benefit of their differentiation and growth from healthy glia and neurons. With a few exceptions, brain tumors show increased activities of one or all of the three arginine (Arg) to NO-converting nitric oxide synthase (NOS) isoforms (iNOS, eNOS, nNOS), but also elevated activities of polyamines-generating and modifying enzymes: arginase I/II, ornithine decarboxylase and spermidine/spermine N1-acetyltransferase. The degree of stimulation of NO- and polyamine synthesis often correlates with brain tumor malignancy. Excess NO, but also spermine, spermidine and their N1-acetylated forms, are tumor- and context-dependently involved in angiogenesis, tumor initiation and growth, and resistance to chemo- or radiotherapy. Hypothetically, increased demand for NO and/or polyamines is likely to contribute to Arg auxotrophy of malignant brain tumors, albeit the causal nexus awaits experimental verification.
Collapse
|
8
|
Dubé MP, Legault MA, Lemaçon A, Lemieux Perreault LP, Fouodjio R, Waters DD, Kouz S, Pinto FJ, Maggioni AP, Diaz R, Berry C, Koenig W, Lopez-Sendon J, Gamra H, Kiwan GS, Asselin G, Provost S, Barhdadi A, Sun M, Cossette M, Blondeau L, Mongrain I, Dubois A, Rhainds D, Bouabdallaoui N, Samuel M, de Denus S, L'Allier PL, Guertin MC, Roubille F, Tardif JC. Pharmacogenomics of the Efficacy and Safety of Colchicine in COLCOT. CIRCULATION-GENOMIC AND PRECISION MEDICINE 2021; 14:e003183. [PMID: 33560138 PMCID: PMC8284376 DOI: 10.1161/circgen.120.003183] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Supplemental Digital Content is available in the text. Background: The randomized, placebo-controlled COLCOT (Colchicine Cardiovascular Outcomes Trial) has shown the benefits of colchicine 0.5 mg daily to lower the rate of ischemic cardiovascular events in patients with a recent myocardial infarction. Here, we conducted a post hoc pharmacogenomic study of COLCOT with the aim to identify genetic predictors of the efficacy and safety of treatment with colchicine. Methods: There were 1522 participants of European ancestry from the COLCOT trial available for the pharmacogenomic study of COLCOT trial. The pharmacogenomic study’s primary cardiovascular end point was defined as for the main trial, as time to first occurrence of cardiovascular death, resuscitated cardiac arrest, myocardial infarction, stroke, or urgent hospitalization for angina requiring coronary revascularization. The safety end point was time to the first report of gastrointestinal events. Patients’ DNA was genotyped using the Illumina Global Screening array followed by imputation. We performed a genome-wide association study in colchicine-treated patients. Results: None of the genetic variants passed the genome-wide association study significance threshold for the primary cardiovascular end point conducted in 702 patients in the colchicine arm who were compliant to medication. The genome-wide association study for gastrointestinal events was conducted in all 767 patients in the colchicine arm and found 2 significant association signals, one with lead variant rs6916345 (hazard ratio, 1.89 [95% CI, 1.52–2.35], P=7.41×10−9) in a locus which colocalizes with Crohn disease, and one with lead variant rs74795203 (hazard ratio, 2.51 [95% CI, 1.82–3.47]; P=2.70×10−8), an intronic variant in gene SEPHS1. The interaction terms between the genetic variants and treatment with colchicine versus placebo were significant. Conclusions: We found 2 genomic regions associated with gastrointestinal events in patients treated with colchicine. Those findings will benefit from replication to confirm that some patients may have genetic predispositions to lower tolerability of treatment with colchicine.
Collapse
Affiliation(s)
- Marie-Pierre Dubé
- Montreal Heart Institute (M.-P.D., M.-A.L., A.L., L.-P.L.P., R.F., G.A., S.P., A.B., M.S., M.C., L.B., I.M., A.D., D.R., N.B., M.S., S.d.D., P.L.L., M.-C.G., J.-C.T.), Université de Montréal, Canada.,Université de Montréal Beaulieu-Saucier Pharmacogenomics Centre (M.-P.D., M.-A.L., A.L., L.-P.L.P., R.F., G.A., S.P., A.B., M.S., I.M., A.D., S.d.D.), Université de Montréal, Canada.,Department of Medicine (M.-P.D., A.L., M.S., N.B., M.S., J.-C.T.), Université de Montréal, Canada
| | - Marc-André Legault
- Montreal Heart Institute (M.-P.D., M.-A.L., A.L., L.-P.L.P., R.F., G.A., S.P., A.B., M.S., M.C., L.B., I.M., A.D., D.R., N.B., M.S., S.d.D., P.L.L., M.-C.G., J.-C.T.), Université de Montréal, Canada.,Université de Montréal Beaulieu-Saucier Pharmacogenomics Centre (M.-P.D., M.-A.L., A.L., L.-P.L.P., R.F., G.A., S.P., A.B., M.S., I.M., A.D., S.d.D.), Université de Montréal, Canada.,Departments of Biochemistry and Molecular Medicine, Faculty of Medicine (M.-A.L.), Université de Montréal, Canada
| | - Audrey Lemaçon
- Montreal Heart Institute (M.-P.D., M.-A.L., A.L., L.-P.L.P., R.F., G.A., S.P., A.B., M.S., M.C., L.B., I.M., A.D., D.R., N.B., M.S., S.d.D., P.L.L., M.-C.G., J.-C.T.), Université de Montréal, Canada.,Université de Montréal Beaulieu-Saucier Pharmacogenomics Centre (M.-P.D., M.-A.L., A.L., L.-P.L.P., R.F., G.A., S.P., A.B., M.S., I.M., A.D., S.d.D.), Université de Montréal, Canada.,Department of Medicine (M.-P.D., A.L., M.S., N.B., M.S., J.-C.T.), Université de Montréal, Canada
| | - Louis-Philippe Lemieux Perreault
- Montreal Heart Institute (M.-P.D., M.-A.L., A.L., L.-P.L.P., R.F., G.A., S.P., A.B., M.S., M.C., L.B., I.M., A.D., D.R., N.B., M.S., S.d.D., P.L.L., M.-C.G., J.-C.T.), Université de Montréal, Canada.,Université de Montréal Beaulieu-Saucier Pharmacogenomics Centre (M.-P.D., M.-A.L., A.L., L.-P.L.P., R.F., G.A., S.P., A.B., M.S., I.M., A.D., S.d.D.), Université de Montréal, Canada
| | - René Fouodjio
- Montreal Heart Institute (M.-P.D., M.-A.L., A.L., L.-P.L.P., R.F., G.A., S.P., A.B., M.S., M.C., L.B., I.M., A.D., D.R., N.B., M.S., S.d.D., P.L.L., M.-C.G., J.-C.T.), Université de Montréal, Canada.,Université de Montréal Beaulieu-Saucier Pharmacogenomics Centre (M.-P.D., M.-A.L., A.L., L.-P.L.P., R.F., G.A., S.P., A.B., M.S., I.M., A.D., S.d.D.), Université de Montréal, Canada
| | | | - Simon Kouz
- Centre Hospitalier Régional de Lanaudière, Joliette, Canada (S.K.)
