1
|
Azam U, Humayun WA, Avathan Veettil AK, Liu Y, Hastürk O, Jiang M, Sievers S, Wu P, Naseer MM. Identification of 5-amino-1,3,4-thiadiazole appended isatins as bioactive small molecules with polypharmacological activities. RSC Med Chem 2025:d4md00770k. [PMID: 39990167 PMCID: PMC11841741 DOI: 10.1039/d4md00770k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Accepted: 01/23/2025] [Indexed: 02/25/2025] Open
Abstract
The identification of heterocyclic small molecules that cover unexplored chemical space is of great importance for the development of new small-molecule therapeutics. In this study, we synthesized a series of 5-amino-1,3,4-thiadiazoles appended isatins (UZ-1-20) that exhibited polypharmacological properties, as evaluated in a cell-painting assay assessing induced cellular morphological changes. A high hit rate ranging from 55% to 80% was observed for the tested compounds at varied concentrations. The most active compounds showed significant activity in inducing cellular morphological changes with a measured induction value of more than 30% and shared a high biological profiling similarity with an antifungal agent itraconazole and a chemokine receptor inhibitor. The synthesized compounds exhibited moderate to good antiproliferative activity against tested cancer cell lines in the MTT assay. Molecular docking studies were performed to theoretically probe and compare the binding modes between the most active UZ compounds and ITZ or BI-6901, respectively. Additionally, ADMET analysis indicated favorable pharmacokinetic parameters including good oral bioavailability, balanced hydrophilicity, and minimal toxicity. Overall, the findings in this study highlight the potential of developing the aminothiadiazole appended isatins as bioactive agents.
Collapse
Affiliation(s)
- Uzma Azam
- Department of Chemistry, Quaid-i-Azam University Islamabad 45320 Pakistan
| | - Waqar Ahmed Humayun
- Department of Medical Oncology & Radiotherapy, King Edward Medical University Lahore 54000 Pakistan
| | - Amrutha K Avathan Veettil
- Chemical Genomics Centre, Max Planck Institute of Molecular Physiology Dortmund 44227 Germany
- Department of Chemical Biology, Max Planck Institute of Molecular Physiology Dortmund 44227 Germany
- Faculty of Chemistry and Chemical Biology, TU Dortmund University Dortmund 44227 Germany
| | - Yang Liu
- Chemical Genomics Centre, Max Planck Institute of Molecular Physiology Dortmund 44227 Germany
- Department of Chemical Biology, Max Planck Institute of Molecular Physiology Dortmund 44227 Germany
- Faculty of Chemistry and Chemical Biology, TU Dortmund University Dortmund 44227 Germany
| | - Oguz Hastürk
- Chemical Genomics Centre, Max Planck Institute of Molecular Physiology Dortmund 44227 Germany
- Department of Chemical Biology, Max Planck Institute of Molecular Physiology Dortmund 44227 Germany
- Faculty of Chemistry and Chemical Biology, TU Dortmund University Dortmund 44227 Germany
| | - Mao Jiang
- Chemical Genomics Centre, Max Planck Institute of Molecular Physiology Dortmund 44227 Germany
- Department of Chemical Biology, Max Planck Institute of Molecular Physiology Dortmund 44227 Germany
- Faculty of Chemistry and Chemical Biology, TU Dortmund University Dortmund 44227 Germany
| | - Sonja Sievers
- Department of Chemical Biology, Max Planck Institute of Molecular Physiology Dortmund 44227 Germany
- Compound Management and Screening Center Dortmund 44227 Germany
| | - Peng Wu
- Chemical Genomics Centre, Max Planck Institute of Molecular Physiology Dortmund 44227 Germany
- Department of Chemical Biology, Max Planck Institute of Molecular Physiology Dortmund 44227 Germany
- Faculty of Chemistry and Chemical Biology, TU Dortmund University Dortmund 44227 Germany
| | - Muhammad Moazzam Naseer
- Department of Chemistry, Quaid-i-Azam University Islamabad 45320 Pakistan
- Chemical Genomics Centre, Max Planck Institute of Molecular Physiology Dortmund 44227 Germany
| |
Collapse
|
2
|
Ogiji ED, Aboheimed N, Ross K, Voller C, Siner R, Jensen RL, Jolly CE, Carr DF. Greater mechanistic understanding of the cutaneous pathogenesis of Stevens-Johnson syndrome/toxic epidermal necrolysis can shed light on novel therapeutic strategies: a comprehensive review. Curr Opin Allergy Clin Immunol 2024; 24:218-227. [PMID: 38753537 PMCID: PMC11213502 DOI: 10.1097/aci.0000000000000993] [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] [Indexed: 05/18/2024]
Abstract
PURPOSE OF REVIEW Stevens-Johnson syndrome/toxic epidermal necrolysis (SJS/TEN) are severe cutaneous adverse drug reactions (SCARs) characterized by widespread epithelial detachment and blistering, which affects the skin and mucocutaneous membranes. To date, therapeutic interventions for SJS/TEN have focused on systematic suppression of the inflammatory response using high-dose corticosteroids or intravenous immunoglobulin G (IgG), for example. No targeted therapies for SJS/TEN currently exist. RECENT FINDINGS Though our understanding of the pathogenesis of SJS/TEN has advanced from both an immunological and dermatological perspective, this knowledge is yet to translate into the development of new targeted therapies. SUMMARY Greater mechanistic insight into SJS/TEN would potentially unlock new opportunities for identifying or repurposing targeted therapies to limit or even prevent epidermal injury and blistering.
