1
|
Januzzi JL, Butler J, Del Prato S, Ezekowitz JA, Ibrahim NE, Lam CSP, Lewis GD, Marwick TH, Perfetti R, Rosenstock J, Solomon SD, Tang WHW, Zannad F. Randomized Trial of a Selective Aldose Reductase Inhibitor in Patients With Diabetic Cardiomyopathy. J Am Coll Cardiol 2024; 84:137-148. [PMID: 38597864 DOI: 10.1016/j.jacc.2024.03.380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 03/13/2024] [Accepted: 03/13/2024] [Indexed: 04/11/2024]
Abstract
BACKGROUND Progression to symptomatic heart failure is a complication of type 2 diabetes; heart failure onset in this setting is commonly preceded by deterioration in exercise capacity. OBJECTIVES This study sought to determine whether AT-001, a highly selective aldose reductase inhibitor, can stabilize exercise capacity among individuals with diabetic cardiomyopathy (DbCM) and reduced peak oxygen uptake (Vo2). METHODS A total of 691 individuals with DbCM meeting inclusion and exclusion criteria were randomized to receive placebo or ascending doses of AT-001 twice daily. Stratification at inclusion included region of enrollment, cardiopulmonary exercise test results, and use of sodium-glucose cotransporter 2 inhibitors or glucagon-like peptide-1 receptor agonists. The primary endpoint was proportional change in peak Vo2 from baseline to 15 months. Subgroup analyses included measures of disease severity and stratification variables. RESULTS The mean age was 67.5 ± 7.2 years, and 50.4% of participants were women. By 15 months, peak Vo2 fell in the placebo-treated patients by -0.31 mL/kg/min (P = 0.005 compared to baseline), whereas in those receiving high-dose AT-001, peak Vo2 fell by -0.01 mL/kg/min (P = 0.21); the difference in peak Vo2 between placebo and high-dose AT-001 was 0.30 (P = 0.19). In prespecified subgroup analyses among those not receiving sodium-glucose cotransporter 2 inhibitors or glucagon-like peptide-1 receptor agonists at baseline, the difference between peak Vo2 in placebo vs high-dose AT-001 at 15 months was 0.62 mL/kg/min (P = 0.04; interaction P = 0.10). CONCLUSIONS Among individuals with DbCM and impaired exercise capacity, treatment with AT-001 for 15 months did not result in significantly better exercise capacity compared with placebo. (Safety and Efficacy of AT-001 in Patients With Diabetic Cardiomyopathy [ARISE-HF]; NCT04083339).
Collapse
Affiliation(s)
- James L Januzzi
- Cardiology Division, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA; Baim Institute for Clinical Research, Boston, Massachusetts, USA.
| | - Javed Butler
- University of Mississippi Medical Center, Jackson, Mississippi, USA; Baylor Scott and White Institute, Dallas, Texas, USA
| | - Stefano Del Prato
- Interdisciplinary Center "Health Sciences," Sant'Anna School of Advanced Studies, Pisa, Italy
| | - Justin A Ezekowitz
- Division of Cardiology, University of Alberta, Edmonton, Alberta, Canada
| | - Nasrien E Ibrahim
- Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Carolyn S P Lam
- Baim Institute for Clinical Research, Boston, Massachusetts, USA; National Heart Centre Singapore and Duke-National University of Singapore, Singapore
| | - Gregory D Lewis
- Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Thomas H Marwick
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | | | - Julio Rosenstock
- Velocity Clinical Research Center at Medical City, Dallas, Texas, USA
| | - Scott D Solomon
- Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - W H Wilson Tang
- Heart, Vascular and Thoracic Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Faiez Zannad
- Université de Lorraine, Inserm CIC and CHRU, Nancy, France
| |
Collapse
|
2
|
Güleç Ö, Türkeş C, Arslan M, Demir Y, Dincer B, Ece A, İrfan Küfrevioğlu Ö, Beydemir Ş. Novel spiroindoline derivatives targeting aldose reductase against diabetic complications: Bioactivity, cytotoxicity, and molecular modeling studies. Bioorg Chem 2024; 145:107221. [PMID: 38387398 DOI: 10.1016/j.bioorg.2024.107221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 02/01/2024] [Accepted: 02/16/2024] [Indexed: 02/24/2024]
Abstract
Despite significant developments in therapeutic strategies, Diabetes Mellitus remains an increasing concern, leading to various complications, e.g., cataracts, neuropathy, retinopathy, nephropathy, and several cardiovascular diseases. The polyol pathway, which involves Aldose reductase (AR) as a critical enzyme, has been focused on by many researchers as a target for intervention. On the other hand, spiroindoline-based compounds possess remarkable biological properties. This guided us to synthesize novel spiroindoline oxadiazolyl-based acetate derivatives and investigate their biological activities. The synthesized molecules' structures were confirmed herein, using IR, NMR (1H and 13C), and Mass spectroscopy. All compounds were potent inhibitors with KI constants spanning from 0.186 ± 0.020 μM to 0.662 ± 0.042 μM versus AR and appeared as better inhibitors than the clinically used drug, Epalrestat (EPR, KI: 0.841 ± 0.051 μM). Besides its remarkable inhibitory profile compared to EPR, compound 6k (KI: 0.186 ± 0.020 μM) was also determined to have an unusual pharmacokinetic profile. The results showed that 6k had less cytotoxic effect on normal mouse fibroblast (L929) cells (IC50 of 569.58 ± 0.80 μM) and reduced the viability of human breast adenocarcinoma (MCF-7) cells (IC50 of 110.87 ± 0.42 μM) more than the reference drug Doxorubicin (IC50s of 98.26 ± 0.45 μM and 158.49 ± 2.73 μM, respectively), thus exhibiting more potent anticancer activity. Moreover, molecular dynamic simulations for 200 ns were conducted to predict the docked complex's stability and reveal significant amino acid residues that 6k interacts with throughout the simulation.
Collapse
Affiliation(s)
- Özcan Güleç
- Department of Chemistry, Faculty of Arts and Sciences, Sakarya University, 54187 Sakarya, Turkey
| | - Cüneyt Türkeş
- Department of Biochemistry, Faculty of Pharmacy, Erzincan Binali Yıldırım University, 24002 Erzincan, Turkey.
| | - Mustafa Arslan
- Department of Chemistry, Faculty of Arts and Sciences, Sakarya University, 54187 Sakarya, Turkey.
| | - Yeliz Demir
- Department of Pharmacy Services, Nihat Delibalta Göle Vocational High School, Ardahan University, 75700 Ardahan, Turkey
| | - Busra Dincer
- Department of Pharmacology, Faculty of Pharmacy, Ondokuz Mayıs University, 55020 Samsun, Turkey
| | - Abdulilah Ece
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Biruni University, 34010 İstanbul, Turkey
| | | | - Şükrü Beydemir
- Department of Biochemistry, Faculty of Pharmacy, Anadolu University, 26470 Eskişehir, Turkey; Bilecik Şeyh Edebali University, 11230 Bilecik, Turkey
| |
Collapse
|
3
|
Carvalho RA. The glycolytic pathway to heart failure. GLYCOLYSIS 2024:235-266. [DOI: 10.1016/b978-0-323-91704-9.00010-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
4
|
Andres-Hernando A, Orlicky DJ, Kuwabara M, Cicerchi C, Pedler M, Petrash MJ, Johnson RJ, Tolan DR, Lanaspa MA. Endogenous Fructose Production and Metabolism Drive Metabolic Dysregulation and Liver Disease in Mice with Hereditary Fructose Intolerance. Nutrients 2023; 15:4376. [PMID: 37892451 PMCID: PMC10609559 DOI: 10.3390/nu15204376] [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: 09/01/2023] [Revised: 10/06/2023] [Accepted: 10/13/2023] [Indexed: 10/29/2023] Open
Abstract
Excessive intake of sugar, and particularly fructose, is closely associated with the development and progression of metabolic syndrome in humans and animal models. However, genetic disorders in fructose metabolism have very different consequences. While the deficiency of fructokinase, the first enzyme involved in fructose metabolism, is benign and somewhat desirable, missense mutations in the second enzyme, aldolase B, causes a very dramatic and sometimes lethal condition known as hereditary fructose intolerance (HFI). To date, there is no cure for HFI, and treatment is limited to avoiding fructose and sugar. Because of this, for subjects with HFI, glucose is their sole source of carbohydrates in the diet. However, clinical symptoms still occur, suggesting that either low amounts of fructose are still being consumed or, alternatively, fructose is being produced endogenously in the body. Here, we demonstrate that as a consequence of consuming high glycemic foods, the polyol pathway, a metabolic route in which fructose is produced from glucose, is activated, triggering a deleterious mechanism whereby glucose, sorbitol and alcohol induce severe liver disease and growth retardation in aldolase B knockout mice. We show that generically and pharmacologically blocking this pathway significantly improves metabolic dysfunction and thriving and increases the tolerance of aldolase B knockout mice to dietary triggers of endogenous fructose production.
Collapse
Affiliation(s)
- Ana Andres-Hernando
- Division of Endocrinology, Metabolism and Diabetes, University of Colorado Denver, Aurora, CO 80045, USA;
| | - David J. Orlicky
- Department of Pathology, University of Colorado School of Medicine, Aurora, CO 80045, USA;
| | - Masanari Kuwabara
- Department of Cardiology, Toranomon Hospital, Tokyo 105-8470, Japan;
- Division of Public Health, Center for Community Medicine, Jichi Medical University, Tochigi 329-0431, Japan
| | - Christina Cicerchi
- Division of Renal Diseases and Hypertension, University of Colorado Denver, Aurora, CO 80045, USA; (C.C.); (R.J.J.)
| | - Michelle Pedler
- Department of Ophthalmology, University of Colorado School of Medicine, Aurora, CO 80045, USA; (M.P.); (M.J.P.)
| | - Mark J. Petrash
- Department of Ophthalmology, University of Colorado School of Medicine, Aurora, CO 80045, USA; (M.P.); (M.J.P.)
| | - Richard J. Johnson
- Division of Renal Diseases and Hypertension, University of Colorado Denver, Aurora, CO 80045, USA; (C.C.); (R.J.J.)
| | - Dean R. Tolan
- Department of Biology, Boston University, Boston, MA 02215, USA;
| | - Miguel A. Lanaspa
- Division of Endocrinology, Metabolism and Diabetes, University of Colorado Denver, Aurora, CO 80045, USA;
| |
Collapse
|
5
|
Gopal K, Karwi QG, Tabatabaei Dakhili SA, Wagg CS, Zhang L, Sun Q, Saed CT, Panidarapu S, Perfetti R, Ramasamy R, Ussher JR, Lopaschuk GD. Aldose reductase inhibition alleviates diabetic cardiomyopathy and is associated with a decrease in myocardial fatty acid oxidation. Cardiovasc Diabetol 2023; 22:73. [PMID: 36978133 PMCID: PMC10053619 DOI: 10.1186/s12933-023-01811-w] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 03/21/2023] [Indexed: 03/30/2023] Open
Abstract
BACKGROUND Cardiovascular diseases, including diabetic cardiomyopathy, are major causes of death in people with type 2 diabetes. Aldose reductase activity is enhanced in hyperglycemic conditions, leading to altered cardiac energy metabolism and deterioration of cardiac function with adverse remodeling. Because disturbances in cardiac energy metabolism can promote cardiac inefficiency, we hypothesized that aldose reductase inhibition may mitigate diabetic cardiomyopathy via normalization of cardiac energy metabolism. METHODS Male C57BL/6J mice (8-week-old) were subjected to experimental type 2 diabetes/diabetic cardiomyopathy (high-fat diet [60% kcal from lard] for 10 weeks with a single intraperitoneal injection of streptozotocin (75 mg/kg) at 4 weeks), following which animals were randomized to treatment with either vehicle or AT-001, a next-generation aldose reductase inhibitor (40 mg/kg/day) for 3 weeks. At study completion, hearts were perfused in the isolated working mode to assess energy metabolism. RESULTS Aldose reductase inhibition by AT-001 treatment improved diastolic function and cardiac efficiency in mice subjected to experimental type 2 diabetes. This attenuation of diabetic cardiomyopathy was associated with decreased myocardial fatty acid oxidation rates (1.15 ± 0.19 vs 0.5 ± 0.1 µmol min-1 g dry wt-1 in the presence of insulin) but no change in glucose oxidation rates compared to the control group. In addition, cardiac fibrosis and hypertrophy were also mitigated via AT-001 treatment in mice with diabetic cardiomyopathy. CONCLUSIONS Inhibiting aldose reductase activity ameliorates diastolic dysfunction in mice with experimental type 2 diabetes, which may be due to the decline in myocardial fatty acid oxidation, indicating that treatment with AT-001 may be a novel approach to alleviate diabetic cardiomyopathy in patients with diabetes.
Collapse
Affiliation(s)
- Keshav Gopal
- Cardiovascular Research Institute, University of Alberta, Edmonton, AB, Canada
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada
- Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada
| | - Qutuba G Karwi
- Cardiovascular Research Institute, University of Alberta, Edmonton, AB, Canada
| | - Seyed Amirhossein Tabatabaei Dakhili
- Cardiovascular Research Institute, University of Alberta, Edmonton, AB, Canada
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada
- Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada
| | - Cory S Wagg
- Cardiovascular Research Institute, University of Alberta, Edmonton, AB, Canada
| | - Liyan Zhang
- Cardiovascular Research Institute, University of Alberta, Edmonton, AB, Canada
| | - Qiuyu Sun
- Cardiovascular Research Institute, University of Alberta, Edmonton, AB, Canada
| | - Christina T Saed
- Cardiovascular Research Institute, University of Alberta, Edmonton, AB, Canada
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada
- Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada
| | - Sai Panidarapu
- Cardiovascular Research Institute, University of Alberta, Edmonton, AB, Canada
| | | | - Ravichandran Ramasamy
- Diabetes Research Program, New York University Grossman Medical Center, New York, NY, USA
| | - John R Ussher
- Cardiovascular Research Institute, University of Alberta, Edmonton, AB, Canada
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada
- Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada
| | - Gary D Lopaschuk
- Cardiovascular Research Institute, University of Alberta, Edmonton, AB, Canada.
| |
Collapse
|
6
|
Januzzi JL, Butler J, Del Prato S, Ezekowitz JA, Ibrahim NE, Lam CSP, Lewis GD, Marwick TH, Rosenstock J, Tang WHW, Zannad F, Lawson F, Perfetti R, Urbinati A. Rationale and design of the Aldose Reductase Inhibition for Stabilization of Exercise Capacity in Heart Failure Trial (ARISE-HF) in patients with high-risk diabetic cardiomyopathy. Am Heart J 2023; 256:25-36. [PMID: 36372245 DOI: 10.1016/j.ahj.2022.11.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 10/28/2022] [Accepted: 11/04/2022] [Indexed: 06/16/2023]
Abstract
BACKGROUND Diabetic cardiomyopathy (DbCM) is a specific form of heart muscle disease that may result in substantial morbidity and mortality in individuals with type 2 diabetes mellitus (T2DM). Hyperactivation of the polyol pathway is one of the primary mechanisms in the pathogenesis of diabetic complications, including development of DbCM. There is an unmet need for therapies targeting the underlying metabolic abnormalities that drive this form of Stage B heart failure (HF). METHODS Aldose reductase (AR) catalyzes the first and rate-limiting step in the polyol pathway, and AR inhibition has been shown to reduce diabetic complications, including DbCM in animal models and in patients with DbCM. Previous AR inhibitors (ARIs) were limited by poor specificity resulting in unacceptable tolerability and safety profile. AT-001 is a novel investigational highly specific ARI with higher binding affinity and greater selectivity than previously studied ARIs. ARISE-HF (NCT04083339) is an ongoing Phase 3 randomized, placebo-controlled, double blind, global clinical study to investigate the efficacy of AT-001 (1000 mg twice daily [BID] and 1500 mg BID) in 675 T2DM patients with DbCM at high risk of progression to overt HF. ARISE-HF assesses the ability of AT-001 to improve or prevent decline in exercise capacity as measured by functional capacity (changes in peak oxygen uptake [peak VO2]) over 15 (and possibly 27) months of treatment. Additional endpoints include percentage of patients progressing to overt HF, health status metrics, echocardiographic measurements, and changes in cardiacbiomarkers. RESULTS The ARISE-HF Trial is fully enrolled. CONCLUSIONS This report describes the rationale and study design of ARISE-HF.