| | - Fausto J Pinto
- Santa Maria University Hospital (CHULN), CAML, CCUL, Faculdade de Medicina da Universidade de Lisboa, Portugal (F.J.P.)
| | - Aldo P Maggioni
- Maria Cecilia Hospital, GVM Care and Research, Italy (A.P.M.)
| | - Rafael Diaz
- Estudios Clinicos Latinoamerica, Rosario, Argentina (R.D.)
| | - Colin Berry
- University of Glasgow, NHS Glasgow Clinical Research Facility, United Kingsom (C.B.)
| | - Wolfgang Koenig
- Deutsches Herzzentrum München, Technische Universität München, Munich, Germany (W.K.).,DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany (W.K.).,Institute of Epidemiology and Medical Biometry, University of Ulm, Germany (W.K.)
| | | | - Habib Gamra
- Fattouma Bourguiba University Hospital, Monastir, Tunisia (H.G.)
| | | | - Géraldine Asselin
- Montreal Heart Institute (M.-P.D., M.-A.L., A.L., L.-P.L.P., R.F., G.A., S.P., A.B., M.S., M.C., L.B., I.M., A.D., D.R., N.B., M.S., S.d.D., P.L.L., M.-C.G., J.-C.T.), Université de Montréal, Canada.,Université de Montréal Beaulieu-Saucier Pharmacogenomics Centre (M.-P.D., M.-A.L., A.L., L.-P.L.P., R.F., G.A., S.P., A.B., M.S., I.M., A.D., S.d.D.), Université de Montréal, Canada
| | - Sylvie Provost
- Montreal Heart Institute (M.-P.D., M.-A.L., A.L., L.-P.L.P., R.F., G.A., S.P., A.B., M.S., M.C., L.B., I.M., A.D., D.R., N.B., M.S., S.d.D., P.L.L., M.-C.G., J.-C.T.), Université de Montréal, Canada.,Université de Montréal Beaulieu-Saucier Pharmacogenomics Centre (M.-P.D., M.-A.L., A.L., L.-P.L.P., R.F., G.A., S.P., A.B., M.S., I.M., A.D., S.d.D.), Université de Montréal, Canada
| | - Amina Barhdadi
- Montreal Heart Institute (M.-P.D., M.-A.L., A.L., L.-P.L.P., R.F., G.A., S.P., A.B., M.S., M.C., L.B., I.M., A.D., D.R., N.B., M.S., S.d.D., P.L.L., M.-C.G., J.-C.T.), Université de Montréal, Canada.,Université de Montréal Beaulieu-Saucier Pharmacogenomics Centre (M.-P.D., M.-A.L., A.L., L.-P.L.P., R.F., G.A., S.P., A.B., M.S., I.M., A.D., S.d.D.), Université de Montréal, Canada
| | - Maxine Sun
- Montreal Heart Institute (M.-P.D., M.-A.L., A.L., L.-P.L.P., R.F., G.A., S.P., A.B., M.S., M.C., L.B., I.M., A.D., D.R., N.B., M.S., S.d.D., P.L.L., M.-C.G., J.-C.T.), Université de Montréal, Canada.,Université de Montréal Beaulieu-Saucier Pharmacogenomics Centre (M.-P.D., M.-A.L., A.L., L.-P.L.P., R.F., G.A., S.P., A.B., M.S., I.M., A.D., S.d.D.), Université de Montréal, Canada.,Department of Medicine (M.-P.D., A.L., M.S., N.B., M.S., J.-C.T.), Université de Montréal, Canada
| | - Mariève Cossette
- Montreal Heart Institute (M.-P.D., M.-A.L., A.L., L.-P.L.P., R.F., G.A., S.P., A.B., M.S., M.C., L.B., I.M., A.D., D.R., N.B., M.S., S.d.D., P.L.L., M.-C.G., J.-C.T.), Université de Montréal, Canada.,Montreal Health Innovation Coordinating Centre, Canada (M.C., L.B., M.-C.G.)
