1
|
Bhuvaneswari D, Riitvek B, Lakshmi BS. Multi Targeted Activity of Cocculus hirsutus through Modulation of DPP-IV and PTP-1B Leading to Enhancement of Glucose Uptake and Attenuation of Lipid Accumulation. Appl Biochem Biotechnol 2025:10.1007/s12010-024-05142-8. [PMID: 39760988 DOI: 10.1007/s12010-024-05142-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/24/2024] [Indexed: 01/07/2025]
Abstract
Multi-targeted therapies are gaining attention in the management of multifactorial diseases due to their poly pharmacology, enhanced potency and reduced toxicity. Metabolic disorders like Type 2 diabetes mellitus (T2DM) and obesity necessitate multi-targeted therapy to improve insulin sensitivity, regulate glucose homeostasis and support weight loss. Medicinal plants rich in bioactive compounds exhibit multi-targetted action with minimal side effects. In the current study, Cocculus hirsutus methanol extract (CME) and its hydromethanolic fraction (HMF) were investigated for their multi-target potential. Significant inhibition of Dipeptidyl peptidase IV (DPP-IV), a key enzyme in glucose metabolism was observed due to CME (54%) and HMF (70%) at 10 µg/ml and 1 µg/ml respectively. Protein Tyrosine Phosphatase 1B (PTP-1B), involved in the regulation of insulin signalling, was also inhibited by CME (67%) and HMF (73%) at 10 µg/ml concentration. An increase in glucose uptake was observed due to CME (62% and 65%) and HMF (63% and 68%) in 3T3-L1 adipocytes and L6 myotubes at 100 ng/ml. Further, investigation of HMF showed a decrease in lipid accumulation by 63% at 1 µg/ml in 3T3-L1 cells. Interestingly, HMF improved insulin sensitivity by upregulating GLUT4 expression (p < 0.05) via the PI3K/AKT pathway in both 3T3-L1 adipocytes and L6 myotubes. An inhibition in lipid accumulation was also observed by suppression of Peroxisome proliferator-activated receptor γ (PPARγ) (p < 0.05), a key regulator of adipogenesis in 3T3-L1 adipocytes. Gas chromatography-mass spectrometry analysis of the HMF showed the major component to be 3-methylmannoside (26.52%).
Collapse
Affiliation(s)
- D Bhuvaneswari
- Tissue Culture and Drug Discovery Laboratory, Department of Biotechnology, Anna University, Chennai, 600 025, India
| | - B Riitvek
- Department of Biochemistry and Biophysics, Oregon State University, Corvallis, OR, USA
| | - B S Lakshmi
- Tissue Culture and Drug Discovery Laboratory, Department of Biotechnology, Anna University, Chennai, 600 025, India.
| |
Collapse
|
2
|
Puranik HH, Thomas AB, Lokhande KB, Shrivastava A, Singh A, Swamy VK, Chitlange SS. Exploring the DPP IV inhibitory potential: molecular docking and dynamic simulations of pyridine-3-carboxylic acid and pyrrolidine-2-carboxylic acid analogs. J Biomol Struct Dyn 2024:1-21. [PMID: 39671243 DOI: 10.1080/07391102.2024.2439579] [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: 12/08/2023] [Accepted: 04/05/2024] [Indexed: 12/14/2024]
Abstract
Diabetes mellitus remains a global challenge, with Type 2 Diabetes Mellitus (T2DM) prevalence increasing from 4% to 6.4% in the past 30 years. Presently oral hypoglycaemic agents like GLP-1 agonists, biguanides, sulphonylureas, glinides, and thiazolidinediones are employed in clinical practice. Very recently, novel targets including Dipeptidyl peptidase IV (DPP IV), PPAR, GIP, FFA1, and melatonin have been in the limelight for the development of novel treatment strategies. The present study focuses on the development of DPP IV inhibitors through computational approaches. DPP IV, also referred to as CD26 (cluster of differentiation 26) or adenosine deaminase complexing protein 2, is a protein that is encoded by the DPP IV gene in humans. This enzyme is involved in the metabolism of incretin hormones such as glucagon-like peptides (GLP-1). DPP IV inhibitors prevent the degradation of GLP-1, glucose-dependent insulinotropic peptide (GIP), thereby controlling the concentration of glucose in the blood. Considering the safety and efficacy of DPP IV inhibitors newer molecules were designed with better binding affinity with the protein as compared to existing Sitagliptin, and Vildagliptin-like drugs. Derivatives of nicotinic acid and proline were designed and studied using molecular docking and dynamic simulations. Docking results demonstrated that the NA-13 molecule possesses potent binding affinity with target protein 6B1E (-38.1498 kcal/mol) as compared to standard Sitagliptin (-33.3187 kcal/mol). MD simulation studies showcased that there are fewer variations of RMSD and RMSF for 6B1E-NA-13, 6B1E-P1, and 6B1E-P7 complexes, suggesting the potential of the designed DPP IV inhibitors in the management of T2DM.
Collapse
Affiliation(s)
- Harshada H Puranik
- Department of Pharmaceutical Chemistry, Dr. D. Y. Patil Institute of Pharmaceutical Science and Research, affiliated to SPPU, Pune, India
| | - Asha B Thomas
- Department of Pharmaceutical Chemistry, Dr. D. Y. Patil Institute of Pharmaceutical Science and Research, affiliated to SPPU, Pune, India
| | - Kiran Bharat Lokhande
- Bioinformatics Research Laboratory, Dr. D. Y. Patil Biotechnology and Bioinformatics Institute, Dr. D. Y. Patil Vidyapeeth, Pune, India
- Translational, Bioinformatics and Computational Genomics Research Lab, Department of Life Sciences, Shiv Nadar Institution of Eminence, Gautam Buddha Nagar, India
| | - Ashish Shrivastava
- Translational, Bioinformatics and Computational Genomics Research Lab, Department of Life Sciences, Shiv Nadar Institution of Eminence, Gautam Buddha Nagar, India
| | - Ashutosh Singh
- Translational, Bioinformatics and Computational Genomics Research Lab, Department of Life Sciences, Shiv Nadar Institution of Eminence, Gautam Buddha Nagar, India
| | - Venkateswara K Swamy
- MIT School of Bioengineering Sciences & Research, MIT Art, Design and Technology University, Pune, India
| | - Sohan S Chitlange
- Department of Pharmaceutical Chemistry, Dr. D. Y. Patil Institute of Pharmaceutical Science and Research, affiliated to SPPU, Pune, India
| |
Collapse
|
3
|
Su J, Xu J, Hu S, Ye H, Xie L, Ouyang S. Advances in small-molecule insulin secretagogues for diabetes treatment. Biomed Pharmacother 2024; 178:117179. [PMID: 39059347 DOI: 10.1016/j.biopha.2024.117179] [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: 05/30/2024] [Revised: 07/16/2024] [Accepted: 07/22/2024] [Indexed: 07/28/2024] Open
Abstract
Diabetes, a metabolic disease caused by abnormally high levels of blood glucose, has a high prevalence rate worldwide and causes a series of complications, including coronary heart disease, stroke, peripheral vascular disease, end-stage renal disease, and retinopathy. Small-molecule compounds have been developed as drugs for the treatment of diabetes because of their oral advantages. Insulin secretagogues are a class of small-molecule drugs used to treat diabetes, and include sulfonylureas, non-sulfonylureas, glucagon-like peptide-1 receptor agonists, dipeptidyl peptidase 4 inhibitors, and other novel small-molecule insulin secretagogues. However, many small-molecule compounds cause different side effects, posing huge challenges to drug monotherapy and drug selection. Therefore, the use of different small-molecule drugs must be improved. This article reviews the mechanism, advantages, limitations, and potential risks of small-molecule insulin secretagogues to provide future research directions on small-molecule drugs for the treatment of diabetes.
