1
|
Vanden Broecke E, Stammeleer L, Stock E, De Paepe E, Daminet S. Efficacy of Urine Asymmetric Dimethylarginine Concentration to Predict Azotemia in Hyperthyroid Cats After Radio-Iodine Treatment. J Vet Intern Med 2025; 39:e70096. [PMID: 40271736 PMCID: PMC12019304 DOI: 10.1111/jvim.70096] [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: 11/13/2024] [Revised: 03/26/2025] [Accepted: 04/01/2025] [Indexed: 04/25/2025] Open
Abstract
BACKGROUND Hyperthyroidism can mask concurrent chronic kidney disease in cats, and no accurate biomarkers are available to predict which cats will develop renal azotemia after radioiodine (131I) treatment. HYPOTHESIS/OBJECTIVES To evaluate the potential of serum and urinary metabolites and metabolite ratios to predict post-131I renal azotemia in hyperthyroid cats. ANIMALS Hyperthyroid cats (n = 31), before and (3-12 months) after treatment with 131I at the Faculty of Veterinary Medicine (Ghent University, Belgium). METHODS Retrospective study. Optimized and validated feline extraction and analysis protocols were employed for metabolic profiling of urine and serum samples using ultra-high performance liquid chromatography-high-resolution mass spectrometry. A dual strategy of cross-validated univariate and penalized multivariate logistic regression was applied to determine predictivity (i.e., area under the curve [AUC], accuracy, sensitivity, and specificity) of individual biomarkers and panels. RESULTS All hyperthyroid cats were non-azotemic before 131I administration. After 131I treatment, 7 cats became persistently (≥ 2 timepoints) azotemic while 24 remained non-azotemic. Urinary asymmetric dimethylarginine (ADMA) was identified as a pivotal predictor of post-131I azotemia in both univariate and multivariate modeling. When employed as a standalone biomarker, an AUC of 0.851, accuracy of 0.903, sensitivity of 0.714, and specificity of 0.958 were achieved. While pre-treatment USG was significantly different (P = 0.002) between both groups, it did not show enhanced prediction over ADMA, nor in multivariate modeling. CONCLUSIONS AND CLINICAL IMPORTANCE Urinary ADMA can accurately predict post-131I azotemia in hyperthyroid cats becoming euthyroid after 131I treatment. These findings can aid clinicians in managing owner expectations and modify treatment plans.
Collapse
Affiliation(s)
- Ellen Vanden Broecke
- Small Animal DepartmentFaculty of Veterinary Medicine, Ghent UniversityMerelbekeBelgium
- Laboratory of Integrative Metabolomics (LIMET), Department of Translational Physiology, Infectiology and Public HealthFaculty of Veterinary Medicine, Ghent UniversityMerelbekeBelgium
| | - Lisa Stammeleer
- Small Animal DepartmentFaculty of Veterinary Medicine, Ghent UniversityMerelbekeBelgium
| | - Emmelie Stock
- Department of Morphology, Imaging, Orthopedics, Rehabilitation and NutritionFaculty of Veterinary Medicine, Ghent UniversityMerelbekeBelgium
| | - Ellen De Paepe
- Laboratory of Integrative Metabolomics (LIMET), Department of Translational Physiology, Infectiology and Public HealthFaculty of Veterinary Medicine, Ghent UniversityMerelbekeBelgium
| | - Sylvie Daminet
- Small Animal DepartmentFaculty of Veterinary Medicine, Ghent UniversityMerelbekeBelgium
| |
Collapse
|
2
|
Sedding D, Schmidt TM, Bähre H, Bavendiek U, Casas AI, Chen S, Thao-Vi Dao V, Elbatreek MH, Gutzki F, Hahn A, Kleikers P, Krahn T, Macchiusi C, Martin C, Mucke H, Nogales C, Schmidt BML, Seifert R, Sonnenschein K, Tongers J, Thol J, van der Arend I, van Kuijk SMJ, Wingler K, Wu M, Bauersachs J, McGrath B, Schmidt HHHW. Nutritional L-Citrulline and Tetrahydrobiopterin in Peripheral Artery Disease: A Phase II Randomized Trial (CIPER Study). JACC. ADVANCES 2025; 4:101590. [PMID: 39985883 PMCID: PMC11904498 DOI: 10.1016/j.jacadv.2025.101590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 12/19/2024] [Accepted: 12/31/2024] [Indexed: 02/24/2025]
Abstract
BACKGROUND Peripheral artery disease (PAD) is a major public health concern due to its high prevalence, severe impact on individuals' health and quality of life, and substantial economic burden. Pharmacological interventions are still limited with numbers needed-to-treat ranging from 6 (cilostazol) to 50 (aspirin, statins, and vorapaxar). OBJECTIVES This randomized, placebo-controlled, double-blinded crossover interventional trial aims to measure the effect of L-citrulline and tetrahydrobiopterin (H4Bip) on walking distance in patients with PAD, stratified by plasma levels of asymmetric dimethyl L-arginine (ADMA), the endogenous inhibitor of endothelial nitric oxide (NO) synthase. METHODS We measured preinterventional ADMA levels in 51 patients with PAD in Australia and Germany with mean changes in absolute claudication distance (dACD) as the primary outcome upon orally supplementing the L-arginine precursor, L-citrulline (3 g) twice daily for 12 weeks, and, in one arm, additionally H4Bip (0.45 g) once per day for a further 2 weeks. RESULTS Preinterventional ADMA levels were pathological (>0.4 μM) in 34 patients. Supplementation with L-citrulline significantly increased the mean plasma levels of both L-citrulline and L-arginine, from 41.8 ± 2.7 μmol/l to 246.3 ± 67.3 μmol/l (P = 0.004) and from 75.2 ± 4.2 μmol/l to 119.2 ± 6.9 μmol/l (P < 0.0001) respectively, when compared with placebo. dACD in % of control was significantly improved by L-citrulline vs placebo (20.11% ± 4.50% vs 5.73% ± 2.74%, respectively; P = 0.011). Further addition of H4Bip increased the mean percentage dACD to 28.15% ± 6.84% (P = 0.021), but only in patients with preinterventional pathological ADMA levels. CONCLUSIONS L-citrulline and, when ADMA levels are pathological, H4Bip are effective nutritional interventions in patients with PAD warranting further confirmatory trials.
Collapse
Affiliation(s)
- Daniel Sedding
- Klinik für Kardiologie, Universitätsklinikum Halle, Halle, Germany; Klinik für Kardiologie und Angiologie, Medizinische Hochschule Hannover, Hannover, Germany
| | - Tim M Schmidt
- Klinik für Kardiologie und Angiologie, Medizinische Hochschule Hannover, Hannover, Germany; Department of Pharmacology and Personalised Medicine, MeHNS, FHML, Maastricht University, Maastricht, the Netherlands
| | - Heike Bähre
- Research Core Unit Metabolomics, Medizinische Hochschule Hannover, Hannover, Germany
| | - Udo Bavendiek
- Klinik für Kardiologie und Angiologie, Medizinische Hochschule Hannover, Hannover, Germany
| | - Ana I Casas
- Department of Pharmacology and Personalised Medicine, MeHNS, FHML, Maastricht University, Maastricht, the Netherlands
| | - Suzi Chen
- Monash Medical Centre, Monash University, Clayton, Melbourne, Australia
| | - Vu Thao-Vi Dao
- Department of Pharmacology and Personalised Medicine, MeHNS, FHML, Maastricht University, Maastricht, the Netherlands
| | - Mahmoud H Elbatreek
- Department of Pharmacology and Personalised Medicine, MeHNS, FHML, Maastricht University, Maastricht, the Netherlands; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
| | - Frank Gutzki
- Research Core Unit Metabolomics, Medizinische Hochschule Hannover, Hannover, Germany
| | - Andreas Hahn
- Leibniz Universität Hannover, Institut für Lebensmittelwissenschaft und Humanernährung, Hannover, Germany
| | - Pamela Kleikers
- Department of Pharmacology and Personalised Medicine, MeHNS, FHML, Maastricht University, Maastricht, the Netherlands
| | - Thomas Krahn
- Department of Pharmacology and Personalised Medicine, MeHNS, FHML, Maastricht University, Maastricht, the Netherlands
| | - Cecilia Macchiusi
- Department of Pharmacology and Personalised Medicine, MeHNS, FHML, Maastricht University, Maastricht, the Netherlands; Department of Biostatistics, Sapienza University, Rome, Italy
| | - Catherine Martin
- Monash Medical Centre, Monash University, Clayton, Melbourne, Australia
| | | | - Cristian Nogales
- Department of Pharmacology and Personalised Medicine, MeHNS, FHML, Maastricht University, Maastricht, the Netherlands
| | - Beate M L Schmidt
- Monash Medical Centre, Monash University, Clayton, Melbourne, Australia
| | - Roland Seifert
- Research Core Unit Metabolomics, Medizinische Hochschule Hannover, Hannover, Germany
| | - Kristina Sonnenschein
- Klinik für Kardiologie und Angiologie, Medizinische Hochschule Hannover, Hannover, Germany
| | - Jörn Tongers
- Klinik für Kardiologie und Angiologie, Medizinische Hochschule Hannover, Hannover, Germany
| | - Jens Thol
- Klinik für Kardiologie und Angiologie, Medizinische Hochschule Hannover, Hannover, Germany
| | - Inés van der Arend
- Department of Pharmacology and Personalised Medicine, MeHNS, FHML, Maastricht University, Maastricht, the Netherlands
| | - Sander M J van Kuijk
- Clinical Epidemiology and Medical Technology Assessment (KEMTA), Maastricht UMC+, Maastricht, the Netherlands
| | - Kirstin Wingler
- Department of Pharmacology and Personalised Medicine, MeHNS, FHML, Maastricht University, Maastricht, the Netherlands
| | - Michael Wu
- Monash Medical Centre, Monash University, Clayton, Melbourne, Australia
| | - Johann Bauersachs
- Klinik für Kardiologie und Angiologie, Medizinische Hochschule Hannover, Hannover, Germany
| | - Barry McGrath
- Monash Medical Centre, Monash University, Clayton, Melbourne, Australia
| | - Harald H H W Schmidt
- Department of Pharmacology and Personalised Medicine, MeHNS, FHML, Maastricht University, Maastricht, the Netherlands; Monash Medical Centre, Monash University, Clayton, Melbourne, Australia.
| |
Collapse
|
3
|
Landowska M, Kałuża B, Watała C, Babula E, Żuk-Łapan A, Woźniak K, Kargul A, Jurek J, Korcz T, Cicha-Brzezińska M, Franek E. Is Insulin Resistance an Independent Predictor of Atherosclerosis? J Clin Med 2025; 14:969. [PMID: 39941639 PMCID: PMC11818120 DOI: 10.3390/jcm14030969] [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: 12/20/2024] [Revised: 01/24/2025] [Accepted: 01/30/2025] [Indexed: 02/16/2025] Open
Abstract
Background: Insulin resistance (IR) is a condition that precedes the onset of type 2 diabetes mellitus (T2DM), which is regarded as an established risk factor for atherosclerosis (AS). Considering that the same metabolic changes as those caused by IR are evidenced to promote the development of AS, we investigated whether IR estimated by the homeostasis model assessment of IR (HOMA-IR) could predict the occurrence of preclinical AS. Methods: The study participants were divided into two groups based on the presence of IR diagnosed during the baseline hospitalization and defined as a HOMA-IR value equal to or higher than 2.5. After a follow-up period of at least four years, a total of 79 (n = 79) were prospectively assessed in terms of the presence of preclinical AS, determined by either an abnormally low ankle-brachial index (ABI) (ABI < 0.9) or an increased carotid intima media thickness (CIMT) (CIMT > 1 mm). Results: Using the multivariate logistic regression analysis, it was demonstrated that the HOMA-IR was associated with an abnormally low ABI (odds ratio: 1.609, 95% confidence interval (CI): [1.041-2.487], p = 0.032). The Cox regression model revealed that the HOMA-IR was a predictor of both an abnormal ABI (hazard ratio: 1.435, CI: [1.076-1.913], p = 0.014) and increased CIMT (hazard ratio: 1.419, CI: [1.033-1.948], p = 0.031), independently of age, sex, dyslipidemia, smoking, triglycerides (TG), low-density lipoproteins (LDL), high-density lipoproteins (HDL), and total cholesterol levels. Conclusions: IR, as estimated by the HOMA-IR, may be considered as a predictor of preclinical AS, independently of cardiovascular risk factors.
Collapse
Affiliation(s)
- Małgorzata Landowska
- Department of Internal Medicine, Endocrinology and Diabetology, National Medical Institute of the Ministry of the Interior and Administration in Warsaw, 02-507 Warsaw, Poland; (M.L.); (E.F.)
- Students Scientific Group of the Medical University of Warsaw at the Department of Internal Medicine, Endocrinology and Diabetology, National Medical Institute of the Ministry of the Interior and Administration in Warsaw, 02-507 Warsaw, Poland; (E.B.); (A.Ż.-Ł.); (K.W.); (A.K.); (J.J.)
| | - Bernadetta Kałuża
- Department of Internal Medicine, Endocrinology and Diabetology, National Medical Institute of the Ministry of the Interior and Administration in Warsaw, 02-507 Warsaw, Poland; (M.L.); (E.F.)
- Students Scientific Group of the Medical University of Warsaw at the Department of Internal Medicine, Endocrinology and Diabetology, National Medical Institute of the Ministry of the Interior and Administration in Warsaw, 02-507 Warsaw, Poland; (E.B.); (A.Ż.-Ł.); (K.W.); (A.K.); (J.J.)
| | - Cezary Watała
- Department of Haemostasis and Haemostatic Disorders, Medical University of Lodz, 92-215 Lodz, Poland;
| | - Emilia Babula
- Students Scientific Group of the Medical University of Warsaw at the Department of Internal Medicine, Endocrinology and Diabetology, National Medical Institute of the Ministry of the Interior and Administration in Warsaw, 02-507 Warsaw, Poland; (E.B.); (A.Ż.-Ł.); (K.W.); (A.K.); (J.J.)
| | - Aleksandra Żuk-Łapan
- Students Scientific Group of the Medical University of Warsaw at the Department of Internal Medicine, Endocrinology and Diabetology, National Medical Institute of the Ministry of the Interior and Administration in Warsaw, 02-507 Warsaw, Poland; (E.B.); (A.Ż.-Ł.); (K.W.); (A.K.); (J.J.)
| | - Kinga Woźniak
- Students Scientific Group of the Medical University of Warsaw at the Department of Internal Medicine, Endocrinology and Diabetology, National Medical Institute of the Ministry of the Interior and Administration in Warsaw, 02-507 Warsaw, Poland; (E.B.); (A.Ż.-Ł.); (K.W.); (A.K.); (J.J.)
| | - Aleksandra Kargul
- Students Scientific Group of the Medical University of Warsaw at the Department of Internal Medicine, Endocrinology and Diabetology, National Medical Institute of the Ministry of the Interior and Administration in Warsaw, 02-507 Warsaw, Poland; (E.B.); (A.Ż.-Ł.); (K.W.); (A.K.); (J.J.)
| | - Jonasz Jurek
- Students Scientific Group of the Medical University of Warsaw at the Department of Internal Medicine, Endocrinology and Diabetology, National Medical Institute of the Ministry of the Interior and Administration in Warsaw, 02-507 Warsaw, Poland; (E.B.); (A.Ż.-Ł.); (K.W.); (A.K.); (J.J.)
| | - Tomasz Korcz
- Laboratory Diagnostics Unit, National Medical Institute of the Ministry of the Interior and Administration in Warsaw, 02-507 Warsaw, Poland; (T.K.); (M.C.-B.)
| | - Małgorzata Cicha-Brzezińska
- Laboratory Diagnostics Unit, National Medical Institute of the Ministry of the Interior and Administration in Warsaw, 02-507 Warsaw, Poland; (T.K.); (M.C.-B.)
| | - Edward Franek
- Department of Internal Medicine, Endocrinology and Diabetology, National Medical Institute of the Ministry of the Interior and Administration in Warsaw, 02-507 Warsaw, Poland; (M.L.); (E.F.)
- Students Scientific Group of the Medical University of Warsaw at the Department of Internal Medicine, Endocrinology and Diabetology, National Medical Institute of the Ministry of the Interior and Administration in Warsaw, 02-507 Warsaw, Poland; (E.B.); (A.Ż.-Ł.); (K.W.); (A.K.); (J.J.)
- Department of Human Epigenetics, Mossakowski Medical Research Centre Polish Academy of Sciences, 02-106 Warsaw, Poland
| |
Collapse
|
4
|
Failla M, Molaro MC, Schiano ME, Serafini M, Tiburtini GA, Gianquinto E, Scoccia R, Battisegola C, Rimoli MG, Chegaev K, Ercolano G, Lazzarato L, Spyrakis F, Sodano F. Opportunities and Challenges of Arginase Inhibitors in Cancer: A Medicinal Chemistry Perspective. J Med Chem 2024; 67:19988-20021. [PMID: 39558532 DOI: 10.1021/acs.jmedchem.4c01429] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2024]
Abstract
The overexpression of two arginase (ARG) isoforms, ARG1 and ARG2, contributes to the onset of numerous disorders, including cardiovascular and immune-mediated diseases, as well as tumors. To elucidate the specific roles of ARG1 and ARG2 without interfering with their physiological functions, it is crucial to develop effective ARG inhibitors that target only one isoform, while maintaining low toxicity and an adequate pharmacokinetic profile. In this context, we present a comprehensive overview of the different generations of ARG inhibitors. Given the general lack of selectivity in most existing inhibitors, we analyzed the structural features and plasticity of the ARG1 and ARG2 binding sites to explore the potential for designing inhibitors with novel binding patterns. We also review ongoing preclinical and clinical studies on selected inhibitors, highlighting both progress and challenges in developing potent, selective ARG inhibitors. Furthermore, we discuss medicinal chemistry strategies that may accelerate the discovery of selective ARG inhibitors.
