101
|
Subramaniyan Y, Khan A, Mujeeburahiman M, Ashmitha, Shivani M, Patil S, Lavanya N, Rekha PD. Urological metabolic conditions alter regulation of quorum-sensing system and virulence genes expression in uropathogenic Pseudomonasaeruginosa. Microb Pathog 2025; 203:107502. [PMID: 40127827 DOI: 10.1016/j.micpath.2025.107502] [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: 09/02/2024] [Revised: 02/24/2025] [Accepted: 03/21/2025] [Indexed: 03/26/2025]
Abstract
Pseudomonas aeruginosa (P. aeruginosa) is responsible for complicated urinary tract infections (UTI) acquired from different transmission routes. Quorum sensing (QS) plays an important role in the regulation of virulence factors in P. aeruginosa and is dependent on the nutritional conditions. Under pathological conditions, urine contains elevated levels of metabolites such as glucose, creatinine, albumin and haem that may alter the QS behavior in P. aeruginosa. Hence, we investigated the role of the QS system in P. aeruginosa on growth and the expression of virulence genes under altered urinary metabolic compositions. We used P. aeruginosa strains isolated from UTI patients, along with PAO1 as a reference strain and grown under simulated metabolic conditions in the synthetic urine. Growth, biofilm formation, motility, siderophore production, rhamnolipid secretion, pyocyanin production, elastase and urease activity were quantified using standard laboratory methods. The expression levels of QS genes, such as lasI/R, rhlI/R, and pqsA/R, alginate synthesis genes (algD/R), the motility-regulating gene (pilA), and urease gene (ureC), were estimated using qRT-PCR for comparison between the growth conditions. The results showed significant differences in growth, biofilm formation and virulence factors between the metabolic conditions and were significantly higher (p<0.001) in simulated glycosuria, haematuria, and creatininuria conditions compared to the control. Under albuminuria, significantly reduced growth and virulence factor production were observed. Compared to control, higher expression levels of lasI/R, rhlI/R, pqsA/R, algD/R, pilA and ureC genes were observed in all conditions except albuminuria. The results highlighted the overexpression of QS-regulating genes and virulence in strains from similar metabolic environments in patients, suggesting possible niche adaptation. Such adaptation may pose considerable challenge in the management of chronic P. aeruginosa infections in urinary tract.
Collapse
Affiliation(s)
- Yuvarajan Subramaniyan
- Division of Microbiology and Biotechnology, Yenepoya Research Centre, Yenepoya (Deemed to Be University), University Road, Deralakatte, Mangalore, 575018, India
| | - Altaf Khan
- Department of Urology, Yenepoya Medical College and Hospital, Yenepoya (Deemed to Be University), University Road, Deralakatte, Mangalore, 575018, India
| | - M Mujeeburahiman
- Department of Urology, Yenepoya Medical College and Hospital, Yenepoya (Deemed to Be University), University Road, Deralakatte, Mangalore, 575018, India
| | - Ashmitha
- Department of Biosciences, Mangalore University, Mangalagangotri, Mangalore, 574199, India
| | - M Shivani
- Division of Microbiology and Biotechnology, Yenepoya Research Centre, Yenepoya (Deemed to Be University), University Road, Deralakatte, Mangalore, 575018, India
| | - Smitha Patil
- Department of Biosciences, Mangalore University, Mangalagangotri, Mangalore, 574199, India
| | - N Lavanya
- Department of Biosciences, Mangalore University, Mangalagangotri, Mangalore, 574199, India
| | - Punchappady Devasya Rekha
- Division of Microbiology and Biotechnology, Yenepoya Research Centre, Yenepoya (Deemed to Be University), University Road, Deralakatte, Mangalore, 575018, India.
| |
Collapse
|
102
|
Manik MRK, Mishu ID, Mahmud Z, Muskan MN, Emon SZ. Association of fluoroquinolone resistance with rare quinolone resistance-determining region (QRDR) mutations and protein-quinolone binding affinity (PQBA) in multidrug-resistant Escherichia coli isolated from patients with urinary tract infection. J Infect Public Health 2025; 18:102766. [PMID: 40153979 DOI: 10.1016/j.jiph.2025.102766] [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: 12/11/2024] [Revised: 03/17/2025] [Accepted: 03/18/2025] [Indexed: 04/01/2025] Open
Abstract
BACKGROUND Urinary tract infections (UTIs) caused by Escherichia coli pose significant public health risks, particularly in developing countries like Bangladesh. This study aimed to elucidate resistance patterns among UTI isolates and comprehensively investigate the mutational spectrum and its impact on drug-microbe interactions. METHODS We collected and identified E. coli isolates from hospitalized UTI patients at Dhaka Medical College Hospital and determined their resistance patterns using the disc diffusion method and broth microdilution. Quinolone resistance-determining regions (QRDRs) of the target genes (gyrA, gyrB, parC, and parE) associated with fluoroquinolone resistance were amplified by polymerase chain reaction (PCR) and analyzed through BTSeq™ sequencing for mutations, followed by molecular docking analysis using PyMOL and AutoDock for the protein-quinolone binding affinity (PQBA) study. RESULTS All isolates (100 %) displayed multidrug resistance, with chloramphenicol (16 % resistant) and colistin (28 % resistant) demonstrating superior efficacy compared to other antibiotics. The isolates resistant to colistin, as determined by disc diffusion testing, exhibited remarkably high minimum inhibitory concentrations (MICs), with one isolate registering an MIC exceeding 512 µg/mL. Alarming resistance rates were observed for five antibiotic classes, except for polymyxins (28 % resistant) and protein synthesis inhibitors (48 % resistant). Fifty-two percent (52 %) of the isolates exhibited resistance to all five tested quinolones. Sequence analysis revealed a novel L88Q mutation in ParC, affecting PQBA and binding conformation. Additionally, three ParC mutations (S80I, E84V, and E84G) and two ParE mutations (S458A and I529L) were identified, which had not been previously reported in Bangladesh. Among these, S80I appeared in all isolates. Double-mutations (S83L+D87N) in GyrA, L88Q and S80I in ParC, and I529L in ParE were identified as key drivers of fluoroquinolone resistance. CONCLUSION Our findings underscore the accumulation of significant mutations within QRDRs of UTI isolates, potentially compromising fluoroquinolone efficacy. The emergence of these novel mutations warrants further investigation to impede their dissemination and combat quinolone resistance.
Collapse
Affiliation(s)
- Md Rasel Khan Manik
- Department of Biochemistry and Molecular Biology, University of Dhaka, Dhaka 1000, Bangladesh
| | | | - Zimam Mahmud
- Department of Biochemistry and Molecular Biology, University of Dhaka, Dhaka 1000, Bangladesh.
| | | | - Sharmin Zaman Emon
- Centre for Advanced Research in Sciences, University of Dhaka, Dhaka 1000, Bangladesh
| |
Collapse
|
103
|
Salazar IL, Curcio M, Mele M, Vetrone R, Frisari S, Costa RO, Caldeira MV, Trader DJ, Duarte CB. Activation of the 20S proteasome core particle prevents cell death induced by oxygen- and glucose deprivation in cultured cortical neurons. Apoptosis 2025; 30:1372-1390. [PMID: 40095265 DOI: 10.1007/s10495-025-02097-x] [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] [Accepted: 02/23/2025] [Indexed: 03/19/2025]
Abstract
Neuronal damage in brain ischemia is characterized by a disassembly of the proteasome and a decrease in its proteolytic activity. However, to what extent these alterations are coupled to neuronal death is controversial since proteasome inhibitors were shown to provide protection in different models of stroke in rodents. This question was addressed in the present work using cultured rat cerebrocortical neurons subjected to transient oxygen- and glucose-deprivation (OGD) as a model for in vitro ischemia. Under the latter conditions there was a time-dependent loss in the proteasome activity, determined by cleavage of the Suc-LLVY-AMC fluorogenic substrate, and the disassembly of the proteasome, as assessed by native-polyacrylamide gel electrophoresis followed by western blot against Psma2 and Rpt6, which are components of the catalytic core and regulatory particle, respectively. Immunocytochemistry experiments against the two proteins also showed differential effects on their dendritic distribution. OGD also downregulated the protein levels of Rpt3 and Rpt10, two components of the regulatory particle, by a mechanism dependent on the activity of NMDA receptors and mediated by calpains. Activation of the proteasome activity, using an inhibitor of USP14, a deubiquitinase enzyme, inhibited OGD-induced cell death, and decreased calpain activity as determined by analysis of spectrin cleavage. Similar results were obtained in the presence of two oleic amide derivatives (B12 and D3) which directly activate the 20S proteasome core particle. Together, these results show that proteasome activation prevents neuronal death in cortical neurons subjected to in vitro ischemia, indicating that inhibition of the proteasome is a mediator of neuronal death in brain ischemia.
Collapse
Affiliation(s)
- Ivan L Salazar
- Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
- CNC-Center for Neuroscience and Cell Biology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- Multidisciplinary Institute of Ageing-MIA Portugal, University of Coimbra, Coimbra, Portugal
| | - Michele Curcio
- CNC-Center for Neuroscience and Cell Biology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Miranda Mele
- Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
- CNC-Center for Neuroscience and Cell Biology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Rossela Vetrone
- Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
- CNC-Center for Neuroscience and Cell Biology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Simone Frisari
- Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
- CNC-Center for Neuroscience and Cell Biology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Rui O Costa
- Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
- CNC-Center for Neuroscience and Cell Biology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Margarida V Caldeira
- Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
- CNC-Center for Neuroscience and Cell Biology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Darci J Trader
- Department of Pharmaceutical Sciences, University of California Irvine, Irvine, USA
| | - Carlos B Duarte
- CNC-Center for Neuroscience and Cell Biology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal.
- Departament of Life Sciences, University of Coimbra, Coimbra, Portugal.
| |
Collapse
|
104
|
Zhang S, Guo R, Liu Y, Wu Z, Song Y. Basic and applied research progress of TRAIL in hematologic malignancies. BLOOD SCIENCE 2025; 7:e00221. [PMID: 40084090 PMCID: PMC11902984 DOI: 10.1097/bs9.0000000000000221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Accepted: 01/20/2025] [Indexed: 03/16/2025] Open
Abstract
Hematological malignancies encompass a diverse range of blood-related cancers characterized by abnormal blood cell production. These cancers, classified by the World Health Organization based on lineage, cell origin, and progression, provide a more comprehensive framework for understanding cancer biology. This classification has significantly advanced cancer research, particularly in genetic analyses for diagnosis and treatment. Despite recent clinical improvements, challenges, such as relapse, resistance, and high mortality, remain unresolved. Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL), a protein that induces apoptosis in cancer cells without affecting normal cells, has emerged as a promising therapeutic target. However, its clinical efficacy is limited by factors, such as tumor heterogeneity and resistance to TRAIL signaling. This review examines the mechanisms of TRAIL in hematological malignancies, factors contributing to resistance, and the current state of preclinical and clinical research, highlighting potential strategies to enhance TRAIL-based therapies in blood cancers.
Collapse
Affiliation(s)
- Sidong Zhang
- Department of Pediatric Hematology-Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Rongqun Guo
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yufeng Liu
- Department of Pediatric Hematology-Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhengyu Wu
- Department of Radiotherapy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yadong Song
- Department of Radiotherapy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
105
|
Sengupta P, Mukhopadhyay D. IGF1R/ARRB1 Mediated Regulation of ERK and cAMP Pathways in Response to Aβ Unfolds Novel Therapeutic Avenue in Alzheimer's Disease. Mol Neurobiol 2025; 62:8065-8083. [PMID: 39969678 PMCID: PMC12078455 DOI: 10.1007/s12035-025-04735-6] [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: 07/17/2024] [Accepted: 01/31/2025] [Indexed: 02/20/2025]
Abstract
IGF1R/INSR signaling is crucial for understanding Alzheimer's disease (AD) and may aid in the development of potent therapeutic strategies. This study investigated the expression and activity of these receptors and their potential to form functional hybrids in response to amyloid beta (Aβ). IGF1R, INSR, and ARRB1 were found to be upregulated in AD. The propensity for functional hybrid formation was also greater in the presence of Aβ. The association of IGF1R with ARRB1 reached a maximum at 60 min of Aβ treatment, which coincided with increased pERK activity at approximately the same time, indicating the importance of this association in pERK regulation. Knocking down IGF1R, INSR, and ARRB1 independently reduced cAMP, whereas overexpressing IGF1R significantly increased cAMP. Knocking down ARRB1 in IGF1R-overexpressing cells led to a reduction in cAMP, indicating that the interaction of ARRB1 and IGF1R possibly contributes to cAMP dysregulation. Since cAMP plays a crucial role in cognition and memory, alterations in cAMP after receptor hybridization could be significant in AD. Additionally, we noted hyperactivation of MAPK, which is associated with aberrant cellular activity, transcriptional control, and stress pathways. This finding highlights the importance of IGF1R and INSR dysregulation, which plays a major role in addition to conventional RTK signaling through multiple pathways. Here, we focused on the ARRB1 and IGF1R interaction and showed that picropodophyllin (PPP), an IGF1R-specific inhibitor, blocks this interaction and alters the ERK and cAMP status under disease conditions. Cell viability studies further revealed that the PPP substantially improved cell viability in the presence of Aβ. This highlights the role of the PPP in regulating these cascades and opens the arena for further therapeutic development for AD.
Collapse
Affiliation(s)
- Priyanka Sengupta
- Biophysical Sciences Group, 1/AF, Biddhanagar, Saha Institute of Nuclear Physics, Kolkata, 700064, India
- Homi Bhabha National Institute, Anushaktinagar, Mumbai, 400094, India
| | - Debashis Mukhopadhyay
- Biophysical Sciences Group, 1/AF, Biddhanagar, Saha Institute of Nuclear Physics, Kolkata, 700064, India.
- Homi Bhabha National Institute, Anushaktinagar, Mumbai, 400094, India.
| |
Collapse
|
106
|
Liu D, Liu L, Zhang X, Zhao X, Li X, Che X, Wu G. Decoding driver and phenotypic genes in cancer: Unveiling the essence behind the phenomenon. Mol Aspects Med 2025; 103:101358. [PMID: 40037122 DOI: 10.1016/j.mam.2025.101358] [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: 10/27/2024] [Revised: 01/25/2025] [Accepted: 02/26/2025] [Indexed: 03/06/2025]
Abstract
Gray hair, widely regarded as a hallmark of aging. While gray hair is associated with aging, reversing this trait through gene targeting does not alter the fundamental biological processes of aging. Similarly, certain oncogenes (such as CXCR4, MMP-related genes, etc.) can serve as markers of tumor behavior, such as malignancy or prognosis, but targeting these genes alone may not lead to tumor regression. We pioneered the name of this class of genes as "phenotypic genes". Historically, cancer genetics research has focused on tumor driver genes, while genes influencing cancer phenotypes have been relatively overlooked. This review explores the critical distinction between driver genes and phenotypic genes in cancer, using the MAPK and PI3K/AKT/mTOR pathways as key examples. We also discuss current research techniques for identifying driver and phenotypic genes, such as whole-genome sequencing (WGS), RNA sequencing (RNA-seq), RNA interference (RNAi), CRISPR-Cas9, and other genomic screening methods, alongside the concept of synthetic lethality in driver genes. The development of these technologies will help develop personalized treatment strategies and precision medicine based on the characteristics of relevant genes. By addressing the gap in discussions on phenotypic genes, this review significantly contributes to clarifying the roles of driver and phenotypic genes, aiming at advancing the field of targeted cancer therapy.
Collapse
Affiliation(s)
- Dequan Liu
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, China
| | - Lei Liu
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, China
| | - Xiaoman Zhang
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, China
| | - Xinming Zhao
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, China
| | - Xiaorui Li
- Department of Oncology, Cancer Hospital of Dalian University of Technology, Shenyang, 110042, China.
| | - Xiangyu Che
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, China.
| | - Guangzhen Wu
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, China.
| |
Collapse
|
107
|
Ipe R, Oh JM, Kumar S, Ahmad I, Nath LR, Bindra S, Patel H, Kolachi KY, Prabhakaran P, Gahtori P, Syed A, Elgorbanh AM, Kim H, Mathew B. Inhibition of monoamine oxidases and neuroprotective effects: chalcones vs. chromones. Mol Divers 2025; 29:2063-2079. [PMID: 39145880 DOI: 10.1007/s11030-024-10959-w] [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: 01/22/2024] [Accepted: 08/05/2024] [Indexed: 08/16/2024]
Abstract
Eighteen compounds derived from two sub-series, (HC1-HC9) and (HF1-HF9), were synthesized and evaluated for their inhibitory activities against monoamine oxidase (MAO). HC (chalcone) series showed higher inhibitory activity against MAO-B than against MAO-A, whereas the HF (chromone) series showed reversed inhibitory activity. Compound HC4 most potently inhibited MAO-B with an IC50 value of 0.040 μM, followed by HC3 (IC50 = 0.049 μM), while compound HF4 most potently inhibited MAO-A (IC50 = 0.046 μM), followed by HF2 (IC50 = 0.075 μM). The selectivity index (SI) values of HC4 and HF4 were 50.40 and 0.59, respectively. Structurally, HC4 (4-OC2H5 in B-ring) showed higher MAO-B inhibition than other derivatives, suggesting that the -OC2H5 substitution of the 4-position in the B-ring contributes to the increase of MAO-B inhibition, especially -OC2H5 (HC4) > -OCH3 (HC3) > -F (HC7) > -CH3 (HC2) > -Br (HC8) > -H (HC1) in order. In MAO-A inhibition, the substituent 4-OC2H5 in the B-ring of HF4 contributed to an increase in inhibitory activity, followed by -CH3 (HF2), -F (HF7), -Br (HF8), -OCH3 (HF3), and-H (HF1). In the enzyme kinetics and reversibility study, the Ki value of HC4 for MAO-B was 0.035 ± 0.005 μM, and that of HF4 for MAO-A was 0.035 ± 0.005 μM, and both were reversible competitive inhibitors. We confirmed that HC4 and HF4 significantly ameliorated rotenone-induced neurotoxicity, as evidenced by the reactive oxygen species and superoxide dismutase assays. This study also supports the significant effect of HC4 and HF4 on mitochondrial membrane potential in rotenone-induced toxicity. A lead molecule was used for molecular docking and dynamic simulation studies. These results show that HC4 is a potent selective MAO-B inhibitor and HF4 is a potent MAO-A inhibitor, suggesting that both compounds can be used as treatment agents for neurological disorders.
