1
|
Jahan I, Harun-Ur-Rashid M, Islam MA, Sharmin F, Al Jaouni SK, Kaki AM, Selim S. Neuronal plasticity and its role in Alzheimer's disease and Parkinson's disease. Neural Regen Res 2026; 21:107-125. [PMID: 39688547 PMCID: PMC12094540 DOI: 10.4103/nrr.nrr-d-24-01019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 11/09/2024] [Accepted: 11/27/2024] [Indexed: 12/18/2024] Open
Abstract
Neuronal plasticity, the brain's ability to adapt structurally and functionally, is essential for learning, memory, and recovery from injuries. In neurodegenerative diseases such as Alzheimer's disease and Parkinson's disease, this plasticity is disrupted, leading to cognitive and motor deficits. This review explores the mechanisms of neuronal plasticity and its effect on Alzheimer's disease and Parkinson's disease. Alzheimer's disease features amyloid-beta plaques and tau tangles that impair synaptic function, while Parkinson's disease involves the loss of dopaminergic neurons affecting motor control. Enhancing neuronal plasticity offers therapeutic potential for these diseases. A systematic literature review was conducted using databases such as PubMed, Scopus, and Google Scholar, focusing on studies of neuronal plasticity in Alzheimer's disease and Parkinson's disease. Data synthesis identified key themes such as synaptic mechanisms, neurogenesis, and therapeutic strategies, linking molecular insights to clinical applications. Results highlight that targeting synaptic plasticity mechanisms, such as long-term potentiation and long-term depression, shows promise. Neurotrophic factors, advanced imaging techniques, and molecular tools (e.g., clustered regularly interspaced short palindromic repeats and optogenetics) are crucial in understanding and enhancing plasticity. Current therapies, including dopamine replacement, deep brain stimulation, and lifestyle interventions, demonstrate the potential to alleviate symptoms and improve outcomes. In conclusion, enhancing neuronal plasticity through targeted therapies holds significant promise for treating neurodegenerative diseases. Future research should integrate multidisciplinary approaches to fully harness the therapeutic potential of neuronal plasticity in Alzheimer's disease and Parkinson's disease.
Collapse
Affiliation(s)
- Israt Jahan
- Genetic Engineering and Biotechnology Research Laboratory (GEBRL), Centre for Advanced Research in Sciences (CARS), University of Dhaka, Dhaka, Bangladesh
| | - Mohammad Harun-Ur-Rashid
- Department of Chemistry, International University of Business Agriculture and Technology (IUBAT), Sector 10, Uttara Model Town, Dhaka, Bangladesh
| | - Md. Aminul Islam
- Genetic Engineering and Biotechnology Research Laboratory (GEBRL), Centre for Advanced Research in Sciences (CARS), University of Dhaka, Dhaka, Bangladesh
| | - Farhana Sharmin
- Department of Anatomy, Shaheed Suhrawardy Medical College, Dhaka, Bangladesh
| | - Soad K. Al Jaouni
- Department of Hematology/Oncology, Yousef Abdulatif Jameel Scientific Chair of Prophetic Medicine Application, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Abdullah M. Kaki
- Department of Anesthesia and Pain Medicine, Director of Pain Clinic, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Samy Selim
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka, Saudi Arabia
| |
Collapse
|
2
|
Zhang X, Di Y, Zhang Y, Hu Y, Chi M, Kang J, Zheng Y, Wang H, Wang Y, Chen J, Zhang X. Hypermutability of Mycolicibacterium smegmatis due to ribonucleotide reductase-mediated oxidative homeostasis and imbalanced dNTP pools. Emerg Microbes Infect 2025; 14:2480698. [PMID: 40099407 PMCID: PMC11948356 DOI: 10.1080/22221751.2025.2480698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 02/09/2025] [Accepted: 03/12/2025] [Indexed: 03/19/2025]
Abstract
Ribonucleotide reductase (RNR) catalyzes the synthesis of four deoxyribonucleoside triphosphates (dNTPs), which are essential for DNA replication. Although dNTP imbalances reduce replication fidelity and elevate mutation rates, the impact of RNR dysfunction on Mycobacterium tuberculosis (Mtb) physiology and drug resistance remains unknown. Here, we constructed inducible knockdown strains for the RNR R1 subunit NrdE in Mtb and Mycolicibacterium smegmatis (Msm). NrdE knockdown significantly impaired growth and metabolic imbalances, indirectly disrupting oxidative homeostasis and mycolic acid synthesis, while increasing levels of intracellular ROS accumulation and enhancing cell wall permeability. Additionally, we developed genomic mutant strains, Msm-Y252A and Msm-Q255A, featuring targeted point mutations in the substrate-specific site (S-site) of the RNR loop domain, which determines NDP reduction specificity. The Msm-Y252A displayed a 1.9-fold decrease in dATP and increases in dGTP (1.6-fold), dTTP (9.0-fold), and dCTP (1.3-fold). In contrast, Msm-Q255A exhibited elevated intracellular levels of dGTP (1.6-fold), dTTP (6.1-fold), and dATP (1.5-fold), while dCTP levels remained unchanged. Neither the NrdE knockdown strain nor the S-site mutants exhibited direct resistance development; however, they both showed genomic instability, enhancing the emergence of rifampicin-resistant mutants, even with a 70-fold and a 25-fold increase in mutation frequency for Msm-Y252A and Msm-Q255A, respectively. This study demonstrates that NrdE is integral to Mycobacterium survival and genomic stability and that its RNR dysfunction creates a mutagenic environment under selective pressure, indirectly contributes to the development of drug resistance, positioning NrdE as an effective target for therapeutic strategies and a valuable molecular marker for early detection of drug-resistant Mtb.
