801
|
Subramani T, Saravanan H, David H, Solanke J, Rajaramon S, Dandela R, Solomon AP. Bioorganic compounds in quorum sensing disruption: strategies, Mechanisms, and future prospects. Bioorg Chem 2025; 156:108192. [PMID: 39874908 DOI: 10.1016/j.bioorg.2025.108192] [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: 08/16/2024] [Revised: 12/04/2024] [Accepted: 01/17/2025] [Indexed: 01/30/2025]
Abstract
Recent research has shed light on the complex world of bacterial communication through quorum sensing. This sophisticated intercellular signalling mechanism, driven by auto-inducers, regulates crucial bacterial community behaviours such as biofilm formation, expression of virulence factors, and resistance mechanisms. The increasing threat of antibiotic resistance, coupled with quorum sensing mediated response, necessitates alternative strategies to combat bacterial infections. Quorum quenching has emerged as a promising approach, utilizing quorum quenching enzymes and quorum sensing inhibitors to disrupt quorum sensing signalling pathways, thus reducing virulence and biofilm formation. This review focuses on natural and synthetic bioorganic compounds that act as quorum-sensing inhibitors, providing insights into their mechanisms, structure-activity relationships, and potential as anti-virulence agents. The review also explores the communication languages of bacteria, including AHLs in gram-negative bacteria, oligopeptides in gram-positive bacteria, and LuxS, a universal microbial language. By highlighting recent advancements and prospects in bioorganic QSIs, this article underscores their crucial role in developing effective anti-virulence therapies and combating the growing threat of antimicrobial resistance.
Collapse
Affiliation(s)
- Tarunkarthick Subramani
- Quorum Sensing Laboratory, Centre for Research in Infectious Diseases (CRID), School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur 613401, India
| | - Harish Saravanan
- Quorum Sensing Laboratory, Centre for Research in Infectious Diseases (CRID), School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur 613401, India
| | - Helma David
- Quorum Sensing Laboratory, Centre for Research in Infectious Diseases (CRID), School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur 613401, India
| | - Jayshree Solanke
- Department of Industrial and Engineering Chemistry, Institute of Chemical Technology, Bhubaneswar, Odisha, India
| | - Shobana Rajaramon
- Quorum Sensing Laboratory, Centre for Research in Infectious Diseases (CRID), School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur 613401, India.
| | - Rambabu Dandela
- Department of Industrial and Engineering Chemistry, Institute of Chemical Technology, Bhubaneswar, Odisha, India.
| | - Adline Princy Solomon
- Quorum Sensing Laboratory, Centre for Research in Infectious Diseases (CRID), School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur 613401, India.
| |
Collapse
|
802
|
Haider MK, Ullah A, Gopiraman M, Kim IS. A review on bioactivity, plant safety, and metal-reducing potential of lignin, its micro/nanostructures, and composites. Int J Biol Macromol 2025; 294:139366. [PMID: 39743055 DOI: 10.1016/j.ijbiomac.2024.139366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 12/20/2024] [Accepted: 12/29/2024] [Indexed: 01/04/2025]
Abstract
Modern science focuses on sustainability-oriented innovation. Structurally sophisticated lignin is a sustainable alternative to non-renewable resources. Over the last several years, a tremendous scientific effort has been made to innovate lignin-based sustainable materials for numerous advanced applications. The lignin's phenolic, methoxyl and aliphatic hydroxyl functional groups are biologically and chemically active, making it conducive to developing state-of-the-art biomedicine, food packaging, crop protection, and catalyst materials. The biocidal effect of lignin rests on the phenolic compounds, specifically the double bond in α, β positions of the side chain, and a methyl group in the γ position. Also, depending on the biomass source and the pulping method, lignins possess different biocidal and antioxidant properties. The abundant hydroxyl groups in lignin are metal reductants and possess capping ability for the nanoparticles (NPs). This review focused on lignin's bioactivity mechanism, including antimicrobial efficacy and antioxidant properties. Lignin-based micro/nanocomposites and their application on food packaging, plant protection, and growth will also be explored. We will also review the application of lignin as a reducing and capping agent for the synthesis of metal NPs.
Collapse
Affiliation(s)
- Md Kaiser Haider
- Nano Fusion Technology Research Group, Institute for Fiber Engineering and Science (IFES), Interdisciplinary Cluster for Cutting Edge Research (ICCER), Shinshu University, Tokida 3-15-1, Ueda, Nagano 386-8567, Japan
| | - Azeem Ullah
- Nano Fusion Technology Research Group, Institute for Fiber Engineering and Science (IFES), Interdisciplinary Cluster for Cutting Edge Research (ICCER), Shinshu University, Tokida 3-15-1, Ueda, Nagano 386-8567, Japan
| | - Mayakrishnan Gopiraman
- Nano Fusion Technology Research Group, Institute for Fiber Engineering and Science (IFES), Interdisciplinary Cluster for Cutting Edge Research (ICCER), Shinshu University, Tokida 3-15-1, Ueda, Nagano 386-8567, Japan
| | - Ick Soo Kim
- Nano Fusion Technology Research Group, Institute for Fiber Engineering and Science (IFES), Interdisciplinary Cluster for Cutting Edge Research (ICCER), Shinshu University, Tokida 3-15-1, Ueda, Nagano 386-8567, Japan.
| |
Collapse
|
803
|
Hellmann A, Zwara A, Weryszko O, Czapiewska M, Korczynska J, Sztendel A, Śledziński T, Mika A. Evaluation of the effect of Hashimoto's Thyroiditis on fatty acids involved in inflammation in the thyroid tissue. Biomed Pharmacother 2025; 184:117894. [PMID: 39913970 DOI: 10.1016/j.biopha.2025.117894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 01/16/2025] [Accepted: 02/03/2025] [Indexed: 03/04/2025] Open
Abstract
Hashimoto's thyroiditis (HT) is one of the most common autoimmune diseases associated with inflammation of the thyroid gland. Some fatty acids (FAs) are involved in inflammation. The aim of the study was to investigate how HT affects FA metabolism in thyroid tissue. We examined normal thyroid tissue from 92 patients with and without HT. We determined total FA panel, pro-inflammatory and specialized pro-resolving lipid mediators (SPMs) concentrations and measured the expression levels of genes encoding the corresponding enzymes of FA metabolism and mediator's synthesis. In HT tissue, we observed increased total lipid levels (p < 0.05), decreased carnitine palmitoyltransferase I (p < 0.05) and increased tumor necrosis factor alpha (p < 0.001) compared to healthy tissue. Simultaneously, we observed an overexpression of enzymes responsible for the synthesis of polyunsaturated FAs and their higher levels in HT tissue. However, despite the overexpression of phospholipase A2 (p < 0.001), FA translocase CD36 (p < 0.05) and higher levels of free arachidonic acid in HT tissue (p < 0.05), we observed no differences in the expression of COX-2 in both tissues and, interestingly, a downregulation of 15-LOX (p = 0.001) and lower concentrations of SPMs. Finally, the opposite effect of HT on enzymes responsible for anti-inflammatory BCFAs synthesis in mitochondria and cytosol could indicate a compensatory mechanism. To summarize, the mechanism of the effect of HT on FA metabolism is certainly complex. Thyrocytes are perturbed by the inflammatory effects associated with HT, leading to mitochondrial dysfunction and consequently decreased β-oxidation and BCFA metabolism in mitochondria. Furthermore, despite a significant synthesis of PUFAs, SPMs are also not produced.
Collapse
Affiliation(s)
- Andrzej Hellmann
- Department of Department of Surgical Oncology, Transplant Surgery and General Surgery, Faculty of Medicine, Medical University of Gdansk, Smoluchowskiego 17, Gdansk 80-214, Poland
| | - Agata Zwara
- Department of Environmental Analytics, Faculty of Chemistry, University of Gdansk, Wita Stwosza 63, Gdansk 80-308, Poland
| | - Oliwia Weryszko
- Department of Pharmaceutical Biochemistry, Medical University of Gdansk, 1 Debinki, Gdansk 80-211, Poland
| | - Monika Czapiewska
- Department of Pharmaceutical Biochemistry, Medical University of Gdansk, 1 Debinki, Gdansk 80-211, Poland
| | - Justyna Korczynska
- Department of Pharmaceutical Biochemistry, Medical University of Gdansk, 1 Debinki, Gdansk 80-211, Poland
| | - Alicja Sztendel
- Department of Pharmaceutical Biochemistry, Medical University of Gdansk, 1 Debinki, Gdansk 80-211, Poland
| | - Tomasz Śledziński
- Department of Pharmaceutical Biochemistry, Medical University of Gdansk, 1 Debinki, Gdansk 80-211, Poland
| | - Adriana Mika
- Department of Environmental Analytics, Faculty of Chemistry, University of Gdansk, Wita Stwosza 63, Gdansk 80-308, Poland; Department of Pharmaceutical Biochemistry, Medical University of Gdansk, 1 Debinki, Gdansk 80-211, Poland.
| |
Collapse
|
804
|
Sharma V, Kumar A. MCL-1 as a potent target for cancer: Recent advancements, structural insights and SAR studies. Bioorg Chem 2025; 156:108211. [PMID: 39889551 DOI: 10.1016/j.bioorg.2025.108211] [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: 12/07/2024] [Revised: 01/16/2025] [Accepted: 01/22/2025] [Indexed: 02/03/2025]
Abstract
The myeloid cell leukemia-1 (Mcl-1) differentiation protein belongs to the B-cell lymphoma 2 (Bcl-2) family of proteins which regulates the apoptosis or cell death. Mcl-1 is known for its pro-survival in response to various stressors. Therefore, it acts as a prominent target in cancer treatment. Mcl-1 has emerged as one of the validated drug targets for anticancer drug discovery as their expression has been implicated in the pathogenesis of cancers. In this review, we have included the various inhibitors based on many heterocyclic rings such as pyrrole, pyrazole, coumarin, quinoline and indole. This manuscript incorporates the anticancer activity, structure activity relationship (SAR) and molecular modelling of recently synthesized Mcl-1 inhibitors. The clinical trial status of Mcl-1 inhibitors is also described. But till now, no Mcl-1 inhibitor has been approved by any drug authority. This review is based on extensive research in the field of designing Mcl-1 inhibitors from 2020 to till now. It will provide extensive information to researchers and scientists for designing of novel Mcl-1 inhibitors.
Collapse
Affiliation(s)
- Vishakha Sharma
- Amity School of Pharmaceutical Sciences, Amity University Punjab, Mohali, India
| | - Ankush Kumar
- Amity School of Pharmaceutical Sciences, Amity University Punjab, Mohali, India.
| |
Collapse
|
805
|
Andrianov AM, Furs KV, Gonchar AV, Skrahina AM, Wang Y, Lyu LD, Tuzikov AV. Virtual screening and identification of promising therapeutic compounds against drug-resistant Mycobacterium tuberculosis β-ketoacyl-acyl carrier protein synthase I (KasA). J Biomol Struct Dyn 2025; 43:2029-2041. [PMID: 38088766 DOI: 10.1080/07391102.2023.2293276] [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: 08/24/2023] [Accepted: 12/01/2023] [Indexed: 02/01/2025]
Abstract
The emergence of new Mycobacterium tuberculosis (Mtb) strains resistant to the key drugs currently used in the clinic for tuberculosis treatment can substantially reduce the probability of therapy success, causing the relevance and importance of studies on the development of novel potent antibacterial agents targeting different vulnerable spots of Mtb. In this study, 28,860 compounds from the library of bioactive molecules were screened to identify novel potential inhibitors of β-ketoacyl-acyl carrier protein synthase I (KasA), one of the key enzymes involved in the biosynthesis of mycolic acids of the Mtb cell wall. In doing so, we used a structure-based virtual screening approach to drug repurposing that included high-throughput docking of the C171Q KasA enzyme with compounds from the library of bioactive molecules including the FDA-approved drugs and investigational drug candidates, assessment of the binding affinity for the docked ligand/C171Q KasA complexes, and molecular dynamics simulations followed by binding free energy calculations. As a result, post-modeling analysis revealed 6 top-ranking compounds exhibiting a strong attachment to the malonyl binding site of the enzyme, as evidenced by the values of binding free energy which are significantly lower than those predicted for the KasA inhibitor TLM5 used in the calculations as a positive control. In light of the data obtained, the identified compounds are suggested to form a good basis for the development of new antitubercular molecules of clinical significance with activity against the KasA enzyme of Mtb.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Alexander M Andrianov
- Institute of Bioorganic Chemistry, National Academy of Sciences of Belarus, Minsk, Republic of Belarus
| | - Konstantin V Furs
- United Institute of Informatics Problems, National Academy of Sciences of Belarus, Minsk, Republic of Belarus
| | - Anna V Gonchar
- United Institute of Informatics Problems, National Academy of Sciences of Belarus, Minsk, Republic of Belarus
| | - Alena M Skrahina
- Republican Scientific and Practical Center of Pulmonology and Tuberculosis, Minsk, Republic of Belarus
| | - Yixin Wang
- Key Laboratory of Medical Molecular Virology of the Ministry of Education/Ministry of Health Commission, School of Basic Medical Sciences, Fudan University, Shanghai, People's Republic of China
| | - Liang-Dong Lyu
- Key Laboratory of Medical Molecular Virology of the Ministry of Education/Ministry of Health Commission, School of Basic Medical Sciences, Fudan University, Shanghai, People's Republic of China
- Shanghai Clinical Research Center for Infectious Disease (Tuberculosis), Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Shanghai, People's Republic of China
| | - Alexander V Tuzikov
- United Institute of Informatics Problems, National Academy of Sciences of Belarus, Minsk, Republic of Belarus
| |
Collapse
|
806
|
Bano N, Mohammed SA, Raza K. Integrating machine learning and multitargeted drug design to combat antimicrobial resistance: a systematic review. J Drug Target 2025; 33:384-396. [PMID: 39535825 DOI: 10.1080/1061186x.2024.2428984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 10/16/2024] [Accepted: 11/07/2024] [Indexed: 11/16/2024]
Abstract
Antimicrobial resistance (AMR) is a critical global health challenge, undermining the efficacy of antimicrobial drugs against microorganisms like bacteria, fungi and viruses. Multidrug resistance (MDR) arises when microorganisms become resistant to multiple antimicrobial agents. The World Health Organisation classifies AMR bacteria into priority list - I (critical), II (high) and III (medium), prompting action from nearly 170 countries. Six priority bacterial strains account for over 70% of AMR-related fatalities, contributing to more than 1.3 million direct deaths annually and linked to over 5 million deaths globally. Enterobacteriaceae, including Escherichia coli, Salmonella enterica and Klebsiella pneumoniae, significantly contribute to AMR fatalities. This systematic literature review explores how machine learning (ML) and multitargeted drug design (MTDD) can combat AMR in Enterobacteriaceae. We followed PRISMA guidelines and comprehensively analysed current prospects and limitations by mining PubMed and Scopus literature databases. Innovative strategies integrating AI algorithms with advanced computational techniques allow for the analysis of vast datasets, identification of novel drug targets, prediction of resistance mechanisms, and optimisation of drug molecules to overcome resistance. Leveraging ML and MTDD is crucial for both advancing our fight against AMR in Enterobacteriaceae, and developing combination therapies that target multiple bacterial survival pathways, reducing the risk of resistance development.