Collapse
Affiliation(s)
- Reshma Ipe
- Department of Pharmaceutical Chemistry, Amrita School of Pharmacy, AIMS Health Sciences Campus, Amrita Vishwa Vidyapeetham, Kochi, 682041, India
| | - Jong Min Oh
- Department of Pharmacy, and Research Institute of Life Pharmaceutical Sciences, Sunchon National University, Suncheon, 57922, Republic of Korea
| | - Sunil Kumar
- Department of Pharmaceutical Chemistry, Amrita School of Pharmacy, AIMS Health Sciences Campus, Amrita Vishwa Vidyapeetham, Kochi, 682041, India
| | - Iqrar Ahmad
- Department of Pharmaceutical Chemistry, Prof. Ravindra Nikam College of Pharmacy, Gondur, Maharashtra, 424002, India
| | - Lekshmi R Nath
- Department of Pharmacognosy, Amrita School of Pharmacy, AIMS Health Sciences Campus, Amrita Vishwa Vidyapeetham, Kochi, 682041, India
| | - Sandeep Bindra
- Department of Pharmaceutical Chemistry, Amrita School of Pharmacy, AIMS Health Sciences Campus, Amrita Vishwa Vidyapeetham, Kochi, 682041, India
| | - Harun Patel
- Division of Computer Aided Drug Design, Department of Pharmaceutical Chemistry, R. C. Patel Institute of Pharmaceutical Education and Research, Shirpur, Maharashtra, 425405, India
| | - Krishna Yallappa Kolachi
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Mysuru, 570015, India
| | - Prabitha Prabhakaran
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Mysuru, 570015, India
| | - Prashant Gahtori
- School of Pharmacy, Graphic Era Hill University, Dehradun, Uttarakhand, 248007, India
| | - Asad Syed
- Department of Botany and Microbiology, College of Science, King Saud University, P.O. 2455, Riyadh, 11451, Saudi Arabia
| | - Abdallah M Elgorbanh
- Department of Botany and Microbiology, College of Science, King Saud University, P.O. 2455, Riyadh, 11451, Saudi Arabia
| | - Hoon Kim
- Department of Pharmacy, and Research Institute of Life Pharmaceutical Sciences, Sunchon National University, Suncheon, 57922, Republic of Korea.
| | - Bijo Mathew
- Department of Pharmaceutical Chemistry, Amrita School of Pharmacy, AIMS Health Sciences Campus, Amrita Vishwa Vidyapeetham, Kochi, 682041, India.
| |
Collapse
|
108
|
Chaudhuri R, Dayal N, Kaiser J, Mohallem R, Brauer NR, Yeboah KS, Aryal UK, Sintim HO. Morpholino nicotinamide analogs of ponatinib, dual MNK, p70S6K inhibitors, display efficacy against lung and breast cancers. Bioorg Chem 2025; 159:108298. [PMID: 40081260 DOI: 10.1016/j.bioorg.2025.108298] [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: 12/04/2024] [Revised: 02/13/2025] [Accepted: 02/18/2025] [Indexed: 03/15/2025]
Abstract
Therapeutic options for aggressive cancer types such as breast and lung remain limited; disease relapse and death occur in 30-60% of non-small cell lung cancer (NSCLC) patients, whereas in triple-negative breast cancer or TNBC, recurrence-free survival occurs in less than 30% patients. The kinases, MNK and p70S6K have been proposed as targets for the potential treatment of breast cancer (BC) and lung cancer but currently, no drug that was purposely designed to inhibit these kinases have been approved by the FDA for the treatment of BC or NSCLC. In this study, we have identified HSND80 (a morpholino nicotinamide analog of ponatinib) as a potent MNK/p70S6K inhibitor that has excellent activity against TNBC and NSCLC cell lines. HSND80 has a longer target residence time (τ) of 45 mins and 58 mins against MNK1 and MNK2 respectively, compared to τ of eFT508 (tomivosertib) against MNK1 and MNK2 (τ = 1 min and 5 min, respectively). Molecular dynamics simulation was used to provide some insights into the binding of HSND80 to MNK and p70S6K kinases. Western blotting analysis and phosphoproteomics analysis of the TNBC cell line, MDA-MB-231, revealed that phosphorylations of elF4E (MNK target) and elF4B and S6 (p70S6K targets) were reduced upon compound treatment, which is in line with the proposed mechanism of action; dual MNK/p70S6K targeting. HSND80 could be dosed orally at 15 and 30 mg/kg and at such doses, could reduce tumor volume in a syngeneic NSCLC mouse model.
Collapse
Affiliation(s)
- Riddhi Chaudhuri
- Department of Chemistry, Purdue University, 560 Oval Drive, West Lafayette, IN 47907, USA; Purdue Institute for Drug Discovery, Purdue University, 720 Clinic Drive, West Lafayette, IN 47907, USA
| | - Neetu Dayal
- Department of Chemistry, Purdue University, 560 Oval Drive, West Lafayette, IN 47907, USA; Purdue Institute for Drug Discovery, Purdue University, 720 Clinic Drive, West Lafayette, IN 47907, USA
| | - Joshua Kaiser
- Department of Chemistry, Purdue University, 560 Oval Drive, West Lafayette, IN 47907, USA; Purdue Institute for Drug Discovery, Purdue University, 720 Clinic Drive, West Lafayette, IN 47907, USA
| | - Rodrigo Mohallem
- Department of Comparative Pathobiology, Purdue University, 1203 W State Street, West Lafayette, IN 47907, USA
| | - Nickolas R Brauer
- Department of Chemistry, Purdue University, 560 Oval Drive, West Lafayette, IN 47907, USA; Purdue Institute for Drug Discovery, Purdue University, 720 Clinic Drive, West Lafayette, IN 47907, USA
| | - Kofi Simpa Yeboah
- Department of Chemistry, Purdue University, 560 Oval Drive, West Lafayette, IN 47907, USA; Purdue Institute for Drug Discovery, Purdue University, 720 Clinic Drive, West Lafayette, IN 47907, USA
| | - Uma K Aryal
- Department of Comparative Pathobiology, Purdue University, 1203 W State Street, West Lafayette, IN 47907, USA; Purdue Proteomics Facility, Bindley Bioscience Center, Purdue University, 1203 W State Street, West Lafayette, IN 47907, USA
| | - Herman O Sintim
- Department of Chemistry, Purdue University, 560 Oval Drive, West Lafayette, IN 47907, USA; Purdue Institute for Drug Discovery, Purdue University, 720 Clinic Drive, West Lafayette, IN 47907, USA; Purdue Institute for Cancer Research, Purdue University, 201 S. University Street, West Lafayette, IN 47907, USA; Department of Chemistry and Biochemistry, University of Notre Dame, 305A McCourtney Hall, Notre Dame, IN 46556, USA; Mike and Josie Harper Cancer Research Institute, 1234 N. Notre Dame Avenue, South Bend, IN 46617, USA.
| |
Collapse
|
109
|
Wang J, Shi X, Wang J, Zheng Q, Shao P, Liu S. Discovery of new covalent inhibitors of monoacylglycerol lipase with the nitrile warhead via SCARdock. Bioorg Chem 2025; 159:108378. [PMID: 40107037 DOI: 10.1016/j.bioorg.2025.108378] [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: 10/30/2024] [Revised: 03/03/2025] [Accepted: 03/12/2025] [Indexed: 03/22/2025]
Abstract
Monoacylglycerol lipase (MAGL) is an important enzyme for endocannabinoid metabolism by converting 2-arachidonoylglycerol (2-AG) into glycerol and free fatty acids. Modulation of the endocannabinoid system by inhibiting MAGL provides a promising therapeutic strategy for various diseases. In this work, we identified five new MAGL inhibitors with the nitrile group by high-throughput screening using SCARdock, a protocol presented by us for covalent drug discovery. Compounds ZQ-4, ZQ-5, ZQ-6, and ZQ-7 inhibit MAGL activity in a time-dependent and concentration-dependent manner. Furthermore, ZQ-7 was confirmed to covalently bind with the residue Ser132 of MAGL. The nitrile group is a new covalent warhead that has never been used in previous covalent MAGL inhibitors. At last, the efficacy of the new MAGL inhibitors on inhibiting breast cancer cells was investigated. Significantly increased 2-AG levels were detected in MDA-MB-231 cells treated with MAGL inhibitor ZQ-5, ZQ-6, ZQ-7, ZQ-19, and KML29, a previously identified MAGL covalent inhibitor. Moreover, these MAGL inhibitors inhibited the proliferation and migration of MDA-MB-231 cells. This work expands the application of SCARdock and provides meaningful clues for developing better MAGL inhibitors.
Collapse
Affiliation(s)
- Juanping Wang
- Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Wuhan 430068, China; Key Laboratory of Fermentation Engineering (Ministry of Education), Wuhan 430068, China, National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Hubei University of Technology, Wuhan 430068, China
| | - Xiaoyu Shi
- Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Wuhan 430068, China; Key Laboratory of Fermentation Engineering (Ministry of Education), Wuhan 430068, China, National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Hubei University of Technology, Wuhan 430068, China
| | - Junlai Wang
- Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Wuhan 430068, China; Key Laboratory of Fermentation Engineering (Ministry of Education), Wuhan 430068, China, National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Hubei University of Technology, Wuhan 430068, China
| | - Qiang Zheng
- Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Wuhan 430068, China; Key Laboratory of Fermentation Engineering (Ministry of Education), Wuhan 430068, China, National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Hubei University of Technology, Wuhan 430068, China
| | - Peipei Shao
- Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Wuhan 430068, China; Key Laboratory of Fermentation Engineering (Ministry of Education), Wuhan 430068, China, National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Hubei University of Technology, Wuhan 430068, China
| | - Sen Liu
- Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Wuhan 430068, China; Key Laboratory of Fermentation Engineering (Ministry of Education), Wuhan 430068, China, National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Hubei University of Technology, Wuhan 430068, China
| |
Collapse
|
110
|
Choi B, Lee H, Popova E, Paik MJ, Kim H. Dynamics of amino acids in the shoot tips of Pogostemon yatabeanus during cryopreservation by droplet-vitrification method and its modifications. Cryobiology 2025; 119:105244. [PMID: 40228354 DOI: 10.1016/j.cryobiol.2025.105244] [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: 01/08/2025] [Revised: 04/03/2025] [Accepted: 04/03/2025] [Indexed: 04/16/2025]
Abstract
Cryopreservation offers effective long-term conservation of the endangered plant species with non-orthodox or unavailable seeds. Shoot tips of Pogostemon yatabeanus, an endemic Korean species, were successfully cryopreserved via the optimized droplet-vitrification (DV) method which included 10 % sucrose preculture, osmoprotection with 17.5 % glycerol + 17.5 % sucrose, vitrification solution (33.3 % glycerol + 13.3 % dimethyl sulfoxide + 13.3 % ethylene glycol + 20.1 % sucrose) treatment and cooling-rewarming using aluminum foil strips. Regrowth was performed in three steps, starting with the ammonium-free medium with growth regulators, followed by full-strength medium with and without growth regulators in the second and the third steps. The contents of 17 amino acids (AA) in shoot tips were analyzed using gas-chromatography-mass-spectrometry (GC-MS/MS) at each step of the optimum DV protocol and in the following modifications: excluding or modifying preculture or osmoprotection, varying vitrification solution composition, cooling-rewarming in vials, change of regrowth conditions. In the optimum DV procedure, the total content of AA increased by 20-40 % at preculture and osmoprotection, dropped to 6 % of their level in freshly excised shoot tips after the first five days of regrowth, and raised again after two weeks of recovery. The contents of alanine, glycine, β-alanine, serine, and γ-aminobutyric acid were maximized during osmoprotection and vitrification solution treatments (6.27-29.75-fold compared to untreated control). In particular, alanine showed a 30-fold peak during osmoprotection. Excluding preculture or osmoprotection decreased total and individual AA concentrations by 40-70 %. Meanwhile, a high (25 %) sucrose concentration during preculture increased AA' contents almost twice compared to the optimum protocol. Modifications of vitrification solutions caused 20-30 % variations in AA contents. Therefore, AA metabolism is essential in shoot tips' response to osmotic and freezing stresses and regeneration.
Collapse
Affiliation(s)
- Byeongchan Choi
- College of Pharmacy, Sunchon National University, Suncheon, 57922, Republic of Korea.
| | - Hyoeun Lee
- Department of Agricultural Life Science, Sunchon National University, Suncheon, 57922, Republic of Korea.
| | - Elena Popova
- K.A. Timiryazev Institute of Plant Physiology of Russian Academy of Sciences, Botanicheskaya 35, Moscow, 127276, Russia.
| | - Man-Jeong Paik
- College of Pharmacy, Sunchon National University, Suncheon, 57922, Republic of Korea.
| | - Haenghoon Kim
- Department of Agricultural Life Science, Sunchon National University, Suncheon, 57922, Republic of Korea.
| |
Collapse
|
111
|
Wang J, Chen M, Wei G, Zou F, Gu J, Cao Y, Deng S, Cai K. From blockage to biology: Unveiling the role of extracellular matrix dynamics in obstructive colorectal cancer pathogenesis. Pathol Res Pract 2025; 270:155994. [PMID: 40306003 DOI: 10.1016/j.prp.2025.155994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2025] [Revised: 04/23/2025] [Accepted: 04/28/2025] [Indexed: 05/02/2025]
Abstract
Colorectal cancer obstruction is a common problem with distinct symptomatic clues on CT/MR images even under incomplete conditions. The choice of management in the emergency setting has a significant effect on the prognosis of obstructive and nonobstructive colorectal cancer patients. Previous studies have demonstrated that obstruction in colorectal cancer is associated with significantly poorer outcomes, alongside distinct alterations in the composition of the extracellular matrix. Based on accumulating evidence, it is hypothesized that ECM remodeling plays a pivotal role in the development of colorectal cancer obstruction. This review explores the pathological features of obstructive colorectal cancer, emphasizing extracellular matrix remodeling as a central process. Key mechanisms include tumor-stromal cell interactions, tumor cell aggregation and migration mediated by the peripheral nervous system, vascular and lymphatic remodeling within the tumor microenvironment, and microbiota-mediated regulation of cancer progression. These findings demonstrate that further remodeling of the extracellular matrix may be a molecular biological feature of obstructive colorectal cancer with poor prognosis.
Collapse
Affiliation(s)
- Jun Wang
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Mian Chen
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Guanxin Wei
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Falong Zou
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Junnan Gu
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yinghao Cao
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore 119074, Singapore; Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
| | - Shenghe Deng
- Center for Liver Transplantation, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - Kailin Cai
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| |
Collapse
|
112
|
Abbas MF, Karim DK, Kareem HR, Kamil MM, Al-Musawi MH, Asker MH, Ghanami M, Shahriari-Khalaji M, Sattar M, Mirhaj M, Sharifianjazi F, Tavamaishvili K, Mohabbatkhah M, Soheily A, Noory P, Tavakoli M. Fucoidan and its derivatives: From extraction to cutting-edge biomedical applications. Carbohydr Polym 2025; 357:123468. [PMID: 40158992 DOI: 10.1016/j.carbpol.2025.123468] [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: 01/04/2025] [Revised: 02/10/2025] [Accepted: 02/28/2025] [Indexed: 04/02/2025]
Abstract
Fucoidan, a sulfated polymeric carbohydrate isolated from various marine brown algae, has attracted the interest of biomedical scientists because of its unique structural features and extensive spectrum of biological activity. This review encompasses a comprehensive insight into fucoidan's extraction procedures, cross-linking strategies, chemical modifications, and biomedical applications. Advanced extraction methods, such as microwave-assisted and enzyme-assisted extraction, are emphasized to get high-quality fucoidan that has augmented bioactivity. Moreover, the production and role of fucoidan-based materials in drug delivery systems are investigated, with a focus on their potential for targeted therapies. The study also explores the strategies to improve fucoidan's bioavailability and mechanical properties via structural modifications, such as Sulfation, desulfation, methylation, benzoylation, sulfation, amination, acetylation and phosphorylation, and cross-linking with other polymers to form films, hydrogels, and nanocomposites. In addition, fucoidan's applications in drug delivery systems, tissue engineering, microneedles, and 3D bioprinting are discussed. By summarizing current research findings, this study seeks to comprehend the mechanisms underpinning fucoidan's therapeutic efficacy and its potential to develop robust biomaterials.
