1
|
Herring P, Roedgaard M, Holst CM, Christensen H, Knudsen BR, Bjergbaek L, Andersen AH. A cellular system to study responses to a collision between the transcription complex and a protein-bound nick in the DNA template. FEBS Lett 2025. [PMID: 40309784 DOI: 10.1002/1873-3468.70053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 03/12/2025] [Accepted: 04/01/2025] [Indexed: 05/02/2025]
Abstract
We present a transcription-coupled Flp-nick system enabling a stable protein-bound nick mimicking a topoisomerase I-DNA cleavage complex. The nick is introduced at a single site within a controllable LacZ gene inserted into the Saccharomyces cerevisiae genome. This system allows unique single-site studies of a frequently occurring damage within a transcription unit in vivo. As proof of principle, we demonstrate RNA polymerase II accumulation at the damage site when MG132 inhibits the proteasome. Similarly, accumulation occurs when polymerase ubiquitination is abolished by deletion of the ubiquitinase ELC1 gene. This indicates that a topoisomerase I-DNA mimicking cleavage complex per se induces RNA polymerase II ubiquitination and degradation. These findings advance understanding of cellular responses to topoisomerase I-targeting drugs used in cancer chemotherapy.
Collapse
Affiliation(s)
- Petra Herring
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus C, Denmark
| | - Morten Roedgaard
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus C, Denmark
| | - Camilla Myrup Holst
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus C, Denmark
| | - Helene Christensen
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus C, Denmark
| | - Birgitta R Knudsen
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus C, Denmark
| | - Lotte Bjergbaek
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus C, Denmark
| | | |
Collapse
|
2
|
El-Saghier AM, Abosella L, Hassan A, Elakesh EO, Bräse S, Abuo-Rahma GEDA, Aziz HA. Design, Synthesis, and In Silico Studies of New Norfloxacin Analogues with Broad Spectrum Antibacterial Activity via Topoisomerase II Inhibition. Pharmaceuticals (Basel) 2025; 18:545. [PMID: 40283980 PMCID: PMC12030355 DOI: 10.3390/ph18040545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2025] [Revised: 04/02/2025] [Accepted: 04/05/2025] [Indexed: 04/29/2025] Open
Abstract
Background: Novel norfloxacin derivatives were synthesized, characterized, and screened for their antibacterial activity against Gram-positive strain S. aureus ATCC 6538 and Gram-negative strains; E. coli ATCC 25923, K. pneumoniae ATCC 10031, and P. aeruginosa ATCC 27853 using the agar cup diffusion method. Results: The results revealed that compounds 6-17 exhibited more potent activity towards S. aureus ATCC 6538 with MIC values of 0.21-3.61 µM than norfloxacin with a MIC of 7.83 µM. The most potent compound, 6, showed 37-fold more potency than norfloxacin. More importantly, compound 7 exhibited more potent activity against MRSA than norfloxacin, with MIC values of 0.80 and 1.96 µM, respectively. Meanwhile, compounds 15 and 16 have potent activity towards the Gram-negative strains with MIC values of 0.20-0.79 µM compared with norfloxacin with a MIC of 0.24 µM. Moreover, the potent compounds showed higher activity towards topoisomerase II enzymes, especially against topoisomerase IV, which confirms the docking study with the S. aureus gyrase enzyme active binding site (PDB ID: 2XCT). In addition, cytotoxicity assays of the most potent compounds showed that compounds 6, 7, 15, and 16 have negligible risks of toxic effects when evaluated against the normal cell line WI 38. Conclusions: The docking study of the most potent compounds 6, 7, 15, and 16 on the gyrase enzyme active site (PDB: 2XCT) aligns their antibacterial activity and topoisomerase inhibition. The physicochemical and pharmacokinetic characteristics of the target derivatives were forecasted via SwissADME. Hence, these compounds are considered promising antibacterial candidates that require further optimization.
Collapse
Affiliation(s)
- Ahmed M. El-Saghier
- Chemistry Department, Faculty of Science, Sohag University, Sohag 82524, Egypt;
| | - Laila Abosella
- Chemistry Department, Faculty of Science, Sohag University, Sohag 82524, Egypt;
- Medicinal Chemistry Department, Faculty of Pharmacy—Al-Jmail, Sabratha University, Sabratha P.O. Box 250, Libya
| | - Abdelfattah Hassan
- Medicinal Chemistry Department, Faculty of Pharmacy, South Valley University, Qena 83523, Egypt;
- Medicinal Chemistry Department, Faculty of Pharmacy, National South Valley University, Qena 83523, Egypt
| | - Esmail O. Elakesh
- Chemistry Department, Faculty of Science, University of Zawia, Al Zawiya 16418, Libya;
| | - Stefan Bräse
- Institute for Biological and Chemical System, Karlsruhe Institute of Technology, 76131 Karlsruhe, Germany
| | - Gamal El-Din A. Abuo-Rahma
- Medicinal Chemistry Department, Faculty of Pharmacy, Minia University, Minia 61519, Egypt;
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Deraya University, New Minia-61768, Egypt
| | - Hossameldin A. Aziz
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, New Vallely University, New Valley 72511, Egypt;
| |
Collapse
|
3
|
Huang C, Tang B, Chen W, Chen J, Zhang H, Bai M. Multiomic traits reveal that critical irinotecan-related core regulator FSTL3 promotes CRC progression and affects ferroptosis. Cancer Cell Int 2025; 25:115. [PMID: 40140870 PMCID: PMC11938592 DOI: 10.1186/s12935-025-03753-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Accepted: 03/18/2025] [Indexed: 03/28/2025] Open
Abstract
BACKGROUND Irinotecan is a widely used chemotherapy drug in colorectal cancer (CRC). The evolution and prognosis of CRC involve complex mechanisms and depend on the drug administered, especially for irinotecan. However, the specific mechanism and prognostic role of irinotecan-related regulators remain to be elucidated. METHODS Data from public databases were used to explore the multiomic traits of irinotecan-related regulators through bioinformatics analysis. RT‒qPCR, western blotting, transmission electron microscopy and flow cytometry were used as experimental validations. RESULTS Iriscore (irinotecan-related score) was constructed based on irinotecan-related regulators, and a high iriscore predicted a poor prognosis, poor therapeutic response and the MSS/MSI-L status. Single-cell analysis revealed that FSTL3 and TMEM98 were mainly expressed in CRC stem cells. Potential transcription factors (E2F1, STAT1, and TTF2) and therapeutic drugs (telatinib) that target irinotecan-related regulators were identified. FSTL3 was the core risk irinotecan-related regulator. Some ferroptosis regulators (GPX4, HSPB1 and RGS4) and related metabolic pathways (lipid oxidation and ROS metabolism) were correlated significantly with FSTL3. In vitro, irinotecan inhibited the expression of FSTL3 and ferroptotic defence proteins (GPX4 and SLC7A11), and induced lipid peroxidation and intracellular Fe (2+) ions concentration increased. CONCLUSIONS We confirmed that irinotecan-related regulators, especially FSTL3, have effective prognostic value in CRC and speculated that FSTL3 may promote CRC progression and affect ferroptosis, which is beneficial for identifying candidate targeted irinotecan-related regulators and accurate individualized treatment strategies for CRC.
Collapse
Affiliation(s)
- Chengyi Huang
- Department of Radiation Oncology, Changhai Hospital Affiliated to Naval Medical University, Shanghai, 200433, China
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, 310022, China
- Zhejiang Key Laboratory of Radiation Oncology, Hangzhou, Zhejiang, 310022, China
| | - Bufu Tang
- Department of Radiation Oncology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Wenjuan Chen
- Department of Radiation Oncology, Changhai Hospital Affiliated to Naval Medical University, Shanghai, 200433, China
| | - Jinggang Chen
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, 310022, China
- Zhejiang Key Laboratory of Radiation Oncology, Hangzhou, Zhejiang, 310022, China
| | - Huojun Zhang
- Department of Radiation Oncology, Changhai Hospital Affiliated to Naval Medical University, Shanghai, 200433, China.
| | - Minghua Bai
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, 310022, China.
- Zhejiang Key Laboratory of Radiation Oncology, Hangzhou, Zhejiang, 310022, China.
| |
Collapse
|
4
|
Oliveira RJ, Oliveira Junior AB, Contessoto VG, Onuchic JN. The synergy between compartmentalization and motorization in chromatin architecture. J Chem Phys 2025; 162:114116. [PMID: 40105139 DOI: 10.1063/5.0239634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 02/20/2025] [Indexed: 03/20/2025] Open
Abstract
High-resolution techniques capable of manipulating from single molecules to millions of cells are combined with three-dimensional modeling followed by simulation to comprehend the specific aspects of chromosomes. From the theoretical perspective, the energy landscape theory from protein folding inspired the development of the minimal chromatin model (MiChroM). In this work, two biologically relevant MiChroM energy terms were minimized under different conditions, revealing a competition between loci compartmentalization and motor-driven activity mechanisms in chromatin folding. Enhancing the motor activity energy baseline increased the lengthwise compaction and reduced the polymer entanglement. Concomitantly, decreasing compartmentalization-related interactions reduced the overall polymer collapse, although compartmentalization given by the microphase separation remained almost intact. For multiple chromosome simulations, increased motorization intensified the territory formation of the different chains and reduced compartmentalization strength lowered the probability of contact formation of different loci between multiple chains, approximating to the experimental inter-contacts of the human chromosomes. These findings have direct implications for experimental data-driven chromosome modeling, specially those involving multiple chromosomes. The interplay between phase-separation and territory formation mechanisms should be properly implemented in order to recover the genome architecture and dynamics, features that might play critical roles in regulating nuclear functions.
Collapse
Affiliation(s)
- Ronaldo J Oliveira
- Laboratório de Biofísica Teórica, Departamento de Física, Instituto de Ciências Exatas, Naturais e Educação, Universidade Federal do Triângulo Mineiro, Uberaba, MG 38064-200, Brazil
- Center for Theoretical Biological Physics, Rice University, Houston, Texas 77030, USA
| | | | - Vinícius G Contessoto
- Center for Theoretical Biological Physics, Rice University, Houston, Texas 77030, USA
| | - José N Onuchic
- Center for Theoretical Biological Physics, Rice University, Houston, Texas 77030, USA
- Department of Physics and Astronomy, Rice University, Houston, Texas 77030, USA
- Department of Chemistry, Rice University, Houston, Texas 77030, USA
- Department of Biosciences, Rice University, Houston, Texas 77030, USA
| |
Collapse
|
5
|
Chang TY, Lin CJ, Wen SN, Wu YC, Wei CY, Huang JY, Tsao YH, Chen YJ, Tang WC, Wu YC, Lee WH, Huang TY, Kuo TM, Li WF, Lai MT. Preclinical evaluation of a novel antibody-drug conjugate OBI-992 for Cancer therapy. Sci Rep 2025; 15:8735. [PMID: 40082588 PMCID: PMC11906863 DOI: 10.1038/s41598-025-92697-z] [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: 11/14/2024] [Accepted: 03/03/2025] [Indexed: 03/16/2025] Open
Abstract
Trophoblast cell surface antigen 2 (TROP2), a transmembrane glycoprotein highly expressed in a variety of epithelial cancers, has been considered as a primary therapeutic target for the development of antibody-drug conjugates (ADCs). OBI-992, an investigational TROP2-targeted ADC, is composed of a novel TROP2 antibody (R4702) conjugated to the topoisomerase I (TOP1) inhibitor exatecan through a hydrophilic enzyme-cleavable linker. This study aimed to characterize R4702 and OBI-992 in vitro. TROP2-targeted antibodies sacituzumab and datopotamab were employed as the comparators for R4702. ADCs sacituzumab govitecan (SG) and datopotamab deruxtecan (Dato-DXd) were used as benchmarks for OBI-992. Results revealed that R4702 binds to an epitope that is distinct from sacituzumab and datopotamab. The cytotoxicity of the OBI-992, SG, and Dato-DXd against different cancer cells is comparable despite they have different internalization profile. Upregulation of breast cancer resistance protein (BCRP) was observed in SG-resistant and Dato-DXd-resistant cells, but not in OBI-992-resistant cells. In addition, significant downregulation of TROP2 expression was detected with Dato-DXd-resistant cells and only slightly downregulation with SG- and OBI-992-resistant cells was observed. Moreover, substantial enhancement of cytotoxicity and DNA damage was found in the combination of OBI-992 with a poly (ADP-ribose) polymerase (PARP) inhibitor (talazoparib). Taken together, the findings in this study support further clinical development of OBI-992.
Collapse
Affiliation(s)
- Ting-Yu Chang
- OBI Pharma, Inc., 6F, No. 508, Section 7 Zhongxiao East Road, Nangang District, Taipei, Taiwan
| | - Chun-Jung Lin
- OBI Pharma, Inc., 6F, No. 508, Section 7 Zhongxiao East Road, Nangang District, Taipei, Taiwan
| | - Shih-Ni Wen
- OBI Pharma, Inc., 6F, No. 508, Section 7 Zhongxiao East Road, Nangang District, Taipei, Taiwan
| | - Yi-Chen Wu
- OBI Pharma, Inc., 6F, No. 508, Section 7 Zhongxiao East Road, Nangang District, Taipei, Taiwan
| | - Cheng-Yen Wei
- OBI Pharma, Inc., 6F, No. 508, Section 7 Zhongxiao East Road, Nangang District, Taipei, Taiwan
| | - Jye-Yu Huang
- OBI Pharma, Inc., 6F, No. 508, Section 7 Zhongxiao East Road, Nangang District, Taipei, Taiwan
| | - Yu-Hsuan Tsao
- OBI Pharma, Inc., 6F, No. 508, Section 7 Zhongxiao East Road, Nangang District, Taipei, Taiwan
| | - Yu-Jung Chen
- OBI Pharma, Inc., 6F, No. 508, Section 7 Zhongxiao East Road, Nangang District, Taipei, Taiwan
| | - Wei-Chien Tang
- OBI Pharma, Inc., 6F, No. 508, Section 7 Zhongxiao East Road, Nangang District, Taipei, Taiwan
| | - Yuen-Chin Wu
- OBI Pharma, Inc., 6F, No. 508, Section 7 Zhongxiao East Road, Nangang District, Taipei, Taiwan
| | - Wei-Han Lee
- OBI Pharma, Inc., 6F, No. 508, Section 7 Zhongxiao East Road, Nangang District, Taipei, Taiwan
| | - Teng-Yi Huang
- OBI Pharma, Inc., 6F, No. 508, Section 7 Zhongxiao East Road, Nangang District, Taipei, Taiwan
| | - Tzer-Min Kuo
- OBI Pharma, Inc., 6F, No. 508, Section 7 Zhongxiao East Road, Nangang District, Taipei, Taiwan
| | - Wan-Fen Li
- OBI Pharma, Inc., 6F, No. 508, Section 7 Zhongxiao East Road, Nangang District, Taipei, Taiwan
| | - Ming-Tain Lai
- OBI Pharma, Inc., 6F, No. 508, Section 7 Zhongxiao East Road, Nangang District, Taipei, Taiwan.
| |
Collapse
|
6
|
Hill E, Hill A, Voisin E, Byrd A, Schoeffler A. Localized Amino Acid Enrichment Analysis as a Tool for Understanding Protein Extremophilicity. Proteins 2025; 93:702-715. [PMID: 39513552 DOI: 10.1002/prot.26760] [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: 09/11/2024] [Revised: 10/13/2024] [Accepted: 10/19/2024] [Indexed: 11/15/2024]
Abstract
Sequence conservation analyses offer us a powerful glimpse of natural selection at work. Standard tools for measuring sequence conservation report conservation as a function of a specific location in a multiple sequence alignment and have proven indispensable in identifying highly constrained features such as active site residues. The advent of large-scale genomic sequencing efforts allows researchers to expand this paradigm and investigate more nuanced relationships between sequence and function. Here, we present a simple tool (SWiLoDD: Sliding Window Localized Differentiation Detection) that allows researchers to analyze local, rather than site-specific, conservation using a sliding window approach. Our tool accepts multiple sequence alignments partitioned based on a biological differentiator and returns alignment position-based, localized differential enrichment metrics for amino acids of choice. We present two case studies of this analysis in action: local-but-diffuse glycine enrichments in the ATPase subunits of thermophilic and psychrophilic bacterial gyrase homologs, and ligand- and interface-specific amino acid enrichments in halophilic bacterial crotonyl-CoA carboxylases/reductases. Though we have described examples of extremophilic bacterial proteins in this study, our tool may be used to investigate any set of homologous sequences from which sub-groups can be meaningfully partitioned. Our results suggest that investigating differential localized conservation in partitioned MSAs will expand our understanding of how sequence conservation and protein function are connected.