Collapse
Affiliation(s)
- James L Januzzi
- Cardiology Division, Massachusetts General Hospital, Baim Institute for Clinical Research and Harvard Medical School, Boston, MA.
| | - Javed Butler
- University of Mississippi Medical Center, Jackson, MS; Baylor Scott and White Institute, Dallas, TX
| | - Stefano Del Prato
- Department of Clinical & Experimental Medicine, Section of Diabetes, University of Pisa, Pisa, Italy
| | | | | | - Carolyn S P Lam
- National Heart Centre Singapore and Duke-National University of Singapore, Singapore, Singapore
| | - Gregory D Lewis
- Cardiology Division, Massachusetts General Hospital, Boston, MA
| | | | | | - W H Wilson Tang
- Heart, Vascular and Thoracic Institute, Cleveland Clinic, Cleveland, OH
| | - Faiez Zannad
- Université de Lorraine, Inserm CIC and CHRU, Nancy, France
| | | | | | | |
Collapse
|
7
|
Shi X, Tuan H, Na X, Yang H, Yang Y, Zhang Y, Xi M, Tan Y, Yang C, Zhang J, Zhao A. The Association between Sugar-Sweetened Beverages and Male Pattern Hair Loss in Young Men. Nutrients 2023; 15:nu15010214. [PMID: 36615870 PMCID: PMC9824121 DOI: 10.3390/nu15010214] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 12/17/2022] [Accepted: 12/29/2022] [Indexed: 01/04/2023] Open
Abstract
We performed this study to investigate the association between sugar-sweetened beverage (SSB) consumption and male pattern hair loss (MPHL) in young men. We conducted this cross-sectional study from January to April 2022 in mainland China. Young people aged 18-45 years (n = 1951) were recruited from 31 provinces in China. We used a self-reported online survey for data collection. We explored the associations between the amount/frequency of SSB consumption and MPHL by using a binary logistic regression model, with adjustments for sociodemographic, hair status, dietary intake, lifestyle, and psychological factors. Among the 1028 participants (27.8 ± 7.2 years) in the final analysis, we found that high SSB consumption is associated with a higher risk of MPHL. We recommend more support to decrease SSB consumption among young people to minimize negative health outcomes.
Collapse
Affiliation(s)
- Xiaojin Shi
- Vanke School of Public Health, Tsinghua University, Beijing 100084, China
| | - Hsiaohan Tuan
- Department of Dermatology, School of Clinical Medicine, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing 102218, China
| | - Xiaona Na
- Vanke School of Public Health, Tsinghua University, Beijing 100084, China
| | - Haibing Yang
- Vanke School of Public Health, Tsinghua University, Beijing 100084, China
| | - Yucheng Yang
- Vanke School of Public Health, Tsinghua University, Beijing 100084, China
| | - Yulin Zhang
- Vanke School of Public Health, Tsinghua University, Beijing 100084, China
| | - Menglu Xi
- Vanke School of Public Health, Tsinghua University, Beijing 100084, China
| | - Yuefeng Tan
- Vanke School of Public Health, Tsinghua University, Beijing 100084, China
| | - Celi Yang
- Vanke School of Public Health, Tsinghua University, Beijing 100084, China
| | - Junhan Zhang
- Vanke School of Public Health, Tsinghua University, Beijing 100084, China
| | - Ai Zhao
- Vanke School of Public Health, Tsinghua University, Beijing 100084, China
- Correspondence: ; Tel.: +86-010-6279-6447
| |
Collapse
|
8
|
Deng M, Sun J, Peng L, Huang Y, Jiang W, Wu S, Zhou L, Chung SK, Cheng X. Scutellarin acts on the AR-NOX axis to remediate oxidative stress injury in a mouse model of cerebral ischemia/reperfusion injury. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 103:154214. [PMID: 35689902 DOI: 10.1016/j.phymed.2022.154214] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 05/11/2022] [Accepted: 05/26/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND Oxidative stress plays an important role in the pathology of ischemic stroke. Studies have confirmedthat scutellarin has antioxidant effects against ischemic injury, and we also reported that the involvement of Aldose reductase (AR) in oxidative stress and cerebral ischemic injury, in this study we furtherly explicit whether the antioxidant effect of scutellarin on cerebral ischemia injury is related to AR gene regulation and its specific mechanism. METHODS C57BL/6N mice (Wild-type, WT) and AR knockout (AR-/-) mice suffered from transient middle cerebral artery occlusion (tMCAO) injury (1 h occlusion followed by 3 days reperfusion), and scutellarin was administered from 2 h before surgery to 3 days after surgery. Subsequently, neurological function was assessed by the modified Longa score method, the histopathological morphology observed with 2,3,5-triphenyltetrazolium chloride (TTC) and hematoxylin-eosin (HE) staining. Enzyme-linked immunosorbent assay (Elisa) was used to detect the levels of ROS, 4-hydroxynonenal (4-HNE), 8-hydroxydeoxyguanosine (8-OHDG), Neurotrophin-3 (NT-3), poly ADP-ribose polymerase-1 (PARP1) and 3-nitrotyrosine (3-NT) in the ischemic penumbra regions. Quantitative proteomics profiling using quantitative nano-HPLC-MS/MS were performed to compare the protein expression difference between AR-/- and WT mice with or without tMCAO injury. The expression of AR, nicotinamide adenine dinucleotide phosphate oxidases (NOX1, NOX2 and NOX4) in the ipsilateral side of ischemic brain were detected by qRT-PCR, Western blot and immunofluorescence co-staining with NeuN. RESULTS Scutellarin treatment alleviated brain damage in tMCAO stroke model such as improved neurological function deficit, brain infarct area and neuronal injury and reduced the expression of oxidation-related products, moreover, also down-regulated tMCAO induced AR mRNA and protein expression. In addition, the therapeutic effect of scutellarin on the reduction of cerebral infarction area and neurological function deficits abolished in AR-/- mice under ischemia cerebral injury, which indicated that the effect of scutellarin treatment on tMCAO injury is through regulating AR gene. Proteomic analysis of AR-/- and WT mice indicated AR knockout would affect oxidation reaction even as NADPH related process and activity in mice under cerebral ischemia conditions. Moreover, NOX isoforms (NOX1, NOX2 and NOX4) mRNA and protein expression were significant decreased in neurons of penumbra region in AR-/- mice compared with that in WT mice at 3d after tMCAO injury, which indicated that AR should be the upstream protein regulating NOX after cerebral ischemia. CONCLUSIONS We first reported that AR directly regulates NOX subtypes (not only NOX2 but also NOX1 and NOX4) after cerebral ischaemic injury. Scutellarin specifically targets the AR-NOX axis and has antioxidant effects in mice with cerebral ischaemic injury, providing a theoretical basis and accurate molecular targets for the clinical application of scutellarin.
Collapse
Affiliation(s)
- Minzhen Deng
- Department of Neurology, Guangdong Provincial Hospital of Traditional Chinese Medicine, Guangzhou, China; Department of Second Institute of Clinical Medicine, Guangzhou University of Traditional Chinese Medicine, Guangzhou, China
| | - Jingbo Sun
- Department of Neurology, Guangdong Provincial Hospital of Traditional Chinese Medicine, Guangzhou, China; State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China; Department of Second Institute of Clinical Medicine, Guangzhou University of Traditional Chinese Medicine, Guangzhou, China; Guangdong Provincial Key Laboratory of Research on Emergency in TCM, Guangzhou, China
| | - Lilin Peng
- Department of Second Institute of Clinical Medicine, Guangzhou University of Traditional Chinese Medicine, Guangzhou, China
| | - Yan Huang
- Department of Neurology, Guangdong Provincial Hospital of Traditional Chinese Medicine, Guangzhou, China; State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China; Department of Second Institute of Clinical Medicine, Guangzhou University of Traditional Chinese Medicine, Guangzhou, China; Guangdong Provincial Key Laboratory of Research on Emergency in TCM, Guangzhou, China
| | - Wen Jiang
- Department of Second Institute of Clinical Medicine, Guangzhou University of Traditional Chinese Medicine, Guangzhou, China
| | - Shuang Wu
- Department of Second Institute of Clinical Medicine, Guangzhou University of Traditional Chinese Medicine, Guangzhou, China
| | - Lihua Zhou
- Department of Anatomy, Sun Yat-Sen School of Medicine, Sun Yat-Sen University, Shenzhen, China
| | - Sookja Kim Chung
- Faculty of Medicine, Macau University of Science and Technology, Macau, China
| | - Xiao Cheng
- Department of Neurology, Guangdong Provincial Hospital of Traditional Chinese Medicine, Guangzhou, China; State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China; Department of Second Institute of Clinical Medicine, Guangzhou University of Traditional Chinese Medicine, Guangzhou, China; Guangdong Provincial Key Laboratory of Research on Emergency in TCM, Guangzhou, China.
| |
Collapse
|
9
|
Zhang X, Xu L, Chen H, Zhang X, Lei Y, Liu W, Xu H, Ma B, Zhu C. Novel Hydroxychalcone-Based Dual Inhibitors of Aldose Reductase and α-Glucosidase as Potential Therapeutic Agents against Diabetes Mellitus and Its Complications. J Med Chem 2022; 65:9174-9192. [PMID: 35749671 DOI: 10.1021/acs.jmedchem.2c00380] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
We designed a novel series of bifunctional inhibitors of α-glucosidase and aldose reductase (ALR2) based on the structure of hydroxychalcone. The two enzymes relate to blood glucose level and anomalously elevated polyol pathway of glucose metabolism under hyperglycemia, respectively. Most compounds in the series exhibited a potent inhibitory activity for both enzymes, and a significant antioxidant property was shown. Further in vivo studies of 11j and 14d using streptozotocin (STZ)-induced diabetic rats as a model found that 11j achieved not only good antihyperglycemic and glucose tolerance effect in a dose-dependent manner (p < 0.01) but also showed effective inhibition of polyol pathway. 14d significantly suppressed the maltose-induced postprandial glucose elevation. Additionally, they effectively improved lipid metabolisms and restored an antioxidant ability. Therefore, the two compounds may be promising agents for the prevention and treatment of diabetic complications.
Collapse
Affiliation(s)
- Xiaonan Zhang
- Key Laboratory of Cluster Science, Ministry of Education of China, Beijing Key Laboratory of Photoelectronic; Electrophotonic Conversion Materials, School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing 100081, China
| | - Long Xu
- Key Laboratory of Cluster Science, Ministry of Education of China, Beijing Key Laboratory of Photoelectronic; Electrophotonic Conversion Materials, School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing 100081, China
| | - Huan Chen
- Key Laboratory of Cluster Science, Ministry of Education of China, Beijing Key Laboratory of Photoelectronic; Electrophotonic Conversion Materials, School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing 100081, China
| | - Xin Zhang
- Key Laboratory of Cluster Science, Ministry of Education of China, Beijing Key Laboratory of Photoelectronic; Electrophotonic Conversion Materials, School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing 100081, China
| | - Yanqi Lei
- Key Laboratory of Cluster Science, Ministry of Education of China, Beijing Key Laboratory of Photoelectronic; Electrophotonic Conversion Materials, School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing 100081, China
| | - Wenchao Liu
- Key Laboratory of Cluster Science, Ministry of Education of China, Beijing Key Laboratory of Photoelectronic; Electrophotonic Conversion Materials, School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing 100081, China
| | - Hulin Xu
- Key Laboratory of Cluster Science, Ministry of Education of China, Beijing Key Laboratory of Photoelectronic; Electrophotonic Conversion Materials, School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing 100081, China
| | - Bing Ma
- Key Laboratory of Cluster Science, Ministry of Education of China, Beijing Key Laboratory of Photoelectronic; Electrophotonic Conversion Materials, School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing 100081, China
| | - Changjin Zhu
- Key Laboratory of Cluster Science, Ministry of Education of China, Beijing Key Laboratory of Photoelectronic; Electrophotonic Conversion Materials, School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing 100081, China
| |
Collapse
|
10
|
Johnson RJ, García-Arroyo FE, Gonzaga-Sánchez G, Vélez-Orozco KA, Álvarez-Álvarez YQ, Aparicio-Trejo OE, Tapia E, Osorio-Alonso H, Andrés-Hernando A, Nakagawa T, Kuwabara M, Kanbay M, Lanaspa MA, Sánchez-Lozada LG. Current Hydration Habits: The Disregarded Factor for the Development of Renal and Cardiometabolic Diseases. Nutrients 2022; 14:2070. [PMID: 35631211 PMCID: PMC9145744 DOI: 10.3390/nu14102070] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 05/12/2022] [Accepted: 05/13/2022] [Indexed: 01/21/2023] Open
Abstract
Improper hydration habits are commonly disregarded as a risk factor for the development of chronic diseases. Consuming an intake of water below recommendations (underhydration) in addition to the substitution of sugar-sweetened beverages (SSB) for water are habits deeply ingrained in several countries. This behavior is due to voluntary and involuntary dehydration; and because young children are exposed to SSB, the preference for a sweet taste is profoundly implanted in the brain. Underhydration and SSB intake lead to mild hyperosmolarity, which stimulates biologic processes, such as the stimulation of vasopressin and the polyol-fructose pathway, which restore osmolarity to normal but at the expense of the continued activation of these biological systems. Unfortunately, chronic activation of the vasopressin and polyol-fructose pathways has been shown to mediate many diseases, such as obesity, diabetes, metabolic syndrome, chronic kidney disease, and cardiovascular disease. It is therefore urgent that we encourage educational and promotional campaigns that promote the evaluation of personal hydration status, a greater intake of potable water, and a reduction or complete halting of the drinking of SSB.