Collapse
Affiliation(s)
- Jingqian Su
- Key Laboratory of Microbial Pathogenesis and Interventions of Fujian Province University, Key Laboratory of Innate Immune Biology of Fujian Province, Biomedical Research Center of South China, Key Laboratory of OptoElectronic Science and Technology for Medicine of the Ministry of Education, College of Life Sciences, Fujian Normal University, Fuzhou 350117, China.
| | - Jingran Xu
- Key Laboratory of Microbial Pathogenesis and Interventions of Fujian Province University, Key Laboratory of Innate Immune Biology of Fujian Province, Biomedical Research Center of South China, Key Laboratory of OptoElectronic Science and Technology for Medicine of the Ministry of Education, College of Life Sciences, Fujian Normal University, Fuzhou 350117, China
| | - Shan Hu
- Key Laboratory of Microbial Pathogenesis and Interventions of Fujian Province University, Key Laboratory of Innate Immune Biology of Fujian Province, Biomedical Research Center of South China, Key Laboratory of OptoElectronic Science and Technology for Medicine of the Ministry of Education, College of Life Sciences, Fujian Normal University, Fuzhou 350117, China
| | - Hui Ye
- Key Laboratory of Microbial Pathogenesis and Interventions of Fujian Province University, Key Laboratory of Innate Immune Biology of Fujian Province, Biomedical Research Center of South China, Key Laboratory of OptoElectronic Science and Technology for Medicine of the Ministry of Education, College of Life Sciences, Fujian Normal University, Fuzhou 350117, China
| | - Lian Xie
- Key Laboratory of Microbial Pathogenesis and Interventions of Fujian Province University, Key Laboratory of Innate Immune Biology of Fujian Province, Biomedical Research Center of South China, Key Laboratory of OptoElectronic Science and Technology for Medicine of the Ministry of Education, College of Life Sciences, Fujian Normal University, Fuzhou 350117, China
| | - Songying Ouyang
- Key Laboratory of Microbial Pathogenesis and Interventions of Fujian Province University, Key Laboratory of Innate Immune Biology of Fujian Province, Biomedical Research Center of South China, Key Laboratory of OptoElectronic Science and Technology for Medicine of the Ministry of Education, College of Life Sciences, Fujian Normal University, Fuzhou 350117, China.
| |
Collapse
|
4
|
Borozdina NA, Dyachenko IA, Popkova DV. Promising Directions for Regulating Signaling Pathways Involved in the Type 2 Diabetes Mellitus Development (A Review). RUSSIAN JOURNAL OF BIOORGANIC CHEMISTRY 2024; 50:1263-1284. [DOI: 10.1134/s1068162024040137] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 11/11/2023] [Accepted: 11/12/2023] [Indexed: 01/04/2025]
|
5
|
Ranade SD, Alegaon SG, Khatib NA, Gharge S, Kavalapure RS. Quinoline-based Schiff bases as possible antidiabetic agents: ligand-based pharmacophore modeling, 3D QSAR, docking, and molecular dynamics simulations study. RSC Med Chem 2024:d4md00344f. [PMID: 39149562 PMCID: PMC11322893 DOI: 10.1039/d4md00344f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 07/03/2024] [Indexed: 08/17/2024] Open
Abstract
α-Glucosidase enzyme inhibition is a legitimate approach to combat type 2 diabetes mellitus as it manages to control postprandial hyperglycemia. In this pursuit, a literature search identified quinoline-based molecules as potential α-glucosidase inhibitors. Thus our intended approach is to identify pharmacophoric features responsible for the α-glucosidase inhibition. This was achieved by performing, ligand-based pharmacophore modeling, 3D QSAR model development, pharmacophore-based screening of a rationally designed quinoline-based benzohydrazide Schiff base library, identifying, synthesizing and characterizing molecules (6a-6j) by IR, (1H and 13C) NMR, and mass studies. Further, these molecules were evaluated for α-glucosidase and α-amylase inhibitory potential. Compound 6c was found to inhibit α-glucosidase enzyme with an IC50 value of 12.95 ± 2.35 μM. Similarly, compound 6b was found to have an IC50 value of 19.37 ± 0.96 μM as compared to acarbose (IC50: 32.63 ± 1.07 μM); the inhibitory kinetics of compounds 6b and 6c revealed a competitive type of inhibition; the inhibitory effect can be attributed to its mapped pharmacophoric feature and model validation with a survival score of 5.0697 and vector score of 0.9552. The QSAR model showed a strong correlation with an R 2 value of 0.96. All the compounds (6a-6j) showed no toxicity in L929 cell lines by the MTT assay method. Further, the binding orientation and stability of the molecules were assessed using molecular docking studies and MD trajectory analysis. The energy profile of the molecules with protein as a complex and molecules alone was evaluated using MM/GBSA and DFT calculations, respectively; finally, the pharmacokinetic profile was computed using ADMET analysis.
Collapse
Affiliation(s)
- Shriram D Ranade
- Department of Pharmaceutical Chemistry, KLE College of Pharmacy, Belagavi, KLE Academy of Higher education and Research Belagavi - 590010 Karnataka India
| | - Shankar G Alegaon
- Department of Pharmaceutical Chemistry, KLE College of Pharmacy, Belagavi, KLE Academy of Higher education and Research Belagavi - 590010 Karnataka India
| | - Nayeem A Khatib
- Department of Pharmacology, KLE College of Pharmacy, Belagavi, KLE Academy of Higher education and Research Belagavi - 590010 Karnataka India
| | - Shankar Gharge
- Department of Pharmaceutical Chemistry, KLE College of Pharmacy, Belagavi, KLE Academy of Higher education and Research Belagavi - 590010 Karnataka India
| | - Rohini S Kavalapure
- Department of Pharmaceutical Chemistry, KLE College of Pharmacy, Belagavi, KLE Academy of Higher education and Research Belagavi - 590010 Karnataka India
| |
Collapse
|
6
|
Nag S, Mitra O, Maturi B, Kaur SP, Saini A, Nama M, Roy S, Samanta S, Chacko L, Dutta R, Sayana SB, Subramaniyan V, Bhatti JS, Kandimalla R. Autophagy and mitophagy as potential therapeutic targets in diabetic heart condition: Harnessing the power of nanotheranostics. Asian J Pharm Sci 2024; 19:100927. [PMID: 38948399 PMCID: PMC11214300 DOI: 10.1016/j.ajps.2024.100927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Revised: 03/29/2024] [Accepted: 04/13/2024] [Indexed: 07/02/2024] Open
Abstract
Autophagy and mitophagy pose unresolved challenges in understanding the pathology of diabetic heart condition (DHC), which encompasses a complex range of cardiovascular issues linked to diabetes and associated cardiomyopathies. Despite significant progress in reducing mortality rates from cardiovascular diseases (CVDs), heart failure remains a major cause of increased morbidity among diabetic patients. These cellular processes are essential for maintaining cellular balance and removing damaged or dysfunctional components, and their involvement in the development of diabetic heart disease makes them attractive targets for diagnosis and treatment. While a variety of conventional diagnostic and therapeutic strategies are available, DHC continues to present a significant challenge. Point-of-care diagnostics, supported by nanobiosensing techniques, offer a promising alternative for these complex scenarios. Although conventional medications have been widely used in DHC patients, they raise several concerns regarding various physiological aspects. Modern medicine places great emphasis on the application of nanotechnology to target autophagy and mitophagy in DHC, offering a promising approach to deliver drugs beyond the limitations of traditional therapies. This article aims to explore the potential connections between autophagy, mitophagy and DHC, while also discussing the promise of nanotechnology-based theranostic interventions that specifically target these molecular pathways.
