1
|
Kumar R, Nidhar M, Tewari AK. Design, Synthesis, in Silico and in Vitro Dipeptidyl Peptidase-4 Activity of Triazole-Based Heterocyclic Compounds. Chem Biodivers 2025; 22:e202402381. [PMID: 39718512 DOI: 10.1002/cbdv.202402381] [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: 09/21/2024] [Revised: 12/05/2024] [Accepted: 12/23/2024] [Indexed: 12/25/2024]
Abstract
1,2,3-triazole-based ring connected with pyridazine, triazine, methyl pyrazole, diphenyl pyrazole, and phthalimide moieties through propylene linker has been synthesized for antidiabetic evaluation via click chemistry. The antidiabetic evaluations have been done by molecular docking studies and in-vitro tests against the dipeptidyl peptidase-4 (DPP-4) enzyme. The molecular docking studies have revealed that compounds 22, 23, 29, and 30 showed hydrogen bonds with the DPP-4 enzyme while in-vitro tests have revealed that compound 30 has (IC50 values 12.82 nM), exhibited potent activity compared to the standard drug, sitagliptin (14.8 nM). Among the heterocyclic compounds, phthalimide and pyrazole derivatives were found more potent for DPP-4 inhibitors.
Collapse
Affiliation(s)
- Ranjeet Kumar
- Department of Chemistry, C.M.P. Degree College, University of Allahabad, Allahabad, India
| | - Manisha Nidhar
- Department of Chemistry, Institute of Science, Banaras Hindu University, Varanasi, India
| | - Ashish Kumar Tewari
- Department of Chemistry, Institute of Science, Banaras Hindu University, Varanasi, India
| |
Collapse
|
2
|
Casertano M, Esposito C, Bello I, Barile M, Izzo L, Mitidieri E, Sorrentino R, Menna M, Panza E, Imperatore C, d'Emmanuele di Villa Bianca R. The Mediterranean Sea on the Bench: Unveiling the Marine Invertebrate Sidnyum elegans as a Source of Novel Promising Therapeutic Tools Against Triple-Negative Breast Cancer. Mar Drugs 2025; 23:195. [PMID: 40422785 DOI: 10.3390/md23050195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2025] [Revised: 04/24/2025] [Accepted: 04/24/2025] [Indexed: 05/28/2025] Open
Abstract
This study aims to unveil the marine invertebrate Sidnyum elegans, a Mediterranean ascidian, as a natural resource for the early development of new treatments for triple-negative breast cancer (TNBC). Nine different fractions obtained via medium-pressure liquid chromatography (MPLC) of the butanol-soluble material of the ascidian were evaluated in proliferating MDA-MB-231 cells in a range of 10-50 µg/mL. Among them, the SEB-5 fraction was found to be the most effective in reducing cell proliferation and concomitantly inducing apoptosis, revealed via MTT assay and FACS analysis using Annexin V/PI dual staining. Furthermore, we investigated the effect of this fraction on cell cycle phases, revealing that SEB-5 can arrest the cells in the G0/G1 phase. This latter effect was then confirmed via transcriptomic analysis, showing that treatment with SEB-5 reduced the expression of cyclinB1, CDC25a, and CDK1. Finally, to evaluate the potential antimetastatic effect of SEB-5, a wound-healing assay was performed showing the ability of SEB-5 to reduce MDA-MB-231 cell migration. The chemical characterization of SEB-5 components was performed using liquid chromatography coupled with high-resolution mass spectrometry (LC-HRMS/MS) and nuclear magnetic resonance (NMR) spectroscopy. This analysis revealed the presence of a terpenoid and polyketide-like compounds, including the alkyl sulfate 1 and phosphoeleganin 2, along with three novel phosphoeleganin-related products 3-5.
Collapse
Affiliation(s)
- Marcello Casertano
- Department of Pharmacy, University of Naples Federico II, Via D. Montesano, 49, 80131 Naples, Italy
| | - Camilla Esposito
- Department of Pharmacy, University of Naples Federico II, Via D. Montesano, 49, 80131 Naples, Italy
| | - Ivana Bello
- Department of Pharmacy, University of Naples Federico II, Via D. Montesano, 49, 80131 Naples, Italy
| | - Martina Barile
- Department of Pharmacy, University of Naples Federico II, Via D. Montesano, 49, 80131 Naples, Italy
| | - Luana Izzo
- Department of Pharmacy, University of Naples Federico II, Via D. Montesano, 49, 80131 Naples, Italy
| | - Emma Mitidieri
- Department of Pharmacy, University of Naples Federico II, Via D. Montesano, 49, 80131 Naples, Italy
| | - Raffaella Sorrentino
- Department of Pharmacy, University of Naples Federico II, Via D. Montesano, 49, 80131 Naples, Italy
| | - Marialuisa Menna
- Department of Pharmacy, University of Naples Federico II, Via D. Montesano, 49, 80131 Naples, Italy
| | - Elisabetta Panza
- Department of Pharmacy, University of Naples Federico II, Via D. Montesano, 49, 80131 Naples, Italy
| | - Concetta Imperatore
- Department of Pharmacy, University of Naples Federico II, Via D. Montesano, 49, 80131 Naples, Italy
| | | |
Collapse
|
3
|
Ishtiaq M, Iqbal N, Imran A, Al-Rashida M, Rana S, Khan MA, Iqbal J, Hameed A. Sustainable approach for synthesis of new coumarin-linked Schiff bases in DABCO-based ionic liquid and their identification as aldose reductase inhibitors. Sci Rep 2025; 15:14400. [PMID: 40275001 PMCID: PMC12022153 DOI: 10.1038/s41598-025-97949-6] [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: 09/10/2024] [Accepted: 04/08/2025] [Indexed: 04/26/2025] Open
Abstract
Aldehyde reductase (ALR1) is the enzyme that speeds up the reduction of many types of aldehydes into sorbitol and D-glucose. The essential enzyme of the polyol pathway, aldose reductase (ALR2), is responsible for the development of chronic complications associated with diabetes when activated under hyperglycemic conditions. Since it is a crucial mediator for the oxidative and inflammatory signaling pathways, ALR is thought to be a target for various diseases. Many medicines are available for the treatment of ALR-associated issues but due to their long term side-effects they are not effectively used. Coumarin is a naturally occurring compound, and its derivatives are widely used in the treatment of many ailments. Therefore, in the pursuit to find potential alternate candidates as drug leads, we have prepared new coumarin-based Schiff base analogues using DABCO-C7-F ionic liquid and compared with conventional method. The sustainable approach making use of DABCO-C7-F ionic liquid, not only made the synthesis easier but also it is cost- and time-effective. The synthesized analogues were further examined for their potentials against ALR2 (IC50 = 1.61 to 11.20 µM) as well as checked selectivity via screening against ALR1 enzyme. Moreover, the molecular docking study was performed to elucidate the binding interactions of active compounds. The results showed that the synthesized compounds may have the potential to be further studied as new and selective anti-diabetic agents.
Collapse
Affiliation(s)
- Marium Ishtiaq
- Third World Center for Science and Technology, International Center for Chemical and Biological Sciences (ICCBS), University of Karachi, Karachi, 75270, Pakistan
- H.E.J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences (ICCBS), University of Karachi, Karachi, 75270, Pakistan
| | - Naved Iqbal
- Third World Center for Science and Technology, International Center for Chemical and Biological Sciences (ICCBS), University of Karachi, Karachi, 75270, Pakistan
- H.E.J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences (ICCBS), University of Karachi, Karachi, 75270, Pakistan
| | - Aqeel Imran
- Center for Advanced Drug Research, COMSATS University Islamabad, Abbottabad Campus, Abbottabad, 22060, Pakistan
- Department of Chemistry, COMSATS University Islamabad, Abbottabad Campus, Abbottabad, 22060, Pakistan
| | - Mariya Al-Rashida
- Department of Chemistry, Forman Christian College (A Chartered University), Lahore, Pakistan
| | - Sobia Rana
- Molecular Biology and Human Genetics Laboratory, Dr. Panjwani Center for Molecular Medicine and Drug Research (PCMD), International Center for Chemical and Biological Sciences (ICCBS), University of Karachi, Karachi, 75270, Pakistan
| | - Maria Aqeel Khan
- Third World Center for Science and Technology, International Center for Chemical and Biological Sciences (ICCBS), University of Karachi, Karachi, 75270, Pakistan.
- H.E.J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences (ICCBS), University of Karachi, Karachi, 75270, Pakistan.
| | - Jamshed Iqbal
- Center for Advanced Drug Research, COMSATS University Islamabad, Abbottabad Campus, Abbottabad, 22060, Pakistan
| | - Abdul Hameed
- H.E.J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences (ICCBS), University of Karachi, Karachi, 75270, Pakistan.
- Department of Chemistry, University of Sahiwal, Sahiwal, 57000, Pakistan.
| |
Collapse
|
4
|
Alvarado Salazar JA, Valdes M, Cruz A, Moreno de Jesús B, Patiño González D, Olivares Corichi IM, Tamay Cach F, Mendieta Wejebe JE. In Silico and In Vivo Evaluation of Novel 2-Aminobenzothiazole Derivative Compounds as Antidiabetic Agents. Int J Mol Sci 2025; 26:909. [PMID: 39940678 PMCID: PMC11817192 DOI: 10.3390/ijms26030909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 01/19/2025] [Accepted: 01/20/2025] [Indexed: 02/16/2025] Open
Abstract
Currently, there are several drugs used for the treatment of type 2 diabetes (T2D); however, all of them have adverse effects. Benzothiazoles have a broad spectrum of biological activities such as antidiabetic. This study aimed to evaluate in silico and in vivo two series of 2-aminobenzothiazole derivatives linked to isothioureas (3a-w) or guanidines (4a-z) for the treatment of T2D. The ADMET properties were determined in silico, from which it was possible to select nine compounds (two isothioureas and seven guanidines), and, with molecular docking, it was shown that compounds methyl (E)-N'-(benzo[d]thiazol-2-yl)-N-methylcarbamimidothioate (3b) and 2-(benzo[d]thiazol-2-yl)-1,3-di-tert-butylguanidine (4y) showed a high affinity for PPARγ (ΔG = -7.8 and -8.4 kcal/mol, respectively). In vivo, the LD50 value was estimated in rats based on OECD Guideline 425, being >1750 mg/kg for both compounds. The pharmacological effect of 3b and 4y was evaluated in the T2D rat model, showing that after oral administration in an equimolar ratio to pioglitazone (15 mg/kg) for 4 weeks, both compounds were able to reduce blood glucose levels (<200 mg/dL) and improve the lipid profile. Therefore, 3b and 4y could be used in the future as antidiabetic agents.
Collapse
Affiliation(s)
- Juan Andres Alvarado Salazar
- Carrera de Química Farmacéutica Biológica, Área Farmacéutica, Facultad de Estudios Superiores (FES)-Zaragoza, Universidad Nacional Autónoma de México, Batalla 5 de mayo s/n, Ejercito de Oriente, Iztapalapa, Mexico City 09230, Mexico;
| | - Miguel Valdes
- Laboratorio de Biofísica y Biocatálisis, Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Av. Salvador Díaz Mirón esq. Plan de San Luis s/n, Casco de Santo Tomás, Miguel Hidalgo, Mexico City 11340, Mexico; (B.M.d.J.); (D.P.G.)
- Unidad de Investigación Médica en Farmacología, UMAE Hospital de Especialidades 2° Piso CORSE Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Av. Cuauhtémoc 330, Col. Doctores, Mexico City 06720, Mexico
| | - Alejandro Cruz
- Laboratorio de Química Supramolecular y Nanociencias, Departamento de Ciencias Básicas, Unidad Profesional Interdisciplinaria de Biotecnología, Instituto Politécnico Nacional, Av. Acueducto s/n, Colonia Barrio La Laguna Ticomán, Mexico City 07340, Mexico;
| | - Brenda Moreno de Jesús
- Laboratorio de Biofísica y Biocatálisis, Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Av. Salvador Díaz Mirón esq. Plan de San Luis s/n, Casco de Santo Tomás, Miguel Hidalgo, Mexico City 11340, Mexico; (B.M.d.J.); (D.P.G.)
| | - David Patiño González
- Laboratorio de Biofísica y Biocatálisis, Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Av. Salvador Díaz Mirón esq. Plan de San Luis s/n, Casco de Santo Tomás, Miguel Hidalgo, Mexico City 11340, Mexico; (B.M.d.J.); (D.P.G.)
| | - Ivonne María Olivares Corichi
- Laboratorio de Bioquímica y Estrés Oxidante, Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Av. Salvador Díaz Mirón esq. Plan de San Luis s/n, Casco de Santo Tomás, Miguel Hidalgo, Mexico City 11340, Mexico;
| | - Feliciano Tamay Cach
- Laboratorio de Investigación de Bioquímica Aplicada, Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Av. Salvador Díaz Mirón esq. Plan de San Luis s/n, Casco de Santo Tomás, Miguel Hidalgo, Mexico City 11340, Mexico;
| | - Jessica Elena Mendieta Wejebe
- Laboratorio de Biofísica y Biocatálisis, Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Av. Salvador Díaz Mirón esq. Plan de San Luis s/n, Casco de Santo Tomás, Miguel Hidalgo, Mexico City 11340, Mexico; (B.M.d.J.); (D.P.G.)
| |
Collapse
|
5
|
Kousaxidis A, Paoli P, Kovacikova L, Genovese M, Santi A, Stefek M, Petrou A, Nicolaou I. Rational design and synthesis of novel N-benzylindole-based epalrestat analogs as selective aldose reductase inhibitors: An unexpected discovery of a new glucose-lowering agent (AK-4) acting as a mitochondrial uncoupler. Eur J Med Chem 2025; 281:117035. [PMID: 39536493 DOI: 10.1016/j.ejmech.2024.117035] [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: 09/23/2024] [Revised: 11/01/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024]
Abstract
Diabetes mellitus is one of the most frequent metabolic diseases associated with hyperglycemia. Although antidiabetic drugs reduce hyperglycemia, diabetic patients suffer from abnormal fluctuations in blood glucose levels leading to the onset of long-term complications. Aldose reductase inhibitors are considered a promising strategy for regulating the occurrence of diabetic-specific comorbidities. So far, epalrestat is the only drug being approved in Asian countries. In this paper, we ground our research in discovering novel epalrestat analogs that prevent chronic complications and normalize hyperglycemia. Herein, we describe the rational design and synthesis of four novel 4-thiazolidinone acetic acid derivatives (AK-1-4) being evaluated for their efficacy against aldose reductase from rat lenses and their specificity over the homologous enzyme from rat kidneys. AK-1-4 were also tested against human recombinant protein tyrosine phosphatase 1B as a key target in insulin sensitization and towards the closely related T-cell-derived enzyme. Docking analyses suggested possible binding modes on examined targets. The promising inhibitory profile of AK-4 sparked our interest in exploring its effect on the insulin-receptor signaling pathway and its ability to stimulate glucose uptake under ex vivo conditions. We further investigated the ability of AK-4 to target mitochondria acting as an uncoupling agent and impairing mitochondrial membrane potential. Herein, we report for the first time a new glucose-lowering agent (AK-4) that can combine alleviation for chronic diabetic complications without off-target adverse effects and antihyperglycemic efficacy through controlled mitochondrial uncoupling activity. Pharmacokinetic and toxicity studies in silico revealed optimal properties of AK-4 for oral administration without potential side effects.
