1
|
Felício Macarini A, Bolda Mariano LN, de Cássia Vilhena da Silva R, Corrêa R, de Souza P. Involvement of Cholinergic and Cyclooxygenase Pathways in the Diuretic Effects of Rosmarinic Acid. Chem Biodivers 2025; 22:e202401634. [PMID: 39417798 DOI: 10.1002/cbdv.202401634] [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: 07/06/2024] [Revised: 10/04/2024] [Accepted: 10/17/2024] [Indexed: 10/19/2024]
Abstract
Rosmarinic acid (RA) is a natural antioxidant known for its diverse biological activities. Although its diuretic activity has been previously established, the mode of action remained unclear. To investigate this, we examined the diuretic activity of RA alone and in combination with hydrochlorothiazide (HCTZ), amiloride (AML), atropine (ATR), and indomethacin (INDO) to see if any of these drugs could interfere with RA's activity in an 8 hour acute diuresis animal model. We observed that RA increases urine excretion and may have a synergistic effect in the HCTZ+RA group, with a potassium-sparing capacity. In the AML+RA group, we also noted increased urine excretion while sodium and potassium excretion decreased. ATR and INDO prevented RA from increasing urine excretion, suggesting a potential interaction with muscarinic receptors or a role in promoting prostaglandin production. Additionally, molecular docking analysis indicated possible interactions with key receptors involved in increased diuresis or free-electrolyte water excretion.
Collapse
Affiliation(s)
- Anelise Felício Macarini
- Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade do Vale do Itajaí, 88302-901 -, Itajaí, Santa Catarina, Brasil
| | - Luísa Nathalia Bolda Mariano
- Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade do Vale do Itajaí, 88302-901 -, Itajaí, Santa Catarina, Brasil
| | - Rita de Cássia Vilhena da Silva
- Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade do Vale do Itajaí, 88302-901 -, Itajaí, Santa Catarina, Brasil
| | - Rogério Corrêa
- Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade do Vale do Itajaí, 88302-901 -, Itajaí, Santa Catarina, Brasil
| | - Priscila de Souza
- Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade do Vale do Itajaí, 88302-901 -, Itajaí, Santa Catarina, Brasil
| |
Collapse
|
2
|
Iqbal T, Khan S, Hussain R, Khan Y, Shoaib K, Rozeena, Saeed M, Darwish HW. Novel indole based fused triazole-thiadiazole derivatives as anti-diabetic agents: in vitro and in silico approaches. Future Med Chem 2024; 16:2475-2486. [PMID: 39508404 PMCID: PMC11622810 DOI: 10.1080/17568919.2024.2419355] [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: 04/26/2024] [Accepted: 09/26/2024] [Indexed: 11/15/2024] Open
Abstract
Aim: The current research presents novel library of indole derived fused triazole-thiadiazole derivatives (1-17) for treatment of diabetes mellitus.Methods & results: These compounds were synthesized by treating 2-(1H-indol-3-yl)acetic acid with hydrazinecarbothiohydrazide followed by treating the resultant compound with substituted benzoic acid. Structural validation was achieved spectroscopically (1HNMR, 13CNMR and HREI-MS). The synthesized compounds were subjected to biological evaluation to assess their potential as anti-diabetic. Molecular docking study was employed to investigate the binding interactions of these analogs with relevant proteins. ADMET analysis was used to predict their drug-like properties. Notably, compound-10 (IC50 = 1.27 ± 1.25 and 2.18 ± 2.45 μM) bearing para-substituted F atom exhibited the highest potency due to strong inhibitory interactions through hydrogen bonding.Conclusion: This study identifies promising compounds with anti-diabetic activity, paving the way for the treatment of diabetes mellitus.