| | - Lucie Blondeau
- Montreal Heart Institute (M.-P.D., M.-A.L., A.L., L.-P.L.P., R.F., G.A., S.P., A.B., M.S., M.C., L.B., I.M., A.D., D.R., N.B., M.S., S.d.D., P.L.L., M.-C.G., J.-C.T.), Université de Montréal, Canada.,Montreal Health Innovation Coordinating Centre, Canada (M.C., L.B., M.-C.G.)
| | - Ian Mongrain
- Montreal Heart Institute (M.-P.D., M.-A.L., A.L., L.-P.L.P., R.F., G.A., S.P., A.B., M.S., M.C., L.B., I.M., A.D., D.R., N.B., M.S., S.d.D., P.L.L., M.-C.G., J.-C.T.), Université de Montréal, Canada.,Université de Montréal Beaulieu-Saucier Pharmacogenomics Centre (M.-P.D., M.-A.L., A.L., L.-P.L.P., R.F., G.A., S.P., A.B., M.S., I.M., A.D., S.d.D.), Université de Montréal, Canada
| | - Anick Dubois
- Montreal Heart Institute (M.-P.D., M.-A.L., A.L., L.-P.L.P., R.F., G.A., S.P., A.B., M.S., M.C., L.B., I.M., A.D., D.R., N.B., M.S., S.d.D., P.L.L., M.-C.G., J.-C.T.), Université de Montréal, Canada.,Université de Montréal Beaulieu-Saucier Pharmacogenomics Centre (M.-P.D., M.-A.L., A.L., L.-P.L.P., R.F., G.A., S.P., A.B., M.S., I.M., A.D., S.d.D.), Université de Montréal, Canada
| | - David Rhainds
- Montreal Heart Institute (M.-P.D., M.-A.L., A.L., L.-P.L.P., R.F., G.A., S.P., A.B., M.S., M.C., L.B., I.M., A.D., D.R., N.B., M.S., S.d.D., P.L.L., M.-C.G., J.-C.T.), Université de Montréal, Canada
| | - Nadia Bouabdallaoui
- Montreal Heart Institute (M.-P.D., M.-A.L., A.L., L.-P.L.P., R.F., G.A., S.P., A.B., M.S., M.C., L.B., I.M., A.D., D.R., N.B., M.S., S.d.D., P.L.L., M.-C.G., J.-C.T.), Université de Montréal, Canada.,Department of Medicine (M.-P.D., A.L., M.S., N.B., M.S., J.-C.T.), Université de Montréal, Canada
| | - Michelle Samuel
- Montreal Heart Institute (M.-P.D., M.-A.L., A.L., L.-P.L.P., R.F., G.A., S.P., A.B., M.S., M.C., L.B., I.M., A.D., D.R., N.B., M.S., S.d.D., P.L.L., M.-C.G., J.-C.T.), Université de Montréal, Canada.,Department of Medicine (M.-P.D., A.L., M.S., N.B., M.S., J.-C.T.), Université de Montréal, Canada
| | - Simon de Denus
- Montreal Heart Institute (M.-P.D., M.-A.L., A.L., L.-P.L.P., R.F., G.A., S.P., A.B., M.S., M.C., L.B., I.M., A.D., D.R., N.B., M.S., S.d.D., P.L.L., M.-C.G., J.-C.T.), Université de Montréal, Canada.,Université de Montréal Beaulieu-Saucier Pharmacogenomics Centre (M.-P.D., M.-A.L., A.L., L.-P.L.P., R.F., G.A., S.P., A.B., M.S., I.M., A.D., S.d.D.), Université de Montréal, Canada.,Université de Montréal, Faculty of Pharmacy, Canada (S.d.D.)
| | - Philippe L L'Allier
- Montreal Heart Institute (M.-P.D., M.-A.L., A.L., L.-P.L.P., R.F., G.A., S.P., A.B., M.S., M.C., L.B., I.M., A.D., D.R., N.B., M.S., S.d.D., P.L.L., M.-C.G., J.-C.T.), Université de Montréal, Canada
| | - Marie-Claude Guertin
- Montreal Heart Institute (M.-P.D., M.-A.L., A.L., L.-P.L.P., R.F., G.A., S.P., A.B., M.S., M.C., L.B., I.M., A.D., D.R., N.B., M.S., S.d.D., P.L.L., M.-C.G., J.-C.T.), Université de Montréal, Canada.,Montreal Health Innovation Coordinating Centre, Canada (M.C., L.B., M.-C.G.)
| | - François Roubille
- PhyMedExp (Physiologie et Médecine Expérimentale du Coeur et des Muscles), Université de Montpellier, INSERM, Centre National de la Recherche Scientifique, Cardiology Department, CHU de Montpellier, France (F.R.)