Collapse
Affiliation(s)
- Antonios Kousaxidis
- Department of Pharmaceutical Chemistry, School of Pharmacy, Aristotle University of Thessaloniki, 54124, Greece
| | - Paolo Paoli
- Dipartimento di Scienze Biomediche Sperimentali e Cliniche, Sezione di Scienze Biochimiche, Università degli Studi di Firenze, Viale Morgagni 50, 50134, Firenze, Italy.
| | - Lucia Kovacikova
- Institute of Experimental Pharmacology and Toxicology, CEM, SAS, Dúbravská cesta 9, 84104, Bratislava, Slovakia
| | - Massimo Genovese
- Dipartimento di Scienze Biomediche Sperimentali e Cliniche, Sezione di Scienze Biochimiche, Università degli Studi di Firenze, Viale Morgagni 50, 50134, Firenze, Italy
| | - Alice Santi
- Dipartimento di Scienze Biomediche Sperimentali e Cliniche, Sezione di Scienze Biochimiche, Università degli Studi di Firenze, Viale Morgagni 50, 50134, Firenze, Italy
| | - Milan Stefek
- Institute of Experimental Pharmacology and Toxicology, CEM, SAS, Dúbravská cesta 9, 84104, Bratislava, Slovakia
| | - Anthi Petrou
- Department of Pharmaceutical Chemistry, School of Pharmacy, Aristotle University of Thessaloniki, 54124, Greece
| | - Ioannis Nicolaou
- Department of Pharmaceutical Chemistry, School of Pharmacy, Aristotle University of Thessaloniki, 54124, Greece.
| |
Collapse
|
6
|
Qin YJ, Zhang P, Zhang P, Li J, Yang Q, Sun JL, Liang YZ, Wang LL, Zhang LZ, Han Y. The impact of endogenous N/OFQ on DPN: Insights into lower limb blood flow regulation in rats. Neuropeptides 2025; 109:102492. [PMID: 39644710 DOI: 10.1016/j.npep.2024.102492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 11/27/2024] [Accepted: 11/27/2024] [Indexed: 12/09/2024]
Abstract
Diabetic peripheral neuropathy (DPN) is a common complication of diabetes, often accompanied by impaired vascular endothelial function in the lower limbs. This dysfunction is characterized by a reduced vasodilatory response, leading to decreased blood flow in the lower limbs and ultimately contributing to the development of diabetic peripheral neuropathy. To delve deeper into this pathological process, the study employed bioinformatics to identify and analyze genes highly active in DPN. The investigation revealed that Membrane metallo-endopeptidase (MME) was effectively mitigated by its antagonist. Male Sprague-Dawley (SD) rats served as the model to systematically explore the intrinsic connection among the nociceptible/orphanin FQ-N/OFQ receptor (N/OFQ-NOP) system, femoral artery blood flow in the lower extremities, MME, and DPN. The rats were randomized into two groups: a control group and a DPN group induced by a single intraperitoneal injection of 55 mg/kg streptozotocin (STZ), with 6 rats in each group. The findings indicated that compared to the control group, the DPN group exhibited a significant reduction in femoral artery blood flow. This was accompanied by a notable increase in serum N/OFQ concentration, heightened expression of opioid-related nociceptive protein receptor 1 (OPRL1) and MME in femoral artery tissues of the lower limbs, and an elevated sciatic nerve stimulation threshold. These results suggest that the serum N/OFQ level in DPN rats is increased, which may promote the occurrence of peripheral neuropathy by up regulating MME and reducing peripheral flow distribution.
Collapse
Affiliation(s)
- Yuan-Jing Qin
- College of Anesthesiology, Shanxi Medical University, Taiyuan 030000, China; Department of Anesthesiology, Second Hospital of Shanxi Medical University, Taiyuan 030000, China
| | - Po Zhang
- College of Anesthesiology, Shanxi Medical University, Taiyuan 030000, China; Department of Anesthesiology, Second Hospital of Shanxi Medical University, Taiyuan 030000, China
| | - Peng Zhang
- College of Anesthesiology, Shanxi Medical University, Taiyuan 030000, China; Department of Anesthesiology, Second Hospital of Shanxi Medical University, Taiyuan 030000, China
| | - Jing Li
- Department of Endocrine, Central Hospital of China Railway 12th Bureau Group, 182 Yingze Road, Taiyuan 030001, Shanxi, China
| | - Qixing Yang
- Department of Anesthesiology, Linfen People's Hospital, Linfen 041000, China
| | - Jun-Li Sun
- College of Anesthesiology, Shanxi Medical University, Taiyuan 030000, China; Department of Anesthesiology, Second Hospital of Shanxi Medical University, Taiyuan 030000, China
| | - Yu-Zhang Liang
- School of Physics, Dalian University of Technology, Dalian 116024, China
| | - Li-Li Wang
- College of Anesthesiology, Shanxi Medical University, Taiyuan 030000, China; Department of Anesthesiology, Second Hospital of Shanxi Medical University, Taiyuan 030000, China
| | - Lin-Zhong Zhang
- College of Anesthesiology, Shanxi Medical University, Taiyuan 030000, China; Department of Anesthesiology, Second Hospital of Shanxi Medical University, Taiyuan 030000, China
| | - Yi Han
- College of Anesthesiology, Shanxi Medical University, Taiyuan 030000, China; Department of Anesthesiology, Second Hospital of Shanxi Medical University, Taiyuan 030000, China.
| |
Collapse
|
7
|
Türkeş C. Aldose reductase with quinolone antibiotics interaction: In vitro and in silico approach of its relationship with diabetic complications. Arch Biochem Biophys 2024; 761:110161. [PMID: 39313142 DOI: 10.1016/j.abb.2024.110161] [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: 07/24/2024] [Revised: 09/18/2024] [Accepted: 09/20/2024] [Indexed: 09/25/2024]
Abstract
Aldose reductase (AR, EC1.1.1.21), a member of the aldo-keto reductase family, is critically implicated in the pathogenesis of chronic complications associated with diabetes mellitus, including neuropathy, nephropathy, and retinopathy. Hyperglycemia-induced AR overactivity results in intracellular sorbitol accumulation, NADPH depletion, and oxidative stress. Consequently, AR is recognized as a key mediator of oxidative and inflammatory signaling pathways involved in diverse human pathologies such as cardiovascular diseases, inflammatory disorders, and cancer. This has sparked renewed interest in developing novel AR inhibitors (ARIs) with enhanced therapeutic profiles. In this study, we evaluated the inhibitory potential of five quinolone antibiotics-gatifloxacin, lomefloxacin, nalidixic acid, norfloxacin, and sparfloxacin-as ARIs relevant to various physiological and pathological conditions. Through comprehensive in vitro and in silico analyses, we explored these antibiotics' binding interactions and affinities within the AR active site. Our findings reveal that these quinolones moderately inhibit AR at micromolar concentrations, with inhibition constants (KIs) ranging from 1.03 ± 0.13 μM to 4.12 ± 0.51 μM, compared to the reference drug epalrestat (KI of 0.85 ± 0.06 μM). The combined in vitro and in silico results underscore significant interactions between these drugs and AR, suggesting their potential as therapeutic agents against the aforementioned pathological conditions. Furthermore, these insights will aid in optimizing clinical dosing regimens and mitigating unexpected drug-drug interactions when these antibiotics are co-administered with other treatments.
Collapse
Affiliation(s)
- Cüneyt Türkeş
- Department of Biochemistry, Faculty of Pharmacy, Erzincan Binali Yıldırım University, Erzincan, 24002, Turkey.
| |
Collapse
|
8
|
Zhao H, Chen S, Lin S, Mei X. Biodegradable microspheres via orally deliver celastrol with ameliorated neuropathic pain in diabetes rats. Regen Biomater 2024; 11:rbae087. [PMID: 39055304 PMCID: PMC11272178 DOI: 10.1093/rb/rbae087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 06/25/2024] [Accepted: 07/03/2024] [Indexed: 07/27/2024] Open
Abstract
The treatment of peripheral neuropathy resulting from diabetes primarily emphasizes neurotrophic medications. However, a growing body of clinical studies indicates that neuroinflammation plays a significant role in the pathogenesis of neuropathic pain. This has spurred active exploration of treatment strategies leveraging nanomedicine for diseases, aiming for superior therapeutic outcomes. In this context, we have developed biodegradable nanoparticles made of polylactic-co-glycolic acid, loaded with triptolide (pCel), designed to alleviate somatic cell neuropathic pain induced by diabetes. Treatment with pCel notably reduced levels of reactive oxygen species and apoptosis in vitro. Furthermore, the progression of streptozotocin-induced diabetes, characterized by elevated renal function indices (blood urea nitrogen, creatinine), liver function indices (bilirubin, alkaline phosphatase) and decreased levels of albumin and globulin, was mitigated following pCel administration. Importantly, oral treatment with pCel significantly inhibited mechanical allodynia and the activation of the sciatic glial cells in diabetic rats. These findings indicate that this synthetic, biodegradable nanomedicine exhibits excellent stability, biocompatibility and catalytic activity, making it a promising and innovative approach for the management of chronic pain conditions associated with diabetic neuropathy.
Collapse
Affiliation(s)
- Haosen Zhao
- Department of Orthopaedic Rehabilitation, Third Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning Province, 121000, P. R. China
| | - Shurui Chen
- Cardiac Intensive Care Unit, Cardiovascular Hospital, Guangdong Second Provincial General Hospital, Guangzhou, Guangdong Province, 510317, P. R. China
| | - Sen Lin
- Department of Orthopaedic Rehabilitation, Third Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning Province, 121000, P. R. China
| | - Xifan Mei
- Department of Orthopaedic Rehabilitation, Third Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning Province, 121000, P. R. China
| |
Collapse
|
9
|
Alkanad M, Hani U, V AH, Ghazwani M, Haider N, Osmani RAM, M D P, Hamsalakshmi, Bhat R. Bitter yet beneficial: The dual role of dietary alkaloids in managing diabetes and enhancing cognitive function. Biofactors 2024; 50:634-673. [PMID: 38169069 DOI: 10.1002/biof.2034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2023] [Accepted: 12/11/2023] [Indexed: 01/05/2024]
Abstract
With the rising prevalence of diabetes and its association with cognitive impairment, interest in the use of dietary alkaloids and other natural products has grown significantly. Understanding how these compounds manage diabetic cognitive dysfunction (DCD) is crucial. This comprehensive review explores the etiology of DCD and the effects of alkaloids in foods and dietary supplements that have been investigated as DCD therapies. Data on how dietary alkaloids like berberine, trigonelline, caffeine, capsaicin, 1-deoxynojirimycin, nuciferine, neferine, aegeline, tetramethylpyrazine, piperine, and others regulate cognition in diabetic disorders were collected from PubMed, Research Gate, Web of Science, Science Direct, and other relevant databases. Dietary alkaloids could improve memory in behavioral models and modulate the mechanisms underlying the cognitive benefits of these compounds, including their effects on glucose metabolism, gut microbiota, vasculopathy, neuroinflammation, and oxidative stress. Evidence suggests that dietary alkaloids hold promise for improving cognition in diabetic patients and could open exciting avenues for future research in diabetes management.