Collapse
Affiliation(s)
- Tayyiaba Iqbal
- Department of Chemistry, Abbottabad University of Science & Technology, Abbottabad, 22500, Pakistan
| | - Shoaib Khan
- Department of Chemistry, Abbottabad University of Science & Technology, Abbottabad, 22500, Pakistan
| | - Rafaqat Hussain
- College of Biology, Hunan University Changsha, Hunan, 410082, P. R. China
| | - Yousaf Khan
- Department of Chemistry, COMSATS University Islamabad, Islamabad, 45550, Pakistan
| | - Khurram Shoaib
- Department of Chemistry, Abbottabad University of Science & Technology, Abbottabad, 22500, Pakistan
| | - Rozeena
- Department of Chemistry, University of Malakand, Pakistan
| | - Masab Saeed
- Department of Chemistry, Hazara University, Mansehra, 21120, Pakistan
| | - Hany W Darwish
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh, 11451, Saudi Arabia
| |
Collapse
|
3
|
Yu Y, Xia Y, Liang G. Exploring novel lead scaffolds for SGLT2 inhibitors: Insights from machine learning and molecular dynamics simulations. Int J Biol Macromol 2024; 263:130375. [PMID: 38403210 DOI: 10.1016/j.ijbiomac.2024.130375] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 01/31/2024] [Accepted: 02/20/2024] [Indexed: 02/27/2024]
Abstract
Sodium-glucose cotransporter 2 (SGLT2) plays a pivotal role in mediating glucose reabsorption within the renal filtrate, representing a well-known target in type 2 diabetes and heart failure. Recent emphasis has been directed toward designing SGLT2 inhibitors, with C-glycoside inhibitors emerging as front-runners. The architecture of SGLT2 has been successfully resolved using cryo-electron microscopy. However, comprehension of the pharmacophores within the binding site of SGLT2 remains unclear. Here, we use machine learning and molecular dynamics simulations on SGLT2 bound with its inhibitors in preclinical or clinical development to shed light on this issue. Our dataset comprises 1240 SGLT2 inhibitors amalgamated from diverse sources, forming the basis for constructing machine learning models. SHapley Additive exPlanation (SHAP) elucidates the crucial fragments that contribute to inhibitor activity, specifically Morgan_3, 162, 310, 325, 366, 470, 597, 714, 926, and 975. Furthermore, the computed binding free energies and per-residue contributions for SGLT2-inhibitor complexes unveil crucial fragments of inhibitors that interact with residues Asn-75, His-80, Val-95, Phe-98, Val-157, Leu-274, and Phe-453 in the binding site of SGLT2. This comprehensive investigation enhances understanding of the binding mechanism for SGLT2 inhibitors, providing a robust framework for evaluating and discovering novel lead scaffolds within this domain.
Collapse
Affiliation(s)
- Yuandong Yu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, Bioengineering College, Chongqing University, Chongqing 400044, China
| | - Yuting Xia
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, Bioengineering College, Chongqing University, Chongqing 400044, China
| | - Guizhao Liang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, Bioengineering College, Chongqing University, Chongqing 400044, China.
| |
Collapse
|
4
|
Alshehri MM, Danazumi AU, Alshammari MK, Bello RO, Alghazwni MK, Alshehri AM, Alshlali OM, Umar HI. Repurposing the inhibitors of MMP-9 and SGLT-2 against ubiquitin specific protease 30 in Parkinson's disease: computational modelling studies. J Biomol Struct Dyn 2024; 42:1307-1318. [PMID: 37139557 DOI: 10.1080/07391102.2023.2208223] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 03/29/2023] [Indexed: 05/05/2023]
Abstract
Ubiquitin specific protease 30 (USP30) has been attributed to mitochondrial dysfunction and impediment of mitophagy in Parkinson's disease (PD). This happens once ubiquitin that supposed to bind with deformed mitochondria at the insistence of Parkin, it's been recruited by USP30 via the distal ubiquitin binding domain. This is a challenge when PINK1 and Parkin loss their functions due to mutation. Although, there are reports on USP30s' inhibitors but no study on the repurposing of inhibitors approved against MMP-9 and SGLT-2 as potential inhibitors of USP30 in PD. Thus, the highlight therein, is to repurpose approved inhibitors of MMP-9 and SGLT-2 against USP30 in PD using extensive computational modelling framework. 3D structures of Ligands and USP30 were obtained from PubChem and protein database (PDB) servers respectively, and were subjected to molecular docking, ADMET evaluation, DFT calculation, molecular dynamics simulation (MDS) and free energy calculations. Out of the 18 drugs, 2 drugs showed good binding affinity to the distal ubiquitin binding domain, moderate pharmacokinetic properties and good stability. The findings showed canagliflozin and empagliflozin as potential inhibitors of USP30. Thus, we present these drugs as repurposing candidates for the treatment of PD. However, the findings in this current study needs to be validated experimentally.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Mohammed M Alshehri
- Pharmaceutical Care Department, Ministry of National Guard-Health Affairs, Riyadh, Saudi Arabia
| | - Ammar Usman Danazumi
- Faculty of Chemistry, Warsaw University of Technology, Warsaw, Poland