Collapse
Affiliation(s)
- Mariacristina Failla
- Department of Drug Science and Technology, University of Turin, 10125 Turin, Italy
| | | | | | - Marta Serafini
- Department of Drug Science and Technology, University of Turin, 10125 Turin, Italy
| | | | - Eleonora Gianquinto
- Department of Drug Science and Technology, University of Turin, 10125 Turin, Italy
| | - Riccardo Scoccia
- Department of Drug Science and Technology, University of Turin, 10125 Turin, Italy
| | - Chiara Battisegola
- Department of Pharmacy, University of Naples Federico II, 80131 Naples, Italy
| | - Maria Grazia Rimoli
- Department of Pharmacy, University of Naples Federico II, 80131 Naples, Italy
| | - Konstantin Chegaev
- Department of Drug Science and Technology, University of Turin, 10125 Turin, Italy
| | - Giuseppe Ercolano
- Department of Pharmacy, University of Naples Federico II, 80131 Naples, Italy
| | - Loretta Lazzarato
- Department of Drug Science and Technology, University of Turin, 10125 Turin, Italy
| | - Francesca Spyrakis
- Department of Drug Science and Technology, University of Turin, 10125 Turin, Italy
| | - Federica Sodano
- Department of Pharmacy, University of Naples Federico II, 80131 Naples, Italy
| |
Collapse
|
5
|
Alipoor E, Salehi S, Dehghani S, Yaseri M, Hosseinzadeh-Attar MJ. Asymmetric dimethylarginine serum concentration in normal weight and obese CKD patients treated with hemodialysis. BMC Nephrol 2024; 25:294. [PMID: 39237885 PMCID: PMC11378466 DOI: 10.1186/s12882-024-03736-2] [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: 01/07/2024] [Accepted: 08/29/2024] [Indexed: 09/07/2024] Open
Abstract
INTRODUCTION Asymmetric dimethylarginine (ADMA), a cardiovascular risk factor, increases in renal failure. The aim of this study was to investigate ADMA levels in normal weight and obese patients on hemodialysis. METHODS In this cross-sectional study, 43 normal weight and 43 obese patients on regular hemodialysis were examined. Malnutrition-inflammation score (MIS), anthropometry, circulating ADMA, lipid profiles including triglycerides (TG), total cholesterol (TC), low-density lipoprotein (LDL), high-density lipoprotein (HDL) and lipid ratios, glucose homeostasis parameters, blood pressure, and high-sensitivity C-reactive protein (hs-CRP) were assessed. RESULTS Serum levels of ADMA were significantly lower in the obese compared to the normal weight patients (10268.2 ± 10092.4 vs. 13765.2 ± 9951.3 ng/l, P = 0.03). At the same time MIS score (6.1 ± 2.4 vs. 10.7 ± 3.2, P < 0.001), systolic blood pressure (119 ± 26.8 vs. 134.2 ± 24.7 mmHg, P = 0.018) and mean arterial pressure (91.3 ± 18.6 vs. 100.9 ± 15.9 mmHg, P = 0.028) were significantly lower in the obese than the normal weight group. Fasting blood glucose (P = 0.045), TG/HDL (P = 0.03), TC/HDL (P = 0.019), and LDL/HDL (P = 0.005) ratios, and hs-CRP (P = 0.015) levels were significantly higher in the obese than in the normal weight group. CONCLUSION Circulating ADMA was significantly lower in obese than in normal weight patients on hemodialysis, which was concomitant with lower MIS, indicating a better nutritional inflammatory status, and lower blood pressure.
Collapse
Affiliation(s)
- Elham Alipoor
- Cardiac Primary Prevention Research Center, Cardiovascular Diseases Research Institute, Tehran University of Medical Sciences, Tehran, Iran
- Department of Clinical Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, No#44, Hojjatdoost St., Naderi St., Keshavarz Blvd, Tehran, Iran
| | - Shiva Salehi
- Department of Clinical Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, No#44, Hojjatdoost St., Naderi St., Keshavarz Blvd, Tehran, Iran
| | - Sahar Dehghani
- Department of Clinical Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, No#44, Hojjatdoost St., Naderi St., Keshavarz Blvd, Tehran, Iran
| | - Mehdi Yaseri
- Department of Epidemiology and Biostatistics, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Javad Hosseinzadeh-Attar
- Cardiac Primary Prevention Research Center, Cardiovascular Diseases Research Institute, Tehran University of Medical Sciences, Tehran, Iran.
- Department of Clinical Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, No#44, Hojjatdoost St., Naderi St., Keshavarz Blvd, Tehran, Iran.
| |
Collapse
|
6
|
Zhao Y, Shen W, Zhang M, Guo M, Dou Y, Han S, Yu J, Cui M, Zhao Y. DDAH-1 maintains endoplasmic reticulum-mitochondria contacts and protects dopaminergic neurons in Parkinson's disease. Cell Death Dis 2024; 15:399. [PMID: 38849335 PMCID: PMC11161642 DOI: 10.1038/s41419-024-06772-w] [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: 11/27/2023] [Revised: 05/21/2024] [Accepted: 05/23/2024] [Indexed: 06/09/2024]
Abstract
The loss of dopaminergic neurons in the substantia nigra is a hallmark of pathology in Parkinson's disease (PD). Dimethylarginine dimethylaminohydrolase-1 (DDAH-1) is the critical enzyme responsible for the degradation of asymmetric dimethylarginine (ADMA) which inhibits nitric oxide (NO) synthase and has been implicated in neurodegeneration. Mitochondrial dysfunction, particularly in the mitochondria-associated endoplasmic reticulum membrane (MAM), plays a critical role in this process, although the specific molecular target has not yet been determined. This study aims to examine the involvement of DDAH-1 in the nigrostriatal dopaminergic pathway and PD pathogenesis. The distribution of DDAH-1 in the brain and its colocalization with dopaminergic neurons were observed. The loss of dopaminergic neurons and aggravated locomotor disability after rotenone (ROT) injection were showed in the DDAH-1 knockout rat. L-arginine (ARG) and NO donors were employed to elucidate the role of NO respectively. In vitro, we investigated the effects of DDAH-1 knockdown or overexpression on cell viability and mitochondrial functions, as well as modulation of ADMA/NO levels using ADMA or ARG. MAM formation was assessed by the Mitofusin2 oligomerization and the mitochondrial ubiquitin ligase (MITOL) phosphorylation. We found that DDAH-1 downregulation resulted in enhanced cell death and mitochondrial dysfunctions, accompanied by elevated ADMA and reduced NO levels. However, the recovered NO level after the ARG supplement failed to exhibit a protective effect on mitochondrial functions and partially restored cell viability. DDAH-1 overexpression prevented ROT toxicity, while ADMA treatment attenuated these protective effects. The declines of MAM formation in ROT-treated cells were exacerbated by DDAH-1 downregulation via reduced MITOL phosphorylation, which was reversed by DDAH-1 overexpression. Together, the abundant expression of DDAH-1 in nigral dopaminergic neurons may exert neuroprotective effects by maintaining MAM formation and mitochondrial function probably via ADMA, indicating the therapeutic potential of targeting DDAH-1 for PD.
Collapse
Affiliation(s)
- Yichen Zhao
- Department of Neurology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Weiwei Shen
- Department of Neurology and Institute of Neurology, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Minjie Zhang
- Department of Neurology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Min Guo
- Department of Neurology and Institute of Neurology, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yunxiao Dou
- Department of Neurology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Sida Han
- Department of Neurology and Institute of Neurology, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jintai Yu
- Department of Neurology, Huashan Hospital, MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China.
| | - Mei Cui
- Department of Neurology and Institute of Neurology, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China.
| | - Yanxin Zhao
- Department of Neurology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.
| |
Collapse
|
7
|
Ng YYH, Dora KA, Lemmey HA, Lin J, Alden J, Wallis L, Donovan L, Shorthose O, Leiper FC, Leiper J, Garland CJ. Asymmetric Dimethylarginine Enables Depolarizing Spikes and Vasospasm in Mesenteric and Coronary Resistance Arteries. Hypertension 2024; 81:764-775. [PMID: 38226470 PMCID: PMC10956675 DOI: 10.1161/hypertensionaha.123.22454] [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: 11/23/2023] [Accepted: 01/03/2024] [Indexed: 01/17/2024]
Abstract
BACKGROUND Increased vasoreactivity due to reduced endothelial NO bioavailability is an underlying feature of cardiovascular disease, including hypertension. In small resistance arteries, declining NO enhances vascular smooth muscle (VSM) reactivity partly by enabling rapid depolarizing Ca2+-based spikes that underlie vasospasm. The endogenous NO synthase inhibitor asymmetric dimethylarginine (ADMA) is metabolized by DDAH1 (dimethylarginine dimethylaminohydrolase 1) and elevated in cardiovascular disease. We hypothesized ADMA might enable VSM spikes and vasospasm by reducing NO bioavailability, which is opposed by DDAH1 activity and L-arginine. METHODS Rat isolated small mesenteric arteries and myogenic rat-isolated intraseptal coronary arteries (RCA) were studied using myography, VSM intracellular recording, Ca2+ imaging, and DDAH1 immunolabeling. Exogenous ADMA was used to inhibit NO synthase and a selective DDAH1 inhibitor, NG-(2-methoxyethyl) arginine, to assess the functional impact of ADMA metabolism. RESULTS ADMA enhanced rat-isolated small mesenteric arteries vasoreactivity to the α1-adrenoceptor agonist, phenylephrine by enabling T-type voltage-gated calcium channel-dependent depolarizing spikes. However, some endothelium-dependent NO-vasorelaxation remained, which was sensitive to DDAH1-inhibition with NG-(2-methoxyethyl) arginine. In myogenically active RCA, ADMA alone stimulated depolarizing Ca2+ spikes and marked vasoconstriction, while NO vasorelaxation was abolished. DDAH1 expression was greater in rat-isolated small mesenteric arteries endothelium compared with RCA, but low in VSM of both arteries. L-arginine prevented depolarizing spikes and protected NO-vasorelaxation in rat-isolated small mesenteric artery and RCA. CONCLUSIONS ADMA increases VSM electrical excitability enhancing vasoreactivity. Endothelial DDAH1 reduces this effect, and low levels of DDAH1 in RCAs may render them susceptible to endothelial dysfunction contributing to vasospasm, changes opposed by L-arginine.
Collapse
Affiliation(s)
- Yu Y. Hanson Ng
- Department of Pharmacology, University of Oxford, United Kingdom (Y.Y.H.N., K.A.D., H.A.L.L., J. Lin, J.A., L.W., LD., O.S., C.J.G.)
| | - Kim A. Dora
- Department of Pharmacology, University of Oxford, United Kingdom (Y.Y.H.N., K.A.D., H.A.L.L., J. Lin, J.A., L.W., LD., O.S., C.J.G.)
| | - Hamish A.L. Lemmey
- Department of Pharmacology, University of Oxford, United Kingdom (Y.Y.H.N., K.A.D., H.A.L.L., J. Lin, J.A., L.W., LD., O.S., C.J.G.)
| | - JinHeng Lin
- Department of Pharmacology, University of Oxford, United Kingdom (Y.Y.H.N., K.A.D., H.A.L.L., J. Lin, J.A., L.W., LD., O.S., C.J.G.)
| | - James Alden
- Department of Pharmacology, University of Oxford, United Kingdom (Y.Y.H.N., K.A.D., H.A.L.L., J. Lin, J.A., L.W., LD., O.S., C.J.G.)
| | - Lillian Wallis
- Department of Pharmacology, University of Oxford, United Kingdom (Y.Y.H.N., K.A.D., H.A.L.L., J. Lin, J.A., L.W., LD., O.S., C.J.G.)
| | - Lucy Donovan
- Department of Pharmacology, University of Oxford, United Kingdom (Y.Y.H.N., K.A.D., H.A.L.L., J. Lin, J.A., L.W., LD., O.S., C.J.G.)
| | - Oliver Shorthose
- Department of Pharmacology, University of Oxford, United Kingdom (Y.Y.H.N., K.A.D., H.A.L.L., J. Lin, J.A., L.W., LD., O.S., C.J.G.)
| | - Fiona C. Leiper
- Institute of Cardiovascular and Medical Sciences, College of Medicine, Veterinary, and Life Sciences, University of Glasgow, United Kingdom (F.C.L., J. Leiper)
| | - James Leiper
- Institute of Cardiovascular and Medical Sciences, College of Medicine, Veterinary, and Life Sciences, University of Glasgow, United Kingdom (F.C.L., J. Leiper)
| | - Christopher J. Garland
- Department of Pharmacology, University of Oxford, United Kingdom (Y.Y.H.N., K.A.D., H.A.L.L., J. Lin, J.A., L.W., LD., O.S., C.J.G.)
| |
Collapse
|
8
|
Azhough R, Jalali P, Dashti MR, Taher S, Aghajani A. Intradermal methylene blue analgesic application in posthemorrhoidectomy pain management: a randomized controlled trial. Front Surg 2024; 11:1354328. [PMID: 38577253 PMCID: PMC10991772 DOI: 10.3389/fsurg.2024.1354328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 03/11/2024] [Indexed: 04/06/2024] Open
Abstract
Introduction Unbearable post-hemorrhoidectomy pain is a well-documented challenge, significantly impacting patient well-being and satisfaction after surgery, often influencing patients to decline in undergoing this procedure. It is widely recognized that methylene blue has an effect of reducing inflammation and pain by reduces the production of nitric oxide and inhibiting the action potentials production in nerves. This study aims to explore the potential benefits of postoperative regional administration of methylene blue in providing extended relief from post-hemorrhoidectomy pain. Methods This study included 97 patients aged 18-75 undergoing hemorrhoidectomy for stage III or IV hemorrhoids. A double-blind, randomized controlled trial compared postoperative intradermal injections of 1% methylene blue to 0.5% Marcaine as the control group. Two-week follow-up assessed pain. Statistical analysis, adherence to ethical standards, and registration were conducted. Result No significant differences were found in baseline demographics, surgical parameters, or complications between the Methylene Blue and control groups. Intervention group remained lower in mean pain score until the 12th day. Methylene blue group reported significantly lower postoperative pain scores from days 1 to 7, with no significant differences afterward. Conclusion This ongoing randomized controlled trial reveals the potential analgesic benefits of intradermal injection 1% methylene blue. It demonstrates comparable efficacy in reducing post-hemorrhoidectomy pain, with negligible side effects and complications.
Collapse
Affiliation(s)
- Ramin Azhough
- Department of General Surgery, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Pooya Jalali
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Centre, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Sahar Taher
- Faculty of Medicine, Islamic Azad University Tabriz Branch, Tabriz, Iran
| | - Ali Aghajani
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Centre, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
9
|
Abolfazli S, Mortazavi P, Kheirandish A, Butler AE, Jamialahmadi T, Sahebkar A. Regulatory effects of curcumin on nitric oxide signaling in the cardiovascular system. Nitric Oxide 2024; 143:16-28. [PMID: 38141926 DOI: 10.1016/j.niox.2023.12.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 11/25/2023] [Accepted: 12/18/2023] [Indexed: 12/25/2023]
Abstract
The continuously rising prevalence of cardiovascular disease (CVD) globally substantially impacts the economic growth of developing countries. Indeed, one of the leading causes of death worldwide is unfavorable cardiovascular events. Reduced nitric oxide (NO) generation is the pathogenic foundation of endothelial dysfunction, which is regarded as the first stage in the development of a number of CVDs. Nitric oxide exerts an array of biological effects, including vasodilation, the suppression of vascular smooth muscle cell proliferation and the functional control of cardiac cells. Numerous treatment strategies aim to increase NO synthesis or upregulate downstream NO signaling pathways. The major component of Curcuma longa, curcumin, has long been utilized in traditional medicine to treat various illnesses, especially CVDs. Curcumin improves CV function as well as having important pleiotropic effects, such as anti-inflammatory and antioxidant, through its ability to increase the bioavailability of NO and to positively impact NO-related signaling pathways. In this review, we discuss the scientific literature relating to curcumin's positive effects on NO signaling and vascular endothelial function.