Collapse
Affiliation(s)
- Maged Alkanad
- Department of Pharmacognosy, Sri Adichunchanagiri College of Pharmacy, Adichunchanagiri University, Mandya, India
| | - Umme Hani
- Department of Pharmaceutics, College of Pharmacy, King Khalid University, Abha, Saudi Arabia
| | - Annegowda H V
- Department of Pharmacognosy, Sri Adichunchanagiri College of Pharmacy, Adichunchanagiri University, Mandya, India
| | - Mohammed Ghazwani
- Department of Pharmaceutics, College of Pharmacy, King Khalid University, Abha, Saudi Arabia
| | - Nazima Haider
- Department of Pathology, College of Medicine, King Khalid University, Abha, Saudi Arabia
| | - Riyaz Ali M Osmani
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Mysuru, India
| | - Pandareesh M D
- Center for Research and Innovations, Adichunchanagiri University, BGSIT, Mandya, India
| | - Hamsalakshmi
- Department of Pharmacognosy, Cauvery College of Pharmacy, Cauvery Group of Institutions, Mysuru, India
| | - Rajeev Bhat
- ERA-Chair in Food By-Products Valorisation Technologies (VALORTECH), Estonian University of Life Sciences, Tartu, Estonia
| |
Collapse
|
10
|
Agognon AL, Casertano M, Vito A, Orso S, Cabaro S, Mormone F, Morelli C, Perruolo G, Formisano P, Menna M, Imperatore C, Oriente F. Marine-Derived Phosphoeleganin and Its Semisynthetic Derivative Decrease IL6 Levels and Improve Insulin Signaling in Human Hepatocellular Carcinoma Cells. Int J Mol Sci 2024; 25:6039. [PMID: 38892230 PMCID: PMC11173279 DOI: 10.3390/ijms25116039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 05/24/2024] [Accepted: 05/29/2024] [Indexed: 06/21/2024] Open
Abstract
Marine natural products constitute a great source of potential new antidiabetic drugs. The aim of this study was to evaluate the role of phosphoeleganin (PE), a polyketide purified from the Mediterranean ascidian Sidnyum elegans, and its derivatives PE/2 and PE/3 on insulin sensitivity in human hepatocellular carcinoma (HepG2) cells. In our experiments, insulin stimulates the phosphorylation of its receptor (INSR) and AKT by 1.5- and 3.5-fold, respectively, whereas in the presence of PE, PE/2, and PE/3, the insulin induced INSR phosphorylation is increased by 2.1-, 2-, and 1.5-fold and AKT phosphorylation by 7.1-, 6.0-, and 5.1-fold, respectively. Interestingly, PE and PE/2 have an additive effect on insulin-mediated reduction of phosphoenolpyruvate carboxykinase (PEPCK) expression. Finally, PE and PE/2, but not PE/3, decrease interleukin 6 (IL6) secretion and expression before and after palmitic acid incubation, while in the presence of high glucose (HG), only PE reduces IL6. Levels of other cytokines are not significantly affected by PE and its derivates. All these data suggest that PE and its synthetic-derived compound, PE/2, significantly decrease IL6 and improve hepatic insulin signaling. As IL6 impairs insulin action, it could be hypothesized that PE and PE/2, by inhibiting IL6, may improve the hepatic insulin pathway.
Collapse
Affiliation(s)
- Ayewa L. Agognon
- Department of Translational Medicine, Federico II University of Naples and URT “Genomic of Diabetes” of Institute of Experimental Endocrinology and Oncology, National Council of Research (CNR), Via Pansini 5, 80131 Naples, Italy; (A.L.A.); (S.O.); (S.C.); (F.M.); (C.M.); (G.P.); (P.F.)
| | - Marcello Casertano
- Department of Pharmacy, University of Naples “Federico II”, Via D. Montesano 49, 80131 Napoli, Italy; (M.C.); (A.V.); (C.I.)
| | - Alessio Vito
- Department of Pharmacy, University of Naples “Federico II”, Via D. Montesano 49, 80131 Napoli, Italy; (M.C.); (A.V.); (C.I.)
| | - Sonia Orso
- Department of Translational Medicine, Federico II University of Naples and URT “Genomic of Diabetes” of Institute of Experimental Endocrinology and Oncology, National Council of Research (CNR), Via Pansini 5, 80131 Naples, Italy; (A.L.A.); (S.O.); (S.C.); (F.M.); (C.M.); (G.P.); (P.F.)
| | - Serena Cabaro
- Department of Translational Medicine, Federico II University of Naples and URT “Genomic of Diabetes” of Institute of Experimental Endocrinology and Oncology, National Council of Research (CNR), Via Pansini 5, 80131 Naples, Italy; (A.L.A.); (S.O.); (S.C.); (F.M.); (C.M.); (G.P.); (P.F.)
| | - Federica Mormone
- Department of Translational Medicine, Federico II University of Naples and URT “Genomic of Diabetes” of Institute of Experimental Endocrinology and Oncology, National Council of Research (CNR), Via Pansini 5, 80131 Naples, Italy; (A.L.A.); (S.O.); (S.C.); (F.M.); (C.M.); (G.P.); (P.F.)
| | - Cristina Morelli
- Department of Translational Medicine, Federico II University of Naples and URT “Genomic of Diabetes” of Institute of Experimental Endocrinology and Oncology, National Council of Research (CNR), Via Pansini 5, 80131 Naples, Italy; (A.L.A.); (S.O.); (S.C.); (F.M.); (C.M.); (G.P.); (P.F.)
| | - Giuseppe Perruolo
- Department of Translational Medicine, Federico II University of Naples and URT “Genomic of Diabetes” of Institute of Experimental Endocrinology and Oncology, National Council of Research (CNR), Via Pansini 5, 80131 Naples, Italy; (A.L.A.); (S.O.); (S.C.); (F.M.); (C.M.); (G.P.); (P.F.)
| | - Pietro Formisano
- Department of Translational Medicine, Federico II University of Naples and URT “Genomic of Diabetes” of Institute of Experimental Endocrinology and Oncology, National Council of Research (CNR), Via Pansini 5, 80131 Naples, Italy; (A.L.A.); (S.O.); (S.C.); (F.M.); (C.M.); (G.P.); (P.F.)
| | - Marialuisa Menna
- Department of Pharmacy, University of Naples “Federico II”, Via D. Montesano 49, 80131 Napoli, Italy; (M.C.); (A.V.); (C.I.)
| | - Concetta Imperatore
- Department of Pharmacy, University of Naples “Federico II”, Via D. Montesano 49, 80131 Napoli, Italy; (M.C.); (A.V.); (C.I.)
| | - Francesco Oriente
- Department of Translational Medicine, Federico II University of Naples and URT “Genomic of Diabetes” of Institute of Experimental Endocrinology and Oncology, National Council of Research (CNR), Via Pansini 5, 80131 Naples, Italy; (A.L.A.); (S.O.); (S.C.); (F.M.); (C.M.); (G.P.); (P.F.)
- Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, 80131 Naples, Italy
| |
Collapse
|
11
|
Parvaiz N, Abro A, Azam SS. Three-state dynamics of zinc(II) complexes yielding significant antidiabetic targets. J Mol Graph Model 2024; 127:108665. [PMID: 38029632 DOI: 10.1016/j.jmgm.2023.108665] [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: 09/07/2023] [Revised: 10/29/2023] [Accepted: 11/01/2023] [Indexed: 12/01/2023]
Abstract
Protein Tyrosine Phosphatase 1B (PTP1B), being negative regulator of insulin signaling pathways is considered as potential medicinal target. Selective and targeted inhibitors for PTP1B can impact the therapeutic options available to cure chronic illness such as diabetes. Significant research evidence including computational studies on the role of Zn2+ in binding and inhibiting the catalytic pocket have been reported along with experimental exploration of zinc(II) complexes as potent inhibitors of the enzyme. The current study has employed advanced computational methods to explore the binding and conformational orientation of zinc(II) complexes in the active site of apoenzyme, phosphoenzyme, and TSA 2 of PTP1B. Metal ion modeling was performed for zinc metal center (Zn-OOOO) utilizing a Python based Metal Center Parameter Builder (MCPB.py). The findings of the study suggest that zinc(II) complex binds to structurally and functionally important residues in open and closed conformation as well as in the phosphorylated state of the enzyme. It was observed that when the catalytic cysteine is phosphorylated in a closed conformation, the zinc(II) complex forms significant interactions with PHE182, VAL184, GLY183, and PRO180 while pushing away Q-loop GLN262 which is crucial for the hydrolysis of phosphoenzyme. Subsequently, the reported inhibitor has also demonstrated its potential to function as allosteric modulator of the enzyme occupying catalytic WPD loop residues. The study uncovers putative binding sites of zinc-containing drugs and gives insight into the size and design of such compounds which keeps them accessible and anchored in the vicinity of active site residues. Reported inhibitor offers enhanced selectivity and inhibition in all three states of the enzyme in contrast to zinc ions which can only impede enzyme in the phosphorylated state. In addition to this, investigation of ASP265→GLU265 mutation reveals the role of GLU265 in affecting the flexibility of WPD loop residues highlighting it as loss-of-function mutation. Our results hints towards a metallodrug approach that builds on the research evidence of inhibition effects of Zn2+ in the binding pocket of PTP1B. The findings presented are noteworthy, not just due to their significant relevance for clinical application, but also for the design and synthesis of novel zinc(II) complexes.
Collapse
Affiliation(s)
- Nousheen Parvaiz
- Computational Biology Lab, National Center for Bioinformatics, Quaid-i-Azam University, Islamabad, 45320, Islamabad, Pakistan
| | - Asma Abro
- Balochistan University of Information Technology Engineering and Management Sciences, Quetta, Pakistan.
| | - Syed Sikander Azam
- Computational Biology Lab, National Center for Bioinformatics, Quaid-i-Azam University, Islamabad, 45320, Islamabad, Pakistan.
| |
Collapse
|
12
|
Lv Y, Yao X, Li X, Ouyang Y, Fan C, Qian Y. Cell metabolism pathways involved in the pathophysiological changes of diabetic peripheral neuropathy. Neural Regen Res 2024; 19:598-605. [PMID: 37721290 PMCID: PMC10581560 DOI: 10.4103/1673-5374.380872] [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: 01/11/2023] [Revised: 03/31/2023] [Accepted: 06/08/2023] [Indexed: 09/19/2023] Open
Abstract
Diabetic peripheral neuropathy is a common complication of diabetes mellitus. Elucidating the pathophysiological metabolic mechanism impels the generation of ideal therapies. However, existing limited treatments for diabetic peripheral neuropathy expose the urgent need for cell metabolism research. Given the lack of comprehensive understanding of energy metabolism changes and related signaling pathways in diabetic peripheral neuropathy, it is essential to explore energy changes and metabolic changes in diabetic peripheral neuropathy to develop suitable treatment methods. This review summarizes the pathophysiological mechanism of diabetic peripheral neuropathy from the perspective of cellular metabolism and the specific interventions for different metabolic pathways to develop effective treatment methods. Various metabolic mechanisms (e.g., polyol, hexosamine, protein kinase C pathway) are associated with diabetic peripheral neuropathy, and researchers are looking for more effective treatments through these pathways.
Collapse
Affiliation(s)
- Yaowei Lv
- Department of Orthopedics, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Shanghai, China
| | - Xiangyun Yao
- Department of Orthopedics, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Shanghai, China
- Youth Science and Technology Innovation Studio of Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiao Li
- Department of Orthopedics, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Shanghai, China
- College of Fisheries and Life Science, Shanghai Ocean University, Shanghai, China
| | - Yuanming Ouyang
- Department of Orthopedics, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Shanghai, China
- Youth Science and Technology Innovation Studio of Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Cunyi Fan
- Department of Orthopedics, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Shanghai, China
- Youth Science and Technology Innovation Studio of Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yun Qian
- Department of Orthopedics, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Shanghai, China
- Youth Science and Technology Innovation Studio of Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
13
|
Jin R, Wang J, Li M, Tang T, Feng Y, Zhou S, Xie H, Feng H, Guo J, Fu R, Liu J, Tang Y, Shi Y, Guo H, Wang Y, Nie F, Li J. Discovery of a Novel Benzothiadiazine-Based Selective Aldose Reductase Inhibitor as Potential Therapy for Diabetic Peripheral Neuropathy. Diabetes 2024; 73:497-510. [PMID: 38127948 DOI: 10.2337/db23-0006] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 12/13/2023] [Indexed: 12/23/2023]
Abstract
Aldose reductase 2 (ALR2), an activated enzyme in the polyol pathway by hyperglycemia, has long been recognized as one of the most promising targets for complications of diabetes, especially in diabetic peripheral neuropathy (DPN). However, many of the ALR2 inhibitors have shown serious side effects due to poor selectivity over aldehyde reductase (ALR1). Herein, we describe the discovery of a series of benzothiadiazine acetic acid derivatives as potent and selective inhibitors against ALR2 and evaluation of their anti-DPN activities in vivo. Compound 15c, carrying a carbonyl group at the 3-position of the thiadiazine ring, showed high potent inhibition against ALR2 (IC50 = 33.19 nmol/L) and ∼16,109-fold selectivity for ALR2 over ALR1. Cytotoxicity assays ensured the primary biosafety of 15c. Further pharmacokinetic assay in rats indicated that 15c had a good pharmacokinetic feature (t1/2 = 5.60 h, area under the plasma concentration time curve [AUC(0-t)] = 598.57 ± 216.5 μg/mL * h), which was superior to epalrestat (t1/2 = 2.23 h, AUC[0-t] = 20.43 ± 3.7 μg/mL * h). Finally, in a streptozotocin-induced diabetic rat model, 15c significantly increased the nerve conduction velocities of impaired sensory and motor nerves, achieved potent inhibition of d-sorbitol production in the sciatic nerves, and significantly increased the paw withdrawal mechanical threshold. By combining the above investigations, we propose that 15c might represent a promising lead compound for the discovery of an antidiabetic peripheral neuropathy drug.