- Groningen Research Institute of Pharmacy, University of Groningen, Groningen, The Netherlands
| | - Mohammed Kanan Alshammari
- Department of Pharmaceutical Care, Rafha Central Hospital, Rafha, North Zone, Kingdom of Saudi Arabia
| | - Ridwan Opeyemi Bello
- Computer-Aided Therapeutic Discovery and Design Group, Federal University of Technology, Akure, Nigeria
| | | | - Ahmed Mughram Alshehri
- Pharmaceutical care Department, Security Forces Hospital - Riyadh, Riyadh, Kingdom of Saudi Arabia
| | | | - Haruna Isiyaku Umar
- Computer-Aided Therapeutic Discovery and Design Group, Federal University of Technology, Akure, Nigeria
- Department of Biochemistry, Federal University of Technology, Akure, Ondo State, Nigeria
| |
Collapse
|
5
|
Gupta S, Saha M, Singh R, Ahmed SB, Asati V. Multistage in silico approach to identify novel quinoline derivatives as potential c-kit kinase inhibitors. J Biomol Struct Dyn 2024:1-18. [PMID: 38287494 DOI: 10.1080/07391102.2024.2308759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 01/15/2024] [Indexed: 01/31/2024]
Abstract
The type II-C-KIT signaling network has been extensively studied for its potential as a target for cancer treatment, leading to the investigation of quinoline derivatives as compounds with inhibitory effects on c-Kit kinase. In this study, a multistage approach was employed, including the creation of pharmacophore models, 3D QSAR analysis, virtual screening, docking investigations, and molecular dynamics stimulation. The pharmacophore evaluation included a data set of 29 ligands, which resulted in the generation of the ADDHR_1pharmacophore model as the most promising, with a survival score of 5.6812. The main objective was to utilize the atom-based 3D-QSAR approach for generating robust 3D-QSAR models aimed at identifying new TypeII-C-kit kinase inhibitors. The evaluations of these models have convincingly demonstrated their high predictive power (Q2 = 0.6547, R2 = 0.9947). Using atom-based 3D-QSAR data, a total of 7564 novel compounds were generated from R-group enumeration. Molecular docking and MM-GBSA study revealed that compound A1 exhibited the highest binding score of -9.30 kcal/mol and a Δ GBind value of -90.56 kcal/mol. The ZINC compounds were then screened using the pharmacophore model, followed by virtual screening, which identified ZINC65798256, ZINC09317958, ZINC73187176, and ZINC76176670 as potential candidates with promising docking scores. Among these, ZINC65798256 demonstrated the best binding interactions with amino acid residues, ASP810, LYS623, CYS673, and THR670 (PDB ID: 1T46). To further analyze the structural features and molecular interactions, molecular dynamics simulation was conducted for a time scale of 100 ns.
Collapse
Affiliation(s)
- Shankar Gupta
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, Moga, Punjab, India
| | - Moumita Saha
- Department of Pharmaceutical Analysis, ISF College of Pharmacy, Moga, Punjab, India
| | - Rajveer Singh
- Department of Pharmacognosy, ISF College of Pharmacy, Moga, Punjab, India
| | - Samia Ben Ahmed
- Department of Chemistry, College of Sciences, King Khalid University, Abha, Saudi Arabia
| | - Vivek Asati
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, Moga, Punjab, India
| |
Collapse
|
6
|
Debnath A, Sharma S, Mazumder R, Mazumder A, Singh R, Kumar A, Dua A, Singhal P, Kumar A, Singh G. In Search of Novel SGLT2 Inhibitors by High-throughput Virtual Screening. Curr Drug Discov Technol 2024; 21:20-31. [PMID: 38047361 DOI: 10.2174/0115701638267615231123160650] [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: 07/13/2023] [Revised: 10/11/2023] [Accepted: 10/23/2023] [Indexed: 12/05/2023]
Abstract
BACKGROUND Type 2 diabetes mellitus constitutes approximately 90% of all reported forms of diabetes mellitus. Insulin resistance characterizes this manifestation of diabetes. The prevalence of this condition is commonly observed in patients aged 45 and above; however, there is an emerging pattern of younger cohorts receiving diagnoses primarily attributed to lifestyle-related variables, including obesity, sedentary behavior, and poor dietary choices. The enzyme SGLT2 exerts a negative regulatory effect on insulin signaling pathways, resulting in the development of insulin resistance and subsequent elevation of blood glucose levels. The maintenance of glucose homeostasis relies on the proper functioning of insulin signaling pathways, while disruptions in insulin signaling can contribute to the development of type 2 diabetes. OBJECTIVE Our study aimed to identify novel SGLT2 inhibitors by high-throughput virtual Screening. METHODS We screened the May bridge Hit Discover database to identify potent hits followed by druglikeness, synthetic accessibility, PAINS alert, toxicity estimation, ADME assessment, and consensus molecular docking. RESULTS The screening process led to the identification of three molecules that demonstrated significant binding affinity, favorable drug-like properties, effective ADME, and minimal toxicity. CONCLUSION The identified molecules could manage T2DM effectively by inhibiting SGLT2, providing a promising avenue for future therapeutic strategies.