Collapse
Affiliation(s)
- Sagnik Nag
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, 47500 Bandar Sunway, Selangor, Malaysia
| | - Oishi Mitra
- Department of Bio-Sciences, School of Bio-Sciences & Technology (SBST), Vellore Institute of Technology (VIT), Tiruvalam Road, Vellore 632014, Tamil Nadu, India
| | - Bhanu Maturi
- Department of Internal Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Simran Preet Kaur
- Department of Microbiology, University of Delhi (South Campus), Benito Juarez Road, New Delhi 110021, India
| | - Ankita Saini
- Department of Microbiology, University of Delhi (South Campus), Benito Juarez Road, New Delhi 110021, India
| | - Muskan Nama
- Department of Bio-Sciences, School of Bio-Sciences & Technology (SBST), Vellore Institute of Technology (VIT), Tiruvalam Road, Vellore 632014, Tamil Nadu, India
| | - Soumik Roy
- Department of Biotechnology, Indian Institute of Technology, Hyderabad (IIT-H), Sangareddy, Telangana 502284, India
| | - Souvik Samanta
- Department of Bio-Sciences, School of Bio-Sciences & Technology (SBST), Vellore Institute of Technology (VIT), Tiruvalam Road, Vellore 632014, Tamil Nadu, India
| | - Leena Chacko
- BioAnalytical Lab, Meso Scale Discovery, 1601 Research Blvd, Rockville, MD, USA
| | - Rohan Dutta
- Department of Bio-Sciences, School of Bio-Sciences & Technology (SBST), Vellore Institute of Technology (VIT), Tiruvalam Road, Vellore 632014, Tamil Nadu, India
| | - Suresh Babu Sayana
- Department of Pharmacology, Government Medical College, Suryapet, Telangana, India
| | - Vetriselvan Subramaniyan
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, 47500 Bandar Sunway, Selangor, Malaysia
| | - Jasvinder Singh Bhatti
- Laboratory of Translational Medicine and Nanotherapeutics, Department of Human Genetics and Molecular Medicine, School of Health Sciences, Central University of Punjab, Bathinda, India
| | - Ramesh Kandimalla
- Department of Biochemistry, Kakatiya Medical College, Warangal 506007, India
| |
Collapse
|
7
|
Kazek G, Głuch-Lutwin M, Mordyl B, Menaszek E, Kubacka M, Jurowska A, Cież D, Trzewik B, Szklarzewicz J, Papież MA. Vanadium Complexes with Thioanilide Derivatives of Amino Acids: Inhibition of Human Phosphatases and Specificity in Various Cell Models of Metabolic Disturbances. Pharmaceuticals (Basel) 2024; 17:229. [PMID: 38399444 PMCID: PMC10892041 DOI: 10.3390/ph17020229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 02/05/2024] [Accepted: 02/06/2024] [Indexed: 02/25/2024] Open
Abstract
In the text, the synthesis and characteristics of the novel ONS-type vanadium (V) complexes with thioanilide derivatives of amino acids are described. They showed the inhibition of human protein tyrosine phosphatases (PTP1B, LAR, SHP1, and SHP2) in the submicromolar range, as well as the inhibition of non-tyrosine phosphatases (CDC25A and PPA2) similar to bis(maltolato)oxidovanadium(IV) (BMOV). The ONS complexes increased [14C]-deoxy-D-glucose transport into C2C12 myocytes, and one of them, VC070, also enhanced this transport in 3T3-L1 adipocytes. These complexes inhibited gluconeogenesis in hepatocytes HepG2, but none of them decreased lipid accumulation in the non-alcoholic fatty liver disease model using the same cells. Compared to the tested ONO-type vanadium complexes with 5-bromosalicylaldehyde and substituted benzhydrazides as Schiff base ligand components, the ONS complexes revealed stronger inhibition of protein tyrosine phosphatases, but the ONO complexes showed greater activity in the cell models in general. Moreover, the majority of the active complexes from both groups showed better effects than VOSO4 and BMOV. Complexes from both groups activated AKT and ERK signaling pathways in hepatocytes to a comparable extent. One of the ONO complexes, VC068, showed activity in all of the above models, including also glucose utilizatiand ONO Complexes are Inhibitors ofon in the myocytes and glucose transport in insulin-resistant hepatocytes. The discussion section explicates the results within the wider scope of the knowledge about vanadium complexes.
Collapse
Affiliation(s)
- Grzegorz Kazek
- Department of Pharmacological Screening, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Krakow, Poland
| | - Monika Głuch-Lutwin
- Department of Radioligands, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Krakow, Poland
| | - Barbara Mordyl
- Department of Radioligands, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Krakow, Poland
| | - Elżbieta Menaszek
- Department of Cytobiology, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Krakow, Poland
| | - Monika Kubacka
- Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Krakow, Poland
| | - Anna Jurowska
- Coordination Chemistry Group, Faculty of Chemistry, Jagiellonian University, Gronostajowa 2, 30-387 Krakow, Poland
| | - Dariusz Cież
- Department of Organic Chemistry, Faculty of Chemistry, Jagiellonian University, Gronostajowa 2, 30-387 Krakow, Poland
| | - Bartosz Trzewik
- Department of Organic Chemistry, Faculty of Chemistry, Jagiellonian University, Gronostajowa 2, 30-387 Krakow, Poland
| | - Janusz Szklarzewicz
- Coordination Chemistry Group, Faculty of Chemistry, Jagiellonian University, Gronostajowa 2, 30-387 Krakow, Poland
| | - Monika A Papież
- Department of Cytobiology, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Krakow, Poland
| |
Collapse
|
8
|
Mkabayi L, Viljoen Z, Krause RW, Lobb KA, Pletschke BI, Frost CL. Inhibitory effects of selected cannabinoids against dipeptidyl peptidase IV, an enzyme linked to type 2 diabetes. Heliyon 2024; 10:e23289. [PMID: 38169946 PMCID: PMC10758829 DOI: 10.1016/j.heliyon.2023.e23289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Revised: 11/30/2023] [Accepted: 11/30/2023] [Indexed: 01/05/2024] Open
Abstract
Ethnopharmacological relevance In recent times the decriminalisation of cannabis globally has increased its use as an alternative medication. Where it has been used in modern medicinal practises since the 1800s, there is limited scientific investigation to understand the biological activities of this plant. Aim of the study Dipeptidyl peptidase IV (DPP-IV) plays a key role in regulating glucose homeostasis, and inhibition of this enzyme has been used as a therapeutic approach to treat type 2 diabetes. However, some of the synthetic inhibitors for this enzyme available on the market may cause undesirable side effects. Therefore, it is important to identify new inhibitors of DPP-IV and to understand their interaction with this enzyme. Methods In this study, four cannabinoids (cannabidiol, cannabigerol, cannabinol and Δ9-tetrahydrocannabinol) were evaluated for their inhibitory effects against recombinant human DPP-IV and their potential inhibition mechanism was explored using both in vitro and in silico approaches. Results All four cannabinoids resulted in a dose-dependent response with IC50 values of between 4.0 and 6.9 μg/mL. Kinetic analysis revealed a mixed mode of inhibition. CD spectra indicated that binding of cannabinoids results in structural and conformational changes in the secondary structure of the enzyme. These findings were supported by molecular docking studies which revealed best docking scores at both active and allosteric sites for all tested inhibitors. Furthermore, molecular dynamics simulations showed that cannabinoids formed a stable complex with DPP-IV protein via hydrogen bonds at an allosteric site, suggesting that cannabinoids act by either inducing conformational changes or blocking the active site of the enzyme. Conclusion These results demonstrated that cannabinoids may modulate DPP-IV activity and thereby potentially assist in improving glycaemic regulation in type 2 diabetes.