Collapse
Affiliation(s)
- Sajad Abolfazli
- Student Research Committee, School of Pharmacy, Mazandaran University of Medical Science, Sari, Iran
| | - Parham Mortazavi
- Isfahan Cardiovascular Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Ali Kheirandish
- Department of Pharmacology, School of Medicine, Iran University of Medical Sciences, Hemmat Highway, Tehran, Iran
| | - Alexandra E Butler
- Research Department, Royal College of Surgeons in Ireland, Bahrain, PO Box, 15503, Adliya, Bahrain
| | - Tannaz Jamialahmadi
- Medical Toxicology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amirhossein Sahebkar
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
10
|
Liu J, Hua Z, Liao S, Li B, Tang S, Huang Q, Wei Z, Lu R, Lin C, Ding X. Prediction of the active compounds and mechanism of Biochanin A in the treatment of Legg-Calvé-Perthes disease based on network pharmacology and molecular docking. BMC Complement Med Ther 2024; 24:26. [PMID: 38195507 PMCID: PMC10775507 DOI: 10.1186/s12906-023-04298-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 12/06/2023] [Indexed: 01/11/2024] Open
Abstract
BACKGROUND Legg-Calvé-Perthes disease is a special self-limited disease in pediatric orthopedics with a high disability rate and a long-term course, and there is still no clear and effective therapeutic drug in clinic. This study aimed to investigate the potential efficacy of biochanin A, a kind of oxygen-methylated isoflavone compound, in treating Perthes disease based on network pharmacology, molecular docking and in vitro experiments. METHODS IL-6 was used to stimulate human umbilical vein endothelial cells to construct endothelial cell dysfunction model. We demonstrated whether biochanin A could alleviate endothelial dysfunction through CCK8 assay, immunofluorescence. Targets of biochanin A from pharmMappeer, SWISS, and TargetNet databases were screened. Targets of endothelial dysfunction were obtained from Genecards and OMIM databases. Protein-protein interaction, Gene Ontology, and Kyoto Encyclopedia of Genes and Genomics analyses were used to analyze the potential target and the key pathway of the anti-endothelial dysfunction activity of biochanin A. To validate the potential target-drug interactions, molecular docking and molecular dynamics simulations were performed and the result was proved by western blot. RESULTS It was found that biochanin A can promote the expression of ZO-1, reduce the expression of ICAM-1, which means improving endothelial dysfunction. A total of 585 targets of biochanin A from pharmMappeer, SWISS, and TargetNet databases were screened. A total of 10,832 targets of endothelial dysfunction were obtained from Genecards and OMIM databases. A total of 527 overlapping targets of endothelial dysfunction and biochanin A were obtained. AKT1, TNF-α, VCAM1, ICAM1, and NOS3 might be the key targets of the anti-endothelial dysfunction activity of biochanin A, and the key pathways might be PI3K-Akt and TNF signaling pathways. Molecular docking results indicated that the AKT1 and TNF-α had the highest affinity binding with biochanin A. CONCLUSION This study indicates that biochanin A can target AKT1 and TNF-α to alleviate endothelial dysfunction induced by IL-6 in Perthes disease, which provides a theoretical basis for the treatment of Perthes disease by using biochanin A.
Collapse
Affiliation(s)
- Jianhong Liu
- Department of Trauma Orthopedic and Hand Surgery, The First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, 530021, Guangxi, China
| | - Zhirui Hua
- Department of Trauma Orthopedic and Hand Surgery, The First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, 530021, Guangxi, China
| | - Shijie Liao
- Department of Trauma Orthopedic and Hand Surgery, The First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, 530021, Guangxi, China
- Guangxi Key Laboratory of Regenerative Medicine, Research Centre for Regenerative Medicine, Guangxi Medical University, Nanning, Guangxi, China
| | - Boxiang Li
- Department of Orthopedics, Minzu Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China
| | - Shengping Tang
- Department of Trauma Orthopedic and Hand Surgery, The First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, 530021, Guangxi, China
| | - Qian Huang
- Department of Trauma Orthopedic and Hand Surgery, The First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, 530021, Guangxi, China
| | - Zhendi Wei
- Department of Trauma Orthopedic and Hand Surgery, The First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, 530021, Guangxi, China
| | - Rongbin Lu
- Department of Trauma Orthopedic and Hand Surgery, The First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, 530021, Guangxi, China
| | - Chengsen Lin
- Trauma Center, Emergency Department, The First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, Guangxi, 530021, China.
| | - Xiaofei Ding
- Department of Trauma Orthopedic and Hand Surgery, The First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, 530021, Guangxi, China.
- Guangxi Key Laboratory of Regenerative Medicine, Research Centre for Regenerative Medicine, Guangxi Medical University, Nanning, Guangxi, China.
| |
Collapse
|
11
|
Ceruti T, Frapolli R, Ghilardi C, Decio A, Dellavedova G, Tommasi S, Zucchetti M, Mangoni AA. Development of a HPLC-MS/MS Method to Assess the Pharmacokinetics and Tumour Distribution of the Dimethylarginine Dimethylaminohydrolase 1 Inhibitors ZST316 and L-257 in a Xenograft Model of Triple-Negative Breast Cancer in Mice. Molecules 2023; 28:8056. [PMID: 38138547 PMCID: PMC10746103 DOI: 10.3390/molecules28248056] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 12/07/2023] [Accepted: 12/12/2023] [Indexed: 12/24/2023] Open
Abstract
We describe the development and validation of an HPLC-MS/MS method to assess the pharmacokinetics and tumour distribution of ZST316, an arginine analogue with inhibitory activity towards dimethylarginine dimethylaminohydrolase 1 (DDAH1) and vasculogenic mimicry, and its active metabolite L-257 in a xenograft model of triple-negative breast cancer (TNBC). The method proved to be reproducible, precise, and highly accurate for the measurement of both compounds in plasma and tumour tissue following acute and chronic (five days) intraperitoneal administration of ZST316 (30 mg/Kg daily) in six-week-old severe combined immunodeficiency disease (SCID) mice inoculated with MDA-MB-231 TNBC cells. ZST316 was detected in tumour tissue and plasma after 1 h (6.47 and 9.01 μM, respectively) and 24 h (0.13 and 0.16 μM, respectively) following acute administration, without accumulation during chronic treatment. Similarly, the metabolite L-257 was found in tumour tissue and plasma after 1 h (15.06 and 8.72 μM, respectively) and 24 h (0.17 and 0.17 μM, respectively) following acute administration of ZST316, without accumulation during chronic treatment. The half-life after acute and chronic treatment ranged between 4.4-7.1 h (plasma) and 4.5-5.0 h (tumour) for ZST316, and 4.2-5.3 h (plasma) and 3.6-4.9 h (tumour) for L-257. The results of our study demonstrate the (a) capacity to accurately measure ZST316 and L-257 concentrations in plasma and tumour tissue in mice using the newly developed HPLC-MS/MS method, (b) rapid conversion of ZST316 into L-257, (c) good intra-tumour penetration of both compounds, and (d) lack of accumulation of both ZST316 and L-257 in plasma and tumour tissue during chronic administration. Compared to a previous method developed by our group to investigate ZST316 in plasma, the main advantages of the new method include a wider range of linearity which reduces the need for dilutions and the combined assessment of ZST316 and L-257 in plasma and tumour tissue which limits the required amount of matrix. The new HPLC-MS/MS method is useful to investigate the in vivo effects of ZST316 and L-257 on vasculogenic mimicry, tumour mass, and metastatic burden in xenograft models of TNBC.
Collapse
Affiliation(s)
- Tommaso Ceruti
- Laboratory of Cancer Pharmacology, Department of Oncology, Instituto di Ricerche Farmacologiche Mario Negri IRCCS, 20156 Milan, Italy; (T.C.); (R.F.); (M.Z.)
| | - Roberta Frapolli
- Laboratory of Cancer Pharmacology, Department of Oncology, Instituto di Ricerche Farmacologiche Mario Negri IRCCS, 20156 Milan, Italy; (T.C.); (R.F.); (M.Z.)
| | - Carmen Ghilardi
- Laboratory of Cancer Metastasis Therapeutics, Department of Oncology, Instituto di Ricerche Farmacologiche Mario Negri IRCCS, 20156 Milan, Italy; (C.G.); (A.D.); (G.D.)
| | - Alessandra Decio
- Laboratory of Cancer Metastasis Therapeutics, Department of Oncology, Instituto di Ricerche Farmacologiche Mario Negri IRCCS, 20156 Milan, Italy; (C.G.); (A.D.); (G.D.)
| | - Giulia Dellavedova
- Laboratory of Cancer Metastasis Therapeutics, Department of Oncology, Instituto di Ricerche Farmacologiche Mario Negri IRCCS, 20156 Milan, Italy; (C.G.); (A.D.); (G.D.)
| | - Sara Tommasi
- Discipline of Clinical Pharmacology, College of Medicine and Public Health, Flinders University, Bedford Park, SA 5042, Australia;
- Department of Clinical Pharmacology, Flinders Medical Centre, Southern Adelaide Local Health Network, Bedford Park, SA 5042, Australia
| | - Massimo Zucchetti
- Laboratory of Cancer Pharmacology, Department of Oncology, Instituto di Ricerche Farmacologiche Mario Negri IRCCS, 20156 Milan, Italy; (T.C.); (R.F.); (M.Z.)
| | - Arduino A. Mangoni
- Discipline of Clinical Pharmacology, College of Medicine and Public Health, Flinders University, Bedford Park, SA 5042, Australia;
- Department of Clinical Pharmacology, Flinders Medical Centre, Southern Adelaide Local Health Network, Bedford Park, SA 5042, Australia
| |
Collapse
|
12
|
Wu Y, Shen S, Chen J, Ni W, Wang Q, Zhou H, Chen J, Zhang H, Mei Z, Sun X, Shen P, Jie Z, Xu W, Hong Z, Ma Y, Wang K, Wan S, Wu H, Xie Z, Qin A, Fan S. Metabolite asymmetric dimethylarginine (ADMA) functions as a destabilization enhancer of SOX9 mediated by DDAH1 in osteoarthritis. SCIENCE ADVANCES 2023; 9:eade5584. [PMID: 36753544 PMCID: PMC9908022 DOI: 10.1126/sciadv.ade5584] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 01/06/2023] [Indexed: 06/18/2023]
Abstract
Osteoarthritis (OA) is a degenerative disease with a series of metabolic changes accompanied by many altered enzymes. Here, we report that the down-regulated dimethylarginine dimethylaminohydrolase-1 (DDAH1) is accompanied by increased asymmetric dimethylarginine (ADMA) in degenerated chondrocytes and in OA samples. Global or chondrocyte-conditional knockout of ADMA hydrolase DDAH1 accelerated OA development in mice. ADMA induces the degeneration and senescence of chondrocytes and reduces the extracellular matrix deposition, thereby accelerating OA progression. ADMA simultaneously binds to SOX9 and its deubiquitinating enzyme USP7, blocking the deubiquitination effects of USP7 on SOX9 and therefore leads to SOX9 degradation. The ADMA level in synovial fluids of patients with OA is increased and has predictive value for OA diagnosis with good sensitivity and specificity. Therefore, activating DDAH1 to reduce ADMA level might be a potential therapeutic strategy for OA treatment.
Collapse
Affiliation(s)
- Yizheng Wu
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Shuying Shen
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Jiaxin Chen
- Department of Breast Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Weiyu Ni
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Qinxin Wang
- Department of Orthopaedic Surgery, China Coast Guard Hospital of the People’s Armed Police Force, Jiaxing, China
| | - Hongyi Zhou
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Junxin Chen
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Haitao Zhang
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Zixuan Mei
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Xuewu Sun
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Panyang Shen
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Zhiwei Jie
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Wenbin Xu
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Zhenghua Hong
- Department of Orthopaedic Surgery, Taizhou Hospital of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Yan Ma
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Kefan Wang
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Shuanglin Wan
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Hongfei Wu
- Department of Orthopaedic Surgery, China Coast Guard Hospital of the People’s Armed Police Force, Jiaxing, China
| | - Ziang Xie
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - An Qin
- Department of Orthopaedics, Shanghai Key Laboratory of Orthopaedic Implant, Shanghai Ninth People’s Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Shunwu Fan
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| |
Collapse
|
13
|
Amirkhizi F, Khalese-Ranjbar B, Mansouri E, Hamedi-Shahraki S, Asghari S. Correlations of selenium and selenoprotein P with asymmetric dimethylarginine and lipid profile in patients with polycystic ovary syndrome. J Trace Elem Med Biol 2023; 75:127101. [PMID: 36395675 DOI: 10.1016/j.jtemb.2022.127101] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 10/25/2022] [Accepted: 11/08/2022] [Indexed: 11/13/2022]
Abstract
BACKGROUND Polycystic ovary syndrome (PCOS) is associated with an increased risk of cardiovascular diseases (CVD). Accumulating evidence has suggested that selenium (Se) is of importance for optimal function of the cardiovascular system. This study aimed to investigate the associations of selenium and selenoprotein P (SePP) with asymmetric dimethylarginine (ADMA) and lipid profile in women with PCOS. METHODS In this cross-sectional study, 125 females aged 18-45 years diagnosed with PCOS were recruited. An interviewer-administered questionnaire was applied to gather the relevant demographic characteristics, detailed clinical information, and lifestyle habits of participants. Fasting blood samples were obtained to measure biochemical parameters. Serum concentrations of total testosterone, sex hormone-binding globulin (SHBG), ADMA, and lipid profiles as well as anthropometric measurements were assessed across tertiles of serum Se and SePP concentrations. RESULTS There was a positive correlation between serum Se and SePP concentrations (r = 0.434, p < 0.001). Serum Se level was inversely correlated with ADMA (r = -0.21, p = 0.025) and TG (r = -0.17, p = 0.041) concentrations. There were also inverse correlations between SePP and ADMA (r = -0.34, p < 0.001), TG (r = -0.21, p = 0.019), and oxidized low density lipoprotein (ox-LDL) (r = -0.25, p = 0.007) levels. No significant relationship was found between serum Se and SePP concentrations with total cholesterol, low density lipoprotein cholesterol (LDL-C), high density lipoprotein cholesterol (HDL-C), apolipoprotein-A1 (Apo-A1), apolipoprotein-B (Apo-B100), total testosterone, SHBG, and free androgen index as well as anthropometric parameters (All p > 0.05). CONCLUSION The present study found that Se and SePP levels were inversely correlated with ADMA and TG concentrations as well as ox-LDL levels.
Collapse
Affiliation(s)
- Farshad Amirkhizi
- Department of Nutrition, Faculty of Public Health, Zabol University of Medical Sciences, Zabol, Iran
| | - Banafshe Khalese-Ranjbar
- Department of Clinical Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Elahe Mansouri
- Department of Clinical Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Soudabeh Hamedi-Shahraki
- Department of Epidemiology and Biostatistics, Faculty of Public Health, Zabol University of Medical Sciences, Zabol, Iran
| | - Somayyeh Asghari
- Department of Clinical Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
14
|
Wu PY, Tan X, Wang M, Zheng X, Lou JH. Selenium supplementation for polycystic ovary syndrome: a meta-analysis of randomized controlled trials. Gynecol Endocrinol 2022; 38:928-934. [PMID: 36050880 DOI: 10.1080/09513590.2022.2118709] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Introduction: The efficacy of selenium supplementation was elusive for polycystic ovary syndrome. This meta-analysis aimed to explore the efficacy of selenium supplementation for polycystic ovary syndrome. Methods: PubMed, EMbase, Web of science, EBSCO, Cochrane library database, CNKI, Chongqing VIP database and Wanfang databases have been searched through July 2022 and we included randomized controlled trials (RCTs) reporting the effect of selenium supplementation versus placebo in patients with polycystic ovary syndrome. Results: Five RCTs were included in the meta-analysis. Compared with placebo group for polycystic ovary syndrome, selenium supplementation was associated with significantly reduced total testosterone (SMD=-0.42; 95% CI=-0.78 to -0.06; p = 0.02) and cholesterol (SMD=-0.71; 95% CI=-1.41 to -0.02; p = 0.04), but revealed no remarkable influence on SHBG (SMD=-0.52; 95% CI=-1.29 to 0.25; p = 0.19), triglyceride (SMD=-1.45; 95% CI=-3.62 to 0.73; p = 0.19), LDL (SMD=-0.17; 95% CI=-0.72 to 0.37; p = 0.53), FPG (SMD=-0.95; 95% CI=-3.72 to 1.82; p = 0.50) or HOMA-IR (SMD=-0.51; 95% CI=-3.79 to 2.77; p = 0.76). Conclusions: Selenium supplementation may be able to improve the metabolic response for polycystic ovary syndrome, and this finding should be interpreted with caution.
Collapse
Affiliation(s)
- Pei-Yu Wu
- Department of VIP, Chongqing General Hospital, Chongqing, China
| | - Xianzu Tan
- Department of VIP, Chongqing General Hospital, Chongqing, China
| | - Min Wang
- Department of VIP, Chongqing General Hospital, Chongqing, China
| | - Xiangqing Zheng
- Department of VIP, Chongqing General Hospital, Chongqing, China
| | - Jin-He Lou
- Department of Health Management Center, Chongqing General Hospital, Chongqing, China
| |
Collapse
|
15
|
Priyadarshini G, Rajappa M. Predictive markers in chronic kidney disease. Clin Chim Acta 2022; 535:180-186. [PMID: 35995274 DOI: 10.1016/j.cca.2022.08.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Revised: 08/13/2022] [Accepted: 08/15/2022] [Indexed: 11/25/2022]
Abstract
Chronic kidney disease (CKD) is defined by gradual deterioration of the renal parenchyma and decline of functioning nephrons. CKD is now recognized as a distinct risk factor for cardiovascular disease (CVD). This risk rises in tandem with the decline in kidney function and peaks at the end-stage. It is important to identify individuals with CKD who are at a higher risk of advancing to end-stage renal disease (ESRD) and the beginning of CVD. This will enhance the clinical benefits and so that evidence-based therapy may be started at the initial stages for those individuals. A promising biomarker must represent tissue damage, and be easy to detect using non-invasive methods. Current CKD progression indicators have difficulties in reaching this aim. Hence this review presents an update on markers studied in the last decade, which help in the prediction of CKD progression such as neutrophil gelatinase-associated lipocalin, kidney injury molecule-1, urinary liver-type fatty acid-binding protein, cystatin-C, asymmetric dimethylarginine, symmetric dimethylarginine, endotrophin, methylglyoxal, sclerostin, uric acid, and miRNA-196a. Additional research is needed to determine the predictive usefulness of these indicators in clinical samples for disease development. Their utility as surrogate markers need to be explored further for the early identification of CKD progression.