Collapse
Affiliation(s)
- Richard J. Johnson
- Division of Renal Diseases and Hypertension, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA;
| | - Fernando E. García-Arroyo
- Department Cardio-Renal Physiopathology, INC Ignacio Chávez, Mexico City 14080, Mexico; (F.E.G.-A.); (G.G.-S.); (K.A.V.-O.); (Y.Q.Á.-Á.); (O.E.A.-T.); (E.T.); (H.O.-A.)
| | - Guillermo Gonzaga-Sánchez
- Department Cardio-Renal Physiopathology, INC Ignacio Chávez, Mexico City 14080, Mexico; (F.E.G.-A.); (G.G.-S.); (K.A.V.-O.); (Y.Q.Á.-Á.); (O.E.A.-T.); (E.T.); (H.O.-A.)
| | - Kevin A. Vélez-Orozco
- Department Cardio-Renal Physiopathology, INC Ignacio Chávez, Mexico City 14080, Mexico; (F.E.G.-A.); (G.G.-S.); (K.A.V.-O.); (Y.Q.Á.-Á.); (O.E.A.-T.); (E.T.); (H.O.-A.)
| | - Yamnia Quetzal Álvarez-Álvarez
- Department Cardio-Renal Physiopathology, INC Ignacio Chávez, Mexico City 14080, Mexico; (F.E.G.-A.); (G.G.-S.); (K.A.V.-O.); (Y.Q.Á.-Á.); (O.E.A.-T.); (E.T.); (H.O.-A.)
| | - Omar Emiliano Aparicio-Trejo
- Department Cardio-Renal Physiopathology, INC Ignacio Chávez, Mexico City 14080, Mexico; (F.E.G.-A.); (G.G.-S.); (K.A.V.-O.); (Y.Q.Á.-Á.); (O.E.A.-T.); (E.T.); (H.O.-A.)
| | - Edilia Tapia
- Department Cardio-Renal Physiopathology, INC Ignacio Chávez, Mexico City 14080, Mexico; (F.E.G.-A.); (G.G.-S.); (K.A.V.-O.); (Y.Q.Á.-Á.); (O.E.A.-T.); (E.T.); (H.O.-A.)
| | - Horacio Osorio-Alonso
- Department Cardio-Renal Physiopathology, INC Ignacio Chávez, Mexico City 14080, Mexico; (F.E.G.-A.); (G.G.-S.); (K.A.V.-O.); (Y.Q.Á.-Á.); (O.E.A.-T.); (E.T.); (H.O.-A.)
| | - Ana Andrés-Hernando
- Division of Nephrology and Hypertension, Oregon Health Sciences University, Portland, OR 97239, USA; (A.A.-H.); (M.A.L.)
| | - Takahiko Nakagawa
- Department of Nephrology, Rakuwakai Otowa Hospital, Kyoto 607-8062, Japan;
| | - Masanari Kuwabara
- Intensive Care Unit, Toranomon Hospital, Tokyo 105-8470, Japan;
- Department of Cardiology, Toranomon Hospital, Tokyo 105-8470, Japan
| | - Mehmet Kanbay
- Division of Nephrology, Department of Internal Medicine, Koc University School of Medicine, Istanbul 34010, Turkey;
| | - Miguel A. Lanaspa
- Division of Nephrology and Hypertension, Oregon Health Sciences University, Portland, OR 97239, USA; (A.A.-H.); (M.A.L.)
| | - Laura Gabriela Sánchez-Lozada
- Department Cardio-Renal Physiopathology, INC Ignacio Chávez, Mexico City 14080, Mexico; (F.E.G.-A.); (G.G.-S.); (K.A.V.-O.); (Y.Q.Á.-Á.); (O.E.A.-T.); (E.T.); (H.O.-A.)
| |
Collapse
|
11
|
Bayrak C, Yildizhan G, Kilinc N, Durdagi S, Menzek A. Synthesis and Aldose Reductase Inhibition Effects of Novel N-Benzyl-4-Methoxyaniline Derivatives. Chem Biodivers 2021; 19:e202100530. [PMID: 34889038 DOI: 10.1002/cbdv.202100530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 11/23/2021] [Indexed: 11/10/2022]
Abstract
In the current study, starting from 4-methoxyaniline, four Schiff bases were synthesized from benzaldehydes with Br and OMe. Corresponding N-benzylanilines and their derivatives were obtained from reductions (by NaBH4 ) and substitutions (by acyl and tosyl chlorides) of these bases, respectively. The inhibitory effects of the sixteen compounds, twelve of which were novel compounds are examined. Then, we conducted molecular docking and binary QSAR studies to determine inhibitory-enzyme interactions of compounds that show an inhibitory effect. Our results reveal that methoxyanilline-derived compounds show good biological activities. The most active compound (22) has IC50 values of 2.83 μM. These novel AR enzyme inhibitors may open new avenues for better AR inhibitors in the future.
Collapse
Affiliation(s)
- Cetin Bayrak
- Department of Chemistry, Faculty of Science, Ataturk University, Erzurum, 25240-, Turkey.,Dogubayazit Ahmed-i Hani Vocational School, Agri Ibrahim Cecen University, Agri, 04400, -Turkey
| | - Gulsah Yildizhan
- Department of Chemistry, Faculty of Science, Ataturk University, Erzurum, 25240-, Turkey
| | - Namik Kilinc
- Department of Medical Services and Techniques, Vocational School of Health Service, Igdir University, Igdir, 76000, Turkey
| | - Serdar Durdagi
- Computational Biology and Molecular Simulations Laboratory, Department of Biophysics, School of Medicine, Bahcesehir University, Istanbul, 34747-, Turkey
| | - Abdullah Menzek
- Department of Chemistry, Faculty of Science, Ataturk University, Erzurum, 25240-, Turkey
| |
Collapse
|
12
|
Sadgrove NJ. The ‘bald’ phenotype (androgenetic alopecia) is caused by the high glycaemic, high cholesterol and low mineral ‘western diet’. Trends Food Sci Technol 2021. [DOI: 10.1016/j.tifs.2021.06.056] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
13
|
Imran A, Tariq Shehzad M, Al Adhami T, Miraz Rahman K, Hussain D, Alharthy RD, Shafiq Z, Iqbal J. Development of coumarin-thiosemicarbazone hybrids as aldose reductase inhibitors: Biological assays, molecular docking, simulation studies and ADME evaluation. Bioorg Chem 2021; 115:105164. [PMID: 34314916 DOI: 10.1016/j.bioorg.2021.105164] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 06/03/2021] [Accepted: 07/08/2021] [Indexed: 11/26/2022]
Abstract
The over expression of aldose reductase (ALR2) in the state of hyperglycemia causes the conversion of glucose into sorbitol and initiates polyol pathway. Accumulation of sorbitol in insulin insensitive tissue like peripheral nerves, glomerulus and eyes, induces diabetic complications like neuropathy, nephropathy and retinopathy. For the treatment of diabetic complications, the inhibition of aldose reductase (ALR2) is a promising approach. A series of coumarin-based thiosemicarbazone derivatives was synthesized as potential inhibitor of aldose reductase. Compound N-(2-fluorophenyl)-2-(1-(2-oxo-2H-chromen-3-yl)ethylidene)hydrazinecarbiothioamide (3n) was found to be the most promising inhibitor of ALR2 with an IC50 in micromolar range (2.07 µM) and high selectivity, relative to ALR1. The crystal structure of ALR2 complexed with 3n explored the types of interaction pattern which further demonstrated its high affinity. Compound 3n has excellent lead-likeness, underlined by its physicochemical parameters, and can be considered as a likely prospect for further structural optimization to get a drugable molecule.
Collapse
Affiliation(s)
- Aqeel Imran
- Center for Advanced Drug Research, COMSATS University Islamabad, Abbottabad Campus, Abbottabad 22060, Pakistan; Institute of Pharmaceutical Science, King's College London, 150 Stamford Street, London SE1 9NH, United Kingdom
| | | | - Taha Al Adhami
- Institute of Pharmaceutical Science, King's College London, 150 Stamford Street, London SE1 9NH, United Kingdom
| | - Khondaker Miraz Rahman
- Institute of Pharmaceutical Science, King's College London, 150 Stamford Street, London SE1 9NH, United Kingdom
| | - Dilawar Hussain
- Center for Advanced Drug Research, COMSATS University Islamabad, Abbottabad Campus, Abbottabad 22060, Pakistan
| | - Rima D Alharthy
- Department of Chemistry, Science and Arts College, Rabigh Campus, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Zahid Shafiq
- Institute of Chemical Sciences, Bahauddin Zakariya University, Multan 60800, Pakistan.
| | - Jamshed Iqbal
- Center for Advanced Drug Research, COMSATS University Islamabad, Abbottabad Campus, Abbottabad 22060, Pakistan.
| |
Collapse
|
14
|
Moiz B, Garcia J, Basehore S, Sun A, Li A, Padmanabhan S, Albus K, Jang C, Sriram G, Clyne AM. 13C Metabolic Flux Analysis Indicates Endothelial Cells Attenuate Metabolic Perturbations by Modulating TCA Activity. Metabolites 2021; 11:metabo11040226. [PMID: 33917224 PMCID: PMC8068087 DOI: 10.3390/metabo11040226] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 03/30/2021] [Accepted: 04/01/2021] [Indexed: 11/16/2022] Open
Abstract
Disrupted endothelial metabolism is linked to endothelial dysfunction and cardiovascular disease. Targeted metabolic inhibitors are potential therapeutics; however, their systemic impact on endothelial metabolism remains unknown. In this study, we combined stable isotope labeling with 13C metabolic flux analysis (13C MFA) to determine how targeted inhibition of the polyol (fidarestat), pentose phosphate (DHEA), and hexosamine biosynthetic (azaserine) pathways alters endothelial metabolism. Glucose, glutamine, and a four-carbon input to the malate shuttle were important carbon sources in the baseline human umbilical vein endothelial cell (HUVEC) 13C MFA model. We observed two to three times higher glutamine uptake in fidarestat and azaserine-treated cells. Fidarestat and DHEA-treated HUVEC showed decreased 13C enrichment of glycolytic and TCA metabolites and amino acids. Azaserine-treated HUVEC primarily showed 13C enrichment differences in UDP-GlcNAc. 13C MFA estimated decreased pentose phosphate pathway flux and increased TCA activity with reversed malate shuttle direction in fidarestat and DHEA-treated HUVEC. In contrast, 13C MFA estimated increases in both pentose phosphate pathway and TCA activity in azaserine-treated cells. These data show the potential importance of endothelial malate shuttle activity and suggest that inhibiting glycolytic side branch pathways can change the metabolic network, highlighting the need to study systemic metabolic therapeutic effects.
Collapse
Affiliation(s)
- Bilal Moiz
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA; (B.M.); (A.S.); (A.L.); (S.P.); (K.A.)
| | - Jonathan Garcia
- School of Bioengineering, Science, and Heath Systems, Drexel University, Philadelphia, PA 19104, USA; (J.G.); (S.B.)
| | - Sarah Basehore
- School of Bioengineering, Science, and Heath Systems, Drexel University, Philadelphia, PA 19104, USA; (J.G.); (S.B.)
| | - Angela Sun
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA; (B.M.); (A.S.); (A.L.); (S.P.); (K.A.)
| | - Andrew Li
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA; (B.M.); (A.S.); (A.L.); (S.P.); (K.A.)
| | - Surya Padmanabhan
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA; (B.M.); (A.S.); (A.L.); (S.P.); (K.A.)
| | - Kaitlyn Albus
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA; (B.M.); (A.S.); (A.L.); (S.P.); (K.A.)
| | - Cholsoon Jang
- Department of Biological Chemistry, Chao Family Comprehensive Cancer Center, University of California Irvine, Irvine, CA 92697, USA;
| | - Ganesh Sriram
- Department of Chemical and Biomolecular Engineering, University of Maryland, College Park, MD 20742, USA;
| | - Alisa Morss Clyne
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA; (B.M.); (A.S.); (A.L.); (S.P.); (K.A.)
- Correspondence: ; Tel.: +1-301-405-9806
| |
Collapse
|
15
|
Jannapureddy S, Sharma M, Yepuri G, Schmidt AM, Ramasamy R. Aldose Reductase: An Emerging Target for Development of Interventions for Diabetic Cardiovascular Complications. Front Endocrinol (Lausanne) 2021; 12:636267. [PMID: 33776930 PMCID: PMC7992003 DOI: 10.3389/fendo.2021.636267] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 01/19/2021] [Indexed: 12/18/2022] Open
Abstract
Diabetes is a leading cause of cardiovascular morbidity and mortality. Despite numerous treatments for cardiovascular disease (CVD), for patients with diabetes, these therapies provide less benefit for protection from CVD. These considerations spur the concept that diabetes-specific, disease-modifying therapies are essential to identify especially as the diabetes epidemic continues to expand. In this context, high levels of blood glucose stimulate the flux via aldose reductase (AR) pathway leading to metabolic and signaling changes in cells of the cardiovascular system. In animal models flux via AR in hearts is increased by diabetes and ischemia and its inhibition protects diabetic and non-diabetic hearts from ischemia-reperfusion injury. In mouse models of diabetic atherosclerosis, human AR expression accelerates progression and impairs regression of atherosclerotic plaques. Genetic studies have revealed that single nucleotide polymorphisms (SNPs) of the ALD2 (human AR gene) is associated with diabetic complications, including cardiorenal complications. This Review presents current knowledge regarding the roles for AR in the causes and consequences of diabetic cardiovascular disease and the status of AR inhibitors in clinical trials. Studies from both human subjects and animal models are presented to highlight the breadth of evidence linking AR to the cardiovascular consequences of diabetes.