Collapse
Affiliation(s)
- Nagmi Bano
- Computational Intelligence and Bioinformatics Lab., Department of Computer Science, Jamia Millia Islamia, New Delhi, India
| | - Salman Arafath Mohammed
- Central Labs, King Khalid University, AlQura'a, Abha, Saudi Arabia
- Electrical Engineering Department, College of Engineering, King Khalid University, Abha, Saudi Arabia
| | - Khalid Raza
- Computational Intelligence and Bioinformatics Lab., Department of Computer Science, Jamia Millia Islamia, New Delhi, India
| |
Collapse
|
807
|
Alam M, Basir MS, Sultan MB, Murshed MF, Hossain S, Anik AH. Ecological footprint of ionophores in livestock production: Environmental pathways and effects. WATER ENVIRONMENT RESEARCH : A RESEARCH PUBLICATION OF THE WATER ENVIRONMENT FEDERATION 2025; 97:e70052. [PMID: 40056089 DOI: 10.1002/wer.70052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 02/12/2025] [Accepted: 02/25/2025] [Indexed: 05/13/2025]
Abstract
Ionophores, a class of animal antibiotics, are widely used in intensive livestock farming to enhance feed efficiency and control coccidiosis. These compounds, known for their ability to transport cations across biological membranes, are crucial in maintaining cellular homeostasis. However, their extensive use raises environmental and human health concerns. This manuscript offers a comprehensive review of ionophores in livestock production, highlighting their environmental impact and potential to contribute to antimicrobial resistance (AMR). It emphasizes the fate and transport of ionophores in various environmental matrices, providing a holistic framework for assessing ecological risks. The study calls for improved management practices like enhanced waste management through anaerobic digestion, and composting is essential. Establishing Maximum Residue Limits (MRLs) and using LC-MS/MS for residue detection will help manage exposure. Educating livestock producers and researching alternatives like probiotics can decrease reliance on ionophores to mitigate the ecological footprint of ionophores, making it a timely and relevant piece of research. Ionophores can persist in the environment, potentially contributing to AMR in gram-positive bacteria. Furthermore, their presence in manure, runoff, and agricultural soils has been documented, leading to contamination of water bodies and sediments. Ionophores pose risks to terrestrial and aquatic ecosystems, with studies revealing hazardous effects even at low concentrations. This review highlights the need for improved management practices to mitigate the environmental impacts of ionophores, particularly regarding AMR development and ecosystem disruption. Careful monitoring and sustainable use of these antibiotics are essential to reduce their ecological footprint in livestock production. PRACTITIONER POINTS: Ionophores enhance feed efficiency, but pose environmental health risks. Their persistence may lead to antimicrobial resistance in gram-positive bacteria. Ionophore contamination threatens both terrestrial and aquatic ecosystems. Monitoring and management are crucial to mitigate ionophore-related risks.
Collapse
Affiliation(s)
- Mahbub Alam
- Department of Environmental Science, Bangladesh University of Professionals (BUP), Dhaka, Bangladesh
- Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, South Carolina, USA
| | - Md Samium Basir
- Department of Environmental Science, Bangladesh University of Professionals (BUP), Dhaka, Bangladesh
| | - Maisha Binte Sultan
- Department of Environmental Science, Bangladesh University of Professionals (BUP), Dhaka, Bangladesh
| | - Md Fahim Murshed
- Department of Environmental Science, Bangladesh University of Professionals (BUP), Dhaka, Bangladesh
| | - Shabiha Hossain
- Department of Geography and Sustainability, University of Tennessee, Knoxville, Tennessee, USA
| | - Amit Hasan Anik
- Department of Environmental Science, Bangladesh University of Professionals (BUP), Dhaka, Bangladesh
| |
Collapse
|
808
|
Fomenko I, Denysenko N, Lozynska I, Kuryk M, Yushyn I, Myhal O, Pinyazhko R, Lozynskyi A, Lesyk R. Efficacy of thiazole derivatives against colorectal cancer induced by dimethylhydrazine in male Wistar rats. Biochem Biophys Res Commun 2025; 750:151424. [PMID: 39892058 DOI: 10.1016/j.bbrc.2025.151424] [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: 11/01/2024] [Revised: 01/21/2025] [Accepted: 01/28/2025] [Indexed: 02/03/2025]
Abstract
Identifying novel biological activities in hit compounds remains a significant challenge in cancer research. In this study, we evaluated the anticancer and safety profiles of two previously studied thiazole-based derivatives, Les-5303 and Les-6485, in a colorectal cancer (CRC) model induced by dimethylhydrazine (DMH) in male Wistar rats. CRC was induced through subcutaneous DMH administration at a 20 mg/kg dose for 20 weeks. Les-5303 and Les-6485 were then administered intrarectally at a 6 mg/kg dose for 5 days. The effects of these compounds on oxidative stress, the antioxidant system, inflammation, NO-synthase activity in the colonic mucosa and blood plasma, and hepatotoxicity markers, were thoroughly assessed. Both compounds demonstrated significant anticancer activity and antioxidant properties. Les-5303, however, exhibited increased hepatotoxicity, as evidenced by elevated AST activity and reduced urea concentration in the blood, indicating its potential for liver damage. In contrast, Les-6485 showed no significant hepatotoxic effects, maintaining normal hepatic enzyme activities and urea levels, suggesting a better safety profile. These findings highlight the distinct biological effects of Les-5303 and Les-6485, with Les-6485 showing promising anticancer activity coupled with minimal toxicity, making it a more favorable candidate for further development in CRC treatment.
Collapse
Affiliation(s)
- Iryna Fomenko
- Department of Biochemistry, Danylo Halytsky Lviv National Medical University, Lviv, 79010, Ukraine
| | - Nataliia Denysenko
- Department of Biochemistry, Danylo Halytsky Lviv National Medical University, Lviv, 79010, Ukraine
| | - Iryna Lozynska
- Department of Biochemistry, Danylo Halytsky Lviv National Medical University, Lviv, 79010, Ukraine
| | - Mariana Kuryk
- Department of Biochemistry, Danylo Halytsky Lviv National Medical University, Lviv, 79010, Ukraine
| | - Ihor Yushyn
- Department of Pharmaceutical, Organic and Bioorganic Chemistry, Danylo Halytsky Lviv National Medical University, Lviv, 79010, Ukraine
| | - Ostap Myhal
- Department of Therapeutic Stomatology, Periodontology and Stomatology FPGE, Danylo Halytsky Lviv National Medical University, Lviv, 79010, Ukraine
| | - Roman Pinyazhko
- Department of Normal Physiology, Danylo Halytsky Lviv National Medical University, Lviv, 79010, Ukraine
| | - Andrii Lozynskyi
- Department of Pharmaceutical, Organic and Bioorganic Chemistry, Danylo Halytsky Lviv National Medical University, Lviv, 79010, Ukraine
| | - Roman Lesyk
- Department of Pharmaceutical, Organic and Bioorganic Chemistry, Danylo Halytsky Lviv National Medical University, Lviv, 79010, Ukraine; Department of Biotechnology and Cell Biology, Medical College, University of Information Technology and Management in Rzeszow, Sucharskiego 2, 35-225, Rzeszow, Poland.
| |
Collapse
|
809
|
Mokos M, Bulat V, Likić R, Bosnić F, Gašparov S. Infliximab-induced symmetrical drug-related intertriginous and flexural exanthema (SDRIFE) in a patient with ulcerative colitis. Br J Clin Pharmacol 2025; 91:903-907. [PMID: 39780340 DOI: 10.1111/bcp.16390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Revised: 12/06/2024] [Accepted: 12/20/2024] [Indexed: 01/11/2025] Open
Abstract
Symmetrical drug-related intertriginous and flexural exanthema (SDRIFE) is a rare, symmetrical skin eruption triggered by various medications, predominantly beta-lactam antibiotics. We report the case of a 69-year-old male with moderate-to-severe ulcerative colitis who developed SDRIFE following the seventh intravenous administration of infliximab. The patient presented with symmetrical, pruritic erythema in the cubital and popliteal fossae, groins, gluteal and retroauricular regions without systemic involvement. Skin biopsy showed mild exocytosis of T lymphocytes in the epidermis and dense superficial perivascular CD3+ and CD4+ infiltration, consistent with a type IVa hypersensitivity reaction. The patient responded well to a regimen of systemic antihistamines, topical corticosteroids and tacrolimus ointment, with complete regression of lesions within one week. Despite mild recurrences of SDRIFE after each infliximab administration, the therapy was not discontinued due to the mild nature of the reaction and favourable prognosis. The authors are not aware of previously published cases of type IVa SDRIFE induced by infliximab. Unlike previous reports of severe type IVc SDRIFE reactions requiring discontinuation of infliximab, our case highlights the predominance of CD4+ cells, which may explain the mild clinical course. Understanding the underlying immunologic mechanisms of infliximab-induced SDRIFE could affect treatment decisions and prevent unnecessary discontinuation of effective therapies.
Collapse
Affiliation(s)
- Mislav Mokos
- Department of Dermatology and Venereology, Sestre Milosrdnice University Hospital Centre, Zagreb, Croatia
| | - Vedrana Bulat
- Department of Dermatology and Venereology, Sestre Milosrdnice University Hospital Centre, Zagreb, Croatia
| | - Robert Likić
- University of Zagreb School of Medicine, Zagreb, Croatia
- Division for Clinical Pharmacology and Therapeutics, Department of Internal Medicine University Hospital Centre Zagreb, Zagreb, Croatia
| | - Filip Bosnić
- Department of Dermatology and Venereology, Sestre Milosrdnice University Hospital Centre, Zagreb, Croatia
| | - Slavko Gašparov
- University of Zagreb School of Medicine, Zagreb, Croatia
- Department of Pathology and Cytology, Merkur University Hospital, Zagreb, Croatia
| |
Collapse
|
810
|
Lv X, Han Z, Huo H, Liu X, Zhang J, Chi J, Han B, Jiang Z. Anti-inflammatory and antioxidant succinyl-chitosan oligosaccharide protects human epidermal cell and mouse skin against ultraviolet B-induced photodamage. Carbohydr Polym 2025; 351:123102. [PMID: 39779016 DOI: 10.1016/j.carbpol.2024.123102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 11/21/2024] [Accepted: 12/02/2024] [Indexed: 01/11/2025]
Abstract
Ultraviolet B (UVB) irradiation from sunlight is one of the primary environmental factors that causes photodamage to the skin. The aim of this study was to prepare succinyl-chitosan oligosaccharide (SU-COS) and evaluate its protective effects and related molecular mechanisms against UVB-induced photodamage for the first time. SU-COS (substitution degree: 69.26 %, molecular weight: 6400 Da) was shown to have excellent biocompatibility and to effectively promote the migration of human epidermal HaCaT cells. UVB irradiation was used to develop photodamage models in HaCaT cells and murine skin. In vitro, treatment with SU-COS significantly increased the cell survival ratio by >11 % while concurrently reducing reactive oxygen species formation, preserving normal cytoskeletal morphology, stabilizing the cell cycle, and decreasing the apoptosis ratio. The SU-COS intervention also demonstrated a reparative effect on UVB-damaged mouse skin by reducing skin erythema and water loss, relieving crusting, and maintaining the epidermis thickness of the skin and the stability of collagen fibers. The observed changes in both in vitro and in vivo results were accompanied by significant modulation of gene and protein expression associated with the cellular cycle, collagen synthesis, extracellular matrix degradation, antioxidant defense, and inflammation. Overall, SU-COS holds immense promise for applications in repairing photodamage.
Collapse
Affiliation(s)
- Xiansen Lv
- Laboratory of Biochemistry and Biomedical Materials, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, PR China
| | - Zi Han
- Laboratory of Biochemistry and Biomedical Materials, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, PR China
| | - Huilin Huo
- Laboratory of Biochemistry and Biomedical Materials, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, PR China
| | - Xiaofei Liu
- Laboratory of Biochemistry and Biomedical Materials, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, PR China
| | - Jinpeng Zhang
- Laboratory of Biochemistry and Biomedical Materials, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, PR China
| | - Jinhua Chi
- Laboratory of Biochemistry and Biomedical Materials, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, PR China
| | - Baoqin Han
- Laboratory of Biochemistry and Biomedical Materials, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, PR China
| | - Zhiwen Jiang
- Laboratory of Biochemistry and Biomedical Materials, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, PR China.
| |
Collapse
|
811
|
Harmon RM, Ayers JL, McCarthy EF, Kowalczyk AP, Green KJ, Simpson CL. Pumping the Breaks on Acantholytic Skin Disorders: Targeting Calcium Pumps, Desmosomes, and Downstream Signaling in Darier, Hailey-Hailey, and Grover Disease. J Invest Dermatol 2025; 145:494-508. [PMID: 39207315 PMCID: PMC11846705 DOI: 10.1016/j.jid.2024.06.1289] [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: 05/08/2024] [Revised: 06/11/2024] [Accepted: 06/14/2024] [Indexed: 09/04/2024]
Abstract
Acantholytic skin disorders, by definition, compromise intercellular adhesion between epidermal keratinocytes. The root cause of blistering in these diseases traces back to direct disruption of adhesive cell-cell junctions, exemplified by autoantibody-mediated attack on desmosomes in pemphigus. However, genetic acantholytic disorders originate from more indirect mechanisms. Darier disease and Hailey-Hailey disease arise from mutations in the endoplasmic reticulum calcium pump, SERCA2, and the Golgi calcium/manganese pump, SPCA1, respectively. Though the disease-causing mutations have been known for nearly 25 years, the mechanistic linkage between dysregulation of intracellular ion stores and weakening of cell-cell junctions at the plasma membrane remains puzzling. The molecular underpinnings of a related idiopathic disorder, Grover disease, are even less understood. Due to an incomplete understanding of acantholytic pathology at the molecular level, these disorders lack proven, targeted treatment options, leaving patients with the significant physical and psychological burdens of chronic skin blistering, infections, and pain. This article aims to review what is known at the molecular, cellular, and clinical levels regarding these under-studied disorders and to highlight knowledge gaps and promising ongoing research. Armed with this knowledge, our goal is to aid investigators in defining essential questions about disease pathogenesis and to accelerate progress toward novel therapeutic strategies.
Collapse
Affiliation(s)
- Robert M Harmon
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA; Department of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA.
| | - Jessica L Ayers
- Molecular Medicine and Mechanisms of Disease PhD Program, Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA; Department of Dermatology, University of Washington, Seattle, Washington, USA; Institute for Stem Cell & Regenerative Medicine, University of Washington, Seattle, Washington, USA
| | - Erin F McCarthy
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA; Department of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Andrew P Kowalczyk
- Department of Dermatology, Pennsylvania State University College of Medicine, Hershey, Pennsylvania, USA; Department of Cellular and Molecular Physiology, Pennsylvania State University College of Medicine, Hershey, Pennsylvania, USA
| | - Kathleen J Green
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA; Department of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Cory L Simpson
- Department of Dermatology, University of Washington, Seattle, Washington, USA; Institute for Stem Cell & Regenerative Medicine, University of Washington, Seattle, Washington, USA.
| |
Collapse
|
812
|
Pyreddy S, Kim S, Miyamoto W, Talib Z, GnanaDev DA, Rahnemai-Azar AA. Current Advances in Immunotherapy Management of Esophageal Cancer. Cancers (Basel) 2025; 17:851. [PMID: 40075698 PMCID: PMC11898678 DOI: 10.3390/cancers17050851] [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: 12/16/2024] [Revised: 02/03/2025] [Accepted: 02/10/2025] [Indexed: 03/14/2025] Open
Abstract
Esophageal cancer is one of the most common and deadliest cancers worldwide. Rates of esophageal cancer worldwide have been steadily rising over the past decade due to higher incidence of gastroesophageal reflux disease (GERD). Current therapies include surgical resection, chemotherapy, and limited targeted therapies. One obstacle to care is tumor cells' ability to evade immune surveillance, which can render certain therapeutics ineffective. Immunotherapy provides a new paradigm to cancer treatment, which has proven to be effective in evasive tumors. In recent years, PD-1/PD-L1 and CLTA-4 inhibitors have been used as frontline treatment and have shown to be extremely effective in the treatment of hard-to-treat tumors. Here, we aim to analyze the current literature regarding current therapeutics along with emerging techniques and future receptor targets for immunotherapy.
Collapse
Affiliation(s)
- Sagar Pyreddy
- School of Medicine, California University of Science and Medicine, Colton, CA 92324, USA; (S.P.); (S.K.); (W.M.); (Z.T.)
| | - Sarah Kim
- School of Medicine, California University of Science and Medicine, Colton, CA 92324, USA; (S.P.); (S.K.); (W.M.); (Z.T.)
| | - William Miyamoto
- School of Medicine, California University of Science and Medicine, Colton, CA 92324, USA; (S.P.); (S.K.); (W.M.); (Z.T.)
| | - Zohray Talib
- School of Medicine, California University of Science and Medicine, Colton, CA 92324, USA; (S.P.); (S.K.); (W.M.); (Z.T.)
| | - Dev A. GnanaDev
- Department of Surgery, Arrowhead Regional Medical Center, Colton, CA 92324, USA;
| | - Amir A. Rahnemai-Azar
- Division of Surgical Oncology, Department of Surgery, Arrowhead Regional Cancer Center, California University of Science and Medicine, Colton, CA 92324, USA
| |
Collapse
|
813
|
Käck H, Sjögren T. Macromolecular crystallography from an industrial perspective - the impact of synchrotron radiation on structure-based drug discovery. JOURNAL OF SYNCHROTRON RADIATION 2025; 32:294-303. [PMID: 39913304 PMCID: PMC11892899 DOI: 10.1107/s1600577524012281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Accepted: 12/19/2024] [Indexed: 03/11/2025]
Abstract
Structure-based drug design has been an integral part of drug discovery for over three decades, contributing to the development of numerous approved drugs. Here we discuss the evolution, as well as the current state, of structure-based drug design within the pharmaceutical industry, using data from AstraZeneca's internal repository for crystal structures to provide additional context. Over the past 20 years, the company has transitioned from a mixed in-house and synchrotron data collection model to a `synchrotron-only' approach, enabled by technological advancements at synchrotron facilities. We provide real-world examples of structure delivery to projects, including a high-throughput project and a case where a single structure was pivotal for discovering a candidate drug. We conclude that, despite recent developments in single-particle cryo-EM and deep-learning structure prediction methods, macromolecular crystallography remains a critical tool for drug discovery.