Collapse
Affiliation(s)
- Lithalethu Mkabayi
- Department of Biochemistry and Microbiology, Rhodes University, Makhanda, 6140, South Africa
| | - Zenobia Viljoen
- Department of Biochemistry and Microbiology, Nelson Mandela University, Port Elizabeth, 6031, South Africa
| | - Rui W.M. Krause
- Department of Chemistry, Rhodes University, Makhanda, 6140, South Africa
| | - Kevin A. Lobb
- Department of Chemistry, Rhodes University, Makhanda, 6140, South Africa
| | - Brett I. Pletschke
- Department of Biochemistry and Microbiology, Rhodes University, Makhanda, 6140, South Africa
| | - Carminita L. Frost
- Department of Biochemistry and Microbiology, Nelson Mandela University, Port Elizabeth, 6031, South Africa
| |
Collapse
|
9
|
Huang L, Cao B, Geng Y, Zhou X, Yang Y, Ma T, Lin H, Huang Z, Zhuo L, Li J. A randomized double-blind phase Ib clinical trial of SY-009 in patients with type 2 diabetes mellitus. Eur J Pharm Sci 2024; 192:106644. [PMID: 37981049 DOI: 10.1016/j.ejps.2023.106644] [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: 11/11/2022] [Revised: 10/18/2023] [Accepted: 11/16/2023] [Indexed: 11/21/2023]
Abstract
INTRODUCTION SY-009 produces a hypoglycemic effect via inhibiting sodium/glucose cotransporter 1 (SGLT1) in type 2 diabetes mellitus (T2DM) patients. This randomized, double-blind, placebo-controlled, and multiple-dose escalation clinical trial aimed to evaluate the pharmacokinetic and pharmacodynamical characteristics as well as the safety and tolerability of SY-009 in T2DM patients. METHOD Fifty T2DM patients were randomized into experimental and placebo groups, and hospitalized for 9 days managed with a unified diet and rest management. Subjects were given SY-009 or placebo from day 1 to day 7 at different frequencies and dosages. Single dose cohort was defined as the first dose on day 1 and multiple dose cohort included all the dose from day 1 to 7. Blood samples were collected for pharmacokinetic analysis. Mixed meal tolerance tests were performed. Blood samples were collected to determine glucose, C-peptide, insulin, glucagon-like peptide-1 (GLP-1), and gastric inhibitory polypeptide (GIP). RESULTS PK parameters were not obtained because blood SY-009 concentrations were below the limit of quantitation in all subjects. SY-009 decreased the postprandial glucose. Blood glucose was controlled within 4 hours after taking the drug. Short-term administration of SY-009 (7 days) had no significant effects on fasting glucose but reduced the secretion of C-peptide, insulin, and GIP and increased GLP-1 secretion. The most common adverse event was gastrointestinal disorder manifesting abdominal pain, diarrhea, and bloating. CONCLUSION Plasma exposure of SY-009 and its metabolites was fairly low in T2DM patients at doses of 1.0-4.0 mg. SY-009 reduced postprandial glucose, C-peptide, and insulin levels, showing relative safety and tolerability in the dose range of 1.0-4.0 mg. TRIALS REGISTRATION ClinicalTrials.gov Identifier: NCT04345107.
Collapse
Affiliation(s)
- Lei Huang
- Phase I Clinical Trials Unit, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Bei Cao
- Phase I Clinical Trials Unit, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Yan Geng
- Phase I Clinical Trials Unit, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Xiaoli Zhou
- Suzhou Yabao Pharmaceutical R&D Co., Ltd., Suzhou 215000, China
| | - Yuanxun Yang
- Phase I Clinical Trials Unit, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Tingting Ma
- Phase I Clinical Trials Unit, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Hui Lin
- Phase I Clinical Trials Unit, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Zhijiang Huang
- Suzhou Yabao Pharmaceutical R&D Co., Ltd., Suzhou 215000, China
| | - Lang Zhuo
- Suzhou Yabao Pharmaceutical R&D Co., Ltd., Suzhou 215000, China.
| | - Juan Li
- Phase I Clinical Trials Unit, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China.
| |
Collapse
|
10
|
Sun HY, Lin XY. Analysis of the management and therapeutic performance of diabetes mellitus employing special target. World J Diabetes 2023; 14:1721-1737. [PMID: 38222785 PMCID: PMC10784800 DOI: 10.4239/wjd.v14.i12.1721] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 08/31/2023] [Accepted: 10/23/2023] [Indexed: 12/14/2023] Open
Abstract
Diabetes mellitus (DM) is a chronic metabolic condition characterized predominantly by hyperglycemia. The most common causes contributing to the pathophysiology of diabetes are insufficient insulin secretion, resistance to insulin's tissue-acting effects, or a combination of both. Over the last 30 years, the global prevalence of diabetes increased from 4% to 6.4%. If no better treatment or cure is found, this amount might climb to 430 million in the coming years. The major factors of the disease's deterioration include age, obesity, and a sedentary lifestyle. Finding new therapies to manage diabetes safely and effectively without jeopardizing patient compliance has always been essential. Among the medications available to manage DM on this journey are glucagon-like peptide-1 agonists, thiazolidinediones, sulphonyl urease, glinides, biguanides, and insulin-targeting receptors discovered more than 10 years ago. Despite the extensive preliminary studies, a few clinical observations suggest this process is still in its early stages. The present review focuses on targets that contribute to insulin regulation and may be employed as targets in treating diabetes since they may be more efficient and secure than current and traditional treatments.
Collapse
Affiliation(s)
- Hong-Yan Sun
- Department of Endocrine and Metabolic Diseases, Yantaishan Hospital, Yantai 264003, Shandong Province, China
| | - Xiao-Yan Lin
- Department of Endocrine and Metabolic Diseases, Yantaishan Hospital, Yantai 264003, Shandong Province, China
| |
Collapse
|
11
|
Soileau LG, Nguyen A, Senthil A, Boullion JA, Talbot NC, Ahmadzadeh S, Shekoohi S, Kaye AD, Varrassi G. Bromocriptine and Colesevelam Hydrochloride: Novel Therapies for Type II Diabetes Mellitus. Cureus 2023; 15:e50138. [PMID: 38192911 PMCID: PMC10771968 DOI: 10.7759/cureus.50138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 12/07/2023] [Indexed: 01/10/2024] Open
Abstract
The increasing prevalence of type II diabetes mellitus (T2DM) is a worldwide healthcare concern. Over the years, our understanding of T2DM has grown considerably in uncovering the pathophysiology of the disease and, in turn, understanding how improved treatment methods can be used to slow disease progression. Some long-term complications that are responsible for most T2DM mortalities include cardiovascular disease, neurological decline, and renal failure. In treating T2DM, it is important that not only glycemic control be obtained but also control of associated complications. Bromocriptine and colesevelam hydrochloride have both been approved by the Food and Drug Administration (FDA) to treat T2DM but are not readily used in practice. These medications are known to treat glycemic dysregulation via unconventional mechanisms, which might contribute to their potential to provide protection against common diabetic complications such as cardiovascular disease. In order to ensure that these overlooked medications become more readily used, it is vital that more research be performed to further elucidate their efficacy in a clinical setting. Future studies should continue to provide clinicians a better understanding of the role these medications have on the treatment of T2DM such as their ability to be used in combination with other commonly used T2DM medications or as monotherapies.