Collapse
Affiliation(s)
| | | | | | | | - Ravichandran Ramasamy
- Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, NYU Grossman School of Medicine, New York, NY, United States
| |
Collapse
|
16
|
Ahmad I, Hoda M. Molecular mechanisms of action of resveratrol in modulation of diabetic and non-diabetic cardiomyopathy. Pharmacol Res 2020; 161:105112. [PMID: 32758636 DOI: 10.1016/j.phrs.2020.105112] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 07/22/2020] [Indexed: 12/18/2022]
Abstract
Cardiomyopathy is among the major clinical manifestations of heart diseases that triggers malfunctioning of the cardiovascular system. Some of the major causal factors of cardiomyopathy includes myocardial ischemia, drug-toxicity, genetic aberrations, abnormal depositions of essential elements, and redox imbalance. Diabetes, being the major comorbid of cardiovascular diseases and vice versa, further contributes to the progression of cardiomyopathy. The molecular mechanisms of action suggest that oxidative stress is among the primary factors that triggers cascading impact on cardiomyopathy. Resveratrol, a phenolic antioxidant, has the potential to quench the excessive free radicals. It is a potent antioxidant supplement that may as well be a therapeutic molecule. The review focuses on the various molecular mechanisms of action that resveratrol potentiates in reversing or attenuating the progress of diabetic and non-diabetic cardiomyopathy triggered by wide range of factors. Additionally, resveratrol also tends to preserve the healthy heart from potential damage that may be triggered by oxidative stress.
Collapse
Affiliation(s)
- Irshad Ahmad
- National Institute of Plant Genome Research, Aruna Asaf Ali Marg, New Delhi, 110067, India
| | - Muddasarul Hoda
- Department of Biological Sciences, Aliah University, IIA/27, Newtown, Kolkata, 700160, India.
| |
Collapse
|
17
|
Nakagawa T, Lanaspa MA, Millan IS, Fini M, Rivard CJ, Sanchez-Lozada LG, Andres-Hernando A, Tolan DR, Johnson RJ. Fructose contributes to the Warburg effect for cancer growth. Cancer Metab 2020; 8:16. [PMID: 32670573 PMCID: PMC7350662 DOI: 10.1186/s40170-020-00222-9] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Accepted: 07/01/2020] [Indexed: 12/13/2022] Open
Abstract
Obesity and metabolic syndrome are strongly associated with cancer, and these disorders may share a common mechanism. Recently, fructose has emerged as a driving force to develop obesity and metabolic syndrome. Thus, we assume that fructose may be the mechanism to explain why obesity and metabolic syndrome are linked with cancer. Clinical and experimental evidence showed that fructose intake was associated with cancer growth and that fructose transporters are upregulated in various malignant tumors. Interestingly, fructose metabolism can be driven under low oxygen conditions, accelerates glucose utilization, and exhibits distinct effects as compared to glucose, including production of uric acid and lactate as major byproducts. Fructose promotes the Warburg effect to preferentially downregulate mitochondrial respiration and increases aerobic glycolysis that may aid metastases that initially have low oxygen supply. In the process, uric acid may facilitate carcinogenesis by inhibiting the TCA cycle, stimulating cell proliferation by mitochondrial ROS, and blocking fatty acid oxidation. Lactate may also contribute to cancer growth by suppressing fat oxidation and inducing oncogene expression. The ability of fructose metabolism to directly stimulate the glycolytic pathway may have been protective for animals living with limited access to oxygen, but may be deleterious toward stimulating cancer growth and metastasis for humans in modern society. Blocking fructose metabolism may be a novel approach for the prevention and treatment of cancer.
Collapse
Affiliation(s)
- Takahiko Nakagawa
- Department of Nephrology, Rakuwakai Otowa Hospital, 2 Otowa-Chinji-cho, Yamashina-ku, Kyoto, Japan
- Department of Stem Cell Biology & Regenerative Medicine, Shiga University of Medical Science, Otsu, Japan
| | - Miguel A. Lanaspa
- Division of Renal Diseases and Hypertension, University of Colorado Denver, Aurora, CO USA
| | - Inigo San Millan
- Department of Medicine, Division of Endocrinology, Metabolism and Diabetes, University of Colorado School of Medicine, Aurora, USA
| | - Mehdi Fini
- University of Colorado Cancer Center, Aurora, CO USA
| | | | - Laura G. Sanchez-Lozada
- Department of Cardio-Renal Physiopathology, Instituto Nacional de Cardiología Ignacio Chavez, 14080 Mexico City, CP Mexico
| | - Ana Andres-Hernando
- Division of Renal Diseases and Hypertension, University of Colorado Denver, Aurora, CO USA
| | - Dean R. Tolan
- Department of Biology, Boston University, Boston, MA USA
| | - Richard J. Johnson
- Division of Renal Diseases and Hypertension, University of Colorado Denver, Aurora, CO USA
| |
Collapse
|
18
|
High Concentrations of Uric Acid and Angiotensin II Act Additively to Produce Endothelial Injury. Mediators Inflamm 2020; 2020:8387654. [PMID: 32565731 PMCID: PMC7261330 DOI: 10.1155/2020/8387654] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 02/22/2020] [Accepted: 03/10/2020] [Indexed: 12/22/2022] Open
Abstract
Renin angiotensin (Ang) system (RAS) activation in metabolic syndrome (MS) patients is associated with elevated uric acid (UA) levels, resulting in endothelial system dysfunction. Our previous study demonstrated that excessive UA could cause endothelial injury through the aldose reductase (AR) pathway. This study is the first to show that a high concentration of Ang II in human umbilical vein endothelial cells (HUVECs) increases reactive oxygen species (ROS) components, including O2·- and H2O2, and further aggravates endothelial system injury induced by high UA (HUA). In a MS/hyperuricemia model, nitric oxide (NO) production was decreased, followed by a decrease in total antioxidant capacity (TAC), and the concentration of the endothelial injury marker von Willebrand factor (vWF) in the serum was increased. Treatment with catalase and polyethylene glycol covalently linked to superoxide dismutase (PEG-SOD) to individually remove H2O2 and O2·- or treatment with the AR inhibitor epalrestat decreased ROS and H2O2, increased NO levels and TAC, and reduced vWF release. Taken together, these data indicate that HUA and Ang II act additively to cause endothelial dysfunction via oxidative stress, and specific elimination of O2·- and H2O2 improves endothelial function. We provide theoretical evidence to prevent or delay endothelial injury caused by metabolic diseases.
Collapse
|
19
|
Affiliation(s)
- Diem H Tran
- 1 Division of Cardiology Department of Internal Medicine University of Texas Southwestern Medical Center Dallas TX
| | - Zhao V Wang
- 1 Division of Cardiology Department of Internal Medicine University of Texas Southwestern Medical Center Dallas TX
| |
Collapse
|
20
|
Zhang D, Bhatnagar A, Baba SP. Inhibition of aldose reductase activity stimulates starvation induced autophagy and clears aldehyde protein adducts. Chem Biol Interact 2019; 306:104-109. [DOI: 10.1016/j.cbi.2019.04.014] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 03/14/2019] [Accepted: 04/11/2019] [Indexed: 12/31/2022]
|
21
|
Chihanga T, Ruby HN, Ma Q, Bashir S, Devarajan P, Kennedy MA. NMR-based urine metabolic profiling and immunohistochemistry analysis of nephron changes in a mouse model of hypoxia-induced acute kidney injury. Am J Physiol Renal Physiol 2018; 315:F1159-F1173. [PMID: 29993280 DOI: 10.1152/ajprenal.00500.2017] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Acute kidney injury can be caused by multiple factors, including sepsis, respiratory failure, heart failure, trauma, or nephrotoxic medications, among others. Here, a mouse model was used to investigate potential urinary metabolic biomarkers of hypoxia-induced AKI. Urine metabolic profiles of 48 Swiss Webster mice were assessed using nuclear magnetic resonance spectroscopy (NMR) for 7 days following 72 h exposure to a hypoxic 6.5% oxygen environment. Histological analyses indicated a lack of gross nephron structural changes in the aftermath of hypoxia. Immunohistochemical (IHC) analyses, however, indicated elevated expression of protein injury biomarkers in distal and proximal tubules but not glomeruli. Kidney injury molecule-1 levels peaked in distal tubules at 72 h and were still increasing in proximal tubules at 7 days posthypoxia, whereas cystatin C levels were elevated at 24 h but decreased thereafter, and were elevated and still increasing in proximal tubules at 7 days posthypoxia. Neutrophil gelatinase-associated lipocalin levels were modestly elevated from 24 h to 7 days posthypoxia. NMR-based metabolic profiling revealed that urine metabolites involved in energy metabolism and associated biosynthetic pathways were initially decreased at 24 h posthypoxia, consistent with metabolic suppression as a mechanism for cell survival, but were significantly elevated at 48 and 72 h posthypoxia, indicating a burst in organism metabolism associated with reactivation of cellular energetics during recovery after cessation of hypoxia and return to a normoxic environment. The IHC results indicated that kidney injury persists long after plasma and urine biomarkers of hypoxia return to normal values.
Collapse
Affiliation(s)
- Tafadzwa Chihanga
- Department of Chemistry and Biochemistry, Miami University , Oxford, Ohio
| | - Hannah N Ruby
- Department of Chemistry and Biochemistry, Miami University , Oxford, Ohio
| | - Qing Ma
- Department of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, University of Cincinnati , Cincinnati, Ohio
| | - Sabina Bashir
- Department of Chemistry and Biochemistry, Miami University , Oxford, Ohio
| | - Prasad Devarajan
- Department of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, University of Cincinnati , Cincinnati, Ohio
| | - Michael A Kennedy
- Department of Chemistry and Biochemistry, Miami University , Oxford, Ohio
| |
Collapse
|
22
|
Abstract
SIGNIFICANCE Pyridine dinucleotides, nicotinamide adenine dinucleotide (NAD) and nicotinamide adenine dinucleotide phosphate (NADP), were discovered more than 100 years ago as necessary cofactors for fermentation in yeast extracts. Since that time, these molecules have been recognized as fundamental players in a variety of cellular processes, including energy metabolism, redox homeostasis, cellular signaling, and gene transcription, among many others. Given their critical role as mediators of cellular responses to metabolic perturbations, it is unsurprising that dysregulation of NAD and NADP metabolism has been associated with the pathobiology of many chronic human diseases. Recent Advances: A biochemistry renaissance in biomedical research, with its increasing focus on the metabolic pathobiology of human disease, has reignited interest in pyridine dinucleotides, which has led to new insights into the cell biology of NAD(P) metabolism, including its cellular pharmacokinetics, biosynthesis, subcellular localization, and regulation. This review highlights these advances to illustrate the importance of NAD(P) metabolism in the molecular pathogenesis of disease. CRITICAL ISSUES Perturbations of NAD(H) and NADP(H) are a prominent feature of human disease; however, fundamental questions regarding the regulation of the absolute levels of these cofactors and the key determinants of their redox ratios remain. Moreover, an integrated topological model of NAD(P) biology that combines the metabolic and other roles remains elusive. FUTURE DIRECTIONS As the complex regulatory network of NAD(P) metabolism becomes illuminated, sophisticated new approaches to manipulating these pathways in specific organs, cells, or organelles will be developed to target the underlying pathogenic mechanisms of disease, opening doors for the next generation of redox-based, metabolism-targeted therapies. Antioxid. Redox Signal. 28, 180-212.
Collapse
Affiliation(s)
- Joshua P Fessel
- 1 Department of Medicine, Vanderbilt University , Nashville, Tennessee
| | - William M Oldham
- 2 Department of Medicine, Brigham and Women's Hospital , Boston, Massachusetts.,3 Department of Medicine, Harvard Medical School , Boston, Massachusetts
| |
Collapse
|
23
|
Putakala M, Gujjala S, Nukala S, Bongu SBR, Chintakunta N, Desireddy S. Cardioprotective effect of Phyllanthus amarus against high fructose diet induced myocardial and aortic stress in rat model. Biomed Pharmacother 2017; 95:1359-1368. [PMID: 28946183 DOI: 10.1016/j.biopha.2017.09.054] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2017] [Revised: 09/08/2017] [Accepted: 09/10/2017] [Indexed: 01/11/2023] Open
Abstract
Increased number of population with heart stroke/attack is attributed to sedentary lifestyle and consumption of high-sugar diets, especially fructose. The objective of this study is to investigate the cardio-protective activity of aqueous extract of Phyllanthus amarus (PAAE) against high-fructose (HF) diet induced cardiac damage in Wistar rats. Male Wistar rats were randomly assigned into five groups of six animals each: Control (C), Control treated with PAAE (C+PAAE), High fructose diet fed (F), High fructose diet fed treated with PAAE (F+PAAE) and High fructose diet fed treated with Pioglitazone (F+Pio). PAAE was orally administered at a dosage of 200mg/kg body weight/day to C+PAAE and F+PAAE group rats for 60days. Pioglitazone (10mg/kg body weight/day) was used to compare the efficacy of PAAE. After 60days, heart and aorta samples were collected for biochemical and histological analysis. Co-administration of PAAE along with HF-diet for 60days prevented the increase in levels of cardiac and aortic lipids i.e., total lipids, triglycerides, total cholesterol and free fatty acids and decreased phospholipids. Further, enhanced activities of cardiac aldose reductase (15.3%) and sorbital dehydrogenase (6.9%) and decreased activity of creatine kinase (35.6%) in group-F were also prevented by PAAE treatment with the recovery of 126% for AR, 122% for SDH and 118% for CK. PAAE treatment showed protection from HF-diet induced increase in stress markers (LPO and PO), decreased non-enzymatic (GSH and Vit-C) and enzymatic (GR, GPx, GST, SOD, and CAT) antioxidants in the heart and aorta. Histopathological examination of the heart and aorta indicated that PAAE/Pio treatment reduced fat deposition and necrosis. The present study clearly indicates the cardio protection efficacy of PAAE against HF-diet induced oxidative stress in rats.
Collapse
Affiliation(s)
- Mallaiah Putakala
- Department of Biochemistry, Sri Krishnadevaraya University, Anantapuramu, Andhra Pradesh, 515 003, India
| | - Sudhakara Gujjala
- Department of Biochemistry, Sri Krishnadevaraya University, Anantapuramu, Andhra Pradesh, 515 003, India
| | - Srinivasulu Nukala
- Department of Biochemistry, Sri Krishnadevaraya University, Anantapuramu, Andhra Pradesh, 515 003, India
| | - Sasi Bhusana Rao Bongu
- Department of Biochemistry, Sri Krishnadevaraya University, Anantapuramu, Andhra Pradesh, 515 003, India
| | - Nagaraju Chintakunta
- Department of Biochemistry, Sri Krishnadevaraya University, Anantapuramu, Andhra Pradesh, 515 003, India
| | - Saralakumari Desireddy
- Department of Biochemistry, Sri Krishnadevaraya University, Anantapuramu, Andhra Pradesh, 515 003, India.
| |
Collapse
|
24
|
Protective role of fructokinase blockade in the pathogenesis of acute kidney injury in mice. Nat Commun 2017; 8:14181. [PMID: 28194018 PMCID: PMC5316807 DOI: 10.1038/ncomms14181] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Accepted: 12/07/2016] [Indexed: 01/10/2023] Open
Abstract
Acute kidney injury is associated with high mortality, especially in intensive care unit patients. The polyol pathway is a metabolic route able to convert glucose into fructose. Here we show the detrimental role of endogenous fructose production by the polyol pathway and its metabolism through fructokinase in the pathogenesis of ischaemic acute kidney injury (iAKI). Consistent with elevated urinary fructose in AKI patients, mice undergoing iAKI show significant polyol pathway activation in the kidney cortex characterized by high levels of aldose reductase, sorbitol and endogenous fructose. Wild type but not fructokinase knockout animals demonstrate severe kidney injury associated with ATP depletion, elevated uric acid, oxidative stress and inflammation. Interestingly, both the renal injury and dysfunction in wild-type mice undergoing iAKI is significantly ameliorated when exposed to luteolin, a recently discovered fructokinase inhibitor. This study demonstrates a role for fructokinase and endogenous fructose as mediators of acute renal disease. The polyol pathway, which converts glucose into sorbitol and fructose, is active in chronic conditions like hepatic steatosis and chronic kidney disease. Here, Andres-Hernando et al. show that fructose production promotes renal injury and fructokinase inhibition protects against kidney damage during ischaemic acute kidney disease.