Collapse
Affiliation(s)
- Elliot Hill
- Department of Chemistry & Biochemistry, Loyola University New Orleans, New Orleans, Louisiana, USA
| | - Avery Hill
- Department of Chemistry & Biochemistry, Loyola University New Orleans, New Orleans, Louisiana, USA
| | - Elena Voisin
- Department of Chemistry & Biochemistry, Loyola University New Orleans, New Orleans, Louisiana, USA
| | - Amber Byrd
- Department of Chemistry & Biochemistry, Loyola University New Orleans, New Orleans, Louisiana, USA
| | - Allyn Schoeffler
- Department of Chemistry & Biochemistry, Loyola University New Orleans, New Orleans, Louisiana, USA
| |
Collapse
|
7
|
Nivón-Ramirez D, Galván-García EA, Ponce-Pérez LD, Gómez-Balderas R. Virtual screening of potential inhibitors of the ATPase site in Acinetobacter baumannii DNA Gyrase. Comput Biol Med 2025; 186:109728. [PMID: 39862471 DOI: 10.1016/j.compbiomed.2025.109728] [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: 10/16/2024] [Revised: 01/16/2025] [Accepted: 01/18/2025] [Indexed: 01/27/2025]
Abstract
Bacterial resistance is a global public health problem because of the ineffectiveness of conventional antibiotics against super pathogens. To counter this situation, the search for or design of new molecules is essential to inhibit the key proteins involved in several stages of bacterial infection. One of these key proteins is DNA gyrase, which is responsible for packaging and unfolding of DNA chains during replication. Virtual screening calculations of 583,900 molecules against the ATPase site of DNA gyrase (PDB ID 7PQM) resulted in three promising molecules (Z927783420, Z4422201766, and Z2440107042) with significant binding modes at the active site, according to molecular docking studies. Additionally, they exhibited lower toxicological profiles than the previously reported 80S inhibitors. Molecular dynamics calculations revealed crucial interactions responsible for the inhibition process, with residues ASP87, GLU94, and ASN60 belonging to the ATPase site. On the other hand, the binding energy calculated using the MM/GBSA protocol highlighted Z2440107042 as the most promising inhibitor, with the best binding energy (-74.77 kcal/mol), suggesting that this molecule is a strong candidate for further biological studies.
Collapse
Affiliation(s)
- Didier Nivón-Ramirez
- Laboratorio de Fisicoquímica Analítica, Unidad de Investigación Multidisciplinaria, Facultad de Estudios Superiores Cuautitlán, Universidad Nacional Autónoma de México, Cuautitlán Izcalli, Estado de México, 54714, Mexico
| | - Emir A Galván-García
- Laboratorio de Fisicoquímica Analítica, Unidad de Investigación Multidisciplinaria, Facultad de Estudios Superiores Cuautitlán, Universidad Nacional Autónoma de México, Cuautitlán Izcalli, Estado de México, 54714, Mexico
| | - León D Ponce-Pérez
- Laboratorio de Fisicoquímica Analítica, Unidad de Investigación Multidisciplinaria, Facultad de Estudios Superiores Cuautitlán, Universidad Nacional Autónoma de México, Cuautitlán Izcalli, Estado de México, 54714, Mexico
| | - Rodolfo Gómez-Balderas
- Laboratorio de Fisicoquímica Analítica, Unidad de Investigación Multidisciplinaria, Facultad de Estudios Superiores Cuautitlán, Universidad Nacional Autónoma de México, Cuautitlán Izcalli, Estado de México, 54714, Mexico.
| |
Collapse
|
8
|
Oger C, Claeys Bouuaert C. SPO11 dimers are sufficient to catalyse DNA double-strand breaks in vitro. Nature 2025; 639:792-799. [PMID: 39972130 PMCID: PMC11922746 DOI: 10.1038/s41586-024-08574-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Accepted: 12/23/2024] [Indexed: 02/21/2025]
Abstract
SPO11 initiates meiotic recombination through the induction of programmed DNA double-strand breaks (DSBs)1,2, but this catalytic activity has never been reconstituted in vitro3,4. Here, using Mus musculus SPO11, we report a biochemical system that recapitulates all the hallmarks of meiotic DSB formation. We show that SPO11 catalyses break formation in the absence of any partners and remains covalently attached to the 5' broken strands. We find that target site selection by SPO11 is influenced by the sequence, bendability and topology of the DNA substrate, and provide evidence that SPO11 can reseal single-strand DNA breaks. In addition, we show that SPO11 is monomeric in solution and that cleavage requires dimerization for the reconstitution of two hybrid active sites. SPO11 and its partner TOP6BL form a 1:1 complex that catalyses DNA cleavage with an activity similar to that of SPO11 alone. However, this complex binds DNA ends with higher affinity, suggesting a potential role after cleavage. We propose a model in which additional partners of SPO11 required for DSB formation in vivo assemble biomolecular condensates that recruit SPO11-TOP6BL, enabling dimerization and cleavage. Our work establishes SPO11 dimerization as the fundamental mechanism that controls the induction of meiotic DSBs.
Collapse
Affiliation(s)
- Cédric Oger
- Louvain Institute of Biomolecular Science and Technology, Université Catholique de Louvain, Louvain-La-Neuve, Belgium
| | - Corentin Claeys Bouuaert
- Louvain Institute of Biomolecular Science and Technology, Université Catholique de Louvain, Louvain-La-Neuve, Belgium.
| |
Collapse
|
9
|
Klein T, Hodgskiss LH, Dreer M, Murrell JC, Hutchings MI, Schleper C, Lehtovirta‐Morley LE. Distinct Patterns of Antibiotic Sensitivities in Ammonia-Oxidising Archaea. Environ Microbiol 2025; 27:e70063. [PMID: 40070055 PMCID: PMC11897584 DOI: 10.1111/1462-2920.70063] [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: 11/25/2024] [Revised: 02/04/2025] [Accepted: 02/06/2025] [Indexed: 03/15/2025]
Abstract
Ammonia-oxidising archaea (AOA) are important microorganisms contributing towards the nitrogen flux in the environment. Unlike archaea from other major phyla, genetic tools are yet to be developed for the AOA, and identification of antibiotic resistance markers for selecting mutants is required for a genetic system. The aim of this study was to test the effects of selected antibiotics (hygromycin B, neomycin, apramycin, puromycin, novobiocin) on pure cultures of three well studied AOA strains, 'Candidatus Nitrosocosmicus franklandianus C13', Nitrososphaera viennensis EN76 and Nitrosopumilus maritimus SCM1. Puromycin, hygromycin B and neomycin inhibited some but not all tested archaeal strains. All strains were resistant to apramycin and inhibited by novobiocin to various degrees. As N. viennensis EN76 was relatively more resistant to the tested antibiotics, a wider range of concentrations and compounds (chloramphenicol, trimethoprim, statins) was tested against this strain. N. viennensis EN76 was inhibited by trimethoprim, but not by chloramphenicol, and growth recovered within days in the presence of simvastatin, suggesting either degradation of, or spontaneous resistance against, this compound. This study highlights the physiological differences between different genera of AOA and has identified new candidate antibiotics for selective enrichment and the development of selectable markers for genetic systems in AOA.
Collapse
Affiliation(s)
- Timothy Klein
- School of Biological SciencesUniversity of East AngliaNorwichUK
| | - Logan H. Hodgskiss
- Department of Functional and Evolutionary Ecology, Archaea Biology and Ecogenomics UnitUniversity of ViennaViennaAustria
| | - Max Dreer
- Department of Functional and Evolutionary Ecology, Archaea Biology and Ecogenomics UnitUniversity of ViennaViennaAustria
| | - J. Colin Murrell
- School of Environmental SciencesUniversity of East AngliaNorwichUK
| | | | - Christa Schleper
- Department of Functional and Evolutionary Ecology, Archaea Biology and Ecogenomics UnitUniversity of ViennaViennaAustria
| | | |
Collapse
|
10
|
Faheem I, Nagaraja V. Multifunctional Mycobacterial Topoisomerases with Distinctive Features. ACS Infect Dis 2025; 11:366-385. [PMID: 39825760 DOI: 10.1021/acsinfecdis.4c00880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2025]
Abstract
Tuberculosis (TB) continues to be a major cause of death worldwide despite having an effective combinatorial therapeutic regimen and vaccine. Being one of the most successful human pathogens, Mycobacterium tuberculosis retains the ability to adapt to diverse intracellular and extracellular environments encountered by it during infection, persistence, and transmission. Designing and developing new therapeutic strategies to counter the emergence of multidrug-resistant and extensively drug-resistant TB remains a major task. DNA topoisomerases make up a unique class of ubiquitous enzymes that ensure steady-state level supercoiling and solve topological problems occurring during DNA transactions in cells. They continue to be attractive targets for the discovery of novel classes of antibacterials and to develop better molecules from existing drugs by virtue of their reaction mechanism. The limited repertoire of topoisomerases in M. tuberculosis, key differences in their properties compared to topoisomerases from other bacteria, their essentiality for the pathogen's survival, and validation as candidates for drug discovery provide an opportunity to exploit them in drug discovery efforts. The present review provides insights into their organization, structure, function, and regulation to further efforts in targeting them for new inhibitor discovery. First, the structure and biochemical properties of DNA gyrase and Topoisomerase I (TopoI) of mycobacteria are described compared to the well-studied counterparts from other bacteria. Next, we provide an overview of known inhibitors of DNA gyrase and emerging novel bacterial topoisomerase inhibitors (NBTIs). We also provide an update on TopoI-specific compounds, highlighting mycobacteria-specific inhibitors.
Collapse
Affiliation(s)
- Iqball Faheem
- Department of Microbiology and Cell Biology, Indian Institute of Science, C.V. Raman Avenue, Bangalore 560012, India
| | - Valakunja Nagaraja
- Department of Microbiology and Cell Biology, Indian Institute of Science, C.V. Raman Avenue, Bangalore 560012, India
- Jawaharlal Nehru Centre for Advanced Scientific Research, Bangalore 560064, India
- Indian Institute of Science Education and Research, Bhopal 462066, India
| |
Collapse
|
11
|
Sofi FA, Mayank, Masoodi MH, Tabassum N. Recent advancements in the development of next-generation dual-targeting antibacterial agents. RSC Med Chem 2025:d4md00934g. [PMID: 40027349 PMCID: PMC11866402 DOI: 10.1039/d4md00934g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Accepted: 02/03/2025] [Indexed: 03/05/2025] Open
Abstract
DNA gyrase and topoisomerase IV are validated targets for developing dual-targeting antibacterial agents. The development of novel molecules targeting both enzymes has gained tremendous importance in circumventing the development of bacterial resistance. In the present review, we highlight the recent developments and discovery of dual-targeting inhibitors over the last five years. The structure-activity relationships, molecular docking analysis, and pharmacological activity are presented to facilitate the rational design and development of novel dual-targeting inhibitors to bridge the gap in antibiotic drug discovery.
Collapse
Affiliation(s)
- Firdoos Ahmad Sofi
- Department of Pharmaceutical Sciences, School of Applied Sciences and Technology, University of Kashmir Hazratbal Srinagar India
| | - Mayank
- Amity Institute of Pharmacy, Amity University Jaipur (Rajasthan) 303002 India
| | - Mubashir H Masoodi
- Department of Pharmaceutical Sciences, School of Applied Sciences and Technology, University of Kashmir Hazratbal Srinagar India
| | - Nahida Tabassum
- Department of Pharmaceutical Sciences, School of Applied Sciences and Technology, University of Kashmir Hazratbal Srinagar India
| |
Collapse
|
12
|
Kost C, Scheffer U, Kalden E, Göbel MW. Efficient Cleavage of pUC19 DNA by Tetraaminonaphthols. ChemistryOpen 2025; 14:e202400157. [PMID: 39460429 PMCID: PMC11808266 DOI: 10.1002/open.202400157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Indexed: 10/28/2024] Open
Abstract
In an attempt to create models of phosphodiesterases, we previously investigated bis(guanidinium) naphthols. Such metal-free anion receptors cleaved aryl phosphates and also plasmid DNA. Observed reaction rates, however, could not compete with those of highly reactive metal complexes. In the present study, we have replaced the guanidines by ethylene diamine side chains which accelerates the plasmid cleavage by compound 13 significantly (1 mM 13: t1/2=22 h). Further gains in reactivity are achieved by azo coupling of the naphthol unit. The electron accepting azo group decreases the pKa of the hydroxy group. It can also serve as a dye label and a handle for attaching DNA binding moieties. The resulting azo naphthol 17 not only nicks (1 mM 17: t1/2~1 h) but also linearizes pUC19 DNA. Although the high reactivity of 17 seems to result in part from aggregation, in the presence of EDTA azo naphthol 17 obeys first order kinetics (1 mM 17: t1/2=4.8 h), reacts four times faster than naphthol 13 and surpasses by far the former bis(guanidinium) naphthols 4 and 5.
Collapse
Affiliation(s)
- Catharina Kost
- Institut für Organische Chemie und Chemische BiologieGoethe-Universität, Frankfurt am MainMax-von-Laue-Str. 7D-60438Frankfurt am MainGermany
| | - Ute Scheffer
- Institut für Organische Chemie und Chemische BiologieGoethe-Universität, Frankfurt am MainMax-von-Laue-Str. 7D-60438Frankfurt am MainGermany
| | - Elisabeth Kalden
- Institut für Organische Chemie und Chemische BiologieGoethe-Universität, Frankfurt am MainMax-von-Laue-Str. 7D-60438Frankfurt am MainGermany
| | - Michael Wilhelm Göbel
- Institut für Organische Chemie und Chemische BiologieGoethe-Universität, Frankfurt am MainMax-von-Laue-Str. 7D-60438Frankfurt am MainGermany
| |
Collapse
|
13
|
Zhou T, Niu Y, Li Y. Advances in research on malignant tumors and targeted agents for TOP2A (Review). Mol Med Rep 2025; 31:50. [PMID: 39670307 DOI: 10.3892/mmr.2024.13415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 11/28/2024] [Indexed: 12/14/2024] Open
Abstract
The DNA topoisomerase isoform topoisomerase IIα (TOP2A) is essential for the condensation and segregation of cellular mitotic chromosomes and the structural maintenance. It has been demonstrated that TOP2A is highly expressed in various malignancies, including lung adenocarcinoma (LUAD), hepatocellular carcinoma (HCC) and breast cancer (BC), associating with poor prognosis and aggressive tumor behavior. Additionally, TOP2A has emerged as a promising target for cancer therapy, with widespread clinical application of associated chemotherapeutic agents. The present study explored the impact of TOP2A on malignant tumor growth and the advancements in research on its targeted drugs. The fundamental mechanisms of TOP2A have been detailed, its specific roles in tumor cells are analyzed, and its potential as a biomarker for tumor prognosis and therapeutic targeting is highlighted. Additionally, the present review compiles findings from the latest clinical trials of relevant targeted agents, information on newly developed inhibitors, and discusses future research directions and clinical application strategies in cancer therapy, aiming to propose novel ideas and methods.
