1
|
Ganapathy A A, Hari Priya VM, Baby K, Bindhu S, Jayan R, Krishnamoorthi R, Somappa SB, Nayak Y, Kumaran A. Flavone-C-glycosides from Cassia auriculata L. as possible inhibitors of phosphodiesterase-5 (PDE5): in vitro, molecular docking and molecular dynamics studies. J Biomol Struct Dyn 2024:1-23. [PMID: 39589221 DOI: 10.1080/07391102.2024.2431659] [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: 11/30/2023] [Accepted: 05/03/2024] [Indexed: 11/27/2024]
Abstract
Phosphodiesterase-5 (PDE5) is a homodimeric enzyme that specifically targets cyclic guanosine monophosphate (cGMP), that mediates many downstream effects such as vasodilation, neurotransmission, and calcium homeostasis. Considering the functions of cGMP, inhibition of PDE5 has been established to have several therapeutic effects in disease conditions such as cancer, cardiovascular diseases and Alzheimer's disease. Consequently, many PDE5 inhibitors were developed but with severe adverse effects such as non-arteritic anterior ischemic optic neuropathy (NAION), priapism, etc. Hence, in our study for the identification of new PDE5 inhibitors from alternative sources, Cassia auriculata L. was identified as a potential PDE5 inhibitors with 56.23% inhibition at 100 μg/mL in vitro. In addition, the respective phytoconstituents were evaluated through molecular docking, interaction studies and MM/GBSA binding free energy calculations, identifying two potential flavone C-glycosides, lucenin-II (-15.977, dG bind = -38.8), stellarin-II (-15.099, dG bind = -34.59), and a flavan derivative (2S)-7,4-dihydroxyflavan(4β-8)-catechin, in comparison to sildenafil (-10.890, dG bind = -75.4) and having frequent contacts with Phe 786, Phe 820, Ser 663, Tyr 664, and other crucial residues at the catalytic site of PDE5. Molecular dynamics simulations performed for 100 ns showed structural stability and compactness of the candidates through RMSD, RMSF which showed less fluctuations. The ADMET analysis revealed favorable pharmacokinetics, and pharmacodynamic properties with no subsequent toxicity in normal cells. The biological target class prediction identified enzymes with similar properties and icariin, which is a well-established natural PDE5 inhibitor was identified as a structurally similar analogue. These findings could lead to the development of novel natural product based PDE5 inhibitors.
Collapse
Affiliation(s)
- Anand Ganapathy A
- Chemical Sciences and Technology Division, CSIR-National Institute for Interdisciplinary Science and Technology, Thiruvananthapuram, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Vijayakumari Mahadevan Hari Priya
- Chemical Sciences and Technology Division, CSIR-National Institute for Interdisciplinary Science and Technology, Thiruvananthapuram, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Krishnaprasad Baby
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Sreelekshmy Bindhu
- Department of Chemistry and Polymer Chemistry, Kumbalathu Sankupillai Memorial Devaswom Board College, Sasthamcotta, India
| | - Raji Jayan
- Department of Chemistry, Sree Narayana College, Punalur, India
| | - Raman Krishnamoorthi
- Chemical Sciences and Technology Division, CSIR-National Institute for Interdisciplinary Science and Technology, Thiruvananthapuram, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Sasidhar Balappa Somappa
- Chemical Sciences and Technology Division, CSIR-National Institute for Interdisciplinary Science and Technology, Thiruvananthapuram, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Yogendra Nayak
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Alaganandam Kumaran
- Chemical Sciences and Technology Division, CSIR-National Institute for Interdisciplinary Science and Technology, Thiruvananthapuram, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| |
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
|
Li L, Ling Z, Wang X, Zhang X, Li Y, Gao G. Proteomics-based screening of AKR1B1 as a therapeutic target and validation study for sepsis-associated acute kidney injury. PeerJ 2024; 12:e16709. [PMID: 38188141 PMCID: PMC10768659 DOI: 10.7717/peerj.16709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 12/01/2023] [Indexed: 01/09/2024] Open
Abstract