Collapse
|
25
|
Singh M, Kapoor A, McCracken J, Hill B, Bhatnagar A. Aldose reductase (AKR1B) deficiency promotes phagocytosis in bone marrow derived mouse macrophages. Chem Biol Interact 2017; 265:16-23. [PMID: 28111134 DOI: 10.1016/j.cbi.2017.01.012] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Accepted: 01/18/2017] [Indexed: 12/31/2022]
Abstract
Macrophages are critical drivers of the immune response during infection and inflammation. The pathogenesis of several inflammatory conditions, such as diabetes, cancer and sepsis has been linked with aldose reductase (AR), a member of the aldo-keto reductase (AKR) superfamily. However, the role of AR in the early stages of innate immunity such as phagocytosis remains unclear. In this study, we examined the role of AR in regulating the growth and the phagocytic activity of bone marrow-derived mouse macrophages (BMMs) from AR-null and wild-type (WT) mice. We found that macrophages derived from AR-null mice were larger in size and had a slower growth rate than those derived from WT mice. The AR-null macrophages also displayed higher basal, and lipopolysaccharide (LPS) stimulated phagocytic activity than WT macrophages. Moreover, absence of AR led to a marked increase in cellular levels of both ATP and NADPH. These data suggest that metabolic pathways involving AR suppress macrophage energy production, and that inhibition of AR could induce a favorable metabolic state that promotes macrophage phagocytosis. Hence, modulation of macrophage metabolism by inhibition of AR might represent a novel strategy to modulate host defense responses and to modify metabolism to promote macrophage hypertrophy and phagocytosis under inflammatory conditions.
Collapse
Affiliation(s)
- Mahavir Singh
- Diabetes and Obesity Center, Institute of Molecular Cardiology, Division of Cardiovascular Medicine, Department of Medicine, University of Louisville, Louisville, KY, USA
| | - Aniruddh Kapoor
- Department of Internal Medicine, SSM Health St. Mary's Hospital, Clayton Rd., Richmond Heights, MO, USA
| | | | - Bradford Hill
- Diabetes and Obesity Center, Institute of Molecular Cardiology, Division of Cardiovascular Medicine, Department of Medicine, University of Louisville, Louisville, KY, USA
| | - Aruni Bhatnagar
- Diabetes and Obesity Center, Institute of Molecular Cardiology, Division of Cardiovascular Medicine, Department of Medicine, University of Louisville, Louisville, KY, USA.
| |
Collapse
|
26
|
The Role of Oxidative Stress in Myocardial Ischemia and Reperfusion Injury and Remodeling: Revisited. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2016; 2016:1656450. [PMID: 27313825 PMCID: PMC4897712 DOI: 10.1155/2016/1656450] [Citation(s) in RCA: 220] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Revised: 04/11/2016] [Accepted: 05/03/2016] [Indexed: 01/11/2023]
Abstract
Oxidative and reductive stress are dual dynamic phases experienced by the cells undergoing adaptation towards endogenous or exogenous noxious stimulus. The former arises due to the imbalance between the reactive oxygen species production and antioxidant defenses, while the latter is due to the aberrant increase in the reducing equivalents. Mitochondrial malfunction is the common denominator arising from the aberrant functioning of the rheostat that maintains the homeostasis between oxidative and reductive stress. Recent experimental evidences suggest that the maladaptation during oxidative stress could play a pivotal role in the pathophysiology of major cardiovascular diseases such as myocardial infraction, atherosclerosis, and diabetic cardiovascular complications. In this review we have discussed the role of oxidative and reductive stress pathways in the pathogenesis of myocardial ischemia/reperfusion injury and diabetic cardiomyopathy (DCM). Furthermore, we have provided impetus for the development of subcellular organelle targeted antioxidant drug therapy for thwarting the deterioration of the failing myocardium in the aforementioned cardiovascular conditions.
Collapse
|
27
|
Mapanga RF, Essop MF. Damaging effects of hyperglycemia on cardiovascular function: spotlight on glucose metabolic pathways. Am J Physiol Heart Circ Physiol 2016; 310:H153-73. [DOI: 10.1152/ajpheart.00206.2015] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Accepted: 10/27/2015] [Indexed: 12/13/2022]
Abstract
The incidence of cardiovascular complications associated with hyperglycemia is a growing global health problem. This review discusses the link between hyperglycemia and cardiovascular diseases onset, focusing on the role of recently emerging downstream mediators, namely, oxidative stress and glucose metabolic pathway perturbations. The role of hyperglycemia-mediated activation of nonoxidative glucose pathways (NOGPs) [i.e., the polyol pathway, hexosamine biosynthetic pathway, advanced glycation end products (AGEs), and protein kinase C] in this process is extensively reviewed. The proposal is made that there is a unique interplay between NOGPs and a downstream convergence of detrimental effects that especially affect cardiac endothelial cells, thereby contributing to contractile dysfunction. In this process the AGE pathway emerges as a crucial mediator of hyperglycemia-mediated detrimental effects. In addition, a vicious metabolic cycle is established whereby hyperglycemia-induced NOGPs further fuel their own activation by generating even more oxidative stress, thereby exacerbating damaging effects on cardiac function. Thus NOGP inhibition, and particularly that of the AGE pathway, emerges as a novel therapeutic intervention for the treatment of cardiovascular complications such as acute myocardial infarction in the presence hyperglycemia.
Collapse
Affiliation(s)
- Rudo F. Mapanga
- Cardio-Metabolic Research Group, Department of Physiological Sciences, Stellenbosch University, Stellenbosch, South Africa
| | - M. Faadiel Essop
- Cardio-Metabolic Research Group, Department of Physiological Sciences, Stellenbosch University, Stellenbosch, South Africa
| |
Collapse
|
28
|
Baba SP, Hoetker JD, Merchant M, Klein JB, Cai J, Barski OA, Conklin DJ, Bhatnagar A. Role of aldose reductase in the metabolism and detoxification of carnosine-acrolein conjugates. J Biol Chem 2013; 288:28163-79. [PMID: 23928303 DOI: 10.1074/jbc.m113.504753] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Oxidation of unsaturated lipids generates reactive aldehydes that accumulate in tissues during inflammation, ischemia, or aging. These aldehydes form covalent adducts with histidine-containing dipeptides such as carnosine and anserine, which are present in high concentration in skeletal muscle, heart, and brain. The metabolic pathways involved in the detoxification and elimination of these conjugates are, however, poorly defined, and their significance in regulating oxidative stress is unclear. Here we report that conjugates of carnosine with aldehydes such as acrolein are produced during normal metabolism and excreted in the urine of mice and adult human non-smokers as carnosine-propanols. Our studies show that the reduction of carnosine-propanals is catalyzed by the enzyme aldose reductase (AR). Carnosine-propanals were converted to carnosine-propanols in the lysates of heart, skeletal muscle, and brain tissue from wild-type (WT) but not AR-null mice. In comparison with WT mice, the urinary excretion of carnosine-propanols was decreased in AR-null mice. Carnosine-propanals formed covalent adducts with nucleophilic amino acids leading to the generation of carnosinylated proteins. Deletion of AR increased the abundance of proteins bound to carnosine in skeletal muscle, brain, and heart of aged mice and promoted the accumulation of carnosinylated proteins in hearts subjected to global ischemia ex vivo. Perfusion with carnosine promoted post-ischemic functional recovery in WT but not in AR-null mouse hearts. Collectively, these findings reveal a previously unknown metabolic pathway for the removal of carnosine-propanal conjugates and suggest a new role of AR as a critical regulator of protein carnosinylation and carnosine-mediated tissue protection.
Collapse
|
29
|
Abstract
The pyridine nucleotides NAD(+) and NADP(+) play a pivotal role in regulating intermediary metabolism in the heart. The intracellular NAD(+)/NADH ratio controls flux through various dehydrogenase enzymes involved in both anaerobic and aerobic metabolism and also regulates posttranslational protein modification. The intracellular NADP(+)/NADPH ratio controls flux through the pentose phosphate pathway (PPP) and the polyol pathway, while also regulating ion channel function and oxidative stress. Not only does the NAD(+)/NADH ratio regulate the rates of ATP production, it can also modify energy substrate preference. For instance, in many forms of heart disease a greater contribution from fatty acids for oxidative energy metabolism increases fatty acid β-oxidation-derived NADH, which can activate pyruvate dehydrogenase (PDH) kinase isoforms that inhibit PDH and subsequent glucose oxidation. As such, novel therapies that overcome fatty acid β-oxidation-induced inhibition of PDH improve cardiac efficiency and subsequent function during ischemia/reperfusion and in heart failure. Furthermore, recent studies have implicated a pivotal role for increased PPP-derived NADPH in mediating oxidative stress observed in heart failure. In this article, we review the multiple actions of NAD(+)/NADH and NADP(+)/NADPH in regulating intermediary metabolism in the heart. A better understanding of the roles of NAD(+)/NADH and NADP(+)/NADPH in cellular physiology and pathology could potentially be used to exploit pyridine nucleotide modification in the treatment of a number of different forms of heart disease.
Collapse
Affiliation(s)
- John R Ussher
- 423 Heritage Medical Research Center, University of Alberta, Edmonton, Canada
| | | | | |
Collapse
|
30
|
Son NH, Ananthakrishnan R, Yu S, Khan RS, Jiang H, Ji R, Akashi H, Li Q, O'Shea K, Homma S, Goldberg IJ, Ramasamy R. Cardiomyocyte aldose reductase causes heart failure and impairs recovery from ischemia. PLoS One 2012; 7:e46549. [PMID: 23029549 PMCID: PMC3459912 DOI: 10.1371/journal.pone.0046549] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2012] [Accepted: 09/02/2012] [Indexed: 01/02/2023] Open
Abstract
Aldose reductase (AR), an enzyme mediating the first step in the polyol pathway of glucose metabolism, is associated with complications of diabetes mellitus and increased cardiac ischemic injury. We investigated whether deleterious effects of AR are due to its actions specifically in cardiomyocytes. We created mice with cardiac specific expression of human AR (hAR) using the α–myosin heavy chain (MHC) promoter and studied these animals during aging and with reduced fatty acid (FA) oxidation. hAR transgenic expression did not alter cardiac function or glucose and FA oxidation gene expression in young mice. However, cardiac overexpression of hAR caused cardiac dysfunction in older mice. We then assessed whether hAR altered heart function during ischemia reperfusion. hAR transgenic mice had greater infarct area and reduced functional recovery than non-transgenic littermates. When the hAR transgene was crossed onto the PPAR alpha knockout background, another example of greater heart glucose oxidation, hAR expressing mice had increased heart fructose content, cardiac fibrosis, ROS, and apoptosis. In conclusion, overexpression of hAR in cardiomyocytes leads to cardiac dysfunction with aging and in the setting of reduced FA and increased glucose metabolism. These results suggest that pharmacological inhibition of AR will be beneficial during ischemia and in some forms of heart failure.
Collapse
Affiliation(s)
- Ni-Huiping Son
- Department of Medicine, Columbia University Medical Center, New York, New York, United States of America
| | - Radha Ananthakrishnan
- Department of Medicine, New York University School of Medicine, New York, New York, United States of America
| | - Shuiqing Yu
- Department of Medicine, Columbia University Medical Center, New York, New York, United States of America
| | - Raffay S. Khan
- Department of Medicine, Columbia University Medical Center, New York, New York, United States of America
| | - Hongfeng Jiang
- Department of Medicine, Columbia University Medical Center, New York, New York, United States of America
| | - Ruiping Ji
- Department of Medicine, Columbia University Medical Center, New York, New York, United States of America
| | - Hirokazu Akashi
- Department of Medicine, Columbia University Medical Center, New York, New York, United States of America
| | - Qing Li
- Department of Medicine, Columbia University Medical Center, New York, New York, United States of America
| | - Karen O'Shea
- Department of Medicine, New York University School of Medicine, New York, New York, United States of America
| | - Shunichi Homma
- Department of Medicine, Columbia University Medical Center, New York, New York, United States of America
| | - Ira J. Goldberg
- Department of Medicine, Columbia University Medical Center, New York, New York, United States of America
| | - Ravichandran Ramasamy
- Department of Medicine, New York University School of Medicine, New York, New York, United States of America
- * E-mail:
| |
Collapse
|
31
|
Varma A, Das A, Hoke NN, Durrant DE, Salloum FN, Kukreja RC. Anti-inflammatory and cardioprotective effects of tadalafil in diabetic mice. PLoS One 2012; 7:e45243. [PMID: 23028874 PMCID: PMC3448606 DOI: 10.1371/journal.pone.0045243] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2012] [Accepted: 08/16/2012] [Indexed: 11/18/2022] Open
Abstract
Background Insulin resistance impairs nitric oxide (NO) bioavailability and obesity promotes a state of chronic inflammation and damages the vascular endothelium. Phosphodiesterase-5 inhibitors restore NO signaling and may reduce circulating inflammatory markers, and improve metabolic parameters through a number of mechanisms. We hypothesized that daily administration of the PDE-5 inhibitor, tadalafil (TAD) will attenuate inflammation, improve fasting plasma glucose and triglyceride levels, body weight, and reduce infarct size after ischemia/reperfusion injury in obese, diabetic mice. Methods Twenty leptin receptor null (db/db) mice underwent treatment with TAD (1 mg/Kg) or 10% DMSO for 28 days. Body weight and fasting plasma glucose levels were determined weekly. Upon completion, hearts were isolated and subjected to 30 min global ischemia followed by 60 min reperfusion in a Langendorff model. Plasma samples were taken for cytokine analysis and fasting triglyceride levels. Infarct size was measured using computer morphometry of tetrazolium stained sections. Additionally, ventricular cardiomyocytes were isolated and subjected to 40 min of simulated ischemia and reoxygenation. Necrosis was determined using trypan blue exclusion and LDH release assay and apoptosis was assessed by TUNEL assay after 1 h or 18 h of reoxygenation, respectively. Results Treatment with TAD caused a reduction in infarct size in the diabetic heart (23.2±1.5 vs. 47.8±3.7%, p<0.01, n = 6/group), reduced fasting glucose levels (292±31.8 vs. 511±19.3 mg/dL, p<0.001) and fasting triglycerides (43.3±21 vs. 129.7±29 mg/dL, p<0.05) as compared to DMSO, however body weight was not significantly reduced. Circulating tumor necrosis factor-α and interleukin-1β were reduced after treatment compared to control (257±16.51 vs. 402.3±17.26 and 150.8±12.55 vs. 264±31.85 pg/mL, respectively; P<0.001) Isolated cardiomyocytes from TAD-treated mice showed reduced apoptosis and necrosis. Conclusion We have provided the first evidence that TAD therapy ameliorates circulating inflammatory cytokines and chemokines in a diabetic animal model while improving fasting glucose levels and reducing infarct size following ischemia-reperfusion injury in the heart.