Collapse
Affiliation(s)
- Tao Zhou
- Department of Hepatobiliary Surgery, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, Shanxi 030032, P.R. China
| | - Yiting Niu
- Department of Hepatobiliary Surgery, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, Shanxi 030032, P.R. China
| | - Yanjun Li
- Department of Hepatobiliary Surgery, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, Shanxi 030032, P.R. China
| |
Collapse
|
14
|
Lu S, Hou BL, Wang T, Ma K, Huang A, Wu X, Liang YN, Wang Z. Antitumor Effects of Tryptanthrin on Colorectal Cancer by Regulating the Mitogen-Activated Protein Kinase Signaling Pathway and Targeting Topo I and IDO1. ACS OMEGA 2025; 10:3206-3221. [PMID: 39895716 PMCID: PMC11780470 DOI: 10.1021/acsomega.4c11189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Accepted: 12/20/2024] [Indexed: 02/04/2025]
Abstract
Tryptanthrin (TRYP) is an indole quinazoline alkaloid with a range of pharmaceutical activities, but the specific mechanism of TRYP against colorectal cancer (CRC) remains obscure. The purpose of this study was to evaluate the antitumor effects of TRYP on CRC models both in vitro and in vivo and further analyze its concrete mechanisms. The results of the in vitro experiment show that TRYP effectively inhibited the proliferation and migration of SW620 cells, arrested the cell cycle at the S phase, and induced cell apoptosis. Deeply, TRYP dramatically increased the expression of Bax and cleaved caspase 3 while decreasing the expression of Bcl-2. The results of transcriptome sequencing implied that the inhibitory effects of TRYP were closely related to the mitogen-activated protein kinase (MAPK) signaling pathway, and the results of western blotting verified that TRYP could decrease the expression of p-Erk and increase the expression of p-p38 and p-Jnk. Besides, our results identified that topoisomerase I (Topo I) and indole amine 2,3-dioxygenase 1 (IDO1) were the targets of TRYP. In vivo, the results showed that different TRYP doses significantly inhibited tumor growth in mice, induced different degrees of necrosis in tumor tissues, decreased the expression level of Ki67 protein, and increased the apoptotic signal in tumor tissues. The findings demonstrated the inhibitory effects of TRYP on CRC, and the mechanisms were tightly connected to inhibiting the activity of Topo I and IDO1 and regulating the expression of the MAPK signaling pathway. Especially, it was first identified that TRYP could directly inhibit Topo I to arrest SW620 at the S phase. Therefore, this work established a scientific basis for the development of TRYP.
Collapse
Affiliation(s)
- Simeng Lu
- Co-construction
Collaborative Innovation Center of Chinese Medicine Resources Industrialization
by Shaanxi & Education Ministry, State Key Laboratory of Research
& Development of Characteristic Qin Medicine Resources (Cultivation), Shaanxi University of Chinese Medicine, Xianyang, Shaanxi712046, China
| | - Bao-Long Hou
- Co-construction
Collaborative Innovation Center of Chinese Medicine Resources Industrialization
by Shaanxi & Education Ministry, State Key Laboratory of Research
& Development of Characteristic Qin Medicine Resources (Cultivation), Shaanxi University of Chinese Medicine, Xianyang, Shaanxi712046, China
| | - Ting Wang
- Co-construction
Collaborative Innovation Center of Chinese Medicine Resources Industrialization
by Shaanxi & Education Ministry, State Key Laboratory of Research
& Development of Characteristic Qin Medicine Resources (Cultivation), Shaanxi University of Chinese Medicine, Xianyang, Shaanxi712046, China
| | - Keyu Ma
- Co-construction
Collaborative Innovation Center of Chinese Medicine Resources Industrialization
by Shaanxi & Education Ministry, State Key Laboratory of Research
& Development of Characteristic Qin Medicine Resources (Cultivation), Shaanxi University of Chinese Medicine, Xianyang, Shaanxi712046, China
| | - Anli Huang
- Co-construction
Collaborative Innovation Center of Chinese Medicine Resources Industrialization
by Shaanxi & Education Ministry, State Key Laboratory of Research
& Development of Characteristic Qin Medicine Resources (Cultivation), Shaanxi University of Chinese Medicine, Xianyang, Shaanxi712046, China
| | - Xue Wu
- Medical
Experiment Center, Shaanxi University of
Chinese Medicine, Xianyang, Shaanxi 712046, China
| | - Yan-Ni Liang
- Co-construction
Collaborative Innovation Center of Chinese Medicine Resources Industrialization
by Shaanxi & Education Ministry, State Key Laboratory of Research
& Development of Characteristic Qin Medicine Resources (Cultivation), Shaanxi University of Chinese Medicine, Xianyang, Shaanxi712046, China
| | - Zheng Wang
- Co-construction
Collaborative Innovation Center of Chinese Medicine Resources Industrialization
by Shaanxi & Education Ministry, State Key Laboratory of Research
& Development of Characteristic Qin Medicine Resources (Cultivation), Shaanxi University of Chinese Medicine, Xianyang, Shaanxi712046, China
| |
Collapse
|
15
|
Wang C, Xiu Y, Zhang Y, Wang Y, Xu J, Yu W, Xing D. Recent advances in biotin-based therapeutic agents for cancer therapy. NANOSCALE 2025; 17:1812-1873. [PMID: 39676680 DOI: 10.1039/d4nr03729d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
Biotin receptors, as biomarkers for cancer cells, are overexpressed in various tumor types. Compared to other vitamin receptors, such as folate receptors and vitamin B12 receptors, biotin receptor-based targeting strategies exhibit superior specificity and broader potential in treating aggressive cancers, including ovarian cancer, leukemia, colon cancer, breast cancer, kidney cancer, and lung cancer. These strategies promote biotin transport via receptor-mediated endocytosis, which is triggered upon ligand binding. Biotin, as the ligand of the biotin receptor, can be conjugated to anti-cancer drugs to form targeted therapies that bind to receptors overexpressed on tumor cells, thus increasing drug uptake. Despite these advantages, many candidate drugs have progressed slowly and remain in the preclinical stage, impeding clinical translation. This is mainly due to the effects of various conjugation methods and drug formulations on their functionality and efficacy. Therefore, developing novel biotin-based therapeutics is crucial. The innovation of this strategy lies in its multifunctionality-researchers can use different conjugation methods to design and synthesize these drugs, enabling precise targeting of various tumor types while minimizing toxicity to normal cells. These drugs include small-molecule-biotin conjugates (SMBCs) and nano-biotin conjugates (NBCs). This dual-platform approach represents a significant advancement in targeted therapy, offering unprecedented flexibility in drug design and delivery. Compared to chemotherapy drugs and traditional delivery systems, biotin-based drugs with tumor-specific targeting demonstrate enhanced targeting, improved efficacy, and reduced toxicity. This review examines strategies and applications for enhancing the delivery of chemotherapy drugs to cancer cells, highlighting the need for high-quality conjugates and strategies. It not only summarizes the latest progress but also provides key insights into how this emerging field could revolutionize personalized cancer treatment, especially in the context of precision medicine. Additionally, it offers perspectives on future research directions in this field.
Collapse
Affiliation(s)
- Chao Wang
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266071, China.
| | - Yutao Xiu
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266071, China.
| | - Yujing Zhang
- The Affiliated Cardiovascular Hospital of Qingdao University, Qingdao University, Qingdao, 266071, China
| | - Yanhong Wang
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266071, China.
| | - Jiazhen Xu
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266071, China.
| | - Wanpeng Yu
- Qingdao Medical College, Qingdao University, Qingdao 266071, China.
| | - Dongming Xing
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266071, China.
- The Affiliated Cardiovascular Hospital of Qingdao University, Qingdao University, Qingdao, 266071, China
- School of Life Sciences, Tsinghua University, Beijing, 100084, China
| |
Collapse
|
16
|
Feng W, Wang Q, Tan Y, Qiao J, Liu Q, Yang B, Yang S, Cui L. Early detection of anthracycline-induced cardiotoxicity. Clin Chim Acta 2025; 565:120000. [PMID: 39401650 DOI: 10.1016/j.cca.2024.120000] [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: 09/14/2024] [Revised: 10/09/2024] [Accepted: 10/10/2024] [Indexed: 10/19/2024]
Abstract
Although anthracyclines are important anticancer agents, their use is limited due to various adverse effects, particularly cardiac toxicity. Mechanisms underlying anthracycline-induced cardiotoxicity (AIC) are complex. Given the irreplaceable role of anthracyclines in treatment of malignancies and other serious diseases, early monitoring of AIC is paramount. In recent years, multiple studies have investigated various biomarkers for early detection of AIC. Currently, the two most common are cardiac troponin and B-type natriuretic peptide. In addition, a range of other molecules, including RNAs, myeloperoxidase (MPO), C-reactive protein (CRP), various genes, and others, also play roles in AIC prediction. Unfortunately, current research indicates a need to validate their sensitivity and specificity of these biomarkers especially in large study populations. In this review, we summarize the mechanisms and potential biomarkers of AIC, although some remain preliminary.
Collapse
Affiliation(s)
- Weimin Feng
- Department of Laboratory Medicine, Peking University Third Hospital, No. 49 Huayuan North Road, 100191, China; Institute of Medical Technology, Peking University Health Science Center, No. 38 Xueyuan Road, Beijing, 100191, China.
| | - Qingchen Wang
- Department of Laboratory Medicine, Peking University Third Hospital, No. 49 Huayuan North Road, 100191, China; Institute of Medical Technology, Peking University Health Science Center, No. 38 Xueyuan Road, Beijing, 100191, China.
| | - Yuan Tan
- Department of Laboratory Medicine, Peking University Third Hospital, No. 49 Huayuan North Road, 100191, China; Institute of Medical Technology, Peking University Health Science Center, No. 38 Xueyuan Road, Beijing, 100191, China.
| | - Jiao Qiao
- Department of Laboratory Medicine, Peking University Third Hospital, No. 49 Huayuan North Road, 100191, China; Institute of Medical Technology, Peking University Health Science Center, No. 38 Xueyuan Road, Beijing, 100191, China.
| | - Qi Liu
- Department of Laboratory Medicine, Peking University Third Hospital, No. 49 Huayuan North Road, 100191, China; Institute of Medical Technology, Peking University Health Science Center, No. 38 Xueyuan Road, Beijing, 100191, China.
| | - Boxin Yang
- Department of Laboratory Medicine, Peking University Third Hospital, No. 49 Huayuan North Road, 100191, China; Institute of Medical Technology, Peking University Health Science Center, No. 38 Xueyuan Road, Beijing, 100191, China.
| | - Shuo Yang
- Department of Laboratory Medicine, Peking University Third Hospital, No. 49 Huayuan North Road, 100191, China; Institute of Medical Technology, Peking University Health Science Center, No. 38 Xueyuan Road, Beijing, 100191, China.
| | - Liyan Cui
- Department of Laboratory Medicine, Peking University Third Hospital, No. 49 Huayuan North Road, 100191, China; Institute of Medical Technology, Peking University Health Science Center, No. 38 Xueyuan Road, Beijing, 100191, China.
| |
Collapse
|
17
|
Goh C, Bader A, Tran TA, Belotserkovskaya R, D’Alessandro G, Jackson S. TDP1 splice-site mutation causes HAP1 cell hypersensitivity to topoisomerase I inhibition. Nucleic Acids Res 2025; 53:gkae1163. [PMID: 39660638 PMCID: PMC11754736 DOI: 10.1093/nar/gkae1163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 10/25/2024] [Accepted: 11/12/2024] [Indexed: 12/12/2024] Open
Abstract
HAP1 is a near-haploid human cell line commonly used for mutagenesis and genome editing studies due to its hemizygous nature. We noticed an unusual hypersensitivity of HAP1 to camptothecin, an antineoplastic drug that stabilizes topoisomerase I cleavage complexes (TOP1ccs). We have attributed this hypersensitivity to a deficiency of TDP1, a key phosphodiesterase involved in resolving abortive TOP1ccs. Through whole-exome sequencing and subsequent restoration of TDP1 protein via CRISPR-Cas9 endogenous genome editing, we demonstrate that TDP1 deficiency and camptothecin hypersensitivity in HAP1 cells are a result of a splice-site mutation (TDP1 c.660-1G > A) that causes exon skipping and TDP1 loss of function. The lack of TDP1 in HAP1 cells should be considered when studying topoisomerase-associated DNA lesions and when generalizing mechanisms of DNA damage repair using HAP1 cells. Finally, we also report the generation of HAP1 STAR clones with restored TDP1 expression and function, which may be useful in further studies to probe cellular phenotypes relating to TOP1cc repair.
Collapse
Affiliation(s)
- Chen Gang Goh
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, CB2 0RE, UK
| | - Aldo S Bader
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, CB2 0RE, UK
| | - Tuan-Anh Tran
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, CB2 0RE, UK
| | | | | | - Stephen P Jackson
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, CB2 0RE, UK
| |
Collapse
|
18
|
Hall PM, Mayse LA, Bai L, Smolka MB, Pugh BF, Wang MD. High-Resolution Genome-Wide Maps Reveal Widespread Presence of Torsional Insulation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.10.11.617876. [PMID: 39416127 PMCID: PMC11482950 DOI: 10.1101/2024.10.11.617876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Torsional stress in chromatin plays a fundamental role in cellular functions, influencing key processes such as transcription, replication, and chromatin organization. Transcription and other processes may generate and be regulated by torsional stress. In the genome, the interplay of these processes creates complicated patterns of both positive (+) and negative (-) torsion. However, a challenge in generating an accurate torsion map is determining the zero-torsion baseline signal, which is conflated with chromatin accessibility. Here, we introduce a high-resolution method based on the intercalator trimethylpsoralen (TMP) to address this challenge. We describe a method to establish the zero-torsion baseline while preserving the chromatin state of the genome of S. cerevisiae. This approach enables both high-resolution mapping of accessibility and torsional stress in chromatin in the cell. Our analysis shows transcription-generated torsional domains consistent with the twin-supercoiled-domain model of transcription and suggests a role for torsional stress in recruiting topoisomerases and in regulating 3D genome architecture via cohesin. Significantly, we reveal that insulator sequence-specific transcription factors decouple torsion between divergent promoters, whereas torsion spreads between divergent promoters lacking these factors, suggesting that torsion serves as a regulatory mechanism in these regions. Although insulators are known to decouple gene expression, our finding provides a physical explanation of how such decoupling may occur. This new method provides a potential path forward for using TMP to measure torsional stress in the genome without the confounding contribution of accessibility in chromatin.
Collapse
Affiliation(s)
- Porter M. Hall
- Department of Physics & LASSP, Cornell University, Ithaca, NY 14853, USA
| | - Lauren A. Mayse
- Department of Physics & LASSP, Cornell University, Ithaca, NY 14853, USA
- Howard Hughes Medical Institute, Cornell University, Ithaca, NY 14853, USA
| | - Lu Bai
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, PA 16802, USA
- Department of Physics, Pennsylvania State University, University Park, PA 16802, USA
| | - Marcus B. Smolka
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY 14853, USA
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY 14853, USA
| | - B. Franklin Pugh
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY 14853, USA
| | - Michelle D. Wang
- Department of Physics & LASSP, Cornell University, Ithaca, NY 14853, USA
- Howard Hughes Medical Institute, Cornell University, Ithaca, NY 14853, USA
| |
Collapse
|
19
|
Fritsch C, Berger JM. Expression, Purification, and Measurement of DNA Supercoil Relaxation Activity by Eukaryotic Topoisomerase II. Methods Mol Biol 2025; 2928:75-87. [PMID: 40372638 DOI: 10.1007/978-1-0716-4550-5_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2025]
Abstract
Topoisomerases play a critical role in safeguarding the genome by resolving topological challenges that both result from and interfere with transcription and replication. These enzymes have been the subject of research not only for understanding their complex mechanisms of action, but also because of their therapeutic utility as drug targets. To gain a better understanding of topoisomerases and how they function, in vitro assays are necessary to measure their activity under various conditions. In this chapter, we describe the expression and purification of a eukaryotic type II topoisomerase (human topo IIα) from budding yeast cells and a gel-based assay that permits measurement of its ability to relax supercoiled DNA.