Collapse
Affiliation(s)
- Xiao Zhang
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, People’s Republic of China
| | - Yuchang Di
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, People’s Republic of China
| | - Yu Zhang
- Shanghai Sci-Tech Inno Center for Infection & Immunity, Shanghai, People’s Republic of China
- Department of Infectious Diseases, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, National Medical Center for Infectious Diseases, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, People’s Republic of China
| | - Youwei Hu
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, People’s Republic of China
| | - Mingzhe Chi
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, People’s Republic of China
| | - Jian Kang
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, People’s Republic of China
| | - Yuqing Zheng
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, People’s Republic of China
| | - Hengyu Wang
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, People’s Republic of China
| | - Yu Wang
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, People’s Republic of China
| | - Jiazhen Chen
- Shanghai Sci-Tech Inno Center for Infection & Immunity, Shanghai, People’s Republic of China
- Department of Infectious Diseases, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, National Medical Center for Infectious Diseases, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, People’s Republic of China
| | - Xuelian Zhang
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, People’s Republic of China
- Shanghai Sci-Tech Inno Center for Infection & Immunity, Shanghai, People’s Republic of China
- MOE Engineering Research Center of Gene Technology and Shanghai Engineering Research Center of Industrial Microorganism, Fudan University, Shanghai, People's Republic of China
| |
Collapse
|
3
|
Haq SU, Ling W, Aqib AI, Danmei H, Aleem MT, Fatima M, Ahmad S, Gao F. Exploring the intricacies of antimicrobial resistance: Understanding mechanisms, overcoming challenges, and pioneering innovative solutions. Eur J Pharmacol 2025; 998:177511. [PMID: 40090539 DOI: 10.1016/j.ejphar.2025.177511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 03/07/2025] [Accepted: 03/14/2025] [Indexed: 03/18/2025]
Abstract
Antimicrobial resistance (AMR) poses a growing global threat. This review examines AMR from diverse angles, tracing the story of antibiotic resistance from its origins to today's crisis. It explores the rise of AMR, from its historical roots to the urgent need to counter this escalating menace. The review explores antibiotic classes, mechanisms, resistance profiles, and genetics. It details bacterial resistance mechanisms with illustrative examples. Multidrug-resistant bacteria spotlight AMR's resilience. Modern AMR control offers hope through precision medicine, stewardship, combination therapy, surveillance, and international cooperation. Converging traditional and innovative treatments presents an exciting frontier as novel compounds seek to enhance antibiotic efficacy. This review calls for global unity and proactive engagement to address AMR collectively, emphasizing the quest for innovative solutions and responsible antibiotic use. It underscores the interconnectedness of science, responsibility, and action in combatting AMR. Humanity faces a choice between antibiotic efficacy and obsolescence. The call is clear: unite, innovate, and prevail against AMR.
Collapse
Affiliation(s)
- Shahbaz Ul Haq
- Department of Pharmacology, Shantou University Medical College, Shantou, 515041, China.
| | - Wang Ling
- Key Laboratory of New Animal Drug Project, Gansu Province, Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agriculture and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of Chinese Academy of Agriculture Sciences, Lanzhou, 730050, China
| | - Amjad Islam Aqib
- Department of Medicine, Cholistan University of Veterinary and Animal Sciences, Bahawalpur, 63100, Pakistan
| | - Huang Danmei
- Department of Pharmacology, Shantou University Medical College, Shantou, 515041, China
| | - Muhammad Tahir Aleem
- Department of Pharmacology, Shantou University Medical College, Shantou, 515041, China
| | - Mahreen Fatima
- Faculty of Biosciences, Cholistan University of Veterinary and Animal Sciences, Bahawalpur, 63100, Pakistan
| | - Saad Ahmad
- Engineering & Technology Research Center of Traditional Chinese Veterinary Medicine of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of Chinese Academy of Agricultural Sciences, Lanzhou, 730050, China
| | - Fenfei Gao
- Department of Pharmacology, Shantou University Medical College, Shantou, 515041, China.
| |
Collapse
|
4
|
Ferreira RL, Parente Rocha JA, Leite VRMC, Moraes D, Graziani D, Pranchevicius MCDS, Soares CMDA. Proteomic profile of multidrug-resistant Serratia marcescens under meropenem challenge. Microb Pathog 2025; 204:107570. [PMID: 40222567 DOI: 10.1016/j.micpath.2025.107570] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 01/13/2025] [Accepted: 04/09/2025] [Indexed: 04/15/2025]
Abstract
Serratia marcescens is an opportunistic bacterium implicated in the prevalence of serious nosocomial infections and increased outbreaks in Intensive Care Units (ICUs) and Neonatal Intensive Care Units (NICUs). S. marcescens strains are resistant to several antimicrobial classes and express numerous virulence factors that promote pathogenicity. In the present study, the proteomic profile of the multidrug-resistant (MDR) S. marcescens clinical isolate challenged with the antimicrobial meropenem was evaluated. The proteins obtained were analyzed using liquid chromatography coupled with tandem mass spectrometry (LC-MSE). A total of 199 induced proteins were identified revealing that multidrug-resistant S. marcescens promotes increasing of proteins related to energy metabolism and efflux pump and decreases synthesis of proteins related to oxidative stress response and cell mobility upon meropenem challenge, shedding some light on the relationship between expressed proteins and bacterial pathogenicity after antimicrobial induction.
Collapse
Affiliation(s)
- Roumayne Lopes Ferreira
- Laboratório de Biologia Molecular, Instituto de Ciências Biológicas, ICB II, Campus II, Universidade Federal de Goiás, 74001-970, Goiânia, GO, Brazil.
| | - Juliana Alves Parente Rocha
- Laboratório de Biologia Molecular, Instituto de Ciências Biológicas, ICB II, Campus II, Universidade Federal de Goiás, 74001-970, Goiânia, GO, Brazil.
| | - Vanessa Rafaela Milhomem Cruz Leite
- Laboratório de Biologia Molecular, Instituto de Ciências Biológicas, ICB II, Campus II, Universidade Federal de Goiás, 74001-970, Goiânia, GO, Brazil.
| | - Dayane Moraes
- Laboratório de Biologia Molecular, Instituto de Ciências Biológicas, ICB II, Campus II, Universidade Federal de Goiás, 74001-970, Goiânia, GO, Brazil.
| | - Daniel Graziani
- Laboratório de Avaliação de Moléculas, Células e Tecidos, Escola de Veterinária e Zootecnia, Universidade Federal de Goiás, Goiânia, GO, Brazil.
| | | | - Célia Maria de Almeida Soares
- Laboratório de Biologia Molecular, Instituto de Ciências Biológicas, ICB II, Campus II, Universidade Federal de Goiás, 74001-970, Goiânia, GO, Brazil.
| |
Collapse
|
5
|
Barrera-Hernández JI, Pérez-Velázquez JR, Ramírez-Trinidad Á, Oria-Hernández J, Hernández-Vázquez E. Imide-based enones: A new scaffold that inhibits biofilm formation in Gram-negative pathogens. Bioorg Med Chem Lett 2025; 122:130206. [PMID: 40132782 DOI: 10.1016/j.bmcl.2025.130206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2025] [Revised: 03/10/2025] [Accepted: 03/21/2025] [Indexed: 03/27/2025]
Abstract
We prepared a series of enones containing different substituents as potential antibiofilm molecules. The design considered the structural features previously found in N-acylhomoserine lactones, but it replaced the labile furanone with different imides portions. After evaluation, some of the analogs inhibited 50 % or more the formation of the biofilm from P. aeruginosa or A. baumannii; moreover, substituents attached at the phenyl ring, the size of the enone as well as the type of imide seemed relevant for the selectivity against the tested pathogens. In the end, we performed a molecular docking study using the crystallized LasR to describe the main interactions of the ligand-receptor complex and propose a plausible mechanism of action.