Collapse
Affiliation(s)
- Amit Varma
- Division of Cardiology, Department of Internal Medicine, VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Anindita Das
- Division of Cardiology, Department of Internal Medicine, VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Nicholas N. Hoke
- Division of Cardiology, Department of Internal Medicine, VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - David E. Durrant
- Division of Cardiology, Department of Internal Medicine, VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Fadi N. Salloum
- Division of Cardiology, Department of Internal Medicine, VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Rakesh C. Kukreja
- Division of Cardiology, Department of Internal Medicine, VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, Virginia, United States of America
- * E-mail:
| |
Collapse
|
32
|
Van der Mieren G, Nevelsteen I, Vanderper A, Oosterlinck W, Flameng W, Herijgers P. Angiotensin-converting enzyme inhibition and food restriction in diabetic mice do not correct the increased sensitivity for ischemia-reperfusion injury. Cardiovasc Diabetol 2012; 11:89. [PMID: 22853195 PMCID: PMC3444392 DOI: 10.1186/1475-2840-11-89] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2012] [Accepted: 07/18/2012] [Indexed: 01/14/2023] Open
Abstract
Background The number of patients with diabetes or the metabolic syndrome reaches epidemic proportions. On top of their diabetic cardiomyopathy, these patients experience frequent and severe cardiac ischemia-reperfusion (IR) insults, which further aggravate their degree of heart failure. Food restriction and angiotensin-converting enzyme inhibition (ACE-I) are standard therapies in these patients but the effects on cardiac IR injury have never been investigated. In this study, we tested the hypothesis that 1° food restriction and 2° ACE-I reduce infarct size and preserve cardiac contractility after IR injury in mouse models of diabetes and the metabolic syndrome. Methods C57Bl6/J wild type (WT) mice, leptin deficient ob/ob (model for type II diabetes) and double knock-out (LDLR-/-;ob/ob, further called DKO) mice with combined leptin and LDL-receptor deficiency (model for metabolic syndrome) were used. The effects of 12 weeks food restriction or ACE-I on infarct size and load-independent left ventricular contractility after 30 min regional cardiac ischemia were investigated. Differences between groups were analyzed for statistical significance by Student’s t-test or factorial ANOVA followed by a Fisher’s LSD post hoc test. Results Infarct size was larger in ob/ob and DKO versus WT. Twelve weeks of ACE-I improved pre-ischemic left ventricular contractility in ob/ob and DKO. Twelve weeks of food restriction, with a weight reduction of 35-40%, or ACE-I did not reduce the effect of IR. Conclusion ACE-I and food restriction do not correct the increased sensitivity for cardiac IR-injury in mouse models of type II diabetes and the metabolic syndrome.
Collapse
Affiliation(s)
- Gerry Van der Mieren
- Department of Cardiovascular Sciences, Research Unit Experimental Cardiac Surgery, K.U. Leuven, Herestraat 49, B-3000, Leuven, Belgium
| | | | | | | | | | | |
Collapse
|
33
|
Abdillahi M, Ananthakrishnan R, Vedantham S, Shang L, Zhu Z, Rosario R, Zirpoli H, Bohren KM, Gabbay KH, Ramasamy R. Aldose reductase modulates cardiac glycogen synthase kinase-3β phosphorylation during ischemia-reperfusion. Am J Physiol Heart Circ Physiol 2012; 303:H297-308. [PMID: 22661511 DOI: 10.1152/ajpheart.00999.2011] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Earlier studies have demonstrated that aldose reductase (AR) plays a key role in mediating ischemia-reperfusion (I/R) injury. Our objective was to investigate if AR mediates I/R injury by influencing phosphorylation of glycogen synthase kinase-3β (p-GSK3β). To investigate this issue, we used three separate models to study the effects of stress injury on the heart. Hearts isolated from wild-type (WT), human expressing AR transgenic (ARTg), and AR knockout (ARKO) mice were perfused with/without GSK3β inhibitors (SB-216763 and LiCl) and subjected to I/R. Ad-human AR (Ad-hAR)-expressing HL-1 cardiac cells were exposed to hypoxia (0.5% O(2)) and reoxygenation (20.9% O(2)) conditions. I/R in a murine model of transient occlusion and reperfusion of the left anterior descending coronary artery (LAD) was used to study if p-GSK3β was affected through increased AR flux. Lactate dehydrogenase (LDH) release and left ventricular developed pressure (LVDP) were measured. LVDP was decreased in hearts from ARTg mice compared with WT and ARKO after I/R, whereas LDH release and apoptotic markers were increased (P < 0.05). p-GSK3β was decreased in ARTg hearts compared with WT and ARKO (P < 0.05). In ARKO, p-GSK3β and apoptotic markers were decreased compared with WT (P < 0.05). WT and ARTg hearts perfused with GSK3β inhibitors improved p-GSK3β expression and LVDP and exhibited decreased LDH release, apoptosis, and mitochondrial pore opening (P < 0.05). Ad-hAR-expressing HL-1 cardiac cells, exposed to hypoxia (0.5% O(2)) and reoxygenation (20.9% O(2)), had greater LDH release compared with control HL-1 cells (P < 0.05). p-GSK3β was decreased and correlated with increased apoptotic markers in Ad-hAR HL-1 cells (P < 0.05). Treatment with phosphatidylinositol 3-kinase (PI3K)/protein kinase B (Akt) inhibitor increased injury demonstrated by increased LDH release in ARTg, WT, and ARKO hearts and in Ad-hAR-expressing HL-1 cells. Cells treated with protein kinase C (PKC) α/β inhibitor displayed significant increases in p-Akt and p-GSK3β expression, and resulted in decreased LDH release. In summary, AR mediates changes in p-GSK3β, in part, via PKCα/β and Akt during I/R.
Collapse
Affiliation(s)
- Mariane Abdillahi
- Diabetes Research Program, Department of Medicine, New York University Langone Medical Center, New York, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Rui T, Zhang J, Xu X, Yao Y, Kao R, Martin CM. Reduction in IL-33 expression exaggerates ischaemia/reperfusion-induced myocardial injury in mice with diabetes mellitus. Cardiovasc Res 2012; 94:370-8. [DOI: 10.1093/cvr/cvs015] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
|
35
|
Osmolarity and glucose differentially regulate aldose reductase activity in cultured mouse podocytes. EXPERIMENTAL DIABETES RESEARCH 2011; 2011:278963. [PMID: 22253613 PMCID: PMC3255165 DOI: 10.1155/2011/278963] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/14/2011] [Revised: 09/05/2011] [Accepted: 09/23/2011] [Indexed: 12/04/2022]
Abstract
Podocyte injury is associated with progression of many renal diseases, including diabetic nephropathy. In this study we examined whether aldose reductase (AR), the enzyme implicated in diabetic complications in different tissues, is modulated by high glucose and osmolarity in podocyte cells. AR mRNA, protein expression, and activity were measured in mouse podocytes cultured in both normal and high glucose and osmolarity for 6 hours to 5 days. Hyperosmolarity acutely stimulated AR expression and activity, with subsequent increase of AR expression but decrease of activity. High glucose also elevated AR protein level; however, this was not accompanied by respective enzyme activation. Furthermore, high glucose appeared to counteract the osmolarity-dependent activation of AR. In conclusion, in podocytes AR is modulated by high glucose and increased osmolarity in a different manner. Posttranslational events may affect AR activity independent of enzyme protein amount. Activation of AR in podocytes may be implicated in diabetic podocytopathy.
Collapse
|
36
|
Vedantham S, Noh H, Ananthakrishnan R, Son N, Hallam K, Hu Y, Yu S, Shen X, Rosario R, Lu Y, Ravindranath T, Drosatos K, Huggins LA, Schmidt AM, Goldberg IJ, Ramasamy R. Human aldose reductase expression accelerates atherosclerosis in diabetic apolipoprotein E-/- mice. Arterioscler Thromb Vasc Biol 2011; 31:1805-13. [PMID: 21636809 DOI: 10.1161/atvbaha.111.226902] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
OBJECTIVE There are several pathways that mediate the aberrant metabolism of glucose and that might induce greater vascular damage in the setting of diabetes. The polyol pathway mediated by aldose reductase (AR) has been postulated to be one such pathway. However, it has been reported that AR reduces toxic lipid aldehydes and, under some circumstances, might be antiatherogenic. METHODS AND RESULTS Atherosclerosis development was quantified in 2 lines of transgenic mice expressing human AR (hAR) crossed on the apolipoprotein E knockout background. The transgenes were used to increase the normally low levels of this enzyme in wild-type mice. Both generalized hAR overexpression and hAR expression via the Tie 2 promoter increased lesion size in streptozotocin diabetic mice. In addition, pharmacological inhibition of AR reduced lesion size. CONCLUSIONS Although in some settings AR expression might reduce levels of toxic aldehydes, transgenic expression of this enzyme within the artery wall leads to greater atherosclerosis.
Collapse
Affiliation(s)
- Srinivasan Vedantham
- Division of Endocrinology, New York University Langone Medical Center, NY 10016, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Ananthakrishnan R, Li Q, Gomes T, Schmidt AM, Ramasamy R. Aldose reductase pathway contributes to vulnerability of aging myocardium to ischemic injury. Exp Gerontol 2011; 46:762-7. [PMID: 21600277 DOI: 10.1016/j.exger.2011.05.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2011] [Revised: 04/29/2011] [Accepted: 05/03/2011] [Indexed: 12/20/2022]
Abstract
Aging men and women display both increased incidence of cardiovascular disease and complications of myocardial infarction and heart failure. We hypothesized that altered glucose metabolism, in particular, flux of glucose via the polyol pathway (PP) may be responsible, in part, for the enhanced vulnerability of aging myocardium to ischemic injury, even in the absence of superimposed disease processes linked to PP flux, such as diabetes. To test our hypothesis, we determined the expression and products of PP enzymes aldose reductase (AR) and sorbitol dehydrogenase (SDH) in hearts from Fischer 344 aged (26 months) and young (4 months) rats subjected to global ischemia followed by reperfusion in the presence or absence of blockers of PP and the measures of ischemic injury and functional recovery were determined. Expression and activities of AR and SDH were significantly higher in aged vs. young hearts, and induction of ischemia further increased AR and SDH activity in the aged hearts. Myocardial ischemic injury was significantly greater in aged vs. young hearts, and blockade of AR reduced ischemic injury and improved cardiac functional recovery on reperfusion in aged hearts. These data indicate that innate increases in activity of the PP enzymes augment myocardial vulnerability to I/R injury in aging, and that blockers of PP protect the vulnerable aging hearts.
Collapse
Affiliation(s)
- Radha Ananthakrishnan
- Diabetes Research Program, Department of Medicine, New York University Langone Medical Center, New York, NY 10016, USA
| | | | | | | | | |
Collapse
|
38
|
Gu J, Wang JJ, Yan J, Cui CF, Wu WH, Li L, Wang ZS, Yu M, Gao N, Liu L, Ouyang DS. Effects of lignans extracted from Eucommia ulmoides and aldose reductase inhibitor epalrestat on hypertensive vascular remodeling. JOURNAL OF ETHNOPHARMACOLOGY 2011; 133:6-13. [PMID: 20817083 DOI: 10.1016/j.jep.2010.08.055] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2010] [Revised: 08/23/2010] [Accepted: 08/26/2010] [Indexed: 05/04/2023]
Abstract
AIM OF THE STUDY To investigate the effects of lignans extracted from Eucommia ulmoides and epalrestat on vascular remodeling in spontaneously hypertensive rats. MATERIALS AND METHODS Ten-week-old male spontaneously hypertensive rats were randomly divided into 3 groups (12 rats each group), and treated orally with 100 mg/kg/d of captopril (an angiotensin-converting enzyme inhibitor), 100 mg/kg/d of epalrestat (an aldose reductase inhibitor) and 300 mg/kg/d of lignans by gavage daily for 16 weeks, respectively. Sex-, age-, and number-matched spontaneously hypertensive rats and normotensive Wistar Kyoto rats, were treated with distilled water (vehicle) as controls. The rats were weighed weekly. Mean arterial blood pressure and heart rate were measured periodically by non-invasive blood pressure monitoring. They were sacrificed at the end of experiment (26-week-old). Superior mesenteric artery and aorta were isolated for determination of histomorphometry and the expression of aldose reductase by immunohistochemistry. RESULTS Captopril and lignans, but not epalrestat, decreased mean arterial blood pressure in spontaneously hypertensive rats. Vascular remodeling was improved in all three treated groups by histomorphometry. CONCLUSIONS Both lignans and epalrestat reversed hypertensive vascular remodeling. Aldose reductase played a vital role in the pathologic process of hypertensive vascular remodeling rather than elevation of blood pressure. These data suggested that aldose reductase could be a new therapeutic target for the treatment of cardiovascular diseases.