Collapse
Affiliation(s)
- Abhijit Debnath
- Department of Pharmacy, Noida Institute of Engineering and Technology (Pharmacy Institute), 19 Knowledge Park-II, Institutional Area, Greater Noida, 201306, Uttar Pradesh, India
| | - Shalini Sharma
- Department of Pharmacy, Noida Institute of Engineering and Technology (Pharmacy Institute), 19 Knowledge Park-II, Institutional Area, Greater Noida, 201306, Uttar Pradesh, India
| | - Rupa Mazumder
- Department of Pharmacy, Noida Institute of Engineering and Technology (Pharmacy Institute), 19 Knowledge Park-II, Institutional Area, Greater Noida, 201306, Uttar Pradesh, India
| | - Avijit Mazumder
- Department of Pharmacy, Noida Institute of Engineering and Technology (Pharmacy Institute), 19 Knowledge Park-II, Institutional Area, Greater Noida, 201306, Uttar Pradesh, India
| | - Rajesh Singh
- Department of Dravyaguna, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Ankit Kumar
- Department of Pharmacy, Noida Institute of Engineering and Technology (Pharmacy Institute), 19 Knowledge Park-II, Institutional Area, Greater Noida, 201306, Uttar Pradesh, India
| | - Arpita Dua
- Department of Pharmacy, Noida Institute of Engineering and Technology (Pharmacy Institute), 19 Knowledge Park-II, Institutional Area, Greater Noida, 201306, Uttar Pradesh, India
| | - Priya Singhal
- Department of Pharmacy, Noida Institute of Engineering and Technology (Pharmacy Institute), 19 Knowledge Park-II, Institutional Area, Greater Noida, 201306, Uttar Pradesh, India
| | - Arvind Kumar
- Department of Biotechnology, Noida Institute of Engineering and Technology, 19 Knowledge Park-II, Institutional Area, Greater Noida, 201306, Uttar Pradesh, India
| | - Gurvinder Singh
- Department of Medicinal Chemistry, Lovely Professional University, Phagwara, 144001, Punjab, India
| |
Collapse
|
7
|
Ritu, Sharma P, Gupta GD, Asati V. Design, synthesis and antidiabetic study of triazole clubbed indole derivatives as α-glucosidase inhibitors. Bioorg Chem 2023; 139:106750. [PMID: 37499530 DOI: 10.1016/j.bioorg.2023.106750] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 06/23/2023] [Accepted: 07/21/2023] [Indexed: 07/29/2023]
Abstract
α -Glucosidase is an enzyme present near the brush boundary of the small intestine that is essential in the hydrolysis of carbohydrates to glucose. Because inhibiting this enzyme slows the release of glucose, α-Glucosidase inhibitors are appealing medications for treating diabetes as a carbohydrate-related illness. The present study includes the design, synthesis and antidiabetic potential of novel triazole based indole derivatives as α-glucosidase inhibitor. Among them, the compound R1 was found to be most potent with promising candidate with IC50 value of 10.1 μM and R2 and R3 showed the good inhibitory potency with IC50 values 12.95 μM, 11.35 μM, respectively when compared to the standard drug acarbose having IC50 value of 13.5 μM. In in vivo studies, body weight of the mice was increased when compared to standard drug acarbose, the blood glucose level of the mice was decreased, same as the total cholesterol level, LDL, and triglycerides level decreased in comparison to standard drug. The level HDL was increased as it is a good cholesterol in comparison to standard drug acarbose. Furthermore, these synthesized compounds were docked with α-glucosidase using PDB ID:3WY1 which showed that compound R1 having good docking score -6.734 kcal/mol and compound R2, R3 showed docking score -6.14, -6.10 kcal/mol, respectively when compared with standard acarbose having docking score -4.55 kcal/mol. R1 showed the similar interaction with amino acid PHE166, GLU271, comparison with standard drug Acarbose. The synthesized compounds have been confirmed for antidiabetic activity and may be used for further development of potent compounds.