Collapse
Affiliation(s)
- Marwa F Abbas
- Department of Clinical Laboratory Sciences, College of Pharmacy, Mustansiriyah University, Baghdad, Iraq.
| | - Dhuha K Karim
- Department of Clinical Laboratory Sciences, College of Pharmacy, Mustansiriyah University, Baghdad, Iraq.
| | - Huda Raad Kareem
- Department of Clinical Laboratory Sciences, College of Pharmacy, Mustansiriyah University, Baghdad, Iraq.
| | - Marwa M Kamil
- Department of Pharmaceutics, College of Pharmacy, Mustansiriyah University, Baghdad, Iraq.
| | - Mastafa H Al-Musawi
- Department of Clinical Laboratory Sciences, College of Pharmacy, Mustansiriyah University, Baghdad, Iraq.
| | - Mohammed Hayder Asker
- Department of pharmacology and toxicology, college of pharmacy, Mustansiriyah University Baghdad, Iraq.
| | - Maral Ghanami
- Department of Mechanical Engineering, Rowan University, Glassboro, NJ 08028, USA.
| | | | - Mamoona Sattar
- College of Biological Science and Medical Engineering, Donghua University, No. 2999 North Renmin Road, Shanghai 201620, China
| | - Marjan Mirhaj
- Department of Materials Engineering, Isfahan University of Technology, Isfahan 84156-83111, Iran
| | - Fariborz Sharifianjazi
- Center for Advanced Materials and Structures, School of Science and Technology, The University of Georgia, Tbilisi 0171, Georgia; Department of Civil Engineering, School of Science and Technology, The University of Georgia, 0171 Tbilisi, Georgia.
| | - Ketevan Tavamaishvili
- Georgian American University, School of Medicine, 10 Merab Aleksidze Str., Tbilisi 0160, Georgia.
| | - Mehdi Mohabbatkhah
- Faculty of Engineering and Natural Sciences, Istinye University, Istanbul, Turkey
| | - Ali Soheily
- Advanced Materials Research Center, Department of Materials Engineering, Najafabad Branch, Islamic Azad University, Najafabad, Iran
| | - Parastoo Noory
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohamadreza Tavakoli
- Department of Materials Engineering, Isfahan University of Technology, Isfahan 84156-83111, Iran.
| |
Collapse
|
113
|
Kumar BVS, Talamadla MK, Nandikolla A, Khetmalis YM, Shetye G, Franzblau SG, Murugesan S, Sekhar KVGC. Exploration of quinoxaline triazoles as antimycobacterial agents: design, synthesis and biological evaluation. Bioorg Med Chem Lett 2025; 121:130177. [PMID: 40058415 DOI: 10.1016/j.bmcl.2025.130177] [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: 01/29/2025] [Revised: 02/25/2025] [Accepted: 03/05/2025] [Indexed: 03/16/2025]
Abstract
In this work, novel 2-substituted-3-((1-substituted-1H-1,2,3-triazol-4-yl) methoxy) quinoxaline analogues were designed, synthesized, and various analytical techniques, viz., 1H NMR, 13C NMR, and Mass spectrometry, were deployed in the structure confirmation of the final compounds. Synthesized derivatives were evaluated for their antimycobacterial activity against Mycobacterium tuberculosis (Mtb) H37Rv. Target molecules mainly consist of methyl substituent in the second position of quinoxaline moiety (QM series) or phenyl substituent in the second position (QP series). Among the forty-two compounds synthesized and evaluated for anti-mycobacterial activity, the MIC values ranged between 5.58 μg/mL to >100 μg/mL. Among QM series compounds, QM7, with MIC 5.58 μg /mL, was the most active compound. Among the QP series derivatives, the intermediate QP-Acy with MIC 23.39 μg /mL was the most promising. Most of the analogues tested in the QP series are less potent than the QM series. All the synthesized molecules showed good drug-likeness when evaluated using the SWISS ADME tool. QM7 was evaluated for docking studies using the crystal structure of enoyl-acyl carrier (INH-A) enzyme PDB: 4TZK, and it showed significant docking scores and interactions. MD simulations were carried out to assess the stability of the protein QM7 complex. Single crystals were grown for QM1, QM6, and QPb from these forty-two compounds, and their structures were solved using OLEX. The corresponding CCDC numbers for these compounds are 2,388,310, 2,388,309, and 2,388,291, respectively.
Collapse
Affiliation(s)
- Boddupalli Venkata Siva Kumar
- Department of Chemistry, Birla Institute of Technology and Science, Pilani, Hyderabad Campus, Jawahar Nagar, Kapra Mandal, Hyderabad 500078, Telangana, India
| | - Mahesh Kumar Talamadla
- Department of Chemistry, Birla Institute of Technology and Science, Pilani, Hyderabad Campus, Jawahar Nagar, Kapra Mandal, Hyderabad 500078, Telangana, India
| | - Adinarayana Nandikolla
- Department of Chemistry, Birla Institute of Technology and Science, Pilani, Hyderabad Campus, Jawahar Nagar, Kapra Mandal, Hyderabad 500078, Telangana, India
| | - Yogesh Mahadu Khetmalis
- Department of Chemistry, Birla Institute of Technology and Science, Pilani, Hyderabad Campus, Jawahar Nagar, Kapra Mandal, Hyderabad 500078, Telangana, India
| | - Gauri Shetye
- Institute for Tuberculosis Research, College of Pharmacy, University of Illinois at Chicago, 833 South Wood Street, Chicago, IL 60612, USA
| | - Scott G Franzblau
- Institute for Tuberculosis Research, College of Pharmacy, University of Illinois at Chicago, 833 South Wood Street, Chicago, IL 60612, USA
| | - Sankaranarayanan Murugesan
- Medicinal Chemistry Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science Pilani, Pilani Campus, Pilani 333031, Rajasthan. India
| | - Kondapalli Venkata Gowri Chandra Sekhar
- Department of Chemistry, Birla Institute of Technology and Science, Pilani, Hyderabad Campus, Jawahar Nagar, Kapra Mandal, Hyderabad 500078, Telangana, India.
| |
Collapse
|
114
|
Muñoz-Osses M, Navarrete E, Morales P, Quiroz J, Silva M, Torres-González S, Vásquez-Martínez Y, Godoy F, Mascayano C. Substituted aryl piperazine ligands as new dual 5-hLOX/COX-2 inhibitors. Synthesis, biological and computational studies. Bioorg Chem 2025; 159:108398. [PMID: 40174530 DOI: 10.1016/j.bioorg.2025.108398] [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: 01/13/2025] [Revised: 03/10/2025] [Accepted: 03/18/2025] [Indexed: 04/04/2025]
Abstract
Two series of cyano (1a-l) and amino (2a-l) aryl piperazines were synthesized and evaluated for their inhibitory activity against 5-lipoxygenase (5-hLOX) and cyclooxygenase-2 (COX-2). The newly designed derivatives feature diphenyl methyl (a-d), phenyl (e-h), or methoxyphenyl (i-l) groups, respectively, and demonstrated significant inhibition of 5-hLOX. Noteworthy were compounds 1b, 1 g, 1 k, 2f, and 2 g, exhibiting IC50 values ranging from 2.2 to 3.3 μM. The most potent inhibitors (1b, 1 g, 1 k, 2c, and 2f) were characterized by a competitive inhibition mechanism, with Ki values ranging between 1.77 μM and 9.50 μM. Additionally, compounds 2a, 2b, 2 g, and 2 h displayed promising dual inhibition of 5-hLOX and COX-2, with IC50 values below 15 μM. Cytotoxicity assessments against HEK293 cells revealed that the cyano derivatives (1a-l) were non-cytotoxic (CC50 > 200 μM), whereas the amino derivatives (2a-l) exhibited moderate cytotoxicity (CC50 < 50 μM). Notably, the most active derivatives against both targets were non-cytotoxic at their respective inhibitory concentrations. Computational studies, including docking and molecular dynamics simulations, indicated that compound 1 g demonstrated greater stability within the catalytic site of 5-hLOX compared to compound 2f, correlating with the higher affinity observed in kinetic assays. Furthermore, quantitative structure-activity relationship (QSAR) analyses revealed strong correlations between theoretical and experimental IC50 values (97 % for 1a-l and 93 % for 2a-l). These findings, combined with absorption, distribution, metabolism, and excretion (ADME) predictions, suggest that these derivatives are promising candidates as dual inhibitors of 5-hLOX and COX-2.
Collapse
Affiliation(s)
| | | | - Pilar Morales
- Departamento Ciencias del Ambiente, Universidad de Santiago de Chile, Chile
| | - Javiera Quiroz
- Departamento Ciencias del Ambiente, Universidad de Santiago de Chile, Chile
| | - Maite Silva
- Departamento Química de los Materiales, Universidad de Santiago de Chile, Chile
| | | | - Yesseny Vásquez-Martínez
- Programa Centro de Investigaciones Biomédicas y Aplicadas (CIBAP), Escuela de Medicina, Facultad de Ciencias Médicas, Universidad de Santiago de Chile, Chile
| | - Fernando Godoy
- Departamento Química de los Materiales, Universidad de Santiago de Chile, Chile
| | - Carolina Mascayano
- Departamento Ciencias del Ambiente, Universidad de Santiago de Chile, Chile
| |
Collapse
|
115
|
Chahat, Nainwal N, Murti Y, Yadav S, Rawat P, Dhiman S, Kumar B. Advancements in targeting tumor suppressor genes (p53 and BRCA 1/2) in breast cancer therapy. Mol Divers 2025; 29:2691-2716. [PMID: 39152355 DOI: 10.1007/s11030-024-10964-z] [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: 06/07/2024] [Accepted: 08/07/2024] [Indexed: 08/19/2024]
Abstract
Globally, among numerous cancer subtypes, breast cancer (BC) is one of the most prevalent forms of cancer affecting the female population. A female's family history significantly increases her risk of developing breast cancer. BC is caused by aberrant breast cells that proliferate and develop into tumors. It is estimated that 5-10% of breast carcinomas are inherited and involve genetic mutations that ensure the survival and prognosis of breast cancer cells. The most common genetic variations are responsible for hereditary breast cancer but are not limited to p53, BRCA1, and BRCA2. BRCA1 and BRCA2 are involved in genomic recombination, cell cycle monitoring, programmed cell death, and transcriptional regulation. When BRCA1 and 2 genetic variations are present in breast carcinoma, p53 irregularities become more prevalent. Both BRCA1/2 and p53 genes are involved in cell cycle monitoring. The present article discusses the current status of breast cancer research, spotlighting the tumor suppressor genes (BRCA1/2 and p53) along with structural activity relationship studies, FDA-approved drugs, and several therapy modalities for treating BC. Breast cancer drugs, accessible today in the market, have different side effects including anemia, pneumonitis, nausea, lethargy, and vomiting. Thus, the development of novel p53 and BRCA1/2 inhibitors with minimal possible side effects is crucial. We have covered compounds that have been examined subsequently (2020 onwards) in this overview which may be utilized as lead compounds. Further, we have covered mechanistic pathways to showcase the critical druggable targets and clinical and post-clinical drugs targeting them for their utility in BC.
Collapse
Affiliation(s)
- Chahat
- Department of Pharmaceutical Sciences, HNB Garhwal University, Chauras Campus, Srinagar, 246174, Uttarakhand, India
| | - Nidhi Nainwal
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Premanagar, Dehradun, 248007, Uttarakhand, India
| | - Yogesh Murti
- Institute of Pharmaceutical Research, GLA University, Mathura, 281406, India
| | - Savita Yadav
- IES Institute of Technology and Management, IES University, Bhopal, 462044, Madhya Pradesh, India
| | - Pramod Rawat
- Graphic Era (Deemed to Be University), Clement Town, Dehradun, 248002, India
- Graphic Era Hill University Clement Town, Dehradun, 248002, India
| | - Sonia Dhiman
- Centre for Research Impact & Outcome, Chitkara College of Pharmacy, Chitkara University, Rajpura, 140401, Punjab, India
| | - Bhupinder Kumar
- Department of Pharmaceutical Sciences, HNB Garhwal University, Chauras Campus, Srinagar, 246174, Uttarakhand, India.
| |
Collapse
|
116
|
Arumugam M, Pachamuthu RS, Rymbai E, Jha AP, Rajagopal K, Kothandan R, Muthu S, Selvaraj D. Gene network analysis combined with preclinical studies to identify and elucidate the mechanism of action of novel irreversible Keap1 inhibitor for Parkinson's disease. Mol Divers 2025; 29:2081-2098. [PMID: 39145879 DOI: 10.1007/s11030-024-10965-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 08/07/2024] [Indexed: 08/16/2024]
Abstract
The cysteine residues of Keap1 such as C151, C273, and C288 are critical for its repressor activity on Nrf2. However, to date, no molecules have been identified to covalently modify all three cysteine residues for Nrf2 activation. Hence, in this study, our goal is to discover new Keap1 covalent inhibitors that can undergo a Michael addition with all three cysteine residues. The Keap1's intervening region was modeled using Modeller v10.4. Covalent docking and binding free energy were calculated using CovDock. Molecular dynamics (MD) was performed using Desmond. Various in-vitro assays were carried out to confirm the neuroprotective effects of the hit molecule in 6-OHDA-treated SH-SY5Y cells. Further, the best hit was evaluated in vivo for its ability to improve rotenone-induced postural instability and cognitive impairment in male rats. Finally, network pharmacology was used to summarize the complete molecular mechanism of the hit molecule. Chalcone and plumbagin were found to form the necessary covalent bonds with all three cysteine residues. However, MD analysis indicated that the binding of plumbagin is more stable than chalcone. Plumbagin displayed neuroprotective effects in 6-OHDA-treated SH-SY5Y cells at concentrations 0.01 and 0.1 μM. Plumbagin at 0.1 µM had positive effects on reactive oxygen species formation and glutathione levels. Plumbagin also improved postural instability and cognitive impairment in rotenone-treated male rats. Our network analysis indicated that plumbagin could also improve dopamine signaling. Additionally, plumbagin could exhibit anti-oxidant and anti-inflammatory activity through the activation of Nrf2. Cumulatively, our study suggests that plumbagin is a novel Keap1 covalent inhibitor for Nrf2-mediated neuroprotection in PD.
Collapse
Affiliation(s)
- Monisha Arumugam
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, Nilgiris, Tamil Nadu, India
| | - Ranjith Sanjeeve Pachamuthu
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, Nilgiris, Tamil Nadu, India
| | - Emdormi Rymbai
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, Nilgiris, Tamil Nadu, India
| | - Aditya Prakash Jha
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, Nilgiris, Tamil Nadu, India
| | - Kalirajan Rajagopal
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Ooty, Nilgiris, Tamil Nadu, India
| | - Ram Kothandan
- Bioinformatics Laboratory, Department of Biotechnology, Kumaraguru College of Technology, Coimbatore, Tamil Nadu, India
| | - Santhoshkumar Muthu
- Department of Biochemistry, Kongunadu Arts and Science College, GN Mills, Coimbatore, Tamil Nadu, India.
| | - Divakar Selvaraj
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, Nilgiris, Tamil Nadu, India.
| |
Collapse
|
117
|
Chen L, Li Q. Nanomaterials in the diagnosis and treatment of gastrointestinal tumors: New clinical choices and treatment strategies. Mater Today Bio 2025; 32:101782. [PMID: 40331152 PMCID: PMC12051065 DOI: 10.1016/j.mtbio.2025.101782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2025] [Revised: 04/14/2025] [Accepted: 04/18/2025] [Indexed: 05/08/2025] Open
Abstract
Nanomaterials have emerged as a promising modality in the diagnosis and treatment of gastrointestinal (GI) tumors, offering significant advancements over conventional methods. In diagnostic applications, nanomaterials facilitate enhanced imaging techniques, including magnetic resonance imaging (MRI), computed tomography (CT), and fluorescence imaging, which provide improved resolution and more accurate detection of early-stage cancers. Nanoparticles (NPs), such as liposomes, dendrimers, and quantum dots, are increasingly employed for the targeted imaging of specific biomarkers associated with GI malignancies, thereby enhancing diagnostic sensitivity and specificity. Liposomes are primarily used for drug delivery due to their ability to encapsulate hydrophobic drugs, dendrimers are useful for both drug delivery and gene therapy due to their highly branched structure, and quantum dots are primarily used in imaging and diagnostics because of their fluorescent properties. We also discuss their respective advantages and limitations. In therapeutic contexts, nanomaterials play a pivotal role in the development of targeted drug delivery systems. These systems address the limitations of traditional chemotherapy by improving drug bioavailability, reducing systemic toxicity, and promoting selective accumulation at tumor sites via both passive and active targeting mechanisms. Nanomedicines, including NPs and nanocarriers, enable the precise delivery of chemotherapeutic agents, nucleic acid -based therapies, and immunomodulators directly to cancer cells, thereby optimizing therapeutic efficacy. Furthermore, nanotechnology offers the potential to modulate the tumor microenvironment (TME), a critical factor in overcoming challenges related to tumor resistance and metastasis. Despite these promising advancements, several challenges persist, including concerns regarding long-term toxicity, stability, and regulatory approval. Nonetheless, the integration of nanomaterials into clinical practice holds substantial potential for revolutionizing the management of GI cancers, paving the way for more precise, personalized, and effective therapeutic strategies.
Collapse
Affiliation(s)
- Liping Chen
- Department of Radiotherapy, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Cancer Hospital of Dalian University of Technology, No.44 Xiaoheyan Road, Dadong District, Shenyang, 110042, Liaoning Province, PR China
| | - Qingqing Li
- Department of Endoscopy, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Cancer Hospital of Dalian University of Technology, No.44 Xiaoheyan Road, Dadong District, Shenyang, 110042, Liaoning Province, PR China
| |
Collapse
|
118
|
Li L, Lu M, Wang H, Ma X, Du W, Zhao Y, Zeng S, Peng Y, Zhang G. A novel MMP-9 inhibitor exhibits selective inhibition in non-small-cell lung cancer harboring EGFR T790M mutation by blocking EGFR/STAT3 signaling pathway. Bioorg Chem 2025; 159:108393. [PMID: 40121769 DOI: 10.1016/j.bioorg.2025.108393] [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: 01/16/2025] [Revised: 03/12/2025] [Accepted: 03/16/2025] [Indexed: 03/25/2025]
Abstract
The T790M secondary mutation in EGFR confers therapeutic resistance to EGFR-TKIs, leading to poor outcomes. Non-small-cell lung cancer (NSCLC) harboring EGFR T790M mutation is incurable and there is an urgent need for improved therapeutics. Here we report the identification of a small compound, MG-3C, that kills NSCLC cells with T790M mutation while sparing lung cancer cells without T790M mutation. We found that MG-3C activity targets EGFR-STAT3 signaling pathway in NSCLC through direct inhibition of matrix metalloproteinase 9 (MMP-9), ultimately leading to G2/M phase arrest, growth inhibition and apoptosis. Compared with the reported MMP-9 inhibitor Ilomastat, MG-3C shows high anticancer activity and affinity for targets. MG-3C forms hydrogen bonds with the ASP-113, ASP-201 and HIS-203 amino acid residues of MMP-9 with a docking fraction of -9.04 kcal/mol, while Ilomastat forms hydrogen bonds with the GLN-169, ASP-201 and HIS-203 amino acid residues of MMP-9 with a docking fraction of -5.98 kcal/mol. The spatial structure composed of ASP-113, ASP-201, and HIS-203 of MMP-9 provides a new coordinate for the design of MMP-9 inhibitors. Most importantly, subcutaneous and oral administration of MG-3C elicit dramatic regression of NSCLC xenograft tumors harboring T790M mutation as well as favorable biosafety profile in vivo, suggesting that MG-3C may be a potential candidate for NSCLC harboring T790M mutation.