Background Sepsis and sepsis-associated acute kidney injury (SA-AKI) pose significant global health challenges, necessitating the development of innovative therapeutic strategies. Dysregulated protein expression has been implicated in the initiation and progression of sepsis and SA-AKI. Identifying potential protein targets and modulating their expression is crucial for exploring alternative therapies. Method We established an SA-AKI rat model using cecum ligation perforation (CLP) and employed differential proteomic techniques to identify protein expression variations in kidney tissues. Aldose reductase (AKR1B1) emerged as a promising target. The SA-AKI rat model received treatment with the aldose reductase inhibitor (ARI), epalrestat. Blood urea nitrogen (BUN) and creatinine (CRE) levels, as well as IL-1β, IL-6 and TNF-α levels in the serum and kidney tissues, were monitored. Hematoxylin-eosin (H-E) staining and a pathological damage scoring scale assessed renal tissue damage, while protein blotting determined PKC (protein kinase C)/NF-κB pathway protein expression. Result Differential proteomics revealed significant downregulation of seven proteins and upregulation of 17 proteins in the SA-AKI rat model renal tissues. AKR1B1 protein expression was notably elevated, confirmed by Western blot. ARI prophylactic administration and ARI treatment groups exhibited reduced renal injury, low BUN and CRE levels and decreased IL-1β, IL-6 and TNF-α levels compared to the CLP group. These changes were statistically significant (P < 0.05). AKR1B1, PKC-α, and NF-κB protein expression levels were also lowered in the ARI prophylactic administration and ARI treatment groups compared to the CLP group (P < 0.05). Conclusions Epalrestat appeared to inhibit the PKC/NF-κB inflammatory pathway by inhibiting AKR1B1, resulting in reduced inflammatory cytokine levels in renal tissues and blood. This mitigated renal tissue injuries and improved the systemic inflammatory response in the severe sepsis rat model. Consequently, AKR1B1 holds promise as a target for treating sepsis-associated acute kidney injuries.
Collapse
Affiliation(s)
- Lei Li
- Intensive Care Unit, Shandong Public Health Clinical Center, Shandong University, Jinan, China
| | - Zaiqin Ling
- Department of Tubercular Medicine, Shandong Public Health Clinical Center, Shandong University, Jinan, China
| | - Xingsheng Wang
- Department of Emergency, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Xinxin Zhang
- Department of Emergency Medicine, Fuyang People’s Hospital of Anhui Medical University, Fuyang, China
| | - Yun Li
- Intensive Care Unit, Central Hospital Affliated to Shandong First Medical University, Jinan, China
| | - Guangsheng Gao
- Neurological Intensive Care Unit, Central Hospital Affliated to Shandong First Medical University, Jinan, China
| |
Collapse
|
4
|
Syamprasad NP, Jain S, Rajdev B, Prasad N, Kallipalli R, Naidu VGM. Aldose reductase and cancer metabolism: The master regulator in the limelight. Biochem Pharmacol 2023; 211:115528. [PMID: 37011733 DOI: 10.1016/j.bcp.2023.115528] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Revised: 03/25/2023] [Accepted: 03/27/2023] [Indexed: 04/03/2023]
Abstract
It is strongly established that metabolic reprogramming mediates the initiation, progression, and metastasis of a variety of cancers. However, there is no common biomarker identified to link the dysregulated metabolism and cancer progression. Recent studies strongly advise the involvement of aldose reductase (AR) in cancer metabolism. AR-mediated glucose metabolism creates a Warburg-like effect and an acidic tumour microenvironment in cancer cells. Moreover, AR overexpression is associated with the impairment of mitochondria and the accumulation of free fatty acids in cancer cells. Further, AR-mediated reduction of lipid aldehydes and chemotherapeutics are involved in the activation of factors promoting proliferation and chemo-resistance. In this review, we have delineated the possible mechanisms by which AR modulates cellular metabolism for cancer proliferation and survival. An in-depth understanding of cancer metabolism and the role of AR might lead to the use of AR inhibitors as metabolic modulating agents for the therapy of cancer.