| | - Jean-Claude Tardif
- Montreal Heart Institute (M.-P.D., M.-A.L., A.L., L.-P.L.P., R.F., G.A., S.P., A.B., M.S., M.C., L.B., I.M., A.D., D.R., N.B., M.S., S.d.D., P.L.L., M.-C.G., J.-C.T.), Université de Montréal, Canada.,Department of Medicine (M.-P.D., A.L., M.S., N.B., M.S., J.-C.T.), Université de Montréal, Canada
| |
Collapse
|
9
|
Transport of L-Arginine Related Cardiovascular Risk Markers. J Clin Med 2020; 9:jcm9123975. [PMID: 33302555 PMCID: PMC7764698 DOI: 10.3390/jcm9123975] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 11/30/2020] [Accepted: 12/01/2020] [Indexed: 12/15/2022] Open
Abstract
L-arginine and its derivatives, asymmetric and symmetric dimethylarginine (ADMA and SDMA) and L-homoarginine, have emerged as cardiovascular biomarkers linked to cardiovascular outcomes and various metabolic and functional pathways such as NO-mediated endothelial function. Cellular uptake and efflux of L-arginine and its derivatives are facilitated by transport proteins. In this respect the cationic amino acid transporters CAT1 and CAT2 (SLC7A1 and SLC7A2) and the system y+L amino acid transporters (SLC7A6 and SLC7A7) have been most extensively investigated, so far, but the number of transporters shown to mediate the transport of L-arginine and its derivatives is constantly increasing. In the present review we assess the growing body of evidence regarding the function, expression, and clinical relevance of these transporters and their possible relation to cardiovascular diseases.
Collapse
|
10
|
Barlow JP, Karstoft K, Vigelsø A, Gram M, Helge JW, Dela F, Pappan K, O'Neil D, Dunn W, Solomon TPJ. Beta-aminoisobutyric acid is released by contracting human skeletal muscle and lowers insulin release from INS-1 832/3 cells by mediating mitochondrial energy metabolism. Metabol Open 2020; 7:100053. [PMID: 32924003 PMCID: PMC7479356 DOI: 10.1016/j.metop.2020.100053] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 08/12/2020] [Accepted: 08/13/2020] [Indexed: 01/01/2023] Open
Abstract
Aims/hypothesis This study aimed to examine if beta-aminoisobutyric acid (BAIBA) is (i) secreted by skeletal muscle in humans during exercise, (ii) associated with insulin secretory function in vivo, and (iii) directly linked with acute glucose-mediated insulin release by pancreatic beta cells in vitro. Methods Following 2-weeks of single-leg immobilization, plasma BAIBA concentrations were measured in the brachial artery and the femoral veins of each leg in healthy male subjects, at rest and during two-legged dynamic knee-extensor exercise. During a 2-h hyperglycamic clamp, insulin secretory function and levels of plasma BAIBA were assessed in non-diabetic individuals, non-diabetic individuals following 24-h hyperglycemia and patients with type 2 diabetes. Direct effects of BAIBA on acute glucose-mediated insulin release were probed in INS-1832/3 cells under normal and ‘diabetes-like’ conditions. Finally, the effect of BAIBA on mitochondrial function was assessed in INS-1832/3 cells using extracellular flux analysis. Results (i) BAIBA is released from skeletal muscle at rest and during exercise under healthy conditions but is suppressed during exercise following leg immobilization, (ii) plasma BAIBA concentrations inversely associate with insulin secretory function in humans, (iii) BAIBA lowers mitochondrial energy metabolism in INS-1 832/3 cells in parallel with decreased insulin secretion Conclusion/interpretation: BAIBA is a myokine released by skeletal muscle during exercise and indepedantly alters the triggering pathway of insulin secretion in cultured INS-1832/3 cells.
Collapse
Affiliation(s)
- Jonathan P Barlow
- School of Sport, Exercise, and Rehabilitation Sciences, College of Life and Environmental Sciences, University of Birmingham, Edgbaston, UK.,Mitochondrial Profiling Centre, College of Life and Environmental Sciences, University of Birmingham, Edgbaston, UK
| | - Kristian Karstoft
- Centre of Inflammation and Metabolism, Rigshospitalet, Copenhagen, Denmark.,Centre for Physical Activity Research, Rigshospitalet, Copenhagen, Denmark
| | - Andreas Vigelsø
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Martin Gram
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jørn W Helge
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Flemming Dela
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark.,Department of Geriatrics, Bispebjerg-Frederiksberg University Hospital, Bispebjerg, Denmark
| | | | - Donna O'Neil
- School of Biosciences and Phenome Centre Birmingham, College of Life and Environmental Sciences, University of Birmingham, Edgbaston, UK
| | - Warwick Dunn
- School of Biosciences and Phenome Centre Birmingham, College of Life and Environmental Sciences, University of Birmingham, Edgbaston, UK.,Institute for Metabolism and Systems Research, College of Medical Sciences, University of Birmingham, Edgbaston, UK
| | - Thomas P J Solomon
- School of Sport, Exercise, and Rehabilitation Sciences, College of Life and Environmental Sciences, University of Birmingham, Edgbaston, UK.,Institute for Metabolism and Systems Research, College of Medical Sciences, University of Birmingham, Edgbaston, UK
| |
Collapse
|
11
|
Zhai W, Lu H, Dong S, Fang J, Yu Z. Identification of potential key genes and key pathways related to clear cell renal cell carcinoma through bioinformatics analysis. Acta Biochim Biophys Sin (Shanghai) 2020; 52:853-863. [PMID: 32556097 DOI: 10.1093/abbs/gmaa068] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2019] [Revised: 10/17/2019] [Accepted: 12/26/2019] [Indexed: 12/16/2022] Open
Abstract
Clear cell renal cell carcinoma (ccRCC) is a common malignancy of the genitourinary system and is associated with high mortality rates. However, the molecular mechanism of ccRCC pathogenesis is still unclear, which translates to few effective diagnostic and prognostic biomarkers. In this study, we conducted a bioinformatics analysis on three Gene Expression Omnibus datasets and identified 437 differentially expressed genes (DEGs) related to ccRCC development and prognosis, of which 311 and 126 genes are respectively down-regulated and up-regulated. The protein-protein interaction network of these DEGs consists of 395 nodes and 1872 interactions and 2 prominent modules. The Staphylococcus aureus infection and complement and coagulation cascades are significantly enriched in module 1 and are likely involved in ccRCC progression. Forty-two hub genes were screened, of which von Willebrand factor, TIMP metallopeptidase inhibitor 1, plasminogen, formimidoyltransferase cyclodeaminase, solute carrier family 34 member 1, hydroxyacid oxidase 2, alanine-glyoxylate aminotransferase 2, phosphoenolpyruvate carboxykinase 1, and 3-hydroxy-3-methylglutaryl-CoA synthase 2 are possibly related to the prognosis of ccRCC. The differential expression of all nine genes was confirmed by quantitative real-time polymerase chain reaction analysis of the ccRCC and normal renal tissues. These key genes are potential biomarkers for the diagnosis and prognosis of ccRCC and warrant further investigation.