Collapse
Affiliation(s)
- G Priyadarshini
- Department of Biochemistry, Jawaharlal Institute of Postgraduate Medical Education and Research, Puducherry, India
| | - Medha Rajappa
- Department of Biochemistry, Jawaharlal Institute of Postgraduate Medical Education and Research, Puducherry, India.
| |
Collapse
|
16
|
Design, synthesis, and molecular docking of novel pyrazole-chalcone analogs of lonazolac as 5-LOX, iNOS and tubulin polymerization inhibitors with potential anticancer and anti-inflammatory activities. Bioorg Chem 2022; 129:106171. [PMID: 36166898 DOI: 10.1016/j.bioorg.2022.106171] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Revised: 09/15/2022] [Accepted: 09/17/2022] [Indexed: 11/22/2022]
Abstract
Uncontrolled inflammation predisposes to pleiotropic effects leading to cancer development thanks to promoting all stages of tumorigenesis. Therefore, cancer-associated inflammation has been delegated as the seventh hallmark of cancer. Thus, raging the war against both inflammation and cancer via the innovation of bioactive agents with dual anti-inflammatory and anticancer activities is a necessity. Herein, a novel series of pyrazole-chalcone analogs of Lonazolac (7a-g and 8a-g) have been synthesized and investigated for their in vitro anticancer activity against four cancer cell lines using the MTT assay method. Among all, hybrid 8g was the most potent against three cancer cell lines, HeLa, HCT-116, and RPMI-822 with IC50 values of 2.41, 2.41, and 3.34 µM, respectively. In contrast, hybrid 8g showed moderate inhibitory activity against MCF-7 with IC50 28.93 μM and with a selectivity profile against MCF-10A (non-cancer cells). Mechanistically, hybrid 8g was the most potent inhibitor against tubulin polymerization (IC50 = 4.77 µM), suggesting tubulin as a molecular target and explaining the observed cytotoxicity of hybrid 8g. This was mirrored by the detected potent pre-G1 apoptosis induction and G2/M cell cycle arrest. Moreover, hybrid8gexhibited selectivity against COX-2 (IC50 = 5.13 µM) more than COX-1 (IC50 = 33.46 µM), indicating that 8g may have lower cardiovascular side effects, but is still not potent as celecoxib (COX-2 IC50 = 0.204 µM, COX-1 = 35.8 µM). Notably, hybrid 8g showed promising inhibitory activity towards 5-LOX (IC50 = 5.88 µM). Finally, the anti-inflammatory activity of hybrid8 g was confirmed by high iNOS and PGE2 inhibitory activities in LPS-stimulated RAW cells with IC50 values of4.93 µM and 10.98 µM, respectively, that accompanied by showingthe most potent inhibition of NO release (70.61 % inhibition rate). Molecular docking studies of hybrid 8g confirmed good correlations with the executed biological results. Furthermore, hybrid 8g had good drug-likeness and suitable physicochemical properties. Taken together, the combined results suggested hybrid8gas a promising orally administered candidate in the journey of repurposing NSAIDs for cancer chemopreventionand treatment.
Collapse
|
17
|
Singh J, Lee Y, Kellum JA. A new perspective on NO pathway in sepsis and ADMA lowering as a potential therapeutic approach. Crit Care 2022; 26:246. [PMID: 35962414 PMCID: PMC9373887 DOI: 10.1186/s13054-022-04075-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 06/23/2022] [Indexed: 11/30/2022] Open
Abstract
The nitric oxide pathway plays a critical role in vascular homeostasis. Increased levels of systemic nitric oxide (NO) are observed in preclinical models of sepsis and endotoxemia. This has led to the postulation that vasodilation by inducible nitric oxide synthase (iNOS) generated NO may be a mechanism of hypotension in sepsis. However, contrary to the expected pharmacological action of a nitric oxide synthase (NOS) inhibitor, clinical studies with L-NAME produced adverse cardiac and pulmonary events, and higher mortality in sepsis patients. Thus, the potential adverse effects of NO in human sepsis and shock have not been fully established. In recent years, the emerging new understanding of the NO pathway has shown that an endogenously produced inhibitor of NOS, asymmetric dimethylarginine (ADMA), a host response to infection, may play an important role in the pathophysiology of sepsis as well as organ damage during ischemia–reperfusion. ADMA induces microvascular dysfunction, proinflammatory and prothrombotic state in endothelium, release of inflammatory cytokines, oxidative stress and mitochondrial dysfunction. High levels of ADMA exist in sepsis patients, which may produce adverse effects like those observed with L-NAME. Several studies have demonstrated the association of plasma ADMA levels with mortality in sepsis patients. Preclinical studies in sepsis and ischemia–reperfusion animal models have shown that lowering of ADMA reduced organ damage and improved survival. The clinical finding with L-NAME and the preclinical research on ADMA “bed to bench” suggest that ADMA lowering could be a potential therapeutic approach to attenuate progressive organ damage and mortality in sepsis. Testing of this approach is now feasible by using the pharmacological molecules that specifically lower ADMA.
Collapse
|
18
|
Pérez-López C, Rodríguez-Mozaz S, Serra-Compte A, Alvarez-Muñoz D, Ginebreda A, Barceló D, Tauler R. Effects of sulfamethoxazole exposure on mussels (Mytilus galloprovincialis) metabolome using retrospective non-target high-resolution mass spectrometry and chemometric tools. Talanta 2022; 252:123804. [DOI: 10.1016/j.talanta.2022.123804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 05/31/2022] [Accepted: 06/03/2022] [Indexed: 11/28/2022]
|
19
|
Johnson CM, Fast W. On the kinetic mechanism of dimethylarginine dimethylaminohydrolase. Bioorg Med Chem 2022; 66:116816. [PMID: 35598478 DOI: 10.1016/j.bmc.2022.116816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 05/06/2022] [Accepted: 05/09/2022] [Indexed: 11/28/2022]
Abstract
Dimethylarginine dimethylaminohydrolase (DDAH, EC 3.5.3.18) catalyzes the hydrolysis of asymmetric Nω,Nω-dimethyl-l-arginine (ADMA), an endogenous inhibitor of human nitric oxide synthases. The active-site cysteine residue has been proposed to serve as the catalytic nucleophile, forming an S-alkylthiourea reaction intermediate, and serving as a target for covalent inhibitors. Inhibition can lead to ADMA accumulation and downstream inhibition of nitric oxide production. Prior studies have provided experimental evidence for formation of this covalent adduct but have not characterized it kinetically. Here, rapid quench-flow is used with ADMA and the DDAH from Pseudomonas aeruginosa to determine the rate constants for formation (k2 = 17 ± 2 s-1) and decay (k3 = 1.5 ± 0.1 s-1) of the covalent S-alkylthiourea adduct. A minimal kinetic mechanism for DDAH is proposed that supports the kinetic competence of this species as a covalent reaction intermediate and assigns the rate-limiting step in substrate turnover as hydrolysis of this intermediate. This work helps elucidate the different reactivities of S-alkylthiourea intermediates found among the mechanistically diverse pentein superfamily of guanidine-modifying enzymes and provides information useful for inhibitor development.
Collapse
Affiliation(s)
- Corey M Johnson
- Division of Chemical Biology and Medicinal Chemistry, College of Pharmacy, University of Texas, Austin, TX 78712, USA; Department of Chemistry and Biochemistry, Howard College of Arts and Sciences, Samford University, Birmingham, AL 35229, USA
| | - Walter Fast
- Division of Chemical Biology and Medicinal Chemistry, College of Pharmacy, University of Texas, Austin, TX 78712, USA.
| |
Collapse
|
20
|
Global analysis of biosynthetic gene clusters reveals conserved and unique natural products in entomopathogenic nematode-symbiotic bacteria. Nat Chem 2022; 14:701-712. [PMID: 35469007 PMCID: PMC9177418 DOI: 10.1038/s41557-022-00923-2] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 02/24/2022] [Indexed: 12/27/2022]
Abstract
Microorganisms contribute to the biology and physiology of eukaryotic hosts and affect other organisms through natural products. Xenorhabdus and Photorhabdus (XP) living in mutualistic symbiosis with entomopathogenic nematodes generate natural products to mediate bacteria–nematode–insect interactions. However, a lack of systematic analysis of the XP biosynthetic gene clusters (BGCs) has limited the understanding of how natural products affect interactions between the organisms. Here we combine pangenome and sequence similarity networks to analyse BGCs from 45 XP strains that cover all sequenced strains in our collection and represent almost all XP taxonomy. The identified 1,000 BGCs belong to 176 families. The most conserved families are denoted by 11 BGC classes. We homologously (over)express the ubiquitous and unique BGCs and identify compounds featuring unusual architectures. The bioactivity evaluation demonstrates that the prevalent compounds are eukaryotic proteasome inhibitors, virulence factors against insects, metallophores and insect immunosuppressants. These findings explain the functional basis of bacterial natural products in this tripartite relationship. ![]()
Entomopathogenic nematodes carrying Xenorhabdus and Photorhabdus bacteria prey on insect larvae in the soil. Now, a comprehensive analysis of the bacterial genome has revealed ubiquitous and unique families of biosynthetic gene clusters. Evaluation of the bioactivity of the natural products expressed by the most prevalent cluster families explains the functional basis of bacterial natural products involved in bacteria–nematode–insect interactions.
Collapse
|
21
|
Lee Y, Singh J, Scott SR, Ellis B, Zorlutuna P, Wang M. A Recombinant Dimethylarginine Dimethylaminohydrolase-1-Based Biotherapeutics to Pharmacologically Lower Asymmetric Dimethyl Arginine, thus Improving Postischemic Cardiac Function and Cardiomyocyte Mitochondrial Activity. Mol Pharmacol 2022; 101:226-235. [PMID: 35042831 PMCID: PMC11033929 DOI: 10.1124/molpharm.121.000394] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 01/16/2022] [Indexed: 11/22/2022] Open
Abstract
High serum levels of asymmetric dimethyl arginine (ADMA) are associated with cardiovascular disease and mortality. Pharmacological agents to specifically lower ADMA and their potential impact on cardiovascular complications are not known. In this study, we aimed to investigate the effect of specific lowering of ADMA on myocardial response to ischemia-reperfusion injury (I/R) and direct effects on cardiomyocyte function. Effects of recombinant dimethylarginine dimethylaminohydrolase (rDDAH)-1 on I/R injury were determined using isolated mouse heart preparation. Respiration capacity and mitochondrial reactive oxygen species (ROS) generation were determined on mouse cardiomyocytes. Our results show that lowering ADMA by rDDAH-1 treatment resulted in improved recovery of cardiac function and reduction in myocardial infarct size in mouse heart response to I/R injury (control 22.24 ±4.60% versus rDDAH-1 15.90 ±4.23%, P < 0.01). In mouse cardiomyocytes, rDDAH-1 treatment improved ADMA-induced dysregulation of respiration capacity and decreased mitochondrial ROS. Furthermore, in human induced pluripotent stem cell (hiPSC)-derived cardiomyocytes with impaired contractility under hypoxia and high ADMA, rDDAH-1 treatment improved recovery and beating frequency (P < 0.05). rDDAH-1 treatment selectively modified I/R-induced myocardial cytokine expression, resulting in reduction in proinflammatory cytokine IL-17A (P < 0.001) and increased expression of anti-inflammatory cytokines IL-10 and IL-13 (P < 0.01). Further in vitro studies showed that IL-17A was the predominant and common cytokine modulated by ADMA-DDAH pathway in heart, cardiomyocytes, and endothelial cells. These studies show that lowering ADMA by pharmacological treatment with rDDAH-1 reduced I/R injury, improved cardiac function, and ameliorated cardiomyocyte bioenergetics and beating activity. These effects may be attributable to ADMA lowering in cardiomyocytes and preservation of cardiomyocyte mitochondrial function. SIGNIFICANCE STATEMENT: The pathological role of asymmetric dimethyl arginine (ADMA) has been demonstrated by its association with cardiovascular disease and mortality. Currently, pharmacological drugs to specifically lower ADMA are not available. The present study provides the first evidence that lowering of ADMA by recombinant recombinant dimethylarginine dimethylaminohydrolase (rDDAH)-1 improved postischemic cardiac function and cardiomyocyte bioenergetics and beating activity. Our studies suggest that lowering of ADMA by pharmacologic treatment offers opportunity to develop new therapies for the treatment of cardiovascular and renal disease.
Collapse
Affiliation(s)
- Young Lee
- Indiana Center for Biomedical Innovation, Indianapolis, Indiana (Y.L., J.S.); Indiana University School of Medicine, Indianapolis, Indiana (J.S.); Department of Surgery, Indiana University, School of Medicine, Indianapolis, Indiana (S.R.S., M.W.); Bioengineering Graduate Program (B.E., P.Z.) and Aerospace and Mechanical Engineering Department (P.Z.), University of Notre Dame, Notre Dame, Indiana; and Vasculonics LLC, Indianapolis, Indiana (J.S.)
| | - Jaipal Singh
- Indiana Center for Biomedical Innovation, Indianapolis, Indiana (Y.L., J.S.); Indiana University School of Medicine, Indianapolis, Indiana (J.S.); Department of Surgery, Indiana University, School of Medicine, Indianapolis, Indiana (S.R.S., M.W.); Bioengineering Graduate Program (B.E., P.Z.) and Aerospace and Mechanical Engineering Department (P.Z.), University of Notre Dame, Notre Dame, Indiana; and Vasculonics LLC, Indianapolis, Indiana (J.S.)
| | - Susan R Scott
- Indiana Center for Biomedical Innovation, Indianapolis, Indiana (Y.L., J.S.); Indiana University School of Medicine, Indianapolis, Indiana (J.S.); Department of Surgery, Indiana University, School of Medicine, Indianapolis, Indiana (S.R.S., M.W.); Bioengineering Graduate Program (B.E., P.Z.) and Aerospace and Mechanical Engineering Department (P.Z.), University of Notre Dame, Notre Dame, Indiana; and Vasculonics LLC, Indianapolis, Indiana (J.S.)
| | - Bradley Ellis
- Indiana Center for Biomedical Innovation, Indianapolis, Indiana (Y.L., J.S.); Indiana University School of Medicine, Indianapolis, Indiana (J.S.); Department of Surgery, Indiana University, School of Medicine, Indianapolis, Indiana (S.R.S., M.W.); Bioengineering Graduate Program (B.E., P.Z.) and Aerospace and Mechanical Engineering Department (P.Z.), University of Notre Dame, Notre Dame, Indiana; and Vasculonics LLC, Indianapolis, Indiana (J.S.)
| | - Pinar Zorlutuna
- Indiana Center for Biomedical Innovation, Indianapolis, Indiana (Y.L., J.S.); Indiana University School of Medicine, Indianapolis, Indiana (J.S.); Department of Surgery, Indiana University, School of Medicine, Indianapolis, Indiana (S.R.S., M.W.); Bioengineering Graduate Program (B.E., P.Z.) and Aerospace and Mechanical Engineering Department (P.Z.), University of Notre Dame, Notre Dame, Indiana; and Vasculonics LLC, Indianapolis, Indiana (J.S.)
| | - Meijing Wang
- Indiana Center for Biomedical Innovation, Indianapolis, Indiana (Y.L., J.S.); Indiana University School of Medicine, Indianapolis, Indiana (J.S.); Department of Surgery, Indiana University, School of Medicine, Indianapolis, Indiana (S.R.S., M.W.); Bioengineering Graduate Program (B.E., P.Z.) and Aerospace and Mechanical Engineering Department (P.Z.), University of Notre Dame, Notre Dame, Indiana; and Vasculonics LLC, Indianapolis, Indiana (J.S.)
| |
Collapse
|
22
|
Lee Y, Steinbach SML, Basile D, Singh J. A Therapeutic Extracorporeal Device for Specific Removal of Pathologic Asymmetric Dimethylarginine from the Blood. Blood Purif 2022; 51:889-898. [PMID: 35249015 PMCID: PMC9440949 DOI: 10.1159/000522294] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 01/25/2022] [Indexed: 12/16/2022]
Abstract
INTRODUCTION Blood levels of uremic toxin, asymmetric dimethylarginine (ADMA), are strongly associated with mortality in sepsis, renal failure, and cardiovascular and renal disease patients. METHODS An extracorporeal approach to reduce pathological ADMA was developed. The dimethylarginine dimethylaminohydrolase (DDAH) was immobilized on agarose beads to prepare a cartridge. The efficacy of cartridge for ADMA lowering in blood was investigated. RESULTS The DDAH beads and cartridge reduced ADMA from solution or plasma. The magnitude of ADMA removal was dependent on the quantity of DDAH linked to the beads and the flow rate. When tested in association with plasmapheresis, the DDAH-cartridge was highly effective in ADMA removal from the blood and improved the arginine/ADMA ratio in a pig model. CONCLUSION A new, safe, and effective extracorporeal approach to lower ADMA was developed which may have application in improving outcomes in patients with vascular complications and risk of mortality associated with high ADMA.