Collapse
Affiliation(s)
- Ruyi Jin
- Shaanxi Key Lab Basic & New Herbal Medicament Research Center, College of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang, China
- Shenzhen Neptunus Pharmaceutical Research Institute Co., Ltd., Shenzhen, China
| | - Jin Wang
- Shenzhen Neptunus Pharmaceutical Research Institute Co., Ltd., Shenzhen, China
- Shenzhen Huahong Marine Biomedicine Co. Ltd., Shenzhen, China
| | - Mingyue Li
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Nankai University, Tianjin, China
| | - Tian Tang
- Shenzhen Neptunus Pharmaceutical Research Institute Co., Ltd., Shenzhen, China
- Cali Biosciences, Shenzhen, China
| | - Yidong Feng
- Shenzhen Neptunus Pharmaceutical Research Institute Co., Ltd., Shenzhen, China
| | - Sha Zhou
- State Key Laboratory of Elemento-Organic Chemistry, Institute of Elemento-Organic Chemistry, Collaborative Innovation Center of Chemical Science and Engineering, College of Chemistry, Nankai University, Tianjin, China
| | - Honglei Xie
- School of Pharmacy, Shandong First Medical University and Shandong Academy of Medical Sciences, Yantai, China
| | - Haiyu Feng
- Shaanxi Key Lab Basic & New Herbal Medicament Research Center, College of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Jianshuang Guo
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Nankai University, Tianjin, China
| | - Ruijia Fu
- Shaanxi Key Lab Basic & New Herbal Medicament Research Center, College of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Jiping Liu
- Shaanxi Key Lab Basic & New Herbal Medicament Research Center, College of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Yuping Tang
- Shaanxi Key Lab Basic & New Herbal Medicament Research Center, College of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Yajun Shi
- Shaanxi Key Lab Basic & New Herbal Medicament Research Center, College of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Hui Guo
- Shaanxi Key Lab Basic & New Herbal Medicament Research Center, College of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Yuwei Wang
- Shaanxi Key Lab Basic & New Herbal Medicament Research Center, College of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Fayi Nie
- Shaanxi Key Laboratory of Acupuncture and Medicine, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Jing Li
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Nankai University, Tianjin, China
- State Key Laboratory of Elemento-Organic Chemistry, Institute of Elemento-Organic Chemistry, Collaborative Innovation Center of Chemical Science and Engineering, College of Chemistry, Nankai University, Tianjin, China
| |
Collapse
|
14
|
Feng B, Zhang J, Liu Z, Xu Y, Hu H. Discovery and biological evaluation of novel dual PTP1B and ACP1 inhibitors for the treatment of insulin resistance. Bioorg Med Chem 2024; 97:117545. [PMID: 38070352 DOI: 10.1016/j.bmc.2023.117545] [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: 10/06/2023] [Revised: 11/20/2023] [Accepted: 12/04/2023] [Indexed: 12/30/2023]
Abstract
In this study, a virtual screening pipeline comprising ligand-based and structure-based approaches was established and applied for the identification of dual PTP1B and ACP1 inhibitors. As a result, a series of benzoic acid derivatives was discovered, and compound H3 and S6 demonstrated PTP1B and ACP1 inhibitory activity, with IC50 values of 3.5 and 8.2 μM for PTP1B, and 2.5 and 5.2 μM for ACP1, respectively. Molecular dynamics simulations illustrated that H3 interacted with critical residues in the active site, such as Cys215 and Arg221 for PTP1B, and Cys17 and Arg18 for ACP1. Enzymatic kinetic research indicated that identified inhibitors competitively inhibited PTP1B and ACP1. Additionally, cellular assays demonstrated that H3 and S6 effectively increased glucose uptake in insulin-resistant HepG2 cells while displaying very limited cytotoxicity at their effective concentrations. In summary, H3 and S6 represent novel dual-target inhibitors for PTP1B and ACP1, warranting further investigation as potential agents for the treatment of diabetes.
Collapse
Affiliation(s)
- Bo Feng
- Department of Pharmacy, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
| | - Jie Zhang
- Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Zhen Liu
- Department of Neurology, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
| | - Yuan Xu
- Department of Pharmacy, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China.
| | - Huabin Hu
- Centre for Cancer Drug Discovery, Division of Cancer Therapeutics, The Institute of Cancer Research, London, UK; Science for Life Laboratory, Department of Cell and Molecular Biology, Uppsala University, BMC, Box 596, SE-751 24 Uppsala, Sweden.
| |
Collapse
|
15
|
Zhao WL, Xu D, Wang JS. Torachrysone-8-O-β-d-glucoside mediates anti-inflammatory effects by blocking aldose reductase-catalyzed metabolism of lipid peroxidation products. Biochem Pharmacol 2023; 218:115931. [PMID: 37981172 DOI: 10.1016/j.bcp.2023.115931] [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: 10/12/2023] [Revised: 11/14/2023] [Accepted: 11/15/2023] [Indexed: 11/21/2023]
Abstract
Aldose reductase (AR) is an important enzyme involved in the reduction of various aldehyde and carbonyl compounds, including the highly reactive and toxic 4-hydroxynonenal (4-HNE), which has been linked to the progression of various pathologies such as atherosclerosis, hyperglycemia, inflammation, and tumors. AR inhibitors have potential therapeutic benefits for these diseases by reducing lipid peroxidation and mitigating the harmful effects of reactive aldehydes. In this study, we found that torachrysone-8-O-β-d-glucoside (TG), a natural product isolated from Polygonum multiflorum Thunb., functions as an effective inhibitor of AR, exhibiting potent effects in clearing reactive aldehydes and reducing inflammation. TG up-regulated the mRNA levels of several antioxidant factors downstream of NRF2, especially glutathione S-transferase (GST), which is significantly increased, thus detoxifying 4-HNE by facilitating the conjugation of 4-HNE to glutathione, forming glutathione-4-hydroxynonenal (GS-HNE). By employing a combination of molecular docking, cellular thermal shift assay, and enzyme activity experiments, we demonstrated that TG exhibited strong binding affinity with AR and inhibited its activity and blocked the conversion of GS-HNE to glutathionyl-1,4-dihydroxynonene (GS-DHN), thereby preventing the formation of protein adducts and inducing severe cellular damage. This study provides novel insights into the anti-inflammatory mechanisms of AR inhibitors and offers potential avenues for developing therapeutic strategies for AR-related pathologies. Our findings suggest that TG, as an AR inhibitor, may hold promise as a therapeutic agent for treating conditions characterized by excessive lipid peroxidation and inflammation. Further investigations are needed to fully explore the clinical potential of TG and evaluate its efficacy in the treatment and management of these complex diseases.
Collapse
Affiliation(s)
- Wen-Long Zhao
- Center for Molecular Metabolism, School of Environmental and Biological Engineering, Nanjing University of Science and Technology, 200 Xiao Ling Wei Street, Nanjing 210094, People's Republic of China
| | - Di Xu
- Center for Molecular Metabolism, School of Environmental and Biological Engineering, Nanjing University of Science and Technology, 200 Xiao Ling Wei Street, Nanjing 210094, People's Republic of China
| | - Jun-Song Wang
- Center for Molecular Metabolism, School of Environmental and Biological Engineering, Nanjing University of Science and Technology, 200 Xiao Ling Wei Street, Nanjing 210094, People's Republic of China.
| |
Collapse
|
16
|
Hamdi A, Yaseen M, Ewes WA, Bhat MA, Ziedan NI, El-Shafey HW, Mohamed AAB, Elnagar MR, Haikal A, Othman DIA, Elgazar AA, Abusabaa AHA, Abdelrahman KS, Soltan OM, Elbadawi MM. Development of new thiazolidine-2,4-dione hybrids as aldose reductase inhibitors endowed with antihyperglycaemic activity: design, synthesis, biological investigations, and in silico insights. J Enzyme Inhib Med Chem 2023; 38:2231170. [PMID: 37470409 PMCID: PMC10361003 DOI: 10.1080/14756366.2023.2231170] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 06/23/2023] [Accepted: 06/25/2023] [Indexed: 07/21/2023] Open
Abstract
This research study describes the development of new small molecules based on 2,4-thiazolidinedione (2,4-TZD) and their aldose reductase (AR) inhibitory activities. The synthesis of 17 new derivatives of 2,4-TZDs hybrids was feasible by incorporating two known bioactive scaffolds, benzothiazole heterocycle, and nitro phenacyl moiety. The most active hybrid (8b) was found to inhibit AR in a non-competitive manner (0.16 µM), as confirmed by kinetic studies and molecular docking simulations. Furthermore, the in vivo experiments demonstrated that compound 8b had a significant hypoglycaemic effect in mice with hyperglycaemia induced by streptozotocin. Fifty milligrams per kilogram dose of 8b produced a marked decrease in blood glucose concentration, and a lower dose of 5 mg/kg demonstrated a noticeable antihyperglycaemic effect. These outcomes suggested that compound 8b may be used as a promising therapeutic agent for the treatment of diabetic complications.
Collapse
Affiliation(s)
- Abdelrahman Hamdi
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| | - Muhammad Yaseen
- Institute of Chemical Sciences, University of Swat, Swat, Pakistan
| | - Wafaa A Ewes
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| | - Mashooq Ahmad Bhat
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Noha I Ziedan
- Department of physical, mathematical and Engineering science, Faculty of science, Business and Enterprise, University of Chester, Chester, UK
| | - Hamed W El-Shafey
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| | - Ahmed A B Mohamed
- Department of Medicinal Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| | - Mohamed R Elnagar
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Al-Azhar University, Cairo, Egypt
- Department of Pharmacology, College of Pharmacy, The Islamic University, Najaf, Iraq
| | - Abdullah Haikal
- Department of Pharmacognosy, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| | - Dina I A Othman
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| | - Abdullah A Elgazar
- Department of Pharmacognosy, Faculty of Pharmacy, Kafrelsheikh University, Kafrelsheikh, Egypt
| | - Ahmed H A Abusabaa
- Department of Organic and Medicinal Chemistry, Faculty of Pharmacy, Fayoum University, Fayoum, Egypt
| | - Kamal S Abdelrahman
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Al-Azhar University, Assiut, Egypt
| | - Osama M Soltan
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Al-Azhar University, Assiut, Egypt
| | - Mostafa M Elbadawi
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Kafrelsheikh University, Kafrelsheikh, Egypt
| |
Collapse
|
17
|
Lin P, Niu Y. Inhibitory selectivity to the AKR1B10 and aldose reductase (AR): insight from molecular dynamics simulations and free energy calculations. RSC Adv 2023; 13:26709-26718. [PMID: 37681045 PMCID: PMC10480703 DOI: 10.1039/d3ra02215c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 06/23/2023] [Indexed: 09/09/2023] Open
Abstract
AKR1B10 is over-expressed in many cancer types and is related to chemotherapy resistance, which makes AKR1B10 a potential anti-cancer target. The high similarity of the protein structure between AKR1B10 and AR makes it difficult to develop highly selective inhibitors against AKR1B10. Understanding the interaction between AKR1B10 and inhibitors is very important for designing selective inhibitors of AKR1B10. In this study, Fidarestat, Zopolrestat, MK184 and MK204 bound to AKR1B10 and AR were used to investigate the selectivity mechanism. The results of MM/PBSA calculations show that van der Waals and electrostatic interaction provide the main contributions of the binding free energy. The hydrogen bonding between residues Y49 and H111 and inhibitors plays a pivotal role in contributing to the high inhibitory activity of AKR1B10 inhibitors. The π-π stacking interaction between residue W112 and inhibitor also plays a key role in the stability of inhibitors and AKR1B10, but W112 should keep its natural conformation to stabilize the inhibitor-AKR1B10 complex. Highly selective AKR1B10 inhibitors should have a bulky moiety like a phenyl group, which can change its binding with ABP in binding with AR and cannot change its binding with AKR1B10. The free energy decomposition shows that residues W21, V48, Y49, K78, W80, H111, R298 and V302 are beneficial to the stability of the inhibitor-AKR1B10. Our work will provide an important in silico basis for researchers to develop highly selective inhibitors of AKR1B10.
Collapse
Affiliation(s)
- Ping Lin
- Weifang University of Science and Technology Weifang 262700 China
- Institute of Modern Physics, Chinese Academy of Science Lanzhou 730000 China
| | - Yuzhen Niu
- Weifang University of Science and Technology Weifang 262700 China
- Shandong Engineering Research Center of Green and High-value Marine Fine Chemical, Weifang University of Science and Technology Weifang 262700 China
| |
Collapse
|
18
|
Wang H, Chen L, Yang B, Du J, Chen L, Li Y, Guo F. Structures, Sources, Identification/Quantification Methods, Health Benefits, Bioaccessibility, and Products of Isorhamnetin Glycosides as Phytonutrients. Nutrients 2023; 15:nu15081947. [PMID: 37111165 PMCID: PMC10143801 DOI: 10.3390/nu15081947] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 04/12/2023] [Accepted: 04/13/2023] [Indexed: 04/29/2023] Open
Abstract
In recent years, people have tended to consume phytonutrients and nutrients in their daily diets. Isorhamnetin glycosides (IGs) are an essential class of flavonoids derived from dietary and medicinal plants such as Opuntia ficus-indica, Hippophae rhamnoides, and Ginkgo biloba. This review summarizes the structures, sources, quantitative and qualitative analysis technologies, health benefits, bioaccessibility, and marketed products of IGs. Routine and innovative assay methods, such as IR, TLC, NMR, UV, MS, HPLC, UPLC, and HSCCC, have been widely used for the characterization and quantification of IGs. All of the therapeutic effects of IGs discovered to date are collected and discussed in this study, with an emphasis on the relevant mechanisms of their health-promoting effects. IGs exhibit diverse biological activities against cancer, diabetes, hepatic diseases, obesity, and thrombosis. They exert therapeutic effects through multiple networks of underlying molecular signaling pathways. Owing to these benefits, IGs could be utilized to make foods and functional foods. IGs exhibit higher bioaccessibility and plasma concentrations and longer average residence time in blood than aglycones. Overall, IGs as phytonutrients are very promising and have excellent application potential.