Collapse
Affiliation(s)
- Clark Fritsch
- Department of Biophysics and Biophysical Chemistry, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - James M Berger
- Department of Biophysics and Biophysical Chemistry, Johns Hopkins School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
20
|
Kumar A, Choubey P, Kaur H, Muralidhara P, Bansal K. Topoisomerase 1 is required for the development and function of thymus. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2025; 214:23-39. [PMID: 39959339 PMCID: PMC7617390 DOI: 10.1093/jimmun/vkae003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 10/16/2024] [Indexed: 02/18/2025]
Abstract
Thymus organogenesis is critical for proper maturation of developing T cells. In this study, we identified Topoisomerase 1 (Top1) as a novel gene involved in thymus development and function. We created a mouse line with deletion of Top1 in thymic epithelial cells (TECs) and our results demonstrate that biallelic loss of Top1 in TECs causes congenital thymic aplasia, precipitating T cell immunodeficiency. Transcriptomic analysis provides insights into the molecular mechanism of Top1 in thymus development as we identify key genes involved in thymus organogenesis as the transcriptional targets of Top1 in TECs. Analysis of peripheral immunological compartments revealed severe loss of αβ T cells complemented with a disproportionate accumulation of γδ T cells and myeloid cells upon deletion of Top1 in TECs. The residual αβ T cells in Top1 knock-out mice were effector and oligoclonal in nature highlighting their self-reactivity. These results reveal a previously unknown role of Top1 in thymus development and T cell homeostasis. We propose Top1 as a genetic target for altered thymic development and T cell lymphopenia.
Collapse
Affiliation(s)
- Amit Kumar
- Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bangalore, India
| | - Pallawi Choubey
- Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bangalore, India
| | - Harshdeep Kaur
- Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bangalore, India
| | - Prerana Muralidhara
- Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bangalore, India
| | - Kushagra Bansal
- Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bangalore, India
| |
Collapse
|
21
|
Martínez-García B, Díaz-Ingelmo O, Ayats-Fraile A, Roca J. Electrophoretic Analysis of DNA Supercoiling Activities. Methods Mol Biol 2025; 2881:259-270. [PMID: 39704948 DOI: 10.1007/978-1-0716-4280-1_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2024]
Abstract
DNA supercoiling in biological systems can occur via three mechanisms. The first is by the activity of DNA topoisomerases, such as DNA gyrases, that can increase or reduce the linking number of relaxed DNA (Lk0). The second is via DNA translocation motors, such as RNA and DNA polymerases, that produce twin supercoiled DNA domains: one positively supercoiled in front and one negatively supercoiled behind. The third is via molecular interactions that constrain DNA supercoils and thereby produce compensatory unconstrained ones. This chapter describes the use of agarose-gel electrophoresis to detect and quantify the DNA supercoils generated by these mechanisms. Particular emphasis is made on the preparation of a relaxed DNA plasmid as initial substrate that marks the position of Lk0 for calculating ΔLk.
Collapse
Affiliation(s)
- Belén Martínez-García
- DNA Topology Lab, Molecular Biology Institute of Barcelona (IBMB-CSIC), Barcelona, Spain
| | - Ofelia Díaz-Ingelmo
- DNA Topology Lab, Molecular Biology Institute of Barcelona (IBMB-CSIC), Barcelona, Spain
| | - Alba Ayats-Fraile
- DNA Topology Lab, Molecular Biology Institute of Barcelona (IBMB-CSIC), Barcelona, Spain
| | - Joaquim Roca
- DNA Topology Lab, Molecular Biology Institute of Barcelona (IBMB-CSIC), Barcelona, Spain.
| |
Collapse
|
22
|
Aldred KJ. Expression and One-Step Purification of His-tagged Bacterial Topoisomerase Subunits from Escherichia coli. Methods Mol Biol 2025; 2928:3-15. [PMID: 40372632 DOI: 10.1007/978-1-0716-4550-5_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2025]
Abstract
Purified topoisomerases are used in a variety of in vitro assays to study their biology as well as their interactions with drugs. The purified enzymes can also be used in structural studies and as standards to quantify immunoblots. The one-step purification of His-tagged subunits from only 500 mL of expression culture described here yields topoisomerase enzymes that are pure enough and concentrated enough for in vitro activity assays and blotting standards.
Collapse
Affiliation(s)
- Katie J Aldred
- Department of Biology, University of Evansville, Evansville, IN, USA.
| |
Collapse
|
23
|
Zhang M, Wang Z, Su Y, Yan W, Ouyang Y, Fan Y, Huang Y, Yang H. TDP1 represents a promising therapeutic target for overcoming tumor resistance to chemotherapeutic agents: progress and potential. Bioorg Chem 2025; 154:108072. [PMID: 39705934 DOI: 10.1016/j.bioorg.2024.108072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Revised: 12/08/2024] [Accepted: 12/15/2024] [Indexed: 12/23/2024]
Abstract
Tyrosyl-DNA phosphodiesterase 1 (TDP1) is an enzyme that plays a crucial role in repairing DNA lesions caused by the entrapment of DNA topoisomerase IB (TOP1)-DNA break-associated crosslinks. TDP1 inhibitors exhibit synergistic effects with TOP1 inhibitors in cancer cells, effectively overcoming resistance to TOP1 inhibitors. Therefore, this approach presents a promising strategy for reversing tumor resistance to TOP1 inhibitors. This review comprehensively outlines the structural and biological features of TDP1, the substrates involved in its catalytic hydrolysis, and its potential as a therapeutic target in oncology. Additionally, we summarize the various screening methods used to identify TDP1 inhibitors, alongside the latest advancements in TDP1 inhibitor research.
Collapse
Affiliation(s)
- Meimei Zhang
- School of Pharmacy, Ningxia Medical University, Yinchuan 750004, PR China
| | - Ziqiang Wang
- School of Pharmacy, Ningxia Medical University, Yinchuan 750004, PR China
| | - Yan Su
- School of Pharmacy, Ningxia Medical University, Yinchuan 750004, PR China
| | - Wenbo Yan
- School of Pharmacy, Ningxia Medical University, Yinchuan 750004, PR China
| | - Yifan Ouyang
- Fujian Key Laboratory of Toxicant and Drug Toxicology, School of Medicine, Ningde Normal University, Ningde, Fujian 352100, People's Republic of China.
| | - Yanru Fan
- School of Pharmacy, Ningxia Medical University, Yinchuan 750004, PR China; Key Laboratory of Protection, Development and Utilization of Medicinal Resources in Liupanshan Area Ministry of Education, Ningxia Medical University, Yinchuan 750004, PR China; Collaborative Innovation Center for Ningxia Characteristic Traditional Chinese Medicine by Ningxia Hui Autonomous Region & Education Ministry of P.R. China, Ningxia Characteristic Traditional Chinese Medicine Modern Engineering and Technique Research Center, Ningxia Key Laboratory of Drug Development and Generic Drug Research, Key Laboratory of Ningxia Ethnomedicine Modernization, Ministry of Education, Yinchuan 750004, PR China.
| | - Yu Huang
- School of Pharmacy, Ningxia Medical University, Yinchuan 750004, PR China; Key Laboratory of Protection, Development and Utilization of Medicinal Resources in Liupanshan Area Ministry of Education, Ningxia Medical University, Yinchuan 750004, PR China; Collaborative Innovation Center for Ningxia Characteristic Traditional Chinese Medicine by Ningxia Hui Autonomous Region & Education Ministry of P.R. China, Ningxia Characteristic Traditional Chinese Medicine Modern Engineering and Technique Research Center, Ningxia Key Laboratory of Drug Development and Generic Drug Research, Key Laboratory of Ningxia Ethnomedicine Modernization, Ministry of Education, Yinchuan 750004, PR China.
| | - Hao Yang
- School of Pharmacy, Ningxia Medical University, Yinchuan 750004, PR China; Key Laboratory of Protection, Development and Utilization of Medicinal Resources in Liupanshan Area Ministry of Education, Ningxia Medical University, Yinchuan 750004, PR China; Collaborative Innovation Center for Ningxia Characteristic Traditional Chinese Medicine by Ningxia Hui Autonomous Region & Education Ministry of P.R. China, Ningxia Characteristic Traditional Chinese Medicine Modern Engineering and Technique Research Center, Ningxia Key Laboratory of Drug Development and Generic Drug Research, Key Laboratory of Ningxia Ethnomedicine Modernization, Ministry of Education, Yinchuan 750004, PR China.
| |
Collapse
|
24
|
Gupta PO, Patil PS, Sekar N. Toxicity of anthraquinone derivatives in relation to non-linear optical properties and electron correlation. J Biomol Struct Dyn 2024:1-12. [PMID: 39681386 DOI: 10.1080/07391102.2024.2439582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 06/03/2024] [Indexed: 12/18/2024]
Abstract
1,4-Dialkylamino -5,8-dihydroxy anthraquinones are investigated using density functional theory (DFT) and time-dependent DFT (TD-DFT) for their growth inhibitory potential. The frontier molecular orbital shows that the electron density is located at the anthraquinone core and at the substituents NH and OH in both HOMO as well as in LUMO. The chemical potential and electrophilicity index showed a direct relation, while hardness and hyperhardness had an inverse association with an energy gap. The results of the molecular docking analysis revealed that the anthraquinone molecules have a high affinity for the primary targets of the DNA topoisomerase IIα enzyme. The docking results showed good binding ability with extremely energetically stable scores ranging from -8.9 to -7.6 kcal/mol. Electron correlation descriptors showed a direct link with NLO properties and toxicity.
Collapse
Affiliation(s)
- Puja O Gupta
- Department of Dyestuff Technology (Currently named Department of Speciality Chemicals Technology), Institute of Chemical Technology, Mumbai, Maharashtra, India
| | - Praful S Patil
- Department of Dyestuff Technology (Currently named Department of Speciality Chemicals Technology), Institute of Chemical Technology, Mumbai, Maharashtra, India
| | - Nagaiyan Sekar
- Department of Dyestuff Technology (Currently named Department of Speciality Chemicals Technology), Institute of Chemical Technology, Mumbai, Maharashtra, India
| |
Collapse
|
25
|
Royzenblat SK, Freddolino L. Spatio-temporal organization of the E. coli chromosome from base to cellular length scales. EcoSal Plus 2024; 12:eesp00012022. [PMID: 38864557 PMCID: PMC11636183 DOI: 10.1128/ecosalplus.esp-0001-2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Accepted: 04/17/2024] [Indexed: 06/13/2024]
Abstract
Escherichia coli has been a vital model organism for studying chromosomal structure, thanks, in part, to its small and circular genome (4.6 million base pairs) and well-characterized biochemical pathways. Over the last several decades, we have made considerable progress in understanding the intricacies of the structure and subsequent function of the E. coli nucleoid. At the smallest scale, DNA, with no physical constraints, takes on a shape reminiscent of a randomly twisted cable, forming mostly random coils but partly affected by its stiffness. This ball-of-spaghetti-like shape forms a structure several times too large to fit into the cell. Once the physiological constraints of the cell are added, the DNA takes on overtwisted (negatively supercoiled) structures, which are shaped by an intricate interplay of many proteins carrying out essential biological processes. At shorter length scales (up to about 1 kb), nucleoid-associated proteins organize and condense the chromosome by inducing loops, bends, and forming bridges. Zooming out further and including cellular processes, topological domains are formed, which are flanked by supercoiling barriers. At the megabase-scale both large, highly self-interacting regions (macrodomains) and strong contacts between distant but co-regulated genes have been observed. At the largest scale, the nucleoid forms a helical ellipsoid. In this review, we will explore the history and recent advances that pave the way for a better understanding of E. coli chromosome organization and structure, discussing the cellular processes that drive changes in DNA shape, and what contributes to compaction and formation of dynamic structures, and in turn how bacterial chromatin affects key processes such as transcription and replication.
Collapse
Affiliation(s)
- Sonya K. Royzenblat
- Cellular and Molecular Biology Program, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Lydia Freddolino
- Cellular and Molecular Biology Program, University of Michigan Medical School, Ann Arbor, Michigan, USA
- Department of Biological Chemistry, University of Michigan Medical School, Ann Arbor, Michigan, USA
- Department of Computational Medicine & Bioinformatics, University of Michigan Medical School, Ann Arbor, Michigan, USA
| |
Collapse
|
26
|
Blackwell AM, Jami-Alahmadi Y, Nasamu AS, Kudo S, Senoo A, Slam C, Tsumoto K, Wohlschlegel JA, Manuel Martinez Caaveiro J, Goldberg DE, Sigala PA. Malaria parasites require a divergent heme oxygenase for apicoplast gene expression and biogenesis. eLife 2024; 13:RP100256. [PMID: 39660822 PMCID: PMC11634067 DOI: 10.7554/elife.100256] [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] [Indexed: 12/12/2024] Open
Abstract
Malaria parasites have evolved unusual metabolic adaptations that specialize them for growth within heme-rich human erythrocytes. During blood-stage infection, Plasmodium falciparum parasites internalize and digest abundant host hemoglobin within the digestive vacuole. This massive catabolic process generates copious free heme, most of which is biomineralized into inert hemozoin. Parasites also express a divergent heme oxygenase (HO)-like protein (PfHO) that lacks key active-site residues and has lost canonical HO activity. The cellular role of this unusual protein that underpins its retention by parasites has been unknown. To unravel PfHO function, we first determined a 2.8 Å-resolution X-ray structure that revealed a highly α-helical fold indicative of distant HO homology. Localization studies unveiled PfHO targeting to the apicoplast organelle, where it is imported and undergoes N-terminal processing but retains most of the electropositive transit peptide. We observed that conditional knockdown of PfHO was lethal to parasites, which died from defective apicoplast biogenesis and impaired isoprenoid-precursor synthesis. Complementation and molecular-interaction studies revealed an essential role for the electropositive N-terminus of PfHO, which selectively associates with the apicoplast genome and enzymes involved in nucleic acid metabolism and gene expression. PfHO knockdown resulted in a specific deficiency in levels of apicoplast-encoded RNA but not DNA. These studies reveal an essential function for PfHO in apicoplast maintenance and suggest that Plasmodium repurposed the conserved HO scaffold from its canonical heme-degrading function in the ancestral chloroplast to fulfill a critical adaptive role in organelle gene expression.