Collapse
Affiliation(s)
- Liangping Li
- Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, Guangxi Key Laboratory of Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, China; School of Pharmacy, Youjiang Medical University for Nationalities, Baise 533000, China
| | - Minghan Lu
- Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, Guangxi Key Laboratory of Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, China
| | - Hui Wang
- Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, Guangxi Key Laboratory of Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, China
| | - Xuesong Ma
- Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, Guangxi Key Laboratory of Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, China
| | - Wenqing Du
- Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, Guangxi Key Laboratory of Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, China
| | - Yufei Zhao
- Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, Guangxi Key Laboratory of Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, China
| | - Shulan Zeng
- Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, Guangxi Key Laboratory of Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, China.
| | - Yan Peng
- Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, Guangxi Key Laboratory of Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, China.
| | - Guohai Zhang
- Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, Guangxi Key Laboratory of Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, China.
| |
Collapse
|
119
|
Szabó R, Hornyánszky Á, Kiss DJ, Keserű GM. Fluorescent tools for imaging class A G-protein coupled receptors. Eur J Pharm Sci 2025; 209:107074. [PMID: 40113106 DOI: 10.1016/j.ejps.2025.107074] [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: 01/29/2025] [Revised: 03/15/2025] [Accepted: 03/17/2025] [Indexed: 03/22/2025]
Abstract
G protein-coupled receptors (GPCRs) are pivotal in biological processes and represent a significant class of drug targets, with 516 approved drugs acting on 121 GPCRs. Many GPCRs, particularly orphan receptors, remain underexplored, emphasizing the need for innovative investigative tools. Fluorescent ligands provide a powerful means to characterize GPCRs including their functional mechanisms and spatial organization, bridging fundamental research and drug discovery. This review presents recent advances (2018-2024) in fluorescent probe development for Class A GPCRs, analyzing over 120 newly developed probes covering 60 GPCRs. We examine their distribution across receptor subclasses, comparing pre-2018 data with contemporary findings and identifying previously uncharted GPCRs that now have fluorescent ligands. Notably, novel probes have been developed for 12 new receptor subtypes and 6 orphan receptors such as GPR6, GPR52, GPR84, MAS1, MRGPRX2, and MRGPRX4. Advances in GPCR structural biology, driven by cryo-EM and AlphaFold technologies, have significantly enhanced probe development, facilitating the design of selective fluorescent ligands across aminergic, peptidergic, lipid, nucleotide, alicarboxylic, melatonin, protein, and orphan GPCRs. These innovations support a broad range of applications, from single-molecule imaging and in vivo bioimaging to diagnostics and fluorescence-guided surgery. By integrating fluorescence-based approaches with structural and pharmacological insights, this field continues to refine polypharmacology profiling, optimize drug-receptor interactions, and accelerate GPCR-targeted drug discovery.
Collapse
Affiliation(s)
- Renáta Szabó
- Medicinal Chemistry Research Group, HUN-REN Research Centre for Natural Sciences, H-1117 Budapest, Hungary; Department of Organic Chemistry and Technology, Budapest University of Technology and Economics, H-1111 Budapest, Hungary; National Laboratory for Drug Research and Development, H-1117 Budapest, Hungary
| | - Ágnes Hornyánszky
- Medicinal Chemistry Research Group, HUN-REN Research Centre for Natural Sciences, H-1117 Budapest, Hungary; Department of Organic Chemistry and Technology, Budapest University of Technology and Economics, H-1111 Budapest, Hungary; National Laboratory for Drug Research and Development, H-1117 Budapest, Hungary
| | - Dóra Judit Kiss
- Medicinal Chemistry Research Group, HUN-REN Research Centre for Natural Sciences, H-1117 Budapest, Hungary; National Laboratory for Drug Research and Development, H-1117 Budapest, Hungary
| | - György Miklós Keserű
- Medicinal Chemistry Research Group, HUN-REN Research Centre for Natural Sciences, H-1117 Budapest, Hungary; Department of Organic Chemistry and Technology, Budapest University of Technology and Economics, H-1111 Budapest, Hungary; National Laboratory for Drug Research and Development, H-1117 Budapest, Hungary.
| |
Collapse
|
120
|
Poggialini F, Governa P, Vagaggini C, Maramai S, Lamponi S, Mugnaini C, Brizzi A, Purgatorio R, de Candia M, Catto M, Dreassi E, Manetti F, Corelli F, Altomare CD, Cappelli A, Paolino M. Light-mediated activation/deactivation control and in vitro ADME-Tox profiling of a donepezil-like Dual AChE/MAO-B Inhibitor. Eur J Pharm Sci 2025; 209:107066. [PMID: 40064401 DOI: 10.1016/j.ejps.2025.107066] [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: 01/07/2025] [Revised: 02/25/2025] [Accepted: 03/07/2025] [Indexed: 03/21/2025]
Abstract
The possibility to control the effects of drugs in time and space represents an ideal condition for developing safer and more personalized therapies against different disorders. In this context, photopharmacology has paved the way for the use of light in the modulation of drugs activity. Our interest is directed to photo-switchable molecules, capable of interconverting between two different isoforms upon light irradiation. We recently reported 1, a donepezil-like compound based on 2-benzylidenindan-1-one structure, as a dual AChE and MAO-B inhibitor, which can be converted into the E- (active form) and Z- (about tenfold less active form) diastereoisomers by irradiating with UV-vis light. Aiming at identifying compounds with remarkable activity in physiological conditions, we herein report a fine characterization of 1 in PBS solutions. First, we evaluated its ability to act as a photoswitch comparing PBS solution with organic solvents (e.g. methanol), demonstrating that a wavelength in the UV range (330 nm) can convert the E- into the Z-diastereoisomer, while the use of a visible light (400 nm) allows the interconversion from Z to E in both media. Along with its photoinducible behavior, we investigated the passive diffusion across cellular membrane with PAMPA experiments, plasma and microsomal stability, and binding to plasma proteins. Interestingly, the results of such studies suggested that 1 could persist in the blood circulation for a long time, which is desirable for application in photopharmacological therapies. Cytotoxicity studies highlighted the potential of our prototypic compound as a lead photodrug against neurodegenerative disorders, deserving to advance in molecular optimization studies and further in vitro and in vivo characterization.
Collapse
Affiliation(s)
- Federica Poggialini
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via A. Moro 2, I-53100, Siena, Italy
| | - Paolo Governa
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via A. Moro 2, I-53100, Siena, Italy
| | - Chiara Vagaggini
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via A. Moro 2, I-53100, Siena, Italy
| | - Samuele Maramai
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via A. Moro 2, I-53100, Siena, Italy
| | - Stefania Lamponi
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via A. Moro 2, I-53100, Siena, Italy
| | - Claudia Mugnaini
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via A. Moro 2, I-53100, Siena, Italy
| | - Antonella Brizzi
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via A. Moro 2, I-53100, Siena, Italy
| | - Rosa Purgatorio
- Department of Pharmacy-Pharmaceutical Sciences, University of Bari Aldo Moro, Via E. Orabona 4, 70125, Bari, Italy
| | - Modesto de Candia
- Department of Pharmacy-Pharmaceutical Sciences, University of Bari Aldo Moro, Via E. Orabona 4, 70125, Bari, Italy
| | - Marco Catto
- Department of Pharmacy-Pharmaceutical Sciences, University of Bari Aldo Moro, Via E. Orabona 4, 70125, Bari, Italy
| | - Elena Dreassi
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via A. Moro 2, I-53100, Siena, Italy
| | - Fabrizio Manetti
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via A. Moro 2, I-53100, Siena, Italy
| | - Federico Corelli
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via A. Moro 2, I-53100, Siena, Italy
| | - Cosimo Damiano Altomare
- Department of Pharmacy-Pharmaceutical Sciences, University of Bari Aldo Moro, Via E. Orabona 4, 70125, Bari, Italy
| | - Andrea Cappelli
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via A. Moro 2, I-53100, Siena, Italy
| | - Marco Paolino
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via A. Moro 2, I-53100, Siena, Italy.
| |
Collapse
|
121
|
Butman HS, Stefaniak MA, Walsh DJ, Gondil VS, Young M, Crow AH, Nemeth AM, Melander RJ, Dunman PM, Melander C. Phenyl urea based adjuvants for β-lactam antibiotics against methicillin resistant Staphylococcus aureus. Bioorg Med Chem Lett 2025; 121:130164. [PMID: 40043820 DOI: 10.1016/j.bmcl.2025.130164] [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: 11/20/2024] [Revised: 02/04/2025] [Accepted: 03/01/2025] [Indexed: 03/10/2025]
Abstract
Penicillin binding protein 4 (PBP4) is essential for Staphylococcus aureus cortical bone osteocyte lacuno-canalicular network (OLCN) invasion, which causes osteomyelitis and serves as a bacterial niche for recurring bone infection. Moreover, PBP4 is also a key determinant of S. aureus resistance to fifth-generation cephalosporins (ceftobiprole and ceftaroline). From these perspectives, the development of S. aureus PBP4 inhibitors may represent dual functional therapeutics that prevent osteomyelitis, and reverse PBP4-mediated β-lactam resistance. A high-throughput screen for small molecules that inhibit S. aureus PBP4 function identified compound 1. We recently described a preliminary structure activity relationship (SAR) study on 1, identifying several compounds with increased PBP4 inhibitory activity, some of which also inhibit PBP2a. Herein, we expand our exploration of phenyl ureas as antibiotic adjuvants, investigating their activity with penicillins and additional cephalosporins against PBP2a-mediated methicillin-resistant S. aureus (MRSA). We screened the previously reported pilot library, and prepared an additional series of phenyl ureas based on compound 1. Lead compounds potentiate multiple β-lactam antibiotics, lowering minimum inhibitory concentrations (MICs) below susceptibility breakpoints, with up to 64-fold reductions in MIC.
Collapse
Affiliation(s)
- Hailey S Butman
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN, 46556, United States
| | - Monica A Stefaniak
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN, 46556, United States
| | - Danica J Walsh
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN, 46556, United States
| | - Vijay S Gondil
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York 14642, United States
| | - Mikaeel Young
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York 14642, United States
| | - Andrew H Crow
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN, 46556, United States
| | - Ansley M Nemeth
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN, 46556, United States
| | - Roberta J Melander
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN, 46556, United States
| | - Paul M Dunman
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York 14642, United States.
| | - Christian Melander
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN, 46556, United States.
| |
Collapse
|
122
|
Alsowayeh N, Alfaiz FA, Ghabban H, Suresh K, Mickymaray S. Design and fabrication of melphalan (MPN) and vincristine (VCR) loaded carboxymethyl chitosan (CMC) nanoparticles for promising antibacterial and anticancer therapy in liver cancer cells. Int J Biol Macromol 2025; 316:144421. [PMID: 40403783 DOI: 10.1016/j.ijbiomac.2025.144421] [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: 02/13/2025] [Revised: 04/25/2025] [Accepted: 05/18/2025] [Indexed: 05/24/2025]
Abstract
Although the literature has shown the use of Vincristine (VCR) in conjunction with other chemotherapeutic agents for liver cancer, the efficiency of VCR combined with Melphalan (MPN). The clinical bioactivity and therapeutic efficacy of anticancer drugs are significantly constrained by inadequate targeting and adverse effects. NDDSs provide a superior approach to mitigate the limitations of small molecule anticancer agents. This investigation developed carboxymethyl chitosan (CMC), an effective aqueous-soluble chitosan (CS) derivative, by a chemical method and utilized it for the combination administration of Melphalan (MPN) and Vincristine (VCR). Carboxymethylation at the -OH and -NH2 groups of CS was verified by the FTIR. The CMC's ability to deliver drugs in a sustained and protracted way is demonstrated by the release profiles obtained. MPN + VCR@CMC NPs show excellent anticancer activity against HepG2 liver cancer cells at 14.9μg/ml and can induce 50 % cell death by generating oxidative stress. MPN + VCR@CMC NPs displays E. coli S. aureus and B. subtilis bacterial strains. Further, the apoptosis was confirmed by AO-EB and flow cytometry analysis. MPN + VCR@CMC NPs show excellent hemocompatibility. Overall, the outcomes revealed that MPN + VCR@CMC NPs for prolonged drug delivery and enhanced apoptotic efficacy as a chemotherapeutic agent.
Collapse
Affiliation(s)
- Noorah Alsowayeh
- Department of Biology, College of Science-Al Zulfi, Majmaah University, Majmaah 11952, Riyadh Region, Saudi Arabia.
| | - Faiz Abdulaziz Alfaiz
- Department of Biology, College of Science-Al Zulfi, Majmaah University, Majmaah 11952, Riyadh Region, Saudi Arabia
| | - Hanaa Ghabban
- Department of Biology, Faculty of Science, University of Tabuk, Tabuk 71491, Saudi Arabia; Biodiversity Genomics Unit, Faculty of Science, University of Tabuk, Tabuk 71491, Saudi Arabia
| | - Kaviya Suresh
- Department of Pharmaceutics, Sri Ramachandra Faculty of Pharmacy, Sri Ramachandra Institute of Higher Education and Research (DU), Porur, Chennai 600 116, India
| | - Suresh Mickymaray
- Department of Biology, College of Science-Al Zulfi, Majmaah University, Majmaah 11952, Riyadh Region, Saudi Arabia.
| |
Collapse
|
123
|
Bani Asadi F, Shirzaei F, Shaterian HR. Fe 3O 4@SiO 2@[Aminoglycol][Formate] as a new superparamagnetic nanocatalyst and [Aminoglycol][Formate] as a novel ionic liquid catalyst for preparation of new dimethyldihydropyrimido[4,5-b]quinolone derivatives. Mol Divers 2025; 29:2601-2616. [PMID: 39546219 DOI: 10.1007/s11030-024-11013-5] [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: 09/03/2024] [Accepted: 10/02/2024] [Indexed: 11/17/2024]
Abstract
Efficient synthesis of novel dimethyldihydropyrimido[4,5-b]quinolones via three-component condensation of barbituric acid, arylaldehydes, and 3,4-dimethylaniline catalyzed by Fe3O4@SiO2@[Aminoglycol][Formate] as a new superparamagnetic nanocatalyst and [Aminoglycol][Formate] as a novel ionic liquid catalyst was described. The new heterogeneous nanocatalyst was characterized by FE-SEM, XRD, FT-IR, TGA-DTG, and VSM techniques. The new ionic liquid was characterized by 13CNMR, 1HNMR, and FT-IR techniques. The present work has advantages, such as excellent yields, short reaction times, environmentally friendly protocol, easy separation, and purification of products. The catalysts kept its catalytic properties after even five recoverability and reusability.
Collapse
Affiliation(s)
- Fatemeh Bani Asadi
- Department of Chemistry, Faculty of Science, University of Sistan and Baluchestan, PO Box: 987-98155, Zahedan, 98197-45845, Iran
| | - Farhad Shirzaei
- Department of Chemistry, Faculty of Science, University of Sistan and Baluchestan, PO Box: 987-98155, Zahedan, 98197-45845, Iran
| | - Hamid Reza Shaterian
- Department of Chemistry, Faculty of Science, University of Sistan and Baluchestan, PO Box: 987-98155, Zahedan, 98197-45845, Iran.
| |
Collapse
|
124
|
Singh P, Singh VK, Gond C, Singh D, Tiwari AK. Current advances in the structure-activity relationship (SAR) analysis of the old/new 18-kDa translocator protein ligands. Mol Divers 2025; 29:2639-2689. [PMID: 39630364 DOI: 10.1007/s11030-024-10963-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 08/07/2024] [Indexed: 05/16/2025]
Abstract
The translocator protein 18 kDa (TSPO) is a crucial external mitochondrial protein involved in cholesterol translocation, which is essential for steroid production. As a primary marker of neuroinflammation, TSPO has been implicated in the development and progression of various neurodegenerative and neuropsychiatric disorders. This review highlights the structural diversity of TSPO ligands, many of which have undergone modifications from selective central benzodiazepine receptor (CBR) ligands to enhance their affinity for TSPO. The paper discusses the significant advancements in the design of these ligands, emphasizing their binding efficacy and specificity. Additionally, it provides an update on the progress of several TSPO ligands that have advanced to clinical trials. The review aims to elucidate the structure-activity relationships (SAR) that govern the interaction between TSPO and its ligands, thereby offering insights into the development of new therapeutic agents targeting TSPO for the treatment of neuroinflammatory conditions. Overall, this work provided an update on previous finding and serves as a valuable resource for researchers in the field.
Collapse
Affiliation(s)
- Priya Singh
- Department of Chemistry, Babasaheb Bhimrao Ambedkar University, Lucknow, Uttar Pradesh, 226025, India
| | - Vijay Kumar Singh
- Department of Chemistry, Babasaheb Bhimrao Ambedkar University, Lucknow, Uttar Pradesh, 226025, India
| | - Chandraprakash Gond
- Department of Chemistry, Babasaheb Bhimrao Ambedkar University, Lucknow, Uttar Pradesh, 226025, India
| | - Deepika Singh
- Department of Chemistry, Babasaheb Bhimrao Ambedkar University, Lucknow, Uttar Pradesh, 226025, India
| | - Anjani Kumar Tiwari
- Department of Chemistry, Babasaheb Bhimrao Ambedkar University, Lucknow, Uttar Pradesh, 226025, India.
| |
Collapse
|
125
|
Odagaki Y, Kinoshita M, Honda M, Meana JJ, Callado LF, García-Sevilla JA, Palkovits M, Borroto-Escuela DO, Fuxe K. Receptor-mediated G i-3 activation in mammalian and human brain membranes: Reestablishment method and its application to nociceptin/orphanin FQ opioid peptide (NOP) receptor/G i-3 interaction. J Pharmacol Sci 2025; 158:131-138. [PMID: 40288823 DOI: 10.1016/j.jphs.2025.03.014] [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: 02/15/2025] [Revised: 03/11/2025] [Accepted: 03/29/2025] [Indexed: 04/29/2025] Open
Abstract
Functional activation of heterotrimeric guanine nucleotide-binding proteins (G-proteins) via G-protein-coupled receptors (GPCRs) has been extensively explored using guanosine-5'-O-(3-[35S]thio)triphosphate ([35S]GTPγS) binding assay. However, the conventional method is primarily applicable to Gi/o family without discrimination among G-protein subtypes. Therefore, this study aims to reestablish a novel method termed "[35S]GTPγS binding/immunoprecipitation assay" by identifying a most suitable anti-Gαi-3 antibody instead of the previously utilized, now withdrawn antibody. In the initial screening of commercially available anti-Gαi-3 antibodies, two were identified and one was selected for further investigations based on efficacy with adenosine-the most potent agonist in our previous research. After optimizing experimental conditions with rat and postmortem human brain membranes, the stimulatory effects of various agonists were evaluated. Some agonists, including nociceptin, exhibited sufficient stimulatory effects for further pharmacological characterization. Nociceptin increased [35S]GTPγS binding to Gαi-3 in a concentration-dependent manner, response that was insensitive to naloxone but potently inhibited using (±)-J-113397. The method described in this study provides a valuable strategy for determining the intrinsic efficacy of ligands at various GPCRs. This includes nociceptin/orphanin FQ opioid peptide (NOP) receptor selectively coupled to Gαi-3, providing insights into the pharmacological concept of "functional selectivity."