Collapse
Affiliation(s)
- H. Käck
- Protein Sciences, Structure and Biophysics, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Pepparedsleden 1, SE-431 50Gothenburg, Sweden
| | - T. Sjögren
- Protein Sciences, Structure and Biophysics, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Pepparedsleden 1, SE-431 50Gothenburg, Sweden
| |
Collapse
|
814
|
Huang S, Xu Z, Zhuang Y. The current landscape of 1,2,3-triazole hybrids with anticancer therapeutic potential: Part I. Arch Pharm (Weinheim) 2025; 358:e2500001. [PMID: 40079707 DOI: 10.1002/ardp.202500001] [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: 01/01/2025] [Revised: 01/26/2025] [Accepted: 02/18/2025] [Indexed: 03/15/2025]
Abstract
Cancer, with its steadily increasing morbidity and mortality, will continue to pose a threat to humanity over an extended period. Chemotherapeutics play an indispensable role in cancer treatment, and hundreds of drugs have been approved for this purpose. Nevertheless, the fight against cancer remains a formidable challenge. This is mainly due to the emergence of multidrug resistance and the severe side effects associated with currently available anticancer drugs. Consequently, there is an urgent imperative to explore novel chemotherapeutic agents. 1,2,3-Triazoles belong to one of the most privileged classes of nitrogen-containing five-membered heterocycles and are regarded as prominent sources for the development of innovative anticancer chemotherapeutics. 1,2,3-Triazole hybrids, which possess multitargeted mechanisms of action within the cancer progression pathway, hold the potential to overcome multidrug resistance and mitigate side effects. Furthermore, several 1,2,3-triazole hybrids have already been approved for cancer therapy or are currently under clinical evaluation. This clearly demonstrates that 1,2,3-triazole hybrids are valuable scaffolds in the treatment and eradication of cancer. This review aims to provide insights into the anticancer therapeutic potential of 1,2,3-triazole hybrids, along with their mechanisms of action, crucial aspects of design, and structure-activity relationships (SARs). It encompasses articles published from 2021 onward.
Collapse
Affiliation(s)
- Shanshan Huang
- School of Chemistry and Pharmaceutical Engineering, Huanghuai University, Zhumadian, Henan, China
| | - Zhi Xu
- Huanghuai University Industry Innovation & Research and Development Institute, Huanghuai University, Zhumadian, Henan, China
| | - Yafei Zhuang
- School of Pharmacy, Xianning Medical College, Hubei University of Science and Technology, Xianning, Hubei, China
| |
Collapse
|
815
|
Kashkush A, Furth‐Lavi J, Hodon J, Benhamou RI. PROTAC and Molecular Glue Degraders of the Oncogenic RNA Binding Protein Lin28. Macromol Biosci 2025; 25:e2400427. [PMID: 39575661 PMCID: PMC11904392 DOI: 10.1002/mabi.202400427] [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: 10/08/2024] [Revised: 11/06/2024] [Indexed: 03/14/2025]
Abstract
The interaction between proteins and RNA is crucial for regulating gene expression, with dysregulation often linked to diseases such as cancer. The RNA-binding protein (RBP) Lin28 inhibits the tumor suppressor microRNA (miRNA) let-7, making it a significant oncogenic factor in tumor progression and metastasis. In this study, a small molecule is used that binds Lin28 and blocks its inhibition of let-7. To enhance its efficay, the inhibitor is transformed into degraders via two degradation approaches: Proteolysis Targeting Chimera (PROTAC) and molecular glue. A series of PROTAC bifunctional molecules and molecular glues capable of degrading Lin28 in cells.is developed Both strategies significantly reduce overexpressed Lin28 and alleviate cancer cellular phenotypes. Notably, the molecular glue approach demonstrates exceptional potency, surpassing PROTAC in several aspects. This outcome underscores the superior efficiency of the molecular glue approach for targeted Lin28 degradation and highlights its potential for addressing associated diseases with small molecules. Innovative small molecule strategies such as molecular glue and PROTAC technology for targeted RBP degradation, hold promise for opening new avenues in RNA modulation and addressing related diseases.
Collapse
Affiliation(s)
- Aseel Kashkush
- The Institute for Drug Research of the School of Pharmacy, Faculty of MedicineThe Hebrew University of JerusalemHadassah‐Ein KeremJerusalem91120Israel
| | - Judith Furth‐Lavi
- The Institute for Drug Research of the School of Pharmacy, Faculty of MedicineThe Hebrew University of JerusalemHadassah‐Ein KeremJerusalem91120Israel
| | - Jiri Hodon
- The Institute for Drug Research of the School of Pharmacy, Faculty of MedicineThe Hebrew University of JerusalemHadassah‐Ein KeremJerusalem91120Israel
| | - Raphael I. Benhamou
- The Institute for Drug Research of the School of Pharmacy, Faculty of MedicineThe Hebrew University of JerusalemHadassah‐Ein KeremJerusalem91120Israel
| |
Collapse
|
816
|
Lu X, Liu F, Chen H, Cai H, Zhang L, Li J. Effects of WN1703 on Cardiovascular Function in Chronic Hyperuricemia Rats and Myocardial Injury Mechanism Exploration in H9C2 Cells. J Appl Toxicol 2025; 45:418-431. [PMID: 39435646 DOI: 10.1002/jat.4710] [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: 08/06/2024] [Revised: 09/09/2024] [Accepted: 09/25/2024] [Indexed: 10/23/2024]
Abstract
Hyperuricemia, a prevalent condition, is typically preceded by disturbances in purine metabolism and is frequently associated with hyperlipidemia and other dysfunctions of metabolism. WN1703 demonstrated an inhibitory activity against xanthine oxidoreductase (XOR) that was comparable to febuxostat in our prior investigation. In this study, we assessed the cardiovascular safety of WN1703 in a chronic hyperuricemia rat model induced by potassium oxonate in combination with hypoxanthine. We investigated the changes in cardiovascular biomarkers in chronic hyperuricemia rats treated with febuxostat and WN1703, including creatine kinase (CK), CK-MB, B type natriuretic peptide (BNP), Corin protein (CRN), Neprilysin (NEP), myeloperoxidase (MPO), 8-hydroxy-2-deoxyguanosine (8-OHdG), tumor necrosis factor (TNF-α), interleukin-1β (IL-1β), and interleukin-8 (IL-8). Additionally, we validated the potential mechanism of cardiac injury induced by WN1703 in H9C2 cells, guided by cardiotoxicity predictions from the cardioToxCSM database and network pharmacology. We observed that excessively rapid urate-lowering, oxidative stress, and inflammation could disrupt myocardial functional homeostasis and increase the risk of cardiovascular injury in hyperuricemia rats, and WN1703 treatment effectively reduced the levels oxidative stress marker 8-OHdG and inflammatory factor TNF-α. Despite the absence of organic damage to the heart with prolonged treatment of febuxostat and WN1703, potential hazard of cardiovascular injury could be associated with the modulation of the TGFβ and RHO/ROCK signaling pathways by febuxostat and WN1703. This could offer new insights into the mechanisms underlying the adverse effects caused by XOR inhibitors.
Collapse
Affiliation(s)
- Xiaodan Lu
- MOE International Joint Research Laboratory on Synthetic Biology and Medicines, School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
| | - Fuyao Liu
- MOE International Joint Research Laboratory on Synthetic Biology and Medicines, School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
| | - Hongming Chen
- MOE International Joint Research Laboratory on Synthetic Biology and Medicines, School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
| | - Haojie Cai
- MOE International Joint Research Laboratory on Synthetic Biology and Medicines, School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
| | - Lei Zhang
- MOE International Joint Research Laboratory on Synthetic Biology and Medicines, School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
| | - Jing Li
- MOE International Joint Research Laboratory on Synthetic Biology and Medicines, School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
| |
Collapse
|
817
|
Angeles Flores G, Cusumano G, Venanzoni R, Angelini P. Advancements in Antibacterial Therapy: Feature Papers. Microorganisms 2025; 13:557. [PMID: 40142450 PMCID: PMC11945154 DOI: 10.3390/microorganisms13030557] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 02/21/2025] [Accepted: 02/27/2025] [Indexed: 03/28/2025] Open
Abstract
Antimicrobial resistance (AMR) is a growing global health crisis that threatens the efficacy of antibiotics and modern medical interventions. The emergence of multidrug-resistant (MDR) pathogens, exacerbated by the misuse of antibiotics in healthcare and agriculture, underscores the urgent need for innovative solutions. (1) Background: AMR arises from complex interactions between human, animal, and environmental health, further aggravated by the overuse and inadequate regulation of antibiotics. Conventional treatments are increasingly ineffective, necessitating alternative strategies. Emerging approaches, including bacteriophage therapy, antimicrobial peptides (AMPs), nanotechnology, microbial extracellular vesicles (EVs), and CRISPR-based antimicrobials, provide novel mechanisms that complement traditional antibiotics in combating resistant pathogens. (2) Methods: This review critically analyzes advanced antibacterial strategies in conjunction with systemic reforms such as antimicrobial stewardship programs, the One Health framework, and advanced surveillance tools. These methods can enhance resistance detection, guide interventions, and promote sustainable practices. Additionally, economic, logistical, and regulatory challenges impeding their implementation are evaluated. (3) Results: Emerging technologies, such as CRISPR and nanotechnology, exhibit promising potential in targeting resistance mechanisms. However, disparities in resource distribution and regulatory barriers hinder widespread adoption. Public-private partnerships and sustainable agriculture practices are critical to overcoming these obstacles. (4) Conclusions: A holistic and integrated approach is essential for mitigating the impact of AMR. By aligning innovative therapeutic strategies with global health policies, fostering interdisciplinary collaboration, and ensuring equitable resource distribution, we can develop a sustainable response to this 21st-century challenge.
Collapse
Affiliation(s)
- Giancarlo Angeles Flores
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Via del Giochetto, 06122 Perugia, Italy; (G.A.F.); (G.C.); (R.V.)
- Centro di Ricerca per l’Innovazione, Digitalizzazione, Valorizzazione e Fruizione del Patrimonio Culturale e Ambientale (CE.D.I.PA.), Piazza San Gabriele dell’Addolorata, 4, 06049 Spoleto, Italy
| | - Gaia Cusumano
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Via del Giochetto, 06122 Perugia, Italy; (G.A.F.); (G.C.); (R.V.)
| | - Roberto Venanzoni
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Via del Giochetto, 06122 Perugia, Italy; (G.A.F.); (G.C.); (R.V.)
| | - Paola Angelini
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Via del Giochetto, 06122 Perugia, Italy; (G.A.F.); (G.C.); (R.V.)
- Centro di Ricerca per l’Innovazione, Digitalizzazione, Valorizzazione e Fruizione del Patrimonio Culturale e Ambientale (CE.D.I.PA.), Piazza San Gabriele dell’Addolorata, 4, 06049 Spoleto, Italy
| |
Collapse
|
818
|
Yang S, Hu J. Comparison of postoperative analgesia between dezocine plus flurbiprofen axetil and sufentanil in patients with CRC undergoing tumor resection: A prospective, observational study. Oncol Lett 2025; 29:121. [PMID: 39807102 PMCID: PMC11726305 DOI: 10.3892/ol.2025.14869] [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: 09/06/2024] [Accepted: 12/04/2024] [Indexed: 01/16/2025] Open
Abstract
Flurbiprofen axetil is a nonsteroidal anti-inflammatory drug used for analgesia. Its combination with dezocine has previously shown a superior postoperative analgesic effect compared with that of opioids. The present study compared the analgesic effect between dezocine plus flurbiprofen axetil (DFA) and sufentanil in patients with colorectal cancer (CRC) following resection of the tumor. The study was performed as a prospective, observational study. It included 107 patients who were treated using a patient-controlled analgesia (PCA) pump following the resection of CRC. Patients in the DFA group were given a loading dose of 5 mg dezocine and 50 mg flurbiprofen axetil, followed by PCA with a combination comprising 30 mg dezocine, 200 mg flurbiprofen axetil and 8 mg ondansetron. Patients in the control group were treated with sufentanil at a loading dose of 5-10 µg followed by PCA with a combination of 100 µg sufentanil and 8 mg ondansetron. The DFA group reported lower pain numerical rating scale scores at 2 h (2.4±1.2 vs. 2.9±1.2) and 12 h (2.0±1.0 vs. 2.5±1.2) and reduced rates of moderate-to-severe pain at 12 h (6.7 vs. 21.0%) compared with those in the control group. In addition, the number of PCA boluses in the DFA group was lower than that in the control group [median (interquartile range), 6.0 (4.5-8.5) vs. 8.5 (5.0-11.0)]. The total satisfaction rate was increased, albeit not significantly, in the DFA group compared with that in the control group (80.0 vs. 62.9%). The levels of tumor necrosis factor-α at 24 and 48 h, and of interleukin-6 at 24 h were decreased in the DFA group compared with those in the control group. The incidences of adverse events did not differ between the groups. These findings indicate that DFA provides more effective analgesia, improves patient satisfaction and reduces the levels of pro-inflammatory cytokines with similar adverse effects compared with those of sufentanil in patients after CRC resection.
Collapse
Affiliation(s)
- Sufang Yang
- Department of Anesthesiology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, Zhejiang 323000, P.R. China
| | - Jingchun Hu
- Department of Anesthesiology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, Zhejiang 323000, P.R. China
| |
Collapse
|
819
|
Wang W, Yi X, Zhou R, Peng W, Huang J, Chen J, Bo R, Liu M, Li J. Tea tree oil nanoemulsion targets AgrA protein potentiates amoxicillin efficacy against methicillin-resistant Staphylococcus aureus. Int J Biol Macromol 2025; 292:139111. [PMID: 39733883 DOI: 10.1016/j.ijbiomac.2024.139111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 12/06/2024] [Accepted: 12/21/2024] [Indexed: 12/31/2024]
Abstract
The excessive utilization of antibiotics gives rise to the development of bacterial resistance, the deterioration of animal immune functions, the increase in mortality rates, and the undermining of human immunity. Therefore, there is an urgent necessity to explore new antimicrobial agents or alternatives to tackle bacterial resistance. We investigated tea tree oil (TTO), a pure natural plant essential oil extracted from Melaleuca leaves, which exerted efficient antibacterial activities. However, the poor solubility and high volatility of TTO limited the clinical application. Therefore, tea tree oil and Tween 80 were formulated into a stable nanoemulsion (Nano TTO). We attested that Nano TTO, as an antibiotic adjuvant, enhanced the antibacterial activity of amoxicillin (AMX) against methicillin-resistant Staphylococcus aureus (MRSA) and inhibited the formation of biofilms. Mechanistic studies proved that the Nano TTO potentiation effect on AMX was primarily the result of inhibition of the Agr expression by targeting the accessory regulator AgrA. Furthermore, Nano TTO effectively boosts the efficacy of amoxicillin in the mouse septicaemia model and mouse skin wound infection model. Overall, these results revealed the potential of Nano TTO as an adjuvant to evade multidrug-resistant bacterial pathogens and improve treatment outcomes for drug-resistant infections.