Collapse
Affiliation(s)
- Lenise G Soileau
- School of Medicine, Louisiana State University Health Sciences Center, Shreveport, USA
| | - Angela Nguyen
- School of Medicine, Louisiana State University Health Sciences Center, Shreveport, USA
| | - Aarthi Senthil
- School of Medicine, Louisiana State University Health Sciences Center, Shreveport, USA
| | - Jolie A Boullion
- School of Medicine, Louisiana State University Health Sciences Center, Shreveport, USA
| | - Norris C Talbot
- School of Medicine, Louisiana State University Health Sciences Center, Shreveport, USA
| | - Shahab Ahmadzadeh
- Department of Anesthesiology, Louisiana State University Health Sciences Center, Shreveport, USA
| | - Sahar Shekoohi
- Department of Anesthesiology, Louisiana State University Health Sciences Center, Shreveport, USA
| | - Alan D Kaye
- Department of Anesthesiology, Louisiana State University Health Sciences Center, Shreveport, USA
| | | |
Collapse
|
12
|
Li B, Li X, Zeng Y, Zhou Z, Zhao D, Qin F, Zhang B, Yao W, Mao Y, Zhou L, Li K, Zhu Q, Rong X, Guo J. Network pharmacology combined with molecular docking and experimental verification to elucidate functional mechanism of Fufang Zhenzhu Tiaozhi against type 2 diabetes mellitus. J Biomol Struct Dyn 2023; 42:13751-13767. [PMID: 37942992 DOI: 10.1080/07391102.2023.2278082] [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: 04/24/2023] [Accepted: 09/29/2023] [Indexed: 11/10/2023]
Abstract
Fufang Zhenzhu Tiaozhi (FTZ) capsules have been prescribed for treating glucose and lipid metabolism disorders such as type 2 diabetes mellitus (T2DM). However, the underlying mechanism remains unknown. In this study, network pharmacology and experimental verification were combined to investigate the mechanisms of FTZ in treating T2DM. A total of 176 active ingredients and 1169 corresponding targets were screened using biological databases. 598 potential targets of T2DM were retrieved from GeneCards, PharmGKB, OMIM, Drugbank, and TTD. The Venn diagram was employed to identify the 194 intersection targets, which were employed to construct the "Herb-Compound-Target" interacting networks. These common targets were also used to prepare a protein-protein interaction (PPI) network to uncover potential targets. The four core targets were docked to their corresponding targets for binding analysis. Additionally, the top-ranked poses of ingredients and the positive compounds from each protein were evaluated for stability using molecular dynamics. Our results suggest that core active ingredients such as kaempferol, luteolin, and baicalein have high binding affinity and stability with AKT1, PTGS2 (also known as COX-2), DPP4, and PAPRG. GO and KEGG analyses indicated that the treatment T2DM by FTZ might be related to different pathway like AMPK and EGFR pathways. The experimental validation results proved that kaempferol, luteolin, and baicalein could significantly inhibit the activity of DPP4 and COX-2, kaempferol and luteolin were also able to activate AKT and AMPK signaling pathway. This study further validated previous findings and enhanced our understanding of the potential effects of FTZ on T2DM.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Bo Li
- Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
- Guangdong Metabolic Diseases Research, Center of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou, China
- Ministry of Education, Key Laboratory of Glucolipid Metabolic Disorder, Guangdong Pharmaceutical University, Guangzhou, China
- Guangdong TCM Key Laboratory for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou, China
| | - Xinying Li
- Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
- Guangdong Metabolic Diseases Research, Center of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou, China
- Ministry of Education, Key Laboratory of Glucolipid Metabolic Disorder, Guangdong Pharmaceutical University, Guangzhou, China
- Guangdong TCM Key Laboratory for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou, China
| | - Youyan Zeng
- Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
- Guangdong Metabolic Diseases Research, Center of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou, China
- Ministry of Education, Key Laboratory of Glucolipid Metabolic Disorder, Guangdong Pharmaceutical University, Guangzhou, China
- Guangdong TCM Key Laboratory for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou, China
| | - Zhenhua Zhou
- Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
- Guangdong Metabolic Diseases Research, Center of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou, China
- Ministry of Education, Key Laboratory of Glucolipid Metabolic Disorder, Guangdong Pharmaceutical University, Guangzhou, China
- Guangdong TCM Key Laboratory for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou, China
| | - Dongyu Zhao
- Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
- Guangdong Metabolic Diseases Research, Center of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou, China
- Ministry of Education, Key Laboratory of Glucolipid Metabolic Disorder, Guangdong Pharmaceutical University, Guangzhou, China
- Guangdong TCM Key Laboratory for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou, China
| | - Fei Qin
- Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
- Guangdong Metabolic Diseases Research, Center of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou, China
- Ministry of Education, Key Laboratory of Glucolipid Metabolic Disorder, Guangdong Pharmaceutical University, Guangzhou, China
- Guangdong TCM Key Laboratory for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou, China
| | - Bin Zhang
- Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
- Guangdong Metabolic Diseases Research, Center of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou, China
- Ministry of Education, Key Laboratory of Glucolipid Metabolic Disorder, Guangdong Pharmaceutical University, Guangzhou, China
- Guangdong TCM Key Laboratory for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou, China
| | - Weiwei Yao
- Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
- Guangdong Metabolic Diseases Research, Center of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou, China
- Ministry of Education, Key Laboratory of Glucolipid Metabolic Disorder, Guangdong Pharmaceutical University, Guangzhou, China
- Guangdong TCM Key Laboratory for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou, China
| | - Yongxin Mao
- Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
- Guangdong Metabolic Diseases Research, Center of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou, China
- Ministry of Education, Key Laboratory of Glucolipid Metabolic Disorder, Guangdong Pharmaceutical University, Guangzhou, China
- Guangdong TCM Key Laboratory for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou, China
| | - Li Zhou
- Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
- Guangdong Metabolic Diseases Research, Center of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou, China
- Ministry of Education, Key Laboratory of Glucolipid Metabolic Disorder, Guangdong Pharmaceutical University, Guangzhou, China
- Guangdong TCM Key Laboratory for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou, China
| | - Kunping Li
- Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
- Guangdong Metabolic Diseases Research, Center of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou, China
- Ministry of Education, Key Laboratory of Glucolipid Metabolic Disorder, Guangdong Pharmaceutical University, Guangzhou, China
- Guangdong TCM Key Laboratory for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou, China
| | - Qing Zhu
- Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
- Guangdong Metabolic Diseases Research, Center of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou, China
- Ministry of Education, Key Laboratory of Glucolipid Metabolic Disorder, Guangdong Pharmaceutical University, Guangzhou, China
- Guangdong TCM Key Laboratory for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou, China
| | - Xianglu Rong
- Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
- Guangdong Metabolic Diseases Research, Center of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou, China
- Ministry of Education, Key Laboratory of Glucolipid Metabolic Disorder, Guangdong Pharmaceutical University, Guangzhou, China
- Guangdong TCM Key Laboratory for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou, China
| | - Jiao Guo
- Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
- Guangdong Metabolic Diseases Research, Center of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou, China
- Ministry of Education, Key Laboratory of Glucolipid Metabolic Disorder, Guangdong Pharmaceutical University, Guangzhou, China
- Guangdong TCM Key Laboratory for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou, China
| |
Collapse
|
13
|
Díaz-Rojas M, González-Andrade M, Aguayo-Ortiz R, Rodríguez-Sotres R, Pérez-Vásquez A, Madariaga-Mazón A, Mata R. Discovery of inhibitors of protein tyrosine phosphatase 1B contained in a natural products library from Mexican medicinal plants and fungi using a combination of enzymatic and in silico methods*. Front Pharmacol 2023; 14:1281045. [PMID: 38027024 PMCID: PMC10644722 DOI: 10.3389/fphar.2023.1281045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 10/13/2023] [Indexed: 12/01/2023] Open
Abstract
This work aimed to discover protein tyrosine phosphatase 1B (PTP1B) inhibitors from a small molecule library of natural products (NPs) derived from selected Mexican medicinal plants and fungi to find new hits for developing antidiabetic drugs. The products showing similar IC50 values to ursolic acid (UA) (positive control, IC50 = 26.5) were considered hits. These compounds were canophyllol (1), 5-O-(β-D-glucopyranosyl)-7-methoxy-3',4'-dihydroxy-4-phenylcoumarin (2), 3,4-dimethoxy-2,5-phenanthrenediol (3), masticadienonic acid (4), 4',5,6-trihydroxy-3',7-dimethoxyflavone (5), E/Z vermelhotin (6), tajixanthone hydrate (7), quercetin-3-O-(6″-benzoyl)-β-D-galactoside (8), lichexanthone (9), melianodiol (10), and confusarin (11). According to the double-reciprocal plots, 1 was a non-competitive inhibitor, 3 a mixed-type, and 6 competitive. The chemical space analysis of the hits (IC50 < 100 μM) and compounds possessing activity (IC50 in the range of 100-1,000 μM) with the BIOFACQUIM library indicated that the active molecules are chemically diverse, covering most of the known Mexican NPs' chemical space. Finally, a structure-activity similarity (SAS) map was built using the Tanimoto similarity index and PTP1B absolute inhibitory activity, which allows the identification of seven scaffold hops, namely, compounds 3, 5, 6, 7, 8, 9, and 11. Canophyllol (1), on the other hand, is a true analog of UA since it is an SAR continuous zone of the SAS map.