Collapse
Affiliation(s)
- J Israel Barrera-Hernández
- Departamento de Química Orgánica, Instituto de Química, Universidad Nacional Autónoma de México (UNAM), CDMX, Mexico
| | - Jesús R Pérez-Velázquez
- Laboratorio de Bioquímica-Genética, Instituto Nacional de Pediatría, Secretaría de Salud Ciudad de México, Mexico
| | - Ángel Ramírez-Trinidad
- Departamento de Química Orgánica, Instituto de Química, Universidad Nacional Autónoma de México (UNAM), CDMX, Mexico
| | - Jesús Oria-Hernández
- Laboratorio de Bioquímica-Genética, Instituto Nacional de Pediatría, Secretaría de Salud Ciudad de México, Mexico
| | - Eduardo Hernández-Vázquez
- Departamento de Química Orgánica, Instituto de Química, Universidad Nacional Autónoma de México (UNAM), CDMX, Mexico.
| |
Collapse
|
6
|
Ji YW, Wen XY, Tang HP, Su WT, Xia ZY, Lei SQ. Necroptosis: a significant and promising target for intervention of cardiovascular disease. Biochem Pharmacol 2025; 237:116951. [PMID: 40268251 DOI: 10.1016/j.bcp.2025.116951] [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: 12/20/2024] [Revised: 03/18/2025] [Accepted: 04/14/2025] [Indexed: 04/25/2025]
Abstract
Due to changes in dietary structures, population aging, and the exacerbation of metabolic risk factors, the incidence of cardiovascular disease continues to rise annually, posing a significant health burden worldwide. Cell death plays a crucial role in the onset and progression of cardiovascular diseases. As a regulated endpoint encountered by cells under adverse stress conditions, the execution of necroptosis is regulated by classicalpathways, the calmodulin-dependent protein kinases (CaMK) pathway, and mitochondria-dependent pathways, and implicated in various cardiovascular diseases, including atherosclerosis, myocardial infarction, myocardial ischemia-reperfusion injury (IRI), heart failure, diabetic cardiomyopathy, dilated cardiomyopathy, hypertrophic cardiomyopathy, chemotherapy drug-induced cardiomyopathy, and abdominal aortic aneurysm (AAA). To further investigate potential therapeutic targets for cardiovascular diseases, we also analyzed the main molecules and their inhibitors involved in necroptosis in an effort to uncover insights for treatment.
Collapse
Affiliation(s)
- Yan-Wei Ji
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xin-Yu Wen
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - He-Peng Tang
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Wa-Ting Su
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhong-Yuan Xia
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Shao-Qing Lei
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China.
| |
Collapse
|
7
|
Zaki RS, Abdelhaseib M, Mahmoud MMA. Optical interferometric: A novel approach for sensitive cost-effective measurement of multispecies biofilm from stress-tolerant field samples. Food Res Int 2025; 212:116378. [PMID: 40382065 DOI: 10.1016/j.foodres.2025.116378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 03/27/2025] [Accepted: 04/15/2025] [Indexed: 05/20/2025]
Abstract
Biofilms significantly impact public health and food safety within the meat industry. Detecting and quantifying biofilms are crucial tasks; however, their inherent complexity and heterogeneity, particularly in immature stages, present substantial challenges. Innovative techniques are necessary for accurate biofilm measurement, despite the presence of diverse interfering agents. An assessment of multispecies biofilms from cattle abattoir environmental samples (wall, basin, and door), equipment (tub, knives, and swivel), and offal (heart, lung, liver, spleen, rumen, and intestine). To elucidate the complexities of biofilm formation, a dual-pronged approach was employed, utilizing epifluorescence microscopy and optical interferometry to conduct a comprehensive analysis of biofilms that had developed on two distinct types of surfaces. Specifically, biotic surfaces that had undergone chilling and abiotic surfaces subjected to chlorination were examined to gain a deeper understanding of the structural and compositional characteristics of these microbial communities. Additionally, the presence of antibiotic-resistant bacteria within the submerged biofilm was evaluated. Despite chilling, the multispecies biofilm continued to grow from 0.2 μm to a maximum of 1.75 μm on the beef spleen surface, demonstrating the interplay of different bacterial species in biofilm formation on biotic surfaces. The highest antibacterial resistance was recorded for Staphylococcus simulans, followed by Staphylococcus saprophyticus and Staphylococcus aureus. In conclusion, the optical interferometer is a straightforward, fast, and sensitive technique for measuring biofilm thickness within the range of 0.1 to 4 μm. Progress in this field can revolutionize food safety, hygiene, biosecurity, and biomedical applications.
Collapse
Affiliation(s)
- Rania S Zaki
- Department of Food Hygiene, Faculty of Veterinary Medicine, New Valley University, El-Kharga 72511, Egypt; Food Science Department, Purdue University, West Lafayette, IN, USA, 47906
| | - Maha Abdelhaseib
- Department of Food Hygiene, Faculty of Veterinary Medicine, Assiut University, Assiut, 71526, Egypt; Animal Science Department, Purdue University, West Lafayette, IN 47906, USA.