Collapse
Affiliation(s)
- Wenxin Zhai
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Haijiao Lu
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai 200000, China
| | - Shenghua Dong
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Jing Fang
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Zhuang Yu
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao 266000, China
| |
Collapse
|
12
|
Holton NW, Singhal M, Kumar A, Ticho AL, Manzella CR, Malhotra P, Jarava D, Saksena S, Dudeja PK, Alrefai WA, Gill RK. Hepatocyte nuclear factor-4α regulates expression of the serotonin transporter in intestinal epithelial cells. Am J Physiol Cell Physiol 2020; 318:C1294-C1304. [PMID: 32348179 PMCID: PMC7311735 DOI: 10.1152/ajpcell.00477.2019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 04/23/2020] [Accepted: 04/23/2020] [Indexed: 12/20/2022]
Abstract
The serotonin transporter (SERT) functions to regulate the availability of serotonin (5-HT) in the brain and intestine. An intestine-specific mRNA variant arising from a unique transcription start site and alternative promoter in the SERT gene has been identified (iSERT; spanning exon 1C). A decrease in SERT is implicated in several gut disorders, including inflammatory bowel diseases (IBD). However, little is known about mechanisms regulating the iSERT variant, and a clearer understanding is warranted for targeting SERT for the treatment of gut disorders. The current studies examined the expression of iSERT across different human intestinal regions and investigated its regulation by HNF4α (hepatic nuclear factor-4α), a transcription factor important for diverse cellular functions. iSERT mRNA abundance was highest in the human ileum and Caco-2 cell line. iSERT mRNA expression was downregulated by loss of HNF4α (but not HNF1α, HNF1β, or FOXA1) in Caco-2 cells. Overexpression of HNF4α increased iSERT mRNA concomitant with an increase in SERT protein. Progressive promoter deletion and site-directed mutagenesis revealed that the HNF4α response element spans nucleotides -1,163 to -1150 relative to the translation start site. SERT mRNA levels in the intestine were drastically reduced in the intestine-specific HNF4α-knockout mice relative to HNF4αFL/FL mice. Both HNF4α and SERT mRNA levels were also downregulated in mouse model of ileitis (SAMP) compared with AKR control mice. These results establish the transcriptional regulation of iSERT at the gut-specific internal promoter (hSERTp2) and have identified HNF4α as a critical modulator of basal SERT expression in the intestine.
Collapse
Affiliation(s)
- Nathaniel W Holton
- Division of Gastroenterology and Hepatology, University of Illinois at Chicago, Chicago, Illinois
| | - Megha Singhal
- Division of Gastroenterology and Hepatology, University of Illinois at Chicago, Chicago, Illinois
| | - Anoop Kumar
- Division of Gastroenterology and Hepatology, University of Illinois at Chicago, Chicago, Illinois
| | - Alexander L Ticho
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, Illinois
| | - Christopher R Manzella
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, Illinois
| | - Pooja Malhotra
- Division of Gastroenterology and Hepatology, University of Illinois at Chicago, Chicago, Illinois
| | - David Jarava
- Division of Gastroenterology and Hepatology, University of Illinois at Chicago, Chicago, Illinois
| | - Seema Saksena
- Division of Gastroenterology and Hepatology, University of Illinois at Chicago, Chicago, Illinois
- Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois
| | - Pradeep K Dudeja
- Division of Gastroenterology and Hepatology, University of Illinois at Chicago, Chicago, Illinois
- Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois
| | - Waddah A Alrefai
- Division of Gastroenterology and Hepatology, University of Illinois at Chicago, Chicago, Illinois
- Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois
| | - Ravinder K Gill
- Division of Gastroenterology and Hepatology, University of Illinois at Chicago, Chicago, Illinois
| |
Collapse
|
13
|
Jarzebska N, Georgi S, Jabs N, Brilloff S, Maas R, Rodionov RN, Zietz C, Montresor S, Hohenstein B, Weiss N. Kidney and liver are the main organs of expression of a key metabolic enzyme alanine:glyoxylate aminotransferase 2 in humans. ATHEROSCLEROSIS SUPP 2020; 40:106-112. [PMID: 31818439 DOI: 10.1016/j.atherosclerosissup.2019.08.041] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
BACKGROUND The metabolic syndrome is a cluster of cardiovascular risk factors and is highly predictive for development of cardiovascular diseases. An association between elevated plasma levels of the endogenous inhibitor of nitric oxide synthases asymmetric dimethylarginine (ADMA) and risk of cardiovascular diseases has been demonstrated in numerous epidemiological studies. ADMA can be catabolized by dimethylarginine dimethylaminohydrolase (DDAH) or metabolized through a much less understood alternative pathway by alanine:glyoxylate aminotransferase 2 (AGXT2) with the formation of α-keto-δ-(N,N-dimethylguanidino)valeric acid (ADGV). Previous RT-PCR and Western Blot studies suggested that Agxt2 is expressed in the mouse kidney and liver at comparable levels, while Northern Blot and in-situ RNA-hybridisation experiments demonstrated that the kidney is the main organ of Agxt2 expression in rats. Given this discrepancy, the goal of the current study was to analyse the expression of AGXT2 in human tissues. MATERIAL AND METHODS We analyzed AGXT2 expression in human tissues from a normal tissue bank by RT-PCR and further validated the results by Western Blot. We also performed immunohistochemical staining for AGXT2 and double fluorescent staining with an anti-AGXT2 antibody and a monoclonal anti-mitochondrial antibody. RESULTS We saw the strongest expression of AGXT2 in the kidney and liver and confirmed this results on protein level. By IHC staining we were able to show that AGXT2 is present in the convoluted tubule in the kidney and in the liver hepatocytes. The double fluorescent staining revealed mitochondrial localization of AGXT2. CONCLUSIONS Our current data suggest that both hepatocytes and kidney tubular epithelial cells are the major sources of AGXT2 in humans. We also demonstrated the mitochondrial localization of human AGXT2 enzyme.