Collapse
Affiliation(s)
- Miriam Díaz-Rojas
- Facultad de Química, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | | | - Rodrigo Aguayo-Ortiz
- Facultad de Química, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | | | | | - Abraham Madariaga-Mazón
- Instituto de Química Unidad Mérida and Instituto de Investigaciones en Matemáticas Aplicadas y en Sistemas Unidad Mérida, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Rachel Mata
- Facultad de Química, Universidad Nacional Autónoma de México, Mexico City, Mexico
| |
Collapse
|
14
|
Kazempour-Dizaji M, Mojtabavi S, Sadri A, Ghanbarpour A, Faramarzi MA, Navidpour L. Arylureidoaurones: Synthesis, in vitro α-glucosidase, and α-amylase inhibition activity. Bioorg Chem 2023; 139:106709. [PMID: 37442042 DOI: 10.1016/j.bioorg.2023.106709] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 06/14/2023] [Accepted: 06/28/2023] [Indexed: 07/15/2023]
Abstract
Because of the colossal global burden of diabetes, there is an urgent need for more effective and safer drugs. We designed and synthesized a new series of aurone derivatives possessing phenylureido or bis-phenylureido moieties as α-glucosidase and α-amylase inhibitors. Most of the synthesized phenylureidoaurones have demonstrated superior inhibition activities (IC50s of 9.6-339.9 μM) against α-glucosidase relative to acarbose (IC50 = 750.0 μM) as the reference drug. Substitution of aurone analogues with two phenylureido substituents at the 5-position of the benzofuranone moiety and the 3' or 4' positions of the 2-phenyl ring resulted in compounds with almost 120-180 times more potent inhibitory activities than acarbose. The aurone analogue possessing two phenylureido substitutions at 5 and 4' positions (13) showed the highest inhibition activity with an IC50 of 4.2 ± 0.1 μM. Kinetic studies suggested their inhibition mode to be competitive. We also investigated the binding mode of the most potent compounds using the consensually docked 4D-QSAR methodology. Furthermore, these analogues showed weak-to-moderate non-competitive inhibitory activity against α-amylase. 5-Methyl substituted aurone with 4'-phenylureido moiety (6e) demonstrated the highest inhibition activity on α-amylase with an IC50 of 142.0 ± 1.6 μM relative to acarbose (IC50 = 108 ± 1.2 μM). Our computational studies suggested that these analogues interact with a hydrophilic allosteric site in α-amylase, located far from the enzyme active site at the N-terminal.
Collapse
Affiliation(s)
- Mohammad Kazempour-Dizaji
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran 14176, Iran
| | - Somayeh Mojtabavi
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, P.O. Box 14155-6451, Tehran 14176, Iran
| | - Arash Sadri
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran 14176, Iran; Interdisciplinary Neuroscience Research Program, Students' Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran; Lyceum Scientific Charity, Iran
| | - Araz Ghanbarpour
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran 14176, Iran
| | - Mohammad Ali Faramarzi
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, P.O. Box 14155-6451, Tehran 14176, Iran
| | - Latifeh Navidpour
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran 14176, Iran.
| |
Collapse
|
15
|
Casertano M, Vito A, Aiello A, Imperatore C, Menna M. Natural Bioactive Compounds from Marine Invertebrates That Modulate Key Targets Implicated in the Onset of Type 2 Diabetes Mellitus (T2DM) and Its Complications. Pharmaceutics 2023; 15:2321. [PMID: 37765290 PMCID: PMC10538088 DOI: 10.3390/pharmaceutics15092321] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 08/24/2023] [Accepted: 09/13/2023] [Indexed: 09/29/2023] Open
Abstract
BACKGROUND Type 2 diabetes mellitus (T2DM) is an ongoing, risky, and costly health problem that therefore always requires new treatment options. Moreover, although several drugs are available, only 36% of patients achieve glycaemic control, and patient adherence is a major obstacle. With monotherapy, T2DM and its comorbidities/complications often cannot be managed, and the concurrent administration of several hypoglycaemic drugs is required, which increases the risk of side effects. In fact, despite the efficacy of the drugs currently on the market, they generally come with serious side effects. Therefore, scientific research must always be active in the discovery of new therapeutic agents. DISCUSSION The present review highlights some of the recent discoveries regarding marine natural products that can modulate the various targets that have been identified as crucial in the establishment of T2DM disease and its complications, with a focus on the compounds isolated from marine invertebrates. The activities of these metabolites are illustrated and discussed. OBJECTIVES The paper aims to capture the relevant evidence of the great chemical diversity of marine natural products as a key tool that can advance understanding in the T2DM research field, as well as in antidiabetic drug discovery. The variety of chemical scaffolds highlighted by the natural hits provides not only a source of chemical probes for the study of specific targets involved in the onset of T2DM, but is also a helpful tool for the development of drugs that are capable of acting via novel mechanisms. Thus, it lays the foundation for the design of multiple ligands that can overcome the drawbacks of polypharmacology.
Collapse
Affiliation(s)
| | | | | | | | - Marialuisa Menna
- Department of Pharmacy, University of Naples “Federico II”, Via D. Montesano 49, 80131 Napoli, Italy; (M.C.); (A.V.); (A.A.); (C.I.)
| |
Collapse
|
16
|
Álvarez-Almazán S, Solís-Domínguez LC, Duperou-Luna P, Fuerte-Gómez T, González-Andrade M, Aranda-Barradas ME, Palacios-Espinosa JF, Pérez-Villanueva J, Matadamas-Martínez F, Miranda-Castro SP, Mercado-Márquez C, Cortés-Benítez F. Anti-Diabetic Activity of Glycyrrhetinic Acid Derivatives FC-114 and FC-122: Scale-Up, In Silico, In Vitro, and In Vivo Studies. Int J Mol Sci 2023; 24:12812. [PMID: 37628991 PMCID: PMC10454726 DOI: 10.3390/ijms241612812] [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: 06/30/2023] [Revised: 08/07/2023] [Accepted: 08/12/2023] [Indexed: 08/27/2023] Open
Abstract
Type 2 diabetes (T2D) is one of the most common diseases and the 8th leading cause of death worldwide. Individuals with T2D are at risk for several health complications that reduce their life expectancy and quality of life. Although several drugs for treating T2D are currently available, many of them have reported side effects ranging from mild to severe. In this work, we present the synthesis in a gram-scale as well as the in silico and in vitro activity of two semisynthetic glycyrrhetinic acid (GA) derivatives (namely FC-114 and FC-122) against Protein Tyrosine Phosphatase 1B (PTP1B) and α-glucosidase enzymes. Furthermore, the in vitro cytotoxicity assay on Human Foreskin fibroblast and the in vivo acute oral toxicity was also conducted. The anti-diabetic activity was determined in streptozotocin-induced diabetic rats after oral administration with FC-114 or FC-122. Results showed that both GA derivatives have potent PTP1B inhibitory activity being FC-122, a dual PTP1B/α-glucosidase inhibitor that could increase insulin sensitivity and reduce intestinal glucose absorption. Molecular docking, molecular dynamics, and enzymatic kinetics studies revealed the inhibition mechanism of FC-122 against α-glucosidase. Both GA derivatives were safe and showed better anti-diabetic activity in vivo than the reference drug acarbose. Moreover, FC-114 improves insulin levels while decreasing LDL and total cholesterol levels without decreasing HDL cholesterol.