Collapse
Affiliation(s)
- Young Lee
- Indiana Center for Biomedical Innovation, Indianapolis, Indiana, USA,
| | - Sarah M L Steinbach
- Department of Veterinary Clinical Sciences, Purdue University, West Lafayette, Indiana, USA
| | - David Basile
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Jaipal Singh
- Indiana Center for Biomedical Innovation, Indianapolis, Indiana, USA
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Vasculonics Inc., Indianapolis, Indiana, USA
| |
Collapse
|
23
|
Xie Z, Hou L, Shen S, Wu Y, Wang J, Jie Z, Zhao X, Li X, Zhang X, Chen J, Xu W, Ning L, Ma Q, Wang S, Wang H, Yuan P, Fang X, Qin A, Fan S. Mechanical force promotes dimethylarginine dimethylaminohydrolase 1-mediated hydrolysis of the metabolite asymmetric dimethylarginine to enhance bone formation. Nat Commun 2022; 13:50. [PMID: 35013196 PMCID: PMC8748781 DOI: 10.1038/s41467-021-27629-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Accepted: 11/30/2021] [Indexed: 12/30/2022] Open
Abstract
Mechanical force is critical for the development and remodeling of bone. Here we report that mechanical force regulates the production of the metabolite asymmetric dimethylarginine (ADMA) via regulating the hydrolytic enzyme dimethylarginine dimethylaminohydrolase 1 (Ddah1) expression in osteoblasts. The presence of -394 4 N del/ins polymorphism of Ddah1 and higher serum ADMA concentration are negatively associated with bone mineral density. Global or osteoblast-specific deletion of Ddah1 leads to increased ADMA level but reduced bone formation. Further molecular study unveils that mechanical stimulation enhances TAZ/SMAD4-induced Ddah1 transcription. Deletion of Ddah1 in osteoblast-lineage cells fails to respond to mechanical stimulus-associated bone formation. Taken together, the study reveals mechanical force is capable of down-regulating ADMA to enhance bone formation.
Collapse
Affiliation(s)
- Ziang Xie
- Department of Orthopedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Lei Hou
- Department of Cardiology, Shanghai Tongren Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Shuying Shen
- Department of Orthopedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Yizheng Wu
- Department of Orthopedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Jian Wang
- Department of Orthopaedics, Tongde Hospital of Zhejiang Province, Hangzhou, China
| | - Zhiwei Jie
- Department of Orthopedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Xiangde Zhao
- Department of Orthopedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Xiang Li
- Department of Orthopedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Xuyang Zhang
- Department of Orthopedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Junxin Chen
- Department of Orthopedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Wenbin Xu
- Department of Orthopedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Lei Ning
- Department of Orthopedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Qingliang Ma
- Department of Orthopedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Shiyu Wang
- Department of Orthopedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Haoming Wang
- Department of Orthopedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Putao Yuan
- Department of Orthopedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Xiangqian Fang
- Department of Orthopedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China.
| | - An Qin
- Department of Orthopaedics, Shanghai Key Laboratory of Orthopaedic Implant, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.
| | - Shunwu Fan
- Department of Orthopedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China.
| |
Collapse
|
24
|
Asymmetric Dimethylarginine (ADMA) in Pediatric Renal Diseases: From Pathophysiological Phenomenon to Clinical Biomarker and Beyond. CHILDREN-BASEL 2021; 8:children8100837. [PMID: 34682102 PMCID: PMC8535118 DOI: 10.3390/children8100837] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 09/20/2021] [Accepted: 09/21/2021] [Indexed: 12/11/2022]
Abstract
Asymmetric dimethylarginine (ADMA), an endogenous nitric oxide (NO) synthase inhibitor, inhibits NO synthesis and contributes to the pathogenesis of many human diseases. In adults, ADMA has been identified as a biomarker for chronic kidney disease (CKD) progression and cardiovascular risk. However, little attention is given to translating the adult experience into the pediatric clinical setting. In the current review, we summarize circulating and urinary ADMA reported thus far in clinical studies relating to kidney disease in children and adolescents, as well as systematize the knowledge on pathophysiological role of ADMA in the kidneys. The aim of this review is also to show the various analytical methods for measuring ADMA and the issues tht need to be addressed before transforming to clinical practice in pediatric medicine. The last task is to suggest that ADMA may not only be suitable as a diagnostic or prognostic biomarker, but also a promising therapeutic strategy to treat pediatric kidney disease in the future.
Collapse
|
25
|
Ushio-Fukai M, Ash D, Nagarkoti S, Belin de Chantemèle EJ, Fulton DJR, Fukai T. Interplay Between Reactive Oxygen/Reactive Nitrogen Species and Metabolism in Vascular Biology and Disease. Antioxid Redox Signal 2021; 34:1319-1354. [PMID: 33899493 PMCID: PMC8418449 DOI: 10.1089/ars.2020.8161] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Reactive oxygen species (ROS; e.g., superoxide [O2•-] and hydrogen peroxide [H2O2]) and reactive nitrogen species (RNS; e.g., nitric oxide [NO•]) at the physiological level function as signaling molecules that mediate many biological responses, including cell proliferation, migration, differentiation, and gene expression. By contrast, excess ROS/RNS, a consequence of dysregulated redox homeostasis, is a hallmark of cardiovascular disease. Accumulating evidence suggests that both ROS and RNS regulate various metabolic pathways and enzymes. Recent studies indicate that cells have mechanisms that fine-tune ROS/RNS levels by tight regulation of metabolic pathways, such as glycolysis and oxidative phosphorylation. The ROS/RNS-mediated inhibition of glycolytic pathways promotes metabolic reprogramming away from glycolytic flux toward the oxidative pentose phosphate pathway to generate nicotinamide adenine dinucleotide phosphate (NADPH) for antioxidant defense. This review summarizes our current knowledge of the mechanisms by which ROS/RNS regulate metabolic enzymes and cellular metabolism and how cellular metabolism influences redox homeostasis and the pathogenesis of disease. A full understanding of these mechanisms will be important for the development of new therapeutic strategies to treat diseases associated with dysregulated redox homeostasis and metabolism. Antioxid. Redox Signal. 34, 1319-1354.
Collapse
Affiliation(s)
- Masuko Ushio-Fukai
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia, USA.,Department of Medicine (Cardiology) and Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Dipankar Ash
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia, USA.,Department of Medicine (Cardiology) and Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Sheela Nagarkoti
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia, USA.,Department of Medicine (Cardiology) and Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Eric J Belin de Chantemèle
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia, USA.,Department of Medicine (Cardiology) and Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - David J R Fulton
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia, USA.,Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Tohru Fukai
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia, USA.,Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, Georgia, USA.,Charlie Norwood Veterans Affairs Medical Center, Augusta, Georgia, USA
| |
Collapse
|
26
|
Lee SW, Han HC. Methylene Blue Application to Lessen Pain: Its Analgesic Effect and Mechanism. Front Neurosci 2021; 15:663650. [PMID: 34079436 PMCID: PMC8165385 DOI: 10.3389/fnins.2021.663650] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 04/16/2021] [Indexed: 12/12/2022] Open
Abstract
Methylene blue (MB) is a cationic thiazine dye, widely used as a biological stain and chemical indicator. Growing evidence have revealed that MB functions to restore abnormal vasodilation and notably it is implicated even in pain relief. Physicians began to inject MB into degenerated disks to relieve pain in patients with chronic discogenic low back pain (CDLBP), and some of them achieved remarkable outcomes. For osteoarthritis and colitis, MB abates inflammation by suppressing nitric oxide production, and ultimately relieves pain. However, despite this clinical efficacy, MB has not attracted much public attention in terms of pain relief. Accordingly, this review focuses on how MB lessens pain, noting three major actions of this dye: anti-inflammation, sodium current reduction, and denervation. Moreover, we showed controversies over the efficacy of MB on CDLBP and raised also toxicity issues to look into the limitation of MB application. This analysis is the first attempt to illustrate its analgesic effects, which may offer a novel insight into MB as a pain-relief dye.
Collapse
Affiliation(s)
- Seung Won Lee
- Good Doctor Research Institute, College of Medicine, Korea University, Seoul, South Korea
| | - Hee Chul Han
- Department of Physiology, College of Medicine and Neuroscience Research Institute, Korea University, Seoul, South Korea
| |
Collapse
|
27
|
Salmani M, Alipoor E, Navid H, Farahbakhsh P, Yaseri M, Imani H. Effect of l-arginine on cardiac reverse remodeling and quality of life in patients with heart failure. Clin Nutr 2021; 40:3037-3044. [PMID: 33610421 DOI: 10.1016/j.clnu.2021.01.044] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 01/14/2021] [Accepted: 01/29/2021] [Indexed: 12/26/2022]
Abstract
BACKGROUND & AIMS Heart failure (HF), as a major cardiac disease, is associated with considerable mortality, morbidities and poor quality of life. The aim of this study was to investigate the effect of l-arginine supplementation on cardiac outcomes and quality of life in patients with ischemic HF. METHODS This double-blind randomized controlled clinical trial was conducted in 50 patients with ischemic HF. Patients were randomly assigned to receive either 3 gr/d l-arginine or placebo, for 10 weeks. Cardiac function (based on echocardiography and six-minute walk test), blood pressure, and quality of life (based on the Minnesota living with heart failure questionnaire) were assessed. RESULTS The results showed significant improvements in ejection fraction (-6.5 ± 8.7 vs. -0.7 ± 7.8%, P = 0.037), left ventricular function (P = 0.043), diastolic dysfunction (P = 0.01) and marginally improvement in changes of left ventricular dimension during diastole (LVDd) (4 ± 6 vs. 0.3 ± 6.9 mm, P = 0.065) in the l-arginine compared to the placebo group. At the end of the study, physical aspect (5.7 ± 3.3 vs. 1.2 ± 6.1, P = 0.002) and total score (10 ± 6.7 vs. 4.1 ± 9.4, P = 0.011) of quality of life improved significantly in the l-arginine compared with the placebo group. Additionally, pre-to post-values of diastolic blood pressure, mean arterial pressure, LVDd, LV ejection fraction, left ventricular function, diastolic dysfunction as well as physical and total scores of quality of life improved significantly within the intervention, but not the placebo, group (all P < 0.05). CONCLUSION This study showed that 3 gr/d l-arginine supplementation for 10 weeks could improve cardiac recovery and function, and quality of life in patients with HF. This study was registered at the Iranian Clinical Trial Registration Center (www.irct.ir) with IRCT20170202032367N4 code.
Collapse
Affiliation(s)
- Mahnaz Salmani
- Department of Clinical Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Elham Alipoor
- Department of Nutrition, School of Public Health, Iran University of Medical Sciences, Tehran, Iran
| | - Hossein Navid
- Department of Heart Failure and Heart Transplantation, Tehran Heart Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Payam Farahbakhsh
- Tehran Heart Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Mehdi Yaseri
- Department of Epidemiology and Biostatistics, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Hossein Imani
- Department of Clinical Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
28
|
Lee Y, Mehrotra P, Basile D, Ullah M, Singh A, Skill N, Younes ST, Sasser J, Shekhar A, Singh J. Specific Lowering of Asymmetric Dimethylarginine by Pharmacological Dimethylarginine Dimethylaminohydrolase Improves Endothelial Function, Reduces Blood Pressure and Ischemia-Reperfusion Injury. J Pharmacol Exp Ther 2021; 376:181-189. [PMID: 33214214 PMCID: PMC7816763 DOI: 10.1124/jpet.120.000212] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 11/13/2020] [Indexed: 12/18/2022] Open
Abstract
Multiple clinical and preclinical studies have demonstrated that plasma levels of asymmetric dimethylarginine (ADMA) are strongly associated with hypertension, diabetes, and cardiovascular and renal disease. Genetic studies in rodents have provided evidence that ADMA metabolizing dimethylarginine dimethylaminohydrolase (DDAH)-1 plays a role in hypertension and cardiovascular disease. However, it remains to be established whether ADMA is a bystander, biomarker, or sufficient contributor to the pathogenesis of hypertension and cardiovascular and renal disease. The goal of the present investigation was to develop a pharmacological molecule to specifically lower ADMA and determine the physiologic consequences of ADMA lowering in animal models. Further, we sought to determine whether ADMA lowering will produce therapeutic benefits in vascular disease in which high ADMA levels are produced. A novel long-acting recombinant DDAH (M-DDAH) was produced by post-translational modification, which effectively lowered ADMA in vitro and in vivo. Treatment with M-DDAH improved endothelial function as measured by increase in cGMP and in vitro angiogenesis. In a rat model of hypertension, M-DDAH significantly reduced blood pressure (vehicle: 187 ± 19 mm Hg vs. M-DDAH: 157 ± 23 mm Hg; P < 0.05). Similarly, in a rat model of ischemia-reperfusion injury, M-DDAH significantly improved renal function as measured by reduction in serum creatinine (vehicle: 3.14 ± 0.74 mg/dl vs. M-DDAH: 1.1 ± 0.75 mg/dl; P < 0.01), inflammation, and injured tubules (vehicle: 73.1 ± 11.1% vs. M-DDAH: 22.1 ± 18.4%; P < 0.001). These pharmacological studies have provided direct evidence for a pathologic role of ADMA and the therapeutic benefits of ADMA lowering in preclinical models of endothelial dysfunction, hypertension, and ischemia-reperfusion injury. SIGNIFICANCE STATEMENT: High levels of ADMA occur in patients with cardiovascular and renal disease. A novel modified dimethylarginine dimethylaminohydrolase by PEGylation effectively lowers ADMA, improves endothelial function, reduces blood pressure and protects from ischemia-reperfusion renal injury.
Collapse
Affiliation(s)
- Young Lee
- Indiana Center for Biomedical Innovation, Indianapolis, Indiana (Y.L., Ar.S., J.S.); Indiana University School of Medicine, Indianapolis, Indiana (P.M., D.B., M.U., N.S., Ar.S., J.S.); University of Mississippi Medical Center, Jackson, Mississippi (S.T.Y., Je.S.); and Vasculonics LLC, Indianapolis, Indiana (J.S.)
| | - Purvi Mehrotra
- Indiana Center for Biomedical Innovation, Indianapolis, Indiana (Y.L., Ar.S., J.S.); Indiana University School of Medicine, Indianapolis, Indiana (P.M., D.B., M.U., N.S., Ar.S., J.S.); University of Mississippi Medical Center, Jackson, Mississippi (S.T.Y., Je.S.); and Vasculonics LLC, Indianapolis, Indiana (J.S.)
| | - David Basile
- Indiana Center for Biomedical Innovation, Indianapolis, Indiana (Y.L., Ar.S., J.S.); Indiana University School of Medicine, Indianapolis, Indiana (P.M., D.B., M.U., N.S., Ar.S., J.S.); University of Mississippi Medical Center, Jackson, Mississippi (S.T.Y., Je.S.); and Vasculonics LLC, Indianapolis, Indiana (J.S.)
| | - Mahbub Ullah
- Indiana Center for Biomedical Innovation, Indianapolis, Indiana (Y.L., Ar.S., J.S.); Indiana University School of Medicine, Indianapolis, Indiana (P.M., D.B., M.U., N.S., Ar.S., J.S.); University of Mississippi Medical Center, Jackson, Mississippi (S.T.Y., Je.S.); and Vasculonics LLC, Indianapolis, Indiana (J.S.)
| | - Arshnoor Singh
- Indiana Center for Biomedical Innovation, Indianapolis, Indiana (Y.L., Ar.S., J.S.); Indiana University School of Medicine, Indianapolis, Indiana (P.M., D.B., M.U., N.S., Ar.S., J.S.); University of Mississippi Medical Center, Jackson, Mississippi (S.T.Y., Je.S.); and Vasculonics LLC, Indianapolis, Indiana (J.S.)
| | - Nicholas Skill
- Indiana Center for Biomedical Innovation, Indianapolis, Indiana (Y.L., Ar.S., J.S.); Indiana University School of Medicine, Indianapolis, Indiana (P.M., D.B., M.U., N.S., Ar.S., J.S.); University of Mississippi Medical Center, Jackson, Mississippi (S.T.Y., Je.S.); and Vasculonics LLC, Indianapolis, Indiana (J.S.)
| | - Subhi Talal Younes
- Indiana Center for Biomedical Innovation, Indianapolis, Indiana (Y.L., Ar.S., J.S.); Indiana University School of Medicine, Indianapolis, Indiana (P.M., D.B., M.U., N.S., Ar.S., J.S.); University of Mississippi Medical Center, Jackson, Mississippi (S.T.Y., Je.S.); and Vasculonics LLC, Indianapolis, Indiana (J.S.)
| | - Jennifer Sasser
- Indiana Center for Biomedical Innovation, Indianapolis, Indiana (Y.L., Ar.S., J.S.); Indiana University School of Medicine, Indianapolis, Indiana (P.M., D.B., M.U., N.S., Ar.S., J.S.); University of Mississippi Medical Center, Jackson, Mississippi (S.T.Y., Je.S.); and Vasculonics LLC, Indianapolis, Indiana (J.S.)
| | - Anantha Shekhar
- Indiana Center for Biomedical Innovation, Indianapolis, Indiana (Y.L., Ar.S., J.S.); Indiana University School of Medicine, Indianapolis, Indiana (P.M., D.B., M.U., N.S., Ar.S., J.S.); University of Mississippi Medical Center, Jackson, Mississippi (S.T.Y., Je.S.); and Vasculonics LLC, Indianapolis, Indiana (J.S.)