Collapse
Affiliation(s)
- Yuji Odagaki
- Department of Psychiatry, Faculty of Medicine, Saitama Medical University, 38 Morohongo, Moroyama-machi, Iruma-gun, 350-0495, Saitama, Japan; Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan.
| | - Masakazu Kinoshita
- Department of Psychiatry, Faculty of Medicine, Saitama Medical University, 38 Morohongo, Moroyama-machi, Iruma-gun, 350-0495, Saitama, Japan
| | - Makoto Honda
- Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - J Javier Meana
- Department of Pharmacology, University of the Basque Country, UPV/EHU, Biobizkaia Health Research Institute, CIBERSAM, Bizkaia, Spain
| | - Luis F Callado
- Department of Pharmacology, University of the Basque Country, UPV/EHU, Biobizkaia Health Research Institute, CIBERSAM, Bizkaia, Spain
| | - Jesús A García-Sevilla
- Laboratory of Neuropharmacology, IUNICS/IdISPa, University of the Balearic Islands, Palma de Mallorca, Spain
| | - Miklós Palkovits
- Human Brain Tissue Bank, Semmelweis University, Budapest, Hungary
| | | | - Kjell Fuxe
- Department of Neuroscience, Biomedicum, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
126
|
Baral H, Kaundal RK. Novel insights into neuroinflammatory mechanisms in traumatic brain injury: Focus on pattern recognition receptors as therapeutic targets. Cytokine Growth Factor Rev 2025; 83:18-34. [PMID: 40169306 DOI: 10.1016/j.cytogfr.2025.03.001] [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: 12/11/2024] [Accepted: 03/14/2025] [Indexed: 04/03/2025]
Abstract
Traumatic brain injury (TBI) is a major global health concern and a leading cause of morbidity and mortality. Neuroinflammation is a pivotal driver of both the acute and chronic phases of TBI, with pattern recognition receptors (PRRs) playing a central role in detecting damage-associated molecular patterns (DAMPs) and initiating immune responses. Key PRR subclasses, including Toll-like receptors (TLRs), NOD-like receptors (NLRs), and cGAS-like receptors (cGLRs), are abundantly expressed in central nervous system (CNS) cells and infiltrating immune cells, where they mediate immune activation, amplify neuroinflammatory cascades, and exacerbate secondary injury mechanisms. This review provides a comprehensive analysis of these PRR subclasses, detailing their distinct structural characteristics, expression patterns, and roles in post-TBI immune responses. We critically examine the molecular mechanisms underlying PRR-mediated signaling and explore their contributions to neuroinflammatory pathways and secondary injury processes. Additionally, preclinical and clinical evidence supporting the therapeutic potential of targeting PRRs to mitigate neuroinflammation and improve neurological outcomes is discussed. By integrating recent advancements, this review offers an in-depth understanding of the role of PRRs in TBI pathobiology and underscores the potential of PRR-targeted therapies in mitigating TBI-associated neurological deficits.
Collapse
Affiliation(s)
- Harapriya Baral
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Raebareli (NIPER-R), Transit Campus, Bijnor-Sisendi Road, Sarojini Nagar, Near CRPF Base Camp, Lucknow, UP 226002, India
| | - Ravinder K Kaundal
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Raebareli (NIPER-R), Transit Campus, Bijnor-Sisendi Road, Sarojini Nagar, Near CRPF Base Camp, Lucknow, UP 226002, India.
| |
Collapse
|
127
|
Janthong A, Utama K, Khamto N, Chawapun P, Siriphong S, Van Doan H, Meerak J, Meepowpan P, Sangthong P. Semi-synthetic flavonoid derivatives from Boesenbergiarotunda induce extrinsic apoptosis pathway via Caspase-3 and Caspase-8 in HCT116 Colon Cancer cell lines. Bioorg Chem 2025; 159:108343. [PMID: 40096806 DOI: 10.1016/j.bioorg.2025.108343] [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: 11/19/2024] [Revised: 02/23/2025] [Accepted: 03/03/2025] [Indexed: 03/19/2025]
Abstract
Colorectal cancer ranks as the second most common cancer and the leading cause of cancer-related deaths globally. Phytochemicals like flavonoids from Boesenbergia rotunda showed potential anti-cancer activities. Chemical structures of the parental compounds of flavonoids were modified by conjugating with an acryloyl group to form semi-synthetic flavonoid derivatives to increasing in anti-colon cancer activities. 7-Acryloyloxypinocembrin (5) showed potential antiproliferative activities of IC50 value of 1.87 ± 0.17 μM in HCT116. In addition, compound 5 showed low cytotoxicity in Vero cells with an IC50 value of 2.84 ± 0.13 μM which is two-fold less cytotoxic than osimertinib. Biological mechanisms studies indicated that compound 5, HCT116 cells demonstrated a two-fold increase in apoptotic cell death. Subsequently, compound 5 upregulated caspase-8 and LC3, triggering the upregulation of caspase-3 and leading to the activation of both the extrinsic apoptosis pathway and the autophagy pathway. Network pharmacology analysis highlighted TNF-α receptor is a key gene associated with the extrinsic apoptosis pathway in HCT116 cells treated with compound 5. Molecular dynamics simulation confirmed the strong interaction between compound 5 and TACE, a crucial element in the EGFR and IL-6 signaling pathway's reduction which may lead to a decline in the survival rate of colon cancer. These findings indicate compound 5 as a promising anti-colon cancer drug candidate.
Collapse
Affiliation(s)
- Atchara Janthong
- Program in Biotechnology, Multidisciplinary and Interdisciplinary School, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Kraikrit Utama
- Office of Research Administration, Chiang Mai University, Chiang Mai 50200, Thailand; Department of Chemistry, Faculty of Science, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Nopawit Khamto
- Department of Chemistry, Faculty of Science, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Pornthip Chawapun
- Program in Biotechnology, Multidisciplinary and Interdisciplinary School, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Sandanon Siriphong
- Program in Biotechnology, Multidisciplinary and Interdisciplinary School, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Hien Van Doan
- Department of Animal and Aquatic Sciences, Faculty of Agriculture, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Jomkhwan Meerak
- Department of Biology, Faculty of Science, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Puttinan Meepowpan
- Department of Chemistry, Faculty of Science, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Padchanee Sangthong
- Research Laboratory on Advanced Materials for Sensor and Biosensor Innovation, Materials Science Research Center, and Center of Excellence for Innovation in Chemistry, Faculty of Science, Chiang Mai University, Chiang Mai, 50200, Thailand; Division of Biochemistry and Biochemical innovation, Department of Chemistry, Faculty of Science, Chiang Mai University, Chiang Mai 50200, Thailand
| |
Collapse
|
128
|
Haripriya E, Hemalatha K, Matada GSP, Pal R, Das PK, Ashadul Sk MD, Mounika S, Viji MP, Aayishamma I, Jayashree KR. Advancements of anticancer agents by targeting the Hippo signalling pathway: biological activity, selectivity, docking analysis, and structure-activity relationship. Mol Divers 2025; 29:2829-2862. [PMID: 39436581 DOI: 10.1007/s11030-024-11009-1] [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: 08/19/2024] [Accepted: 09/30/2024] [Indexed: 10/23/2024]
Abstract
The Hippo signalling pathway is prominent and governs cell proliferation and stem cell activity, acting as a growth regulator and tumour suppressor. Defects in Hippo signalling and hyperactivation of its downstream effector's Yes-associated protein (YAP) and transcriptional co-activator with PDZ-binding motif (TAZ) play roles in cancer development, implying that pharmacological inhibition of YAP and TAZ activity could be an effective cancer treatment strategy. Conversely, YAP and TAZ can also have beneficial effects in promoting tissue repair and regeneration following damage, therefore their activation may be therapeutically effective in certain instances. Recently, a complex network of intracellular and extracellular signalling mechanisms that affect YAP and TAZ activity has been uncovered. The YAP/TAZ-TEAD interaction leads to tumour development and the protein structure of YAP/TAZ-TEAD includes three interfaces and one hydrophobic pocket. There are clinical and preclinical trial drugs available to inhibit the hippo signalling pathway, but these drugs have moderate to severe side effects, so researchers are in search of novel, potent, and selective hippo signalling pathway inhibitors. In this review, we have discussed the hippo pathway in detail, including its structure, activation, and role in cancer. We have also provided the various inhibitors under clinical and preclinical trials, and advancement of small molecules their detailed docking analysis, structure-activity relationship, and biological activity. We anticipate that the current study will be a helpful resource for researchers.
Collapse
Affiliation(s)
- E Haripriya
- Integrated Drug Discovery Centre, Department of Pharmaceutical Chemistry, Acharya & BM Reddy College of Pharmacy, Bengaluru, 560107, Karnataka, India
| | - K Hemalatha
- Integrated Drug Discovery Centre, Department of Pharmaceutical Chemistry, Acharya & BM Reddy College of Pharmacy, Bengaluru, 560107, Karnataka, India.
| | - Gurubasavaraja Swamy Purawarga Matada
- Integrated Drug Discovery Centre, Department of Pharmaceutical Chemistry, Acharya & BM Reddy College of Pharmacy, Bengaluru, 560107, Karnataka, India
| | - Rohit Pal
- Integrated Drug Discovery Centre, Department of Pharmaceutical Chemistry, Acharya & BM Reddy College of Pharmacy, Bengaluru, 560107, Karnataka, India.
| | - Pronoy Kanti Das
- Integrated Drug Discovery Centre, Department of Pharmaceutical Chemistry, Acharya & BM Reddy College of Pharmacy, Bengaluru, 560107, Karnataka, India
| | - M D Ashadul Sk
- Integrated Drug Discovery Centre, Department of Pharmaceutical Chemistry, Acharya & BM Reddy College of Pharmacy, Bengaluru, 560107, Karnataka, India
| | - S Mounika
- Integrated Drug Discovery Centre, Department of Pharmaceutical Chemistry, Acharya & BM Reddy College of Pharmacy, Bengaluru, 560107, Karnataka, India
| | - M P Viji
- Integrated Drug Discovery Centre, Department of Pharmaceutical Chemistry, Acharya & BM Reddy College of Pharmacy, Bengaluru, 560107, Karnataka, India
| | - I Aayishamma
- Integrated Drug Discovery Centre, Department of Pharmaceutical Chemistry, Acharya & BM Reddy College of Pharmacy, Bengaluru, 560107, Karnataka, India
| | - K R Jayashree
- Integrated Drug Discovery Centre, Department of Pharmaceutical Chemistry, Acharya & BM Reddy College of Pharmacy, Bengaluru, 560107, Karnataka, India
| |
Collapse
|
129
|
Amberntsson S, Foster AJ, Chouhan B, Wilkinson S, Harlfinger S, Smith G, Kettle JG, Niedbala M, Kavanagh S, Williams DP. Use of new approach methodology for hepatic safety assessment of covalent inhibitor drug candidates. Toxicol Res (Camb) 2025; 14:tfaf054. [PMID: 40421426 PMCID: PMC12103896 DOI: 10.1093/toxres/tfaf054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 03/21/2025] [Accepted: 04/07/2025] [Indexed: 05/28/2025] Open
Abstract
Interest in inhibiting target proteins through covalent binding mechanisms has increased in the last decade due to the potential for beneficial pharmacological properties. However, the inherent targeted covalent inhibitor (TCI) adverse off-target reactivity risk requires a mitigation strategy early during drug discovery. The aim of this research was to design a pre-clinical hepatic safety assessment strategy for TCIs considering risk associated with electrophilic warhead reactivity and reactive metabolites formation at clinically-relevant plasma concentrations. The mitigation strategy was applied to compound 35, a potent irreversible inhibitor to KRASG12C. Drug induced liver injury was assessed in primary human hepatocyte spheroids. GSH and ATP depletion were investigated for compound 35 and 6 other marketed TCIs containing an acrylamide warhead which binds irreversibly to cysteine-containing target proteins. None of the TCIs showed GSH depletion prior to ATP depletion after 7-days exposure, suggesting that GSH depletion was not driving cytotoxicity in the spheroids. The calculated hepatotoxicity margin towards plasma exposure of 2.5 for compound 35 was found to be in the same range as for the two KRASG12Cinhibitors adagrasib and sotorasib, with clinically reported treatment-related adverse aminotransferase elevations leading to dose modifications. The safety evaluation reported here suggests no negative discrepancy in liver toxicity for compound 35 versus similar approved TCI's. Finally, the risk associated with detected oxidative metabolites was further mitigated as the pan-CYP450 inhibitor 1-aminobenzotriazole (ABT) had no effect on the cytotoxicity response following incubation of compound 35 in the presence and absence of ABT.
Collapse
Affiliation(s)
- Sara Amberntsson
- Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Pepparredsleden 1, SE-431 83 Mölndal, Sweden
| | - Alison J Foster
- Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, 1 Francis Crick Avenue, Cambridge CB2 0AA, United Kingdom
| | - Bhavik Chouhan
- Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Pepparredsleden 1, SE-431 83 Mölndal, Sweden
| | - Stephen Wilkinson
- DMPK, Oncology, R&D, AstraZeneca, 1 Francis Crick Avenue, Cambridge CB2 0AA, United Kingdom
| | - Stephanie Harlfinger
- DMPK, Oncology, R&D, AstraZeneca, 1 Francis Crick Avenue, Cambridge CB2 0AA, United Kingdom
| | - Graham Smith
- Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, 1 Francis Crick Avenue, Cambridge CB2 0AA, United Kingdom
| | - Jason G Kettle
- Oncology R&D, AstraZeneca, 1 Francis Crick Avenue, Cambridge CB2 0AA, United Kingdom
| | - Michael Niedbala
- Oncology R&D, AstraZeneca, 35 Gatehouse Dr, Waltham, MA 02145 US
| | - Stefan Kavanagh
- Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, 1 Francis Crick Avenue, Cambridge CB2 0AA, United Kingdom
| | - Dominic P Williams
- Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, 1 Francis Crick Avenue, Cambridge CB2 0AA, United Kingdom
| |
Collapse
|
130
|
Muteeb G, Kazi RNA, Aatif M, Azhar A, Oirdi ME, Farhan M. Antimicrobial resistance: Linking molecular mechanisms to public health impact. SLAS DISCOVERY : ADVANCING LIFE SCIENCES R & D 2025; 33:100232. [PMID: 40216324 DOI: 10.1016/j.slasd.2025.100232] [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: 02/15/2025] [Revised: 03/20/2025] [Accepted: 04/08/2025] [Indexed: 04/21/2025]
Abstract
BACKGROUND Antimicrobial resistance (AMR) develops into a worldwide health emergency through genetic and biochemical adaptations which enable microorganisms to resist antimicrobial treatment. β-lactamases (blaNDM, blaKPC) and efflux pumps (MexAB-OprM) working with mobile genetic elements facilitate fast proliferation of multidrug-resistant (MDR) and exttreme drug-resistant (XDR) phenotypes thus creating major concerns for healthcare systems and community health as well as the agricultural sector. OBJECTIVES The review dissimilarly unifies molecular resistance pathways with public health implications through the study of epidemiological data and monitoring approaches and innovative therapeutic solutions. Previous studies separating their attention between molecular genetics and clinical outcomes have been combined into our approach which delivers an all-encompassing analysis of AMR. KEY INSIGHTS The report investigates the resistance mechanisms which feature enzymatic degradation and efflux pump overexpression together with target modification and horizontal gene transfer because these factors represent important contributors to present-day AMR developments. This review investigates AMR effects on hospital and community environments where it affects pathogens including MRSA, carbapenem-resistant Klebsiella pneumoniae, and drug-resistant Pseudomonas aeruginosa. This document explores modern AMR management methods that comprise WHO GLASS molecular surveillance systems and three innovative strategies such as CRISPR-modified genome editing and bacteriophage treatments along with antimicrobial peptides and artificial intelligence diagnostic tools. CONCLUSION The resolution of AMR needs complete scientific and global operational methods alongside state-of-the-art therapeutic approaches. Worldwide management of drug-resistant infection burden requires both enhanced infection prevention procedures with next-generation antimicrobial strategies to reduce cases effectively.