Collapse
Affiliation(s)
- Juan Gu
- Institute of Clinical Pharmacology, Central South University, Changsha, Hunan, PR China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Hallam KM, Li Q, Ananthakrishnan R, Kalea A, Zou YS, Vedantham S, Schmidt AM, Yan SF, Ramasamy R. Aldose reductase and AGE-RAGE pathways: central roles in the pathogenesis of vascular dysfunction in aging rats. Aging Cell 2010; 9:776-84. [PMID: 20670350 DOI: 10.1111/j.1474-9726.2010.00606.x] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Aging is inevitably accompanied by gradual and irreversible innate endothelial dysfunction. In this study, we tested the hypothesis that accentuation of glucose metabolism via the aldose reductase (AR) pathway contributes to age-related vascular dysfunction. AR protein and activity levels were significantly increased in aged vs. young aortic homogenates from Fischer 344 rats. Immunostaining revealed that the principal site of increased AR protein was the aortic endothelium as well as smooth muscle cells. Studies revealed that endothelial-dependent relaxation (EDR) in response to acetylcholine was impaired in aged rats compared to young rats and that treatment with the AR inhibitor (ARI) zopolrestat significantly improved EDR in aged rats. Methylglyoxal (MG), a key precursor of advanced glycation endproducts (AGEs), was significantly increased in the aortas of aged rats vs. young rats. Consistent with central roles for AR in generation of MG in aging, ARI treatment significantly reduced MG levels in aged rat aorta to those in young rats. Treatment of aged rats with soluble(s) RAGE, a soluble form of the chief signal transduction receptor for AGEs, RAGE, significantly improved EDR in aged rats, thus establishing the contribution of age-related increases in AGEs to endothelial dysfunction. These findings reveal that significant increases in AR expression and activity in aged rat vasculature linked to endothelial dysfunction may be mitigated, at least in part, via ARI and that aging-linked increased flux via AR generates AGEs; species which transduce endothelial injury consequent to their interaction with RAGE. These data demonstrate for the first time that AR mediates aging-related vascular dysfunction, at least in part, via RAGE.
Collapse
|
40
|
Wu LY, Ma ZM, Fan XL, Zhao T, Liu ZH, Huang X, Li MM, Xiong L, Zhang K, Zhu LL, Fan M. The anti-necrosis role of hypoxic preconditioning after acute anoxia is mediated by aldose reductase and sorbitol pathway in PC12 cells. Cell Stress Chaperones 2010; 15:387-94. [PMID: 19902381 PMCID: PMC3082650 DOI: 10.1007/s12192-009-0153-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2009] [Revised: 09/14/2009] [Accepted: 10/21/2009] [Indexed: 12/14/2022] Open
Abstract
It has been demonstrated that hypoxic preconditioning (HP) enhances the survival ability of the organism against the subsequent acute anoxia (AA). However, it is not yet clear whether necrosis induced by AA can be prevented by HP, and what are the underlying mechanisms. In this study, we examined the effect of HP (10% O(2), 48 h) on necrosis induced by AA (0% O(2), 24 h) in PC12 cells. We found that HP delayed the regulatory volume decrease and reduced cell swelling after 24 h of exposure to AA. Since aldose reductase (AR) is involved in cell volume regulation, we detected AR mRNA expression with reverse transcription-polymerase chain reaction (RT-PCR) techniques. The AR mRNA level was dramatically elevated by HP. Furthermore, an HP-induced decrease in cell injury was reversed by berberine chloride (BB), the inhibitor of AR. In addition, sorbitol synthesized from glucose catalyzed by AR is directly related to cell volume regulation. Subsequently, we tested sorbitol content in the cytoplasm. HP clearly elevated sorbitol content, while BB inhibited the elevation induced by HP. Further study showed that a strong inhibitor of sorbitol permease, quinidine, completely reversed the protection induced by HP after AA. These data provide evidence that HP prevents necrosis induced by AA and is mediated by AR and sorbitol pathway.
Collapse
Affiliation(s)
- Li-Ying Wu
- Department of Brain Protection and Plasticity, Institute of Basic Medical Sciences, Taiping Rd 27, Beijing, 100850 People’s Republic of China
| | - Zi-Min Ma
- Department of Brain Protection and Plasticity, Institute of Basic Medical Sciences, Taiping Rd 27, Beijing, 100850 People’s Republic of China
| | - Xue-Lai Fan
- Department of Brain Protection and Plasticity, Institute of Basic Medical Sciences, Taiping Rd 27, Beijing, 100850 People’s Republic of China
| | - Tong Zhao
- Department of Brain Protection and Plasticity, Institute of Basic Medical Sciences, Taiping Rd 27, Beijing, 100850 People’s Republic of China
| | - Zhao-Hui Liu
- Department of Brain Protection and Plasticity, Institute of Basic Medical Sciences, Taiping Rd 27, Beijing, 100850 People’s Republic of China
| | - Xin Huang
- Department of Brain Protection and Plasticity, Institute of Basic Medical Sciences, Taiping Rd 27, Beijing, 100850 People’s Republic of China
| | - Ming-Ming Li
- Department of Brain Protection and Plasticity, Institute of Basic Medical Sciences, Taiping Rd 27, Beijing, 100850 People’s Republic of China
| | - Lei Xiong
- Department of Brain Protection and Plasticity, Institute of Basic Medical Sciences, Taiping Rd 27, Beijing, 100850 People’s Republic of China
| | - Kuan Zhang
- Department of Brain Protection and Plasticity, Institute of Basic Medical Sciences, Taiping Rd 27, Beijing, 100850 People’s Republic of China
| | - Ling-Ling Zhu
- Department of Brain Protection and Plasticity, Institute of Basic Medical Sciences, Taiping Rd 27, Beijing, 100850 People’s Republic of China
| | - Ming Fan
- Department of Brain Protection and Plasticity, Institute of Basic Medical Sciences, Taiping Rd 27, Beijing, 100850 People’s Republic of China
| |
Collapse
|
41
|
Wetzelberger K, Baba SP, Thirunavukkarasu M, Ho YS, Maulik N, Barski OA, Conklin DJ, Bhatnagar A. Postischemic deactivation of cardiac aldose reductase: role of glutathione S-transferase P and glutaredoxin in regeneration of reduced thiols from sulfenic acids. J Biol Chem 2010; 285:26135-48. [PMID: 20538586 DOI: 10.1074/jbc.m110.146423] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Aldose reductase (AR) is a multifunctional enzyme that catalyzes the reduction of glucose and lipid peroxidation-derived aldehydes. During myocardial ischemia, the activity of AR is increased due to the oxidation of its cysteine residues to sulfenic acids. It is not known, however, whether the activated, sulfenic form of the protein (AR-SOH) is converted back to its reduced, unactivated state (AR-SH). We report here that in perfused mouse hearts activation of AR during 15 min of global ischemia is completely reversed by 30 min of reperfusion. During reperfusion, AR-SOH was converted to a mixed disulfide (AR-SSG). Deactivation of AR and the appearance of AR-SSG during reperfusion were delayed in hearts of mice lacking glutathione S-transferase P (GSTP). In vitro, GSTP accelerated glutathiolation and inactivation of AR-SOH. Reduction of AR-SSG to AR-SH was facilitated by glutaredoxin (GRX). Ischemic activation of AR was increased in GRX-null hearts but was attenuated in the hearts of cardiospecific GRX transgenic mice. Incubation of AR-SSG with GRX led to the regeneration of the reduced form of the enzyme. In ischemic cardiospecific AR transgenic hearts, AR was co-immunoprecipitated with GSTP, whereas in reperfused hearts, the association of AR with GRX was increased. These findings suggest that upon reperfusion of the ischemic heart AR-SOH is converted to AR-SSG via GSTP-assisted glutathiolation. AR-SSG is then reduced by GRX to AR-SH. Sequential catalysis by GSTP and GRX may be a general redox switching mechanism that regulates the reduction of protein sulfenic acids to cysteines.
Collapse
Affiliation(s)
- Karin Wetzelberger
- Diabetes and Obesity Center, University of Louisville, Louisville, Kentucky 40202, USA
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Ramasamy R, Goldberg IJ. Aldose reductase and cardiovascular diseases, creating human-like diabetic complications in an experimental model. Circ Res 2010; 106:1449-58. [PMID: 20466987 DOI: 10.1161/circresaha.109.213447] [Citation(s) in RCA: 104] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Hyperglycemia and reduced insulin actions affect many biological processes. One theory is that aberrant metabolism of glucose via several pathways including the polyol pathway causes cellular toxicity. Aldose reductase (AR) is a multifunctional enzyme that reduces aldehydes. Under diabetic conditions AR converts glucose into sorbitol, which is then converted to fructose. This article reviews the biology and pathobiology of AR actions. AR expression varies considerably among species. In humans and rats, the higher level of AR expression is associated with toxicity. Flux via AR is increased by ischemia and its inhibition during ischemia reperfusion reduces injury. However, similar pharmacological effects are not observed in mice unless they express a human AR transgene. This is because mice have much lower levels of AR expression, probably insufficient to generate toxic byproducts. Human AR expression in LDL receptor knockout mice exacerbates vascular disease, but only under diabetic conditions. In contrast, a recent report suggests that genetic ablation of AR increased atherosclerosis and increased hydroxynonenal in arteries. It was hypothesized that AR knockout prevented reduction of toxic aldehydes. Like many in vivo effects found in genetically manipulated animals, interpretation requires the reproduction of human-like physiology. For AR, this will require tissue specific expression of AR in sites and at levels that approximate those in humans.
Collapse
Affiliation(s)
- Ravichandran Ramasamy
- Department of Surgery, Columbia University College of Physicians and Surgeons, New York, NY 10032, USA.
| | | |
Collapse
|
43
|
Calderone V, Testai L, Martelli A, Motta CL, Sartini S, Da Settimo F, Breschi MC. Anti-ischaemic activity of an antioxidant aldose reductase inhibitor on diabetic and non-diabetic rat hearts. J Pharm Pharmacol 2010; 62:107-13. [DOI: 10.1211/jpp.62.01.0012] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
Abstract
Objectives
Many observations report the cardioprotective effects of inhibitors of aldose reductase in different models of ischaemia–reperfusion injury in diabetic myocardium. In this paper, the inhibitory effects of the new pyrido[1,2-a]-pyrimidin-4-one derivative PPO, whose aldose reductase-inhibitory and antioxidant effects were shown in a previous study, were evaluated.
Methods
The effect of PPO was evaluated on aldose reductase from hearts of diabetic and non-diabetic rats, and compared with that of the reference drug epalrestat. Moreover, the two drugs were tested on isolated and Langendorff-perfused diabetic and non-diabetic hearts submitted to ischaemia–reperfusion cycle.
Key findings
Epalrestat showed equivalent levels of potency in inhibiting the activity of the enzyme in the diabetic and in the non-diabetic hearts. On the contrary, the inhibitory potency of PPO was decreased in the diabetic organs. In the diabetic hearts submitted to ischaemia–reperfusion, an increased level of heart aldose reductase activity was recorded, and both PPO and epalrestat produced cardioprotective effects, suggesting that aldose reductase is deeply involved in the process of ischaemia–reperfusion injury in diabetic myocardium. In non-diabetic hearts, where aldose reductase has a lower activity, epalrestat failed to produce significant protection, while PPO still maintained cardioprotective effects, which may be reasonably attributed to useful ‘ancillary’ effects – such as antioxidant activity – independent from the aldose reductase inhibition.
Conclusions
Therefore PPO, a new molecule endowed with both aldose reductase-inhibitory effects and antioxidant activity, may represent the prototype of a new class of multitarget drugs, focused on two different steps deeply involved in the pathogenesis of ischaemic injury of diabetic hearts.
Collapse
Affiliation(s)
- Vincenzo Calderone
- Dipartimento di Psichiatria, Neurobiologia, Farmacologia, Biotecnologie, Università di Pisa, Pisa, Italy
| | - Lara Testai
- Dipartimento di Psichiatria, Neurobiologia, Farmacologia, Biotecnologie, Università di Pisa, Pisa, Italy
| | - Alma Martelli
- Dipartimento di Psichiatria, Neurobiologia, Farmacologia, Biotecnologie, Università di Pisa, Pisa, Italy
| | | | - Stefania Sartini
- Dipartimento di Scienze Farmaceutiche, Università di Pisa, Pisa, Italy
| | | | - Maria C Breschi
- Dipartimento di Psichiatria, Neurobiologia, Farmacologia, Biotecnologie, Università di Pisa, Pisa, Italy
| |
Collapse
|
44
|
Cipriani V, Ranzato E, Balbo V, Mazzucco L, Cavaletto M, Patrone M. Long-term effect of platelet lysate on primary fibroblasts highlighted with a proteomic approach. J Tissue Eng Regen Med 2009; 3:531-8. [DOI: 10.1002/term.195] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|
45
|
Ananthakrishnan R, Kaneko M, Hwang YC, Quadri N, Gomez T, Li Q, Caspersen C, Ramasamy R. Aldose reductase mediates myocardial ischemia-reperfusion injury in part by opening mitochondrial permeability transition pore. Am J Physiol Heart Circ Physiol 2008; 296:H333-41. [PMID: 19060123 DOI: 10.1152/ajpheart.01012.2008] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Aldose reductase (AR), a member of the aldo-keto reductase family, has been demonstrated to play a central role in mediating myocardial ischemia-reperfusion (I/R) injury. Recently, using transgenic mice broadly overexpressing human AR (ARTg), we demonstrated that AR is an important component of myocardial I/R injury and that inhibition of this enzyme protects heart from I/R injury (20-22, 48, 49, 56). To rigorously delineate mechanisms by which AR pathway influences myocardial ischemic injury, we investigated the role played by reactive oxygen species (ROS), antioxidant enzymes, and mitochondrial permeability transition (MPT) pore opening in hearts from ARTg or littermates [wild type (WT)] subjected to I/R. MPT pore opening after I/R was determined using mitochondrial uptake of 2-deoxyglucose ratio, while H2O2 was measured as a key indicator of ROS. Myocardial 2-deoxyglucose uptake ratio and calcium-induced swelling were significantly greater in mitochondria from ARTg mice than in WT mice. Blockade of MPT pore with cyclosphorin A during I/R reduced ischemic injury significantly in ARTg mice hearts. H2O2 measurements indicated mitochondrial ROS generation after I/R was significantly greater in ARTg mitochondria than in WT mice hearts. Furthermore, the levels of antioxidant GSH were significantly reduced in ARTg mitochondria than in WT. Resveratrol treatment or pharmacological blockade of AR significantly reduced ROS generation and MPT pore opening in mitochondria of ARTg mice hearts exposed to I/R stress. This study demonstrates that MPT pore opening is a key event by which AR pathway mediates myocardial I/R injury, and that the MPT pore opening after I/R is triggered, in part, by increases in ROS generation in ARTg mice hearts. Therefore, inhibition of AR pathway protects mitochondria and hence may be a useful adjunct for salvaging ischemic myocardium.