Collapse
Affiliation(s)
- Ritu
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, Moga, Punjab, India
| | - Priyanka Sharma
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, Moga, Punjab, India
| | - G D Gupta
- Department of Pharmaceutics, ISF College of Pharmacy, Moga, Punjab, India
| | - Vivek Asati
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, Moga, Punjab, India.
| |
Collapse
|
8
|
Kailasam Natesan V, Balaraman S, KuppannaGounder Pitchaimuthu E. Insilico design of an allosteric modulator targeting the protein-protein interaction site of 3 Phosphoinositide dependent Kinase-1: design, synthesis and biological activity. In Silico Pharmacol 2023; 11:26. [PMID: 37767119 PMCID: PMC10519888 DOI: 10.1007/s40203-023-00160-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 08/24/2023] [Indexed: 09/29/2023] Open
Abstract
The signalling pathways in human cells mostly rely on protein-protein interactions (PPI) for their function. Such a PPI site in 3 Phosphoinositide dependent Kinase-1 (PDK1) is targeted to design the small molecule modulators. Based on the hotspot residues in its PPI site, a pharmacophore with seven different features was developed and screened against 2.9 million lead like compounds in Zinc database. A phthalazine derivative was identified as a potent allosteric inhibitor through virtual screening, molecular docking and 100 ns dynamics simulations. The modified hit possessed hydrogen bonds with Lys115, Arg131, Thr148, Glu150 as well as pi-pi stacking interactions with Phe157 which are the key residues in the PIF pocket of PDK1. Comparison between the free energy profiles by metadynamics simulation with the presence and absence of the modified ligand (MH) in the binding pocket indicates that the binding of MH enhances the hinge motion making PDK1 to adopt open conformation also and stabilizes the fluctuation of the end-to-end distance in αB helix of PDK1. The modified hit compound was synthesized, characterized and found to be cytotoxic to cancerous cells that are rich in PDK1 expression. These results propose that MH can serve as a new scaffold template for the design of novel drugs targeting PDK1 as well as promising allosteric regulator of PDK1 targeting its protein-protein interface. Supplementary Information The online version contains supplementary material available at 10.1007/s40203-023-00160-6.