Collapse
Affiliation(s)
- N P Syamprasad
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research Guwahati, Sila Village, Changsari, Assam 781101, India
| | - Siddhi Jain
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research Guwahati, Sila Village, Changsari, Assam 781101, India
| | - Bishal Rajdev
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research Guwahati, Sila Village, Changsari, Assam 781101, India
| | - Neethu Prasad
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research Guwahati, Sila Village, Changsari, Assam 781101, India
| | - Ravindra Kallipalli
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research Guwahati, Sila Village, Changsari, Assam 781101, India
| | - V G M Naidu
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research Guwahati, Sila Village, Changsari, Assam 781101, India.
| |
Collapse
|
5
|
Trigonella foenum-graecum L. and Psoralea corylifolia L. Improve Erectile Dysfunction in Streptozotocin-Induced Diabetic Rats through Suppression of Oxidative Stress. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:4187359. [PMID: 35707467 PMCID: PMC9192318 DOI: 10.1155/2022/4187359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 03/23/2022] [Accepted: 04/21/2022] [Indexed: 11/26/2022]
Abstract
Background Diabetes mellitus-induced erectile dysfunction (DMED) is one of the most common complications of diabetes and is mainly attributed to oxidative stress. Hu-Lu-Ba-Wan (HLBW) is a classic Chinese formulation consisting of Trigonella foenum-graecum L. (TFG) and Psoralea corylifolia L. (PC). HLBW has been used not only for the treatment of diabetes but also for the treatment of erectile dysfunction in clinics. This study aimed to explore the efficacy and underlying mechanism of HLBW in ameliorating erectile function in streptozotocin-induced diabetic rats. Methods The diabetic model was established by tail vein injection of streptozotocin (26 mg/kg), and then DMED rats screened by the apomorphine test were randomly divided into two groups: the model group and the HLBW group. The rats in the HLBW group were administered HLBW granules daily for 12 weeks. Fasting blood glucose and fasting insulin were tested by a commercial kit. Intracavernous pressure (ICP) and mean arterial pressure (MAP) were measured by cavernous nerve electrostimulation before the rats were killed. Erectile function was evaluated with ICP/MAP. The markers of oxidative stress in the corpus cavernosum (CC) were assayed by assay kits. Apoptosis in cavernosal tissue was detected by Western blotting (WB). The expression levels of vascular endothelial marker (vWF), α-smooth muscle actin (α-SMA), endothelial nitric oxide synthase (eNOS), and NADPH oxidase subunit P47phox were determined by WB and PCR. Furthermore, the structure of the CC was further confirmed by Masson's trichrome staining. Results The results showed that HLBW significantly reduced blood glucose and increased insulin sensitivity. HLBW reduced oxidative stress and apoptosis. In addition, we observed that the expression levels of vWF, α-SMA, and eNOS as well as the ratio of smooth muscle to collagen increased in the HLBW group. Conclusions Our results demonstrated that HLBW could reduce oxidative stress damage in CC to improve diabetes mellitus-induced erectile dysfunction in rats by inhibiting NADPH oxidase.
Collapse
|
6
|
Zhou Y, Wang R, Han F, Zhang J. Efficacy of epalrestat combined with alprostadil for diabetic nephropathy and its impacts on renal fibrosis and related factors of inflammation and oxidative stress. Am J Transl Res 2022; 14:3172-3179. [PMID: 35702110 PMCID: PMC9185026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 03/30/2022] [Indexed: 06/15/2023]
Abstract
OBJECTIVE To explore the efficacy of epalrestat (Ep) combined with alprostadil (Alp) in the treatment of diabetic nephropathy (DN) and its impacts on renal fibrosis (RF) and inflammation and oxidative stress (OS)-related factors. METHODS In this retrospective study, 120 patients with DN treated in the Cangzhou Central Hospital from January 2020 to January 2021 were selected as the research subjects. Among them, 80 cases treated with Ep combined with Alp were assigned to group A, and the rest 40 patients treated with Alp only were assigned to group B. The two groups were compared with respect to the following items: serum OS indexes (malondialdehyde, MDA; superoxide dismutase, SOD; total antioxidant capacity, TAOC), inflammatory factors (tumor necrosis factor-α, TNF-α; interleukin-2, IL-2), RF index transforming growth factor-β1 (TGF-β1), urinary protein indexes (urinary albumin excretion, UAE; serum albumin, ALB), blood glucose (fasting blood glucose, FBG), fasting C-peptide, postprandial 2hC peptide levels, overall response rate (ORR) and incidence of adverse reactions. RESULTS Compared with group B, the levels of MDA, TNF-α, IL-2 and TGF-β1 were lower, while SOD and TAOC were higher in group A. In addition, ALB was higher, while UAE and FBG were lower in group A as compared with group B. Moreover, group A had a higher ORR and fewer adverse reactions as compared with group B. CONCLUSION The combined therapy of Ep and Alp is more effective in the treatment of DN. This combination can effectively reduce RF and better alleviate inflammation and OS.