| | - Manal M A Mahmoud
- Department of Animal Hygiene, Faculty of Veterinary Medicine, Assiut University, Assiut 71526, Egypt
| |
Collapse
|
8
|
Schofs L, Sparo MD, Lissarrague S, de Yaniz MG, Bistoletti M, Sánchez Bruni SF. Bacteriocin AP7121 as a potential treatment for surgical site infections by Staphylococcus aureus: in vitro/in vivo models. Microb Pathog 2025; 204:107573. [PMID: 40222566 DOI: 10.1016/j.micpath.2025.107573] [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: 09/30/2024] [Revised: 03/07/2025] [Accepted: 04/09/2025] [Indexed: 04/15/2025]
Abstract
Surgical site infections (SSIs) are among the leading healthcare-associated infections worldwide, and S. aureus is the most prevalent cause. Antimicrobial resistance, dormant cells, and biofilm formation contribute to treatment failure in SSIs. Therefore, new therapeutic approaches are needed to fight SSIs. The bacteriocin AP7121 has previously shown in vitro bactericidal and anti-biofilm activity against multi-resistant S. aureus. This study aimed to advance on the characterization of the in vitro activity of AP7121 against dormant forms of methicillin-resistant S. aureus (MRSA) and its effect on the adherence of a biofilm-producing strain to sutures. Additionally, a preliminary murine model of SSIs was utilized to proceed toward the in vivo application of AP7121, comparing its antimicrobial potency with the commercial antibiotic Cefazolin. Initially, MRSA cultures were grown to the logarithmic growth phase and subsequently exposed to varying concentrations of AP7121. Viable bacterial counts were assessed at different times of incubation. AP7121 demonstrated a concentration-dependent effect on dormant cells of MRSA when using 8xMIC. The effect of AP7121 on the adherence of biofilm-producing S. aureus to suture surfaces was subsequently evaluated using scanning electron microscopy. AP7121 showed significant inhibitory effects on the adherence of S. aureus in suture threads. Finally, AP7121 demonstrated a significant in vivo bactericidal effect against S. aureus in SSI model. The reduction in viable bacterial counts compared to the control group exceeded 90 % for both Cefazolin and AP7121 treatments. These preliminary findings highlight AP7121 as a novel and promising antimicrobial peptide for potential applications in human and veterinary medicine.
Collapse
Affiliation(s)
- Laureano Schofs
- Universidad Nacional del Centro de la Provincia de Buenos Aires (UNCPBA), Facultad de Ciencias Veterinarias. Paraje Arroyo Seco. Campus Universitario (B7000), Tandil, Buenos Aires, Argentina; Centro de Investigación Veterinaria Tandil (CIVETAN). Universidad Nacional del Centro de la Provincia de Buenos Aires (UNCPBA)- Comisión de investigaciones científicas de la Provincia de Buenos Aires (CICPBA)- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Paraje Arroyo Seco. Campus Universitario (B7000), Tandil, Buenos Aires, Argentina.
| | - Mónica D Sparo
- Centro de Investigación Veterinaria Tandil (CIVETAN). Universidad Nacional del Centro de la Provincia de Buenos Aires (UNCPBA)- Comisión de investigaciones científicas de la Provincia de Buenos Aires (CICPBA)- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Paraje Arroyo Seco. Campus Universitario (B7000), Tandil, Buenos Aires, Argentina; Universidad Nacional del Centro de la Provincia de Buenos Aires, Facultad de Ciencias de la Salud, Instituto de Investigación en Ciencias de la Salud. Av. Pringles N° 4375 (B7400), Olavarría, Buenos Aires, Argentina
| | - Sabina Lissarrague
- Universidad Nacional del Centro de la Provincia de Buenos Aires, Facultad de Ciencias de la Salud, Instituto de Investigación en Ciencias de la Salud. Av. Pringles N° 4375 (B7400), Olavarría, Buenos Aires, Argentina
| | - María G de Yaniz
- Universidad Nacional del Centro de la Provincia de Buenos Aires (UNCPBA), Facultad de Ciencias Veterinarias. Paraje Arroyo Seco. Campus Universitario (B7000), Tandil, Buenos Aires, Argentina; Centro de Investigación Veterinaria Tandil (CIVETAN). Universidad Nacional del Centro de la Provincia de Buenos Aires (UNCPBA)- Comisión de investigaciones científicas de la Provincia de Buenos Aires (CICPBA)- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Paraje Arroyo Seco. Campus Universitario (B7000), Tandil, Buenos Aires, Argentina
| | - Mariana Bistoletti
- Instituto de Investigación en Ciencias de la Salud (IICS). Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET). Facultad de Ciencias de la Salud, Universidad Nacional del Centro de la Provincia de Buenos Aires. Av. Pringles N° 4375 (B7400), Olavarría, Buenos Aires, Argentina
| | - Sergio F Sánchez Bruni
- Universidad Nacional del Centro de la Provincia de Buenos Aires (UNCPBA), Facultad de Ciencias Veterinarias. Paraje Arroyo Seco. Campus Universitario (B7000), Tandil, Buenos Aires, Argentina; Centro de Investigación Veterinaria Tandil (CIVETAN). Universidad Nacional del Centro de la Provincia de Buenos Aires (UNCPBA)- Comisión de investigaciones científicas de la Provincia de Buenos Aires (CICPBA)- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Paraje Arroyo Seco. Campus Universitario (B7000), Tandil, Buenos Aires, Argentina
| |
Collapse
|
9
|
Sahu A, Ruhal R. Immune system dynamics in response to Pseudomonas aeruginosa biofilms. NPJ Biofilms Microbiomes 2025; 11:104. [PMID: 40506442 PMCID: PMC12162861 DOI: 10.1038/s41522-025-00738-2] [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: 10/27/2024] [Accepted: 05/24/2025] [Indexed: 06/16/2025] Open
Abstract
Pseudomonas aeruginosa biofilms contribute to chronic infections by resisting immune attacks and antibiotics. This review explores how innate immunity, including neutrophils, macrophages, and dendritic cells, responds to biofilms and how adaptive mechanisms involving T cells, B cells, and immunoglobulins contribute to infection persistence. Additionally, it highlights immune evasion strategies and discusses emerging therapies such as immunotherapy, monoclonal antibodies, and vaccines, offering insights into enhancing biofilm clearance and improving treatment outcomes.
Collapse
Affiliation(s)
- Abhijeet Sahu
- School of Bio Science and Technology, VIT Vellore, Vellore, Tamil Nadu, India
| | - Rohit Ruhal
- School of Bio Science and Technology, VIT Vellore, Vellore, Tamil Nadu, India.