Collapse
Affiliation(s)
- Hong Wang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Lijia Chen
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Binrui Yang
- Nutrition Science, Amway (Shanghai) Innovation & Science Co., Ltd., Shanghai 201203, China
| | - Jun Du
- Nutrition Science, Amway (Shanghai) Innovation & Science Co., Ltd., Shanghai 201203, China
| | - Liang Chen
- Nutrition Science, Amway (Shanghai) Innovation & Science Co., Ltd., Shanghai 201203, China
| | - Yiming Li
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Fujiang Guo
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| |
Collapse
|
19
|
Chaurasyia A, Chawla P, Monga V, Singh G. Rhodanine derivatives: An insight into the synthetic and medicinal perspectives as antimicrobial and antiviral agents. Chem Biol Drug Des 2023; 101:500-549. [PMID: 36447391 DOI: 10.1111/cbdd.14163] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 10/18/2022] [Accepted: 10/22/2022] [Indexed: 12/02/2022]
Abstract
Rhodanine or 2-Thioxothiazolidin-4-one is a privileged heterocyclic compound offering a wide opportunity for structural modification, lead development, and modification. It is one of the highly decorated scaffolds in the drug discovery process. Rhodanine derivatives possess a plethora of biological activities due to their ability to interact with a diverse range of protein targets, which provide tremendous opportunities to discover new drugs with different modes of action. The most common strategy for developing novel rhodanine derivatives is the introduction of structurally diverse substituents at the C-5 or N-3, or both positions. Since the inception of Epralestat into the market in 1992, the exploration of rhodanine-3-acetic acids has led to the development of novel leads against different biological targets such as MRSA, HHV-6, Mycobacterial tuberculosis, dengue, etc. In the current pandemic era, some rhodanine compounds have been explored against SARS-CoV-2. In recent years, rhodanine and its derivatives have witnessed significant progress in developing new drug leads as potential antimicrobial and antiviral agents. Different synthetic methodologies and recent developments in the medicinal chemistry of rhodanine derivatives, including biological activities, their mechanistic aspects, structure-activity relationships, and in silico findings, have been compiled in the present review. This article will benefit the scientific community and offer perspectives on how these scaffolds as privileged structures might be exploited in the future for rational design and discovery of rhodanine-based bio-active molecules.
Collapse
Affiliation(s)
- Abhishek Chaurasyia
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, Moga, Punjab, India
| | - Pooja Chawla
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, Moga, Punjab, India
| | - Vikramdeep Monga
- Department of Pharmaceutical Sciences and Natural Products, Central University of Punjab, Bathinda, India
| | - Gurpreet Singh
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, Moga, Punjab, India.,Research Scholar, IK Gujral Punjab Technical University, Kapurthala, Punjab, India
| |
Collapse
|
20
|
Lin J, Xiao D, Lu L, Liang B, Xiong Z, Xu X. New β-carboline derivatives as potential α-glucosidase inhibitor: Synthesis and biological activity evaluation. J Mol Struct 2023. [DOI: 10.1016/j.molstruc.2023.135279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/09/2023]
|
21
|
Ishibashi F, Zha S, Kondo T, Sakamoto M, Ueno M, Fukuda T. Synthesis and structure-activity relationship study of aldose reductase inhibiting marine alkaloid lukianol A and its derivatives. Biosci Biotechnol Biochem 2023; 87:148-157. [PMID: 36441013 DOI: 10.1093/bbb/zbac193] [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: 10/26/2022] [Accepted: 11/21/2022] [Indexed: 11/30/2022]
Abstract
Lukianol A (1a) and its six derivatives 1b-1g, in which each hydroxyl groups of 1a was individually modified, were synthesized via the common intermediate 7a, which was obtained by condensation of the styryl carbazate 10 with p-hydroxyphenylpyruvic acid and subsequent [3,3]-sigmatropic rearrangement. The synthesized lukianol derivatives were evaluated for their ability to inhibit human aldose reductase. 4'-O-methyl (1b) and 4'-dehydroxy (1g) derivatives showed the same level of inhibitory activity as 1a (IC50 2.2 µm), indicating that the 4'-OH is irrelevant for the activity. In contrast, methylation of the hydroxyl group at the 4″'-position (1d) resulted in the loss of activity at a concentration of 10 µm, and masking the hydroxyl group at the 4″-position (1e) caused a 9-fold decrease in activity compared with that of 1b, suggesting that the 4″-OH is an essential group, and the 4″'-OH is required for higher activity.
Collapse
Affiliation(s)
- Fumito Ishibashi
- Graduate School of Fisheries and Environmental Sciences, Nagasaki University, 1-14 Bunkyo-Machi, Nagasaki, Japan.,Faculty of Fisheries, Nagasaki University, 1-14 Bunkyo-Machi, Nagasaki, Japan
| | - Shijiao Zha
- School of Earth and Environment, Anhui University of Science and Technology, Huainan, China
| | - Taiyo Kondo
- Faculty of Fisheries, Nagasaki University, 1-14 Bunkyo-Machi, Nagasaki, Japan
| | - Mayu Sakamoto
- Faculty of Fisheries, Nagasaki University, 1-14 Bunkyo-Machi, Nagasaki, Japan
| | - Mikinori Ueno
- Graduate School of Fisheries and Environmental Sciences, Nagasaki University, 1-14 Bunkyo-Machi, Nagasaki, Japan.,Faculty of Fisheries, Nagasaki University, 1-14 Bunkyo-Machi, Nagasaki, Japan
| | - Tsutomu Fukuda
- Environmental Protection Center, Nagasaki University, 1-14 Bunkyo-Machi, Nagasaki, Japan
| |
Collapse
|
22
|
Altıntop MD, Demir Y, Türkeş C, Öztürk RB, Cantürk Z, Beydemir Ş, Özdemir A. A new series of hydrazones as small-molecule aldose reductase inhibitors. Arch Pharm (Weinheim) 2023; 356:e2200570. [PMID: 36603162 DOI: 10.1002/ardp.202200570] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 12/08/2022] [Accepted: 12/12/2022] [Indexed: 01/07/2023]
Abstract
In the search for small-molecule aldose reductase (AR) inhibitors, new tetrazole-hydrazone hybrids (1-15) were designed. An efficient procedure was employed for the synthesis of compounds 1-15. All hydrazones were subjected to an in vitro assay to assess their AR inhibitory profiles. Compounds 1-15 caused AR inhibition with Ki values ranging between 0.177 and 6.322 µM and IC50 values ranging between 0.210 and 0.676 µM. 2-[(1-(4-Hydroxyphenyl)-1H-tetrazol-5-yl)thio]-N'-(4-fluorobenzylidene)acetohydrazide (4) was the most potent inhibitor of AR in this series. Compound 4 markedly inhibited AR (IC50 = 0.297 µM) in a competitive manner (Ki = 0.177 µM) compared to epalrestat (Ki = 0.857 µM, IC50 = 0.267 µM). Based on the in vitro data obtained by applying the MTT test, compound 4 showed no cytotoxic activity toward normal (NIH/3T3) cells at the tested concentrations, indicating its safety as an AR inhibitor. Compound 4 exhibited proper interactions with crucial amino acid residues within the active site of AR. In silico QikProp data of all hydrazones (1-15) were also determined to assess their pharmacokinetic profiles. Taken together, compound 4 stands out as a promising inhibitor of AR for further in vivo studies.
Collapse
Affiliation(s)
- Mehlika D Altıntop
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Anadolu University, Eskişehir, Turkey
| | - Yeliz Demir
- Department of Pharmacy Services, Nihat Delibalta Göle Vocational High School, Ardahan University, Ardahan, Turkey
| | - Cüneyt Türkeş
- Department of Biochemistry, Faculty of Pharmacy, Erzincan Binali Yıldırım University, Erzincan, Turkey
| | - Remzi B Öztürk
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Anadolu University, Eskişehir, Turkey
| | - Zerrin Cantürk
- Department of Pharmaceutical Microbiology, Faculty of Pharmacy, Anadolu University, Eskişehir, Turkey
| | - Şükrü Beydemir
- Department of Biochemistry, Faculty of Pharmacy, Anadolu University, Eskişehir, Turkey.,The Rectorate of Bilecik Şeyh Edebali University, Bilecik, Turkey
| | - Ahmet Özdemir
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Anadolu University, Eskişehir, Turkey
| |
Collapse
|
23
|
Feng B, Dong X, Liu Z, Zhang J, Liu H, Xu Y. Virtual Screening and Biological Evaluation of Novel Low Molecular Weight Protein Tyrosine Phosphatase Inhibitor for the Treatment of Insulin Resistance. Drug Des Devel Ther 2023; 17:1191-1201. [PMID: 37113468 PMCID: PMC10128076 DOI: 10.2147/dddt.s406956] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 04/07/2023] [Indexed: 04/29/2023] Open
Abstract
Purpose Protein tyrosine phosphatases (PTPs) play an essential way in diseases including cancer, obesity, diabetes and autoimmune disorders. As a member of PTPs, low molecular weight PTP (LMPTP) has been a well-recognized anti-insulin resistance target in obesity. However, the number of reported LMPTP inhibitors is limited. Our research aims to discover a novel LMPTP inhibitor and evaluate its biological activity against insulin resistance. Methods A virtual screening pipeline based on the X-ray co-crystal complex of LMPTP was constructed. Enzyme inhibition assay and cellular bioassay were used to evaluate the activity of screened compounds. Results The screening pipeline rendered 15 potential hits from Specs chemical library. Enzyme inhibition assay identified compound F9 (AN-465/41163730) as a potential LMPTP inhibitor with a K i value of 21.5 ± 7.3 μM. Cellular bioassay showed F9 could effectively increase the glucose consumption of HepG2 cells as a result of releasing insulin resistance by regulating PI3K-Akt pathway. Conclusion In summary, this study presents a versatile virtual screening pipeline for potential LMPTP inhibitor discovery and provides a novel-scaffold lead compound that is worthy of further modification to get more potent LMPTP inhibitors.
Collapse
Affiliation(s)
- Bo Feng
- Department of Pharmacy, The Affiliated Hospital of Yangzhou University, Yangzhou, People’s Republic of China
| | - Xu Dong
- Department of Pharmacy, The Affiliated Hospital of Yangzhou University, Yangzhou, People’s Republic of China
| | - Zhen Liu
- Department of Neurology, The Affiliated Hospital of Yangzhou University, Yangzhou, People’s Republic of China
| | - Jie Zhang
- Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, People’s Republic of China
| | - Hongyu Liu
- Department of Pharmacy, The Affiliated Hospital of Yangzhou University, Yangzhou, People’s Republic of China
- Correspondence: Hongyu Liu; Yuan Xu, Department of Pharmacy, The Affiliated Hospital of Yangzhou University, Yangzhou, People’s Republic of China, Email ;
| | - Yuan Xu
- Department of Pharmacy, The Affiliated Hospital of Yangzhou University, Yangzhou, People’s Republic of China
| |
Collapse
|
24
|
Hu CM, Zheng YY, Lin AT, Zhang X, Wu XZ, Lin J, Xu XT, Xiong Z. Design, synthesis and evaluation of indole-based bisacylhydrazone derivatives as α-glucosidase inhibitors. J Mol Struct 2023. [DOI: 10.1016/j.molstruc.2022.134124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
|
25
|
Morikawa T. Pharmaceutical Food Science: Search for Bio-Functional Molecules Obtained from Natural Resources to Prevent and Ameliorate Lifestyle Diseases. Chem Pharm Bull (Tokyo) 2023; 71:756-765. [PMID: 37779077 DOI: 10.1248/cpb.c23-00518] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/03/2023]
Abstract
In this review, our resent pharmaceutical food science research for bio-functional molecules obtained from natural resources that contribute to i) suppression of postprandial blood glucose elevation and/or improvement of glucose tolerance and ii) reduction of visceral fat accumulation and improvement of lipid metabolism were summarized. Based on studies using MONOTORI science, salacinol (1), neokotalanol (4), and trans-tiliroside (20) have been approved or notified by the Consumer Affairs Agency in Japan as functional substances in food with health claims, Food for Specified Health Use and Food with Functional Claims.