| | - Jaipal Singh
- Indiana Center for Biomedical Innovation, Indianapolis, Indiana (Y.L., Ar.S., J.S.); Indiana University School of Medicine, Indianapolis, Indiana (P.M., D.B., M.U., N.S., Ar.S., J.S.); University of Mississippi Medical Center, Jackson, Mississippi (S.T.Y., Je.S.); and Vasculonics LLC, Indianapolis, Indiana (J.S.)
| |
Collapse
|
29
|
Kobayashi T, Ueda S, Takagi M, Kihara M, Suzuki Y. Pathophysiological roles of ADMA-mediated endothelial injury in hypertensive disorders of pregnancy. HYPERTENSION RESEARCH IN PREGNANCY 2020. [DOI: 10.14390/jsshp.hrp2019-013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
| | - Seiji Ueda
- Department of Nephrology, Juntendo University Faculty of Medicine
| | - Miyuki Takagi
- Department of Nephrology, Juntendo University Faculty of Medicine
| | - Masao Kihara
- Department of Nephrology, Juntendo University Faculty of Medicine
| | - Yusuke Suzuki
- Department of Nephrology, Juntendo University Faculty of Medicine
| |
Collapse
|
30
|
Ammar YA, Awad A. Effect of a Supervised Peridialytic Exercise Program on Serum Asymmetric Dimethylarginine in Maintenance Hemodialysis Patients. Int J Nephrol 2020; 2020:8878306. [PMID: 33163233 PMCID: PMC7604598 DOI: 10.1155/2020/8878306] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 09/12/2020] [Accepted: 10/05/2020] [Indexed: 12/03/2022] Open
Abstract
End-stage renal disease (ESRD) patients treated with maintenance haemodialysis (MHD) have alarmingly high atherosclerotic cardiovascular disease morbidity and mortality. Nitric oxide (NO) is the principal endogenous antiatherosclerotic molecule. Increased asymmetric dimethylarginine (ADMA), an endogenous NO synthase inhibitor, was strongly implicated in endothelial dysfunction, premature atherosclerosis, vascular events, and mortality. Regular physical exercise effectively decreased serum ADMA in several patient cohorts, but this potential benefit has not been specifically explored among MHD patients. Forty-four middle-aged ESRD patients treated with thrice-weekly MHD for ≥6 months completed a 6-months regimen of peridialytic lower limb exercise comprising predialytic 10-12 stretching cycles and 20-30 minutes of intradialytic pedaling cycles. Before and after the study, predialytic haemoglobin, serum ADMA, urea, creatinine, calcium, phosphorus, and C-reactive protein (CRP) were measured. Dialysis adequacy was assessed by single-pool Kt/V. The average total physical activity (PA) level was assessed by the International Physical Activity Questionnaire (IPAQ). P values <0.05 denoted a statistical significance. The overall level of PA, on both categorical and continuous scales, has significantly increased after application of the exercise program. However, S. ADMA increased from a median of 2375 to 3000 ng/mL (P=0.016). Thirty-one patients sustained an increase in S. ADMA (ADMA_Inc), whereas 13 patients had a declining or stable S. ADMA (ADMA_Dec). Compared with ADMA_Inc, ADMA_Dec patients had significantly higher Kt/V (P=0.02), higher grade of the basal general PA level (P=0.017), and significantly fewer intradialytic hypotension episodes (IDHs) (P=0.019). The increase in the S. ADMA and the poststudy S. ADMA level had statistically significant positive correlations with the number of IDHs (r = 0.401, P=0.007 and r = 0.305, P=0.044, respectively). A 6-month program of combined aerobic and resistance peridialytic exercise failed to reduce S. ADMA in most MHD patients studied. A modest S. ADMA decline, however, occurred in patients with higher basal PA levels, higher Kt/V, and less IDHs. A potential exercise benefit may be promoted by a multidisciplinary approach targeting increased PA, improved dialysis efficiency, and prevention of IDHs.
Collapse
Affiliation(s)
- Yaser A. Ammar
- Internal Medicine Department, Medical Research Institute, Alexandria University, Alexandria, Egypt
| | - Ahmad Awad
- Chemical Pathology Department, Medical Research Institute, Alexandria University, Alexandria, Egypt
| |
Collapse
|
31
|
Malden DE, Mangoni AA, Woodman RJ, Thies F, McNeil C, Murray AD, Soiza RL. Circulating asymmetric dimethylarginine and cognitive decline: A 4-year follow-up study of the 1936 Aberdeen Birth Cohort. Int J Geriatr Psychiatry 2020; 35:1181-1188. [PMID: 32452069 DOI: 10.1002/gps.5355] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 04/27/2020] [Accepted: 05/17/2020] [Indexed: 12/24/2022]
Abstract
BACKGROUND The underlying mechanisms leading to dementia and Alzheimer's disease (AD) are unclear. Asymmetric dimethylarginine (ADMA), an endogenous inhibitor of nitric oxide synthase, may be associated with cognitive decline, but population-based evidence is lacking. METHODS Change in cognitive performance was assessed in participants of the Aberdeen Birth Cohort of 1936 using longitudinal Raven's progressive matrices (RPM) between 2000 and 2004. Multiple linear regression was used to estimate the association between ADMA concentrations in 2000 and change in cognitive performance after adjustment for potential confounders. RESULTS A total of 93 participants had complete information on cognitive performance between 2000 and 2004. Mean plasma ADMA concentrations were approximately 0.4 μmol/L lower in those participants with stable or improved RPM scores over follow-up compared with participants whose cognitive performance worsened. In confounder-adjusted analysis, one SD (0.06 μmol/L) increase in ADMA at 63 years of age was associated with an average reduction in RPM of 1.26 points (95% CI 0.14-2.26) after 4 years. CONCLUSION Raised plasma ADMA concentrations predicted worsening cognitive performance after approximately 4 years in this cohort of adults in late-middle age. These findings have implications for future research, including presymptomatic diagnosis or novel therapeutic targets for dementia and AD.
Collapse
Affiliation(s)
- Deborah E Malden
- Clinical Trial Service Unit & Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health (NDPH), University of Oxford, Oxford, UK
| | - Arduino A Mangoni
- Discipline of Clinical Pharmacology, College of Medicine and Public Health, Flinders University and Flinders Medical Centre, Adelaide, Australia.,Medizinische Fakultät Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Richard J Woodman
- Flinders Centre for Epidemiology and Biostatistics, College of Medicine and Public Health, Flinders University, Adelaide, Australia
| | - Frank Thies
- Rowett institute, University of Aberdeen, Aberdeen, UK
| | - Chris McNeil
- Aberdeen Biomedical Imaging Centre, University of Aberdeen, Aberdeen, UK
| | - Alison D Murray
- Aberdeen Biomedical Imaging Centre, University of Aberdeen, Aberdeen, UK
| | - Roy L Soiza
- Ageing Clinical & Experimental Research (ACER), University of Aberdeen, Aberdeen, UK
| |
Collapse
|
32
|
Rashidi BH, Mohammad Hosseinzadeh F, Alipoor E, Asghari S, Yekaninejad MS, Hosseinzadeh-Attar MJ. Effects of Selenium Supplementation on Asymmetric Dimethylarginine and Cardiometabolic Risk Factors in Patients with Polycystic Ovary Syndrome. Biol Trace Elem Res 2020; 196:430-437. [PMID: 31667685 DOI: 10.1007/s12011-019-01954-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Accepted: 10/23/2019] [Indexed: 12/26/2022]
Abstract
Polycystic ovary syndrome (PCOS) is characterized by various reproductive and cardiometabolic disorders. Asymmetric dimethylarginine (ADMA) is associated with cardiovascular, metabolic, and hormonal status. Selenium, a micronutrient with antioxidant properties, could affect multiple physiological pathways. This study aimed to investigate the effect of selenium supplementation on ADMA, cardiometabolic risk factors, and hormonal status in women with PCOS. In this randomized, double-blind, placebo-controlled clinical trial, 66 women with PCOS, aged 18-45 years, were randomly assigned to receive either 200 μg/day selenium or placebo, for 12 weeks. Circulating concentrations of ADMA, testosterone, sex hormone-binding globulin (SHBG), lipid profiles, and glycemic parameters were assessed at baseline and following supplementation. ADMA concentration decreased significantly compared to baseline values (85.14 ± 75 to 56.4 ± 38.64 ng/l, p = 0.02) in the selenium group. This change was marginally significant compared with the placebo group (28.74 ± 68.63 vs. - 1.77 ± 52.88 ng/l, p = 0.056). Serum testosterone levels declined significantly in the intervention compared to the placebo group (0.01 ± 0.17 vs. - 0.08 ± 0.18 ng/ml, p = 0.038). Pre- to post-Apo-B100/Apo-A1 ratio declined considerably in the intervention group (0.72 ± 0.16 to 0.65 ± 0.16, p = 0.003). No further differences were observed in SHBG, lipid profiles, Apo-A1, Apo-B100, Apo-B100/Apo-A1 ratio, and glycemic control between the two groups at the end of the study. Selenium supplementation for 12 weeks had beneficial effects on reduction of circulating ADMA and total testosterone levels in women with PCOS. No significant improvements were seen in other cardiometabolic risk factors. The effects of selenium supplementation on hormonal, reproductive, and cardiometabolic disorders, considering the potential mediating role of ADMA, should be further investigated.
Collapse
Affiliation(s)
- Batool Hossein Rashidi
- Vali-E-Asr Reproductive Health Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Mohammad Hosseinzadeh
- Department of Clinical Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, No#44, Hojjatdoust St., Naderi St., Keshavarz Blvd, Tehran, 141556117, Iran
| | - Elham Alipoor
- Department of Nutrition, School of Public Health, Iran University of Medical Sciences, Tehran, Iran
| | - Somayyeh Asghari
- Department of Clinical Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, No#44, Hojjatdoust St., Naderi St., Keshavarz Blvd, Tehran, 141556117, Iran
| | - Mir Saeed Yekaninejad
- Department of Epidemiology and Biostatistics, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Javad Hosseinzadeh-Attar
- Department of Clinical Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, No#44, Hojjatdoust St., Naderi St., Keshavarz Blvd, Tehran, 141556117, Iran.
- Cardiac Primary Prevention Research Center (CPPRC), Tehran Heart Center, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
33
|
Urinary Dimethylamine (DMA) and Its Precursor Asymmetric Dimethylarginine (ADMA) in Clinical Medicine, in the Context of Nitric Oxide (NO) and Beyond. J Clin Med 2020; 9:jcm9061843. [PMID: 32545708 PMCID: PMC7356952 DOI: 10.3390/jcm9061843] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 06/08/2020] [Accepted: 06/09/2020] [Indexed: 02/08/2023] Open
Abstract
Asymmetric protein-arginine dimethylation is a major post-translational modification (PTM) catalyzed by protein-arginine methyltransferase (PRMT). Regular proteolysis releases asymmetric dimethylarginine (ADMA). Of the daily produced ADMA, about 10% are excreted unchanged in the urine. The remaining 90% are hydrolyzed by dimethylarginine dimethylaminohydrolase (DDAH) to L-citrulline and dimethylamine (DMA), which is readily excreted in the urine. The PRMT/DDAH pathway is almost the exclusive origin of urinary ADMA and the major source of urinary DMA. Dietary fish and seafood represent additional abundant sources of urinary DMA. The present article provides an overview of urinary ADMA and DMA reported thus far in epidemiological, clinical and pharmacological studies, in connection with the L-arginine/nitric oxide (NO) pathway and beyond, in neonates, children and adolescents, young and elderly subjects, males and females. Discussed diseases mainly include those relating to the renal and cardiovascular systems such as peripheral arterial occlusive disease, coronary artery disease, chronic kidney disease, rheumatoid arthritis, Becker muscular disease, Duchenne muscular disease (DMD), attention deficit hyperactivity disorder (ADHD), and type I diabetes. Under standardized conditions involving the abstinence of DMA-rich fresh and canned fish and seafood, urinary DMA and ADMA are useful as measures of whole-body asymmetric arginine-dimethylation in health and disease. The creatinine-corrected excretion rates of DMA range from 10 to 80 µmol/mmol in adults and up to 400 µmol/mmol in children and adolescents. The creatinine-corrected excretion rates of ADMA are on average 10 times lower. In general, diseases are associated with higher urinary DMA and ADMA excretion rates, and pharmacological treatment, such as with steroids and creatine (in DMD), decreases their excretion rates, which may be accompanied by a decreased urinary excretion of nitrate, the major metabolite of NO. In healthy subjects and in rheumatoid arthritis patients, the urinary excretion rate of DMA correlates positively with the excretion rate of dihydroxyphenylglycol (DHPG), the major urinary catecholamines metabolite, suggesting a potential interplay in the PRMT/DDAH/NO pathway.
Collapse
|
34
|
Discovery of N-(4-Aminobutyl)- N'-(2-methoxyethyl)guanidine as the First Selective, Nonamino Acid, Catalytic Site Inhibitor of Human Dimethylarginine Dimethylaminohydrolase-1 ( hDDAH-1). J Med Chem 2020; 63:425-432. [PMID: 31841335 DOI: 10.1021/acs.jmedchem.9b01230] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
N-(4-Aminobutyl)-N'-(2-methoxyethyl)guanidine (8a) is a potent inhibitor targeting the hDDAH-1 active site (Ki = 18 μM) and derived from a series of guanidine- and amidine-based inhibitors. Its nonamino acid nature leads to high selectivities toward other enzymes of the nitric oxide-modulating system. Crystallographic data of 8a-bound hDDAH-1 illuminated a unique binding mode. Together with its developed N-hydroxyguanidine prodrug 11, 8a will serve as a most widely applicable, pharmacological tool to target DDAH-1-associated diseases.
Collapse
|
35
|
Inhibition of Dimethylarginine Dimethylaminohydrolase 1 Improves the Outcome of Sepsis in Pregnant Mice. Shock 2019; 54:498-506. [PMID: 31821207 DOI: 10.1097/shk.0000000000001490] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
36
|
Premont RT, Reynolds JD, Zhang R, Stamler JS. Role of Nitric Oxide Carried by Hemoglobin in Cardiovascular Physiology: Developments on a Three-Gas Respiratory Cycle. Circ Res 2019; 126:129-158. [PMID: 31590598 DOI: 10.1161/circresaha.119.315626] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
A continuous supply of oxygen is essential for the survival of multicellular organisms. The understanding of how this supply is regulated in the microvasculature has evolved from viewing erythrocytes (red blood cells [RBCs]) as passive carriers of oxygen to recognizing the complex interplay between Hb (hemoglobin) and oxygen, carbon dioxide, and nitric oxide-the three-gas respiratory cycle-that insures adequate oxygen and nutrient delivery to meet local metabolic demand. In this context, it is blood flow and not blood oxygen content that is the main driver of tissue oxygenation by RBCs. Herein, we review the lines of experimentation that led to this understanding of RBC function; from the foundational understanding of allosteric regulation of oxygen binding in Hb in the stereochemical model of Perutz, to blood flow autoregulation (hypoxic vasodilation governing oxygen delivery) observed by Guyton, to current understanding that centers on S-nitrosylation of Hb (ie, S-nitrosohemoglobin; SNO-Hb) as a purveyor of oxygen-dependent vasodilatory activity. Notably, hypoxic vasodilation is recapitulated by native S-nitrosothiol (SNO)-replete RBCs and by SNO-Hb itself, whereby SNO is released from Hb and RBCs during deoxygenation, in proportion to the degree of Hb deoxygenation, to regulate vessels directly. In addition, we discuss how dysregulation of this system through genetic mutation in Hb or through disease is a common factor in oxygenation pathologies resulting from microcirculatory impairment, including sickle cell disease, ischemic heart disease, and heart failure. We then conclude by identifying potential therapeutic interventions to correct deficits in RBC-mediated vasodilation to improve oxygen delivery-steps toward effective microvasculature-targeted therapies. To the extent that diseases of the heart, lungs, and blood are associated with impaired tissue oxygenation, the development of new therapies based on the three-gas respiratory system have the potential to improve the well-being of millions of patients.
Collapse
Affiliation(s)
- Richard T Premont
- From the Institute for Transformative Molecular Medicine (R.T.P., J.D.R., R.Z., J.S.S.), Case Western Reserve University School of Medicine, OH.,Harrington Discovery Institute (R.T.P., J.D.R., J.S.S.), University Hospitals Cleveland Medical Center, OH
| | - James D Reynolds
- From the Institute for Transformative Molecular Medicine (R.T.P., J.D.R., R.Z., J.S.S.), Case Western Reserve University School of Medicine, OH.,Department of Anesthesiology and Perioperative Medicine (J.D.R.), Case Western Reserve University School of Medicine, OH.,Harrington Discovery Institute (R.T.P., J.D.R., J.S.S.), University Hospitals Cleveland Medical Center, OH
| | - Rongli Zhang
- From the Institute for Transformative Molecular Medicine (R.T.P., J.D.R., R.Z., J.S.S.), Case Western Reserve University School of Medicine, OH.,Department of Medicine, Cardiovascular Research Institute (R.Z., J.S.S.), Case Western Reserve University School of Medicine, OH
| | - Jonathan S Stamler
- From the Institute for Transformative Molecular Medicine (R.T.P., J.D.R., R.Z., J.S.S.), Case Western Reserve University School of Medicine, OH.,Department of Medicine, Cardiovascular Research Institute (R.Z., J.S.S.), Case Western Reserve University School of Medicine, OH.,Harrington Discovery Institute (R.T.P., J.D.R., J.S.S.), University Hospitals Cleveland Medical Center, OH
| |
Collapse
|
37
|
Huang H, Huang M, Lv W, Hu Y, Wang R, Zheng X, Ma Y, Chen C, Tang H. Inhibition of Trichophyton rubrum by 420-nm Intense Pulsed Light: In Vitro Activity and the Role of Nitric Oxide in Fungal Death. Front Pharmacol 2019; 10:1143. [PMID: 31632277 PMCID: PMC6785631 DOI: 10.3389/fphar.2019.01143] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2019] [Accepted: 09/04/2019] [Indexed: 01/06/2023] Open
Abstract
Trichophyton rubrum is a common dermatophyte of the skin. The aim of this experiment was to explore the role of nitric oxide (NO) in the inhibition of T. rubrum growth induced by 420-nm intense pulsed light (IPL). This study found that nitric oxide synthase (NOS) and NO levels were increased, whereas asymmetric dimethylarginine (ADMA) level, keratinase activity, and fungal viability were decreased after IPL treatment compared with the control condition in vitro. Moreover, micromorphology was damaged by IPL treatment. Fungal viability was increased, and the damage to the fungal structure was reduced after pretreatment with an NOS inhibitor (L-NMMA) compared with IPL treatment alone. Compared with IPL alone, pretreatment with L-NMMA decreased NOS expression and NO level and increased keratinase activity. We found that 420-nm IPL treatment can inhibit the growth of T. rubrum by regulating NO in vitro.