Collapse
Affiliation(s)
- Amanda Mixon Blackwell
- Department of Biochemistry, University of Utah School of MedicineSalt Lake CityUnited States
| | - Yasaman Jami-Alahmadi
- Department of Biological Chemistry, University of California, Los AngelesLos AngelesUnited States
| | - Armiyaw S Nasamu
- Departments of Medicine and Molecular Microbiology, Washington University School of MedicineSt. LouisUnited States
| | - Shota Kudo
- Department of Chemistry & Biotechnology, The University of TokyoTokyoJapan
| | - Akinobu Senoo
- Department of Protein Drug Discovery, Graduate School of Pharmaceutical Sciences, Kyushu UniversityFukuokaJapan
| | - Celine Slam
- Department of Biochemistry, University of Utah School of MedicineSalt Lake CityUnited States
| | - Kouhei Tsumoto
- Department of Chemistry & Biotechnology, The University of TokyoTokyoJapan
- Department of Bioengineering, University of TokyoTokyoJapan
| | - James A Wohlschlegel
- Department of Biological Chemistry, University of California, Los AngelesLos AngelesUnited States
| | | | - Daniel E Goldberg
- Departments of Medicine and Molecular Microbiology, Washington University School of MedicineSt. LouisUnited States
| | - Paul A Sigala
- Department of Biochemistry, University of Utah School of MedicineSalt Lake CityUnited States
- Departments of Medicine and Molecular Microbiology, Washington University School of MedicineSt. LouisUnited States
| |
Collapse
|
27
|
Zhang H, Tian L, Ma Y, Xu J, Bai T, Wang Q, Liu X, Guo L. Not only the top: Type I topoisomerases function in multiple tissues and organs development in plants. J Adv Res 2024:S2090-1232(24)00588-5. [PMID: 39662729 DOI: 10.1016/j.jare.2024.12.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 11/24/2024] [Accepted: 12/07/2024] [Indexed: 12/13/2024] Open
Abstract
BACKGROUND DNA topoisomerases (TOPs) are essential components in a diverse range of biological processes including DNA replication, transcription and genome integrity. Although the functions and mechanisms of TOPs, particularly type I TOP (TOP1s), have been extensively studied in bacteria, yeast and animals, researches on these proteins in plants have only recently commenced. AIM OF REVIEW In this review, the function and mechanism studies of TOP1s in plants and the structural biology of plant TOP1 are presented, providing readers with a comprehensive understanding of the current research status of this essential enzyme.The future research directions for exploring the working mechanism of plant TOP1s are also discussed. KEY SCIENTIFIC CONCEPTS OF REVIEW Over the past decade, it has been discovered TOP1s play a vital role in multiphasic processes of plant development, such as maintaining meristem activity, gametogenesis, flowering time, gravitropic response and so on. Plant TOP1s affects gene transcription by modulating chromatin status, including chromatin accessibility, DNA/RNA structure, and nucleosome positioning. However, the function and mechanism of this vital enzyme is poorly summarized although it has been systematically summarized in other species. This review summarized the research progresses of plant TOP1s according to the diverse functions and working mechanism in different tissues.
Collapse
Affiliation(s)
- Hao Zhang
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Hebei Research Center of the Basic Discipline of Cell Biology, Hebei Collaboration Innovation Center for Cell Signaling and Environmental Adaptation, Hebei Key Laboratory of Molecular and Cellular Biology, College of Life Sciences, Hebei Normal University, 050024, Shijiazhuang, China.
| | - Lirong Tian
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Hebei Research Center of the Basic Discipline of Cell Biology, Hebei Collaboration Innovation Center for Cell Signaling and Environmental Adaptation, Hebei Key Laboratory of Molecular and Cellular Biology, College of Life Sciences, Hebei Normal University, 050024, Shijiazhuang, China.
| | - Yuru Ma
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Hebei Research Center of the Basic Discipline of Cell Biology, Hebei Collaboration Innovation Center for Cell Signaling and Environmental Adaptation, Hebei Key Laboratory of Molecular and Cellular Biology, College of Life Sciences, Hebei Normal University, 050024, Shijiazhuang, China.
| | - Jiahui Xu
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Hebei Research Center of the Basic Discipline of Cell Biology, Hebei Collaboration Innovation Center for Cell Signaling and Environmental Adaptation, Hebei Key Laboratory of Molecular and Cellular Biology, College of Life Sciences, Hebei Normal University, 050024, Shijiazhuang, China.
| | - Tianyu Bai
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Hebei Research Center of the Basic Discipline of Cell Biology, Hebei Collaboration Innovation Center for Cell Signaling and Environmental Adaptation, Hebei Key Laboratory of Molecular and Cellular Biology, College of Life Sciences, Hebei Normal University, 050024, Shijiazhuang, China.
| | - Qian Wang
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Hebei Research Center of the Basic Discipline of Cell Biology, Hebei Collaboration Innovation Center for Cell Signaling and Environmental Adaptation, Hebei Key Laboratory of Molecular and Cellular Biology, College of Life Sciences, Hebei Normal University, 050024, Shijiazhuang, China.
| | - Xigang Liu
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Hebei Research Center of the Basic Discipline of Cell Biology, Hebei Collaboration Innovation Center for Cell Signaling and Environmental Adaptation, Hebei Key Laboratory of Molecular and Cellular Biology, College of Life Sciences, Hebei Normal University, 050024, Shijiazhuang, China.
| | - Lin Guo
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Hebei Research Center of the Basic Discipline of Cell Biology, Hebei Collaboration Innovation Center for Cell Signaling and Environmental Adaptation, Hebei Key Laboratory of Molecular and Cellular Biology, College of Life Sciences, Hebei Normal University, 050024, Shijiazhuang, China.
| |
Collapse
|
28
|
Ren Q, Chen G, Wan Q, Xiao L, Zhang Z, Feng Y. Unravelling the role of natural and synthetic products as DNA topoisomerase inhibitors in hepatocellular carcinoma. Bioorg Chem 2024; 153:107860. [PMID: 39442463 DOI: 10.1016/j.bioorg.2024.107860] [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: 05/17/2024] [Revised: 09/11/2024] [Accepted: 09/29/2024] [Indexed: 10/25/2024]
Abstract
Topoisomerase is a ubiquitous enzyme in the control of DNA chain topology. There have been extensive research on topoisomerase inhibitors derived from natural sources, which act as partial inducers of tumor cell apoptosis. However, their specific efficacy in treating hepatocellular carcinoma is relatively unexplored. Hence, this comprehensive review focuses on the structural characteristics and anti-cancer properties of topoisomerase inhibitors in hepatocellular carcinoma. Furthermore, this review is also elucidating the mechanism of action, structure-activity relationships, therapeutic limitations, stage of clinical trials of described classes of natural bioactive compounds as well as their potential application in cancer chemotherapies. This broad understanding of topoisomerase medical biology will provide indispensable framework for enhancing the efficiency of rational anti-hepatocellular carcinoma drug discovery.
Collapse
Affiliation(s)
- Qing Ren
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR 999077, China
| | - Guoming Chen
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR 999077, China
| | - Qi Wan
- Jiangxi University of Chinese Medicine, Nanchang 330006, China
| | - Liangman Xiao
- Acupuncture Rehabilitation Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Zhitong Zhang
- Fourth Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Yibin Feng
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR 999077, China.
| |
Collapse
|
29
|
Kitaoka M, Yamashita YM. Running the gauntlet: challenges to genome integrity in spermiogenesis. Nucleus 2024; 15:2339220. [PMID: 38594652 PMCID: PMC11005813 DOI: 10.1080/19491034.2024.2339220] [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: 02/23/2024] [Accepted: 04/02/2024] [Indexed: 04/11/2024] Open
Abstract
Species' continuity depends on gametogenesis to produce the only cell types that can transmit genetic information across generations. Spermiogenesis, which encompasses post-meiotic, haploid stages of male gametogenesis, is a process that leads to the formation of sperm cells well-known for their motility. Spermiogenesis faces three major challenges. First, after two rounds of meiotic divisions, the genome lacks repair templates (no sister chromatids, no homologous chromosomes), making it incredibly vulnerable to any genomic insults over an extended time (typically days-weeks). Second, the sperm genome becomes transcriptionally silent, making it difficult to respond to new perturbations as spermiogenesis progresses. Third, the histone-to-protamine transition, which is essential to package the sperm genome, counterintuitively involves DNA break formation. How spermiogenesis handles these challenges remains poorly understood. In this review, we discuss each challenge and their intersection with the biology of protamines. Finally, we discuss the implication of protamines in the process of evolution.
Collapse
Affiliation(s)
- Maiko Kitaoka
- Whitehead Institute for Biomedical Research and Howard Hughes Medical Institute, Cambridge, MA, USA
| | - Yukiko M. Yamashita
- Whitehead Institute for Biomedical Research and Howard Hughes Medical Institute, Cambridge, MA, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| |
Collapse
|
30
|
Kim Y, Kim S, Heo K, Lee S. Single-molecule FRET-based approach for protein-targeted drug discovery. Mol Cells 2024; 47:100150. [PMID: 39549747 DOI: 10.1016/j.mocell.2024.100150] [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: 10/24/2024] [Revised: 11/08/2024] [Accepted: 11/08/2024] [Indexed: 11/18/2024] Open
Abstract
Many therapeutic drugs target various proteins involved in diverse biological processes. Among these proteins, type II topoisomerases are critical targets for anticancer and antibacterial chemotherapies, yet the action mechanisms of many type II topoisomerase-targeting drugs have not been fully elucidated. In this regard, the development of rapid and accurate methods to identify the mode of action of potential drug candidates is crucial to improve the efficiency of drug screening and discovery. Here, using type II topoisomerase as a model system, we present a single-molecule fluorescence resonance energy transfer-based drug screening method capable of delineating when and how the drug candidates participate in the entire reaction steps of the target protein. This unique capability has been demonstrated to be applicable to the identification of representative types of widely prescribed drugs targeting type II topoisomerase: etoposide which stabilizes the enzyme-DNA cleavage complex, and bisdioxopiperazines (ICRF-I93) which lock the N-terminal gate of the enzyme into the closed state. Based on this demonstration experiment, we expect that our proposed method will be extended to broad applications in the screening of potent drugs targeting various proteins.
Collapse
Affiliation(s)
- Yuyoung Kim
- Department of Medical Life Sciences, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; Department of Medical Sciences, Graduate School of The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Surim Kim
- Department of Medical Life Sciences, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; Department of Medical Sciences, Graduate School of The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Kang Heo
- Strategic Development Team, Vieworks Company, Anyang-si, Gyeonggi-do 14055, Republic of Korea
| | - Sanghwa Lee
- Department of Medical Life Sciences, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; Department of Medical Sciences, Graduate School of The Catholic University of Korea, Seoul 06591, Republic of Korea.
| |
Collapse
|
31
|
Mukhametzyanova L, Schmitt LT, Torres-Rivera J, Rojo-Romanos T, Lansing F, Paszkowski-Rogacz M, Hollak H, Brux M, Augsburg M, Schneider PM, Buchholz F. Activation of recombinases at specific DNA loci by zinc-finger domain insertions. Nat Biotechnol 2024; 42:1844-1854. [PMID: 38297187 PMCID: PMC11631766 DOI: 10.1038/s41587-023-02121-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 12/22/2023] [Indexed: 02/02/2024]
Abstract
Recombinases have several potential advantages as genome editing tools compared to nucleases and other editing enzymes, but the process of engineering them to efficiently recombine predetermined DNA targets demands considerable investment of time and labor. Here we sought to harness zinc-finger DNA-binding domains (ZFDs) to program recombinase binding by developing fusions, in which ZFDs are inserted into recombinase coding sequences. By screening libraries of hybrid proteins, we optimized the insertion site, linker length, spacing and ZFD orientation and generated Cre-type recombinases that remain dormant unless the insertionally fused ZFD binds its target site placed in the vicinity of the recombinase binding site. The developed fusion improved targeted editing efficiencies of recombinases by four-fold and abolished measurable off-target activity in mammalian cells. The ZFD-dependent activity is transferable to a recombinase with relaxed specificity, providing the means for developing fully programmable recombinases. Our engineered recombinases provide improved genome editing tools with increased precision and efficiency.
Collapse
Affiliation(s)
- Liliya Mukhametzyanova
- Medical Systems Biology, Medical Faculty, Technical University Dresden, Dresden, Germany
| | - Lukas Theo Schmitt
- Medical Systems Biology, Medical Faculty, Technical University Dresden, Dresden, Germany
- Seamless Therapeutics GmbH, Dresden, Germany
| | - Julia Torres-Rivera
- Medical Systems Biology, Medical Faculty, Technical University Dresden, Dresden, Germany
| | - Teresa Rojo-Romanos
- Medical Systems Biology, Medical Faculty, Technical University Dresden, Dresden, Germany
- Seamless Therapeutics GmbH, Dresden, Germany
| | - Felix Lansing
- Medical Systems Biology, Medical Faculty, Technical University Dresden, Dresden, Germany
- Seamless Therapeutics GmbH, Dresden, Germany
| | | | - Heike Hollak
- Medical Systems Biology, Medical Faculty, Technical University Dresden, Dresden, Germany
- Seamless Therapeutics GmbH, Dresden, Germany
| | - Melanie Brux
- Medical Systems Biology, Medical Faculty, Technical University Dresden, Dresden, Germany
| | - Martina Augsburg
- Medical Systems Biology, Medical Faculty, Technical University Dresden, Dresden, Germany
| | - Paul Martin Schneider
- Medical Systems Biology, Medical Faculty, Technical University Dresden, Dresden, Germany
- Seamless Therapeutics GmbH, Dresden, Germany
| | - Frank Buchholz
- Medical Systems Biology, Medical Faculty, Technical University Dresden, Dresden, Germany.
| |
Collapse
|
32
|
Rusu A, Oancea OL, Tanase C, Uncu L. Unlocking the Potential of Pyrrole: Recent Advances in New Pyrrole-Containing Compounds with Antibacterial Potential. Int J Mol Sci 2024; 25:12873. [PMID: 39684580 PMCID: PMC11640851 DOI: 10.3390/ijms252312873] [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: 10/10/2024] [Revised: 11/25/2024] [Accepted: 11/26/2024] [Indexed: 12/18/2024] Open
Abstract
Nitrogen heterocycles are valuable structural elements in the molecules of antibacterial drugs approved and used to treat bacterial infections. Pyrrole is a five-atom heterocycle found in many natural compounds with biological activity, including antibacterial activity. Numerous compounds are being develop based on the pyrrole heterocycle as new potential antibacterial drugs. Due to the phenomenon of antibacterial resistance, there is a continuous need to create new effective antibacterials. In the scientific literature, we have identified the most relevant studies that aim to develop new compounds, such as pyrrole derivatives, that are proven to have antibacterial activity. Nature is an endless reservoir of inspiration for designing new compounds based on the structure of pyrrole heterocycles such as calcimycin, lynamycins, marinopyrroles, nargenicines, phallusialides, and others. However, many other synthetic compounds based on the pyrrole heterocycle have been developed and can be optimized in the future. The identified compounds were classified according to the type of chemical structure. The chemical structure-activity relationships, mechanisms of action, and antibacterial effectiveness of the most valuable compounds were highlighted. This review highlights scientific progress in designing new pyrrole-containing compounds and provides examples of lead compounds that can be successfully optimized further.
Collapse
Affiliation(s)
- Aura Rusu
- Pharmaceutical and Therapeutic Chemistry Department, Faculty of Pharmacy, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540142 Targu Mures, Romania;
| | - Octavia-Laura Oancea
- Organic Chemistry Department, Faculty of Pharmacy, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540142 Targu Mures, Romania;
| | - Corneliu Tanase
- Pharmaceutical Botany Department, Faculty of Pharmacy, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540142 Targu Mures, Romania
| | - Livia Uncu
- Scientific Center for Drug Research, Pharmaceutical and Toxicological Chemistry Department, “Nicolae Testemitanu” State University of Medicine and Pharmacy, 165 Bd. Stefan Cel Mare si Sfant, MD-2004 Chisinau, Moldova;
| |
Collapse
|
33
|
Kassab AE, Gomaa RM, Gedawy EM. Drug repurposing of fluoroquinolones as anticancer agents in 2023. RSC Adv 2024; 14:37114-37130. [PMID: 39569131 PMCID: PMC11578043 DOI: 10.1039/d4ra03571b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 11/06/2024] [Indexed: 11/22/2024] Open
Abstract
Drug developers are currently focusing on investigating alternative strategies, such as "drug repositioning", to address issues associated with productivity, regulatory obstacles, and the steadily rising cost of pharmaceuticals. Repositioning is the best strategy to stop searching for new drugs because it takes less time and money to investigate new indications for already approved or unsuccessful drugs. Although there are several potent Topo II inhibitors available on the market as important drugs used in the therapy of many types of cancer, more may be required in the future. The current inhibitors have drawbacks including acquired resistance and unfavorable side effects such as cardiotoxicity and subsequent malignancy. A substantial body of research documented the cytotoxic potential of experimental fluoroquinolones (FQs) on tumor cell lines and their remarkable efficacy against eukaryotic Topo II in addition to optimized physical and metabolic characteristics. The FQ scaffold has a unique ability to potentially resolve every major issue associated with traditional Topo II inhibitors while maintaining a highly desirable profile in crucial drug-likeness parameters; therefore, there is a significant chance that FQs will be repositioned as anticancer candidates. This review offers a summary of the most recent research on the anticancer potential of FQs that was published in 2023. Along with discussing structural activity relationship studies and the mechanism underlying their antiproliferative activity, this review aims to provide up-to-date information that will spur the development of more potent FQs as viable cancer treatment candidates.