Collapse
Affiliation(s)
- Radha Ananthakrishnan
- Division of Surgical Science , Department of Surgery, College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Regenold WT, Hisley KC, Phatak P, Marano CM, Obuchowski A, Lefkowitz DM, Sassan A, Ohri S, Phillips TL, Dosanjh N, Conley RR, Gullapalli R. Relationship of cerebrospinal fluid glucose metabolites to MRI deep white matter hyperintensities and treatment resistance in bipolar disorder patients. Bipolar Disord 2008; 10:753-64. [PMID: 19032707 PMCID: PMC3753008 DOI: 10.1111/j.1399-5618.2008.00626.x] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
OBJECTIVES Both diabetes mellitus and magnetic resonance image (MRI) deep white matter hyperintensities (WMHs) are more common in bipolar disorder (BD) patients than in matched controls. Deep-as opposed to periventricular--WMHs and diabetes are associated with treatment resistance and poorer outcome. This study investigated whether brain glucose metabolism by the polyol pathway--a pathway linked to nervous tissue disease in diabetes--is related to deep WMH volume and treatment resistance in BD patients. METHODS Volumes of fluid-attenuated inversion recovery WMHs were quantified and correlated with cerebrospinal fluid (CSF) concentrations of glucose metabolites in 20 nondiabetic patients with BD and nondiabetic comparison subjects with schizophrenia (n = 15) or transient neurologic symptoms (neurologic controls, n = 15). RESULTS BD patients, but not schizophrenic patients, had significantly greater volumes of deep but not periventricular WMHs compared to neurologic controls. BD subjects also had significantly greater CSF concentrations of sorbitol and fructose (the polyol pathway metabolites of glucose) compared to controls. Significant positive correlations between CSF metabolites and WMH volumes were found only in the BD group and were between deep WMH volumes and CSF sorbitol (rho = 0.487, p = 0.029) and fructose (rho = 0.474, p = 0.035). An index of treatment resistance correlated significantly with deep WMH volume (rho = 0.578, p = 0.008), sorbitol (rho = 0.542, p = 0.013), and fructose (rho = 0.692, p = 0.001) in BD subjects but not in other subjects. CONCLUSIONS This is the first reported evidence of relationships between abnormal brain glucose metabolism and both deep WMHs and treatment resistance in a group of BD patients. Further studies are necessary to determine the significance of these findings to BD pathophysiology.
Collapse
Affiliation(s)
- William T Regenold
- Department of Psychiatry, Division of Geriatric Psychiatry, University of Maryland School of Medicine, Baltimore, MD,
| | - K Calvin Hisley
- Department of Diagnostic Radiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Pornima Phatak
- Division of Geriatric Psychiatry, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Christopher M Marano
- Division of Geriatric Psychiatry, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Abraham Obuchowski
- Department of Diagnostic Radiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - David M Lefkowitz
- Department of Diagnostic Radiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Amritpal Sassan
- Division of Geriatric Psychiatry, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Sameer Ohri
- Division of Geriatric Psychiatry, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Tony L Phillips
- Division of Geriatric Psychiatry, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Narveen Dosanjh
- Division of Geriatric Psychiatry, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Robert R Conley
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Rao Gullapalli
- Department of Diagnostic Radiology, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
47
|
Noh HL, Hu Y, Park TS, DiCioccio T, Nichols AJ, Okajima K, Homma S, Goldberg IJ. Regulation of plasma fructose and mortality in mice by the aldose reductase inhibitor lidorestat. J Pharmacol Exp Ther 2008; 328:496-503. [PMID: 18974362 DOI: 10.1124/jpet.108.136283] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Aldose reductase (AR), an enzyme widely believed to be involved in the aberrant metabolism of glucose and development of diabetic complications, is expressed at low levels in the mouse. We studied whether expression of human AR (hAR), its inhibition with lidorestat, which is an AR inhibitor (ARI), and the presence of streptozotocin (STZ)-induced diabetes altered plasma fructose, mortality, and/or vascular lesions in low-density lipoprotein (LDL) receptor-deficient [Ldlr(-/-)] mice. Mice were made diabetic at 12 weeks of age with low-dose STZ treatment. Four weeks later, the diabetic animals (glucose > 20 mM) were blindly assigned to a 0.15% cholesterol diet with or without ARI. After 4 and 6 weeks, there were no significant differences in body weights or plasma cholesterol, triglyceride, and glucose levels between the groups. Diabetic Ldlr(-/-) mice receiving ARI had plasma fructose levels of 5.2 +/- 2.3 microg/ml; placebo-treated mice had plasma fructose levels of 12.08 +/- 7.4 microg/ml, p < 0.01, despite the induction of fructose-metabolizing enzymes, fructose kinase and adolase B. After 6 weeks, hAR/Ldlr(-/-) mice on the placebo-containing diet had greater mortality (31%, n = 9/26 versus 6%, n = 1/21, p < 0.05). The mortality rate in the ARI-treated group was similar to that in non-hAR-expressing mice. Therefore, diabetic hAR-expressing mice had increased fructose and greater mortality that was corrected by inclusion of lidorestat, an ARI, in the diet. If similar effects are found in humans, such treatment could improve clinical outcome in diabetic patients.
Collapse
Affiliation(s)
- Hye-Lim Noh
- Department of Medicine, Columbia University College of Physicians and Surgeons, New York, New York 10032, USA
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Li Q, Hwang YC, Ananthakrishnan R, Oates PJ, Guberski D, Ramasamy R. Polyol pathway and modulation of ischemia-reperfusion injury in Type 2 diabetic BBZ rat hearts. Cardiovasc Diabetol 2008; 7:33. [PMID: 18957123 PMCID: PMC2584021 DOI: 10.1186/1475-2840-7-33] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2008] [Accepted: 10/28/2008] [Indexed: 12/03/2022] Open
Abstract
We investigated the role of polyol pathway enzymes aldose reductase (AR) and sorbitol dehydrogenase (SDH) in mediating injury due to ischemia-reperfusion (IR) in Type 2 diabetic BBZ rat hearts. Specifically, we investigated, (a) changes in glucose flux via cardiac AR and SDH as a function of diabetes duration, (b) ischemic injury and function after IR, (c) the effect of inhibition of AR or SDH on ischemic injury and function. Hearts isolated from BBZ rats, after 12 weeks or 48 weeks diabetes duration, and their non-diabetic littermates, were subjected to IR protocol. Myocardial function, substrate flux via AR and SDH, and tissue lactate:pyruvate (L/P) ratio (a measure of cytosolic NADH/NAD+), and lactate dehydrogenase (LDH) release (a marker of IR injury) were measured. Zopolrestat, and CP-470,711 were used to inhibit AR and SDH, respectively. Myocardial sorbitol and fructose content, and associated changes in L/P ratios were significantly higher in BBZ rats compared to non-diabetics, and increased with disease duration. Induction of IR resulted in increased ischemic injury, reduced ATP levels, increases in L/P ratio, and poor cardiac function in BBZ rat hearts, while inhibition of AR or SDH attenuated these changes and protected hearts from IR injury. These data indicate that AR and SDH are key modulators of myocardial IR injury in BBZ rat hearts and that inhibition of polyol pathway could in principle be used as a therapeutic adjunct for protection of ischemic myocardium in Type 2 diabetic patients.
Collapse
Affiliation(s)
- Qing Li
- Division of Surgical Science, Department of Surgery, Columbia University College of Physicians and Surgeons, New York, NY 10032, USA
| | - Yuying C Hwang
- Division of Surgical Science, Department of Surgery, Columbia University College of Physicians and Surgeons, New York, NY 10032, USA
| | - Radha Ananthakrishnan
- Division of Surgical Science, Department of Surgery, Columbia University College of Physicians and Surgeons, New York, NY 10032, USA
| | | | | | - Ravichandran Ramasamy
- Division of Surgical Science, Department of Surgery, Columbia University College of Physicians and Surgeons, New York, NY 10032, USA
| |
Collapse
|
49
|
Bucciarelli LG, Ananthakrishnan R, Hwang YC, Kaneko M, Song F, Sell DR, Strauch C, Monnier VM, Yan SF, Schmidt AM, Ramasamy R. RAGE and modulation of ischemic injury in the diabetic myocardium. Diabetes 2008; 57:1941-51. [PMID: 18420491 PMCID: PMC2453611 DOI: 10.2337/db07-0326] [Citation(s) in RCA: 90] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2007] [Accepted: 04/09/2008] [Indexed: 01/18/2023]
Abstract
OBJECTIVE Subjects with diabetes experience an increased risk of myocardial infarction and cardiac failure compared with nondiabetic age-matched individuals. The receptor for advanced glycation end products (RAGE) is upregulated in diabetic tissues. In this study, we tested the hypothesis that RAGE affected ischemia/reperfusion (I/R) injury in the diabetic myocardium. In diabetic rat hearts, expression of RAGE and its ligands was enhanced and localized particularly to both endothelial cells and mononuclear phagocytes. RESEARCH DESIGN AND METHODS To specifically dissect the impact of RAGE, homozygous RAGE-null mice and transgenic (Tg) mice expressing cytoplasmic domain-deleted RAGE (DN RAGE), in which RAGE-dependent signal transduction was deficient in endothelial cells or mononuclear phagocytes, were rendered diabetic with streptozotocin. Isolated perfused hearts were subjected to I/R. RESULTS Diabetic RAGE-null mice were significantly protected from the adverse impact of I/R injury in the heart, as indicated by decreased release of LDH and lower glycoxidation products carboxymethyl-lysine (CML) and pentosidine, improved functional recovery, and increased ATP. In diabetic Tg mice expressing DN RAGE in endothelial cells or mononuclear phagocytes, markers of ischemic injury and CML were significantly reduced, and levels of ATP were increased in heart tissue compared with littermate diabetic controls. Furthermore, key markers of apoptosis, caspase-3 activity and cytochrome c release, were reduced in the hearts of diabetic RAGE-modified mice compared with wild-type diabetic littermates in I/R. CONCLUSIONS These findings demonstrate novel and key roles for RAGE in I/R injury in the diabetic heart.
Collapse
MESH Headings
- Animals
- Diabetes Mellitus, Experimental/complications
- Diabetes Mellitus, Experimental/drug therapy
- Diabetes Mellitus, Experimental/genetics
- Diabetic Angiopathies/genetics
- Insulin/therapeutic use
- Male
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Transgenic
- Myocardial Ischemia/genetics
- Rats
- Rats, Inbred BB
- Receptor for Advanced Glycation End Products
- Receptors, Immunologic/deficiency
- Receptors, Immunologic/genetics
- Receptors, Immunologic/metabolism
- Ventricular Dysfunction, Left/genetics
Collapse
Affiliation(s)
- Loredana G. Bucciarelli
- From the Division of Surgical Science, Department of Surgery, Columbia University Medical Center, New York, New York; and the Department of Pathology, Case Western Reserve University, Cleveland, Ohio
| | - Radha Ananthakrishnan
- From the Division of Surgical Science, Department of Surgery, Columbia University Medical Center, New York, New York; and the Department of Pathology, Case Western Reserve University, Cleveland, Ohio
| | - Yuying C. Hwang
- From the Division of Surgical Science, Department of Surgery, Columbia University Medical Center, New York, New York; and the Department of Pathology, Case Western Reserve University, Cleveland, Ohio
| | - Michiyo Kaneko
- From the Division of Surgical Science, Department of Surgery, Columbia University Medical Center, New York, New York; and the Department of Pathology, Case Western Reserve University, Cleveland, Ohio
| | - Fei Song
- From the Division of Surgical Science, Department of Surgery, Columbia University Medical Center, New York, New York; and the Department of Pathology, Case Western Reserve University, Cleveland, Ohio
| | - David R. Sell
- From the Division of Surgical Science, Department of Surgery, Columbia University Medical Center, New York, New York; and the Department of Pathology, Case Western Reserve University, Cleveland, Ohio
| | - Christopher Strauch
- From the Division of Surgical Science, Department of Surgery, Columbia University Medical Center, New York, New York; and the Department of Pathology, Case Western Reserve University, Cleveland, Ohio
| | - Vincent M. Monnier
- From the Division of Surgical Science, Department of Surgery, Columbia University Medical Center, New York, New York; and the Department of Pathology, Case Western Reserve University, Cleveland, Ohio
| | - Shi Fang Yan
- From the Division of Surgical Science, Department of Surgery, Columbia University Medical Center, New York, New York; and the Department of Pathology, Case Western Reserve University, Cleveland, Ohio
| | - Ann Marie Schmidt
- From the Division of Surgical Science, Department of Surgery, Columbia University Medical Center, New York, New York; and the Department of Pathology, Case Western Reserve University, Cleveland, Ohio
| | - Ravichandran Ramasamy
- From the Division of Surgical Science, Department of Surgery, Columbia University Medical Center, New York, New York; and the Department of Pathology, Case Western Reserve University, Cleveland, Ohio
| |
Collapse
|
50
|
Kaiserova K, Tang XL, Srivastava S, Bhatnagar A. Role of nitric oxide in regulating aldose reductase activation in the ischemic heart. J Biol Chem 2008; 283:9101-12. [PMID: 18223294 PMCID: PMC2431016 DOI: 10.1074/jbc.m709671200] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2007] [Revised: 01/24/2008] [Indexed: 01/04/2023] Open
Abstract
Aldose reductase (AR) catalyzes the reduction of several aldehydes ranging from lipid peroxidation products to glucose. The activity of AR is increased in the ischemic heart due to oxidation of its cysteine residues, but the underlying mechanisms remain unclear. To examine signaling mechanisms regulating AR activation, we studied the role of nitric oxide (NO). Treatment with the NO synthase (NOS) inhibitor, N-nitro-l-arginine methyl ester prevented ischemia-induced AR activation and myocardial sorbitol accumulation in rat hearts subjected to global ischemia ex vivo or coronary ligation in situ, whereas inhibition of inducible NOS and neuronal NOS had no effect. Activation of AR in the ischemic heart was abolished by pretreatment with peroxynitrite scavengers hesperetin or 5, 10, 15, 20-tetrakis-[4-sulfonatophenyl]-porphyrinato-iron [III]. Site-directed mutagenesis and electrospray ionization mass spectrometry analyses showed that Cys-298 of AR was readily oxidized to sulfenic acid by peroxynitrite. Treatment with bradykinin and insulin led to a phosphatidylinositol 3-kinase (PI3K)-dependent increase in the phosphorylation of endothelial NOS at Ser-1177 and, even in the absence of ischemia, was sufficient in activating AR. Activation of AR by bradykinin and insulin was reversed upon reduction with dithiothreitol or by inhibiting NOS or PI3K. Treatment with AR inhibitors sorbinil or tolrestat reduced post-ischemic recovery in the rat hearts subjected to global ischemia and increased the infarct size when given before ischemia or upon reperfusion. These results suggest that AR is a cardioprotective protein and that its activation in the ischemic heart is due to peroxynitrite-mediated oxidation of Cys-298 to sulfenic acid via the PI3K/Akt/endothelial NOS pathway.
Collapse
Affiliation(s)
- Karin Kaiserova
- Institute of Molecular Cardiology, University of Louisville, Louisville, KY 40202, USA
| | | | | | | |
Collapse
|