| |
Collapse
|
10
|
Zheng YD, Xu J, Wu J, Zhong T, He QY, Sun X. Oleanolic acid derivative bardoxolone combats multidrug-resistant Staphylococcus aureus by destroying cell membrane and pyruvate metabolism pathway. J Antimicrob Chemother 2025:dkaf190. [PMID: 40492526 DOI: 10.1093/jac/dkaf190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2025] [Accepted: 05/29/2025] [Indexed: 06/12/2025] Open
Abstract
INTRODUCTION Staphylococcus aureus poses a significant threat to human health, making it imperative to develop novel antimicrobial agents to combat infections caused by this pathogen. OBJECTIVES To evaluate the antibacterial efficacy and elucidate the mechanism of bardoxolone as a potential agent against multidrug-resistant S. aureus. METHODS Natural products and their derivatives were systematically evaluated for antibacterial activity. The antibacterial activity of bardoxolone was assessed in vitro against planktonic bacteria, internalized bacteria and biofilm-forming multidrug-resistant S. aureus, as well as in vivo using mouse pneumonia and thigh abscess infection models. The underlying antibacterial mechanisms were investigated through quantitative proteomics and a series of biochemical assays. RESULTS Bardoxolone exhibited potent antibacterial efficacy against S. aureus and other Gram-positive pathogens. It demonstrated strong antibacterial activity against internalized and biofilm-associated multidrug-resistant S. aureus, showing low resistance potential. In murine infection models, treatment significantly enhanced survival rates while reducing bacterial burden and attenuating inflammatory responses in pulmonary and femoral tissues. Mechanistic analyses revealed dual antibacterial actions: membrane integrity disruption and suppression of pyruvate metabolism, manifesting as diminished activity of pivotal enzymes, reduced acetyl-CoA/ATP synthesis and consequent growth inhibition of S. aureus. CONCLUSIONS These findings suggest that bardoxolone holds promise as a candidate drug for treating refractory multidrug-resistant S. aureus infections.
Collapse
Affiliation(s)
- Yun-Dan Zheng
- MOE Key Laboratory of Tumor Molecular Biology and State Key Laboratory of Bioactive Molecules and Druggability Assessment, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Jiayi Xu
- MOE Key Laboratory of Tumor Molecular Biology and State Key Laboratory of Bioactive Molecules and Druggability Assessment, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Jiayi Wu
- MOE Key Laboratory of Tumor Molecular Biology and State Key Laboratory of Bioactive Molecules and Druggability Assessment, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Tairan Zhong
- MOE Key Laboratory of Tumor Molecular Biology and State Key Laboratory of Bioactive Molecules and Druggability Assessment, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Qing-Yu He
- MOE Key Laboratory of Tumor Molecular Biology and State Key Laboratory of Bioactive Molecules and Druggability Assessment, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Xuesong Sun
- MOE Key Laboratory of Tumor Molecular Biology and State Key Laboratory of Bioactive Molecules and Druggability Assessment, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou, China
| |
Collapse
|
11
|
Kang A, Qiao Y, Pan S, Yan F, Chen H, Bai Y. From RIPK1 to Necroptosis: Pathogenic Mechanisms in Neurodegenerative Diseases. Neurochem Res 2025; 50:194. [PMID: 40493155 DOI: 10.1007/s11064-025-04448-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2025] [Revised: 05/26/2025] [Accepted: 05/30/2025] [Indexed: 06/12/2025]
Abstract
Receptor-interacting protein kinase 1 (RIPK1)-mediated necroptosis, a newly identified mode of regulated cell death, represents a significant pathogenic mechanism in multiple neurodegenerative disorders. Substantial experimental evidence indicates that RIPK1 regulates necroptotic cell death pathways in both neuronal and glial cell populations through activation of the canonical RIPK3-MLKL signaling cascade, thereby exacerbating neuroinflammatory responses and accelerating neurodegenerative progression. The pathological relevance of this molecular pathway has been extensively validated across multiple major neurodegenerative disorders, including Alzheimer's disease (AD), Parkinson's disease (PD), amyotrophic lateral sclerosis (ALS), and multiple sclerosis (MS). Pharmacological interventions targeting RIPK1 or its downstream effectors-particularly RIPK3 and MLKL-have demonstrated significant efficacy in mitigating disease-associated pathological manifestations. This highlights the RIPK1 signaling axis as a promising therapeutic target for neuroprotective strategies. Consequently, thorough investigation of RIPK1-mediated necroptosis in neurodegenerative settings holds considerable translational potential. Such inquiry deepens mechanistic understanding of disease pathogenesis while accelerating the advancement of innovative therapeutic approaches with direct clinical relevance.
Collapse
Affiliation(s)
- Anjuan Kang
- School of Pharmacy, Lanzhou University, Lanzhou, 730000, China
- Cuiying Biomedical Research Center, Lanzhou University Second Hospital, Lanzhou, 730000, China
| | - Yujun Qiao
- Department of Pharmacy, Lanzhou University Second Hospital, Lanzhou, 730000, China
- Cuiying Biomedical Research Center, Lanzhou University Second Hospital, Lanzhou, 730000, China
| | - Shunli Pan
- Department of Pharmacy, Lanzhou University Second Hospital, Lanzhou, 730000, China
- Cuiying Biomedical Research Center, Lanzhou University Second Hospital, Lanzhou, 730000, China
| | - Feixing Yan
- School of Pharmacy, Lanzhou University, Lanzhou, 730000, China
- Cuiying Biomedical Research Center, Lanzhou University Second Hospital, Lanzhou, 730000, China
| | - Haile Chen
- Department of Pharmacy, Lanzhou University Second Hospital, Lanzhou, 730000, China
- Cuiying Biomedical Research Center, Lanzhou University Second Hospital, Lanzhou, 730000, China
| | - Yinliang Bai
- School of Pharmacy, Lanzhou University, Lanzhou, 730000, China.
- Department of Pharmacy, Lanzhou University Second Hospital, Lanzhou, 730000, China.
- Cuiying Biomedical Research Center, Lanzhou University Second Hospital, Lanzhou, 730000, China.
| |
Collapse
|
12
|
Aghaali Z, Zargar M, Naghavi MR. Encapsulation of Ferula-derived bioactive compounds in nanoparticles: A promising therapeutic route for cancers and infectious diseases. Int Immunopharmacol 2025; 157:114705. [PMID: 40306115 DOI: 10.1016/j.intimp.2025.114705] [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: 02/18/2025] [Revised: 04/03/2025] [Accepted: 04/19/2025] [Indexed: 05/02/2025]
Abstract
The biomedical sector is constantly searching for new drugs that efficiently improve human health and well-being with minimum side effects. Bio-inspired nanomedicine has emerged as a feasible alternative to chemotherapeutic agents for diagnosis and therapy due to its safety, biocompatibility, affordability, and sustainability. Among biological sources available for green nanomedicine are plants offering the avoidance of the labor-intensive and time-consuming processes of cultivation and maintenance compared to microorganisms. With a long history of treating over a hundred health-related issues, Ferula-derived metabolites have received special consideration for combining with nanoparticles (NPs) since they have been reported to enhance the therapeutic efficiency of NPs, enable targeted drug delivery, and ensure controlled release, which make them elusive candidates for green nanotechnology. This review aimed to provide comprehensive information about the inhibitory effects of NPs carrying Ferula-originated bioactive compounds on several cancers and pathogenic bacteria. Plus, it explores the potential of these NPs in addressing different viral diseases, such as HIV, SARS-CoV2, and hepatitis. The anticancer, antibacterial, and antiviral mechanisms of action are also briefed. The valuable insights provided by this article may result in the development of designer Ferula-based NPs that satisfy the growing needs of the pharmaceutical industry for innovative and effective medications.