Collapse
Affiliation(s)
- Hao Huang
- Department of Dermatology, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde Foshan), Foshan, China
| | - Meiling Huang
- Department of Dermatology, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde Foshan), Foshan, China
| | - Wenyi Lv
- Department of Dermatology, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde Foshan), Foshan, China
| | - Yong Hu
- Department of Dermatology, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde Foshan), Foshan, China
| | - Ruihua Wang
- Department of Dermatology, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde Foshan), Foshan, China
| | - Xiufen Zheng
- Department of Dermatology, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde Foshan), Foshan, China
| | - Yuetang Ma
- Department of Dermatology, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde Foshan), Foshan, China
| | - Chunmei Chen
- Department of Dermatology, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde Foshan), Foshan, China
| | - Hongfeng Tang
- Department of Dermatology, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde Foshan), Foshan, China
| |
Collapse
|
38
|
Ricciotti E, Castro C, Tang SY, Briggs WTE, West JA, Malik D, Rhoades SD, Meng H, Li X, Lahens NF, Sparks JA, Karlson EW, Weljie AM, Griffin JL, FitzGerald GA. Cyclooxygenase-2, Asymmetric Dimethylarginine, and the Cardiovascular Hazard From Nonsteroidal Anti-Inflammatory Drugs. Circulation 2019; 138:2367-2378. [PMID: 29930022 DOI: 10.1161/circulationaha.118.033540] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND Large-scale, placebo-controlled trials established that nonsteroidal anti-inflammatory drugs confer a cardiovascular hazard: this has been attributed to depression of cardioprotective products of cyclooxygenase (COX)-2, especially prostacyclin. An alternative mechanism by which nonsteroidal anti-inflammatory drugs might constrain cardioprotection is by enhancing the formation of methylarginines in the kidney that would limit the action of nitric oxide throughout the vasculature. METHODS Targeted and untargeted metabolomics were used to investigate the effect of COX-2 deletion or inhibition in mice and in osteoarthritis patients exposed to nonsteroidal anti-inflammatory drugs on the l-arginine/nitric oxide pathway. RESULTS Analysis of the plasma and renal metabolome was performed in postnatal tamoxifen-inducible Cox-2 knockout mice, which exhibit normal renal function and blood pressure. This revealed no changes in arginine and methylarginines compared with their wild-type controls. Moreover, the expression of genes in the l-arginine/nitric oxide pathway was not altered in the renal medulla or cortex of tamoxifen inducible Cox-2 knockout mice. Therapeutic concentrations of the selective COX-2 inhibitors, rofecoxib, celecoxib, and parecoxib, none of which altered basal blood pressure or renal function as reflected by plasma creatinine, failed to elevate plasma arginine and methylarginines in mice. Finally, plasma arginine or methylarginines were not altered in osteoarthritis patients with confirmed exposure to nonsteroidal anti-inflammatory drugs that inhibit COX-1 and COX-2. By contrast, plasma asymmetrical dimethylarginine was increased in mice infused with angiotensin II sufficient to elevate blood pressure and impair renal function. Four weeks later, blood pressure, plasma creatinine, and asymmetrical dimethylarginine were restored to normal levels. The increase in asymmetrical dimethylarginine in response to infusion with angiotensin II in celecoxib-treated mice was also related to transient impairment of renal function. CONCLUSIONS Plasma methylarginines are not altered by COX-2 deletion or inhibition but rather are elevated coincident with renal compromise.
Collapse
Affiliation(s)
- Emanuela Ricciotti
- Department of Systems Pharmacology and Translational Therapeutics and the Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, Philadelphia, PA (E.R., S.Y.T., D.M., S.D.R., H.M., X.L., N.F.L., A.M.W., G.A.F.)
| | - Cecilia Castro
- Department of Biochemistry, Cambridge Systems Biology Centre, University of Cambridge, United Kingdom (C.C., W.T.E.B., J.A.W., J.L.G.)
| | - Soon Yew Tang
- Department of Systems Pharmacology and Translational Therapeutics and the Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, Philadelphia, PA (E.R., S.Y.T., D.M., S.D.R., H.M., X.L., N.F.L., A.M.W., G.A.F.)
| | - William T E Briggs
- Department of Biochemistry, Cambridge Systems Biology Centre, University of Cambridge, United Kingdom (C.C., W.T.E.B., J.A.W., J.L.G.)
| | - James A West
- Department of Biochemistry, Cambridge Systems Biology Centre, University of Cambridge, United Kingdom (C.C., W.T.E.B., J.A.W., J.L.G.)
| | - Dania Malik
- Department of Systems Pharmacology and Translational Therapeutics and the Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, Philadelphia, PA (E.R., S.Y.T., D.M., S.D.R., H.M., X.L., N.F.L., A.M.W., G.A.F.)
| | - Seth D Rhoades
- Department of Systems Pharmacology and Translational Therapeutics and the Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, Philadelphia, PA (E.R., S.Y.T., D.M., S.D.R., H.M., X.L., N.F.L., A.M.W., G.A.F.)
| | - Hu Meng
- Department of Systems Pharmacology and Translational Therapeutics and the Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, Philadelphia, PA (E.R., S.Y.T., D.M., S.D.R., H.M., X.L., N.F.L., A.M.W., G.A.F.)
| | - Xuanwen Li
- Department of Systems Pharmacology and Translational Therapeutics and the Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, Philadelphia, PA (E.R., S.Y.T., D.M., S.D.R., H.M., X.L., N.F.L., A.M.W., G.A.F.)
| | - Nicholas F Lahens
- Department of Systems Pharmacology and Translational Therapeutics and the Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, Philadelphia, PA (E.R., S.Y.T., D.M., S.D.R., H.M., X.L., N.F.L., A.M.W., G.A.F.)
| | - Jeffrey A Sparks
- Department of Medicine, Division of Rheumatology, Immunology, and Allergy, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (J.A.S., E.W.K.)
| | - Elizabeth W Karlson
- Department of Medicine, Division of Rheumatology, Immunology, and Allergy, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (J.A.S., E.W.K.)
| | - Aalim M Weljie
- Department of Systems Pharmacology and Translational Therapeutics and the Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, Philadelphia, PA (E.R., S.Y.T., D.M., S.D.R., H.M., X.L., N.F.L., A.M.W., G.A.F.)
| | - Julian L Griffin
- Department of Biochemistry, Cambridge Systems Biology Centre, University of Cambridge, United Kingdom (C.C., W.T.E.B., J.A.W., J.L.G.)
| | - Garret A FitzGerald
- Department of Systems Pharmacology and Translational Therapeutics and the Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, Philadelphia, PA (E.R., S.Y.T., D.M., S.D.R., H.M., X.L., N.F.L., A.M.W., G.A.F.)
| |
Collapse
|
39
|
Joris V, Gomez EL, Menchi L, Lobysheva I, Di Mauro V, Esfahani H, Condorelli G, Balligand JL, Catalucci D, Dessy C. MicroRNA-199a-3p and MicroRNA-199a-5p Take Part to a Redundant Network of Regulation of the NOS (NO Synthase)/NO Pathway in the Endothelium. Arterioscler Thromb Vasc Biol 2019; 38:2345-2357. [PMID: 29976767 DOI: 10.1161/atvbaha.118.311145] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Objective- Members of the microRNA (miR)-199a family, namely miR-199a-5p and miR-199a-3p, have been recently identified as potential regulators of cardiac homeostasis. Also, upregulation of miR-199a expression in cardiomyocytes was reported to influence endothelial cells. Whether miR-199a is expressed by endothelial cells and, if so, whether it directly regulates endothelial function remains unknown. We investigate the implication of miR-199a products on endothelial function by focusing on the NOS (nitric oxide synthase)/NO pathway. Approach and Results- Bovine aortic endothelial cells were transfected with specific miRNA inhibitors (locked-nucleic acids), and potential molecular targets identified with prediction algorithms were evaluated by Western blot or immunofluorescence. Ex vivo experiments were performed with mice treated with antagomiRs targeting miR-199a-3p or -5p. Isolated vessels and blood were used for electron paramagnetic resonance or myograph experiments. eNOS (endothelial NO synthase) activity (through phosphorylations Ser1177/Thr495) is increased by miR-199a-3p/-5p inhibition through an upregulation of the PI3K (phosphoinositide 3-kinase)/Akt (protein kinase B) and calcineurin pathways. SOD1 (superoxide dismutase 1) and PRDX1 (peroxiredoxin 1) upregulation was also observed in locked-nucleic acid-treated cells. Moreover, miR-199a-5p controls angiogenesis and VEGFA (vascular endothelial growth factor A) production and upregulation of NO-dependent relaxation were observed in vessels from antagomiR-treated mice. This was correlated with increased circulated hemoglobin-NO levels and decreased superoxide production. Angiotensin infusion for 2 weeks also revealed an upregulation of miR-199a-3p/-5p in vascular tissues. Conclusions- Our study reveals that miR-199a-3p and miR-199a-5p participate in a redundant network of regulation of the NOS/NO pathway in the endothelium. We highlighted that inhibition of miR-199a-3p and -5p independently increases NO bioavailability by promoting eNOS activity and reducing its degradation, thereby supporting VEGF-induced endothelial tubulogenesis and modulating vessel contractile tone.
Collapse
Affiliation(s)
- Virginie Joris
- From the Pole of Pharmacology and Therapeutics, Experimental and Clinical Research Institute (IREC), Université catholique de Louvain, Brussels, Belgium (V.J., E.L.G., L.M., H.E., J.-L.B., C.D.)
| | - Elvira Leon Gomez
- From the Pole of Pharmacology and Therapeutics, Experimental and Clinical Research Institute (IREC), Université catholique de Louvain, Brussels, Belgium (V.J., E.L.G., L.M., H.E., J.-L.B., C.D.)
| | - Lisa Menchi
- From the Pole of Pharmacology and Therapeutics, Experimental and Clinical Research Institute (IREC), Université catholique de Louvain, Brussels, Belgium (V.J., E.L.G., L.M., H.E., J.-L.B., C.D.)
| | | | - Vittoria Di Mauro
- Humanitas University, Rozzano (Milan), Italy (V.D.M., G.C.).,Humanitas#8232, Clinical and Research Center, Rozzano (Milan), Italy (V.D.M., G.C., D.C.).,Institute of Genetics and Biomedical Research, Milan Unit, National Research Council, Italy (V.D.M., G.C., D.C.)
| | - Hrag Esfahani
- From the Pole of Pharmacology and Therapeutics, Experimental and Clinical Research Institute (IREC), Université catholique de Louvain, Brussels, Belgium (V.J., E.L.G., L.M., H.E., J.-L.B., C.D.)
| | - Gianluigi Condorelli
- Humanitas University, Rozzano (Milan), Italy (V.D.M., G.C.).,Humanitas#8232, Clinical and Research Center, Rozzano (Milan), Italy (V.D.M., G.C., D.C.).,Institute of Genetics and Biomedical Research, Milan Unit, National Research Council, Italy (V.D.M., G.C., D.C.)
| | - Jean-Luc Balligand
- From the Pole of Pharmacology and Therapeutics, Experimental and Clinical Research Institute (IREC), Université catholique de Louvain, Brussels, Belgium (V.J., E.L.G., L.M., H.E., J.-L.B., C.D.)
| | - Daniele Catalucci
- Humanitas#8232, Clinical and Research Center, Rozzano (Milan), Italy (V.D.M., G.C., D.C.).,Institute of Genetics and Biomedical Research, Milan Unit, National Research Council, Italy (V.D.M., G.C., D.C.)
| | - Chantal Dessy
- From the Pole of Pharmacology and Therapeutics, Experimental and Clinical Research Institute (IREC), Université catholique de Louvain, Brussels, Belgium (V.J., E.L.G., L.M., H.E., J.-L.B., C.D.)
| |
Collapse
|
40
|
Genetic regulation of dimethylarginines and endothelial dysfunction in rheumatoid arthritis. Amino Acids 2019; 51:983-990. [PMID: 31062169 DOI: 10.1007/s00726-019-02740-3] [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: 03/03/2019] [Accepted: 05/01/2019] [Indexed: 12/21/2022]
Abstract
Rheumatoid Arthritis (RA) confers an increased cardiovascular disease (CVD) risk which accounts for much of the premature morbidity and mortality observed in this population. Alterations in vascular function and morphology leading to increased atherosclerotic burden are considered the main drivers of CVD in RA individuals with systemic inflammation playing a key role in the dysregulation of endothelial homeostasis and initiation of vascular injury. Dimethylarginines are endogenous inhibitors of nitric oxide (NO) synthase and have emerged as novel, independent biomarkers of CVD in a wide range of conditions associated with vascular pathology. In RA several reports have demonstrated abnormal dimethylarginine metabolism attributable to various factors such as systemic inflammation, decreased degradation or upregulated synthesis. Although a causal relationship between dimethylarginines and vascular damage in RA has not been established, the tight interrelations between inflammation, dimethylarginines and endothelial dysfunction suggest that determination of dimethylarginine regulators may shed more light in the pathophysiology of the atherosclerotic process in RA and may also provide new therapeutic targets. The Alanine-Glyoxylate Aminotransferase 2 (AGTX2)-dependent pathway is a relatively recently discovered alternative pathway of dimethylarginine catabolism and its role on RA-related atherosclerotic disease is yet to be established. As factors affecting dimethylarginine concentrations linked to CVD risk and endothelial dysfunction are of prominent clinical relevance in RA, we present preliminary evidence that gene variants of AGTX-2 may influence dimethylarginine levels in RA patients and provide the rationale for larger studies in this field.
Collapse
|
41
|
Zhao WC, Li G, Huang CY, Jiang JL. Asymmetric dimethylarginine: An crucial regulator in tissue fibrosis. Eur J Pharmacol 2019; 854:54-61. [PMID: 30951718 DOI: 10.1016/j.ejphar.2019.03.055] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 03/21/2019] [Accepted: 03/28/2019] [Indexed: 02/06/2023]
Abstract
Fibrosis is a reparative process with very few therapeutic options to prevent its progression to organ dysfunction. Chronic fibrotic diseases contribute to an estimated 45% of all death in the industrialized world. Asymmetric dimethylarginine (ADMA), an endothelial nitric oxide synthase inhibitor, plays a crucial role in the pathogenesis of various cardiovascular diseases associated with endothelial dysfunction. Recent reports have focused on ADMA in the pathogenesis of tissue fibrosis. This review discusses the current knowledge about ADMA biology, its association with risk factors of established fibrotic diseases and the potential pathophysiological mechanisms implicating ADMA in the process of tissue fibrosis.
Collapse
Affiliation(s)
- Wei-Chen Zhao
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410078, China
| | - Ge Li
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410078, China; Faculty of Medical Public Courses, Xinhua College of Sun Yat-sen University, Guangzhou, Guangdong, 510520, China
| | - Chu-Yi Huang
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410078, China
| | - Jun-Lin Jiang
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410078, China; Provincial Key Laboratory of Cardiovascular Research, Central South University, Changsha, 410078, China.
| |
Collapse
|
42
|
Hulin JA, Tommasi S, Elliot D, Mangoni AA. Small molecule inhibition of DDAH1 significantly attenuates triple negative breast cancer cell vasculogenic mimicry in vitro. Biomed Pharmacother 2019; 111:602-612. [PMID: 30611984 DOI: 10.1016/j.biopha.2018.12.117] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Revised: 12/14/2018] [Accepted: 12/29/2018] [Indexed: 12/21/2022] Open
Abstract
Dimethylarginine dimethylaminohydrolase 1 (DDAH1) is a key enzyme involved in the metabolism of the endogenous nitric oxide synthase (NOS) inhibitors asymmetric dimethylarginine (ADMA) and monomethyl arginine (L-NMMA). Increased DDAH1 expression and subsequent increased NO production have been recently linked to cancer. Specifically, DDAH1 is implicated in establishment of a vascular network by tumour cells, vasculogenic mimicry (VM), which is strongly associated with tumour progression and poor patient prognosis. The use of DDAH1 inhibitors as potential therapeutic agents thus represents a growing field of interest. Here we describe a UPLC-MS assay to quantify stability and intracellular concentration of two small molecule DDAH1 inhibitors synthesised by our group, ZST316 and ZST152, following incubation with MDA-MB-231 breast cancer cells. In an in vitro assay of VM, both DDAH1 inhibitors significantly attenuated formation of capillary-like tube structures in a dose-dependent fashion. This was not due to cell toxicity or altered cell proliferation, but may be due in part to inhibition of cell migration. Mechanistically, we demonstrate significant modulation of the endogenous DDAH/ADMA/NO pathway following exposure of 100 μM ZST316 or ZST152: a 40% increase in the DDAH1 substrate ADMA, and a 38% decrease in the DDAH1 product l-citrulline. This study represents the first evidence for therapeutic inhibition of DDAH1 by small molecules in breast cancer.