Collapse
Affiliation(s)
- Weimei Wang
- School of Veterinary Medicine, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, PR China
| | - Xiaobin Yi
- School of Veterinary Medicine, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, PR China
| | - Ruigang Zhou
- School of Veterinary Medicine, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, PR China
| | - Weilong Peng
- School of Veterinary Medicine, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, PR China
| | - Junjie Huang
- School of Veterinary Medicine, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, PR China
| | - Jun Chen
- School of Veterinary Medicine, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, PR China
| | - Ruonan Bo
- School of Veterinary Medicine, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, PR China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu 225009, PR China
| | - Mingjiang Liu
- School of Veterinary Medicine, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, PR China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu 225009, PR China
| | - Jingui Li
- School of Veterinary Medicine, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, PR China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu 225009, PR China.
| |
Collapse
|
820
|
Dalix E, Marotte H. From a better knowledge of periodontal disease to Porphyromonas gingivalis target for rheumatoid arthritis disease activity. Joint Bone Spine 2025; 92:105822. [PMID: 39551151 DOI: 10.1016/j.jbspin.2024.105822] [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: 02/19/2024] [Revised: 10/19/2024] [Accepted: 10/23/2024] [Indexed: 11/19/2024]
Abstract
Periodontal disease (PD) and rheumatoid arthritis (RA) are both inflammatory diseases affecting the tooth and joint, with local inflammation associated with bone loss. Bacterial infections by oral bacteria are involved in periodontal inflammation, and the best known to be associated with PD is Porphyromonas gingivalis (Pg). A large body of recent data suggests a strong involvement of this specific bacteria, Pg, in PD outcomes, but also in RA. The aim of this review is to discuss the association between PD and Pg, RA and its mechanisms, and to determine whether targeting Pg bacteria could improve RA. Numerous epidemiological studies have already confirmed the association between PD and Pg, as well as between PD and RA, which is mainly associated with a common genetic background, the shared epitope. The involvement of Pg in pathogenesis of RA is supported by the induction of gingival citrullinated proteins and therefore of anti-citrullinated proteins antibodies, which constitute the most specific biomarker of RA. The prevalence of Pg in RA is still controversial, but studies should include patients with preclinical and early RA. Finally, recent data confirmed that targeting Pg is highly effective in improving RA.
Collapse
Affiliation(s)
- Elisa Dalix
- Inserm, SAINBIOSE U1059, Mines Saint-Étienne, Université Jean-Monnet Saint-Étienne, 42023 Saint-Étienne, France.
| | - Hubert Marotte
- Inserm, SAINBIOSE U1059, Rheumatology Departement, Mines Saint-Étienne, Université Jean-Monnet Saint-Étienne, CHU de Saint-Etienne, 42023 Saint-Étienne, France.
| |
Collapse
|
821
|
Sun R, Wang Z, Li M, Du T, Jia S, Yang W, Yang L. Regulatory Effects of Copper on Ghrelin Secretion in Rat Fundic Glands. J Anim Physiol Anim Nutr (Berl) 2025; 109:521-532. [PMID: 39545633 DOI: 10.1111/jpn.14068] [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: 08/06/2024] [Revised: 10/25/2024] [Accepted: 10/28/2024] [Indexed: 11/17/2024]
Abstract
Copper (Cu) is an effective additive in feed for promoting growth. Growth dan axis comprising growth hormone (GH), somatostatin (SS) and GH-releasing hormone (GHRH), with ghrelin regulating their release. The growth-promoting effects of Cu are closely related to ghrelin, but the specific mechanism behind the relationship remains unknown. We investigated the adjustment of ghrelin synthesis and secretion by Cu. Sprague-Dawley rats were fed basal diets with an addition of 0, 120 or 240 mg/kg Cu sulfate for 28 day to establish a growth-promoting model. Signalling molecules relevant to ghrelin synthesis and secretion were detected and mechanistically explored using enzyme-linked immunosorbent assay, quantitative reverse-transcription polymerase chain reaction and Western blot analysis. The 120 mg/kg supplement improved growth performance; significantly increased the serum levels of ghrelin, ghrelin O-acyltransferase (GOAT), acylated ghrelin (AG), GH, and reactive oxygen species (ROS) and decreased those of SS; significantly increased the mRNA and protein expression of ghrelin, GOAT, ghrelin receptor (GHS-R1α), and activator protein 1 (AP-1); increased the phosphorylation ratio of JNK and p38 MAPK; and inhibited the mRNA and protein expression of SS and SS receptor subtype 2 (SSTR2) in gastric fundic gland tissues. Thus, Cu may affect gastric ghrelin synthesis at the transcriptional level by activating the JNK/p38 MAPK pathway through increased ROS levels and regulating the activation of the downstream redox-sensitive transcription factor AP-1. SS plays a crucial determinant role in ghrelin regulation via intragastric Cu. Cu promotes GOAT activity and ghrelin secretion by inhibiting SS secretion, affecting AG levels, and promoting ghrelin acylation through ghrelin/GOAT/GHS-R1α system, modulating ghrelin secretion.
Collapse
Affiliation(s)
- Rui Sun
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, Jilin Province, China
- Jilin Provincial Key Lab of Animal Nutrition and Feed Science, Jilin Agricultural University, Changchun, Jilin Province, China
- Key Laboratory of Animal Production, Product Quality and Security of Ministry of Education, Changchun, Jilin Province, China
| | - Zhongshen Wang
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, Jilin Province, China
- Jilin Provincial Key Lab of Animal Nutrition and Feed Science, Jilin Agricultural University, Changchun, Jilin Province, China
- Key Laboratory of Animal Production, Product Quality and Security of Ministry of Education, Changchun, Jilin Province, China
| | - Meng Li
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, Jilin Province, China
- Jilin Provincial Key Lab of Animal Nutrition and Feed Science, Jilin Agricultural University, Changchun, Jilin Province, China
- Key Laboratory of Animal Production, Product Quality and Security of Ministry of Education, Changchun, Jilin Province, China
| | - Tianyang Du
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, Jilin Province, China
- Jilin Provincial Key Lab of Animal Nutrition and Feed Science, Jilin Agricultural University, Changchun, Jilin Province, China
- Key Laboratory of Animal Production, Product Quality and Security of Ministry of Education, Changchun, Jilin Province, China
| | - Shuang Jia
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, Jilin Province, China
- Jilin Provincial Key Lab of Animal Nutrition and Feed Science, Jilin Agricultural University, Changchun, Jilin Province, China
- Key Laboratory of Animal Production, Product Quality and Security of Ministry of Education, Changchun, Jilin Province, China
| | - Wenyan Yang
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, Jilin Province, China
- Jilin Provincial Key Lab of Animal Nutrition and Feed Science, Jilin Agricultural University, Changchun, Jilin Province, China
- Key Laboratory of Animal Production, Product Quality and Security of Ministry of Education, Changchun, Jilin Province, China
| | - Lianyu Yang
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, Jilin Province, China
- Jilin Provincial Key Lab of Animal Nutrition and Feed Science, Jilin Agricultural University, Changchun, Jilin Province, China
- Key Laboratory of Animal Production, Product Quality and Security of Ministry of Education, Changchun, Jilin Province, China
| |
Collapse
|
822
|
Maehara S, Kumamoto M, Nakajima S, Hieda Y, Watashi K, Hata T. Potent SARS-CoV-2 3C-like protease inhibitor ( +)-eupenoxide-3,6-diketone (IC 50: 0.048 μM) was synthesized based on ( +)-eupenoxide; lead from ( +)-eupenoxide analogs study by endophytic fermentation. J Nat Med 2025; 79:357-370. [PMID: 39899217 DOI: 10.1007/s11418-024-01874-3] [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: 11/24/2024] [Accepted: 12/21/2024] [Indexed: 02/04/2025]
Abstract
Since the coronavirus disease 2019 (COVID-19) outbreak, research has been conducted on treatment and countermeasures against the causative severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). However, the development of new seeds is urgently needed because viruses have the characteristic of becoming resistant through mutation. We hypothesize that endophytes produce antiviral substances to combat foreign viruses in host plants. According to this hypothesis, the seeds of therapeutic agents for infectious diseases could be obtained from endophytes by culture experiments. This report found that Aspergillus sp. endophyte isolated from Catharanthus roseus produced ( +)-eupenoxide and its 3-ketone form with anti-SARS-CoV-2 activity. In addition, ( +)-eupenoxide-3,6-diketon was discovered as a new compound with potent 3C-like protease inhibitory activity (IC50: 0.048 μM) by synthesis based on ( +)-eupenoxide. This finding could be an important evidence that endophytic fungi symbiosis with medicinal plants is useful as antiviral producers.
Collapse
Affiliation(s)
- Shoji Maehara
- Faculty of Pharmacy and Pharmaceutical Sciences, Fukuyama University, Sanzo, 1 Gakuen-cho, Fukuyama, Hiroshima, 729-0292, Japan.
| | - Moeka Kumamoto
- Faculty of Pharmacy and Pharmaceutical Sciences, Fukuyama University, Sanzo, 1 Gakuen-cho, Fukuyama, Hiroshima, 729-0292, Japan
| | - Shogo Nakajima
- Department of Virology II, National Institute of Infectious Diseases, Toyama 1-23-1, Shinjuku-ku, Tokyo, 162-8640, Japan
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Toyama 1-23-1, Shinjuku-ku, Tokyo, 162-8640, Japan
| | - Yuhzo Hieda
- Common Resources Center, Fukuyama University, 985-1, Sanzo, Higashimura-cho, Fukuyama, Hiroshima, 729-0292, Japan
| | - Koichi Watashi
- Department of Virology II, National Institute of Infectious Diseases, Toyama 1-23-1, Shinjuku-ku, Tokyo, 162-8640, Japan
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Toyama 1-23-1, Shinjuku-ku, Tokyo, 162-8640, Japan
| | - Toshiyuki Hata
- Faculty of Pharmacy and Pharmaceutical Sciences, Fukuyama University, Sanzo, 1 Gakuen-cho, Fukuyama, Hiroshima, 729-0292, Japan
| |
Collapse
|
823
|
Jia D, Deng R, Wang W, Hu H, Zhang X. Metabolic engineering of Pseudomonas chlororaphis P3 for high-level and directed production of phenazine-1,6-dicarboxylic acid from crude glycerol. BIORESOURCE TECHNOLOGY 2025; 419:132053. [PMID: 39798811 DOI: 10.1016/j.biortech.2025.132053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 12/01/2024] [Accepted: 01/08/2025] [Indexed: 01/15/2025]
Abstract
Phenazine-1,6-dicarboxylic acid (PDC) is a precursor of complex substituted phenazines used as pesticides and pharmaceuticals. The PDC biosynthesis exists the low production and the high proportion of by-products phenazine-1-carboxylic acid (PCA) derivatives in Pseudomonas P3△A. Herein, PDC production were improved by systematic metabolic engineering and synthetic regulation. The directed PDC biosynthesis was achieved by introducing the isozymes of PhzF', and PCA derivatives was barely detectable. Subsequently, a high-level PDC-producing strain P3FK2E-aF'EC was obtained by co-overexpression of aroE, phzE, phzC, and aphzF' in a multi-knockout strain. Through scale-up culture, the highest PDC production and proportion reached 6,447.05 mg/L and 99.68 %, with the productivity of 89.54 mg/L·h using KB. Economically, PDC production achieved 5,584.35 mg/L accounting for 99.43 % with the highest productivity of 108.32 mg/L·h from crude glycerol. This study first achieved the directed high-level production of PDC from renewable energy, and presented a potential biosynthesis platform for PDC derivatives in Pseudomonas.
Collapse
Affiliation(s)
- Dan Jia
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Ruxiang Deng
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Wei Wang
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Hongbo Hu
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China; National Experimental Teaching Center for Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Xuehong Zhang
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China; National Experimental Teaching Center for Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China.
| |
Collapse
|
824
|
Levinstein MR, Budinich RC, Bonaventura J, Schatzberg AF, Zarate CA, Michaelides M. Redefining Ketamine Pharmacology for Antidepressant Action: Synergistic NMDA and Opioid Receptor Interactions? Am J Psychiatry 2025; 182:247-258. [PMID: 39810555 PMCID: PMC11872000 DOI: 10.1176/appi.ajp.20240378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/16/2025]
Abstract
Ketamine is a racemic compound and medication comprised of (S)-ketamine and (R)-ketamine enantiomers and its metabolites. It has been used for decades as a dissociative anesthetic, analgesic, and recreational drug. More recently, ketamine, its enantiomers, and its metabolites have been used or are being investigated for the treatment of refractory depression, as well as for comorbid disorders such as anxiety, obsessive-compulsive, and opioid use disorders. Despite its complex pharmacology, ketamine is referred to as an N-methyl-d-aspartate (NMDA) receptor antagonist. In this review, the authors argue that ketamine's pharmacology should be redefined to include opioid receptors and the endogenous opioid system. They also highlight a potential mechanism of action of ketamine for depression that is attributed to bifunctional, synergistic interactions involving NMDA and opioid receptors.
Collapse
Affiliation(s)
- Marjorie R. Levinstein
- Biobehavioral Imaging & Molecular Neuropsychopharmacology Section, National Institute on Drug Abuse, Baltimore, MD, USA
| | - Reece C. Budinich
- Biobehavioral Imaging & Molecular Neuropsychopharmacology Section, National Institute on Drug Abuse, Baltimore, MD, USA
| | - Jordi Bonaventura
- Departament de Patologia i Terapèutica Experimental, Institut de Neurociències, Universitat de Barcelona, L’Hospitalet de Llobregat
- Neuropharmacology and Pain Group, Neuroscience Program, IDIBELL-Institut d’Investigació Biomèdica de Bellvitge, L’Hospitalet de Llobregat
| | - Alan F. Schatzberg
- Department of Psychiatry and Behavioral Sciences, Stanford School of Medicine, Stanford University, Stanford, California, USA
| | - Carlos A. Zarate
- Experimental Therapeutics & Pathophysiology Branch, National Institute of Mental Health, Bethesda, MD, USA
| | - Michael Michaelides
- Biobehavioral Imaging & Molecular Neuropsychopharmacology Section, National Institute on Drug Abuse, Baltimore, MD, USA
- Department of Psychiatry & Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
825
|
Yu Z, Lepoitevin M, Serre C. Iron-MOFs for Biomedical Applications. Adv Healthc Mater 2025; 14:e2402630. [PMID: 39388416 PMCID: PMC11937880 DOI: 10.1002/adhm.202402630] [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: 07/17/2024] [Revised: 08/29/2024] [Indexed: 10/12/2024]
Abstract
Over the past two decades, iron-based metal-organic frameworks (Fe-MOFs) have attracted significant research interest in biomedicine due to their low toxicity, tunable degradability, substantial drug loading capacity, versatile structures, and multimodal functionalities. Despite their great potential, the transition of Fe-MOFs-based composites from laboratory research to clinical products remains challenging. This review evaluates the key properties that distinguish Fe-MOFs from other MOFs and highlights recent advances in synthesis routes, surface engineering, and shaping technologies. In particular, it focuses on their applications in biosensing, antimicrobial, and anticancer therapies. In addition, the review emphasizes the need to develop scalable, environmentally friendly, and cost-effective production methods for additional Fe-MOFs to meet the specific requirements of various biomedical applications. Despite the ability of Fe-MOFs-based composites to combine therapies, significant hurdles still remain, including the need for a deeper understanding of their therapeutic mechanisms and potential risks of resistance and overdose. Systematically addressing these challenges could significantly enhance the prospects of Fe-MOFs in biomedicine and potentially facilitate their integration into mainstream clinical practice.
Collapse
Affiliation(s)
- Zhihao Yu
- Institut des Matériaux Poreux de ParisENSESPCI ParisCNRSPSL UniversityParisFrance
| | - Mathilde Lepoitevin
- Institut des Matériaux Poreux de ParisENSESPCI ParisCNRSPSL UniversityParisFrance
| | - Christian Serre
- Institut des Matériaux Poreux de ParisENSESPCI ParisCNRSPSL UniversityParisFrance
| |
Collapse
|
826
|
Myrsing E, Mouli HMC, Nikhil P, Deepali, Sahu A, Jana A, Ramalingam P. Protein profiling uncovers IGF-1R inhibition potential of 3-(2-furoyl)-indole scaffolds in hepatocellular carcinoma. Future Med Chem 2025; 17:513-528. [PMID: 40028717 PMCID: PMC11906113 DOI: 10.1080/17568919.2025.2467616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Accepted: 02/04/2025] [Indexed: 03/05/2025] Open
Abstract
AIM This study investigates the anti-proliferative potential and possible molecular mechanisms of 3-(2-furoyl)-indole derivatives against HepG2. METHOD Identified hit compounds (4a, 4b, 4c) using MTT screening, were further investigated for their efficacy and mechanism of action through FACS studies, in-silico molecular docking, molecular dynamics (MD) simulations, and label-free quantitative proteome and ADMET prediction. RESULTS Lead compound 4a, showed IC50 of 27 µM against HepG2 cells and a binding score of -8.077 kcal/mol against IGF-1 R (PDB ID: 5XFS) and formed a stable complex 100 ns. Proteomic study revealed significant downregulation of the IGF-1 R downstream signaling molecules and showed minimal toxicity and favorable drug-like properties. CONCLUSION These findings suggest that 4a is a promising IGF-1 R inhibitor and potential drug candidate against drug resistance hepatocellular carcinoma (HCC).