Collapse
Affiliation(s)
- Emeka D. Ogiji
- Department of Pharmacology and Therapeutics, University of Liverpool, Liverpool, UK
- Department of Pharmacology and Therapeutics, Ebonyi State University, Abakaliki, Nigeria
| | - Nourah Aboheimed
- Department of Pharmacology and Therapeutics, University of Liverpool, Liverpool, UK
- Department of Pharmacy Practice, Princess Nourah bint Abdulrahman University, Saudi Arabia
| | - Kehinde Ross
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University
| | - Calum Voller
- School of Medicine, University of Liverpool, Liverpool, UK
| | - Ryan Siner
- Department of Pharmacology and Therapeutics, University of Liverpool, Liverpool, UK
| | - Rebecca L. Jensen
- Department of Pharmacology and Therapeutics, University of Liverpool, Liverpool, UK
| | - Carol E. Jolly
- Department of Pharmacology and Therapeutics, University of Liverpool, Liverpool, UK
| | - Daniel F. Carr
- Department of Pharmacology and Therapeutics, University of Liverpool, Liverpool, UK
| |
Collapse
|
3
|
Eiger DS, Boldizsar N, Honeycutt CC, Gardner J, Rajagopal S. Biased agonism at chemokine receptors. Cell Signal 2020; 78:109862. [PMID: 33249087 DOI: 10.1016/j.cellsig.2020.109862] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 11/07/2020] [Accepted: 11/24/2020] [Indexed: 12/11/2022]
Abstract
In the human chemokine system, interactions between the approximately 50 known endogenous chemokine ligands and 20 known chemokine receptors (CKRs) regulate a wide range of cellular functions and biological processes including immune cell activation and homeostasis, development, angiogenesis, and neuromodulation. CKRs are a family of G protein-coupled receptors (GPCR), which represent the most common and versatile class of receptors in the human genome and the targets of approximately one third of all Food and Drug Administration-approved drugs. Chemokines and CKRs bind with significant promiscuity, as most CKRs can be activated by multiple chemokines and most chemokines can activate multiple CKRs. While these ligand-receptor interactions were previously regarded as redundant, it is now appreciated that many chemokine:CKR interactions display biased agonism, the phenomenon in which different ligands binding to the same receptor signal through different pathways with different efficacies, leading to distinct biological effects. Notably, these biased responses can be modulated through changes in ligand, receptor, and or the specific cellular context (system). In this review, we explore the biochemical mechanisms, functional consequences, and therapeutic potential of biased agonism in the chemokine system. An enhanced understanding of biased agonism in the chemokine system may prove transformative in the understanding of the mechanisms and consequences of biased signaling across all GPCR subtypes and aid in the development of biased pharmaceuticals with increased therapeutic efficacy and safer side effect profiles.