Collapse
Affiliation(s)
- Asmaa E Kassab
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Cairo University Kasr El-Aini Street, P. O. Box 11562 Cairo Egypt
| | - Rania M Gomaa
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Mansoura University P. O. Box 35516 Mansoura Egypt
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Pharmaceutical Industries, Badr University in Cairo (BUC) Badr City, P. O. Box 11829 Cairo Egypt +2023635140 +2023639307
| | - Ehab M Gedawy
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Cairo University Kasr El-Aini Street, P. O. Box 11562 Cairo Egypt
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Pharmaceutical Industries, Badr University in Cairo (BUC) Badr City, P. O. Box 11829 Cairo Egypt +2023635140 +2023639307
| |
Collapse
|
34
|
Yesodi D, Katz A, Weizmann Y. Advancing Topoisomerase Research Using DNA Nanotechnology. SMALL METHODS 2024:e2401113. [PMID: 39526512 DOI: 10.1002/smtd.202401113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Revised: 10/30/2024] [Indexed: 11/16/2024]
Abstract
In this Perspective, the use of DNA nanotechnology is explored as a powerful tool for studying a family of enzymes known as topoisomerases. These enzymes regulate DNA topology within a living cell and play a major role in the pharmaceutical field, serving as anti-cancer and anti-bacterial targets. This Perspective will provide a short historical overview of the methods employed in studying these enzymes and emphasizing recent advancements in assays using DNA nanotechnology. These innovations have substantially improved accuracy and expanded the understanding of enzyme activity. This perspective will showcase the versatile utility of DNA nanotechnology in advancing scientific knowledge and its application in exploring new drug candidates, particularly in the study of topoisomerase enzymes.
Collapse
Affiliation(s)
- Doron Yesodi
- Department of Chemistry, Ben-Gurion University of the Negev, Beer-Sheva, 84105, Israel
| | - Adi Katz
- Department of Chemistry, Ben-Gurion University of the Negev, Beer-Sheva, 84105, Israel
| | - Yossi Weizmann
- Department of Chemistry, Ben-Gurion University of the Negev, Beer-Sheva, 84105, Israel
- Ilse Katz Institute for Nanotechnology Science, Ben-Gurion University of the Negev, Beer-Sheva, 84105, Israel
- Goldman Sonnenfeldt School of Sustainability and Climate Change, Ben-Gurion University of the Negev, Beer-Sheva, 84105, Israel
| |
Collapse
|
35
|
Bondarev AD, Jonsson J, Chubarev VN, Tarasov VV, Lagunas-Rangel FA, Schiöth HB. Recent developments of topoisomerase inhibitors: Clinical trials, emerging indications, novel molecules and global sales. Pharmacol Res 2024; 209:107431. [PMID: 39307213 DOI: 10.1016/j.phrs.2024.107431] [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: 06/14/2024] [Revised: 09/09/2024] [Accepted: 09/19/2024] [Indexed: 11/11/2024]
Abstract
The nucleic acid topoisomerases (TOP) are an evolutionary conserved mechanism to solve topological problems within DNA and RNA that have been historically well-established as a chemotherapeutic target. During investigation of trends within clinical trials, we have identified a very high number of clinical trials involving TOP inhibitors, prompting us to further evaluate the current status of this class of therapeutic agents. In total, we have identified 233 unique molecules with TOP-inhibiting activity. In this review, we provide an overview of the clinical drug development highlighting advances in current clinical uses and discussing novel drugs and indications under development. A wide range of bacterial infections, along with solid and hematologic neoplasms, represent the bulk of clinically approved indications. Negative ADR profile and drug resistance among the antibacterial TOP inhibitors and anthracycline-mediated cardiotoxicity in the antineoplastic TOP inhibitors are major points of concern, subject to continuous research efforts. Ongoing development continues to focus on bacterial infections and cancer; however, there is a degree of diversification in terms of novel drug classes and previously uncovered indications, such as glioblastoma multiforme or Clostridium difficile infections. Preclinical studies show potential in viral, protozoal, parasitic and fungal infections as well and suggest the emergence of a novel target, TOP IIIβ. We predict further growth and diversification of the field thanks to the large number of experimental TOP inhibitors emerging.
Collapse
Affiliation(s)
- Andrey D Bondarev
- Department of Surgical Sciences, Functional Pharmacology and Neuroscience, Uppsala University, Uppsala, Sweden
| | - Jörgen Jonsson
- Department of Surgical Sciences, Functional Pharmacology and Neuroscience, Uppsala University, Uppsala, Sweden
| | - Vladimir N Chubarev
- Advanced Molecular Technologies, Limited Liability Company (LLC), Moscow 354340, Russia
| | - Vadim V Tarasov
- Advanced Molecular Technologies, Limited Liability Company (LLC), Moscow 354340, Russia
| | - Francisco Alejandro Lagunas-Rangel
- Department of Surgical Sciences, Functional Pharmacology and Neuroscience, Uppsala University, Uppsala, Sweden; Laboratory of Pharmaceutical Pharmacology, Latvian Institute of Organic Synthesis, Riga, Latvia.
| | - Helgi B Schiöth
- Department of Surgical Sciences, Functional Pharmacology and Neuroscience, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
36
|
van Oostveen WM, Huizinga TWJ, Fehres CM. Pathogenic role of anti-nuclear autoantibodies in systemic sclerosis: Insights from other rheumatic diseases. Immunol Rev 2024; 328:265-282. [PMID: 39248128 PMCID: PMC11659924 DOI: 10.1111/imr.13390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/10/2024]
Abstract
Systemic sclerosis (SSc) is a severe autoimmune disease characterized by vasculopathy, fibrosis, and dysregulated immunity, with hallmark autoantibodies targeting nuclear antigens such as centromere protein (ACA) and topoisomerase I (ATA). These autoantibodies are highly prevalent and disease-specific, rarely coexisting, thus serving as crucial biomarkers for SSc diagnosis. Despite their diagnostic value, their roles in SSc pathogenesis remain unclear. This review summarizes current literature on ACA and ATA in SSc, comparing them to autoantibodies in other rheumatic diseases to elucidate their potential pathogenic roles. Similarities are drawn with anti-citrullinated protein antibodies (ACPA) in rheumatoid arthritis, particularly regarding disease specificity and minimal pathogenic impact of antigen binding. In addition, differences between ANA and ACPA in therapeutic responses and Fab glycosylation patterns are reviewed. While ACA and ATA are valuable for disease stratification and monitoring activity, understanding their origins and the associated B cell responses is critical for advancing therapeutic strategies for SSc.
Collapse
Affiliation(s)
| | - Tom W. J. Huizinga
- Department of RheumatologyLeiden University Medical CenterLeidenThe Netherlands
| | - Cynthia M. Fehres
- Department of RheumatologyLeiden University Medical CenterLeidenThe Netherlands
| |
Collapse
|
37
|
Lee CC, Chang CH, Huang YC, Shih TL. Novel 1,8-Naphthalimide Derivatives Inhibit Growth and Induce Apoptosis in Human Glioblastoma. Int J Mol Sci 2024; 25:11593. [PMID: 39519145 PMCID: PMC11546702 DOI: 10.3390/ijms252111593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Revised: 10/18/2024] [Accepted: 10/22/2024] [Indexed: 11/16/2024] Open
Abstract
Given the rapid advancement of functional 1,8-Naphthalimide derivatives in anticancer research, we synthesized these two novel naphthalimide derivatives with diverse substituents and investigated the effect on glioblastoma multiforme (GBM) cells. Cytotoxicity, apoptosis, cell cycle, topoisomerase II and Western blotting assays were evaluated for these compounds against GBM in vitro. A human GBM xenograft mouse model established by subcutaneously injecting U87-MG cells and the treatment responses were assessed. Both compounds 3 and 4 exhibited significant antiproliferative activities, inducing apoptosis and cell death. Only compound 3 notably induced G2/M phase cell cycle arrest in the U87-MG GBM cells. Both compounds inhibited DNA topoisomerase II activity, resulting in DNA damage. The in vivo antiproliferative potential of compound 3 was further validated in a U87-MG GBM xenograft mouse model, without any discernible loss of body weight or kidney toxicity noted. This study presents novel findings demonstrating that 1,8-Naphthalimide derivatives exhibited significant GBM cell suppression in vitro and in vivo without causing adverse effects on body weight or kidney function. Further experiments, including investigations into mechanisms and pathways, as well as preclinical studies on the pharmacokinetics and pharmacodynamics, may be instrumental to the development of a new anti-GBM compound.
Collapse
Affiliation(s)
- Cheng-Chi Lee
- Department of Neurosurgery, Chang Gung Memorial Hospital, Linkou Medical Center, Chang Gung University, Taoyuan 333423, Taiwan;
| | - Chuan-Hsin Chang
- Department of Research, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City 231016, Taiwan;
- Research Center for Chinese Herbal Medicine, Graduate Institute of Healthy Industry Technology, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan 333324, Taiwan
| | - Yin-Cheng Huang
- Department of Neurosurgery, Chang Gung Memorial Hospital, Linkou Medical Center, Chang Gung University, Taoyuan 333423, Taiwan;
| | - Tzenge-Lien Shih
- Department of Chemistry, Tamkang University, Tamsui Dist., New Taipei City 251301, Taiwan
| |
Collapse
|
38
|
Bagattin A, Tammaccaro SL, Chiral M, Makinistoglu MP, Zimmermann N, Lerner J, Garbay S, Kuperwasser N, Pontoglio M. HNF1β bookmarking involves Topoisomerase 1 activation and DNA topology relaxation in mitotic chromatin. Cell Rep 2024; 43:114805. [PMID: 39388351 DOI: 10.1016/j.celrep.2024.114805] [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: 12/04/2023] [Revised: 07/03/2024] [Accepted: 09/12/2024] [Indexed: 10/12/2024] Open
Abstract
HNF1β (HNF1B) is a transcription factor frequently mutated in patients with developmental renal disease. It binds to mitotic chromatin and reactivates gene expression after mitosis, a phenomenon referred to as bookmarking. Using a crosslinking method that circumvents the artifacts of formaldehyde, we demonstrate that HNF1β remains associated with chromatin in a sequence-specific way in both interphase and mitosis. We identify an HNF1β-interacting protein, BTBD2, that enables the interaction and activation of Topoisomerase 1 (TOP1) exclusively during mitosis. Our study identifies a shared microhomology domain between HNF1β and TOP1, where a mutation, found in "maturity onset diabetes of the young" patients, disrupts their interaction. Importantly, HNF1β recruits TOP1 and induces DNA relaxation around HNF1β mitotic chromatin sites, elucidating its crucial role in chromatin remodeling and gene reactivation after mitotic exit. These findings shed light on how HNF1β reactivates target gene expression after mitosis, providing insights into its crucial role in maintenance of cellular identity.
Collapse
Affiliation(s)
- Alessia Bagattin
- Epigenetics and Development Laboratory, Growth and Signaling Department, Université Paris Cité, CNRS, INSERM, Institut Necker-Enfants Malades, 75015 Paris, France.
| | - Salvina Laura Tammaccaro
- Epigenetics and Development Laboratory, Growth and Signaling Department, Université Paris Cité, CNRS, INSERM, Institut Necker-Enfants Malades, 75015 Paris, France
| | - Magali Chiral
- Epigenetics and Development Laboratory, Growth and Signaling Department, Université Paris Cité, CNRS, INSERM, Institut Necker-Enfants Malades, 75015 Paris, France
| | - Munevver Parla Makinistoglu
- Epigenetics and Development Laboratory, Growth and Signaling Department, Université Paris Cité, CNRS, INSERM, Institut Necker-Enfants Malades, 75015 Paris, France
| | - Nicolas Zimmermann
- Epigenetics and Development Laboratory, Growth and Signaling Department, Université Paris Cité, CNRS, INSERM, Institut Necker-Enfants Malades, 75015 Paris, France
| | - Jonathan Lerner
- Epigenetics and Development Laboratory, Growth and Signaling Department, Université Paris Cité, CNRS, INSERM, Institut Necker-Enfants Malades, 75015 Paris, France
| | - Serge Garbay
- Epigenetics and Development Laboratory, Growth and Signaling Department, Université Paris Cité, CNRS, INSERM, Institut Necker-Enfants Malades, 75015 Paris, France
| | - Nicolas Kuperwasser
- Epigenetics and Development Laboratory, Growth and Signaling Department, Université Paris Cité, CNRS, INSERM, Institut Necker-Enfants Malades, 75015 Paris, France
| | - Marco Pontoglio
- Epigenetics and Development Laboratory, Growth and Signaling Department, Université Paris Cité, CNRS, INSERM, Institut Necker-Enfants Malades, 75015 Paris, France.
| |
Collapse
|
39
|
Wu M, Beck C, Lee JH, Fulbright RM, Jeong J, Inman JT, Woodhouse MV, Berger JM, Wang MD. Human Topoisomerase IIα Promotes Chromatin Condensation Via a Phase Transition. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.15.618281. [PMID: 39464128 PMCID: PMC11507700 DOI: 10.1101/2024.10.15.618281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
Topoisomerase II (topo II) enzymes are essential enzymes known to resolve topological entanglements during DNA processing. Curiously, while yeast expresses a single topo II, humans express two topo II isozymes, topo IIα and topo IIβ, which share a similar catalytic domain but differ in their intrinsically disordered C-terminal domains (CTDs). During mitosis, topo IIα and condensin I constitute the most abundant chromosome scaffolding proteins essential for chromosome condensation. However, how topo IIα enables this function is poorly understood. Here, we discovered a new and functionally distinct role for human topo IIα - it condenses DNA and chromatin at a low topo IIα concentration (100 pM or less) during a polymer-collapse phase transition. The removal of the topo IIα CTDs effectively abolishes its condensation ability, indicating that the condensation is mediated by the CTDs. Although topo IIβ can also perform condensation, it is about 4-fold less effective. During the condensation, topo IIα-DNA condensates form along DNA, working against a DNA tension of up to 1.5 pN, greater than that previously reported for yeast condensin. In addition, this condensation does not require ATP and thus is independent of topo IIα's catalytic activity. We also found that condensation and catalysis can concurrently proceed with minimal mutual interference. Our findings suggest topo IIα may directly participate in chromosome condensation during mitosis.
Collapse
Affiliation(s)
- Meiling Wu
- Department of Physics & LASSP, Cornell University, Ithaca, NY 14853, USA
- Howard Hughes Medical Institute, Cornell University, Ithaca, NY 14853, USA
| | - Curtis Beck
- Department of Biophysics and Biophysical Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Joyce H. Lee
- Department of Biophysics and Biophysical Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | | | - Joshua Jeong
- Department of Biophysics and Biophysical Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - James T. Inman
- Department of Physics & LASSP, Cornell University, Ithaca, NY 14853, USA
- Howard Hughes Medical Institute, Cornell University, Ithaca, NY 14853, USA
| | | | - James M. Berger
- Department of Biophysics and Biophysical Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Michelle D. Wang
- Department of Physics & LASSP, Cornell University, Ithaca, NY 14853, USA
- Howard Hughes Medical Institute, Cornell University, Ithaca, NY 14853, USA
| |
Collapse
|
40
|
Liu R, Sun J, Li LF, Cheng Y, Li M, Fu L, Li S, Peng G, Wang Y, Liu S, Qu X, Ran J, Li X, Pang E, Qiu HJ, Wang Y, Qi J, Wang H, Gao GF. Structural basis for difunctional mechanism of m-AMSA against African swine fever virus pP1192R. Nucleic Acids Res 2024; 52:11301-11316. [PMID: 39166497 PMCID: PMC11472052 DOI: 10.1093/nar/gkae703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 07/24/2024] [Accepted: 08/07/2024] [Indexed: 08/23/2024] Open
Abstract
The African swine fever virus (ASFV) type II topoisomerase (Topo II), pP1192R, is the only known Topo II expressed by mammalian viruses and is essential for ASFV replication in the host cytoplasm. Herein, we report the structures of pP1192R in various enzymatic stages using both X-ray crystallography and single-particle cryo-electron microscopy. Our data structurally define the pP1192R-modulated DNA topology changes. By presenting the A2+-like metal ion at the pre-cleavage site, the pP1192R-DNA-m-AMSA complex structure provides support for the classical two-metal mechanism in Topo II-mediated DNA cleavage and a better explanation for nucleophile formation. The unique inhibitor selectivity of pP1192R and the difunctional mechanism of pP1192R inhibition by m-AMSA highlight the specificity of viral Topo II in the poison binding site. Altogether, this study provides the information applicable to the development of a pP1192R-targeting anti-ASFV strategy.