Collapse
Affiliation(s)
- Zahra Aghaali
- Division of Plant Biotechnology, Department of Agronomy and Plant Breeding, College of Agricultural and Natural Resources, University of Tehran, Karaj, Iran.
| | - Meisam Zargar
- Department of Agrobiotechnology, Agrarian Technological Institute, RUDN University, Moscow, Russia
| | - Mohammad Reza Naghavi
- Division of Plant Biotechnology, Department of Agronomy and Plant Breeding, College of Agricultural and Natural Resources, University of Tehran, Karaj, Iran; Department of Agrobiotechnology, Agrarian Technological Institute, RUDN University, Moscow, Russia.
| |
Collapse
|
13
|
Rupa N, Donthineni PR, Basu S, Arunasri K. The burden of antimicrobial resistance in biofilm forming Staphylococcus spp. from Vernal Keratoconjunctivitis patients eyes. Biofilm 2025; 9:100278. [PMID: 40276588 PMCID: PMC12018567 DOI: 10.1016/j.bioflm.2025.100278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 04/02/2025] [Accepted: 04/04/2025] [Indexed: 04/26/2025] Open
Abstract
Vernal keratoconjunctivitis (VKC) is a chronic allergic ocular surface disease with seasonal recurrences and severe forms showing vision threatening complications. The purpose of the study is to understand the prevalence and diversity of biofilm-forming bacteria and antimicrobial resistance in VKC compared to healthy individuals (HC). For this, conjunctival swab samples were collected from VKC (n = 26) and HC (n = 23), of which culture positive samples were 77 % and 78.26 % respectively. The 16S rRNA gene sequencing revealed a significant increase in bacterial diversity in VKC compared to HC (p < 0.05), identifying 16 and 9 bacterial species, respectively. Staphylococcus epidermidis emerged as the predominant bacterium in both groups, with relative abundances of 52.8 % in HC and 30.2 % in VKC (p < 0.001). Biofilm formation was observed in 64.15 % of bacterial species in VKC and 31 % in HC (p < 0.001). Scanning electron microscopy analysis confirmed temporal biofilm formation by Staphylococcus spp. in both groups. Minimum inhibitory concentration testing showed that biofilm forming Staphylococcus spp. from VKC exhibited multidrug resistance (>2 antibiotics) more frequently than those from HC. Additionally, Staphylococcus spp. in VKC demonstrated higher resistance to fluoroquinolones compared to HC. These findings indicate a significantly greater prevalence of biofilm-forming and antimicrobial resistant Staphylococcus bacteria in VKC Patients compared with HC.
Collapse
Affiliation(s)
- Nelaveni Rupa
- Prof. Brien Holden Eye Research Centre, L V Prasad Eye Institute, Banjara Hills, Hyderabad, 500034, Telangana, India
| | - Pragnya Rao Donthineni
- Shantilal Shanghvi Cornea Institute, L V Prasad Eye Institute, Hyderabad, 500034, Telangana, India
| | - Sayan Basu
- Prof. Brien Holden Eye Research Centre, L V Prasad Eye Institute, Banjara Hills, Hyderabad, 500034, Telangana, India
- Shantilal Shanghvi Cornea Institute, L V Prasad Eye Institute, Hyderabad, 500034, Telangana, India
| | - Kotakonda Arunasri
- Prof. Brien Holden Eye Research Centre, L V Prasad Eye Institute, Banjara Hills, Hyderabad, 500034, Telangana, India
| |
Collapse
|
14
|
Thakur B, Verma R, Bhatia A. Mutations in Necroptosis-Related Genes Reported in Breast Cancer: A Cosmic and Uniport Database-Based Study. Clin Breast Cancer 2025; 25:e341-e359. [PMID: 39794252 DOI: 10.1016/j.clbc.2024.12.008] [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: 05/18/2024] [Revised: 11/27/2024] [Accepted: 12/05/2024] [Indexed: 01/13/2025]
Abstract
Breast cancer (BC) now holds the top position as the primary reason of cancer-related fatalities worldwide, overtaking lung cancer. BC is classified into diverse categories depending on histopathological type, hormone receptor status, and gene expression profile, with ongoing evolution in their classifications. Cancer initiates and advances when there is a disruption in cell death pathways. In BC, the primary cell death pathway, apoptosis, experiences dysregulation across multiple stages. Ongoing studies aim to discover therapeutic targets that enhance cancer cell susceptibility to apoptosis. However, resistance to this therapy remains a significant challenge in treating BC. If apoptosis is hindered, investigating alternative pathways for cell death that can effectively eradicate BC cells during treatment becomes a valuable endeavor. In this context, necroptosis is gaining considerable focus as an alternative cell death pathway. Necroptosis represents a programmed version of necrosis which shares its key regulators with apoptosis. When apoptosis is hampered, necroptosis serves as an alternative cell death pathway even in physiological conditions like formation of limbs during embryonic development. Additionally, it comes into play during bacterial and viral infections when the apoptosis machinery is hijacked and inhibited by proteins from these pathogens. Studies reveal that in BC, mutations significantly impact molecules in the apoptosis pathway, contributing to the onset, advancement, and multiplication of cancer cells. Although some studies do indicate that the functionality of necroptosis pathway may be compromised in malignancy the status of its key molecules remains largely unknown. In this article, we aim to gather the known mutations present in key molecules of necroptosis among various subtypes of BC, utilizing data from the Cosmic and UniProt databases. The same may help to enhance the development of therapeutic strategies to effectively induce necroptosis in apoptosis-resistant BCs.