Collapse
Affiliation(s)
- Efficiency Myrsing
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research (NIPER), Hajipur, India
| | - H M Chandra Mouli
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research (NIPER), Hajipur, India
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Pallaprolu Nikhil
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research (NIPER), Hajipur, India
| | - Deepali
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research (NIPER), Hajipur, India
| | - Abhishek Sahu
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research (NIPER), Hajipur, India
| | - Anupam Jana
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research (NIPER), Hajipur, India
| | - P Ramalingam
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research (NIPER), Hajipur, India
| |
Collapse
|
827
|
Wang L, Sooram B, Kumar R, Schedin‐Weiss S, Tjernberg LO, Winblad B. Tau degradation in Alzheimer's disease: Mechanisms and therapeutic opportunities. Alzheimers Dement 2025; 21:e70048. [PMID: 40109019 PMCID: PMC11923393 DOI: 10.1002/alz.70048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 02/05/2025] [Accepted: 02/05/2025] [Indexed: 03/22/2025]
Abstract
In Alzheimer's disease (AD), tau undergoes abnormal post-translational modifications and aggregations. Impaired intracellular degradation pathways further exacerbate the accumulation of pathological tau. A new strategy - targeted protein degradation - recently emerged as a modality in drug discovery where bifunctional molecules bring the target protein close to the degradation machinery to promote clearance. Since 2016, this strategy has been applied to tau pathologies and attracted broad interest in academia and the pharmaceutical industry. However, a systematic review of recent studies on tau degradation mechanisms is lacking. Here we review tau degradation mechanisms (the ubiquitin-proteasome system and the autophagy-lysosome pathway), their dysfunction in AD, and tau-targeted degraders, such as proteolysis-targeting chimeras and autophagy-targeting chimeras. We emphasize the need for a continuous exploration of tau degradation mechanisms and provide a future perspective for developing tau-targeted degraders, encouraging researchers to work on new treatment options for AD patients. HIGHLIGHTS: Post-translational modifications, aggregation, and mutations affect tau degradation. A vicious circle exists between impaired degradation pathways and tau pathologies. Ubiquitin plays an important role in complex degradation pathways. Tau-targeted degraders provide promising strategies for novel AD treatment.
Collapse
Affiliation(s)
- Lisha Wang
- Division of NeurogeriatricsDepartment of Neurobiology, Care Sciences and SocietyKarolinska InstitutetSolnaSweden
| | - Banesh Sooram
- Division of NeurogeriatricsDepartment of Neurobiology, Care Sciences and SocietyKarolinska InstitutetSolnaSweden
| | - Rajnish Kumar
- Division of NeurogeriatricsDepartment of Neurobiology, Care Sciences and SocietyKarolinska InstitutetSolnaSweden
- Department of Pharmaceutical Engineering & TechnologyIndian Institute of Technology (BHU)VaranasiIndia
| | - Sophia Schedin‐Weiss
- Division of NeurogeriatricsDepartment of Neurobiology, Care Sciences and SocietyKarolinska InstitutetSolnaSweden
| | - Lars O. Tjernberg
- Division of NeurogeriatricsDepartment of Neurobiology, Care Sciences and SocietyKarolinska InstitutetSolnaSweden
| | - Bengt Winblad
- Division of NeurogeriatricsDepartment of Neurobiology, Care Sciences and SocietyKarolinska InstitutetSolnaSweden
- Theme Inflammation and AgingKarolinska University HospitalHuddingeSweden
| |
Collapse
|
828
|
Ebrahim MA, Ramsis TM, Gohar NA, Metwally SA, Rushdi A, Fayed EA. Novel Pyrrolidine-bearing quinoxaline inhibitors of DNA Gyrase, RNA polymerase and spike glycoprotein. Bioorg Chem 2025; 156:108218. [PMID: 39879826 DOI: 10.1016/j.bioorg.2025.108218] [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: 11/24/2024] [Revised: 12/31/2024] [Accepted: 01/25/2025] [Indexed: 01/31/2025]
Abstract
Anti-infective agents are a class of drugs used to prevent, treat, or control infections caused by microorganisms such as bacteria, viruses, fungi, and parasites. They play a crucial role in modern medicine, helping to reduce the severity of infections and, in many cases, save lives. This study aims at the design and synthesis of hybrid compounds containing quinoxaline, pyrrolidine, and an azo bridge to combat antimicrobial resistance, and evaluating their antimicrobial, antifungal, and antiviral activities against various pathogenic strains. Eight most potent bactericidal derivatives 2, 4, 5, 7, 9, 11, 12, and 13 were further assessed for their antibiofilm activity. Additionally, these compounds were tested for their inhibitory effects on DNA gyrase using a DNA supercoiling assay with IC50 ranging from 26.57 to 84.84 μM when compared to ciprofloxacin as standard drug. The antiviral activities were performed against HSV-1, H1N1 and SARS-CoV-2 viruses, which showed that compound 9 has the highest antiviral activity with IC50 = 0.32 µM, IC50 = 1.76 µM and 1.06 µM, respectively, as well as the best safety profile with CC50 = 30000 µM. Compound 9 displayed the highest SI value against HSV-1, H1N1 and SARS-CoV-2 with values of 93685, 17,034 and 28368, respectively. Compound 9 inhibited RdRp and spike glycoprotein (IC50 = 2.437 ± 0.102 and 1425.1 ± 55.3 nM; respectively). The physicochemical and pharmacokinetic properties of the most active compounds were screened to identify those with optimal drug-like characteristics. Molecular docking studies were conducted on the most effective compounds to elucidate their binding interactions and mechanisms of action.
Collapse
Affiliation(s)
- Maha A Ebrahim
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo 11754 Egypt
| | - Triveena M Ramsis
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Sinai University, Kantara Branch, Ismailia 41636 Egypt
| | - Nirvana A Gohar
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Modern University for Technology and Information (MTI), Cairo 11571 Egypt
| | - Shimaa A Metwally
- Microbiology and Immunology Department, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo 11884 Egypt
| | - Areej Rushdi
- Department of Medical Microbiology and Immunology, Faculty of Medicine for Girls, Al-Azhar University, Cairo 11651 Egypt
| | - Eman A Fayed
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo 11754 Egypt.
| |
Collapse
|
829
|
Lemos R, Pérez-Badell Y, De Nisco M, Carpentieri A, Suárez M, Pedatella S. Organic Chimeras Based on Selenosugars, Steroids, and Fullerenes as Potential Inhibitors of the β-amyloid Peptide Aggregation. Chempluschem 2025; 90:e202400404. [PMID: 39235155 DOI: 10.1002/cplu.202400404] [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: 06/10/2024] [Revised: 09/04/2024] [Accepted: 09/04/2024] [Indexed: 09/06/2024]
Abstract
The aggregation of β-amyloid peptide (Aβ) is associated with neurodegenerative diseases such as Alzheimer's disease (AD). Several therapies aimed at reducing the aggregation of this peptide have emerged as potential strategies for the treatment of AD. This paper describes the design and preparation of new hybrid molecules based on steroids, selenosugars, and [60]fullerene as potential inhibitors of Aβ oligomerization. These moieties were selected based on their antioxidant properties and possible areas of interaction with the Aβ. Cyclopropanations between C60 and malonates bearing different steroid and selenosugar moieties using the Bingel-Hirsch protocol have enabled the synthesis of functionalized molecular hybrids. The obtained derivatives were characterized by physical and spectroscopic techniques. Theoretical calculations for all the selenium compounds were performed using the density functional theory DFT/B3LYP-D3(BJ)/6-311G(2d,p) predicting the most stable conformations of the synthesized derivatives. Relevant geometrical parameters were investigated to relate the stereochemical behavior and the spectroscopic data obtained. The affinity of the compounds for Aβ-peptide was estimated by molecular docking simulation, which predicted an increase in affinity and interactions for Aβ for the hybrids containing the C60 core. In addition, parameters such as lipophilicity, polar surface area, and dipole moment were calculated to predict their potential interaction with membrane cells.
Collapse
Affiliation(s)
- Reinier Lemos
- Laboratorio de Síntesis Orgánica, Facultad de Química, Universidad de la Habana, 10400, La Habana, Cuba
- Department of Chemical Sciences, University of Napoli Federico II, Via Cintia 4, I-80126, Napoli, Italy
| | - Yoana Pérez-Badell
- Laboratorio de Química Computacional y Teórica, Facultad de Química, Universidad de La Habana, 10400, La Habana, Cuba
| | - Mauro De Nisco
- Department of Chemical Sciences, University of Basilicata, Viale dell'Ateneo Lucano 10, I-85100, Potenza, Italy
| | - Andrea Carpentieri
- Department of Chemical Sciences, University of Napoli Federico II, Via Cintia 4, I-80126, Napoli, Italy
| | - Margarita Suárez
- Laboratorio de Síntesis Orgánica, Facultad de Química, Universidad de la Habana, 10400, La Habana, Cuba
| | - Silvana Pedatella
- Department of Chemical Sciences, University of Napoli Federico II, Via Cintia 4, I-80126, Napoli, Italy
| |
Collapse
|
830
|
Shin JH, Yoo HB, Roe JS. Current advances and future directions in targeting histone demethylases for cancer therapy. Mol Cells 2025; 48:100192. [PMID: 39938867 PMCID: PMC11889978 DOI: 10.1016/j.mocell.2025.100192] [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: 12/13/2024] [Revised: 01/23/2025] [Accepted: 01/24/2025] [Indexed: 02/14/2025] Open
Abstract
Epigenetic regulators, known as "writers," erasers," and "readers," are essential for controlling gene expression by adding, removing, or recognizing post-translational modifications to histone tails, respectively. These regulators significantly affect genes involved in cancer initiation and maintenance. Recently, several clinical strategies targeting these epigenetic enzymes have emerged and some trials have demonstrated promising results for cancer treatment. Histone lysine demethylases (KDMs) yield distinct transcriptional outcomes that depend on the position of the methylated lysine and the specific genotype or lineage of the cancer cells. Due to their diverse roles in transcription, KDMs offer valuable opportunities for precision oncology, allowing treatments to be tailored to meet individual patient needs. This review emphasizes our current understanding of the functional relationship between KDMs and cancer as well as the development and application of small-molecule compounds that target KDMs.
Collapse
Affiliation(s)
- June-Ha Shin
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Hye-Been Yoo
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Jae-Seok Roe
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea.
| |
Collapse
|
831
|
Mou LL, Wu XM, Bibi A, Wang JX, Zhou CH. A comprehensive insight into naphthalimides as novel structural skeleton of multitargeting promising antibiotics. Future Med Chem 2025; 17:575-590. [PMID: 39957205 PMCID: PMC11901364 DOI: 10.1080/17568919.2025.2463872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Accepted: 01/27/2025] [Indexed: 02/18/2025] Open
Abstract
The globally growing antimicrobial resistance seriously threatens human health, increasing efforts have been devoting to the development of novel antibiotics. Naphthalimides contain a special skeleton of cyclic double imides and the naphthalene framework, this unique structure can exert multitargeting abilities which are helpful to overcome the escalating issue of resistance. Therefore, research in connection with the development of naphthalimides as novel antimicrobial agents is becoming progressively active. It has been revealed that naphthalimides as novel structural skeleton of multitargeting promising antibiotics could not only target DNAs and enzymes, disturb membrane, produce reactive oxygen species, etc. suggesting the multitargeting actions which do not induce resistance, but also show a broad antimicrobial spectrum with safety profile and pharmacokinetic characteristics, implying large potential as a new type of antibiotics via continuous efforts toward antimicrobial naphthalimides. This review presents naphthalimides as a new type of potential antimicrobial agents and discusses rational design strategies, structure-activity relationships, and mechanisms of action, with a comprehensive view to providing a new insight for in the rational design of efficient, broad-spectrum, and low-toxic naphthalimide antibiotics.
Collapse
Affiliation(s)
- Lin-Li Mou
- School of Chemistry and Chemical Engineering, Southwest University, Chongqing, China
| | - Xin-Miao Wu
- School of Chemistry and Chemical Engineering, Southwest University, Chongqing, China
| | - Aisha Bibi
- School of Chemistry and Chemical Engineering, Southwest University, Chongqing, China
| | - Jin-Xin Wang
- School of Chemistry and Chemical Engineering, Southwest University, Chongqing, China
- Chongqing Research Institute, Chongqing Werlchem New Materials Technology Co., Ltd., Chongqing, China
| | - Cheng-He Zhou
- School of Chemistry and Chemical Engineering, Southwest University, Chongqing, China
| |
Collapse
|
832
|
Wang X, Zhang Y, Liu H, Wang J, Zhang B, He T, Zhang H, Xiong Z, Liu X, Li J, Zhao W, Liu X, Zhang W, Yang L, Li Q, Zhang H, Qi J, Jia Q. A Novel Compound QO-83 Alleviates Acute and Chronic Epileptic Seizures in Rodents by Modulating K V7 Channel Activity. CNS Neurosci Ther 2025; 31:e70334. [PMID: 40125810 PMCID: PMC11931445 DOI: 10.1111/cns.70334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 12/04/2024] [Accepted: 03/01/2025] [Indexed: 03/25/2025] Open
Abstract
AIMS KV7 channels are promising targets for antiepileptic therapy. However, the classic KV7 channel opener retigabine has been withdrawn due to severe adverse reactions. We developed a novel KV7 channel opener, QO-83, with good chemical stability and blood-brain barrier penetration, and sought to evaluate its KV7-opening activity, antiepileptic effects, and mechanisms of action. METHODS We used patch-clamp electrophysiology, electroencephalogram recordings, dynamic simulations, and various epilepsy models to investigate the mechanisms and antiepileptic activity of QO-83. RESULTS Compound QO-83 exhibits greater potency at KV7.2/7.3 channels compared to KV7.4 or KV7.5 channels. It shows superior efficacy for KV7.2 with voltage-dependent opening than retigabine, with W236 identified as the key binding site for the KV7.2 channel. QO-83 significantly inhibited epileptiform discharge and influenced hippocampal sEPSC and sIPSC amplitudes. QO-83 has a more effective dose of 1 mg/kg in acute and chronic epilepsy models smaller than that of retigabine (10 mg/kg). The higher potency of QO-83 may be attributed to its greater stability at the KV7.2 binding pocket compared to retigabine. CONCLUSION QO-83, as a newly developed Kv7.2 opener, has the advantages of stable properties, strong affinity, and high activity compared with retigabine, and is expected to become a new antiepileptic drug.
Collapse
Affiliation(s)
- Xiangyu Wang
- Hebei Medical University, Postdoctoral Mobile Station of Basic MedicalHebei Medical UniversityShijiazhuangChina
- College of PharmacyHebei Medical UniversityShijiazhuangChina
| | - Yang Zhang
- College of PharmacyHebei Medical UniversityShijiazhuangChina
| | - Hui Liu
- Department of NeurobiologyHebei Medical UniversityShijiazhuangChina
| | - Jiahao Wang
- Department of PharmacologyHebei Medical UniversityShijiazhuangChina
| | - Boxuan Zhang
- Department of PharmacologyHebei Medical UniversityShijiazhuangChina
| | - Tenghui He
- Department of PharmacologyHebei Medical UniversityShijiazhuangChina
| | - Huiran Zhang
- College of PharmacyHebei Medical UniversityShijiazhuangChina
| | - Zhumei Xiong
- Department of PharmacologyHebei Medical UniversityShijiazhuangChina
| | - Xingang Liu
- College of PharmacyHebei Medical UniversityShijiazhuangChina
| | - Jincan Li
- College of PharmacyHebei Medical UniversityShijiazhuangChina
| | - Weidong Zhao
- Department of PharmacologyHebei Medical UniversityShijiazhuangChina
| | - Xiao Liu
- Department of PharmacologyHebei Medical UniversityShijiazhuangChina
| | - Wei Zhang
- Department of PharmacologyHebei Medical UniversityShijiazhuangChina
| | - Le Yang
- Department of PharmacologyHebei Medical UniversityShijiazhuangChina
| | - Qian Li
- Department of PharmacyShijiazhuang Fifth HospitalShijiazhuangChina
| | - Hailin Zhang
- Hebei Medical University, Postdoctoral Mobile Station of Basic MedicalHebei Medical UniversityShijiazhuangChina
- Department of PharmacologyHebei Medical UniversityShijiazhuangChina
- The Key Laboratory of Neural and Vascular BiologyMinistry of Education, Hebei Medical UniversityShijiazhuangChina
- Collaborative Innovation Center of Hebei Province for Mechanism, Diagnosis and Treatment of Neuropsychiatric DiseasesHebei Medical UniversityShijiazhuangChina
| | - Jinlong Qi
- Department of PharmacologyHebei Medical UniversityShijiazhuangChina
- The Key Laboratory of New Drug Pharmacology and ToxicologyHebei Medical UniversityShijiazhuangChina
| | - Qingzhong Jia
- Department of PharmacologyHebei Medical UniversityShijiazhuangChina
- The Key Laboratory of New Drug Pharmacology and ToxicologyHebei Medical UniversityShijiazhuangChina
- National Key Laboratory of New Pharmaceutical Preparations and ExcipientsHebei Medical UniversityShijiazhuangChina
| |
Collapse
|
833
|
Kızılyıldırım S, Sucu B, Muhammed MT, Akkoç S, Esatbeyoglu T, Ozogul F. Experimental and theoretical studies on antituberculosis activity of different benzimidazole derivatives. Heliyon 2025; 11:e42674. [PMID: 40051852 PMCID: PMC11883371 DOI: 10.1016/j.heliyon.2025.e42674] [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: 09/08/2024] [Revised: 02/11/2025] [Accepted: 02/12/2025] [Indexed: 03/09/2025] Open
Abstract
Tuberculosis (TB) continues to be one of the deadliest infectious diseases with a rapid increase in multidrug-resistant cases. The discovery of new agents against tuberculosis is urgently needed. Thus, the research article focuses on the antituberculosis activity of a series of benzimidazolium compounds. The antituberculosis activities of compounds including benzimidazole core (7a-h) against Mycobacterium tuberculosis H37Rv strain were tested in vitro using the BACTEC MGIT 960 system. The concentrations of benzimidazole compounds were adjusted to range from 0.25 to 4 μg/ml. The antituberculosis interactions of the compounds were investigated by molecular docking and molecular dynamics simulation. The results revealed that only benzimidazolium salt 7h showed antituberculosis activity at MIC value of 2 μg/ml although the other compounds showed no antituberculosis activity. The docking data revealed that 7h could bind to InhA thus indicating its inhibition potential on the enzyme. Molecular dynamics simulation exhibited that 7h formed a stable complex with the enzyme and was able to remain inside the binding region of the enzyme. Besides, the pharmacokinetic and drug-likeness properties of the compounds were assessed through computational approaches. The compounds exhibited drug-like properties. Consequently, 7h could be a good candidate for the development of new TB drugs.