Collapse
Affiliation(s)
- Samuel Álvarez-Almazán
- Laboratory of Biotechnology, Unidad de Posgrado, Facultad de Estudios Superiores Cuautitlán Campus 1, Universidad Nacional Autónoma de México, Cuautitlán Izcalli 54740, Mexico; (S.Á.-A.); (L.C.S.-D.); (T.F.-G.); (M.E.A.-B.); (S.P.M.-C.)
| | - Luz Cassandra Solís-Domínguez
- Laboratory of Biotechnology, Unidad de Posgrado, Facultad de Estudios Superiores Cuautitlán Campus 1, Universidad Nacional Autónoma de México, Cuautitlán Izcalli 54740, Mexico; (S.Á.-A.); (L.C.S.-D.); (T.F.-G.); (M.E.A.-B.); (S.P.M.-C.)
| | - Paulina Duperou-Luna
- Laboratory of Synthesis and Isolation of Bioactive Substances, Departamento de Sistemas Biológicos, División de Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana–Xochimilco (UAM–X), Mexico City 04960, Mexico; (P.D.-L.); (J.F.P.-E.); (J.P.-V.); (F.M.-M.)
| | - Teresa Fuerte-Gómez
- Laboratory of Biotechnology, Unidad de Posgrado, Facultad de Estudios Superiores Cuautitlán Campus 1, Universidad Nacional Autónoma de México, Cuautitlán Izcalli 54740, Mexico; (S.Á.-A.); (L.C.S.-D.); (T.F.-G.); (M.E.A.-B.); (S.P.M.-C.)
| | - Martin González-Andrade
- Laboratory of Biosensors and Molecular Modelling, Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico;
| | - María E. Aranda-Barradas
- Laboratory of Biotechnology, Unidad de Posgrado, Facultad de Estudios Superiores Cuautitlán Campus 1, Universidad Nacional Autónoma de México, Cuautitlán Izcalli 54740, Mexico; (S.Á.-A.); (L.C.S.-D.); (T.F.-G.); (M.E.A.-B.); (S.P.M.-C.)
| | - Juan Francisco Palacios-Espinosa
- Laboratory of Synthesis and Isolation of Bioactive Substances, Departamento de Sistemas Biológicos, División de Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana–Xochimilco (UAM–X), Mexico City 04960, Mexico; (P.D.-L.); (J.F.P.-E.); (J.P.-V.); (F.M.-M.)
| | - Jaime Pérez-Villanueva
- Laboratory of Synthesis and Isolation of Bioactive Substances, Departamento de Sistemas Biológicos, División de Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana–Xochimilco (UAM–X), Mexico City 04960, Mexico; (P.D.-L.); (J.F.P.-E.); (J.P.-V.); (F.M.-M.)
| | - Félix Matadamas-Martínez
- Laboratory of Synthesis and Isolation of Bioactive Substances, Departamento de Sistemas Biológicos, División de Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana–Xochimilco (UAM–X), Mexico City 04960, Mexico; (P.D.-L.); (J.F.P.-E.); (J.P.-V.); (F.M.-M.)
| | - Susana Patricia Miranda-Castro
- Laboratory of Biotechnology, Unidad de Posgrado, Facultad de Estudios Superiores Cuautitlán Campus 1, Universidad Nacional Autónoma de México, Cuautitlán Izcalli 54740, Mexico; (S.Á.-A.); (L.C.S.-D.); (T.F.-G.); (M.E.A.-B.); (S.P.M.-C.)
| | - Crisóforo Mercado-Márquez
- Isolation and Animal Facility Unit, Facultad de Estudios Superiores Cuautitlán 28, Universidad Nacional Autónoma de México, Cuautitlán Izcalli 54714, Mexico;
| | - Francisco Cortés-Benítez
- Laboratory of Synthesis and Isolation of Bioactive Substances, Departamento de Sistemas Biológicos, División de Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana–Xochimilco (UAM–X), Mexico City 04960, Mexico; (P.D.-L.); (J.F.P.-E.); (J.P.-V.); (F.M.-M.)
| |
Collapse
|
17
|
Han JY, Park M, Lee HJ. Stevia (Stevia rebaudiana) extract ameliorates insulin resistance by regulating mitochondrial function and oxidative stress in the skeletal muscle of db/db mice. BMC Complement Med Ther 2023; 23:264. [PMID: 37488560 PMCID: PMC10367355 DOI: 10.1186/s12906-023-04033-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 06/09/2023] [Indexed: 07/26/2023] Open
Abstract
BACKGROUND Type 2 diabetes mellitus (T2DM), a growing health problem worldwide, is a metabolic disorder characterized by hyperglycemia due to insulin resistance and defective insulin secretion by pancreatic β-cells. The skeletal muscle is a central organ that consumes most of the insulin-stimulated glucose in the body, and insulin resistance can damage muscles in T2DM. Based on a strong correlation between diabetes and muscles, we investigated the effects of stevia extract (SE) and stevioside (SV) on the skeletal muscle of diabetic db/db mice. METHODS The mice were administered saline, metformin (200 mg/kg/day), SE (200 and 500 mg/kg/day), and SV (40 mg/kg/day) for 35 days. During administration, we checked the levels of fasting blood glucose twice a week and conducted the oral glucose tolerance test (OGTT) and insulin tolerance test (ITT). After administration, we analyzed serum biochemical parameters, triglyceride (TG), total cholesterol (TC), insulin and antioxidant enzymes, and the cross-sectional area of skeletal muscle fibers of db/db mice. Western blots were conducted using the skeletal muscle of mice to examine the effect of SE and SV on protein expression of insulin signaling, mitochondrial function, and oxidative stress. RESULTS SE and SV administration lowered the levels of fasting blood glucose, OGTT, and ITT in db/db mice. The administration also decreased serum levels of TG, TC, and insulin while increasing those of superoxide dismutase (SOD) and glutathione peroxidase (GPx). Interestingly, muscle fiber size was significantly increased in db/db mice treated with SE500 and SV. In the skeletal muscle of db/db mice, SE and SV administration activated insulin signaling by increasing the protein expression of insulin receptor substrate, Akt, and glucose transporter type 4. Furthermore, SE500 administration markedly increased the protein expression of AMP-activated protein kinase-α, sirtuin-1, and peroxisome proliferator-activated receptor-γ coactivator-1α. SV administration significantly reduced oxidative stress by down-regulating the protein expression of 4-hydroxynonenal, heme oxygenase-1, SOD, and GPx. In addition, SE500 and SV administration suppressed the expression of apoptosis-related proteins in the skeletal muscle of db/db mice. CONCLUSION SE and SV administration attenuated hyperglycemia in diabetic mice. Moreover, the administration ameliorated insulin resistance by regulating mitochondrial function and oxidative stress, increasing muscle fiber size. Overall, this study suggests that SE and SV administration may serve as a potential strategy for the treatment of diabetic muscles.
Collapse
Affiliation(s)
- Jin-Young Han
- Institute for Aging and Clinical Nutrition Research, Gachon University, Seongnam-Si, 13120, Gyeonggi-Do, Korea
| | - Miey Park
- Institute for Aging and Clinical Nutrition Research, Gachon University, Seongnam-Si, 13120, Gyeonggi-Do, Korea.
- Department of Food and Nutrition, College of BioNano Technology, Gachon University, Seongnam-Si, 13120, Gyeonggi-Do, Korea.
| | - Hae-Jeung Lee
- Institute for Aging and Clinical Nutrition Research, Gachon University, Seongnam-Si, 13120, Gyeonggi-Do, Korea.