Collapse
Affiliation(s)
- Ruili Liu
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan Province 450046, China
- Beijing Life Science Academy, Beijing 102200, China
| | - Junqing Sun
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, Shanxi Province 030801, China
| | - Lian-Feng Li
- State Key Laboratory for Animal Disease Control and Prevention, National African Swine Fever Para-Reference Laboratory, National High-Containment Facilities for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin Province 150069, China
| | - Yingxian Cheng
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan Province 450046, China
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
| | - Meilin Li
- State Key Laboratory for Animal Disease Control and Prevention, National African Swine Fever Para-Reference Laboratory, National High-Containment Facilities for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin Province 150069, China
| | - Lifeng Fu
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
| | - Su Li
- State Key Laboratory for Animal Disease Control and Prevention, National African Swine Fever Para-Reference Laboratory, National High-Containment Facilities for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin Province 150069, China
| | - Guorui Peng
- China/WOAH Reference Laboratory for Classical Swine Fever, China Institute of Veterinary Drug Control, Beijing 100081, China
| | - Yanjin Wang
- State Key Laboratory for Animal Disease Control and Prevention, National African Swine Fever Para-Reference Laboratory, National High-Containment Facilities for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin Province 150069, China
| | - Sheng Liu
- SUSTech Cryo-EM Centre, Southern University of Science and Technology, Shenzhen 518055, China
| | - Xiao Qu
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
| | - Jiaqi Ran
- Department of Biological Sciences, School of life Science, Liaoning University, Shenyang, Liaoning Province 110030, China
| | - Xiaomei Li
- Shanxi Academy of Advanced Research and Innovation, Taiyuan, Shanxi Province 030032, China
| | - Erqi Pang
- Shanxi Academy of Advanced Research and Innovation, Taiyuan, Shanxi Province 030032, China
| | - Hua-Ji Qiu
- State Key Laboratory for Animal Disease Control and Prevention, National African Swine Fever Para-Reference Laboratory, National High-Containment Facilities for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin Province 150069, China
| | - Yanli Wang
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Jianxun Qi
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
| | - Han Wang
- Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing 100091, China
| | - George Fu Gao
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
| |
Collapse
|
41
|
Blackwell AM, Jami-Alahmadi Y, Nasamu AS, Kudo S, Senoo A, Slam C, Tsumoto K, Wohlschlegel JA, Caaveiro JMM, Goldberg DE, Sigala PA. Malaria parasites require a divergent heme oxygenase for apicoplast gene expression and biogenesis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.30.596652. [PMID: 38853871 PMCID: PMC11160694 DOI: 10.1101/2024.05.30.596652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
Malaria parasites have evolved unusual metabolic adaptations that specialize them for growth within heme-rich human erythrocytes. During blood-stage infection, Plasmodium falciparum parasites internalize and digest abundant host hemoglobin within the digestive vacuole. This massive catabolic process generates copious free heme, most of which is biomineralized into inert hemozoin. Parasites also express a divergent heme oxygenase (HO)-like protein (PfHO) that lacks key active-site residues and has lost canonical HO activity. The cellular role of this unusual protein that underpins its retention by parasites has been unknown. To unravel PfHO function, we first determined a 2.8 Å-resolution X-ray structure that revealed a highly α-helical fold indicative of distant HO homology. Localization studies unveiled PfHO targeting to the apicoplast organelle, where it is imported and undergoes N-terminal processing but retains most of the electropositive transit peptide. We observed that conditional knockdown of PfHO was lethal to parasites, which died from defective apicoplast biogenesis and impaired isoprenoid-precursor synthesis. Complementation and molecular-interaction studies revealed an essential role for the electropositive N-terminus of PfHO, which selectively associates with the apicoplast genome and enzymes involved in nucleic acid metabolism and gene expression. PfHO knockdown resulted in a specific deficiency in levels of apicoplast-encoded RNA but not DNA. These studies reveal an essential function for PfHO in apicoplast maintenance and suggest that Plasmodium repurposed the conserved HO scaffold from its canonical heme-degrading function in the ancestral chloroplast to fulfill a critical adaptive role in organelle gene expression.
Collapse
Affiliation(s)
| | | | - Armiyaw S. Nasamu
- Departments of Medicine and Molecular Microbiology, Washington University School of Medicine, St. Louis, MO
| | - Shota Kudo
- Department of Chemistry & Biotechnology, The University of Tokyo, Tokyo, Japan
| | - Akinobu Senoo
- Department of Protein Drug Discovery, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Celine Slam
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT
| | - Kouhei Tsumoto
- Department of Chemistry & Biotechnology, The University of Tokyo, Tokyo, Japan
- Department of Bioengineering, University of Tokyo, Tokyo, Japan
| | | | - Jose M. M. Caaveiro
- Department of Chemistry & Biotechnology, The University of Tokyo, Tokyo, Japan
| | - Daniel E. Goldberg
- Departments of Medicine and Molecular Microbiology, Washington University School of Medicine, St. Louis, MO
| | - Paul A. Sigala
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT
- Departments of Medicine and Molecular Microbiology, Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
42
|
Hong Y, Park SH, Wang H, Wang MD. Geometry of Braided DNA Dictates Supercoiling Partition. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.08.617221. [PMID: 39416030 PMCID: PMC11482784 DOI: 10.1101/2024.10.08.617221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
During DNA replication, the replisome must rotate relative to the DNA substrate, generating supercoiling that must be partitioned in front of or behind the replisome. Supercoiling partitioned behind the replisome may intertwine (or braid) daughter DNA molecules and restrict chromosome segregation. Supercoiling partitioning and torsional resistance at the replisome should depend on the geometry of the two daughter DNA molecules, determined by their end separations. However, experimental investigation of DNA braiding under well-defined DNA geometry has proven challenging. Here, we present methods to engineer braiding substrates of defined geometry, from minimal to significant end separations. We then directly measured the torque required to braid these substrates using an angular optical trap (AOT) and found that the torque required to initiate the braiding during the first 0.5 turn critically depends on the end separation. Once braiding started, we found that the subsequent effective twist persistence length of DNA braiding is about 20-30 nm, insensitive to the end separations. Our work highlights the crucial role of braiding geometry in dictating supercoiling partitioning and torque build-up during replication. It suggests that dynamic modulation of end separation on the daughter DNA molecules could serve as a mechanism to regulate replication progression in vivo.
Collapse
Affiliation(s)
- Yifeng Hong
- Department of Electrical and Computer Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Seong ha Park
- Department of Physics & LASSP, Cornell University, Ithaca, NY 14853, USA
- Howard Hughes Medical Institute, Cornell University, Ithaca, NY 14853, USA
| | - Hanjie Wang
- Department of Physics & LASSP, Cornell University, Ithaca, NY 14853, USA
| | - Michelle D. Wang
- Department of Physics & LASSP, Cornell University, Ithaca, NY 14853, USA
- Howard Hughes Medical Institute, Cornell University, Ithaca, NY 14853, USA
| |
Collapse
|
43
|
Xin Y, Xian R, Yang Y, Cong J, Rao Z, Li X, Chen Y. Structural and functional insights into the T-even type bacteriophage topoisomerase II. Nat Commun 2024; 15:8719. [PMID: 39379365 PMCID: PMC11461880 DOI: 10.1038/s41467-024-53037-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 09/26/2024] [Indexed: 10/10/2024] Open
Abstract
T-even type bacteriophages are virulent phages commonly used as model organisms, playing a crucial role in understanding various biological processes. One such process involves the regulation of DNA topology during phage replication upon host infection, governed by type IIA DNA topoisomerases. In spite of various studies on prokaryotic and eukaryotic counterparts, viral topoisomerase II remains insufficiently understood, especially the unique domain composition of T4 phage. In this study, we determine the cryo-EM structures of topoisomerase II from T4 and T6 phages, including full-length structures of both apo and DNA-binding states which have never been determined before. Together with other conformational states, these structures provide an explicit blueprint of mechanisms of phage topoisomerase II. Particularly, the asymmetric dimeric interactions observed in cryo-EM structures of T6 phage topoisomerase II ATPase domain and central domain bound with DNA shed light on the asynchronous ATP usage and asynchronous cleavage of the G-segment DNA, respectively. The elucidation of phage topoisomerase II's structures and functions not only enhances our understanding of mechanisms and evolutionary parallels with prokaryotic and eukaryotic homologs but also highlights its potential as a model for developing type IIA topoisomerase inhibitors.
Collapse
Affiliation(s)
- Yuhui Xin
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Runqi Xian
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yunge Yang
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Jingyuan Cong
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Zihe Rao
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China.
- Laboratory of Structural Biology, School of Medicine, Tsinghua University, Beijing, China.
| | - Xuemei Li
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China.
| | - Yutao Chen
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
44
|
Koide K, Kim H, Whelan MVX, Belotindos LP, Tanomsridachchai W, Changkwanyeun R, Usui M, Ó Cróinín T, Thapa J, Nakajima C, Suzuki Y. WQ-3810, a fluoroquinolone with difluoropyridine derivative as the R1 group exerts high potency against quinolone-resistant Campylobacter jejuni. Microbiol Spectr 2024; 12:e0432223. [PMID: 39162520 PMCID: PMC11448395 DOI: 10.1128/spectrum.04322-23] [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: 12/31/2023] [Accepted: 07/17/2024] [Indexed: 08/21/2024] Open
Abstract
Quinolone-resistant Campylobacter jejuni have been increasing worldwide. Quinolones exert their antibacterial activity by inhibiting DNA gyrase, but most of the isolates acquire quinolone resistance via an amino acid substitution in the A subunit of DNA gyrase. WQ-3810 is a quinolone antibiotic that has been reported to have high potency even to DNA gyrase with amino acid substitutions in several bacterial species; however, there was no information on C. jejuni. Hence, this study aimed to evaluate the activity of WQ-3810 to inhibit wild-type/mutant DNA gyrases of C. jejuni and the bacterial growth for accessing the potency for the treatment of quinolone-resistant C. jejuni infection. The inhibitory activity of WQ-3810 was assessed and compared with ciprofloxacin and nalidixic acid by calculating the half maximal inhibitory concentration (IC50) against wild-type/mutant DNA gyrases. Next, the minimum inhibitory concentration (MIC) of WQ-3810 and five other quinolones was determined for C. jejuni including quinolone-resistant strains with amino acid substitutions in GyrA. Furthermore, the interaction between WQ-3810 and wild-type/mutant DNA gyrase was speculated using docking simulations. The IC50 of WQ-3810 against wild-type DNA gyrase was 1.03 µg/mL and not different from that of ciprofloxacin. However, those of WQ-3810 against mutant DNA gyrases were much lower than ciprofloxacin. The MICs of WQ-3810 ranged <0.016-0.031 µg/mL and were the lowest against both quinolone-susceptible and quinolone-resistant strains among the examined quinolones. The results obtained by the docking simulation agreed well with this observation. WQ-3810 seems to be a promising antimicrobial agent for the infections caused by quinolone-resistant C. jejuni. IMPORTANCE WQ-3810, a relatively new quinolone antibiotic, demonstrates exceptional antibacterial properties against certain pathogens in previous studies. However, its efficacy against quinolone-resistant Campylobacter jejuni was not previously reported. The prevalence of quinolone-resistant C. jejuni as a cause of foodborne illnesses is increasing, prompting this investigation into the effectiveness of WQ-3810 as a countermeasure. This study revealed high inhibitory activity of WQ-3810 against both wild-type and mutant DNA gyrases of C. jejuni. WQ-3810 was equally efficacious as ciprofloxacin against wild-type DNA gyrases but showed superior effectiveness against mutant DNA gyrases. WQ-3810 also demonstrated the lowest minimum inhibitory concentrations, highlighting its enhanced potency against both susceptible and resistant strains of C. jejuni. This observation was well supported by the results of the in silico analysis. Consequently, WQ-3810 exhibits a higher level of bactericidal activity compared to existing quinolones in combating both susceptible and resistant C. jejuni isolates.
Collapse
Affiliation(s)
- Kentaro Koide
- Division of Bioresources, Hokkaido University International Institute for Zoonosis Control, Sapporo, Japan
- Department of Bacteriology II, National Institute of Infectious Diseases, Musashimurayama, Japan
| | - Hyun Kim
- Department of Bacteriology II, National Institute of Infectious Diseases, Musashimurayama, Japan
| | - Matthew V X Whelan
- School of Biomolecular and Biomedical Science, University College Dublin, Belfield, Ireland
| | - Lawrence P Belotindos
- Division of Bioresources, Hokkaido University International Institute for Zoonosis Control, Sapporo, Japan
| | - Wimonrat Tanomsridachchai
- Division of Bioresources, Hokkaido University International Institute for Zoonosis Control, Sapporo, Japan
| | | | - Masaru Usui
- School of Veterinary Medicine, Rakuno Gakuen University, Ebetsu, Japan
| | - Tadhg Ó Cróinín
- School of Biomolecular and Biomedical Science, University College Dublin, Belfield, Ireland
| | - Jeewan Thapa
- Division of Bioresources, Hokkaido University International Institute for Zoonosis Control, Sapporo, Japan
| | - Chie Nakajima
- Division of Bioresources, Hokkaido University International Institute for Zoonosis Control, Sapporo, Japan
- International Collaboration Unit, Hokkaido University, Sapporo, Japan
- Hokkaido University Institute for Vaccine Research and Development, Sapporo, Japan
| | - Yasuhiko Suzuki
- Division of Bioresources, Hokkaido University International Institute for Zoonosis Control, Sapporo, Japan
- International Collaboration Unit, Hokkaido University, Sapporo, Japan
- Hokkaido University Institute for Vaccine Research and Development, Sapporo, Japan
| |
Collapse
|
45
|
Desai J, Patel B, Panchal N, Gite A, Darji B, Viswanathan K, Trivedi J, Vyas P, Pawar V, Giri P, S S, Sharma R, Jain M, Iyer P, Kumar S. Discovery of aminopiperidine based potent & novel topoisomerase inhibitor with broad spectrum anti-bacterial activity. Bioorg Med Chem Lett 2024; 111:129911. [PMID: 39067715 DOI: 10.1016/j.bmcl.2024.129911] [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: 03/26/2024] [Revised: 07/13/2024] [Accepted: 07/24/2024] [Indexed: 07/30/2024]
Abstract
Bacterial DNA gyrase and topoisomerase IV inhibition has emerged as a promising strategy for the cure of infections caused by antibiotic-resistant bacteria. The Novel Bacterial Topoisomerase Inhibitors (NBTIs) bind to a different site from that of the quinolones with novel mechanism of action. This evades the existing target-mediated bacterial resistance associated with quinolones. This article presents our efforts to identify in vitro potent and broad-spectrum antibacterial agent 4l.