Collapse
Affiliation(s)
- Suna Kızılyıldırım
- Cukurova University, Faculty of Pharmacy, Department of Pharmaceutical Microbiology, Adana, Türkiye
| | - Berfin Sucu
- Cukurova University, Institute of Science and Technology, Department of Biotechnology, Adana, Türkiye
| | - Muhammed Tilahun Muhammed
- Süleyman Demirel University, Faculty of Pharmacy, Department of Pharmaceutical Chemistry, 32260, Isparta, Türkiye
| | - Senem Akkoç
- Süleyman Demirel University, Faculty of Pharmacy, Department of Basic Pharmaceutical Sciences, 32260, Isparta, Türkiye
- Bahçeşehir University, Faculty of Engineering and Natural Sciences, Istanbul, 34353, Türkiye
| | - Tuba Esatbeyoglu
- Gottfried Wilhelm Leibniz University Hannover, Institute of Food and One Health, Department of Molecular Food Chemistry and Food Development, Am Kleinen Felde 30, 30167, Hannover, Germany
| | - Fatih Ozogul
- Cukurova University, Faculty of Fisheries, Department of Seafood Processing Technology, Adana, Türkiye
- Cukurova University, Biotechnology Research and Application Center, Adana, Türkiye
| |
Collapse
|
834
|
Zhang H, Zhang Q, Zuo T, Wang Z, Liao J, Lu Y. 2-Chloromethyl anthraquinone inhibits Candida albicans biofilm formation by inhibiting the Ras1-cAMP-Efg1 pathway. Res Microbiol 2025:104280. [PMID: 40024356 DOI: 10.1016/j.resmic.2025.104280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 02/17/2025] [Accepted: 02/27/2025] [Indexed: 03/04/2025]
Abstract
Candida albicans is an opportunistic pathogen, and the formation of its biofilm makes it resistant to traditional antifungal therapy. Anthraquinones have universal antibacterial activity. We evaluated the inhibitory effects of 2-chloromethyl anthraquinone on C. albicans adhesion, mycelial morphology transformation, and biofilm formation. The results showed that 2-chloromethyl anthraquinone could inhibit C. albicans adhesion, mycelium formation, and biofilm formation in a dose-dependent manner at 2 μg/mL. In addition, 2-chloromethyl anthraquinone significantly inhibited the expression of biofilm formation-related genes in C. albicans, including ALS1, CPH1, ECE1, HWP1, TEC1, BCR1, and UME6. In addition, Ras1-cAMP-Efg1 pathway-related genes (RAC1, CYR1, and TPK2) were also significantly down-regulated, indicating that the inhibitory effect of 2-chloromethyl anthraquinone on C. albicans biofilms may be related to the Ras1-cAMP-Efg1 signaling pathway. In summary, the results of this study confirmed the inhibitory mechanism of 2-chloromethyl anthraquinone on the virulence factors of C. albicans, which laid a theoretical foundation for its use as an anti-biofilm agent against C. albicans.
Collapse
Affiliation(s)
- Haoying Zhang
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, 210009, China
| | - Qi Zhang
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, 210009, China
| | - Ting Zuo
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, 210009, China
| | - Ziqi Wang
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, 210009, China
| | - Jianmin Liao
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, 210009, China.
| | - Yuanyuan Lu
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, 210009, China.
| |
Collapse
|
835
|
Ataollahi E, Emami L, Al-Dies AAM, Zare F, Poustforoosh A, Emami M, Saadat F, Motamen F, Rezaei Z, Khabnadideh S. Design, synthesis, in silico studies and antiproliferative evaluation of some novel hybrids of pyrimidine-morpholine. Front Chem 2025; 13:1537261. [PMID: 40093994 PMCID: PMC11906455 DOI: 10.3389/fchem.2025.1537261] [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: 11/30/2024] [Accepted: 02/06/2025] [Indexed: 03/19/2025] Open
Abstract
Introduction Cancer is a complex group of diseases characterized by the uncontrolled growth and spread of abnormal cells in the body. These cells can invade nearby tissues and organs, or they may metastasize to other parts of the body through the bloodstream or lymphatic system. Methods In this study, eight novel pyrimidine-morpholine hybrides (2a-2h) were designed and synthesized based on molecular hybridization approach to identify potent cytotoxic agents. Spectroscopic methods, including infrared spectroscopy (IR), proton and carbon nuclear magnetic resonance (1HNMR & 13CNMR), and mass spectrometry, were employed to confirm the structures of the compounds. The cytotoxic effects of the derivatives were evaluated against cancerous cell lines, including MCF-7 and SW480, using the MTT assay. Results and discussion It was demonstrated that all derivatives had appropriate cytotoxic potential with IC50 in range of 5.12-117.04 μM. Compound 2g was identified as the most potent compound, exhibiting IC50 values of 5.10 ± 2.12 μM and 19.60 ± 1.13 μM toward the SW480 and MCF-7 cell lines, respectively. Cell cycle analysis showed that 2g could induces phase arrest in MCF-7 breast cancer cells. The apoptosis assay demonstrated the induction of apoptosis in the SW480 cell line. The biological activity of the compounds was confirmed by the docking studies. DFT analysis for compounds 2g and 2h was conducted at the B3LYP/6-31+G** level of theory. It was concluded that 2g is both thermodynamically and kinetically more stable than 2h. Moreover, the interpretation of ADME (Absorption, Distribution, Metabolism, and Excretion) indicates that these new series of compounds possess acceptable prognostic physicochemical properties. These synthesized compounds may serve as promising candidates for further investigation as anticancer agents.
Collapse
Affiliation(s)
- Elaheh Ataollahi
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Leila Emami
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Al-Anood Mohammad Al-Dies
- Department of Chemistry, Al Qunfudah University College, UMM Al-Qura University, Mecca, Saudi Arabia
| | - Fateme Zare
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Alireza Poustforoosh
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mina Emami
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Fateme Saadat
- Student Research Committee, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Fateme Motamen
- Student Research Committee, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Zahra Rezaei
- Department of Medicinal Chemistry, Faculty of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Soghra Khabnadideh
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Medicinal Chemistry, Faculty of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
836
|
Piškor M, Milić A, Koštrun S, Majerić Elenkov M, Grbčić P, Kraljević Pavelić S, Pavelić K, Raić-Malić S. Synthesis, Antiproliferative Activity, and ADME Profiling of Novel Racemic and Optically Pure Aryl-Substituted Purines and Purine Bioisosteres. Biomolecules 2025; 15:351. [PMID: 40149888 PMCID: PMC11940194 DOI: 10.3390/biom15030351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 02/25/2025] [Accepted: 02/26/2025] [Indexed: 03/29/2025] Open
Abstract
The aim of this study was to synthesize new racemic and optically pure aryl-substituted purine bioisosteres using ultrasound-assisted Cu(I)-catalyzed Huisgen 1,3-dipolar cycloaddition. Regioselective synthesis of α-azido alcohols was applied to afford heterocycles with a 2-hydroxyeth-1-yl linker. Catalytic asymmetric synthesis using halohydrin dehalogenase in the ring-opening of epoxides gave enantioenriched azido alcohols, which subsequently afforded R- and S-enantiomers of purine and pyrrolo[2,3-d]pyrimidines with a 1-hydroxyeth-2-yl linker. The newly synthesized compounds were evaluated in vitro for their antiproliferative activity against four malignant tumor cell lines. The influence of regioisomerism and the stereochemistry of the hydroxyethyl group, as well as a N-heterocyclic scaffold linked to the aryl moiety on cytostatic activity was evaluated. Of all the compounds tested, purine 40a and pyrrolo[2,3-d]pyrimidine 45a derivatives with p-trifluoromethyl-substituted aryl connected to 1,2,3-triazole via a 2-hydroxyeth-1-yl spacer showed promising submicromolar antiproliferative activity. In addition, compound 45a exhibited selectivity towards the tumor cell line, with a selectivity index (SI) of 40, moderate clearance, and good membrane permeability.
Collapse
Affiliation(s)
- Martina Piškor
- Department of Organic Chemistry, University of Zagreb, Faculty of Chemical Engineering and Technology, Marulićev trg 19, 10000 Zagreb, Croatia;
| | | | | | - Maja Majerić Elenkov
- Division of Organic Chemistry and Biochemistry, Ruder Bošković Institute, Bijenička 54, 10000 Zagreb, Croatia;
| | - Petra Grbčić
- Juraj Dobrila University of Pula, Faculty of Medicine, 52100 Pula, Croatia; (P.G.); (K.P.)
| | | | - Krešimir Pavelić
- Juraj Dobrila University of Pula, Faculty of Medicine, 52100 Pula, Croatia; (P.G.); (K.P.)
| | - Silvana Raić-Malić
- Department of Organic Chemistry, University of Zagreb, Faculty of Chemical Engineering and Technology, Marulićev trg 19, 10000 Zagreb, Croatia;
| |
Collapse
|
837
|
Di Stefano M, Piazza L, Poles C, Galati S, Granchi C, Giordano A, Campisi L, Macchia M, Poli G, Tuccinardi T. KinasePred: A Computational Tool for Small-Molecule Kinase Target Prediction. Int J Mol Sci 2025; 26:2157. [PMID: 40076779 PMCID: PMC11900317 DOI: 10.3390/ijms26052157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 02/25/2025] [Accepted: 02/26/2025] [Indexed: 03/14/2025] Open
Abstract
Protein kinases are key regulators of cellular processes and critical therapeutic targets in diseases like cancer, making them a focal point for drug discovery efforts. In this context, we developed KinasePred, a robust computational workflow that combines machine learning and explainable artificial intelligence to predict the kinase activity of small molecules while providing detailed insights into the structural features driving ligand-target interactions. Our kinase-family predictive tool demonstrated significant performance, validated through virtual screening, where it successfully identified six kinase inhibitors. Target-focused operational models were subsequently developed to refine target-specific predictions, enabling the identification of molecular determinants of kinase selectivity. This integrated framework not only accelerates the screening and identification of kinase-targeting compounds but also supports broader applications in target identification, polypharmacology studies, and off-target effect analysis, providing a versatile tool for streamlining the drug discovery process.
Collapse
Affiliation(s)
- Miriana Di Stefano
- Department of Pharmacy, University of Pisa, 56124 Pisa, Italy; (M.D.S.); (L.P.); (C.G.); (M.M.); (T.T.)
| | - Lisa Piazza
- Department of Pharmacy, University of Pisa, 56124 Pisa, Italy; (M.D.S.); (L.P.); (C.G.); (M.M.); (T.T.)
| | - Clarissa Poles
- Telethon Institute of Genetics and Medicine, 80078 Naples, Italy;
- Genomics and Experimental Medicine Program, Scuola Superiore Meridionale (SSM, School of Advanced Studies), 80078 Naples, Italy
| | - Salvatore Galati
- Department of Pharmacy, University of Pisa, 56124 Pisa, Italy; (M.D.S.); (L.P.); (C.G.); (M.M.); (T.T.)
| | - Carlotta Granchi
- Department of Pharmacy, University of Pisa, 56124 Pisa, Italy; (M.D.S.); (L.P.); (C.G.); (M.M.); (T.T.)
| | - Antonio Giordano
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA 19122, USA;
| | - Luca Campisi
- Flashtox srl, Via Tosco Romagnola 136, 56025 Pontedera, Italy;
| | - Marco Macchia
- Department of Pharmacy, University of Pisa, 56124 Pisa, Italy; (M.D.S.); (L.P.); (C.G.); (M.M.); (T.T.)
| | - Giulio Poli
- Department of Pharmacy, University of Pisa, 56124 Pisa, Italy; (M.D.S.); (L.P.); (C.G.); (M.M.); (T.T.)
| | - Tiziano Tuccinardi
- Department of Pharmacy, University of Pisa, 56124 Pisa, Italy; (M.D.S.); (L.P.); (C.G.); (M.M.); (T.T.)
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA 19122, USA;
| |
Collapse
|
838
|
Ou H, Wu Z, Ning J, Huang Q, Wang W, Yang G, Zhou Y, Hou A, Li P, Chen L, Jin WB. In vitro and in vivo characterization of oridonin analogs as anti-inflammatory agents that regulate the NF-κB and NLRP3 inflammasome axis. Front Pharmacol 2025; 16:1512740. [PMID: 40083382 PMCID: PMC11903421 DOI: 10.3389/fphar.2025.1512740] [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: 10/17/2024] [Accepted: 01/28/2025] [Indexed: 03/16/2025] Open
Abstract
Introduction A series of oridonin hybrids were synthesized and evaluated for anti-inflammatory potential, focusing on their ability to inhibit NO production in RAW264.7 cells and their therapeutic prospects for NLRP3-driven disorders. Methods Anti-inflammatory activity was assessed by measuring NO inhibition in LPS-stimulated RAW264.7 cells. The most active compound, 4c, was further analyzed using ELISA and WB to evaluate its effects on inflammatory proteins (p-NF-κB, p-IκB, NLRP3, IL-6, IL-1β, COX-2, iNOS). In vivo efficacy was tested in a murine acute lung injury model, with RT‒qPCR and WB used to assess inflammatory markers in lung tissues. Molecular docking predicted 4c's binding mode with NLRP3, while RNA-seq and RT‒qPCR identified differentially expressed genes. Results Compound 4c significantly inhibited NO production and suppressed key inflammatory proteins in vitro. In vivo, it alleviated acute lung injury, reduced IL-6 and TNF-α mRNA levels, and inhibited NLRP3, p-NF-κB, and IL-6 protein expression. Docking suggested covalent binding to NLRP3. RNA-seq revealed 4c upregulated Trdc, Stfa2, and Gsta2 while downregulating Spib, Csf2, and Nr4a1. Discussion Compound 4c demonstrates potent anti-inflammatory effects via NLRP3 pathway inhibition and modulation of inflammatory genes. These findings highlight oridonin hybrids, particularly 4c, as promising candidates for NLRP3-driven inflammatory disorders, warranting further investigation.