- Department of Food and Nutrition, College of BioNano Technology, Gachon University, Seongnam-Si, 13120, Gyeonggi-Do, Korea.
| |
Collapse
|
18
|
Ali MY, Park SE, Seong SH, Zamponi GW, Jung HA, Choi JS. Ursonic acid from Artemisia montana exerts anti-diabetic effects through anti-glycating properties, and by inhibiting PTP1B and activating the PI3K/Akt signaling pathway in insulin-resistant C2C12 cells. Chem Biol Interact 2023; 376:110452. [PMID: 36933777 DOI: 10.1016/j.cbi.2023.110452] [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: 01/17/2023] [Accepted: 03/13/2023] [Indexed: 03/18/2023]
Abstract
Artemisia is one of the largest genera in the plant family Asteraceae and has long been used in traditional medicine for its antitussive, analgesic, antihypertensive, antitoxic, antiviral, antimalarial, and anti-inflammatory properties. However, the anti-diabetic activity of Artemisia montana has not been broadly studied. The goal of this study was to determine whether extracts of the aerial parts of A. montana and its main constituents inhibit protein tyrosine phosphatase 1B (PTP1B) and α-glucosidase activities. We isolated nine compounds from A. montana including ursonic acid (UNA) and ursolic acid (ULA), which significantly inhibited PTP1B with IC50 values of 11.68 and 8.73 μM, respectively. In addition, UNA showed potent inhibitory activity against α-glucosidase (IC50 = 61.85 μM). Kinetic analysis of PTP1B and α-glucosidase inhibition revealed that UNA was a non-competitive inhibitor of both enzymes. Docking simulations of UNA demonstrated negative binding energies and close proximity to residues in the binding pockets of PTP1B and α-glucosidase. Molecular docking simulations between UNA and human serum albumin (HSA) revealed that UNA binds tightly to all three domains of HSA. Furthermore, UNA significantly inhibited fluorescent AGE formation (IC50 = 4.16 μM) in a glucose-fructose-induced HSA glycation model over the course of four weeks. Additionally, we investigated the molecular mechanisms underlying the anti-diabetic effects of UNA in insulin-resistant C2C12 skeletal muscle cells and discovered that UNA significantly increased glucose uptake and decreased PTP1B expression. Further, UNA increased GLUT-4 expression level by activating the IRS-1/PI3K/Akt/GSK-3 signaling pathway. These findings clearly demonstrate that UNA from A. montana shows great potential for treatment of diabetes and its complications.
Collapse
Affiliation(s)
- Md Yousof Ali
- Department of Clinical Neurosciences, Hotchkiss Brain Institute and Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Se Eun Park
- Department of Food and Life Science, Pukyong National University, Busan, 48513, Republic of Korea
| | - Su Hui Seong
- Department of Food and Life Science, Pukyong National University, Busan, 48513, Republic of Korea; Division of Natural Products Research, Honam National Institute of Biological Resource, Mokpo, 58762, Republic of Korea
| | - Gerald W Zamponi
- Department of Clinical Neurosciences, Hotchkiss Brain Institute and Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Hyun Ah Jung
- Department of Food Science and Human Nutrition, Jeonbuk National University, Jeonju, 54896, Republic of Korea.
| | - Jae Sue Choi
- Department of Food and Life Science, Pukyong National University, Busan, 48513, Republic of Korea.
| |
Collapse
|
19
|
He S, Lim GE. The Application of High-Throughput Approaches in Identifying Novel Therapeutic Targets and Agents to Treat Diabetes. Adv Biol (Weinh) 2023; 7:e2200151. [PMID: 36398493 DOI: 10.1002/adbi.202200151] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 10/04/2022] [Indexed: 11/19/2022]
Abstract
During the past decades, unprecedented progress in technologies has revolutionized traditional research methodologies. Among these, advances in high-throughput drug screening approaches have permitted the rapid identification of potential therapeutic agents from drug libraries that contain thousands or millions of molecules. Moreover, high-throughput-based therapeutic target discovery strategies can comprehensively interrogate relationships between biomolecules (e.g., gene, RNA, and protein) and diseases and significantly increase the authors' knowledge of disease mechanisms. Diabetes is a chronic disease primarily characterized by the incapacity of the body to maintain normoglycemia. The prevalence of diabetes in modern society has become a severe public health issue that threatens the well-being of millions of patients. Although a number of pharmacological treatments are available, there is no permanent cure for diabetes, and discovering novel therapeutic targets and agents continues to be an urgent need. The present review discusses the technical details of high-throughput screening approaches in drug discovery, followed by introducing the applications of such approaches to diabetes research. This review aims to provide an example of the applicability of high-throughput technologies in facilitating different aspects of disease research.
Collapse
Affiliation(s)
- Siyi He
- Department of Medicine, Université de Montréal, Pavillon Roger-Gaudry, 2900 Edouard Montpetit Blvd, Montreal, Québec, H3T 1J4, Canada.,Cardiometabolic Axis, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), 900 rue St Denis, Montreal, Québec, H2X 0A9, Canada
| | - Gareth E Lim
- Department of Medicine, Université de Montréal, Pavillon Roger-Gaudry, 2900 Edouard Montpetit Blvd, Montreal, Québec, H3T 1J4, Canada.,Cardiometabolic Axis, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), 900 rue St Denis, Montreal, Québec, H2X 0A9, Canada
| |
Collapse
|
20
|
Anton IC, Mititelu-Tartau L, Popa EG, Poroch M, Poroch V, Pintilei DR, Botnariu GE. Clinical Parameters Affecting the Therapeutic Efficacy of SGLT-2-Comparative Effectiveness and Safety of Dapagliflozin and Empagliflozin in Patients with Type 2 Diabetes. Healthcare (Basel) 2022; 10:1153. [PMID: 35885680 PMCID: PMC9316348 DOI: 10.3390/healthcare10071153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 06/08/2022] [Accepted: 06/16/2022] [Indexed: 11/30/2022] Open
Abstract
(1) Background. We aimed to assess long-term efficacy and safety in inadequately controlled type 2 diabetes (T2DM) of two SGLT-2 inhibitors: empagliflozin (Empa) and dapagliflozin (Dapa), combined with metformin, other oral antidiabetics or insulin, according to the protocols in Romania. (2) Methods. The data of 100 patients treated for T2DM with associated dyslipidemia and/or cardiovascular diseases at the University Hospital and Consultmed Medical Center in Iasi were retrospectively reviewed (2017-2021). In total, 48 patients had received dapagliflozin (10 mg with oral antidiabetics or insulin) and 52 patients received empagliflozin (10 mg /25 mg with oral antidiabetics). (3) Results. In both groups, the lowering of BMI was significant: Dapa group (32.04 ± 4.49 vs. 31.40 ± 4.18 kg/m2; p = 0.006), and Empa group (34.16 ± 5.08 vs. 33.17 ± 4.99 kg/m2; p = 0.002). Blood sugar average levels decreased significantly (170 vs. 136 mg/dL; p = 0.001 for Dapa; 163 vs. 140 mg/dL; p = 0.002 for Empa) and also average levels of HbA1c (7.90% vs. 7.51%; p = 0,01 for Dapa; 7.72% vs. 7.35%; p = 0.004 for Empa). (4) Conclusions. Better results in all variables were observed in younger male patients with a shorter duration of diabetes and threshold BMI levels of 34.1, treated with SGLT2, and more significantly with Empa.
Collapse
Affiliation(s)
- Irina Claudia Anton
- Department of Pharmacology, Clinical Pharmacology and Algesiology, Faculty of Medicine, ‘Grigore T. Popa’ University of Medicine and Pharmacy, Universitatii St. 16, 700115 Iasi, Romania;
| | - Liliana Mititelu-Tartau
- Department of Pharmacology, Clinical Pharmacology and Algesiology, Faculty of Medicine, ‘Grigore T. Popa’ University of Medicine and Pharmacy, Universitatii St. 16, 700115 Iasi, Romania;
| | - Eliza Gratiela Popa
- Department of Pharmaceutical Technology, Faculty of Pharmacy, ‘Grigore T. Popa’ University of Medicine and Pharmacy, Universitatii St. 16, 700115 Iasi, Romania
| | - Mihaela Poroch
- Department of Family Medicine, Preventive Medicine and Interdisciplinarity, Faculty of Medicine, ‘Grigore T. Popa’ University of Medicine and Pharmacy, Universitatii St. 16, 700115 Iasi, Romania;
| | - Vladimir Poroch
- 2nd Department of Internal Medicine, Faculty of Medicine, ‘Grigore T. Popa’ University of Medicine and Pharmacy, Universitatii St. 16, 700115 Iasi, Romania;
| | - Delia Reurean Pintilei
- Department of Diabetes, Nutrition and Metabolic Disease, Consultmed Medical Center, Pacurari St. 70, 700544 Iasi, Romania;
| | - Gina Eosefina Botnariu
- Department of Diabetes, Nutrition and Metabolic Disease, ‘Grigore T. Popa’ University of Medicine and Pharmacy, Universitatii St. 16, 700115 Iasi, Romania;
| |
Collapse
|