Collapse
Affiliation(s)
- Natalia Jarzebska
- University Center for Vascular Medicine & Department of Medicine III - Section Angiology, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany; Department of Anesthesiology and Intensive Care Medicine, Pulmonary Engineering Group, University Hospital Carl Gustav Carus, Technische Universität Dresden, Germany
| | - Sophia Georgi
- University Center for Vascular Medicine & Department of Medicine III - Section Angiology, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Normund Jabs
- University Center for Vascular Medicine & Department of Medicine III - Section Angiology, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Silke Brilloff
- University Center for Vascular Medicine & Department of Medicine III - Section Angiology, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Renke Maas
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Roman N Rodionov
- University Center for Vascular Medicine & Department of Medicine III - Section Angiology, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Christian Zietz
- Institute of Pathology, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Sabrina Montresor
- University Center for Vascular Medicine & Department of Medicine III - Section Angiology, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Bernd Hohenstein
- Division of Nephrology, Department of Internal Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Norbert Weiss
- University Center for Vascular Medicine & Department of Medicine III - Section Angiology, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.
| |
Collapse
|
14
|
Early Transcriptional Changes within Liver, Adrenal Gland, and Lymphoid Tissues Significantly Contribute to Ebola Virus Pathogenesis in Cynomolgus Macaques. J Virol 2020; 94:JVI.00250-20. [PMID: 32213610 DOI: 10.1128/jvi.00250-20] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Accepted: 03/11/2020] [Indexed: 01/05/2023] Open
Abstract
Ebola virus (EBOV) continues to pose a significant threat to human health, as evidenced by the 2013-2016 epidemic in West Africa and the ongoing outbreak in the Democratic Republic of the Congo. EBOV causes hemorrhagic fever, organ damage, and shock culminating in death, with case fatality rates as high as 90%. This high lethality combined with the paucity of licensed medical countermeasures makes EBOV a critical human pathogen. Although EBOV infection results in significant damage to the liver and the adrenal glands, little is known about the molecular signatures of injury in these organs. Moreover, while changes in peripheral blood cells are becoming increasingly understood, the host responses within organs and lymphoid tissues remain poorly characterized. To address this knowledge gap, we tracked longitudinal transcriptional changes in tissues collected from EBOV-Makona-infected cynomolgus macaques. Following infection, both liver and adrenal glands exhibited significant and early downregulation of genes involved in metabolism, coagulation, hormone synthesis, and angiogenesis; upregulated genes were associated with inflammation. Analysis of lymphoid tissues showed early upregulation of genes that play a role in innate immunity and inflammation and downregulation of genes associated with cell cycle and adaptive immunity. Moreover, transient activation of innate immune responses and downregulation of humoral immune responses in lymphoid tissues were confirmed with flow cytometry. Together, these data suggest that the liver, adrenal gland, and lymphatic organs are important sites of EBOV infection and that dysregulating the function of these vital organs contributes to the development of Ebola virus disease.IMPORTANCE Ebola virus (EBOV) remains a high-priority pathogen since it continues to cause outbreaks with high case fatality rates. Although it is well established that EBOV results in severe organ damage, our understanding of tissue injury in the liver, adrenal glands, and lymphoid tissues remains limited. We begin to address this knowledge gap by conducting longitudinal gene expression studies in these tissues, which were collected from EBOV-infected cynomolgus macaques. We report robust and early gene expression changes within these tissues, indicating they are primary sites of EBOV infection. Furthermore, genes involved in metabolism, coagulation, and adaptive immunity were downregulated, while inflammation-related genes were upregulated. These results indicate significant tissue damage consistent with the development of hemorrhagic fever and lymphopenia. Our study provides novel insight into EBOV-host interactions and elucidates how host responses within the liver, adrenal glands, and lymphoid tissues contribute to EBOV pathogenesis.