Collapse
Affiliation(s)
- Ghazala Muteeb
- Department of Nursing, College of Applied Medical Science, King Faisal University, Al-Ahsa, Saudi Arabia.
| | - Raisa Nazir Ahmed Kazi
- Department of Respiratory Therapy, College of Applied Medical Science, King Faisal, University, Al-Ahsa, Saudi Arabia
| | - Mohammad Aatif
- Department of Public Health, College of Applied Medical Science, King Faisal University, Al-Ahsa, Saudi Arabia
| | - Asim Azhar
- NAP Life Sciences; Metropolitan Region, Maharashtra 401208, India
| | - Mohamed El Oirdi
- Department of Biological Sciences, College of Science, King Faisal University, Al Ahsa, Saudi Arabia; Department of Basic Sciences, Preparatory Year, King Faisal University, Al-Ahsa, Saudi Arabia
| | - Mohd Farhan
- Department of Basic Sciences, Preparatory Year, King Faisal University, Al-Ahsa, Saudi Arabia; Department of Chemistry, College of Science, King Faisal University, Al Ahsa, Saudi Arabia.
| |
Collapse
|
131
|
Li X, Li J, Ji Z, Li C, Wu T, Song H, Yang X. Magnetic Nanoparticles Combined With Pulsed Electromagnetic Field Alleviate Chondrocyte Necroptosis in Osteoarthritis. FASEB J 2025; 39:e70652. [PMID: 40366222 PMCID: PMC12077385 DOI: 10.1096/fj.202401959rr] [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: 08/20/2024] [Revised: 04/11/2025] [Accepted: 05/07/2025] [Indexed: 05/15/2025]
Abstract
Chondrocyte necroptosis contributes to the pathogenesis of osteoarthritis (OA). Pulsed electromagnetic field (PEMF) is a potentially useful treatment for OA. Here, magnetic nanoparticles and PEMF generate magneto-mechanical forces for regulating signaling pathways, but their effectiveness remains unclear. This study investigated whether magnetic nanoparticles (MIL-101(Fe)) combined with PEMF alleviate chondrocyte necroptosis in OA. Destabilization of the medial meniscus (DMM) surgery was performed to induce OA in 10-week-old wild-type mice. MIL-101(Fe) and PEMF were applied in human OA chondrocytes and experimental OA mice. Characterization and biocompatibility of MIL-101(Fe) were examined. Chondrocyte necroptosis was analyzed by western blotting, immunofluorescence, TUNEL, and transmission electron microscopy. OA severity was assessed by RT-PCR, immunofluorescence, histology, and micro-CT. We found that MIL-101(Fe) had no obvious cytotoxicity and presented biocompatibility. The combination of MIL-101(Fe) and PEMF ameliorated cartilage metabolism. PEMF attenuated cartilage degeneration and trabecular bone microarchitecture; these protective effects were enhanced by MIL-101(Fe). Further, the combination therapy markedly inhibited chondrocyte necroptosis and significantly decreased phosphorylation of RIP1, RIP3, and MLKL. Together, our findings indicate that MIL-101(Fe) combined with PEMF synergistically ameliorates chondrocyte necroptosis and OA progression without severe side effects, suggesting that this combination therapy may offer a novel strategy for treating OA.
Collapse
Affiliation(s)
- Xuqing Li
- Department of Rehabilitation MedicineSir Run Run Shaw Hospital, Zhejiang University School of MedicineHangzhouChina
| | - Jianhua Li
- Department of Rehabilitation MedicineSir Run Run Shaw Hospital, Zhejiang University School of MedicineHangzhouChina
| | - Zhongyin Ji
- Department of Orthopaedic SurgerySir Run Run Shaw Hospital, Zhejiang University School of MedicineHangzhouChina
| | - Changsheng Li
- Department of Rehabilitation MedicineSir Run Run Shaw Hospital, Zhejiang University School of MedicineHangzhouChina
| | - Tao Wu
- Department of Rehabilitation MedicineSir Run Run Shaw Hospital, Zhejiang University School of MedicineHangzhouChina
| | - Haixin Song
- Department of Rehabilitation MedicineSir Run Run Shaw Hospital, Zhejiang University School of MedicineHangzhouChina
| | - Xiaotian Yang
- Department of Rehabilitation MedicineSir Run Run Shaw Hospital, Zhejiang University School of MedicineHangzhouChina
| |
Collapse
|
132
|
Li Y, Ladd Z, Xiong Z, Bui-Linh C, Paiboonrungruang C, Subramaniyan B, Li H, Wang H, Balch C, Shersher DD, Spitz F, Chen X. Lymphatic Metastasis of Esophageal Squamous Cell Carcinoma: The Role of NRF2 and Therapeutic Strategies. Cancers (Basel) 2025; 17:1853. [PMID: 40507333 PMCID: PMC12153707 DOI: 10.3390/cancers17111853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2025] [Revised: 05/26/2025] [Accepted: 05/28/2025] [Indexed: 06/16/2025] Open
Abstract
The lethality of esophageal squamous cell carcinoma (ESCC), and other epithelial cancers, is primarily due to its aggressive nature and frequent lymphatic metastasis, both of which impact prognosis. In this review, we explore the underlying molecular mechanisms of ESCC lymphatic metastasis, specifically, the functional role of NRF2 and therapeutic strategies. Current data suggest that NRF2 hyperactivation (NRF2high) may promote lymphatic metastasis of ESCC by affecting the extracellular matrix (ECM), epithelial-mesenchymal transition (EMT), lymphangiogenesis, immune evasion, metabolic programming, and Hippo signaling. We also update the latest developments in NRF2 inhibitors, their mechanisms of action, screening strategies, and approaches for evaluating compound efficacy. Finally, we highlight the utility of animal models for mechanistic studies and therapeutic development. We believe elucidation of the functional role of NRF2 in ESCC lymphatic metastasis and developing proper NRF2 inhibitors will greatly improve the clinical prognosis of ESCC in human patients.
Collapse
Affiliation(s)
- Yahui Li
- Surgical Research Lab, Department of Surgery, Cooper University Health Care, Camden, NJ 08103, USA; (Y.L.); (Z.L.); (B.S.); (H.L.); (D.D.S.); (F.S.)
| | - Zachary Ladd
- Surgical Research Lab, Department of Surgery, Cooper University Health Care, Camden, NJ 08103, USA; (Y.L.); (Z.L.); (B.S.); (H.L.); (D.D.S.); (F.S.)
- Cooper Medical School of Rowan University, Camden, NJ 08103, USA
| | - Zhaohui Xiong
- Coriell Institute for Medical Research, Camden, NJ 08103, USA; (Z.X.); (C.B.-L.); (C.P.); (C.B.)
| | - Candice Bui-Linh
- Coriell Institute for Medical Research, Camden, NJ 08103, USA; (Z.X.); (C.B.-L.); (C.P.); (C.B.)
| | | | - Boopathi Subramaniyan
- Surgical Research Lab, Department of Surgery, Cooper University Health Care, Camden, NJ 08103, USA; (Y.L.); (Z.L.); (B.S.); (H.L.); (D.D.S.); (F.S.)
| | - Huan Li
- Surgical Research Lab, Department of Surgery, Cooper University Health Care, Camden, NJ 08103, USA; (Y.L.); (Z.L.); (B.S.); (H.L.); (D.D.S.); (F.S.)
| | - Haining Wang
- Insilico Medicine Canada Inc., Montreal, QC H3B 4W8, Canada;
| | - Curt Balch
- Coriell Institute for Medical Research, Camden, NJ 08103, USA; (Z.X.); (C.B.-L.); (C.P.); (C.B.)
| | - David D. Shersher
- Surgical Research Lab, Department of Surgery, Cooper University Health Care, Camden, NJ 08103, USA; (Y.L.); (Z.L.); (B.S.); (H.L.); (D.D.S.); (F.S.)
- Cooper Medical School of Rowan University, Camden, NJ 08103, USA
- MD Anderson Cancer Center at Cooper, Camden, NJ 08103, USA
| | - Francis Spitz
- Surgical Research Lab, Department of Surgery, Cooper University Health Care, Camden, NJ 08103, USA; (Y.L.); (Z.L.); (B.S.); (H.L.); (D.D.S.); (F.S.)
- Cooper Medical School of Rowan University, Camden, NJ 08103, USA
- MD Anderson Cancer Center at Cooper, Camden, NJ 08103, USA
| | - Xiaoxin Chen
- Surgical Research Lab, Department of Surgery, Cooper University Health Care, Camden, NJ 08103, USA; (Y.L.); (Z.L.); (B.S.); (H.L.); (D.D.S.); (F.S.)
- Cooper Medical School of Rowan University, Camden, NJ 08103, USA
- Coriell Institute for Medical Research, Camden, NJ 08103, USA; (Z.X.); (C.B.-L.); (C.P.); (C.B.)
- MD Anderson Cancer Center at Cooper, Camden, NJ 08103, USA
| |
Collapse
|
133
|
Siva S, Han SS. Fabrication of tolmetin/cyclodextrin nanofibers for potential wound healing applications: Physicochemical, in vitro characterization, and scratch assay. Int J Pharm 2025; 677:125644. [PMID: 40288507 DOI: 10.1016/j.ijpharm.2025.125644] [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: 02/24/2025] [Revised: 04/23/2025] [Accepted: 04/24/2025] [Indexed: 04/29/2025]
Abstract
The biocompatibility and sustained delivery of bioactive materials are crucial for maximum therapeutic benefits in the medical field. In this study, high-porous biocompatible nanofibers (NFs) were fabricated from inclusion complexes (ICs) of tolmetin (TMN) with 2-hydroxypropyl and methyl-β-cyclodextrin (CD) using an electrospinning method for sustained drug delivery at the wound site. The TMN-CD-IC-NFs were uniform, bead-free fibers with average diameters of 350 ± 55 nm and 225 ± 40 nm, exhibiting improved thermal properties (degradation temperatures of 317.0 °C and 329.2 °C) compared with pristine TMN. X-ray diffraction analysis revealed an amorphous distribution of TMN within the TMN-CD-IC-NFs. Fourier transform infrared and 1H nuclear magnetic resonance spectral analyses confirmed that the phenyl ring of TMN was deeply penetrated and strongly interacted with the atoms inside the CD cavities. Sustained TMN release was observed from TMN-CD-IC-NFs compared to the pure drug. The TMN-CD-IC-NFs had no adverse effects on skin fibroblast cells (NIH/3T3), demonstrating good biocompatibility. Molecular docking analysis demonstrated strong interactions between inflammation-responsible enzyme 6-COX and TMN-CD-ICs. Cultured cells treated with TMN-CD-IC-NFs were well proliferated towards the scratched area and the wound closure rate was higher (80-90%) compared to the control. Thus, fabricated TMN-CD-IC-NFs have potential as sustained drug nanocarriers for wound treatment.
Collapse
Affiliation(s)
- Subramanian Siva
- School of Chemical Engineering, Yeungnam University, 280 Daehak-Ro, Gyeongsan-si, Gyeongsangbuk-do 38541, Republic of Korea.
| | - Sung Soo Han
- School of Chemical Engineering, Yeungnam University, 280 Daehak-Ro, Gyeongsan-si, Gyeongsangbuk-do 38541, Republic of Korea; Research Institute of Cell Culture, Yeungnam University, 280 Daehak-Ro, Gyeongsan-si, Gyeongsangbuk-do 38541, Republic of Korea.
| |
Collapse
|
134
|
Wdowiak M, Raza S, Grotek M, Zbonikowski R, Nowakowska J, Doligalska M, Cai N, Luo Z, Paczesny J. Phage/nanoparticle cocktails for a biocompatible and environmentally friendly antibacterial therapy. Appl Microbiol Biotechnol 2025; 109:129. [PMID: 40442509 PMCID: PMC12122614 DOI: 10.1007/s00253-025-13526-x] [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: 03/14/2025] [Revised: 05/11/2025] [Accepted: 05/17/2025] [Indexed: 06/02/2025]
Abstract
Antibiotic resistance continues to rise, necessitating alternative strategies. Bacteriophages have emerged as promising natural antibacterial agents, offering a targeted approach to combating bacterial infections. Combining bacteriophages with nanoparticles presents a novel approach that could enhance antibacterial potency while reducing the risk of resistance. While phage/antibiotic cocktails are widely explored to enhance antibacterial efficacy and prevent resistance, research on phage/nanoparticle combinations remains limited. We explore the synergy between green tea extract-capped silver nanoparticles (G-TeaNPs) and bacteriophages in combating pathogenic bacteria (methicillin-resistant Staphylococcus aureus, Salmonella enterica). G-TeaNPs show minimal antiphage activity, ensuring compatibility in phage-NP formulations. These combinations significantly reduce bacterial counts in a short time (only 3 h), e.g., S. aureus survival is around 30% after incubation with just 0.001 mg/mL of G-TeaNPs, while G-TeaNPs and phages alone result in around 80% and 70% survival, respectively. Cytotoxicity tests against eukaryotic 3T3 NIH fibroblast cells confirm biocompatibility at effective concentrations. Additionally, we examine G-TeaNPs' impact on the free-living protist Acanthamoeba castellanii. Both green tea extract and G-TeaNPs can reduce A. castellanii cell counts by 80%, but only at high concentrations. Microscopy revealed nanoparticle uptake by amoebae, causing intracellular accumulation and vacuolization, while green tea extract induced similar changes without uptake. Our findings highlight G-TeaNPs as safe, effective agents in phage/nanoparticle antibacterial formulations with dual antimicrobial and amoebicidal properties for therapeutic and environmental applications. KEYPOINTS: • Silver nanoparticles synthesized with tea extracts (G-TeaNPs) have a minimal effect on the tested viruses. • Combining G-TeaNP with bacteriophages offers new-generation antibacterial cocktails. • Green tea extracts and AgNPs present concentration-dependent anti-amoebic activity.
Collapse
Affiliation(s)
- Mateusz Wdowiak
- Institute of Physical Chemistry, Polish Academy of Sciences, Marcina Kasprzaka 44/52, 01-224, Warsaw, Poland
- Centre of New Technologies, University of Warsaw, Stefana Banacha 2 C, 02-097, Warsaw, Poland
| | - Sada Raza
- Institute of Physical Chemistry, Polish Academy of Sciences, Marcina Kasprzaka 44/52, 01-224, Warsaw, Poland.
| | - Mateusz Grotek
- Institute of Physical Chemistry, Polish Academy of Sciences, Marcina Kasprzaka 44/52, 01-224, Warsaw, Poland
- Military University of Technology, Gen. Sylwestra Kaliskiego 2, 00-908, Warsaw, Poland
| | - Rafał Zbonikowski
- Institute of Physical Chemistry, Polish Academy of Sciences, Marcina Kasprzaka 44/52, 01-224, Warsaw, Poland
| | - Julita Nowakowska
- Faculty of Biology, University of Warsaw, Ilii Miecznikowa 1, 02-096, Warsaw, Poland
| | - Maria Doligalska
- Faculty of Biology, University of Warsaw, Ilii Miecznikowa 1, 02-096, Warsaw, Poland.
| | - Ningjing Cai
- Laboratory of Bioinspired Medicine and Materials, Southern University of Science and Technology, 1088 Xueyuan Avenue, Shenzhen, 518055, People's Republic of China
| | - Zhi Luo
- Laboratory of Bioinspired Medicine and Materials, Southern University of Science and Technology, 1088 Xueyuan Avenue, Shenzhen, 518055, People's Republic of China
| | - Jan Paczesny
- Institute of Physical Chemistry, Polish Academy of Sciences, Marcina Kasprzaka 44/52, 01-224, Warsaw, Poland.
| |
Collapse
|
135
|
Bae HE, Jung J, Sung JS, Kwon S, Kang MJ, Jose J, Pyun JC. Penicillin-like mimotopes from autodisplayed Fv-antibody library inhibiting β-lactamase activity. J Mater Chem B 2025; 13:6154-6163. [PMID: 40326485 DOI: 10.1039/d4tb02793k] [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: 05/07/2025]
Abstract
A penicillin-like mimotope was screened from an Fv-antibody library which had the inhibition activity of β-lactamase. Fv-antibody indicated the variable region (VH) of the immunoglobulin G, which includes three complementarity determining regions (CDRs). The Fv-antibody library was then prepared by randomizing the complementarity determining region 3 (CDR3), and it was expressed on the outer membrane of E. coli. The penicillin-like mimotopes were screened from the Fv-antibody library using magnetic beads with an immobilized monoclonal anti-penicillin antibody. The screened mimotopes were expressed as soluble Fv-antibodies and were also synthesized into peptides (11-mer). The binding affinity (KD) of the expressed Fv-antibodies and synthesized peptides was estimated using SPR measurements. The β-lactamase inhibition activity of the Fv-antibodies and synthetic peptides was estimated using colorimetry based on the formation of penicilloic acid. The penicillin-like mimotopes of the expressed Fv-antibodies and synthesized peptides were demonstrated to have β-lactamase inhibition activity in the bacterial lysates. Finally, the docking analysis of β-lactamase and the screened CDR3 sequences demonstrated that the screened CDR3 sequences were specifically bound to the binding sites of β-lactamase.
Collapse
Affiliation(s)
- Hyung Eun Bae
- Department of Materials Science and Engineering, Yonsei University, 50 Yonsei-Ro, Seodaemun-Gu, Seoul, 03722, Korea.
| | - Jaeyong Jung
- Department of Materials Science and Engineering, Yonsei University, 50 Yonsei-Ro, Seodaemun-Gu, Seoul, 03722, Korea.
| | - Jeong Soo Sung
- Department of Materials Science and Engineering, Yonsei University, 50 Yonsei-Ro, Seodaemun-Gu, Seoul, 03722, Korea.
| | - Soonil Kwon
- Department of Materials Science and Engineering, Yonsei University, 50 Yonsei-Ro, Seodaemun-Gu, Seoul, 03722, Korea.
| | - Min-Jung Kang
- Korea Institute of Science and Technology (KIST), Seoul, 02792, Korea
| | - Joachim Jose
- Institute of Pharmaceutical and Medical Chemistry, Westfälischen Wilhelms-Universität Münster, Muenster, 48149, Germany
| | - Jae-Chul Pyun
- Department of Materials Science and Engineering, Yonsei University, 50 Yonsei-Ro, Seodaemun-Gu, Seoul, 03722, Korea.
| |
Collapse
|
136
|
Swinand G, Rowe M, Bowen K, Olatunji S, Caffrey M, Scanlan EM. Late-stage lipidation of peptides via aqueous thiol-michael addition to dehydroalanine (Dha). Chem Commun (Camb) 2025; 61:8083-8086. [PMID: 40331307 DOI: 10.1039/d5cc01085c] [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: 05/08/2025]
Abstract
This work presents a late-stage aqueous peptide lipidation strategy via the thiol-Michael addition of thiolated lipids at dehydroalanine (Dha). This strategy was used to synthesise lipopeptides containing diacylglycerol (DAG), saturated, unsaturated and cholesterol lipid motifs. The DAG lipopeptide product was found to be a substrate for the lipoprotein processing enzyme, LspA.