Collapse
Affiliation(s)
- Huiping Ou
- Faculty of Chinese Materia Medica, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
- School of Food and Drug, Shenzhen Polytechnic University, Shenzhen, Guangdong, China
| | - Zhanpan Wu
- Faculty of Chinese Materia Medica, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Jinhua Ning
- Faculty of Chinese Materia Medica, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Qiufeng Huang
- School of Food and Drug, Shenzhen Polytechnic University, Shenzhen, Guangdong, China
| | - Wancun Wang
- Faculty of Chinese Materia Medica, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Guochun Yang
- Faculty of Chinese Materia Medica, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Yingxun Zhou
- Faculty of Chinese Materia Medica, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Anguo Hou
- Faculty of Chinese Materia Medica, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Peng Li
- School of Food and Drug, Shenzhen Polytechnic University, Shenzhen, Guangdong, China
| | - Lingyun Chen
- Faculty of Chinese Materia Medica, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Wen Bin Jin
- Faculty of Chinese Materia Medica, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| |
Collapse
|
839
|
Li F, Li M, Nie L, Zuo J, Fan W, Lian L, Hu J, Chen S, Jiang W, Han X, Wang H. Molecular Epidemiology and Antibiotic Resistance Associated with Avian Pathogenic Escherichia coli in Shanxi Province, China, from 2021 to 2023. Microorganisms 2025; 13:541. [PMID: 40142434 PMCID: PMC11946381 DOI: 10.3390/microorganisms13030541] [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: 01/25/2025] [Revised: 02/20/2025] [Accepted: 02/25/2025] [Indexed: 03/28/2025] Open
Abstract
Avian Pathogenic Escherichia coli (APEC) constitutes a major etiological agent of avian colibacillosis, which significantly hinders the development of the poultry industry. Conducting molecular epidemiological studies of APEC plays a crucial role in its prevention and control. This study aims to elucidate the molecular epidemiological characteristics of Avian Pathogenic Escherichia coli in Shanxi Province. In this study, 135 APEC strains were isolated and identified from 150 liver samples of diseased and deceased chickens exhibiting clinical symptoms, which were collected from farms in Shanxi Province between 2021 and 2023. The isolates were then analyzed for phylogenetic clustering, drug resistance, resistance genes, virulence genes, and biofilm formation capabilities. The results revealed that the proportions of the A, B1, B2, and D evolutionary subgroups were 26.67%, 32.59%, 17.78%, and 15.56%, respectively. The drug resistance testing results indicated that 92% of the isolates exhibited resistance to cotrimoxazole, kanamycin, chloramphenicol, amoxicillin, tetracycline, and other antibiotics. In contrast, 95% of the strains were sensitive to ofloxacin, amikacin, and ceftazidime. The most prevalent resistance genes included tetracycline-related (tetA) at 88.15%, followed by beta-lactam-related (bla-TEM) at 85.19%, and peptide-related (mcr1) at 12.59%. The virulence gene analysis revealed that ibeB, ompA, iucD, and mat were present in more than 90% of the isolates. The results revealed that 110 strains were biofilm-positive, corresponding to a detection rate of 81.48%. No significant correlation was found between the drug resistance genes, virulence genes, and the drug resistance phenotype. A moderate negative correlation was observed between the adhesion-related gene tsh and biofilm formation ability (r = -0.38). This study provides valuable insights into the prevention and control of avian colibacillosis in Shanxi Province.
Collapse
Affiliation(s)
- Fangfang Li
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong 030801, China; (F.L.); (M.L.); (L.N.); (W.F.); (S.C.)
- Shanghai Veterinary Research Institute, The Chinese Academy of Agricultural Sciences (CAAS), 518 Ziyue Road, Shanghai 200241, China; (J.Z.); (L.L.); (J.H.); (W.J.)
| | - Mengya Li
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong 030801, China; (F.L.); (M.L.); (L.N.); (W.F.); (S.C.)
- Shanghai Veterinary Research Institute, The Chinese Academy of Agricultural Sciences (CAAS), 518 Ziyue Road, Shanghai 200241, China; (J.Z.); (L.L.); (J.H.); (W.J.)
| | - Lianhua Nie
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong 030801, China; (F.L.); (M.L.); (L.N.); (W.F.); (S.C.)
- Shanghai Veterinary Research Institute, The Chinese Academy of Agricultural Sciences (CAAS), 518 Ziyue Road, Shanghai 200241, China; (J.Z.); (L.L.); (J.H.); (W.J.)
| | - Jiakun Zuo
- Shanghai Veterinary Research Institute, The Chinese Academy of Agricultural Sciences (CAAS), 518 Ziyue Road, Shanghai 200241, China; (J.Z.); (L.L.); (J.H.); (W.J.)
| | - Wenyan Fan
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong 030801, China; (F.L.); (M.L.); (L.N.); (W.F.); (S.C.)
- Shanghai Veterinary Research Institute, The Chinese Academy of Agricultural Sciences (CAAS), 518 Ziyue Road, Shanghai 200241, China; (J.Z.); (L.L.); (J.H.); (W.J.)
| | - Liyan Lian
- Shanghai Veterinary Research Institute, The Chinese Academy of Agricultural Sciences (CAAS), 518 Ziyue Road, Shanghai 200241, China; (J.Z.); (L.L.); (J.H.); (W.J.)
| | - Jiangang Hu
- Shanghai Veterinary Research Institute, The Chinese Academy of Agricultural Sciences (CAAS), 518 Ziyue Road, Shanghai 200241, China; (J.Z.); (L.L.); (J.H.); (W.J.)
| | - Shuming Chen
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong 030801, China; (F.L.); (M.L.); (L.N.); (W.F.); (S.C.)
| | - Wei Jiang
- Shanghai Veterinary Research Institute, The Chinese Academy of Agricultural Sciences (CAAS), 518 Ziyue Road, Shanghai 200241, China; (J.Z.); (L.L.); (J.H.); (W.J.)
| | - Xiangan Han
- Shanghai Veterinary Research Institute, The Chinese Academy of Agricultural Sciences (CAAS), 518 Ziyue Road, Shanghai 200241, China; (J.Z.); (L.L.); (J.H.); (W.J.)
| | - Haidong Wang
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong 030801, China; (F.L.); (M.L.); (L.N.); (W.F.); (S.C.)
| |
Collapse
|
840
|
Tao H, Yang B, Farhangian A, Xu K, Li T, Zhang ZY, Li J. Covalent-Allosteric Inhibitors: Do We Get the Best of Both Worlds? J Med Chem 2025; 68:4040-4052. [PMID: 39937154 DOI: 10.1021/acs.jmedchem.4c02760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/13/2025]
Abstract
Covalent-allosteric inhibitors (CAIs) may achieve the best of both worlds: increased potency, long-lasting effects, and reduced drug resistance typical of covalent ligands, along with enhanced specificity and decreased toxicity inherent in allosteric modulators. Therefore, CAIs can be an effective strategy to transform many undruggable targets into druggable ones. However, CAIs are challenging to design. In this perspective, we analyze the discovery of known CAIs targeting three protein families: protein phosphatases, protein kinases, and GTPases. We also discuss how computational methods and tools can play a role in addressing the practical challenges of rational CAI design.
Collapse
Affiliation(s)
- Hui Tao
- Borch Department of Medicinal Chemistry and Molecular Pharmacology, Purdue Institute for Drug Discovery, Purdue University, West Lafayette, Indiana 47907, United States
| | - Bo Yang
- Borch Department of Medicinal Chemistry and Molecular Pharmacology, Purdue Institute for Drug Discovery, Purdue University, West Lafayette, Indiana 47907, United States
| | - Atena Farhangian
- Borch Department of Medicinal Chemistry and Molecular Pharmacology, Purdue Institute for Drug Discovery, Purdue University, West Lafayette, Indiana 47907, United States
| | - Ke Xu
- Borch Department of Medicinal Chemistry and Molecular Pharmacology, Purdue Institute for Drug Discovery, Purdue University, West Lafayette, Indiana 47907, United States
| | - Tongtong Li
- Borch Department of Medicinal Chemistry and Molecular Pharmacology, Purdue Institute for Drug Discovery, Purdue University, West Lafayette, Indiana 47907, United States
| | - Zhong-Yin Zhang
- Borch Department of Medicinal Chemistry and Molecular Pharmacology, Purdue Institute for Drug Discovery, Purdue University, West Lafayette, Indiana 47907, United States
| | - Jianing Li
- Borch Department of Medicinal Chemistry and Molecular Pharmacology, Purdue Institute for Drug Discovery, Purdue University, West Lafayette, Indiana 47907, United States
| |
Collapse
|
841
|
Bilgin N, Hintzen JCJ, Mecinović J. Chemical tools for probing histidine modifications. Chem Commun (Camb) 2025; 61:3805-3820. [PMID: 39936705 DOI: 10.1039/d4cc06586g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/13/2025]
Abstract
Histidine is a unique amino acid with critical roles in protein structure and function, ranging from metal ion binding to enzyme catalysis. Histidine residues in proteins also undergo diverse posttranslational modifications, including methylation, phosphorylation and hydroxylation, by various enzymes, some of them being only recently identified and characterised. In this review, we describe the development of chemical tools for understanding the role of histidine residues in chemical and biological systems. We spotlight the application of histidine analogues in probing biomedically important posttranslational modifications of histidine residues in proteins, and we highlight novel bioconjugation methods that enable chemoselective modifications of histidine residues in peptides and proteins.
Collapse
Affiliation(s)
- Nurgül Bilgin
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, Campusvej 55, 5230 Odense, Denmark.
| | - Jordi C J Hintzen
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jasmin Mecinović
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, Campusvej 55, 5230 Odense, Denmark.
| |
Collapse
|
842
|
Herth MM, Hvass L, Poulie CBM, Müller M, García-Vázquez R, Gustavsson T, Shalgunov V, Clausen AS, Jørgensen JT, Hansson E, Jensen H, Aneheim E, Lindegren S, Kjaer A, Battisti UM. An 211At-labeled Tetrazine for Pretargeted Therapy. J Med Chem 2025; 68:4410-4425. [PMID: 39963966 DOI: 10.1021/acs.jmedchem.4c02281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/20/2025]
Abstract
Pretargeted radioimmunoimaging has been shown to enhance tumor-to-background ratios by up to 125-fold at early time points, leading to more efficient and less toxic radionuclide therapies, particularly with shorter half-lives such as astatine-211 (211At). The tetrazine ligation is the most utilized bioorthogonal reaction in these strategies, making tetrazines ideal for 211At labeling and controlling the biodistribution. We developed a 211At-labeled pretargeting agent for alpha-radionuclide therapy, achieving a radiochemical yield of approximately 65% and purity over 99%. Our results showed higher tumor-to-blood ratios within the first 24 h compared to directly labeled monoclonal antibodies. This suggests that pretargeted therapy may deliver better tumor doses than conventional methods, although the deastatination observed will need to be addressed in future tetrazine developments.
Collapse
Affiliation(s)
- Matthias M Herth
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark
- Cluster for Molecular Imaging, Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3, 2100 Copenhagen Ø, Denmark
| | - Lars Hvass
- Department of Clinical Physiology, Nuclear Medicine, Cyclotron and Radiochemistry Unit, Rigshospitalet, Blegdamsvej 9, 2100 Copenhagen, Denmark
| | - Christian B M Poulie
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark
| | - Marius Müller
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark
| | - Rocio García-Vázquez
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark
| | - Tobias Gustavsson
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark
| | - Vladimir Shalgunov
- Cluster for Molecular Imaging, Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3, 2100 Copenhagen Ø, Denmark
| | - Anne S Clausen
- Cluster for Molecular Imaging, Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3, 2100 Copenhagen Ø, Denmark
- Department of Clinical Physiology, Nuclear Medicine, Cyclotron and Radiochemistry Unit, Rigshospitalet, Blegdamsvej 9, 2100 Copenhagen, Denmark
| | - Jesper T Jørgensen
- Department of Clinical Physiology, Nuclear Medicine, Cyclotron and Radiochemistry Unit, Rigshospitalet, Blegdamsvej 9, 2100 Copenhagen, Denmark
| | - Ellinor Hansson
- Departments of Radiation Physics, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gula Stråket 2b, 41345 Gothenburg, Sweden
| | - Holger Jensen
- Cluster for Molecular Imaging, Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3, 2100 Copenhagen Ø, Denmark
| | - Emma Aneheim
- Departments of Radiation Physics, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gula Stråket 2b, 41345 Gothenburg, Sweden
| | - Sture Lindegren
- Departments of Radiation Physics, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gula Stråket 2b, 41345 Gothenburg, Sweden
| | - Andreas Kjaer
- Department of Clinical Physiology, Nuclear Medicine, Cyclotron and Radiochemistry Unit, Rigshospitalet, Blegdamsvej 9, 2100 Copenhagen, Denmark
| | - Umberto M Battisti
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark
| |
Collapse
|
843
|
Dhanda G, Singh H, Gupta A, Abdul Mohid S, Biswas K, Mukherjee R, Mukherjee S, Bhunia A, Nair NN, Haldar J. Dual-Functional Antibiotic Adjuvant Displays Potency against Complicated Gram-Negative Bacterial Infections and Exhibits Immunomodulatory Properties. ACS CENTRAL SCIENCE 2025; 11:279-293. [PMID: 40028349 PMCID: PMC11868958 DOI: 10.1021/acscentsci.4c02060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Revised: 12/31/2024] [Accepted: 01/03/2025] [Indexed: 03/05/2025]
Abstract
The treatment of Gram-negative bacterial infections is challenged by antibiotic resistance and complicated forms of infection like persistence, multispecies biofilms, intracellular infection, as well as infection-associated hyperinflammation and sepsis. To overcome these challenges, a dual-functional antibiotic adjuvant has been developed as a novel strategy to target complicated forms of bacterial infection and exhibit immunomodulatory properties. The lead adjuvant, D-LBDiphe showed multimodal mechanisms of action like weak outer membrane permeabilization, weak membrane depolarization, and inhibition of efflux machinery, guided primarily by hydrogen bonding and electrostatic interactions, along with weak van der Waals forces. D-LBDiphe potentiated antibiotics up to ∼4100-fold, targeted phenotypic forms of antibiotic tolerance, and revitalized antibiotics against topical and systemic infections of P. aeruginosa in mice. The aromatic moiety in D-LBDiphe was instrumental for interaction with lipopolysaccharide (LPS) micelles, and this interaction was the driving factor in reducing pro-inflammatory cytokines by 61.8-79% in mice challenged with LPS. Such multifarious properties of a weak-membrane perturbing, nonactive and nontoxic adjuvant have been discussed for the first time, supported by detailed mechanistic understanding and elucidation of structure-guided properties. This work expands the scope of antibiotic adjuvants and validates them as a promising approach for treatment of complicated bacterial infections and inflammation.
Collapse
Affiliation(s)
- Geetika Dhanda
- Antimicrobial
Research Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur, Bengaluru 560064, Karnataka, India
| | - Himani Singh
- Antimicrobial
Research Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur, Bengaluru 560064, Karnataka, India
| | - Abhinav Gupta
- Department
of Chemistry, Indian Institute of Technology
Kanpur, Kanpur 208016, India
| | - Sk Abdul Mohid
- Department
of Chemical Sciences, Bose Institute, Kolkata 700091, India
| | - Karishma Biswas
- Department
of Chemical Sciences, Bose Institute, Kolkata 700091, India
| | - Riya Mukherjee
- Antimicrobial
Research Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur, Bengaluru 560064, Karnataka, India
| | - Smriti Mukherjee
- Antimicrobial
Research Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur, Bengaluru 560064, Karnataka, India
| | - Anirban Bhunia
- Department
of Chemical Sciences, Bose Institute, Kolkata 700091, India
| | - Nisanth N. Nair
- Department
of Chemistry, Indian Institute of Technology
Kanpur, Kanpur 208016, India
| | - Jayanta Haldar
- Antimicrobial
Research Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur, Bengaluru 560064, Karnataka, India
- School
of Advanced Materials, Jawaharlal Nehru
Centre for Advanced Scientific Research, Jakkur, Bengaluru 560064, Karnataka, India
| |
Collapse
|
844
|
Zhu C, Liu X. Behavioral and pathological characteristics of 5xFAD female mice in the early stage. Sci Rep 2025; 15:6924. [PMID: 40011556 PMCID: PMC11865263 DOI: 10.1038/s41598-025-90335-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Accepted: 02/12/2025] [Indexed: 02/28/2025] Open
Abstract
Alzheimer's disease (AD) is a central nervous system degenerative disease with insidious onset and gradual development caused by selective and progressive loss of neurons. The 5xFAD mouse is a relatively mature disease model of AD. However, the behavioral research on 5xFAD female mouse is more focused on the changes of late memory function, and the exploration of its early behavioral and pathological changes is still incomplete. This research aims to explore the changes in memory function, emotional function (including anxiety and depression), motor ability, amyloid plaques, glial cell response and neurogenesis in the hippocampus of female 5xFAD mice in the early stage, laying a foundation for a comprehensive exploration of the disease mechanism of AD. The results of this study found that early 4-month-old female 5xFAD mice mainly showed a decline in memory function without other dysfunction. Accompanied by a large amount of amyloid protein plaques deposited in the hippocampus, it induced the response of microglia and astrocytes, and neurogenesis decreased significantly with age, especially in early female 5xFAD mice, which resulted in a decrease in the number of new neurons. This may be an important reason for the decline in memory function of female 5xFAD mice in the early stage.