Collapse
Affiliation(s)
- Toshio Morikawa
- Pharmaceutical Research and Technology Institute, Kindai University
- Antiaging Center, Kindai University
| |
Collapse
|
26
|
A molecular hybridization approach for the design of selective aldose reductase (ALR2) inhibitors and exploration of their activities against protein tyrosine phosphatase 1B (PTP1B). J Mol Struct 2023. [DOI: 10.1016/j.molstruc.2022.134116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
27
|
Yuan H, Tang H, Shi L. Low expression of lncRNA UCA1 assists the diagnosis of type 2 diabetes mellitus and predicts an increased risk of cardiovascular complications. ALL LIFE 2022. [DOI: 10.1080/26895293.2022.2138561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Affiliation(s)
- Hui Yuan
- Department of Endocrinology, Daqing Oilfield General Hospital, Daqing, People’s Republic of China
| | - Haiyan Tang
- Department of Infectious Diseases, Daqing Oilfield General Hospital, Daqing, People’s Republic of China
| | - Lili Shi
- Department of Endocrinology, The First Affiliated Hospital of Harbin Medical University, Harbin, People’s Republic of China
| |
Collapse
|
28
|
Zhu H, Zhong X. Synthesis of activity evaluation of flavonoid derivatives as ɑ-glucosidase inhibitors. Front Chem 2022; 10:1041328. [DOI: 10.3389/fchem.2022.1041328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Accepted: 10/04/2022] [Indexed: 11/16/2022] Open
Abstract
Six flavonoid derivatives were synthesized and tested for anti-α-glucosidase activities. All derivatives were confirmed using NMR and HRMS and exhibited excellent inhibitory effects on α-glucosidase. Derivative four exhibited the highest anti-α-glucosidase activity (IC50: 15.71 ± 0.21 μM). Structure-activity relationship results showed that bromine group would be the most beneficial group to anti-α-glucosidase activity. Inhibitory mechnism and inhibition kinetics results showed derivative four was a reversible and mixed-type inhibitor. Molecular docking revealed that derivative four was tightly bind to the amino acid residues of active pocket of α-glucosidase and formed hydrogen bond, π-π stacking, and Pi-Donor hydrogen with α-glucosidase. Moreover, the physicochemical parameters of all derivatives were assessed using SwissADME software. This results also showed that the hybridization of flavonoid and phenylpropionic acid would be a useful strategy for the development of α-glucosidase inhibitors.
Collapse
|
29
|
Xie Q, Wang C. Polyacetylenes in herbal medicine: A comprehensive review of its occurrence, pharmacology, toxicology, and pharmacokinetics (2014-2021). PHYTOCHEMISTRY 2022; 201:113288. [PMID: 35718132 DOI: 10.1016/j.phytochem.2022.113288] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Revised: 05/16/2022] [Accepted: 06/13/2022] [Indexed: 06/15/2023]
Abstract
Polyacetylenes are a kind of small active compounds with carbon-carbon triple bond with vast occurrence in plants. Polyacetylenes have attracted considerable attention owing to their diverse biofunctions like tumor suppression, immunity regulation, depression resistance and neural protection. The present review intends to reconstruct data concerning the occurrence, pharmacology, toxicology and pharmacokinetics of polyacetylenes from herbal medicine in a systematic and integrated way, with a view to backing up their curative potential and healthcare properties (2014-2021). The natural polyacetylene-related data were all acquired from the scientific search engines and databases that are globally recognized, such as PubMed, Web of Science, Elsevier, Google Scholar, ResearchGate, SciFindern and CNKI. A total of 183 polyacetylenes were summarized in this paper. Modern pharmacological studies indicated that polyacetylenes possess multiple biological activities including antitumor, immunomodulatory, neuroprotective, anti-depression, anti-obesity, hypoglycemic, antiviral, antibacterial, antifungal, hepatoprotective and renoprotective activities. As important bioactive components of herbal medicine, the pharmacological curative potential of polyacetylenes has been described against carcinomas, inflammatory responses, central nervous system, endocrine disorders and microbial infection in this review. While, further in-depth studies on the aspects of polyacetylenes for toxicity, pharmacokinetics, and molecular mechanisms are still limited, thereby intensive research and assessments should be performed.
Collapse
Affiliation(s)
- Qi Xie
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai R&D Centre for Standardization of Chinese Medicines, 1200 Cailun Road, Shanghai, 201203, China
| | - Changhong Wang
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai R&D Centre for Standardization of Chinese Medicines, 1200 Cailun Road, Shanghai, 201203, China.
| |
Collapse
|
30
|
Pandey AR, Ahmad S, Singh SP, Mishra A, Bisen AC, Sharma G, Ahmad I, Shukla SK, Bhatta RS, Kanojiya S, Tamrakar AK, Sashidhara KV. Furostanol saponins from Asparagus racemosus as potential hypoglycemic agents. PHYTOCHEMISTRY 2022; 201:113286. [PMID: 35752344 DOI: 10.1016/j.phytochem.2022.113286] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 06/13/2022] [Accepted: 06/13/2022] [Indexed: 06/15/2023]
Abstract
Bioactivity guided phytochemical investigation led to isolation of six undescribed furostanol saponins, furoasparoside A-F along with five known compounds, gallic acid, methyl gallate, quercetin-3-O-β-glucopyranoside, liquiritigenin 4׳-O-β-apiofuranosyl-(1 → 2)-β-glucopyranoside and β-glucogallin for the first time from the roots of Asparagus racemosus. Isolated saponins were screened for their antidiabetic potential in L6-GLUT4myc myotubes in vitro followed by an in vivo evaluation in streptozocin-induced diabetic rats and db/db mice. Furoasparoside E produced a notable decrease in the postprandial blood glucose profile, in leptin receptor-deficient db/db mice, type 2 diabetes model. The effect of furoasparoside E on GLUT4 translocation was found to be mediated by the AMPK-dependent signaling pathway in L6-GLUT4myc myotubes. Moreover, it emerged as a stable plant metabolite with higher bioavailability and efficacy in in vivo pharmacokinetic studies. Therefore, these studies indicated that furoasparoside E may serve as a propitious lead for the management of type 2 diabetes and its secondary complications from natural source.
Collapse
Affiliation(s)
- Alka Raj Pandey
- Medicinal and Process Chemistry Division, CSIR-Central Drug Research Institute, BS-10/1, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, U.P., India
| | - Shadab Ahmad
- Division of Biochemistry & Structural Biology, CSIR-Central Drug Research Institute, BS-10/1, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, U.P., India
| | - Suriya Pratap Singh
- Medicinal and Process Chemistry Division, CSIR-Central Drug Research Institute, BS-10/1, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, 226031, India
| | - Anjali Mishra
- Pharmaceutics and Pharmacokinetics Division, CSIR-Central Drug Research Institute, BS-10/1, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, U.P., India
| | - Amol Chhatrapati Bisen
- Pharmaceutics and Pharmacokinetics Division, CSIR-Central Drug Research Institute, BS-10/1, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, U.P., India
| | - Gaurav Sharma
- Sophisticated Analytical Instrument Facility & Research, CSIR-Central Drug Research Institute, BS-10/1, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, 226031, U.P., India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, U.P., India
| | - Ishbal Ahmad
- Division of Biochemistry & Structural Biology, CSIR-Central Drug Research Institute, BS-10/1, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, 226031, India
| | - Sanjeev K Shukla
- Sophisticated Analytical Instrument Facility & Research, CSIR-Central Drug Research Institute, BS-10/1, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, 226031, U.P., India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, U.P., India
| | - Rabi Sankar Bhatta
- Pharmaceutics and Pharmacokinetics Division, CSIR-Central Drug Research Institute, BS-10/1, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, U.P., India
| | - Sanjeev Kanojiya
- Sophisticated Analytical Instrument Facility & Research, CSIR-Central Drug Research Institute, BS-10/1, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, 226031, U.P., India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, U.P., India
| | - Akhilesh Kumar Tamrakar
- Division of Biochemistry & Structural Biology, CSIR-Central Drug Research Institute, BS-10/1, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, U.P., India
| | - Koneni V Sashidhara
- Medicinal and Process Chemistry Division, CSIR-Central Drug Research Institute, BS-10/1, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, 226031, India; Sophisticated Analytical Instrument Facility & Research, CSIR-Central Drug Research Institute, BS-10/1, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, 226031, U.P., India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, U.P., India.
| |
Collapse
|
31
|
Yang Y, He J, Jiang Z, Du X, Chen F, Wang J, Ni H. Characterization of the inhibition of aldose reductase with
p
‐coumaric acid ethyl ester. J Food Biochem 2022; 46:e14370. [DOI: 10.1111/jfbc.14370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 06/14/2022] [Accepted: 07/22/2022] [Indexed: 12/01/2022]
Affiliation(s)
- Yuanfan Yang
- College of Ocean Food and Biological Engineering Jimei University Xiamen China
- Fujian Provincial Key Laboratory of Food Microbiology and Enzyme Engineering Xiamen China
- Research Center of Food Biotechnology of Xiamen City Xiamen China
| | - Junzhu He
- College of Ocean Food and Biological Engineering Jimei University Xiamen China
| | - Zedong Jiang
- College of Ocean Food and Biological Engineering Jimei University Xiamen China
- Fujian Provincial Key Laboratory of Food Microbiology and Enzyme Engineering Xiamen China
- Research Center of Food Biotechnology of Xiamen City Xiamen China
| | - Xiping Du
- College of Ocean Food and Biological Engineering Jimei University Xiamen China
- Fujian Provincial Key Laboratory of Food Microbiology and Enzyme Engineering Xiamen China
- Research Center of Food Biotechnology of Xiamen City Xiamen China
| | - Feng Chen
- Department of Food, Nutrition and Packaging Sciences Clemson University Clemson South Carolina USA
| | - Jinling Wang
- School of Forestry Northeast Forestry University Harbin China
| | - Hui Ni
- College of Ocean Food and Biological Engineering Jimei University Xiamen China
- Fujian Provincial Key Laboratory of Food Microbiology and Enzyme Engineering Xiamen China
- Research Center of Food Biotechnology of Xiamen City Xiamen China
| |
Collapse
|
32
|
Saeed A, Ejaz SA, Sarfraz M, Tamam N, Siddique F, Riaz N, Qais FA, Chtita S, Iqbal J. Discovery of Phenylcarbamoylazinane-1,2,4-Triazole Amides Derivatives as the Potential Inhibitors of Aldo-Keto Reductases (AKR1B1 & AKRB10): Potential Lead Molecules for Treatment of Colon Cancer. Molecules 2022; 27:molecules27133981. [PMID: 35807227 PMCID: PMC9268700 DOI: 10.3390/molecules27133981] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Revised: 05/19/2022] [Accepted: 05/23/2022] [Indexed: 12/12/2022] Open
Abstract
Both members of the aldo-keto reductases (AKRs) family, AKR1B1 and AKR1B10, are over-expressed in various type of cancer, making them potential targets for inflammation-mediated cancers such as colon, lung, breast, and prostate cancers. This is the first comprehensive study which focused on the identification of phenylcarbamoylazinane-1, 2,4-triazole amides (7a−o) as the inhibitors of aldo-keto reductases (AKR1B1, AKR1B10) via detailed computational analysis. Firstly, the stability and reactivity of compounds were determined by using the Guassian09 programme in which the density functional theory (DFT) calculations were performed by using the B3LYP/SVP level. Among all the derivatives, the 7d, 7e, 7f, 7h, 7j, 7k, and 7m were found chemically reactive. Then the binding interactions of the optimized compounds within the active pocket of the selected targets were carried out by using molecular docking software: AutoDock tools and Molecular operation environment (MOE) software, and during analysis, the Autodock (academic software) results were found to be reproducible, suggesting this software is best over the MOE (commercial software). The results were found in correlation with the DFT results, suggesting 7d as the best inhibitor of AKR1B1 with the energy value of −49.40 kJ/mol and 7f as the best inhibitor of AKR1B10 with the energy value of −52.84 kJ/mol. The other potent compounds also showed comparable binding energies. The best inhibitors of both targets were validated by the molecular dynamics simulation studies where the root mean square value of <2 along with the other physicochemical properties, hydrogen bond interactions, and binding energies were observed. Furthermore, the anticancer potential of the potent compounds was confirmed by cell viability (MTT) assay. The studied compounds fall into the category of drug-like properties and also supported by physicochemical and pharmacological ADMET properties. It can be suggested that the further synthesis of derivatives of 7d and 7f may lead to the potential drug-like molecules for the treatment of colon cancer associated with the aberrant expression of either AKR1B1 or AKR1B10 and other associated malignancies.