Collapse
Affiliation(s)
| | - Noelia Boldizsar
- Trinity College of Arts and Sciences, Duke University, Durham, NC 27710, USA.
| | | | - Julia Gardner
- Trinity College of Arts and Sciences, Duke University, Durham, NC 27710, USA.
| | - Sudarshan Rajagopal
- Department of Biochemistry, Duke University, Durham, NC 27710, USA; Department of Medicine, Duke University, Durham, NC 27710, USA.
| |
Collapse
|
4
|
Ribeiro CJA, Kankanala J, Xie J, Williams J, Aihara H, Wang Z. Triazolopyrimidine and triazolopyridine scaffolds as TDP2 inhibitors. Bioorg Med Chem Lett 2018; 29:257-261. [PMID: 30522956 DOI: 10.1016/j.bmcl.2018.11.044] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Revised: 11/12/2018] [Accepted: 11/21/2018] [Indexed: 01/09/2023]
Abstract
Tyrosyl-DNA phosphodiesterase 2 (TDP2) repairs topoisomerase II (TOP2) mediated DNA damages and causes cellular resistance to clinically used TOP2 poisons. Inhibiting TDP2 can potentially sensitize cancer cells toward TOP2 poisons. Commercial compound P10A10, to which the structure was assigned as 7-phenyl triazolopyrimidine analogue 6a, was previously identified as a TDP2 inhibitor hit in our virtual and fluorescence-based biochemical screening campaign. We report herein that the hit validation through resynthesis and structure elucidation revealed the correct structure of P10A10 (Chembridge ID 7236827) to be the 5-phenyl triazolopyrimidine regioisomer 7a. Subsequent structure-activity relationship (SAR) via the synthesis of a total of 47 analogues of both the 5-phenyl triazolopyrimidine scaffold (7) and its bioisosteric triazolopyridine scaffold (17) identified four derivatives (7a, 17a, 17e, and 17z) with significant TDP2 inhibition (IC50 < 50 µM), with 17z showing excellent cell permeability and no cytotoxicity.
Collapse
Affiliation(s)
- Carlos J A Ribeiro
- Center for Drug Design, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, United States
| | - Jayakanth Kankanala
- Center for Drug Design, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, United States
| | - Jiashu Xie
- Center for Drug Design, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, United States
| | - Jessica Williams
- Center for Drug Design, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, United States
| | - Hideki Aihara
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN 55455, United States
| | - Zhengqiang Wang
- Center for Drug Design, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, United States.
| |
Collapse
|
5
|
Gao YY, Jiang Y, Chen GC, Li SS, Yang F, Ma Q. A Sensitive and Rapid UPLC-MS/MS Method for Determination of Monosaccharides and Anti-Allergic Effect of the Polysaccharides Extracted from Saposhnikoviae Radix. Molecules 2018; 23:molecules23081924. [PMID: 30071672 PMCID: PMC6222413 DOI: 10.3390/molecules23081924] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2018] [Revised: 07/24/2018] [Accepted: 07/26/2018] [Indexed: 12/21/2022] Open
Abstract
Background: Allergic disease is a common clinical disease. Natural products provide an important source for a wide range of potential anti-allergic agents. This study was designed to evaluate the anti-allergic activities of the water-soluble polysaccharides extracted and purified from Saposhnikoviae Radix (SRPS). The composition and content of monosaccharides were determined to provide a material basis. Methods: An ultra-high-performance liquid chromatography-tandem mass spectrometry (UPLC-MS/MS) method was established to determine the composition and content of SRPS. 2,4-dinitrofluorobenzene (DNFB) induced a delayed-type hypersensitivity (DTH) mouse model orally administrated SRPS for seven consecutive days. Ear swelling, organ index, and serum IgE levels were observed to evaluate the anti-allergic activities. Results: The UPLC-MS/MS analysis showed that SRPS was consisted of eight monosaccharides including galacturonic acid, mannose, glucose, galactose, rhamnose, fucose, ribose, and arabinose with a relative molar ratio of 4.42%, 7.86%, 23.69%, 12.06%, 3.10%, 0.45%, 0.71%, and 47.70%, respectively. SRPS could effectively reduce ear swelling, a thymus index, and a serum IgE levels. Conclusions: The method was simple, rapid, sensitive, and reproducible, which could be used to analyze and determine the monosaccharide composition of SRPS. The vivo experiments demonstrated that SRPS may effectively inhibit development of DNFB-induced DTH. SRPS is a novel potential resource for natural anti-allergic drugs.
Collapse
Affiliation(s)
- Yan-Yan Gao
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China.
| | - Yue Jiang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China.
| | - Guo-Chao Chen
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China.
| | - Shuang-Shuang Li
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China.
| | - Fei Yang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China.
| | - Qun Ma
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China.
| |
Collapse
|