Collapse
Affiliation(s)
- Yanan Zhou
- Endocrinology and Diabetes Department, Cangzhou Central HospitalCangzhou 061001, Hebei, China
| | - Rongrong Wang
- Endocrinology and Diabetes Department, Cangzhou Central HospitalCangzhou 061001, Hebei, China
| | - Fengmei Han
- Ophthalmology Department, Cangzhou Central HospitalCangzhou 061001, Hebei, China
| | - Jincheng Zhang
- Endocrinology and Diabetes Department, Cangzhou Central HospitalCangzhou 061001, Hebei, China
| |
Collapse
|
7
|
Ligezka AN, Radenkovic S, Saraswat M, Garapati K, Ranatunga W, Krzysciak W, Yanaihara H, Preston G, Brucker W, McGovern RM, Reid JM, Cassiman D, Muthusamy K, Johnsen C, Mercimek-Andrews S, Larson A, Lam C, Edmondson AC, Ghesquière B, Witters P, Raymond K, Oglesbee D, Pandey A, Perlstein EO, Kozicz T, Morava E. Sorbitol Is a Severity Biomarker for PMM2-CDG with Therapeutic Implications. Ann Neurol 2021; 90:887-900. [PMID: 34652821 DOI: 10.1002/ana.26245] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 10/07/2021] [Accepted: 10/07/2021] [Indexed: 01/27/2023]
Abstract
OBJECTIVE Epalrestat, an aldose reductase inhibitor increases phosphomannomutase (PMM) enzyme activity in a PMM2-congenital disorders of glycosylation (CDG) worm model. Epalrestat also decreases sorbitol level in diabetic neuropathy. We evaluated the genetic, biochemical, and clinical characteristics, including the Nijmegen Progression CDG Rating Scale (NPCRS), urine polyol levels and fibroblast glycoproteomics in patients with PMM2-CDG. METHODS We performed PMM enzyme measurements, multiplexed proteomics, and glycoproteomics in PMM2-deficient fibroblasts before and after epalrestat treatment. Safety and efficacy of 0.8 mg/kg/day oral epalrestat were studied in a child with PMM2-CDG for 12 months. RESULTS PMM enzyme activity increased post-epalrestat treatment. Compared with controls, 24% of glycopeptides had reduced abundance in PMM2-deficient fibroblasts, 46% of which improved upon treatment. Total protein N-glycosylation improved upon epalrestat treatment bringing overall glycosylation toward the control fibroblasts' glycosylation profile. Sorbitol levels were increased in the urine of 74% of patients with PMM2-CDG and correlated with the presence of peripheral neuropathy, and CDG severity rating scale. In the child with PMM2-CDG on epalrestat treatment, ataxia scores improved together with significant growth improvement. Urinary sorbitol levels nearly normalized in 3 months and blood transferrin glycosylation normalized in 6 months. INTERPRETATION Epalrestat improved PMM enzyme activity, N-glycosylation, and glycosylation biomarkers in vitro. Leveraging cellular glycoproteome assessment, we provided a systems-level view of treatment efficacy and discovered potential novel biosignatures of therapy response. Epalrestat was well-tolerated and led to significant clinical improvements in the first pediatric patient with PMM2-CDG treated with epalrestat. We also propose urinary sorbitol as a novel biomarker for disease severity and treatment response in future clinical trials in PMM2-CDG. ANN NEUROL 2021.