Collapse
Affiliation(s)
- Glenna Swinand
- School of Chemistry, Trinity College Dublin, Trinity Biomedical Sciences Institute, Pearse St, Dublin 2, Ireland.
| | - Matthew Rowe
- School of Chemistry, Trinity College Dublin, Trinity Biomedical Sciences Institute, Pearse St, Dublin 2, Ireland.
| | - Katherine Bowen
- School of Chemistry, Trinity College Dublin, Trinity Biomedical Sciences Institute, Pearse St, Dublin 2, Ireland.
| | - Samir Olatunji
- School of Biochemistry and Immunology, Trinity College Dublin, Trinity Biomedical Sciences Institute, Pearse St, Dublin 2, Ireland
| | - Martin Caffrey
- School of Biochemistry and Immunology, Trinity College Dublin, Trinity Biomedical Sciences Institute, Pearse St, Dublin 2, Ireland
| | - Eoin M Scanlan
- School of Chemistry, Trinity College Dublin, Trinity Biomedical Sciences Institute, Pearse St, Dublin 2, Ireland.
| |
Collapse
|
137
|
Nakadate K, Ito N, Kawakami K, Yamazaki N. Anti-Inflammatory Actions of Plant-Derived Compounds and Prevention of Chronic Diseases: From Molecular Mechanisms to Applications. Int J Mol Sci 2025; 26:5206. [PMID: 40508016 PMCID: PMC12154257 DOI: 10.3390/ijms26115206] [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: 02/28/2025] [Revised: 05/23/2025] [Accepted: 05/27/2025] [Indexed: 06/16/2025] Open
Abstract
Chronic inflammation is a key contributor to the development and progression of numerous chronic diseases, including cardiovascular diseases, type 2 diabetes, neurodegenerative disorders, cancer, and obesity. As the side effects of conventional anti-inflammatory drugs pose challenges, plant-derived compounds have emerged as promising alternatives due to their potent anti-inflammatory properties and minimal adverse effects. This review explores the molecular mechanisms by which these compounds alleviate chronic inflammation and highlights their potential role in disease prevention. Polyphenols (e.g., quercetin and resveratrol), flavonoids (e.g., luteolin and apigenin), carotenoids (e.g., β-carotene and lycopene), and other phytochemicals (e.g., curcumin and gingerol) modulate inflammatory pathways, such as nuclear factor-κB and mitogen-activated protein kinase, reduce oxidative stress, and inhibit pro-inflammatory cytokines. Plant-derived compounds interact with the gut microbiota, enhancing anti-inflammatory effects. Evidence from animal studies and clinical trials has demonstrated their efficacy in reducing inflammation-related biomarkers and improving health outcomes. However, challenges such as low bioavailability and determination of the optimal dosage require further investigation. Advancing delivery technologies and personalized nutrition strategies may help overcome these barriers. This review emphasizes the therapeutic potential of plant-derived compounds in preventing chronic diseases and underscores the need for continued research to translate these findings into practical applications for public health.
Collapse
Affiliation(s)
- Kazuhiko Nakadate
- Department of Functional Morphology, Meiji Pharmaceutical University, 2-522-1, Noshio, Kiyose 204-8588, Tokyo, Japan; (N.I.); (K.K.)
| | - Nozomi Ito
- Department of Functional Morphology, Meiji Pharmaceutical University, 2-522-1, Noshio, Kiyose 204-8588, Tokyo, Japan; (N.I.); (K.K.)
| | - Kiyoharu Kawakami
- Department of Functional Morphology, Meiji Pharmaceutical University, 2-522-1, Noshio, Kiyose 204-8588, Tokyo, Japan; (N.I.); (K.K.)
| | - Noriko Yamazaki
- Department of Community Health Care and Sciences, Meiji Pharmaceutical University, 2-522-1, Noshio, Kiyose 204-8588, Tokyo, Japan;
| |
Collapse
|
138
|
Rana N, Chaudhary PK, Prasad R, Sankar M. Photodynamic Evaluation of A 2BC Aminoporphyrins: Synthesis, Characterization, and Cellular Impact. ACS APPLIED BIO MATERIALS 2025. [PMID: 40421616 DOI: 10.1021/acsabm.5c00411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/28/2025]
Abstract
This study focuses on the design and evaluation of a series of A2BC aminoporphyrins, featuring electron-donating substituents like pyrene, carbazole, and phenothiazine to enhance their photophysical and biological performance. Detailed characterization through spectroscopic methods, single-crystal X-ray diffraction, and computational analyses revealed insights into their electronic structure and planarity. Photophysical investigations revealed characteristic Soret and Q bands, along with tunable fluorescence and excited-state lifetimes influenced by the meso substituents. Biological evaluation was conducted using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assays and fluorescence microscopy to assess the photodynamic therapeutic efficacy against T24 bladder cancer cells. The porphyrins exhibited pronounced photocytotoxicity upon 660 nm light activation, attributed to reactive oxygen species (ROS) generation. Cellular analysis, including acridine orange/ethidium bromide and 4',6-diamidino-2-phenylindole staining, confirmed apoptosis induction through chromatin condensation and nuclear fragmentation. The findings highlight the potential of these porphyrins as effective photosensitizers for photodynamic therapy, demonstrating enhanced stability and ROS generation efficiency.
Collapse
Affiliation(s)
- Nivedita Rana
- Department of Chemistry, Indian Institute of Technology Roorkee, Roorkee 247667, India
| | - Pankaj Kumar Chaudhary
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee 247667, India
| | - Ramasare Prasad
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee 247667, India
| | - Muniappan Sankar
- Department of Chemistry, Indian Institute of Technology Roorkee, Roorkee 247667, India
| |
Collapse
|
139
|
Yang Z, Peng Y, Qu M, Yang H, Jia X, Liu X. Rational design of near-infrared carbon dots as polarity-sensitive fluorescent probes for imaging of lipid droplets. J Mater Chem B 2025. [PMID: 40420739 DOI: 10.1039/d5tb00521c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/28/2025]
Abstract
Polarity plays important roles in establishing and reflecting numerous complex physiological functions and pathological effects associated with energy metabolism and cell signaling. Monitoring the variations in polarity, particularly the polarity of lipid droplets (LDs), is of great significance in biomedical research and clinical diagnosis. Herein, a novel near-infrared (NIR) carbon dots (CDs)-based fluorescent nanoprobe is presented to serve the stringent requirements of polarity targeting and imaging with high sensitivity, superior photostability, excellent permeability and biocompatibility. The absorption and emission wavelength shifts were in response to an increased ambient polarity (Δf), in which the emission reached to the NIR region near 800 nm with the maximum emission wavelength located around 700 nm. This nanoprobe can specifically colocalize with LDs with a high correlation coefficient of 0.96 and effectively image the polarity changes in LDs and living cells. This work presents effective strategies and foundations for the construction of NIR CDs, helps in the design of polarity selective probes, and has implications for accelerating the development of polarity-related processes for disease diagnosis.
Collapse
Affiliation(s)
- Zheng Yang
- College of Chemistry and Chemical Engineering, Xi'an University of Science and Technology, Xi'an 710054, P. R. China.
- Key Laboratory of Coal Resources Exploration and Comprehensive Utilization, Ministry of Land and Resources, Xi'an 710012, P. R. China
| | - Ying Peng
- College of Chemistry and Chemical Engineering, Xi'an University of Science and Technology, Xi'an 710054, P. R. China.
| | - Mengyao Qu
- College of Chemistry and Chemical Engineering, Xi'an University of Science and Technology, Xi'an 710054, P. R. China.
| | - Haonan Yang
- College of Chemistry and Chemical Engineering, Xi'an University of Science and Technology, Xi'an 710054, P. R. China.
| | - Xiaodan Jia
- College of Chemistry and Chemical Engineering, Xi'an University of Science and Technology, Xi'an 710054, P. R. China.
- Key Laboratory of Coal Resources Exploration and Comprehensive Utilization, Ministry of Land and Resources, Xi'an 710012, P. R. China
| | - Xiangrong Liu
- College of Chemistry and Chemical Engineering, Xi'an University of Science and Technology, Xi'an 710054, P. R. China.
- Key Laboratory of Coal Resources Exploration and Comprehensive Utilization, Ministry of Land and Resources, Xi'an 710012, P. R. China
| |
Collapse
|
140
|
Srikanth D, Shejul GD, Joshi SV, Kalam A, Jahnavi AS, Dikundwar AG, Chopra S, Yaddanapudi MV, Nanduri S. Transition metal-free one-pot tandem chemoselective reduction and cyclization of 3/5-(2-nitrophenyl)-1 H-pyrazoles using sodium dithionite. Org Biomol Chem 2025. [PMID: 40424067 DOI: 10.1039/d5ob00610d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/29/2025]
Abstract
A sodium dithionite (Na2S2O4) mediated tandem chemoselective reductive cyclization of 5-(2-nitrophenyl)-1H-pyrazoles with aldehydes/carbon disulfide is developed for the synthesis of pyrazolo[1,5-c]quinazolines. The protocol involves one pot in situ reduction of 5-(2-nitrophenyl)-1H-pyrazoles, followed by intermolecular cyclization with aldehydes or carbon disulfide to afford the pyrazolo[1,5-c]quinazolines. The protocol is further expanded for the synthesis of pyrazolo[4,3-c]quinolines from 3-(2-nitrophenyl)-1-phenyl-1H-pyrazole-4-carbaldehyde via one pot in situ reduction followed by intramolecular cyclization. Notably, the use of Na2S2O4 as the reducing agent enables a metal, ligand, and additive-free approach, wide substrate scope, and scalability up to gram scale. The synthesized compounds were studied for photophysical properties in ACN and MeOH, the compound 7c, which contains an electron-donating methoxy group, exhibited a greater bathochromic shift. Notably, the pyrazolo[1,5-c]quinazoline-5(6H)-thione derivatives 5a and 5d demonstrated selective inhibition of Staphylococcus aureus, with minimum inhibitory concentrations (MICs) of 2 and 4 μg mL-1, respectively. ESI-MS and DFT studies were conducted for the identification of the key intermediates and to elucidate the plausible reaction mechanism.
Collapse
Affiliation(s)
- Danaboina Srikanth
- Department of Chemical Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500 037, India.
| | - Gayatri Diliprao Shejul
- Department of Chemical Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500 037, India.
| | - Swanand Vinayak Joshi
- Department of Chemical Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500 037, India.
| | - Abdul Kalam
- Department of Chemical Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500 037, India.
| | - Avvaru Subha Jahnavi
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500 037, India
| | - Amol G Dikundwar
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500 037, India
| | - Sidharth Chopra
- Division of Molecular Microbiology and Immunology, CSIR-Central Drug Research Institute, Sitapur Road, Sector 10, Janakipuram Extension, Lucknow-226031, Uttar Pradesh, India
- AcSIR: Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Madhavi Venkata Yaddanapudi
- Department of Chemical Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500 037, India.
| | - Srinivas Nanduri
- Department of Chemical Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500 037, India.
| |
Collapse
|
141
|
van den Maagdenberg HW, de Mol van Otterloo J, van Hasselt JGC, van der Graaf PH, van Westen GJP. Integrating Pharmacokinetics and Quantitative Systems Pharmacology Approaches in Generative Drug Design. J Chem Inf Model 2025; 65:4783-4796. [PMID: 40343729 PMCID: PMC12117666 DOI: 10.1021/acs.jcim.5c00107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Revised: 04/09/2025] [Accepted: 04/10/2025] [Indexed: 05/11/2025]
Abstract
Integrated understanding of pharmacokinetics (PK) and pharmacodynamics (PD) is a key aspect of successful drug discovery. Yet in generative computational drug design, the focus often lies on optimizing potency. Here we integrate PK property predictions in DrugEx, a generative drug design framework and we explore the generated compounds' PD through simulations with a quantitative systems pharmacology (QSP) model. Quantitative structure-property relationship models were developed to predict molecule PK (clearance, volume of distribution and unbound fraction) and affinity for the Adenosine A2AR receptor (A2AR), a drug target in immuno-oncology. These models were used to score compounds in a reinforcement learning framework to generate molecules with a specific PK profile and high affinity for the A2AR. We predicted the expected tumor growth inhibition profiles using the QSP model for selected candidate molecules with varying PK and affinity profiles. We show that optimizing affinity to the A2AR, while minimizing or maximizing a PK property, shifts the type of molecular scaffolds that are generated. The difference in physicochemical properties of the compounds with different predicted PK parameters was found to correspond with the differences observed in the PK data set. We demonstrated the use of the QSP model by simulating the effect of a broad range of compound properties on the predicted tumor volume. In conclusion, our proposed integrated workflow incorporating affinity predictions with PKPD may provide a template for the next generation of advanced generative computational drug design.
Collapse
Affiliation(s)
| | | | - J. G. Coen van Hasselt
- Leiden
Academic Centre of Drug Research, Leiden
University, 2333, Leiden, The Netherlands
| | - Piet H. van der Graaf
- Leiden
Academic Centre of Drug Research, Leiden
University, 2333, Leiden, The Netherlands
- Certara, CT2 7FGCanterbury, U.K.
| | | |
Collapse
|
142
|
Sharma S, Bose A, Biswas S, Sen S, Roy I. Cyperus rotundus mediated green synthesis of silver nanoparticles for antibacterial wound dressing applications. Sci Rep 2025; 15:18394. [PMID: 40419643 DOI: 10.1038/s41598-025-03555-x] [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: 03/14/2025] [Accepted: 05/21/2025] [Indexed: 05/28/2025] Open
Abstract
Wound healing is a complex biological process that can be hindered by persistent infections and inflammation, especially in the presence of multidrug-resistant (MDR) bacteria. Silver nanoparticles (AgNPs) have demonstrated significant antimicrobial efficacy; however, concerns regarding their toxicity have limited their therapeutic application. METHODS In this study, we developed a biocompatible Ag-NPs-based hydrogel using Cyperus rotundus extract via a green synthesis approach for prospective wound healing applications. The synthesized AgNPs were characterized for their physicochemical properties, confirming their stability and antibacterial potency against E. coli and S. epidermidis. The Ag-NPs-loaded hydrogel was formulated using Carbopol 974P and evaluated for its physicochemical properties, antibacterial activity, anti-inflammatory potential, and cytotoxicity. RESULTS Characterization studies confirmed the successful synthesis of AgNPs, exhibiting potent antibacterial, antioxidant, and anti-inflammatory properties. The Ag-NPs-loaded hydrogel demonstrated significant wound contraction in an excision wound model, comparable to standard treatment. Additionally, in vitro safety evaluations confirmed excellent biocompatibility, minimizing toxicity concerns associated with conventional silver formulations. CONCLUSIONS These findings suggest that the developed Ag-NPs-based hydrogel is an effective, natural, and safer alternative for advanced wound care, warranting further clinical validation.
Collapse
Affiliation(s)
- Susrita Sharma
- School of Pharmaceutical Sciences, Siksha O Anusandhan (Deemed to be University), Bhubaneswar, Odisha, 751003, India
| | - Anindya Bose
- School of Pharmaceutical Sciences, Siksha O Anusandhan (Deemed to be University), Bhubaneswar, Odisha, 751003, India.
| | - Soumojit Biswas
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, S.A.S. Nagar, S.A.S. Nagar, Punjab, 160062, India
| | - Shreeja Sen
- School of Pharmaceutical Sciences, Siksha O Anusandhan (Deemed to be University), Bhubaneswar, Odisha, 751003, India
| | - Ipsita Roy
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, S.A.S. Nagar, S.A.S. Nagar, Punjab, 160062, India
| |
Collapse
|
143
|
Jiang M, Alqahtani SA, Seto WK, Yilmaz Y, Pan Z, Valenti L, Eslam M. Alternative splicing: hallmark and therapeutic opportunity in metabolic liver disease. Gastroenterol Rep (Oxf) 2025; 13:goaf044. [PMID: 40438258 PMCID: PMC12116422 DOI: 10.1093/gastro/goaf044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Revised: 03/23/2025] [Accepted: 04/15/2025] [Indexed: 06/01/2025] Open
Abstract
Metabolic dysfunction-associated fatty liver disease (MAFLD) has become the leading cause of chronic liver disease worldwide, with fibrosis recognized as the main prognostic factor and therapeutic target. While early-stage fibrosis is reversible, advanced fibrosis poses a significant clinical challenge due to limited treatment options, highlighting the need for innovative management strategies. Recent studies have shown that alternative pre-mRNA splicing, a critical mechanism regulating gene expression and protein diversity, plays a fundamental role in the pathogenesis of MAFLD and associated fibrosis. Understanding the complex relationship between alternative splicing and fibrosis progression in MAFLD could pave the way for novel therapeutic approaches and improve clinical outcomes. In this review, we describe the intricate mechanisms of alternative splicing in fibrosis associated with MAFLD. Specifically, we explored the pivotal of splicing factors, and RNA-binding proteins, highlighting their critical interactions with metabolic and epigenetic regulators. Furthermore, we provide an overview of the latest advancements in splicing-based therapeutic strategies and biomarker development. Particular emphasis is placed on the potential application of antisense oligonucleotides for rectifying splicing anomalies, thereby laying the foundation for precision medicine approaches in the treatment of MAFLD-associated fibrosis.