Collapse
Affiliation(s)
- Banita Thakur
- Department of General Surgery, Stanford university, CA, USA
| | - Rohit Verma
- Department of Neurosurgery, Stanford University, CA, USA
| | - Alka Bhatia
- Department of Experimental Medicine & Biotechnology, PGIMER, Chandigarh, India.
| |
Collapse
|
15
|
Monteiro R, Sousa AM, Pereira MO. Aspartic acid unveils as antibiofilm agent and tobramycin adjuvant against mucoid and small colony variants of Pseudomonas aeruginosa isolates in vitro within cystic fibrosis airway mucus. Biofilm 2025; 9:100252. [PMID: 39866543 PMCID: PMC11759549 DOI: 10.1016/j.bioflm.2024.100252] [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: 08/06/2024] [Revised: 12/07/2024] [Accepted: 12/29/2024] [Indexed: 01/28/2025] Open
Abstract
Antibiotics are central to managing airway infections in cystic fibrosis (CF), yet current treatments often fail due to the presence of Pseudomonas aeruginosa biofilms, settling down the need for seeking therapies targeting biofilms. This study aimed to investigate the antibiofilm activity of aspartic acid and its potential as an adjuvant to tobramycin against P. aeruginosa biofilms formed by mucoid and small colony variant (SCV) tobramycin tolerant strain. We assessed the effect of aspartic acid on both surface-attached and suspended P. aeruginosa biofilms within CF artificial mucus and investigated the synergistic impact of combining it with non-lethal tobramycin concentrations. Our findings showed that aspartic acid inhibited planktonic P. aeruginosa without affecting its viability and prevented biofilm formation by hindering bacterial adhesion or interfering with EPS production, depending on the experimental conditions. In CF mucus, aspartic acid significantly reduced bacterial growth, with the highest inhibition observed when combined with tobramycin, showing notable effects against the mucoid and tolerant SCV strain. Despite these reductions, P. aeruginosa repopulated the mucus within 24 h of stress withdrawal. Additional strategies, including delayed tobramycin application and a second dose of co-application of aspartic acid and tobramycin were explored to address bacterial survival and recovery. Although none of the strategies eradicated P. aeruginosa, the second co-application resulted in slower bacterial recovery rates. In conclusion, this study highlighted aspartic acid as an effective antibiofilm agent and demonstrated for the first time its potential as an adjuvant to tobramycin. The combined use of aspartic acid and tobramycin offers a promising advancement in CF therapeutics, particularly against P. aeruginosa biofilms formed by mucoid and SCV strains, mitigating their antibiotic resistance.
Collapse
Affiliation(s)
- Rosana Monteiro
- CEB - Centre of Biological Engineering, University of Minho, Campus de Gualtar, 4710-057, Braga, Portugal
| | - Ana Margarida Sousa
- CEB - Centre of Biological Engineering, University of Minho, Campus de Gualtar, 4710-057, Braga, Portugal
- LABBELS – Associate Laboratory, Braga/Guimarães, Portugal
| | - Maria Olívia Pereira
- CEB - Centre of Biological Engineering, University of Minho, Campus de Gualtar, 4710-057, Braga, Portugal
- LABBELS – Associate Laboratory, Braga/Guimarães, Portugal
| |
Collapse
|
16
|
Manik MRK, Mishu ID, Mahmud Z, Muskan MN, Emon SZ. Association of fluoroquinolone resistance with rare quinolone resistance-determining region (QRDR) mutations and protein-quinolone binding affinity (PQBA) in multidrug-resistant Escherichia coli isolated from patients with urinary tract infection. J Infect Public Health 2025; 18:102766. [PMID: 40153979 DOI: 10.1016/j.jiph.2025.102766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 03/17/2025] [Accepted: 03/18/2025] [Indexed: 04/01/2025] Open
Abstract
BACKGROUND Urinary tract infections (UTIs) caused by Escherichia coli pose significant public health risks, particularly in developing countries like Bangladesh. This study aimed to elucidate resistance patterns among UTI isolates and comprehensively investigate the mutational spectrum and its impact on drug-microbe interactions. METHODS We collected and identified E. coli isolates from hospitalized UTI patients at Dhaka Medical College Hospital and determined their resistance patterns using the disc diffusion method and broth microdilution. Quinolone resistance-determining regions (QRDRs) of the target genes (gyrA, gyrB, parC, and parE) associated with fluoroquinolone resistance were amplified by polymerase chain reaction (PCR) and analyzed through BTSeq™ sequencing for mutations, followed by molecular docking analysis using PyMOL and AutoDock for the protein-quinolone binding affinity (PQBA) study. RESULTS All isolates (100 %) displayed multidrug resistance, with chloramphenicol (16 % resistant) and colistin (28 % resistant) demonstrating superior efficacy compared to other antibiotics. The isolates resistant to colistin, as determined by disc diffusion testing, exhibited remarkably high minimum inhibitory concentrations (MICs), with one isolate registering an MIC exceeding 512 µg/mL. Alarming resistance rates were observed for five antibiotic classes, except for polymyxins (28 % resistant) and protein synthesis inhibitors (48 % resistant). Fifty-two percent (52 %) of the isolates exhibited resistance to all five tested quinolones. Sequence analysis revealed a novel L88Q mutation in ParC, affecting PQBA and binding conformation. Additionally, three ParC mutations (S80I, E84V, and E84G) and two ParE mutations (S458A and I529L) were identified, which had not been previously reported in Bangladesh. Among these, S80I appeared in all isolates. Double-mutations (S83L+D87N) in GyrA, L88Q and S80I in ParC, and I529L in ParE were identified as key drivers of fluoroquinolone resistance. CONCLUSION Our findings underscore the accumulation of significant mutations within QRDRs of UTI isolates, potentially compromising fluoroquinolone efficacy. The emergence of these novel mutations warrants further investigation to impede their dissemination and combat quinolone resistance.
Collapse
Affiliation(s)
- Md Rasel Khan Manik
- Department of Biochemistry and Molecular Biology, University of Dhaka, Dhaka 1000, Bangladesh
| | | | - Zimam Mahmud
- Department of Biochemistry and Molecular Biology, University of Dhaka, Dhaka 1000, Bangladesh.