Collapse
Affiliation(s)
- Anna N Ligezka
- Department of Clinical Genomics, Mayo Clinic, Rochester, MN.,Department of Medical Diagnostics, Faculty of Pharmacy, Jagiellonian University Medical College, Krakow, Poland
| | - Silvia Radenkovic
- Department of Clinical Genomics, Mayo Clinic, Rochester, MN.,Laboratory of Hepatology, Department of CHROMETA, KU Leuven, Leuven, Belgium.,Department of Oncology, KU Leuven, Leuven, Belgium.,Metabolomics Expertise Center, VIB-KU Leuven, Leuven, Belgium
| | - Mayank Saraswat
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN.,Institute of Bioinformatics, Bangalore, India.,Manipal Academy of Higher Education (MAHE), Manipal, India
| | - Kishore Garapati
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN.,Institute of Bioinformatics, Bangalore, India.,Manipal Academy of Higher Education (MAHE), Manipal, India.,Center for Molecular Medicine, National Institute of Mental Health and Neurosciences (NIMHANS), Bangalore, India
| | | | - Wirginia Krzysciak
- Department of Medical Diagnostics, Faculty of Pharmacy, Jagiellonian University Medical College, Krakow, Poland
| | | | - Graeme Preston
- Department of Clinical Genomics, Mayo Clinic, Rochester, MN
| | - William Brucker
- Department of Pediatrics, Human Genetics, Rhode Island Hospital, Providence, RI
| | - Renee M McGovern
- Division of Oncology Research, Mayo Clinic College of Medicine, Rochester, MN
| | - Joel M Reid
- Division of Oncology Research, Mayo Clinic College of Medicine, Rochester, MN
| | - David Cassiman
- Laboratory of Hepatology, Department of CHROMETA, KU Leuven, Leuven, Belgium.,Department of Paediatrics, Metabolic Disease Center, University Hospitals Leuven, Leuven, Belgium
| | | | | | - Saadet Mercimek-Andrews
- Division of Clinical and Metabolic Genetics, The Hospital for Sick Children, Toronto, ON, Canada.,Department of Medical Genetics, University of Alberta, Stollery Children's Hospital, Alberta Health Services, Edmonton, AB, Canada
| | - Austin Larson
- Section of Clinical Genetics and Metabolism, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO
| | - Christina Lam
- Division of Genetic Medicine, Department of Pediatrics, University of Washington School of Medicine, Seattle, WA.,Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA
| | - Andrew C Edmondson
- Section of Biochemical Genetics, Division of Human Genetics, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA
| | - Bart Ghesquière
- Department of Oncology, KU Leuven, Leuven, Belgium.,Metabolomics Expertise Center, VIB-KU Leuven, Leuven, Belgium
| | - Peter Witters
- Department of Paediatrics, Metabolic Disease Center, University Hospitals Leuven, Leuven, Belgium.,Department of Development and Regeneration, Faculty of Medicine, KU Leuven, Leuven, Belgium
| | - Kimiyo Raymond
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
| | - Devin Oglesbee
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
| | - Akhilesh Pandey
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
| | | | - Tamas Kozicz
- Department of Clinical Genomics, Mayo Clinic, Rochester, MN.,Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
| | - Eva Morava
- Department of Clinical Genomics, Mayo Clinic, Rochester, MN.,Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN.,Department of Paediatrics, Metabolic Disease Center, University Hospitals Leuven, Leuven, Belgium
| |
Collapse
|
8
|
Singh M, Kapoor A, Bhatnagar A. Physiological and Pathological Roles of Aldose Reductase. Metabolites 2021; 11:655. [PMID: 34677370 PMCID: PMC8541668 DOI: 10.3390/metabo11100655] [Citation(s) in RCA: 60] [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: 08/23/2021] [Revised: 09/17/2021] [Accepted: 09/17/2021] [Indexed: 12/15/2022] Open
Abstract