Collapse
|
9
|
Raut DG, Bhosale RB, Lawand AS, Hublikar MG, Kadu VD, Patil SB, Choudhari PB. Syntheses, Molecular Docking and Biological Evaluation of 2-(2- hydrazinyl)thiazoles as Potential Antioxidant, Anti-Inflammatory and Significant Anticancer Agents. RECENT ADVANCES IN INFLAMMATION & ALLERGY DRUG DISCOVERY 2022; 16:96-106. [PMID: 36056853 DOI: 10.2174/2772270816666220902094019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 05/31/2022] [Accepted: 06/06/2022] [Indexed: 01/20/2023]
Abstract
BACKGROUND Recently, researchers have worked on the development of new methods for the synthesis of bioactive heterocycles using polyethylene glycol as a green solvent. In this context, we report the synthesized 2-(2-hydrazinyl) thiazoles for their in vitro antioxidant, in vitro anti-inflammatory and in vitro anti-cancer activities. OBJECTIVE The objective of the study was to develop novel antioxidant, anti-inflammatory and anti-cancer drugs. METHODS At the outset, the condensation of substituted acetophenones 1, thiosemicarbazide 2, and α-haloketones 3 was carried out using PEG-400 (20 mL) in the presence of 5 mol% glacial acetic acid to afford thiosemicarbazones intermediate. Furthermore, these thiosemicarbazones were reacted with α-haloketones 3 to obtain appropriate 2-(2-hydrazinyl) thiazoles. The synthesized compounds were in vitro tested for their antioxidant, anti-inflammatory, and anti-cancer activity. RESULTS In vitro evaluation report showed that nearly all molecules possessed potential antioxidant activity against 2,2-Diphenyl-1-picrylhydrazyl (DPPH), nitric oxide (NO), superoxide radical (SOR) and hydrogen peroxide (H2O2) radical scavenging activity. Most 2-(2-hydrazinyl) thiazoles derivatives have shown potential anti-inflammatory activity as compared to diclofenac sodium as a reference standard. 2-(2-Hydrazinyl) thiazoles derivatives showed significant anticancer activity for human leukemia cell line K-562 compared to adriamycin as a reference standard. CONCLUSION All tested compounds showed potential 2,2-Diphenyl-1-picrylhydrazyl (DPPH) and nitric oxide (NO) radical scavenging activity. Among the tested series, 4b, 4d and 4e exhibited good hydrogen peroxide and 4b, 4e, 4f and 4g showed excellent superoxide radical scavenging activity. In addition, the 4b, 4e and 4g compounds revealed potent in vitro anti-inflammatory activity against standard diclofenac sodium drug. 2-(2-Hydrazinyl) thiazole derivatives, such as 4c and 4d, showed significant anticancer activity against human leukemia cell line K-562. Thus, these molecules provide an interesting template for the design and development of new antioxidant, anti-inflammatory, and anti-cancer agents.
Collapse
Affiliation(s)
- Dattatraya G Raut
- Organic Chemistry Research Laboratory, School of Chemical Sciences, Punyashlok Ahilyadevi Holkar Solapur University, Solapur - 413255 Maharashtra, India
| | - Raghunath B Bhosale
- Organic Chemistry Research Laboratory, School of Chemical Sciences, Punyashlok Ahilyadevi Holkar Solapur University, Solapur - 413255 Maharashtra, India
| | - Anjana S Lawand
- Organic Chemistry Research Laboratory, School of Chemical Sciences, Punyashlok Ahilyadevi Holkar Solapur University, Solapur - 413255 Maharashtra, India
| | - Mahesh G Hublikar
- Organic Chemistry Research Laboratory, School of Chemical Sciences, Punyashlok Ahilyadevi Holkar Solapur University, Solapur - 413255 Maharashtra, India
| | - Vikas D Kadu
- Organic Chemistry Research Laboratory, School of Chemical Sciences, Punyashlok Ahilyadevi Holkar Solapur University, Solapur - 413255 Maharashtra, India
| | - Sandeep B Patil
- Department of Pharmacology, Dr. Shivajirao Kadam College of Pharmacy Kasbe Digraj, Sangli, Maharashtra, India
| | - Prafulla B Choudhari
- Computational Chemistry Research Lab, Department of Pharmaceutical Chemistry, Bharati Vidyapeeth College of Pharmacy, Kolhapur, 416013 Maharashtra, India
| |
Collapse
|
10
|
Romanello Joaquim M, Furth EE, Fan Y, Song HK, Pickup S, Cao J, Choi H, Gupta M, Cao Q, Shinohara R, McMenamin D, Clendenin C, Karasic TB, Duda J, Gee JC, O’Dwyer PJ, Rosen MA, Zhou R. DWI Metrics Differentiating Benign Intraductal Papillary Mucinous Neoplasms from Invasive Pancreatic Cancer: A Study in GEM Models. Cancers (Basel) 2022; 14:cancers14164017. [PMID: 36011011 PMCID: PMC9406679 DOI: 10.3390/cancers14164017] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 07/26/2022] [Accepted: 08/17/2022] [Indexed: 11/16/2022] Open
Abstract
KPC (KrasG12D:Trp53R172H:Pdx1-Cre) and CKS (KrasG12D:Smad4L/L:Ptf1a-Cre) mice are genetically engineered mouse (GEM) models that capture features of human pancreatic ductal adenocarcinoma (PDAC) and intraductal papillary mucinous neoplasms (IPMN), respectively. We compared these autochthonous tumors using quantitative imaging metrics from diffusion-weighted MRI (DW-MRI) and dynamic contrast enhanced (DCE)-MRI in reference to quantitative histological metrics including cell density, fibrosis, and microvasculature density. Our results revealed distinct DW-MRI metrics between the KPC vs. CKS model (mimicking human PDAC vs. IPMN lesion): the apparent diffusion coefficient (ADC) of CKS tumors is significantly higher than that of KPC, with little overlap (mean ± SD 2.24±0.2 vs. 1.66±0.2, p<10−10) despite intratumor and intertumor variability. Kurtosis index (KI) is also distinctively separated in the two models. DW imaging metrics are consistent with growth pattern, cell density, and the cystic nature of the CKS tumors. Coregistration of ex vivo ADC maps with H&E-stained sections allowed for regional comparison and showed a correlation between local cell density and ADC value. In conclusion, studies in GEM models demonstrate the potential utility of diffusion-weighted MRI metrics for distinguishing pancreatic cancer from benign pancreatic cysts such as IPMN.