Collapse
Affiliation(s)
- Julie-Ann Hulin
- Clinical Pharmacology, College of Medicine and Public Health, Flinders University and Flinders Medical Centre, Bedford Park, South Australia, Australia; Flinders Centre for Innovation in Cancer, Bedford Park, South Australia, Australia.
| | - Sara Tommasi
- Clinical Pharmacology, College of Medicine and Public Health, Flinders University and Flinders Medical Centre, Bedford Park, South Australia, Australia
| | - David Elliot
- Clinical Pharmacology, College of Medicine and Public Health, Flinders University and Flinders Medical Centre, Bedford Park, South Australia, Australia
| | - Arduino A Mangoni
- Clinical Pharmacology, College of Medicine and Public Health, Flinders University and Flinders Medical Centre, Bedford Park, South Australia, Australia
| |
Collapse
|
43
|
Azizi F, Omrani MD, Amiri V, Mirfakhraie R, Dodangeh F, Shahmirzadi SA, Gargari SS. Altered methylation and expression patterns of genes regulating placental nitric oxide pathway in patients with severe preeclampsia. Hum Antibodies 2018; 27:117-124. [PMID: 30594923 DOI: 10.3233/hab-180356] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Pre-eclampsia is a common pregnancy disorder syndrome whose molecular mechanism is not clear. Nitric oxide (NO) is a key regulator of placentation. Reduction of NO has previously been associated with endothelial dysfunction in pre-eclamptic women. Therefore, we measured expression and methylation of some placental genes that were involved in NO pathway like named ARG II, PRMT1 and DDAH2 in pre-eclampsia and normal pregnancies in order to determine whether impairment of expression of these genes in the pre-eclamptic placenta could contribute to development of disease. ARG II, PRMT1 expressions as well as DDAH2 expression and methylation, in placentas collected from 59 patients with preeclampsia and 40 normotensive pregnancies were measured using real-time PCR and methylation specific PCR, respectively. The relationship among ARG II, PRMT1 and DDAH2 expressions was analyzed statistically. ARG II expression was increased, PRMT1 expression was not significantly changed. DDAH2 expression was decreased and qualitative methylation patterns were 32/59 and 21/40 in placentas from patients with pre-eclampsia compared with control group, respectively. The alterations in ARG II and DDAH2 expressions in pre-eclampsia patients maybe correlated with decreased eNOS expression. These findings indicate that ARG II and DDAH2 may be involved in pre-eclampsia pathogenesis and could be potential therapeutic targets for this disease.
Collapse
Affiliation(s)
- Faezeh Azizi
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mir Davood Omrani
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Vahid Amiri
- School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Reza Mirfakhraie
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Dodangeh
- Feto-Maternal Unit, Mahdiyeh Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Soraya Saleh Gargari
- Feto-Maternal Unit, Shohadaye Tajrish Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
44
|
Alipoor E, Mohammad Hosseinzadeh F, Hosseinzadeh-Attar MJ. Adipokines in critical illness: A review of the evidence and knowledge gaps. Biomed Pharmacother 2018; 108:1739-1750. [PMID: 30372877 DOI: 10.1016/j.biopha.2018.09.165] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Revised: 09/27/2018] [Accepted: 09/28/2018] [Indexed: 12/28/2022] Open
Abstract
Adipose tissue products or adipokines play a major role in chronic endocrine and metabolic disorders; however, little is known about critical conditions. In this article, the experimental and clinical evidence of alterations of adipokines, adiponectin, leptin, resistin, visfatin, asymmetric dimethylarginine (ADMA), and ghrelin in critical illness, their potential metabolic, diagnostic, and prognostic value, and the gaps in the field have been reviewed. The results showed considerable changes in the concentration of the adipokines; while the impact of adipokines on metabolic disorders such as insulin resistance and inflammation has not been well documented in critically ill patients. There is no consensus about the circulatory and functional changes of leptin and adiponectin. However, it seems that lower concentrations of adiponectin at admission with gradual consequent increase might be a useful pattern in determining better outcomes of critical illness. Some evidence has suggested the adverse effects of elevated resistin concentration, potential prognostic importance of visfatin, and therapeutic value of ghrelin. High ADMA levels and low arginine:ADMA ratio were also proposed as predictors of ICU mortality and morbidities. However, there is no consensus on these findings. Although primary data indicated the role of adipokines in critical illness, further studies are required to clarify whether the reason of these changes is pathophysiological or compensatory. The relationship of pathophysiological background, disease severity, baseline nutritional status and nutrition support during hospitalization, and variations in body fat percentage and distribution with adipokines, as well as the potential prognostic or therapeutic role of these peptides should be further investigated in critically ill patients.
Collapse
Affiliation(s)
- Elham Alipoor
- Department of Nutrition, School of Public Health, Iran University of Medical Sciences, Tehran, Iran; Department of Clinical Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Mohammad Hosseinzadeh
- Department of Clinical Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Javad Hosseinzadeh-Attar
- Department of Clinical Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran; Centre of Research Excellence in Translating Nutritional Science to Good Health, University of Adelaide, Adelaide, Australia.
| |
Collapse
|
45
|
Burstein-Teitelbaum G, Er JAV, Monzingo AF, Tuley A, Fast W. Dissection, Optimization, and Structural Analysis of a Covalent Irreversible DDAH1 Inhibitor. Biochemistry 2018; 57:4574-4582. [PMID: 29983043 DOI: 10.1021/acs.biochem.8b00554] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Inhibitors of the human enzyme dimethylarginine dimethylaminohydrolase-1 (DDAH1) can control endogenous nitric oxide production. A time-dependent covalent inactivator of DDAH1, N5-(1-imino-2-chloroethyl)-l-ornithine ( KI = 1.3 μM, kinact = 0.34 min-1), was conceptually dissected into two fragments and each characterized separately: l-norvaline ( Ki = 470 μM) and 2-chloroacetamidine ( KI = 310 μM, kinact = 4.0 min-1). This analysis suggested that the two fragments were not linked in a manner that allows either to reach full affinity or reactivity, prompting the synthesis and characterization of three analogues: two that mimic the dimethylation status of the substrate, N5-(1-imino-2-chloroisopropyl)-l-ornithine ( kinact /KI = 208 M-1 s-1) and N5-(1-imino-2-chlorisopropyl)-l-lysine ( kinact /KI = 440 M-1 s-1), and one that lengthens the linker beyond that found in the substrate, N5-(1-imino-2-chloroethyl)-l-lysine (Cl-NIL, KI = 0.19 μM, kinact = 0.22 min-1). Cl-NIL is one of the most potent inhibitors reported for DDAH1, inactivates with a second order rate constant (1.9 × 104 M-1 s-1) larger than the catalytic efficiency of DDAH1 for its endogenous substrate (1.6 × 102 M-1 s-1), and has a partition ratio of 1 with a >100 000-fold selectivity for DDAH1 over arginase. An activity-based protein-profiling probe is used to show inhibition of DDAH1 within cultured HEK293T cells (IC50 = 10 μM) with cytotoxicity appearing only at higher concentrations (ED50 = 118 μM). A 1.91 Å resolution X-ray crystal structure reveals specific interactions made with DDAH1 upon covalent inactivation by Cl-NIL. Dissecting a covalent inactivator and analysis of its constituent fragments proved useful for the design and optimization of this potent and effective DDAH1 inhibitor.
Collapse
Affiliation(s)
- Gayle Burstein-Teitelbaum
- Division of Chemical Biology & Medicinal Chemistry, College of Pharmacy , University of Texas , Austin , Texas 78712 , United States
| | - Joyce A V Er
- Division of Chemical Biology & Medicinal Chemistry, College of Pharmacy , University of Texas , Austin , Texas 78712 , United States
| | - Arthur F Monzingo
- Center for Biomedical Research Support , University of Texas , Austin , Texas 78712 , United States
| | - Alfred Tuley
- Division of Chemical Biology & Medicinal Chemistry, College of Pharmacy , University of Texas , Austin , Texas 78712 , United States
| | - Walter Fast
- Division of Chemical Biology & Medicinal Chemistry, College of Pharmacy , University of Texas , Austin , Texas 78712 , United States
| |
Collapse
|
46
|
Li H, Sun W, Deng M, Zhou Q, Wang J, Liu J, Chen C, Qi C, Luo Z, Xue Y, Zhu H, Zhang Y. Asperversiamides, Linearly Fused Prenylated Indole Alkaloids from the Marine-Derived Fungus Aspergillus versicolor. J Org Chem 2018; 83:8483-8492. [PMID: 30016097 DOI: 10.1021/acs.joc.8b01087] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Asperversiamides A-H (1-8), eight linearly fused prenylated indole alkaloids featuring an unusual pyrano[3,2- f]indole unit, were isolated from the marine-derived fungus Aspergillus versicolor. The structures and absolute configurations of these compounds were elucidated by extensive spectroscopic analyses, single-crystal X-ray diffraction, electronic circular dichroism (ECD) calculations, and optical rotation (OR) calculations. The relative configuration of C-21 of iso-notoamide B was herein revised, and a new methodology for preliminarily determining if the relative configuration of the bicyclo[2.2.2]diazaoctane moiety of a spiro-bicyclo[2.2.2]diazaoctane-type indole alkaloid is syn or anti was developed. The anti-inflammatory activities of the isolated compounds were all tested, and of these compounds, 7 exhibited a potent inhibitory effect against iNOS with an IC50 value of 5.39 μM.
Collapse
Affiliation(s)
- Huaqiang Li
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College , Huazhong University of Science and Technology Wuhan 430030 , China
| | - Weiguang Sun
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College , Huazhong University of Science and Technology Wuhan 430030 , China
| | - Mengyi Deng
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College , Huazhong University of Science and Technology Wuhan 430030 , China
| | - Qun Zhou
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College , Huazhong University of Science and Technology Wuhan 430030 , China
| | - Jianping Wang
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College , Huazhong University of Science and Technology Wuhan 430030 , China
| | - Junjun Liu
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College , Huazhong University of Science and Technology Wuhan 430030 , China
| | - Chunmei Chen
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College , Huazhong University of Science and Technology Wuhan 430030 , China
| | - Changxing Qi
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College , Huazhong University of Science and Technology Wuhan 430030 , China
| | - Zengwei Luo
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College , Huazhong University of Science and Technology Wuhan 430030 , China
| | - Yongbo Xue
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College , Huazhong University of Science and Technology Wuhan 430030 , China
| | - Hucheng Zhu
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College , Huazhong University of Science and Technology Wuhan 430030 , China
| | - Yonghui Zhang
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College , Huazhong University of Science and Technology Wuhan 430030 , China
| |
Collapse
|
47
|
Assessment of the direct effects of DDAH I on tumour angiogenesis in vivo. Angiogenesis 2018; 21:737-749. [PMID: 29721731 PMCID: PMC6208893 DOI: 10.1007/s10456-018-9617-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Accepted: 04/24/2018] [Indexed: 02/07/2023]
Abstract
Nitric oxide (NO) has been strongly implicated in glioma progression and angiogenesis. The endogenous inhibitors of NO synthesis, asymmetric dimethylarginine (ADMA) and N-monomethyl-l-arginine (l-NMMA), are metabolized by dimethylarginine dimethylaminohydrolase (DDAH), and hence, DDAH is an intracellular factor that regulates NO. However, DDAH may also have an NO-independent action. We aimed to investigate whether DDAH I has any direct role in tumour vascular development and growth independent of its NO-mediated effects, in order to establish the future potential of DDAH inhibition as an anti-angiogenic treatment strategy. A clone of rat C6 glioma cells deficient in NO production expressing a pTet Off regulatable element was identified and engineered to overexpress DDAH I in the absence of doxycycline. Xenografts derived from these cells were propagated in the presence or absence of doxycycline and susceptibility magnetic resonance imaging used to assess functional vasculature in vivo. Pathological correlates of tumour vascular density, maturation and function were also sought. In the absence of doxycycline, tumours exhibited high DDAH I expression and activity, which was suppressed in its presence. However, overexpression of DDAH I had no measurable effect on tumour growth, vessel density, function or maturation. These data suggest that in C6 gliomas DDAH has no NO-independent effects on tumour growth and angiogenesis, and that the therapeutic potential of targeting DDAH in gliomas should only be considered in the context of NO regulation.
Collapse
|
48
|
Dumitrescu E, Wallace KN, Andreescu S. Real time electrochemical investigation of the release, distribution and modulation of nitric oxide in the intestine of individual zebrafish embryos. Nitric Oxide 2018; 74:32-38. [DOI: 10.1016/j.niox.2018.01.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Revised: 01/03/2018] [Accepted: 01/10/2018] [Indexed: 12/16/2022]
|
49
|
Sayols-Baixeras S, Subirana I, Fernández-Sanlés A, Sentí M, Lluís-Ganella C, Marrugat J, Elosua R. DNA methylation and obesity traits: An epigenome-wide association study. The REGICOR study. Epigenetics 2017; 12:909-916. [PMID: 29099282 DOI: 10.1080/15592294.2017.1363951] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
Abstract
Obesity is associated with increased risk of several diseases and has become epidemic. Obesity is highly heritable but the genetic variants identified by genome-wide association studies explain only limited variability. Epigenetics could contribute to explain the missing variability. The study aim was to discover differential methylation patterns related to obesity. We designed an epigenome-wide association study with a discovery phase in a subsample of 641 REGICOR study participants, validated by analysis of 2,515 participants in the Framingham Offspring Study. Blood DNA methylation was assessed using Illumina HumanMethylation450 BeadChip. Next, we meta-analyzed the data using the fixed effects method and performed a functional and pathway analysis using the Ingenuity Pathway Analysis software. We were able to validate 94 CpGs associated with body mass index (BMI) and 49 CpGs associated with waist circumference, located in 95 loci. In addition, we newly discovered 70 CpGs associated with BMI and 33 CpGs related to waist circumference. These CpGs explained 25.94% and 29.22% of the variability of BMI and waist circumference, respectively, in the REGICOR sample. We also evaluated 65 of the 95 validated loci in the GIANT genome-wide association data; 10 of them had Tag SNPs associated with BMI. The top-ranked diseases and functions identified in the functional and pathway analysis were neurologic, psychological, endocrine, and metabolic.
Collapse
Affiliation(s)
- Sergi Sayols-Baixeras
- a Cardiovascular Epidemiology and Genetics Research Group , IMIM (Hospital del Mar Medical Research Institute) , Barcelona , Catalonia , Spain.,b Universitat Pompeu Fabra (UPF) , Barcelona , Catalonia , Spain.,c CIBER Cardiovascular diseases (CIBERCV) , Barcelona , Catalonia , Spain
| | - Isaac Subirana
- a Cardiovascular Epidemiology and Genetics Research Group , IMIM (Hospital del Mar Medical Research Institute) , Barcelona , Catalonia , Spain.,d CIBER Epidemiology and Public Health (CIBERESP) , Barcelona , Catalonia , Spain
| | - Alba Fernández-Sanlés
- a Cardiovascular Epidemiology and Genetics Research Group , IMIM (Hospital del Mar Medical Research Institute) , Barcelona , Catalonia , Spain.,b Universitat Pompeu Fabra (UPF) , Barcelona , Catalonia , Spain
| | - Mariano Sentí
- b Universitat Pompeu Fabra (UPF) , Barcelona , Catalonia , Spain.,c CIBER Cardiovascular diseases (CIBERCV) , Barcelona , Catalonia , Spain
| | - Carla Lluís-Ganella
- a Cardiovascular Epidemiology and Genetics Research Group , IMIM (Hospital del Mar Medical Research Institute) , Barcelona , Catalonia , Spain
| | - Jaume Marrugat
- a Cardiovascular Epidemiology and Genetics Research Group , IMIM (Hospital del Mar Medical Research Institute) , Barcelona , Catalonia , Spain.,c CIBER Cardiovascular diseases (CIBERCV) , Barcelona , Catalonia , Spain
| | - Roberto Elosua
- a Cardiovascular Epidemiology and Genetics Research Group , IMIM (Hospital del Mar Medical Research Institute) , Barcelona , Catalonia , Spain.,c CIBER Cardiovascular diseases (CIBERCV) , Barcelona , Catalonia , Spain
| |
Collapse
|
50
|
Reddy KRK, Dasari C, Duscharla D, Supriya B, Ram NS, Surekha MV, Kumar JM, Ummanni R. Dimethylarginine dimethylaminohydrolase-1 (DDAH1) is frequently upregulated in prostate cancer, and its overexpression conveys tumor growth and angiogenesis by metabolizing asymmetric dimethylarginine (ADMA). Angiogenesis 2017; 21:79-94. [DOI: 10.1007/s10456-017-9587-0] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Accepted: 11/13/2017] [Indexed: 12/20/2022]
|