Aldose reductase (AR) is an aldo-keto reductase that catalyzes the first step in the polyol pathway which converts glucose to sorbitol. Under normal glucose homeostasis the pathway represents a minor route of glucose metabolism that operates in parallel with glycolysis. However, during hyperglycemia the flux of glucose via the polyol pathway increases significantly, leading to excessive formation of sorbitol. The polyol pathway-driven accumulation of osmotically active sorbitol has been implicated in the development of secondary diabetic complications such as retinopathy, nephropathy, and neuropathy. Based on the notion that inhibition of AR could prevent these complications a range of AR inhibitors have been developed and tested; however, their clinical efficacy has been found to be marginal at best. Moreover, recent work has shown that AR participates in the detoxification of aldehydes that are derived from lipid peroxidation and their glutathione conjugates. Although in some contexts this antioxidant function of AR helps protect against tissue injury and dysfunction, the metabolic transformation of the glutathione conjugates of lipid peroxidation-derived aldehydes could also lead to the generation of reactive metabolites that can stimulate mitogenic or inflammatory signaling events. Thus, inhibition of AR could have both salutary and injurious outcomes. Nevertheless, accumulating evidence suggests that inhibition of AR could modify the effects of cardiovascular disease, asthma, neuropathy, sepsis, and cancer; therefore, additional work is required to selectively target AR inhibitors to specific disease states. Despite past challenges, we opine that a more gainful consideration of therapeutic modulation of AR activity awaits clearer identification of the specific role(s) of the AR enzyme in health and disease.
Collapse
Affiliation(s)
- Mahavir Singh
- Eye and Vision Science Laboratory, Department of Physiology, School of Medicine, University of Louisville, Louisville, KY 40202, USA
| | - Aniruddh Kapoor
- Internal Medicine—Critical Care, School of Medicine, Saint Louis University, St. Louis, MO 63141, USA;
| | - Aruni Bhatnagar
- Christina Lee Brown Envirome Institute, School of Medicine, University of Louisville, Louisville, KY 40202, USA;
| |
Collapse
|
9
|
Kousaxidis A, Petrou A, Lavrentaki V, Fesatidou M, Nicolaou I, Geronikaki A. Aldose reductase and protein tyrosine phosphatase 1B inhibitors as a promising therapeutic approach for diabetes mellitus. Eur J Med Chem 2020; 207:112742. [PMID: 32871344 DOI: 10.1016/j.ejmech.2020.112742] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 08/06/2020] [Indexed: 02/07/2023]
Abstract
Diabetes mellitus is a metabolic disease characterized by high blood glucose levels and usually associated with several chronic pathologies. Aldose reductase and protein tyrosine phosphatase 1B enzymes have identified as two novel molecular targets associated with the onset and progression of type II diabetes and related comorbidities. Although many inhibitors against these enzymes have already found in the field of diabetic mellitus, the research for discovering more effective and selective agents with optimal pharmacokinetic properties continues. In addition, dual inhibition of these target proteins has proved as a promising therapeutic approach. A variety of diverse scaffolds are presented in this review for the future design of potent and selective inhibitors of aldose reductase and protein tyrosine phosphatase 1B based on the most important structural features of both enzymes. The discovery of novel dual aldose reductase and protein tyrosine phosphatase 1B inhibitors could be effective therapeutic molecules for the treatment of insulin-resistant type II diabetes mellitus. The methods used comprise a literature survey and X-ray crystal structures derived from Protein Databank (PDB). Despite the available therapeutic options for type II diabetes mellitus, the inhibitors of aldose reductase and protein tyrosine phosphatase 1B could be two promising approaches for the effective treatment of hyperglycemia and diabetes-associated pathologies. Due to the poor pharmacokinetic profile and low in vivo efficacy of existing inhibitors of both targets, the research turned to more selective and cell-permeable agents as well as multi-target molecules.