Collapse
Affiliation(s)
- Amna Saeed
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, The Islamia University of Bahawalpur, Bahawalpur 63100, Pakistan;
| | - Syeda Abida Ejaz
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, The Islamia University of Bahawalpur, Bahawalpur 63100, Pakistan;
- Correspondence: (S.A.E.); (J.I.)
| | - Muhammad Sarfraz
- College of Pharmacy, Al Ain Campus, Al Ain University, Al Ain P.O. Box 64141, United Arab Emirates;
| | - Nissren Tamam
- Department of Physics, College of Science, Princess Nourah bint Abdulrahman University, P.O Box 84428, Riyadh 11671, Saudi Arabia;
| | - Farhan Siddique
- Laboratory of Organic Electronics, Department of Science and Technology, Linköping University, SE-60174 Norrköping, Sweden;
- Department of Pharmacy, Royal Institute of Medical Sciences (RIMS), Multan 60000, Pakistan
| | - Naheed Riaz
- Department of Chemistry, Baghdad-ul-Jadeed Campus, The Islamia University of Bahawalpur, Bahawalpur 63100, Pakistan;
| | - Faizan Abul Qais
- Department of Agricultural Microbiology, Faculty of Agricultural Sciences, Aligarh Muslim University, Aligarh 202002, UP, India;
| | - Samir Chtita
- Laboratory of Analytical and Molecular Chemistry, Faculty of Sciences Ben M’Sik, Hassan II University of Casablanca, Sidi Othmane, Casablanca BP7955, Morocco;
| | - Jamshed Iqbal
- Centre for Advanced Drug Research, Abbottabad Campus, COMSATS University Islamabad, Abbotabad 22060, Pakistan
- Correspondence: (S.A.E.); (J.I.)
| |
Collapse
|
33
|
Ghemrawi R, Khair M, Hasan S, Aldulaymi R, AlNeyadi SS, Atatreh N, Ghattas MA. The Discovery of Potent SHP2 Inhibitors with Anti-Proliferative Activity in Breast Cancer Cell Lines. Int J Mol Sci 2022; 23:ijms23084468. [PMID: 35457286 PMCID: PMC9030381 DOI: 10.3390/ijms23084468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 04/14/2022] [Accepted: 04/17/2022] [Indexed: 02/04/2023] Open
Abstract
Despite available treatments, breast cancer is the leading cause of cancer-related death. Knowing that the tyrosine phosphatase SHP2 is a regulator in tumorigenesis, developing inhibitors of SHP2 in breast cells is crucial. Our study investigated the effects of new compounds, purchased from NSC, on the phosphatase activity of SHP2 and the modulation of breast cancer cell lines’ proliferation and viability. A combined ligand-based and structure-based virtual screening protocol was validated, then performed, against SHP2 active site. Top ranked compounds were tested via SHP2 enzymatic assay, followed by measuring IC50 values. Subsequently, hits were tested for their anti-breast cancer viability and proliferative activity. Our experiments identified three compounds 13030, 24198, and 57774 as SHP2 inhibitors, with IC50 values in micromolar levels and considerable selectivity over the analogous enzyme SHP1. Long MD simulations of 500 ns showed a very promising binding mode in the SHP2 catalytic pocket. Furthermore, these compounds significantly reduced MCF-7 breast cancer cells’ proliferation and viability. Interestingly, two of our hits can have acridine or phenoxazine cyclic system known to intercalate in ds DNA. Therefore, our novel approach led to the discovery of SHP2 inhibitors, which could act as a starting point in the future for clinically useful anticancer agents.
Collapse
Affiliation(s)
- Rose Ghemrawi
- College of Pharmacy, Al Ain University, Abu Dhabi 112612, United Arab Emirates; (R.G.); (S.H.)
- AAU Health and Biomedical Research Center, Al Ain University, Abu Dhabi 112612, United Arab Emirates;
| | - Mostafa Khair
- Core Technology Platforms, New York University Abu Dhabi, Abu Dhabi 129188, United Arab Emirates;
| | - Shaima Hasan
- College of Pharmacy, Al Ain University, Abu Dhabi 112612, United Arab Emirates; (R.G.); (S.H.)
| | - Raghad Aldulaymi
- AAU Health and Biomedical Research Center, Al Ain University, Abu Dhabi 112612, United Arab Emirates;
| | - Shaikha S. AlNeyadi
- Department of Chemistry, College of Science, UAE University Al-Ain, Abu Dhabi 15551, United Arab Emirates;
| | - Noor Atatreh
- College of Pharmacy, Al Ain University, Abu Dhabi 112612, United Arab Emirates; (R.G.); (S.H.)
- AAU Health and Biomedical Research Center, Al Ain University, Abu Dhabi 112612, United Arab Emirates;
- Correspondence: (N.A.); (M.A.G.)
| | - Mohammad A. Ghattas
- College of Pharmacy, Al Ain University, Abu Dhabi 112612, United Arab Emirates; (R.G.); (S.H.)
- AAU Health and Biomedical Research Center, Al Ain University, Abu Dhabi 112612, United Arab Emirates;
- Correspondence: (N.A.); (M.A.G.)
| |
Collapse
|
34
|
Zhang H, Chen R, Xu C, Zhang G, Guan Y, Feng Q, Yao J, Yan J. An integrated approach to discriminate the quality markers of Traditional Chinese medicine preparation based on multi-dimensional characteristic network: Shenqi Jiangtang Granule as a case. JOURNAL OF ETHNOPHARMACOLOGY 2021; 278:114277. [PMID: 34089811 DOI: 10.1016/j.jep.2021.114277] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 05/23/2021] [Accepted: 05/30/2021] [Indexed: 06/12/2023]
Abstract
BACKGROUND Due to the complexity of traditional Chinese medicine (TCM), the current quality evaluation of TCM are difficult to associate with clinical efficacy. Shenqi Jiangtang Granule (SJG), a classical TCM formula, is proven as a therapy for treatment of type II diabetes mellitus (DM) and complications while the substantial basis of the therapeutic effects is not clear. PURPOSE The present study proposed an integrated approach to discriminate the quality markers (Q-markers) based on multi-dimensional characteristic network for quality control of TCM. METHODS The multi-dimensional characteristic network was established by "Spider-web" mode, which was comprehensively integrating "compatibility-content-activity- efficiency-stability" of the candidate ingredients. The activity dimension was evaluated by the inhibitory activity of SJG on α-glucosidase and aldose reductase. The efficacy dimension was assessed through the association between the compounds and the target pathway of diabetic nephropathy (DN) based on integrated pharmacology platform. Each dimension for the feature network was quantified by multivariate statistical analysis, and regression area of the candidate compounds was constructed in the network. Finally, the candidate compounds were sorted comprehensively by the regression area. RESULTS A total of 30 chemical compounds with effective hypoglycemic activity were identified as the potential Q-markers. From the data analysis, three dimensions of activity, efficacy and content performed a greater impact on the regression area of the characteristic network. Among these compounds, ginsenoside Re, ginsenoside Rd, ginsenoside Rg1, calycosin, ginsenoside Rb1, formononetin, astragaloside IV, ginsenoside Rf, ginsenoside Rc, notoginsenoside Fe, schisandrol A, gomisin D were screened out as the candidate Q-markers of SJG. CONCLUSION The multi-dimensional characteristic network integrating compatibility, content, activity, efficiency and stability is efficient to discriminate the potential Q-markers of TCM prescription. Our results demonstrated that 12 candidate compounds from Panax Ginseng, Radix Astragali and Schisandrae Chinensis might select as Q-markers for qualitative evaluation of SJG.
Collapse
Affiliation(s)
- Hui Zhang
- College of Pharmaceutical Science, Zhejiang University of Technology, No. 18, Chaowang Road, Hangzhou 310014, China.
| | - Ruoyu Chen
- College of Pharmaceutical Science, Zhejiang University of Technology, No. 18, Chaowang Road, Hangzhou 310014, China
| | - Cong Xu
- College of Pharmaceutical Science, Zhejiang University of Technology, No. 18, Chaowang Road, Hangzhou 310014, China
| | - Guimin Zhang
- State Key Laboratory of Generic Manufacture Technology of Chinese Traditional Medicine, Shandong, 276006, China; Lunan Pharmaceutical Group Co., Ltd., Shandong, 276006, China
| | - Yongxia Guan
- State Key Laboratory of Generic Manufacture Technology of Chinese Traditional Medicine, Shandong, 276006, China; Lunan Pharmaceutical Group Co., Ltd., Shandong, 276006, China
| | - Qun Feng
- State Key Laboratory of Generic Manufacture Technology of Chinese Traditional Medicine, Shandong, 276006, China; Lunan Pharmaceutical Group Co., Ltd., Shandong, 276006, China
| | - Jingchun Yao
- State Key Laboratory of Generic Manufacture Technology of Chinese Traditional Medicine, Shandong, 276006, China; Lunan Pharmaceutical Group Co., Ltd., Shandong, 276006, China
| | - Jizhong Yan
- College of Pharmaceutical Science, Zhejiang University of Technology, No. 18, Chaowang Road, Hangzhou 310014, China.
| |
Collapse
|
35
|
Dual Targeting of PTP1B and Aldose Reductase with Marine Drug Phosphoeleganin: A Promising Strategy for Treatment of Type 2 Diabetes. Mar Drugs 2021; 19:md19100535. [PMID: 34677434 PMCID: PMC8540617 DOI: 10.3390/md19100535] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 09/21/2021] [Accepted: 09/21/2021] [Indexed: 12/14/2022] Open
Abstract
An in-depth study on the inhibitory mechanism on protein tyrosine phosphatase 1B (PTP1B) and aldose reductase (AR) enzymes, including analysis of the insulin signalling pathway, of phosphoeleganin, a marine-derived phosphorylated polyketide, was achieved. Phosphoeleganin was demonstrated to inhibit both enzymes, acting respectively as a pure non-competitive inhibitor of PTP1B and a mixed-type inhibitor of AR. In addition, in silico docking analyses to evaluate the interaction mode of phosphoeleganin with both enzymes were performed. Interestingly, this study showed that phosphoeleganin is the first example of a dual inhibitor polyketide extracted from a marine invertebrate, and it could be used as a versatile scaffold structure for the synthesis of new designed multiple ligands.
Collapse
|
36
|
Non-acidic bifunctional benzothiazole-based thiazolidinones with antimicrobial and aldose reductase inhibitory activity as a promising therapeutic strategy for sepsis. Med Chem Res 2021; 30:1837-1848. [PMID: 34366640 PMCID: PMC8335715 DOI: 10.1007/s00044-021-02778-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 07/27/2021] [Indexed: 12/24/2022]
Abstract
Sepsis is a life-threatening disease that affects millions of people worldwide. Microbial infections that lead to sepsis syndrome are associated with an increased production of inflammatory molecules. Aldose reductase has recently emerged as a molecular target that is involved in various inflammatory diseases, including sepsis. Herein, a series of previously synthesized benzothiazole-based thiazolidinones that exhibited strong antibacterial and antifungal activities has been evaluated for inhibition efficacy against aldose reductase and selectivity toward aldehyde reductase under in vitro conditions. The most promising inhibitor 5 was characterized with IC50 value of 3.99 μM and a moderate selectivity. Molecular docking simulations revealed the binding mode of compounds at the active site of human aldose reductase. Moreover, owning to the absence of an acidic pharmacophore, good membrane permeation of the novel aldose reductase inhibitors was predicted. Excellent “drug-likeness” was assessed for most of the compounds by applying the criteria of Lipinski’s “rule of five”. ![]()
Collapse
|
37
|
Sever B, Altıntop MD, Demir Y, Yılmaz N, Akalın Çiftçi G, Beydemir Ş, Özdemir A. Identification of a new class of potent aldose reductase inhibitors: Design, microwave-assisted synthesis, in vitro and in silico evaluation of 2-pyrazolines. Chem Biol Interact 2021; 345:109576. [PMID: 34252406 DOI: 10.1016/j.cbi.2021.109576] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Revised: 06/17/2021] [Accepted: 07/08/2021] [Indexed: 02/05/2023]
Abstract
Aldose reductase (AR) acts as a multi-disease target for the design and development of therapeutic agents for the management of diabetic complications as well as non-diabetic diseases. In the search for potent AR inhibitors, the microwave-assisted synthesis of twenty new compounds with a 1,3-diaryl-5-(4-fluorophenyl)-2-pyrazoline moiety as a common fragment in their structure (1-20) was carried out efficiently. Compounds 1-20 were subjected to in vitro studies, which were conducted to assess their AR inhibitory effects and cytotoxicity towards L929 mouse fibroblast (normal) cells. Among these compounds, 1-(3-bromophenyl)-3-(4-piperidinophenyl)-5-(4-fluorophenyl)-2-pyrazoline (20) was identified as the most promising AR inhibitor with an IC50 value of 0.160 ± 0.005 μM exerting competitive inhibition with a Ki value of 0.019 ± 0.001 μM as compared to epalrestat (IC50 = 0.279 ± 0.001 μM; Ki = 0.801 ± 0.023 μM) and quercetin (IC50 = 4.120 ± 0.123 μM; Ki = 6.082 ± 0.272 μM). Compound 20 displayed cytotoxicity towards L929 cells with an IC50 value of 18.75 ± 1.06 μM highlighting its safety as an AR inhibitor. Molecular docking studies suggested that π-π stacking interactions occurred between the m-bromophenyl moiety of compound 20 and Trp21. Based on in silico pharmacokinetic studies, compound 20 was found to possess favorable oral bioavailability and drug-like properties. It can be concluded that compound 20 is a potential orally bioavailable AR inhibitor for the management of diabetic complications as well as non-diabetic diseases.
Collapse
Affiliation(s)
- Belgin Sever
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Anadolu University, 26470, Eskişehir, Turkey
| | - Mehlika Dilek Altıntop
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Anadolu University, 26470, Eskişehir, Turkey.
| | - Yeliz Demir
- Department of Pharmacy Services, Nihat Delibalta Göle Vocational High School, Ardahan University, 75700, Ardahan, Turkey
| | - Nalan Yılmaz
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Anadolu University, 26470, Eskişehir, Turkey
| | - Gülşen Akalın Çiftçi
- Department of Biochemistry, Faculty of Pharmacy, Anadolu University, 26470, Eskişehir, Turkey
| | - Şükrü Beydemir
- Department of Biochemistry, Faculty of Pharmacy, Anadolu University, 26470, Eskişehir, Turkey; The Rectorate of Bilecik Şeyh Edebali University, 11230, Bilecik, Turkey
| | - Ahmet Özdemir
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Anadolu University, 26470, Eskişehir, Turkey
| |
Collapse
|
38
|
Kovacikova L, Prnova MS, Majekova M, Bohac A, Karasu C, Stefek M. Development of Novel Indole-Based Bifunctional Aldose Reductase Inhibitors/Antioxidants as Promising Drugs for the Treatment of Diabetic Complications. Molecules 2021; 26:molecules26102867. [PMID: 34066081 PMCID: PMC8151378 DOI: 10.3390/molecules26102867] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 04/29/2021] [Accepted: 05/04/2021] [Indexed: 01/16/2023] Open
Abstract
Aldose reductase (AR, ALR2), the first enzyme of the polyol pathway, is implicated in the pathophysiology of diabetic complications. Aldose reductase inhibitors (ARIs) thus present a promising therapeutic approach to treat a wide array of diabetic complications. Moreover, a therapeutic potential of ARIs in the treatment of chronic inflammation-related pathologies and several genetic metabolic disorders has been recently indicated. Substituted indoles are an interesting group of compounds with a plethora of biological activities. This article reviews a series of indole-based bifunctional aldose reductase inhibitors/antioxidants (ARIs/AOs) developed during recent years. Experimental results obtained in in vitro, ex vivo, and in vivo models of diabetic complications are presented. Structure–activity relationships with respect to carboxymethyl pharmacophore regioisomerization and core scaffold modification are discussed along with the criteria of ‘drug-likeness”. Novel promising structures of putative multifunctional ARIs/AOs are designed.