Collapse
|
15
|
Protein Arginine Methyltransferases in Cardiovascular and Neuronal Function. Mol Neurobiol 2019; 57:1716-1732. [PMID: 31823198 DOI: 10.1007/s12035-019-01850-z] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Accepted: 12/01/2019] [Indexed: 12/16/2022]
Abstract
The methylation of arginine residues by protein arginine methyltransferases (PRMTs) is a type of post-translational modification which is important for numerous cellular processes, including mRNA splicing, DNA repair, signal transduction, protein interaction, and transport. PRMTs have been extensively associated with various pathologies, including cancer, inflammation, and immunity response. However, the role of PRMTs has not been well described in vascular and neurological function. Aberrant expression of PRMTs can alter its metabolic products, asymmetric dimethylarginine (ADMA), and symmetric dimethylarginine (SDMA). Increased ADMA levels are recognized as an independent risk factor for cardiovascular disease and mortality. Recent studies have provided considerable advances in the development of small-molecule inhibitors of PRMTs to study their function under normal and pathological states. In this review, we aim to elucidate the particular roles of PRMTs in vascular and neuronal function as a potential target for cardiovascular and neurological diseases.
Collapse
|
16
|
Chai X, Yan J, Gao Y, Jin J. Endothelial HNF4α potentiates angiogenic dysfunction via enhancement of vascular endothelial growth factor resistance in T2DM. J Cell Biochem 2019; 120:12989-13000. [PMID: 30873661 DOI: 10.1002/jcb.28570] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Revised: 12/18/2018] [Accepted: 01/02/2019] [Indexed: 12/24/2022]
Abstract
Although both hyperprocoagulant status, characterized by elevated thrombin levels, and vascular endothelial growth factor (VEGF) resistance, marked by attenuated expression of VEGFR2 (also called FLK1 or KDR), are known to contribute importantly to an increased risk of vascular events in diabetes mellitus type 2 (T2DM), it remains obscure whether these two biological events regulate angiogenic response in a coordinated manner. We show here that endothelial expression of hepatocyte nuclear factor 4α (HNF4α) was significantly upregulated in rodents and humans with T2DM, and HNF4α upregulation by thrombin was dependent on activation of multiple pathways, including protein kinase B, c-Jun N-terminal kinase, p38, oxidative stress, protein kinase C, and AMPK (5'-adenosine monophosphate (AMP)-activated protein kinase). Functionally, HNF4α inhibited VEGF-mediated endothelial proliferation and migration, and blunted VEGF-stimulated in vitro angiogenesis, thus rendering endothelial cells unresponsive to established angiogenic VEGF stimulation. Mechanistically, HNF4α potentiated the endothelial VEGF resistance through the direct transcriptional repression of FLK1 gene. From a therapeutic standpoint, overexpression of the exogenous FLK1 successfully rescued HNF4α-inhibited angiogenic response to VEGF and potentiated VEGF-stimulated in vitro tube formation. Considering a strong association between HNF4A deregulation and increased risk of T2DM, our findings suggest that HNF4α may act as a critical converging point linking hyperprocoagulant condition to VEGF resistance in diabetic ECs, and repression of FLK1 expression by thrombin-induced HNF4α mediates, at least partially, the vascular dysfunction caused by T2DM.
Collapse
Affiliation(s)
- Xubing Chai
- Department of Endocrinology, Xi'an Institute of Rheumatolog, Xi'an No. 5 Hospital, Xi'an, Shaanxi, China
| | - Jun Yan
- Department of Endocrinology, Xi'an Institute of Rheumatolog, Xi'an No. 5 Hospital, Xi'an, Shaanxi, China
| | - Yaya Gao
- Department of Endocrinology, Xi'an Institute of Rheumatolog, Xi'an No. 5 Hospital, Xi'an, Shaanxi, China
| | - Jing Jin
- Department of Geriatric, Xi'an Institute of Rheumatolog, Xi'an No. 5 Hospital, Xi'an, Shaanxi, China
| |
Collapse
|
17
|
Meeting report of the "Symposium on kidney stones and mineral metabolism: calcium kidney stones in 2017". J Nephrol 2019; 32:681-698. [PMID: 30680550 DOI: 10.1007/s40620-019-00587-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Accepted: 01/16/2019] [Indexed: 02/06/2023]
Abstract
A symposium on kidney stones and mineral metabolism held on December 2017 in Brussels, Belgium was the first international multidisciplinary conference of the International Collaborative Network on Kidney Stones and Mineral Metabolism. This meeting addressed epidemiology, underlying pathophysiological mechanisms, genetics, pathological, as well as clinical and research topics. The participants included clinicians and recognized experts in the field from Europe and the United States interacted closely during the symposium which promoted a chance to explore new frontiers in the field of kidney stone disease. This manuscript summarizes some of the major highlights of the meeting.
Collapse
|
18
|
Maas R, Mieth M, Titze SI, Hübner S, Fromm MF, Kielstein JT, Schmid M, Köttgen A, Kronenberg F, Krane V, Hausknecht B, Eckardt KU, Schneider MP. Drugs linked to plasma homoarginine in chronic kidney disease patients—a cross-sectional analysis of the German Chronic Kidney Disease cohort. Nephrol Dial Transplant 2018; 35:1187-1195. [DOI: 10.1093/ndt/gfy342] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Accepted: 09/25/2018] [Indexed: 01/16/2023] Open
Abstract
Abstract
Background
Elevated plasma concentrations of symmetric and asymmetric dimethylarginine (SDMA and ADMA, respectively) and a lower plasma concentration of the structurally related homoarginine are commonly observed in patients with chronic kidney disease (CKD) and independently predict total mortality as well as progression of renal disease. We aimed to identify drugs that may alter this adverse metabolite pattern in a favourable fashion.
Methods
Plasma ADMA, SDMA, homoarginine and l-arginine were determined by liquid chromatography–tandem mass spectrometry in 4756 CKD patients ages 18–74 years with an estimated glomerular filtration rate (eGFR) of 30–60 mL/min/1.73 m2 or an eGFR >60 mL/min/1.73 m2 and overt proteinuria who were enrolled in the German Chronic Kidney Disease (GCKD) study. Associations between laboratory, clinical and medication data were assessed.