Collapse
Affiliation(s)
- Jigar Desai
- Zydus Research Centre, Sarkhej Bavla NH No 8A, Moraiya, Ahmedabad 382213, Gujarat, India.
| | - Bhaumin Patel
- Zydus Research Centre, Sarkhej Bavla NH No 8A, Moraiya, Ahmedabad 382213, Gujarat, India
| | - Nandini Panchal
- Zydus Research Centre, Sarkhej Bavla NH No 8A, Moraiya, Ahmedabad 382213, Gujarat, India
| | - Archana Gite
- Zydus Research Centre, Sarkhej Bavla NH No 8A, Moraiya, Ahmedabad 382213, Gujarat, India
| | - Brijesh Darji
- Zydus Research Centre, Sarkhej Bavla NH No 8A, Moraiya, Ahmedabad 382213, Gujarat, India
| | - Kasinath Viswanathan
- Zydus Research Centre, Sarkhej Bavla NH No 8A, Moraiya, Ahmedabad 382213, Gujarat, India
| | - Jinal Trivedi
- Zydus Research Centre, Sarkhej Bavla NH No 8A, Moraiya, Ahmedabad 382213, Gujarat, India
| | - Purvi Vyas
- Zydus Research Centre, Sarkhej Bavla NH No 8A, Moraiya, Ahmedabad 382213, Gujarat, India
| | - Vishwanath Pawar
- Zydus Research Centre, Sarkhej Bavla NH No 8A, Moraiya, Ahmedabad 382213, Gujarat, India
| | - Poonam Giri
- Zydus Research Centre, Sarkhej Bavla NH No 8A, Moraiya, Ahmedabad 382213, Gujarat, India
| | - Sachchidanand S
- Zydus Research Centre, Sarkhej Bavla NH No 8A, Moraiya, Ahmedabad 382213, Gujarat, India
| | - Rajiv Sharma
- Zydus Research Centre, Sarkhej Bavla NH No 8A, Moraiya, Ahmedabad 382213, Gujarat, India
| | - Mukul Jain
- Zydus Research Centre, Sarkhej Bavla NH No 8A, Moraiya, Ahmedabad 382213, Gujarat, India
| | - Pravin Iyer
- Zydus Research Centre, Sarkhej Bavla NH No 8A, Moraiya, Ahmedabad 382213, Gujarat, India
| | - Sanjay Kumar
- Zydus Research Centre, Sarkhej Bavla NH No 8A, Moraiya, Ahmedabad 382213, Gujarat, India
| |
Collapse
|
46
|
Chen R, Ren Z, Bai L, Hu X, Chen Y, Ye Q, Hu Y, Shi J. Novel antibody-drug conjugates based on DXd-ADC technology. Bioorg Chem 2024; 151:107697. [PMID: 39121594 DOI: 10.1016/j.bioorg.2024.107697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Revised: 07/27/2024] [Accepted: 08/04/2024] [Indexed: 08/12/2024]
Abstract
In recent years, antibody-drug conjugate (ADC) technology, which uses monoclonal antibodies (mAbs) to specifically deliver effective cytotoxic payloads to tumor cells, has become a promising method of tumor targeted therapy. ADCs are a powerful class of biopharmaceuticals that link antibodies targeting specific antigens and small molecule drugs with potent cytotoxicity via a linker, thus enabling selective destruction of cancer cells while minimizing systemic toxicity. DXd is a topoisomerase I inhibitor that induces DNA damage leading to cell cycle arrest, making it an option for ADC payloads. The DXd-ADC technology, developed by Daiichi Sankyo, is a cutting-edge platform that produces a new generation of ADCs with improved therapeutic metrics and has shown significant therapeutic potential in various types of cancer. This review provides a comprehensive assessment of drugs developed with DXd-ADC technology, with a focus on mechanisms of action, pharmacokinetics studies, preclinical data, and clinical outcomes for DS-8201a, U3-1402, DS-1062a, DS-7300a, DS-6157a, and DS-6000a. By integrating existing data, we aim to provide valuable insights into the current therapeutic status and future prospects of these novel agents.
Collapse
Affiliation(s)
- Rong Chen
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, Sichuan, China
| | - Zhiwen Ren
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Lan Bai
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Xuefang Hu
- Key Laboratory of Agro-Products Postharvest Handling, Ministry of Agriculture, Academy of Agricultural Planning and Engineering Mara, Beijing 100121, China
| | - Yuchen Chen
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, Sichuan, China
| | - Qiang Ye
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, Sichuan, China.
| | - Yuan Hu
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China.
| | - Jianyou Shi
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China.
| |
Collapse
|
47
|
Dernovšek J, Urbančič D, Zajec Ž, Sturtzel C, Grissenberger S, Wenninger-Weinzierl A, Gedgaudas M, Zubrienė A, Goričan T, Golič Grdadolnik S, Skok Ž, Ilaš J, Distel M, Zidar N, Tomašič T. First dual inhibitors of human topoisomerase IIα and Hsp90 C-terminal domain inhibit the growth of Ewing sarcoma in vitro and in vivo. Bioorg Chem 2024; 153:107850. [PMID: 39395321 DOI: 10.1016/j.bioorg.2024.107850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 09/19/2024] [Accepted: 09/26/2024] [Indexed: 10/14/2024]
Abstract
Heat shock protein 90 (Hsp90) and topoisomerase IIα (TopoIIα) are members of the GHKL protein superfamily, both with clinically validated roles as anticancer drug targets. We report the discovery of the first class of dual inhibitors targeting the ATP-binding site of TopoIIα and the C-terminal domain of Hsp90, displaying potent cancer growth inhibition both in vitro and in vivo. Initially, a known TopoIIα inhibitor, compound 3, was shown to bind to the C-terminal domain of Hsp90, but not to its ATP-binding N-terminal domain. Nineteen analogs were then prepared and evaluated to investigate the structure-activity relationships, several of which inhibited the growth of SK-N-MC Ewing sarcoma cells in vitro. Compound 3 emerged as one of the most potent growth inhibitors (IC50 = 0.33 ± 0.04 µM), demonstrating the ability to induce apoptosis and cell cycle arrest in SK-N-MC cells in vitro, and to slow the growth of Ewing sarcoma in vivo in a zebrafish model.
Collapse
Affiliation(s)
- Jaka Dernovšek
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva cesta 7, 1000 Ljubljana, Slovenia
| | - Dunja Urbančič
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva cesta 7, 1000 Ljubljana, Slovenia
| | - Živa Zajec
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva cesta 7, 1000 Ljubljana, Slovenia
| | - Caterina Sturtzel
- St. Anna Children's Cancer Research Institute, Zimmermannplatz 10, 1090 Vienna, Austria
| | - Sarah Grissenberger
- St. Anna Children's Cancer Research Institute, Zimmermannplatz 10, 1090 Vienna, Austria
| | | | - Marius Gedgaudas
- Department of Biothermodynamics and Drug Design, Institute of Biotechnology, Life Sciences Center, Vilnius University, Saulėtekio al. 7, LT-10257 Vilnius, Lithuania
| | - Asta Zubrienė
- Department of Biothermodynamics and Drug Design, Institute of Biotechnology, Life Sciences Center, Vilnius University, Saulėtekio al. 7, LT-10257 Vilnius, Lithuania
| | - Tjaša Goričan
- Laboratory for Molecular Structural Dynamics, Theory Department, National Institute of Chemistry, Hajdrihova 19, 1001 Ljubljana, Slovenia
| | - Simona Golič Grdadolnik
- Laboratory for Molecular Structural Dynamics, Theory Department, National Institute of Chemistry, Hajdrihova 19, 1001 Ljubljana, Slovenia
| | - Žiga Skok
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva cesta 7, 1000 Ljubljana, Slovenia
| | - Janez Ilaš
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva cesta 7, 1000 Ljubljana, Slovenia
| | - Martin Distel
- St. Anna Children's Cancer Research Institute, Zimmermannplatz 10, 1090 Vienna, Austria
| | - Nace Zidar
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva cesta 7, 1000 Ljubljana, Slovenia.
| | - Tihomir Tomašič
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva cesta 7, 1000 Ljubljana, Slovenia.
| |
Collapse
|
48
|
Andrade-Pavón D, Gómez-García O, Villa-Tanaca L. Review and Current Perspectives on DNA Topoisomerase I and II Enzymes of Fungi as Study Models for the Development of New Antifungal Drugs. J Fungi (Basel) 2024; 10:629. [PMID: 39330389 PMCID: PMC11432948 DOI: 10.3390/jof10090629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 08/25/2024] [Accepted: 08/29/2024] [Indexed: 09/28/2024] Open
Abstract
Fungal infections represent a growing public health problem, mainly stemming from two phenomena. Firstly, certain diseases (e.g., AIDS and COVID-19) have emerged that weaken the immune system, leaving patients susceptible to opportunistic pathogens. Secondly, an increasing number of pathogenic fungi are developing multi-drug resistance. Consequently, there is a need for new antifungal drugs with novel therapeutic targets, such as type I and II DNA topoisomerase enzymes of fungal organisms. This contribution summarizes the available information in the literature on the biology, topology, structural characteristics, and genes of topoisomerase (Topo) I and II enzymes in humans, two other mammals, and 29 fungi (including Basidiomycetes and Ascomycetes). The evidence of these enzymes as alternative targets for antifungal therapy is presented, as is a broad spectrum of Topo I and II inhibitors. Research has revealed the genes responsible for encoding the Topo I and II enzymes of fungal organisms and the amino acid residues and nucleotide residues at the active sites of the enzymes that are involved in the binding mode of topoisomerase inhibitors. Such residues are highly conserved. According to molecular docking studies, antifungal Topo I and II inhibitors have good affinity for the active site of the respective enzymes. The evidence presented in the current review supports the proposal of the suitability of Topo I and II enzymes as molecular targets for new antifungal drugs, which may be used in the future in combined therapies for the treatment of infections caused by fungal organisms.
Collapse
Affiliation(s)
- Dulce Andrade-Pavón
- Laboratorio de Biología Molecular de Bacterias y Levaduras, Departamento de Microbiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Prol. de Carpio y Plan de Ayala. Col. Sto. Tomás, Ciudad de México 11340, Mexico;
- Departamento de Fisiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Av. Wilfrido Massieu 399, Nueva Industrial Vallejo, Gustavo A. Madero, Ciudad de México 07738, Mexico
| | - Omar Gómez-García
- Departamento de Química Orgánica, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Prol. de Carpio y Plan de Ayala. Col. Sto. Tomás, Ciudad de México 11340, Mexico
| | - Lourdes Villa-Tanaca
- Laboratorio de Biología Molecular de Bacterias y Levaduras, Departamento de Microbiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Prol. de Carpio y Plan de Ayala. Col. Sto. Tomás, Ciudad de México 11340, Mexico;
| |
Collapse
|
49
|
Paul Chowdhuri S, Das BB. TDP1 phosphorylation by CDK1 in mitosis promotes MUS81-dependent repair of trapped Top1-DNA covalent complexes. EMBO J 2024; 43:3710-3732. [PMID: 39014228 PMCID: PMC11377750 DOI: 10.1038/s44318-024-00169-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 06/26/2024] [Accepted: 06/28/2024] [Indexed: 07/18/2024] Open
Abstract
Topoisomerase 1 (Top1) controls DNA topology, relieves DNA supercoiling during replication and transcription, and is critical for mitotic progression to the G1 phase. Tyrosyl-DNA phosphodiesterase 1 (TDP1) mediates the removal of trapped Top1-DNA covalent complexes (Top1cc). Here, we identify CDK1-dependent phosphorylation of TDP1 at residue S61 during mitosis. A TDP1 variant defective for S61 phosphorylation (TDP1-S61A) is trapped on the mitotic chromosomes, triggering DNA damage and mitotic defects. Moreover, we show that Top1cc repair in mitosis occurs via a MUS81-dependent DNA repair mechanism. Replication stress induced by camptothecin or aphidicolin leads to TDP1-S61A enrichment at common fragile sites, which over-stimulates MUS81-dependent chromatid breaks, anaphase bridges, and micronuclei, ultimately culminating in the formation of 53BP1 nuclear bodies during G1 phase. Our findings provide new insights into the cell cycle-dependent regulation of TDP1 dynamics for the repair of trapped Top1-DNA covalent complexes during mitosis that prevents genomic instability following replication stress.
Collapse
Affiliation(s)
- Srijita Paul Chowdhuri
- Laboratory of Molecular Biology, School of Biological Sciences, Indian Association for the Cultivation of Science, 2A & B, Raja S. C. Mullick Road, Jadavpur, Kolkata, West Bengal, 700032, India
| | - Benu Brata Das
- Laboratory of Molecular Biology, School of Biological Sciences, Indian Association for the Cultivation of Science, 2A & B, Raja S. C. Mullick Road, Jadavpur, Kolkata, West Bengal, 700032, India.
| |
Collapse
|
50
|
Nofal HR, Al-Karmalawy AA, Elmaaty AA, Ismail MF, Ali AK, Abbass EM. Pharmacophore-based, rationale design, and efficient synthesis of novel tetrahydrobenzo[b]thiophene candidates as potential dual Topo I/II inhibitors and DNA intercalators. Arch Pharm (Weinheim) 2024; 357:e2400217. [PMID: 38864845 DOI: 10.1002/ardp.202400217] [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: 03/25/2024] [Revised: 05/23/2024] [Accepted: 05/26/2024] [Indexed: 06/13/2024]
Abstract
A series of tetrahydrobenzo[b]thiophene derivatives was designed and synthesized as dual topoisomerase (Topo) I/II inhibitors implicating potential DNA intercalation. Ethyl-2-amino-3-cyano-4,5,6,7-tetrahydrobenzo[b]thiophene-4-carboxylate (1) was prepared by modification of the Gewald reaction procedure using a Fe2O3 nanocatalyst and then it was used as a building block for the synthesis of tetrahydrobenzo[b]thiophene candidates (2-14). Interestingly, compound 14 showed the best cytotoxic potential against hepatocellular, colorectal, and breast cancer cell lines (IC50 = 7.79, 8.10, and 3.53 µM), respectively, surpassing doxorubicin at breast cancer (IC50 = 4.17 µM). Meanwhile, the Topo I and II inhibition assay displayed that compound 3 could exhibit the best inhibitory potential among the investigated candidates (IC50 = 25.26 and 10.01 nM), respectively, in comparison to camptothecin (IC50 = 28.34 nM) and doxorubicin (IC50 = 11.01 nM), as reference standards. In addition, the DNA intercalation assay showed that compound 14 could display the best binding affinity with an IC50 value of 77.82 µM in comparison to doxorubicin (IC50 = 58.03 µM). Furthermore, cell cycle and apoptosis analyses described that compound 3 prompts the G1 phase arrest in michigan cancer foundation-7 cancer cells and increases the apoptosis ratio by 29.31% with respect to untreated cells (2.25%). Additionally, the conducted molecular docking assured the promising binding of the investigated members toward Topo I and II with potential DNA intercalation. Accordingly, the synthesized compounds could be treated as promising anticancer candidates for future optimization.
Collapse
Affiliation(s)
- Hager R Nofal
- Department of Chemistry, Faculty of Science, Ain Shams University, Abbassia, Cairo, Egypt
| | - Ahmed A Al-Karmalawy
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Horus University-Egypt, New Damietta, Egypt
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Ahram Canadian University, Giza, Egypt
| | - Ayman Abo Elmaaty
- Medicinal Chemistry Department, Faculty of Pharmacy, Port Said University, Port Said, Egypt
| | - Mahmoud F Ismail
- Department of Chemistry, Faculty of Science, Ain Shams University, Abbassia, Cairo, Egypt
| | - Ali Khalil Ali
- Department of Chemistry, Faculty of Science, Ain Shams University, Abbassia, Cairo, Egypt
| | - Eslam M Abbass
- Department of Chemistry, Faculty of Science, Ain Shams University, Abbassia, Cairo, Egypt
| |
Collapse
|