Collapse
Affiliation(s)
- Lucia Kovacikova
- Institute of Experimental Pharmacology and Toxicology, CEM SAS, Dúbravská Cesta 9, 841 04 Bratislava, Slovakia; (L.K.); (M.S.P.); (M.M.)
| | - Marta Soltesova Prnova
- Institute of Experimental Pharmacology and Toxicology, CEM SAS, Dúbravská Cesta 9, 841 04 Bratislava, Slovakia; (L.K.); (M.S.P.); (M.M.)
| | - Magdalena Majekova
- Institute of Experimental Pharmacology and Toxicology, CEM SAS, Dúbravská Cesta 9, 841 04 Bratislava, Slovakia; (L.K.); (M.S.P.); (M.M.)
| | - Andrej Bohac
- Department of Organic Chemistry, Faculty of Natural Sciences, Comenius University in Bratislava, Ilkovičova 6, 842 15 Bratislava, Slovakia;
- Biomagi, Inc., Mamateyova 26, 851 04 Bratislava, Slovakia
| | - Cimen Karasu
- Cellular Stress Response and Signal Transduction Research Laboratory, Department of Medical Pharmacology, Faculty of Medicine, Gazi University, Beşevler, 06500 Ankara, Turkey;
| | - Milan Stefek
- Institute of Experimental Pharmacology and Toxicology, CEM SAS, Dúbravská Cesta 9, 841 04 Bratislava, Slovakia; (L.K.); (M.S.P.); (M.M.)
- Correspondence:
| |
Collapse
|
39
|
Sipos Á, Szennyes E, Hajnal NÉ, Kun S, Szabó KE, Uray K, Somsák L, Docsa T, Bokor É. Dual-Target Compounds against Type 2 Diabetes Mellitus: Proof of Concept for Sodium Dependent Glucose Transporter (SGLT) and Glycogen Phosphorylase (GP) Inhibitors. Pharmaceuticals (Basel) 2021; 14:ph14040364. [PMID: 33920838 PMCID: PMC8071193 DOI: 10.3390/ph14040364] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 04/07/2021] [Accepted: 04/13/2021] [Indexed: 12/15/2022] Open
Abstract
A current trend in the quest for new therapies for complex, multifactorial diseases, such as diabetes mellitus (DM), is to find dual or even multi-target inhibitors. In DM, the sodium dependent glucose cotransporter 2 (SGLT2) in the kidneys and the glycogen phosphorylase (GP) in the liver are validated targets. Several (β-D-glucopyranosylaryl)methyl (het)arene type compounds, called gliflozins, are marketed drugs that target SGLT2. For GP, low nanomolar glucose analogue inhibitors exist. The purpose of this study was to identify dual acting compounds which inhibit both SGLTs and GP. To this end, we have extended the structure-activity relationships of SGLT2 and GP inhibitors to scarcely known (C-β-D-glucopyranosylhetaryl)methyl arene type compounds and studied several (C-β-D-glucopyranosylhetaryl)arene type GP inhibitors against SGLT. New compounds, such as 5-arylmethyl-3-(β-D-glucopyranosyl)-1,2,4-oxadiazoles, 5-arylmethyl-2-(β-D-glucopyranosyl)-1,3,4-oxadiazoles, 4-arylmethyl-2-(β-D-glucopyranosyl)pyrimidines and 4(5)-benzyl-2-(β-D-glucopyranosyl)imidazole were prepared by adapting our previous synthetic methods. None of the studied compounds exhibited cytotoxicity and all of them were assayed for their SGLT1 and 2 inhibitory potentials in a SGLT-overexpressing TSA201 cell system. GP inhibition was also determined by known methods. Several newly synthesized (C-β-D-glucopyranosylhetaryl)methyl arene derivatives had low micromolar SGLT2 inhibitory activity; however, none of these compounds inhibited GP. On the other hand, several (C-β-D-glucopyranosylhetaryl)arene type GP inhibitor compounds with low micromolar efficacy against SGLT2 were identified. The best dual inhibitor, 2-(β-D-glucopyranosyl)-4(5)-(2-naphthyl)-imidazole, had a Ki of 31 nM for GP and IC50 of 3.5 μM for SGLT2. This first example of an SGLT-GP dual inhibitor can prospectively be developed into even more efficient dual-target compounds with potential applications in future antidiabetic therapy.
Collapse
Affiliation(s)
- Ádám Sipos
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Egyetem tér 1, H-4032 Debrecen, Hungary; (Á.S.); (K.U.)
- Doctoral School of Molecular Medicine, University of Debrecen, Egyetem tér 1, H-4032 Debrecen, Hungary
| | - Eszter Szennyes
- Department of Organic Chemistry, University of Debrecen, POB 400, H-4002 Debrecen, Hungary; (E.S.); (N.É.H.); (S.K.); (K.E.S.)
| | - Nikolett Éva Hajnal
- Department of Organic Chemistry, University of Debrecen, POB 400, H-4002 Debrecen, Hungary; (E.S.); (N.É.H.); (S.K.); (K.E.S.)
| | - Sándor Kun
- Department of Organic Chemistry, University of Debrecen, POB 400, H-4002 Debrecen, Hungary; (E.S.); (N.É.H.); (S.K.); (K.E.S.)
| | - Katalin E. Szabó
- Department of Organic Chemistry, University of Debrecen, POB 400, H-4002 Debrecen, Hungary; (E.S.); (N.É.H.); (S.K.); (K.E.S.)
| | - Karen Uray
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Egyetem tér 1, H-4032 Debrecen, Hungary; (Á.S.); (K.U.)
| | - László Somsák
- Department of Organic Chemistry, University of Debrecen, POB 400, H-4002 Debrecen, Hungary; (E.S.); (N.É.H.); (S.K.); (K.E.S.)
- Correspondence: (L.S.); (T.D.); (É.B.); Tel.: +36-525-129-00 (ext. 22348) (L.S.); +36-525-186-00 (ext. 61192) (T.D.); +36-525-129-00 (ext. 22474) (É.B.)
| | - Tibor Docsa
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Egyetem tér 1, H-4032 Debrecen, Hungary; (Á.S.); (K.U.)
- Correspondence: (L.S.); (T.D.); (É.B.); Tel.: +36-525-129-00 (ext. 22348) (L.S.); +36-525-186-00 (ext. 61192) (T.D.); +36-525-129-00 (ext. 22474) (É.B.)
| | - Éva Bokor
- Department of Organic Chemistry, University of Debrecen, POB 400, H-4002 Debrecen, Hungary; (E.S.); (N.É.H.); (S.K.); (K.E.S.)
- Correspondence: (L.S.); (T.D.); (É.B.); Tel.: +36-525-129-00 (ext. 22348) (L.S.); +36-525-186-00 (ext. 61192) (T.D.); +36-525-129-00 (ext. 22474) (É.B.)
| |
Collapse
|
40
|
Sever B, Altıntop MD, Demir Y, Türkeş C, Özbaş K, Çiftçi GA, Beydemir Ş, Özdemir A. A new series of 2,4-thiazolidinediones endowed with potent aldose reductase inhibitory activity. OPEN CHEM 2021. [DOI: 10.1515/chem-2021-0032] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Abstract
In an effort to identify potent aldose reductase (AR) inhibitors, 5-(arylidene)thiazolidine-2,4-diones (1–8), which were prepared by the solvent-free reaction of 2,4-thiazolidinedione with aromatic aldehydes in the presence of urea, were examined for their in vitro AR inhibitory activities and cytotoxicity. 5-(2-Hydroxy-3-methylbenzylidene)thiazolidine-2,4-dione (3) was the most potent AR inhibitor in this series, exerting uncompetitive inhibition with a K
i value of 0.445 ± 0.013 µM. The IC50 value of compound 3 for L929 mouse fibroblast cells was determined as 8.9 ± 0.66 µM, pointing out its safety as an AR inhibitor. Molecular docking studies suggested that compound 3 exhibited good affinity to the binding site of AR (PDB ID: 4JIR). Based upon in silico absorption, distribution, metabolism, and excretion data, the compound is predicted to have favorable pharmacokinetic features. Taking into account the in silico and in vitro data, compound 3 stands out as a potential orally bioavailable AR inhibitor for the management of diabetic complications as well as nondiabetic diseases.
Collapse
Affiliation(s)
- Belgin Sever
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Anadolu University , 26470 Eskişehir , Turkey
| | - Mehlika Dilek Altıntop
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Anadolu University , 26470 Eskişehir , Turkey
| | - Yeliz Demir
- Department of Pharmacy Services, Nihat Delibalta Göle Vocational High School, Ardahan University , 75700 Ardahan , Turkey
| | - Cüneyt Türkeş
- Department of Biochemistry, Faculty of Pharmacy, Erzincan Binali Yıldırım University , 24100 Erzincan , Turkey
| | - Kaan Özbaş
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Anadolu University , 26470 Eskişehir , Turkey
| | - Gülşen Akalın Çiftçi
- Department of Biochemistry, Faculty of Pharmacy, Anadolu University , 26470 Eskişehir , Turkey
| | - Şükrü Beydemir
- Department of Biochemistry, Faculty of Pharmacy, Anadolu University , 26470 Eskişehir , Turkey
- The Rectorate of Bilecik Şeyh Edebali University , 11230 Bilecik , Turkey
| | - Ahmet Özdemir
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Anadolu University , 26470 Eskişehir , Turkey
| |
Collapse
|
41
|
Wang Z, Wu Z, Zuo G, Lim SS, Yan H. Defatted Seeds of Oenothera biennis as a Potential Functional Food Ingredient for Diabetes. Foods 2021; 10:foods10030538. [PMID: 33807644 PMCID: PMC8002154 DOI: 10.3390/foods10030538] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 02/22/2021] [Accepted: 03/02/2021] [Indexed: 12/23/2022] Open
Abstract
The defatted seeds of Oenothera biennis (DSOB) are a by-product of evening primrose oil production that are currently not effectively used. In this study, α-glucosidase inhibition, aldose reductase inhibition, antioxidant capacity, polyphenol composition, and nutritional value (carbohydrates, proteins, minerals, fat, organic acid, and tocopherols) of DSOB were evaluated using the seeds of Oenothera biennis (SOB) as a reference. DSOB was an excellent inhibitor of α-glucosidase (IC50 = 3.31 μg/mL) and aldose reductase (IC50 = 2.56 μg/mL). DSOB also showed considerable antioxidant capacities (scavenging of 2,2-diphenyl-1-picrylhydrazyl, 2,2’-azino-bis(3-ethylbenzothiazoline-6-sulfonic acid, nitric oxide, peroxynitrite, and hydroxyl radicals). DSOB was a reservoir of polyphenols, and 25 compounds in DSOB were temporarily identified by liquid chromatography coupled with electrospray ionization–quadrupole time of flight–mass spectrometry analysis. Moreover, the carbohydrate, protein, and mineral content of DSOB were increased compared to that of SOB. DSOB contained large amounts of fiber and low levels of sugars, and was rich in calcium and iron. These results imply that DSOB may be a potential functional food ingredient for diabetes, providing excellent economic and environmental benefits.
Collapse
Affiliation(s)
- Zhiqiang Wang
- Key Laboratory of Public Health Safety of Hebei Province, College of Public Health, Hebei University, Baoding 071002, China;
- Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, Institute of Life Science and Green Development, Hebei University, Baoding 071002, China
- Correspondence: (Z.W.); (H.Y.); Tel.: +86-312-5079010 (Z.W.); +86-312-5078507 (H.Y.)
| | - Zhaoyang Wu
- Key Laboratory of Public Health Safety of Hebei Province, College of Public Health, Hebei University, Baoding 071002, China;
| | - Guanglei Zuo
- Department of Food Science and Nutrition, Hallym University, 1 Hallymdeahak-gil, Chuncheon 24252, Korea; (G.Z.); (S.S.L.)
| | - Soon Sung Lim
- Department of Food Science and Nutrition, Hallym University, 1 Hallymdeahak-gil, Chuncheon 24252, Korea; (G.Z.); (S.S.L.)
| | - Hongyuan Yan
- Key Laboratory of Public Health Safety of Hebei Province, College of Public Health, Hebei University, Baoding 071002, China;
- Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, Institute of Life Science and Green Development, Hebei University, Baoding 071002, China
- Correspondence: (Z.W.); (H.Y.); Tel.: +86-312-5079010 (Z.W.); +86-312-5078507 (H.Y.)
| |
Collapse
|