Results
Intake of several commonly used drugs was independently associated with plasma concentrations of homoarginine and/or related metabolites. Among these, the peroxisome proliferator-activated receptor alpha (PPAR-α) agonist fenofibrate was associated with the most profound differences in ADMA, SDMA and homoarginine plasma concentrations: 66 patients taking fenofibrate had a multivariable adjusted odds ratio (OR) of 5.83 [95% confidence interval (CI) 2.82–12.03, P < 0.001] to have a plasma homoarginine concentration above the median. The median homoarginine plasma concentration in patients taking fenofibrate was 2.30 µmol/L versus 1.55 in patients not taking the drug (P < 0.001). In addition, fibrates were significantly associated with lower plasma SDMA and higher l-arginine concentrations. In contrast, glucocorticoids were associated with lower plasma homoarginine, with adjusted ORs of 0.52 (95% CI 0.40–0.67, P < 0.001) and 0.53 (95% CI 0.31–0.90, P = 0.018) for prednisolone and methylprednisolone, respectively.
Conclusions
In a large cohort of CKD patients, intake of fenofibrate and glucocorticoids were independently associated with higher and lower plasma homoarginine concentrations, respectively. Effects on plasma homoarginine and methylarginines warrant further investigation as potential mechanisms mediating beneficial or adverse drug effects.
Collapse
Affiliation(s)
- Renke Maas
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Maren Mieth
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Stephanie I Titze
- Department of Nephrology and Hypertension, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Silvia Hübner
- Department of Nephrology and Hypertension, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Martin F Fromm
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Jan T Kielstein
- Divison of Nephrology, Medical School Hannover, Hannover, Germany
- Medical Clinic V Nephrology Rheumatology Blood Purification, Klinikum Braunschweig, Braunschweig, Germany
| | - Matthias Schmid
- Department of Medical Biometry, Informatics and Epidemiology, University Hospital Bonn, Bonn, Germany
| | - Anna Köttgen
- Institute of Genetic Epidemiology, Faculty of Medicine and Medical Centre, University of Freiburg, Freiburg, Germany
| | - Florian Kronenberg
- Division of Genetic Epidemiology, Department of Medical Genetics, Molecular and Clinical Pharmacology, Medical University of Innsbruck, Innsbruck, Austria
| | - Vera Krane
- Department of Medicine I, Division of Nephrology, University Hospital Würzburg, Würzburg, Germany
| | - Birgit Hausknecht
- Department of Nephrology and Hypertension, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Kai-Uwe Eckardt
- Department of Nephrology and Hypertension, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Department of Nephrology and Medical Intensive Care, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Markus P Schneider
- Department of Nephrology and Hypertension, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
19
|
Hu XL, Li MP, Song PY, Tang J, Chen XP. AGXT2: An unnegligible aminotransferase in cardiovascular and urinary systems. J Mol Cell Cardiol 2017; 113:33-38. [PMID: 28970090 DOI: 10.1016/j.yjmcc.2017.09.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2017] [Revised: 09/22/2017] [Accepted: 09/27/2017] [Indexed: 01/07/2023]
Abstract
Cardiovascular diseases (CVDs) and renal impairment interact in a complex and interdependent manner, which makes clarification of possible pathogenesis between CVDs and renal diseases very challenging and important. There is increasing evidence showing that both asymmetric dimethylarginine (ADMA) and symmetric dimethylarginine (SDMA) play a crucial role in the development of CVDs as well as in the prediction of cardiovascular events. Also, the plasma levels of ADMA and SDMA were reported to be significantly associated with renal function. Alanine-glyoxylate aminotransferase 2 (AGXT2) is reported to be involved in ADMA and SDMA metabolism, thus deficiency in the expression or activity of AGXT2 may play a part in the progression of cardiovascular or renal diseases through affecting ADMA/SDMA levels. Here, we focused our attention on AGXT2 and discussed its potential impact on CVDs and renal diseases. Meanwhile, the review also summarized the functions and recent advances of AGXT2, as well as the clinical association studies of AGXT2 in cardiovascular and urinary systems, which might arouse the interest of researchers in these fields.
Collapse
Affiliation(s)
- Xiao-Lei Hu
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, China; Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha 410078, China
| | - Mu-Peng Li
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, China; Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha 410078, China
| | - Pei-Yuan Song
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, China; Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha 410078, China
| | - Jie Tang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, China; Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha 410078, China.
| | - Xiao-Ping Chen
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, China; Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha 410078, China.
| |
Collapse
|
20
|
Establishment of human retinal mitoscriptome gene expression signature for diabetic retinopathy using cadaver eyes. Mitochondrion 2017; 36:150-181. [PMID: 28729194 DOI: 10.1016/j.mito.2017.07.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Revised: 07/09/2017] [Accepted: 07/14/2017] [Indexed: 11/20/2022]
Abstract
Diabetic retinopathy (DR) is a leading cause of blindness due to retinal microvasculature. We used microarray analysis for the first time to establish the retinal mitoscriptome gene expression signature for DR using human cadaver eyes. Among the 1042 genes, 60 (52-down, 8-up) and 39 (36-down, 3-up) genes were differentially expressed in the DR as compared to normal control and diabetic retinas respectively. These genes were mainly responsible for regulating angiogenesis, anti-oxidant defense mechanism, ATP production and apoptosis contributing to the disease pathology of DR. These findings might be useful for the discovery of biomarker and developing therapeutic regimen.
Collapse
|