Collapse
Affiliation(s)
| | - Emma E. Furth
- Pancreatic Cancer Research Center, University of Pennsylvania, Philadelphia, PA 19104, USA
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Yong Fan
- Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Hee Kwon Song
- Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Stephen Pickup
- Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jianbo Cao
- Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Hoon Choi
- Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Mamta Gupta
- Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Quy Cao
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Russell Shinohara
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Deirdre McMenamin
- Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Cynthia Clendenin
- Pancreatic Cancer Research Center, University of Pennsylvania, Philadelphia, PA 19104, USA
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Thomas B. Karasic
- Pancreatic Cancer Research Center, University of Pennsylvania, Philadelphia, PA 19104, USA
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jeffrey Duda
- Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - James C. Gee
- Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Peter J. O’Dwyer
- Pancreatic Cancer Research Center, University of Pennsylvania, Philadelphia, PA 19104, USA
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Mark A. Rosen
- Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104, USA
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Rong Zhou
- Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104, USA
- Pancreatic Cancer Research Center, University of Pennsylvania, Philadelphia, PA 19104, USA
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA 19104, USA
- Correspondence:
| |
Collapse
|
11
|
Arumugaperumal A, Sudalaimani DK, Arumugaswami V, Sivasubramaniam S. Draft Genome Sequence of the Earthworm Eudrilus eugeniae. Curr Genomics 2022; 23:118-125. [PMID: 36778974 PMCID: PMC9878837 DOI: 10.2174/1389202923666220401095626] [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: 12/01/2021] [Revised: 01/30/2022] [Accepted: 02/14/2022] [Indexed: 11/22/2022] Open
Abstract
Background: Earthworms are annelids. They play a major role in agriculture and soil fertility. Vermicompost is the best organic manure for plant crops. Eudrilus eugeniae is an earthworm well suited for efficient vermicompost production. The worm is also used to study the cell and molecular biology of regeneration, molecular toxicology, developmental biology, etc., because of its abilities like high growth rate, rapid reproduction, tolerability toward wide temperature range, and less cost of maintenance. Objective: The whole genome has been revealed only for Eisenia andrei and Eisenia fetida. Methods: In the present work, we sequenced the genome of E. eugeniae using the Illumina platform and generated 160,684,383 paired-end reads. Results: The reads were assembled into a draft genome of size 488 Mb with 743,870 contigs and successfully annotated 24,599 genes. Further, 208 stem cell-specific genes and 3,432 non-coding genes were identified. Conclusion: The sequence and annotation details were hosted in a web application available at https://sudhakar-sivasubramaniam-labs.shinyapps.io/eudrilus_genome/.