Collapse
Affiliation(s)
- Antonios Kousaxidis
- School of Health, Department of Pharmacy, Aristotle University of Thessaloniki, 54124, Greece
| | - Anthi Petrou
- School of Health, Department of Pharmacy, Aristotle University of Thessaloniki, 54124, Greece
| | - Vasiliki Lavrentaki
- School of Health, Department of Pharmacy, Aristotle University of Thessaloniki, 54124, Greece
| | - Maria Fesatidou
- School of Health, Department of Pharmacy, Aristotle University of Thessaloniki, 54124, Greece
| | - Ioannis Nicolaou
- School of Health, Department of Pharmacy, Aristotle University of Thessaloniki, 54124, Greece
| | - Athina Geronikaki
- School of Health, Department of Pharmacy, Aristotle University of Thessaloniki, 54124, Greece.
| |
Collapse
|
10
|
Addressing selectivity issues of aldose reductase 2 inhibitors for the management of diabetic complications. Future Med Chem 2020; 12:1327-1358. [PMID: 32602375 DOI: 10.4155/fmc-2020-0032] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Aldose Reductase 2 (ALR2), the rate-limiting enzyme of the polyol pathway, plays an important role in detoxification of some toxic aldehydes. Under hyperglycemia, this enzyme overactivates and causes diabetic complications (DC). Therefore, ALR2 inhibition has been established as a potential approach to manage these complications. Several ALR2 inhibitors have been reported, but none of them could reach US FDA approval. One of the main reasons is their poor selectivity over ALR1, which leads to the toxicity. The current review underlines the molecular connectivity of ALR2 with DC and comparative analysis of the catalytic domains of ALR2 and ALR1, to better understand the selectivity issues. This report also discusses the key features required for ALR2 inhibition and to limit toxicity due to off-target activity.
Collapse
|
11
|
Oduro PK, Fang J, Niu L, Li Y, Li L, Zhao X, Wang Q. Pharmacological management of vascular endothelial dysfunction in diabetes: TCM and western medicine compared based on biomarkers and biochemical parameters. Pharmacol Res 2020; 158:104893. [PMID: 32434053 DOI: 10.1016/j.phrs.2020.104893] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 04/18/2020] [Accepted: 05/03/2020] [Indexed: 12/20/2022]
Abstract
Diabetes, a worldwide health concern while burdening significant populace of countries with time due to a hefty increase in both incidence and prevalence rates. Hyperglycemia has been buttressed both in clinical and experimental studies to modulate widespread molecular actions that effect macro and microvascular dysfunctions. Endothelial dysfunction, activation, inflammation, and endothelial barrier leakage are key factors contributing to vascular complications in diabetes, plus the development of diabetes-induced cardiovascular diseases. The recent increase in molecular, transcriptional, and clinical studies has brought a new scope to the understanding of molecular mechanisms and the therapeutic targets for endothelial dysfunction in diabetes. In this review, an attempt made to discuss up to date critical and emerging molecular signaling pathways involved in the pathophysiology of endothelial dysfunction and viable pharmacological management targets. Importantly, we exploit some Traditional Chinese Medicines (TCM)/TCM isolated bioactive compounds modulating effects on endothelial dysfunction in diabetes. Finally, clinical studies data on biomarkers and biochemical parameters involved in the assessment of the efficacy of treatment in vascular endothelial dysfunction in diabetes was compared between clinically used western hypoglycemic drugs and TCM formulas.
Collapse
Affiliation(s)
- Patrick Kwabena Oduro
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin 301617, PR China
| | - Jingmei Fang
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin 301617, PR China
| | - Lu Niu
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin 301617, PR China
| | - Yuhong Li
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin 301617, PR China; Tianjin Key Laboratory of Chinese medicine Pharmacology, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China
| | - Lin Li
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin 301617, PR China; Tianjin Key Laboratory of Chinese medicine Pharmacology, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China
| | - Xin Zhao
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin 301617, PR China; Tianjin Key Laboratory of Chinese medicine Pharmacology, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China
| | - Qilong Wang
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin 301617, PR China; Tianjin Key Laboratory of Chinese medicine Pharmacology, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China.