Collapse
Affiliation(s)
- Chenlu Zhu
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China.
| | - Xuejiao Liu
- Department of Hyperbaric Medicine, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
| |
Collapse
|
845
|
Gorachand B, Lakshmi PR, Ramachary DB. Direct organocatalytic chemoselective synthesis of pharmaceutically active benzothiazole/benzoxazole-triazoles. Org Biomol Chem 2025; 23:2142-2152. [PMID: 39849920 DOI: 10.1039/d4ob01527d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2025]
Abstract
Benzothiazole and benzoxazole heterocyclic ring-containing 1,4,5-trisubstituted-1,2,3-triazoles are well known for their wide range of applications in pharmaceutical and medicinal chemistry, but their high-yielding metal-free selective synthesis has always remained challenging as no comprehensive simple protocol has been outlined to date. Owing to their structural and medicinal importance, herein, we synthesized various benzothiazole and benzoxazole heterocyclic ring-containing 1,4,5-trisubstituted-1,2,3-triazoles in high to excellent yields with chemo-/regioselectivity from the library of benzothiazole/benzoxazole-ketones and aryl/alkyl-azides through an enolate-mediated organocatalytic azide-ketone [3 + 2]-cycloaddition under ambient conditions in a few hours. The commercial availability or quick synthesis of the starting materials and catalysts, a diverse substrate scope, chemo-/regioselectivity, quick synthesis of pharmaceutically active known compounds and their analogues, and numerous medicinal applications of functionalized benzothiazole/benzoxazole-triazoles are the key attractions of this metal-free organo-click reaction.
Collapse
Affiliation(s)
- Badaraita Gorachand
- Catalysis Laboratory, School of Chemistry, University of Hyderabad, Hyderabad-500 046, India.
| | - Pandhiti R Lakshmi
- Catalysis Laboratory, School of Chemistry, University of Hyderabad, Hyderabad-500 046, India.
| | | |
Collapse
|
846
|
Ou L, Setegne MT, Elliot J, Shen F, Dassama LMK. Protein-Based Degraders: From Chemical Biology Tools to Neo-Therapeutics. Chem Rev 2025; 125:2120-2183. [PMID: 39818743 PMCID: PMC11870016 DOI: 10.1021/acs.chemrev.4c00595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 12/26/2024] [Accepted: 12/30/2024] [Indexed: 01/19/2025]
Abstract
The nascent field of targeted protein degradation (TPD) could revolutionize biomedicine due to the ability of degrader molecules to selectively modulate disease-relevant proteins. A key limitation to the broad application of TPD is its dependence on small-molecule ligands to target proteins of interest. This leaves unstructured proteins or those lacking defined cavities for small-molecule binding out of the scope of many TPD technologies. The use of proteins, peptides, and nucleic acids (otherwise known as "biologics") as the protein-targeting moieties in degraders addresses this limitation. In the following sections, we provide a comprehensive and critical review of studies that have used proteins and peptides to mediate the degradation and hence the functional control of otherwise challenging disease-relevant protein targets. We describe existing platforms for protein/peptide-based ligand identification and the drug delivery systems that might be exploited for the delivery of biologic-based degraders. Throughout the Review, we underscore the successes, challenges, and opportunities of using protein-based degraders as chemical biology tools to spur discoveries, elucidate mechanisms, and act as a new therapeutic modality.
Collapse
Affiliation(s)
- Lisha Ou
- Department
of Chemistry, Stanford University, Stanford, California 94305, United States
- Sarafan
ChEM-H Institute, Stanford University, Stanford, California 94305, United States
| | - Mekedlawit T. Setegne
- Department
of Chemistry, Stanford University, Stanford, California 94305, United States
- Sarafan
ChEM-H Institute, Stanford University, Stanford, California 94305, United States
| | - Jeandele Elliot
- Department
of Chemical Engineering, Stanford University, Stanford, California 94305, United States
| | - Fangfang Shen
- Department
of Chemistry, Stanford University, Stanford, California 94305, United States
| | - Laura M. K. Dassama
- Department
of Chemistry, Stanford University, Stanford, California 94305, United States
- Sarafan
ChEM-H Institute, Stanford University, Stanford, California 94305, United States
- Department
of Microbiology & Immunology, Stanford
School of Medicine, Stanford, California 94305, United States
| |
Collapse
|
847
|
Yuan Y, Li P, Shen W, Li M, He X, Zhou B. Genomic identification of a pair of multidrug-resistant but non-pathogenic Salmonella enterica serovar Goldcoast isolates in southeast China. Front Microbiol 2025; 16:1540843. [PMID: 40078548 PMCID: PMC11897504 DOI: 10.3389/fmicb.2025.1540843] [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: 12/17/2024] [Accepted: 01/24/2025] [Indexed: 03/14/2025] Open
Abstract
Introduction Salmonella is an important foodborne pathogen that can induce severe diseases such as gastrointestinal disease and typhoid fever. Accumulating evidence revealed that Salmonella's resistance to antibiotics also seriously affects human health. Pathogenic Salmonella enterica serovar Goldcoast (S. Goldcoast) was first detected in 2010 in China and was predicted to have an increasing tendency. Methods The MacConkey agar, Salmonella Shigella agar, three-sugar iron agar slant, and Gram-stained microscopic examination were used for strain identification. Gram-negative bacteria identification cards explored more properties of the isolates, while antimicrobial susceptibility testing was used to examine the multidrug resistance. The 2nd and 3rd generation sequencing revealed the genetic information of the isolates. Results Two non-pathogenic isolates with multidrug resistance, JS33 and JS34, harbored 42 antibiotic-resistant genes (ARGs) in contig1 and 13 ARGs in contig2, were isolated from a healthy donor living in southeast China and identified as S. Goldcoast (6,8:r:l,w). Interestingly, JS33 and JS34 showed identical responses to more than 20 antimicrobial agents and were resistant to ampicillin, selectrin, chloramphenicol, tetracycline, and streptomycin. However, JS33 differed from JS34 in hydrogen sulfide (H2S) generation. The genomic sequencing identified a deletion in thiosulfate reductase (K08352) in JS34. Discussion H2S is an essential physiological regulator linked to inflammation and cancer. Therefore, genomic identification of JS33 and JS34 provided us with a better understanding of drug resistance and could be used as model strains to study the effects of microbial H2S production on the host. Since JS33 and JS34 did not induce gastrointestinal infection or other clinical symptoms as previously reported, the appearance of non-pathogenic S. Goldcoast in southeast China warned us to prepare for the prevalence of antimicrobial-resistant S. Goldcoast in China.
Collapse
Affiliation(s)
- Yongjuan Yuan
- Jiashan County Center for Disease Control and Prevention, Jiaxing, China
| | - Ping Li
- Jiaxing Center for Disease Control and Prevention, Jiaxing, China
| | - Wei Shen
- Jiashan County Center for Disease Control and Prevention, Jiaxing, China
| | - Min Li
- Jiashan County Center for Disease Control and Prevention, Jiaxing, China
| | - Xiaofei He
- Medical Research Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Bin Zhou
- Medical Research Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| |
Collapse
|
848
|
Becerra D, Castillo JC. Recent advances in the synthesis of anticancer pyrazole derivatives using microwave, ultrasound, and mechanochemical techniques. RSC Adv 2025; 15:7018-7038. [PMID: 40041378 PMCID: PMC11878059 DOI: 10.1039/d4ra08866b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Accepted: 02/21/2025] [Indexed: 03/06/2025] Open
Abstract
Pyrazole and its derivatives have attracted considerable attention in pharmaceutical and medicinal chemistry, as reflected in their presence in numerous FDA-approved drugs and clinical candidates. This review presents a comprehensive analysis of articles published between 2014 and 2024, focusing on the microwave-, ultrasound-, and mechanochemical-assisted synthesis of pyrazole derivatives with anticancer activity. It explores synthetic methodologies, anticancer efficacy, and molecular docking studies, underscoring the significance of pyrazole derivatives in drug discovery and medicinal chemistry. Notably, microwave irradiation stands out as the most widely employed technique, providing high efficiency by significantly reducing reaction times while maintaining moderate temperatures. Ultrasound irradiation serves as a valuable alternative, particularly for processes that require milder conditions, whereas mechanochemical activation, though less frequently employed, offers distinct advantages in terms of sustainability.
Collapse
Affiliation(s)
- Diana Becerra
- Escuela de Ciencias Químicas, Universidad Pedagógica y Tecnológica de Colombia Avenida Central del Norte 39-115 Tunja Colombia
| | - Juan-Carlos Castillo
- Escuela de Ciencias Químicas, Universidad Pedagógica y Tecnológica de Colombia Avenida Central del Norte 39-115 Tunja Colombia
| |
Collapse
|
849
|
Liu XH, Fu CH, Wang J, Wei YC, Tan CX, Weng JQ, Min LJ, Xu TM, Wu N. Novel 5-(Trifluoromethyl)-1,2,4-oxadiazole-Based Pyrimidin-4-ether Histone Deacetylase Inhibitors for Controlling Rust Disease: Design, Synthesis, Activity, and Structure-Activity Relationship. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:4563-4573. [PMID: 39939838 DOI: 10.1021/acs.jafc.4c09039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/14/2025]
Abstract
Rust disease, an important plant pathogen, can lead to reduced crop or fruit production. Trifluoromethyloxadiazole (TFMO) is a class of histone deacetylase inhibitors (HDACs). Herein, a series of 5-(trifluoromethyl)-1,2,4-oxadiazole (TFMO)-based pyrimidin-4-ether derivatives were designed and synthesized. Antirust bioassay results of TFMOs showed that some of them possessed excellent activities against plant rust pathogens, such as Puccinia sorghi, Phakopsora pachyrhizi, and Puccinia rubigo. The most active compound, 3-(5-(((6-(difluoromethyl)pyrimidin-4-yl)oxy)methyl)thiophen-2-yl)-5-(trifluoromethyl)-1,2,4-oxadiazole (XII6), exhibited 50% control against P. pachyrhizi at 0.780 mg/L, which was significantly better than the commercial fungicide azoxystrobin (0%) at the same concentration. The field trial results indicated that the compound exhibited an excellent control effect against P. rubigo at 116 g a.i./ha. The acute toxic results indicated that compound XII6 has low toxicity. Furthermore, the enzyme activity results showed that compound XII6 is a strong, nonselective HDAC inhibitor. Finally, the structure-activity relationship was established, and the compound XII6-HDAC binding mode was carried out based on the crystal structure of hHDAC1, hHDAC4, and hHDAC6. This work provided an excellent fungicide against rust for further optimization.
Collapse
Affiliation(s)
- Xing-Hai Liu
- College of Chemical Engineering, Zhejiang University of Technology, Hangzhou 310014, China
| | - Chen-Hao Fu
- College of Chemical Engineering, Zhejiang University of Technology, Hangzhou 310014, China
- Zhejiang Base of National Southern Pesticide Research Centre, Zhejiang Research Institute of Chemical Industry, Hangzhou 310023, China
| | - Jian Wang
- College of Chemical Engineering, Zhejiang University of Technology, Hangzhou 310014, China
- Zhejiang Base of National Southern Pesticide Research Centre, Zhejiang Research Institute of Chemical Industry, Hangzhou 310023, China
| | - You-Chang Wei
- Zhejiang Base of National Southern Pesticide Research Centre, Zhejiang Research Institute of Chemical Industry, Hangzhou 310023, China
| | - Cheng-Xia Tan
- College of Chemical Engineering, Zhejiang University of Technology, Hangzhou 310014, China
| | - Jian-Quan Weng
- College of Chemical Engineering, Zhejiang University of Technology, Hangzhou 310014, China
| | - Li-Jing Min
- College of Life Science, Key Laboratory of Vector Biology and Pathogen Control of Zhejiang Province, Huzhou University, Huzhou, Zhejiang 313000, China
| | - Tian-Ming Xu
- Zhejiang Base of National Southern Pesticide Research Centre, Zhejiang Research Institute of Chemical Industry, Hangzhou 310023, China
| | - Ningjie Wu
- Zhejiang Base of National Southern Pesticide Research Centre, Zhejiang Research Institute of Chemical Industry, Hangzhou 310023, China
| |
Collapse
|
850
|
Mulligan M, Boudreau MW, Bouwens BA, Lee Y, Carrell HW, Zhu J, Mousses S, Shapiro DJ, Nelson ER, Fan TM, Hergenrother PJ. Single Dose of a Small Molecule Leads to Complete Regressions of Large Breast Tumors in Mice. ACS CENTRAL SCIENCE 2025; 11:228-238. [PMID: 40028352 PMCID: PMC11869136 DOI: 10.1021/acscentsci.4c01628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 11/25/2024] [Accepted: 12/19/2024] [Indexed: 03/05/2025]
Abstract
Patients with estrogen receptor α positive (ERα+) breast cancer typically undergo surgical resection, followed by 5-10 years of treatment with adjuvant endocrine therapy. This prolonged intervention is associated with a host of undesired side effects that reduce patient compliance, and ultimately therapeutic resistance and disease relapse/progression are common. An ideal anticancer therapy would be effective against recurrent and refractory disease with minimal dosing; however, there is little precedent for marked tumor regression with a single dose of a small molecule therapeutic. Herein we report ErSO-TFPy as a small molecule that induces quantitative or near-quantitative regression of tumors in multiple mouse models of breast cancer with a single dose. Importantly, this effect is robust and independent of tumor size with eradication of even very large tumors (500-1500 mm3) observed. Mechanistically, these tumor regressions are a consequence of rapid induction of necrotic cell death in the tumor and are immune cell independent. If successfully translated to human cancer patients, the benefits of such an anticancer drug that is effective with a single dose would be significant.
Collapse
Affiliation(s)
- Michael
P. Mulligan
- Department
of Chemistry, University of Illinois at
Urbana-Champaign, Urbana, Illinois 61801, United States
- Carl
R. Woese Institute for Genomic Biology, University of Illinois at
Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Matthew W. Boudreau
- Department
of Chemistry, University of Illinois at
Urbana-Champaign, Urbana, Illinois 61801, United States
- Carl
R. Woese Institute for Genomic Biology, University of Illinois at
Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Brooke A. Bouwens
- Carl
R. Woese Institute for Genomic Biology, University of Illinois at
Urbana-Champaign, Urbana, Illinois 61801, United States
- Department
of Biochemistry, University of Illinois
at Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Yoongyeong Lee
- Department
of Comparative Biosciences, University of
Illinois at Urbana-Champaign, Urbana, Illinois 61802, United States
| | - Hunter W. Carrell
- Department
of Chemistry, University of Illinois at
Urbana-Champaign, Urbana, Illinois 61801, United States
- Carl
R. Woese Institute for Genomic Biology, University of Illinois at
Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Junyao Zhu
- Department
of Biochemistry, University of Illinois
at Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Spyro Mousses
- Systems
Oncology, Scottsdale, Arizona 85255, United States
| | - David J. Shapiro
- Department
of Biochemistry, University of Illinois
at Urbana-Champaign, Urbana, Illinois 61801, United States
- Cancer Center
at Illinois, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Erik R. Nelson
- Carl
R. Woese Institute for Genomic Biology, University of Illinois at
Urbana-Champaign, Urbana, Illinois 61801, United States
- Cancer Center
at Illinois, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
- Department
of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
- Division
of Nutritional Sciences, University of Illinois
at Urbana-Champaign, Urbana, Illinois 61801, United States
- Beckman Institute
for Advanced Science and Technology, University
of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Timothy M. Fan
- Carl
R. Woese Institute for Genomic Biology, University of Illinois at
Urbana-Champaign, Urbana, Illinois 61801, United States
- Cancer Center
at Illinois, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
- Department
of Veterinary Clinical Medicine, University
of Illinois at Urbana-Champaign, Urbana, Illinois 61802, United States
| | - Paul J. Hergenrother
- Department
of Chemistry, University of Illinois at
Urbana-Champaign, Urbana, Illinois 61801, United States
- Carl
R. Woese Institute for Genomic Biology, University of Illinois at
Urbana-Champaign, Urbana, Illinois 61801, United States
- Cancer Center
at Illinois, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
| |
Collapse
|