Collapse
Affiliation(s)
| | | | | | - Sudhakar Sivasubramaniam
- Address correspondence to this author at the Department of Biotechnology, Manonmaniam Sundaranar University, Tirunelveli, Tamilnadu- 627012, India; Tel: +91 9940998936; E-mail:
| |
Collapse
|
12
|
Das S, Akbar S, Ahmed B, Dewangan RP, Iqubal MK, Iqubal A, Chawla P, Pottoo FH, Joseph A. Recent Advancement of Pyrazole Scaffold Based Neuroprotective Agents: A Review. CNS & NEUROLOGICAL DISORDERS-DRUG TARGETS 2021; 21:940-951. [PMID: 34080970 DOI: 10.2174/1871527320666210602152308] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Revised: 02/17/2021] [Accepted: 04/06/2021] [Indexed: 11/22/2022]
Abstract
As a source of therapeutic agents, heterocyclic nitrogen-containing compounds and their derivatives are still interesting and essential. Pyrazole, a five-member heteroaromatic ring with two nitrogen atoms, has a major impact on chemical industries as well as pharmaceutical industries. Due to its wide range of biological activities against various diseases, it has been identified as a biologically important heterocyclic scaffold. The treatment of neurological disorders has always been a difficult task. Therefore, identifying therapeutically effective molecules for neurological conditions remains an open challenge in biomedical research and development. For developing novel entities as neuroprotective agents, recently, pyrazole scaffold has attracted medicinal chemists worldwide. The major focus of research in this area is to discover novel molecules as neuroprotective agents with minimal adverse effects and better effectiveness in improving the neurological condition. This review mainly covers recent developments in the neuropharmacological role of pyrazole incorporated compounds, including their structural-activity relationship (SAR), which also further includes IC50 values (in mM as well as in μM), recent patents, and a brief history as neuroprotective agents.
Collapse
Affiliation(s)
- Subham Das
- Department of Pharmaceutical Chemistry, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka-576104, India
| | - Saleem Akbar
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, Delhi-110062, India
| | - Bahar Ahmed
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, Delhi-110062, India
| | - Rikeshwar Prasad Dewangan
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, Delhi-110062, India
| | - Mohammad Kashif Iqubal
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, Delhi-110062, India
| | - Ashif Iqubal
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, Delhi-110062, India
| | - Pooja Chawla
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, Ghal Kalan, Moga, Punjab-142001, India
| | - Faheem Hyder Pottoo
- Department of Pharmacology, College of Clinical Pharmacy, Imam Abdulrahman Bin Faisal University, 2835 King Faisal Road, Dammam 31441. Saudi Arabia
| | - Alex Joseph
- Department of Pharmaceutical Chemistry, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka-576104, India
| |
Collapse
|
13
|
Xin Y, Zhou S, Wang H, Hu B, Zhang Z, Wang J, Sun T. Comprehensive structure–activity relationship (SAR) investigation of C-aryl glycoside derivatives for the development of SGLT1/SGLT2 dual inhibitors. NEW J CHEM 2021. [DOI: 10.1039/d1nj02510d] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Multi-combined computational approaches were used to explore the SAR and design novel potential SGLT1/SGLT2 dual inhibitors.
Collapse
Affiliation(s)
- Yunting Xin
- Key Laboratory of Structure-Based Drugs Design and Discovery of Ministry of Education, Shenyang Pharmaceutical University
- Shenyang 110016
- People's Republic of China
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University
- Shenyang 110016
| | - Shuhao Zhou
- Key Laboratory of Structure-Based Drugs Design and Discovery of Ministry of Education, Shenyang Pharmaceutical University
- Shenyang 110016
- People's Republic of China
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University
- Shenyang 110016
| | - Huibin Wang
- Key Laboratory of Structure-Based Drugs Design and Discovery of Ministry of Education, Shenyang Pharmaceutical University
- Shenyang 110016
- People's Republic of China
- School of Pharmacy, Shenyang Pharmaceutical University
- Shenyang 110016
| | - Baichun Hu
- Key Laboratory of Structure-Based Drugs Design and Discovery of Ministry of Education, Shenyang Pharmaceutical University
- Shenyang 110016
- People's Republic of China
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University
- Shenyang 110016
| | - Zhigang Zhang
- Key Laboratory of Structure-Based Drugs Design and Discovery of Ministry of Education, Shenyang Pharmaceutical University
- Shenyang 110016
- People's Republic of China
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University
- Shenyang 110016
| | - Jian Wang
- Key Laboratory of Structure-Based Drugs Design and Discovery of Ministry of Education, Shenyang Pharmaceutical University
- Shenyang 110016
- People's Republic of China
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University
- Shenyang 110016
| | - Tiemin Sun
- Key Laboratory of Structure-Based Drugs Design and Discovery of Ministry of Education, Shenyang Pharmaceutical University
- Shenyang 110016
- People's Republic of China
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University
- Shenyang 110016
| |
Collapse
|