| |
Collapse
|
12
|
Skała E, Makowczyńska J, Wieczfinska J, Kowalczyk T, Sitarek P. Caffeoylquinic Acids with Potential Biological Activity from Plant In vitro Cultures as Alternative Sources of Valuable Natural Products. Curr Pharm Des 2020; 26:2817-2842. [PMID: 32048962 DOI: 10.2174/1381612826666200212115826] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 12/05/2019] [Indexed: 02/07/2023]
Abstract
BACKGROUND For a long time, the researchers have been looking for new efficient methods to enhance production and obtain valuable plant secondary metabolites, which would contribute to the protection of the natural environment through the preservation of various plant species, often rare and endangered. These possibilities offer plant in vitro cultures which can be performed under strictly-controlled conditions, regardless of the season or climate and environmental factors. Biotechnological methods are promising strategies for obtaining the valuable plant secondary metabolites with various classes of chemical compounds including caffeoylquinic acids (CQAs) and their derivatives. CQAs have been found in many plant species which are components in the daily diet and exhibit a wide spectrum of biological activities, including antioxidant, immunomodulatory, antihypertensive, analgesic, anti-inflammatory, hepato- and neuroprotective, anti-hyperglycemic, anticancer, antiviral and antimicrobial activities. They have also been found to offer protection against Alzheimer's disease, and play a role in weight reduction and lipid metabolism control, as well as modulating the activity of glucose-6-phosphatase involved in glucose metabolism. METHODS This work presents the review of the recent advances in use in vitro cultures of various plant species for the alternative system to the production of CQAs and their derivatives. Production of the secondary metabolites in in vitro culture is usually performed with cell suspension or organ cultures, such as shoots and adventitious or transformed roots. To achieve high production of valuable secondary metabolites in in vitro cultures, the optimization of the culture condition is necessary with respect to both biomass accumulation and metabolite content. The optimization of the culture conditions can be achieved by choosing the type of medium, growth regulators or growth conditions, selection of high-productivity lines or culture period, supplementation of the culture medium with precursors or elicitor treatments. Cultivation for large-scale in bioreactors and genetic engineering: Agrobacterium rhizogenes transformation and expression improvement of transcriptional factor or genes involved in the secondary metabolite production pathway are also efficient strategies for enhancement of the valuable secondary metabolites. RESULTS Many studies have been reported to obtain highly productive plant in vitro cultures with respect to CQAs. Among these valuable secondary metabolites, the most abundant compound accumulated in in vitro cultures was 5-CQA (chlorogenic acid). Highly productive cultures with respect to this phenolic acid were Leonurus sibiricus AtPAP1 transgenic roots, Lonicera macranthoides and Eucomia ulmoides cell suspension cultures which accumulated above 20 mg g-1 DW 5-CQA. It is known that di- and triCQAs are less common in plants than monoCQAs, but it was also possible to obtain them by biotechnological methods. CONCLUSION The results indicate that the various in vitro cultures of different plant species can be a profitable approach for the production of CQAs. In particular, an efficient production of these valuable compounds is possible by Lonicera macranthoides and Eucomia ulmoides cell suspension cultures, Leonurus sibiricus transformed roots and AtPAP1 transgenic roots, Echinacea angustifolia adventitious shoots, Rhaponticum carthamoides transformed plants, Lavandula viridis shoots, Sausera involucrata cell suspension and Cichorium intybus transformed roots.
Collapse
Affiliation(s)
- Ewa Skała
- Department of Biology and Pharmaceutical Botany, Medical University of Lodz, Muszynskiego 1, 90-151 Lodz, Poland
| | - Joanna Makowczyńska
- Department of Biology and Pharmaceutical Botany, Medical University of Lodz, Muszynskiego 1, 90-151 Lodz, Poland
| | - Joanna Wieczfinska
- Department of Immunopathology, Medical University of Lodz, Zeligowskiego 7/9, 90-752 Lodz, Poland
| | - Tomasz Kowalczyk
- Department of Molecular Biotechnology and Genetics, University of Lodz, Banacha 12/16, 90-237, Lodz, Poland
| | - Przemysław Sitarek
- Department of Biology and Pharmaceutical Botany, Medical University of Lodz, Muszynskiego 1, 90-151 Lodz, Poland
| |
Collapse
|