Collapse
Affiliation(s)
- Mingqian Jiang
- Department of Endocrinology and Metabolism, People’s Hospital Affiliated to Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, P. R. China
- Storr Liver Centre, Westmead Institute for Medical Research, Westmead Hospital and University of Sydney, NSW, Australia
| | - Saleh A Alqahtani
- Liver, Digestive, & Lifestyle Health Research Section, and Organ Transplant Center of Excellence, King Faisal Specialist Hospital & Research Center, Riyadh, Saudi Arabia
- Division of Gastroenterology and Hepatology, Weill Cornell Medicine, New York, NY, USA
| | - Wai-Kay Seto
- Department of Medicine, The University of Hong Kong, Hong Kong, P. R. China
- State Key Laboratory of Liver Research, The University of Hong Kong, Hong Kong, P. R. China
| | - Yusuf Yilmaz
- Department of Gastroenterology, School of Medicine, Recep Tayyip Erdoğan University, Rize, Türkiye
| | - Ziyan Pan
- Storr Liver Centre, Westmead Institute for Medical Research, Westmead Hospital and University of Sydney, NSW, Australia
| | - Luca Valenti
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy
- Precision Medicine, Biological Resource Center, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico Milano, Milan, Italy
| | - Mohammed Eslam
- Storr Liver Centre, Westmead Institute for Medical Research, Westmead Hospital and University of Sydney, NSW, Australia
| |
Collapse
|
144
|
Marco-Contelles JL, Oset-Gasque MJ. Cholinesterase-based inhibitors as multitarget small molecules for the therapy of Alzheimer's disease. Chem Biol Interact 2025; 413:111497. [PMID: 40158868 DOI: 10.1016/j.cbi.2025.111497] [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: 12/17/2024] [Revised: 03/14/2025] [Accepted: 03/27/2025] [Indexed: 04/02/2025]
Abstract
Herein, we have summarized the most significant results that we have communicated from our laboratories in the last thirty years, highlighting the most potent and attractive ChEIs based hit(lead)-Multitarget Small Molecules, such as (S)-p-methoxytacripyrine (1), ASS234 (2), Contilisant (3), and Contilistat (4), that we have identified in the search for new chemical entities for the therapy of Alzheimer's disease.
Collapse
Affiliation(s)
- José Luis Marco-Contelles
- Laboratory of Medicinal Chemistry, Institute of General Organic Chemistry (CSIC), C/ Juan de La Cierva, 3, 28006, Madrid, Spain.
| | - María Jesús Oset-Gasque
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Complutense University of Madrid, Plaza Ramón y Cajal S/n, Ciudad Universitaria, 28040, Madrid, Spain; Instituto de Investigación Sanitaria del Hospital Clínico San Carlos, 28040, Madrid, Spain; Instituto Universitario de Investigación en Neuroquímica, Complutense University of Madrid, Ciudad Universitaria, 28040, Madrid, Spain
| |
Collapse
|
145
|
Jia H, Wei J, Zheng W, Li Z. The dual role of autophagy in cancer stem cells: implications for tumor progression and therapy resistance. J Transl Med 2025; 23:583. [PMID: 40414839 DOI: 10.1186/s12967-025-06595-z] [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: 02/24/2025] [Accepted: 05/08/2025] [Indexed: 05/27/2025] Open
Abstract
Cancer stem cells (CSCs) constitute a small yet crucial subgroup in tumors, known for their capacity to self-renew, differentiate, and promote tumor growth, metastasis, and resistance to therapy. These characteristics position CSCs as significant factors in tumor recurrence and unfavorable clinical results, emphasizing their role as targets for therapy. Autophagy, an evolutionarily preserved cellular mechanism for degradation and recycling, has a complex function in cancer by aiding cell survival during stress and preserving balance by eliminating damaged organelles and proteins. Although autophagy can hinder tumor growth by reducing genomic instability, it also aids tumor advancement, particularly in harsh microenvironments, highlighting its dual characteristics. Recent research has highlighted the complex interactions between autophagy and CSCs, showing that autophagy governs CSC maintenance, boosts survival, and aids in resistance to chemotherapy and radiotherapy. On the other hand, in specific situations, autophagy may restrict CSC growth by increasing differentiation or inducing cell death. These intricate interactions offer both obstacles and possibilities for therapeutic intervention. Pharmacological modulation of autophagy, via inhibitors like chloroquine or by enhancing autophagy when advantageous, has demonstrated potential in making CSCs more responsive to standard treatments. Nonetheless, applying these strategies in clinical settings necessitates a better understanding of context-dependent autophagy dynamics and the discovery of dependable biomarkers indicating autophagic activity in CSCs. Progressing in this area might unveil novel, accurate strategies to tackle therapy resistance, lessen tumor recurrence, and ultimately enhance patient outcomes.
Collapse
Affiliation(s)
- Haiqing Jia
- Department of Gynecology, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, No.44 xiaoheyan road, Shenyang, 110042, China
| | - Jing Wei
- Department of Gynecology, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, No.44 xiaoheyan road, Shenyang, 110042, China
| | - Wei Zheng
- Department of Gynecology, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, No.44 xiaoheyan road, Shenyang, 110042, China.
| | - Zhuo Li
- Department of Gynecology, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, No.44 xiaoheyan road, Shenyang, 110042, China.
| |
Collapse
|
146
|
Qayyum N, Seo H, Khan N, Manan A, Ramachandran R, Haseeb M, Kim E, Choi S. A hybrid protocol for peptide development: integrating deep generative models and physics simulations for biomolecular design targeting IL23R/IL23. Int J Biol Macromol 2025; 316:144652. [PMID: 40419055 DOI: 10.1016/j.ijbiomac.2025.144652] [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: 03/22/2025] [Revised: 05/13/2025] [Accepted: 05/23/2025] [Indexed: 05/28/2025]
Abstract
Recent advances in machine learning have revolutionized molecular design; however, a gap remains in integrating generative models with physics-based simulations to develop functional modulators, such as stable peptides, for challenging targets like the interleukin-23 receptor (IL23R) and its associated cytokine, interleukin-23 (IL23). The IL23R/IL23 axis plays a critical role in autoimmune diseases, and current therapies have largely been limited to antibody-based approaches. To address this gap, we employed a hybrid computational approach that combines Long Short-Term Memory (LSTM) networks for peptide generation, a Gated Recurrent Unit (GRU)-based classifier for anti-inflammatory property prediction, and molecular dynamics (MD) simulations to assess structural dynamics, binding interactions, as well as key properties such as binding affinity and stability. Using this hybrid framework, we identified novel inhibitory peptides, particularly P4, with an IC50 of 2 μM. Systematic experimental validation established its inhibitory activity, elucidated its binding mechanism, confirmed its specificity toward the IL23R, and demonstrated its ability to disrupt IL23R/IL23 interaction. This integrated approach highlights the significant potential of combining deep learning and simulations to accelerate the identification of peptide-based therapeutics targeting key protein targets.
Collapse
Affiliation(s)
- Naila Qayyum
- Department of Molecular Science and Technology, Ajou University, Suwon 16499, South Korea.
| | - Hana Seo
- Department of Molecular Science and Technology, Ajou University, Suwon 16499, South Korea
| | - Noman Khan
- Department of Computer Science, Yonsei University, Seoul 03722, South Korea
| | - Abdul Manan
- Department of Molecular Science and Technology, Ajou University, Suwon 16499, South Korea
| | - Rajath Ramachandran
- Department of Molecular Science and Technology, Ajou University, Suwon 16499, South Korea
| | - Muhammad Haseeb
- S&K Therapeutics, Ajou University Campus Plaza 418, Worldcup-ro 199, Yeongtong-gu, Suwon 16502, South Korea
| | - Eunha Kim
- Department of Molecular Science and Technology, Ajou University, Suwon 16499, South Korea
| | - Sangdun Choi
- S&K Therapeutics, Ajou University Campus Plaza 418, Worldcup-ro 199, Yeongtong-gu, Suwon 16502, South Korea.
| |
Collapse
|
147
|
Rekik N. Theoretical elucidation of the IR spectra of 4-bromo-3, 5-dimethylpyrazole crystals and their deuterium-bonded analogues: Multi-objective analysis from a quantum modeling perspective. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2025; 341:126328. [PMID: 40418868 DOI: 10.1016/j.saa.2025.126328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2025] [Revised: 04/18/2025] [Accepted: 05/01/2025] [Indexed: 05/28/2025]
Abstract
In the field of medicine, compounds that contain pyrazole and their derivatives are recognized as some of the earliest anti-inflammatory and analgesic agents. Consequently, the development of novel pyrazole-containing synthons represents a promising avenue in the quest for new biologically active substances. The supramolecular aspect plays a crucial role in comprehending the chemistry of pyrazoles. The primary driving force behind the self-assembly of pyrazolyl molecules is attributed to hydrogen bonds. In this context, we propose a quantum theoretical approach to hydrogen bonding in order to elucidate the infrared (IR) spectra of 4-bromo-3,5-dimethylpyrazole (4-Br,3,5-DMPz) crystals. Our ultimate objective is to elucidate the hydrogen-bond dynamics and illustrate the main mechanisms that govern the generation of the IR band contours. The approach addresses the impact of the anharmonic vibrational coupling within the dimeric units, the Davydov coupling and the combined influences of damping mechanisms on theυSX-H IR band contours. The exchange interaction between the two hydrogen bonds of the dimeric units is analyzed through the non-adiabatic coupling framework. The direct damping mechanism is addressed within the theoretical framework established by Rösch and Ratner, while the indirect damping associated with the two hydrogen-bonded bridges is integrated using non-Hermitian Hamiltonians. The contours of the IR bands are obtained through linear response theory by applying a Fourier transform to the dipole moment operator autocorrelation function associated with the high-frequency mode. This theoretical framework reverts to the model proposed by Maréchal and Witkowski regarding Davydov coupling in damping lack, and aligns with the foundational quantum approach of indirect damping when Davydov coupling is not present. The methodology is applied to the study the IR spectra of crystals of 4-Br,3,5-DMPz and their deuterium-bonded analogues at 293 K and 77 K. Numerical simulations demonstrate that the congregated influence of Davydov coupling, the linear interaction within the dimeric units, and the various damping mechanisms can effectively account for the intricate characteristics of the experimental IR band contours. The notable distinctions identified in the fine structure patterns of theυSX-H(D) band contours, along with the temperature effects in these crystals, have been clarified and interpreted from a physical perspective.
Collapse
Affiliation(s)
- Najeh Rekik
- Physics Department, College of Science, King Faisal University, Al Ahsa, 31982, Saudi Arabia.
| |
Collapse
|
148
|
Khakshur AA, Khodaverdi E, Kamali H, Nokhodchi A. An insight into cell-penetrating peptides: perspectives on design, optimization, and targeting in drug delivery systems. Pharm Dev Technol 2025:1-27. [PMID: 40356455 DOI: 10.1080/10837450.2025.2505000] [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: 02/12/2025] [Revised: 05/06/2025] [Accepted: 05/08/2025] [Indexed: 05/15/2025]
Abstract
The authors carried out a comprehensive review of the application of peptides known as cell-penetrating peptides (CPPs) in various drug delivery systems (DDS), with the prospect of achieving novel solutions and ideas to overcome the challenges of DDS, by making them more able to penetrate cells and biological membranes. A conceptual search was conducted in relevant literature databases (Scopus, PubMed, Web of Science, and Google Scholar) up to 1 April 2025 using keywords such as drug delivery systems, cell-penetrating peptides, CPPs, complexes, conjugates, nanoparticles, dendrimers, exosomes, liquid crystalline, liposomes, micelles, nanospheres and lipid nanoparticles. The studies demonstrate that the antimicrobial effect of drugs, including curcumin, gentamicin, and antifungal drugs like imidazoacridinone derivatives, can be enhanced when they are conjugated or complexed with CPPs. CPPs possess positive charges, which make them suitable for gene therapy applications by facilitating the delivery of plasmids and siRNAs with negative charges in modern delivery systems. Medicinal formulations containing CPPs in combination with liquid crystals or nanostructured lipid carriers (NLCs) increase drugs penetration to the skin. Additionally, several investigations showed that CPPs could have a positive impact on the pharmacokinetic and pharmacodynamic of chemotherapy agents, reducing their side effects. CPPs have significant potential in enhancing penetration, bioavailability, targeting, and optimization of DDS. By using computer modeling and designing CPPs with more desirable features and conducting more clinical studies, new methods for treating diseases and better formulations can be achieved.
Collapse
Affiliation(s)
- Ali Asghar Khakshur
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
- Targeted Drug Delivery Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Elham Khodaverdi
- Targeted Drug Delivery Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Pharmaceutics, School of Pharmacy, Mashhad University of Medical Science, Mashhad, Iran
| | - Hossein Kamali
- Targeted Drug Delivery Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Pharmaceutics, School of Pharmacy, Mashhad University of Medical Science, Mashhad, Iran
| | - Ali Nokhodchi
- Pharmaceutics Research Laboratory, School of Life Sciences, University of Sussex, Brighton, UK
| |
Collapse
|
149
|
Yang Y, Qing L, You C, Li Q, Xu W, Dong Z. Methuosis key gene ARF6 as a diagnostic, prognostic and immunotherapeutic marker for prostate cancer: based on a comprehensive pan-cancer multi-omics analysis. Discov Oncol 2025; 16:882. [PMID: 40410613 PMCID: PMC12102050 DOI: 10.1007/s12672-025-02275-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Accepted: 04/01/2025] [Indexed: 05/25/2025] Open
Abstract
BACKGROUND Prostate cancer (PCa) is a leading cause of cancer-related mortality among men worldwide. Despite progress in the understanding of tumor biology, the prognosis for advanced prostate cancer remains poor, necessitating the identification of novel diagnostic, prognostic, and therapeutic biomarkers. Methuosis, a recently identified form of programmed cell death (PCD), is characterized by cytoplasmic vacuole accumulation and subsequent cell rupture, distinct from classical apoptosis and necrosis. The key regulatory gene in Methuosis, ARF6 (ADP-ribosylation factor 6), has emerged as a potential marker for cancer diagnosis and treatment. However, its role in prostate cancer and other malignancies remains insufficiently understood. METHODS In this study, we performed a comprehensive pan-cancer multi-omics analysis to investigate the role of ARF6 in Methuosis across multiple cancer types, with a specific focus on PCa as the primary context. Using data from public databases, including RNA sequencing, gene expression profiling, and clinical outcomes, we assessed the association between ARF6 expression and patient prognosis in PCa within this broader pan-cancer framework. Additionally, we employed functional enrichment analyses and survival analysis to explore the potential of ARF6 as a diagnostic and prognostic marker for prostate cancer. Immunotherapy-related gene expression signatures were also evaluated to determine the therapeutic relevance of ARF6. RESULTS ARF6 was significantly overexpressed in PCa tissues compared to normal tissues and was associated with poor prognosis (p < 0.05), particularly in advanced and metastatic stages. Receiver operating characteristic (ROC) analysis revealed a diagnostic AUC of 0.792 for ARF6. Functional analyses indicated that ARF6 regulates pathways critical to cell migration, invasion, and drug resistance. Moreover, ARF6 expression showed a strong negative correlation with immune checkpoint markers, such as PD-L1 (r = - 0.74), suggesting its potential as an immunotherapy target. These findings underscore ARF6's pivotal role in Methuosis and its promise as a biomarker in PCa. CONCLUSION ARF6 is a key regulator of Methuosis in prostate cancer, contributing to tumor progression, metastasis, and resistance to treatment. Our findings support the potential of ARF6 as a diagnostic, prognostic, and immunotherapeutic target in prostate cancer. Further experimental validation is needed to confirm these observations and to explore the therapeutic implications of targeting ARF6 in cancer treatment.
Collapse
Affiliation(s)
- Yongjin Yang
- Department of Urology, the Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, 730000, Gansu, China
- Gansu Province Clinical Research Center for Urinary System Disease, Lanzhou, 730000, Gansu, China
| | - Liangliang Qing
- Department of Urology, Zigong Fourth People's Hospital, No.19 Tanmulin Street, Ziliujing District, Zigong, 643000, China
| | - Chengyu You
- Department of Urology, the Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, 730000, Gansu, China
- Gansu Province Clinical Research Center for Urinary System Disease, Lanzhou, 730000, Gansu, China
| | - Qingchao Li
- Department of Urology, the Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, 730000, Gansu, China
- Gansu Province Clinical Research Center for Urinary System Disease, Lanzhou, 730000, Gansu, China
| | - Wenbo Xu
- Department of Urology, the Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, 730000, Gansu, China
- Gansu Province Clinical Research Center for Urinary System Disease, Lanzhou, 730000, Gansu, China
| | - Zhilong Dong
- Department of Urology, the Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, 730000, Gansu, China.
- Gansu Province Clinical Research Center for Urinary System Disease, Lanzhou, 730000, Gansu, China.
| |
Collapse
|
150
|
Chen X, Au CM, Fang P, Xue Y, Leung KCF, Chan WL. Late-Stage N-Alkenylative Modifications of Indolic Scaffolds with Propiolates: Toward Bisconjugation and Macrocyclization. Org Lett 2025; 27:5081-5086. [PMID: 40368809 DOI: 10.1021/acs.orglett.5c01162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2025]
Abstract
A facile, mild, and scalable late-stage N-alkenylative modification strategy is introduced on 1H-indoles, 9H-carbazoles, and their structural derivatives and analogues, including alkaloids, bioactive agents, and tryptophan motifs, via chemo- and regioselective phosphine-mediated propiolate hydroamination. Saliently, through this protocol, bisconjugation and macrocyclization on (bis)indolic scaffolds can also be accomplished, with the installation of new α,β-unsaturated ester handles for potential further versatile synthetic manipulations.
Collapse
Affiliation(s)
- Xiaoye Chen
- The International Joint Institute of Tianjin University-National University of Singapore in Fuzhou, Tianjin University, Tianjin 300072, China
- Department of Chemistry, National University of Singapore, 3 Science Drive, Singapore 117543
| | - Chi-Ming Au
- Department of Chemistry, National University of Singapore, 3 Science Drive, Singapore 117543
| | - Pengyuan Fang
- The International Joint Institute of Tianjin University-National University of Singapore in Fuzhou, Tianjin University, Tianjin 300072, China
| | - Yunsheng Xue
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, School of Pharmacy, Xuzhou Medical University, No. 209, Tongshan Road, Xuzhou, Jiangsu 221004, China
| | - Ken Cham-Fai Leung
- Department of Chemistry, Hong Kong Baptist University, Kowloon Tong, Kowloon 999077, Hong Kong SAR, China
| | - Wai-Lun Chan
- The International Joint Institute of Tianjin University-National University of Singapore in Fuzhou, Tianjin University, Tianjin 300072, China
- Department of Chemistry, National University of Singapore, 3 Science Drive, Singapore 117543
| |
Collapse
|