| | | | - Sharmin Zaman Emon
- Centre for Advanced Research in Sciences, University of Dhaka, Dhaka 1000, Bangladesh
| |
Collapse
|
17
|
Guo Y, Lu J, Jin P, Qiu Z, Yu F, Zhu Y, Huang J. Genomic characterization of multidrug-resistant tuberculosis in Shanghai, China: antibiotic resistance, virulence and transmission. JAC Antimicrob Resist 2025; 7:dlaf064. [PMID: 40342723 PMCID: PMC12059630 DOI: 10.1093/jacamr/dlaf064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Accepted: 04/10/2025] [Indexed: 05/11/2025] Open
Abstract
Objectives Whole-genome sequencing (WGS) was employed to investigate antibiotic resistance, virulence and transmission profiles of multidrug-resistant tuberculosis (MDR-TB) isolates from Shanghai, China. Methods A total of 306 MDR-TB clinical isolates were collected from Shanghai Pulmonary Hospital and underwent phenotypic drug susceptibility testing (DST) for common anti-TB drugs and WGS. Combined 778 published bacterial sequences, we performed phylogenetic analysis, resistance and virulence gene identification to understand the genetic relationships and resistance mechanisms among those strains. Results WGS determination, supported by DST, revealed high resistance rates for isoniazid (83.66%) and rifampicin (90.20%) among the MDR-TB isolates. Key resistance-associated mutations included katG Ser315Thr for isoniazid, rpoB mutations for rifampicin, and embB Met306Val for ethambutol. WGS demonstrated >90% concordance with culture-based DST for most drugs, except ethambutol that showed a 76.80% concordance. Analyses of virulence factors and phylogenetics revealed the genetically homogeneous, endemic MDR-TB population in Shanghai, with no evidence of recent transmission. Conclusions This study highlights the genetic homogeneity and endemic nature of MDR-TB in Shanghai, providing insights into key resistance mechanisms of TB.
Collapse
Affiliation(s)
- Yang Guo
- School of Medicine, Shenzhen Campus of Sun Yat-Sen University, Shenzhen, Guangdong 518107, China
- State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510120, China
| | - Jing Lu
- Institute of Infectious Disease, The Second Affiliated Hospital of Tianjin Medical University, Tianjin 300222, China
| | - Peipei Jin
- Department of Laboratory Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201801, China
| | - Zhipeng Qiu
- Independent Researcher, Shenzhen, Guangdong 518000, China
| | - Fangyou Yu
- Department of Laboratory Medicine, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
| | - Yan Zhu
- Systems Biology Center, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin 300308, China
| | - Jiayuan Huang
- School of Medicine, Shenzhen Campus of Sun Yat-Sen University, Shenzhen, Guangdong 518107, China
| |
Collapse
|
18
|
Roy R, Das A, Ganguly D, Chakraborty P, Paul P, Das S, Maity A, Malik M, Tribedi P. Cuminaldehyde synergistically enhances the antimicrobial and antibiofilm potential of gentamicin: A direction towards an effective combination for the control of biofilm-linked threats of Staphylococcus aureus. Braz J Microbiol 2025; 56:1033-1048. [PMID: 39934529 PMCID: PMC12095763 DOI: 10.1007/s42770-025-01628-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 01/13/2025] [Indexed: 02/13/2025] Open
Abstract
Staphylococcus aureus, a Gram-positive, coccus-shaped bacterium often causes several infections on human hosts by exploiting biofilm. This current work investigates a potential strategy to manage the threats of biofilm-linked infections by embracing a combinatorial approach involving cuminaldehyde (phytochemical) and gentamicin (antibiotic). Despite showing antimicrobial properties individually, cuminaldehyde and gentamicin could exhibit enhanced antimicrobial potential when used together against S. aureus. The fractional inhibitory concentration index (FICI = 0.36) suggested that the selected compounds (cuminaldehyde and gentamicin) offered synergistic interaction while showing antimicrobial potential against the same organism. A series of experiments indicated that the selected compounds (cuminaldehyde and gentamicin) showed substantial antibiofilm potential against S. aureus when combined. The increased antibiofilm potential was linked to the accumulation of reactive oxygen species (ROS) and increased cell membrane permeability. Additionally, the combination of the selected compounds (cuminaldehyde and gentamicin) also impeded the cell surface hydrophobicity of S. aureus, aiding in the prevention of biofilm formation. The present study also showed that combining the mentioned compounds (cuminaldehyde and gentamicin) notably reduced the secretion of several virulence factors from S. aureus. Furthermore, the current research showed that these compounds (cuminaldehyde and gentamicin) could also exhibit antibiofilm potential against the clinical strains of Methicillin-Resistant S. aureus (MRSA). Taken together, this innovative approach not only enhances the potential of existing standard antibiotics but also opens up new therapeutic possibilities for combating biofilm-related infections.
Collapse
Affiliation(s)
- Ritwik Roy
- Microbial Ecology Research Laboratory, Department of Biotechnology, The Neotia University, Sarisha, West Bengal, 743368, India
| | - Awantika Das
- Microbiology Department, Suraksha Diagnostic Pvt. Ltd., Newtown, Kolkata, West Bengal, 700156, India
| | - Debolina Ganguly
- Microbial Ecology Research Laboratory, Department of Biotechnology, The Neotia University, Sarisha, West Bengal, 743368, India
| | - Poulomi Chakraborty
- Microbial Ecology Research Laboratory, Department of Biotechnology, The Neotia University, Sarisha, West Bengal, 743368, India
| | - Payel Paul
- Microbial Ecology Research Laboratory, Department of Biotechnology, The Neotia University, Sarisha, West Bengal, 743368, India.
| | - Sharmistha Das
- Microbial Ecology Research Laboratory, Department of Biotechnology, The Neotia University, Sarisha, West Bengal, 743368, India
| | - Alakesh Maity
- Microbial Ecology Research Laboratory, Department of Biotechnology, The Neotia University, Sarisha, West Bengal, 743368, India
| | - Moumita Malik
- Microbial Ecology Research Laboratory, Department of Biotechnology, The Neotia University, Sarisha, West Bengal, 743368, India
| | - Prosun Tribedi
- Microbial Ecology Research Laboratory, Department of Biotechnology, The Neotia University, Sarisha, West Bengal, 743368, India.
| |
Collapse
|
19
|
Wang H, Bian C, Zhang Y, Zhang L, Wang F. Circular RNAs in glioma progression: Fundamental mechanisms and therapeutic potential: A review. Int J Biol Macromol 2025; 313:144360. [PMID: 40388873 DOI: 10.1016/j.ijbiomac.2025.144360] [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: 04/28/2025] [Revised: 05/14/2025] [Accepted: 05/16/2025] [Indexed: 05/21/2025]
Abstract
Gliomas are the most common primary malignant brain tumors, characterized by aggressive invasion, limited therapeutic options, and poor prognosis. Despite advances in surgery, radiotherapy, and chemotherapy, the median survival of glioma patients remains disappointingly low. Therefore, identifying glioma-associated therapeutic targets and biomarkers is of significant clinical importance. Circular RNAs (circRNAs) are a class of naturally occurring long non-coding RNAs (lncRNAs), notable for their stability and evolutionary conservation. Increasing evidence indicates that circRNA expression is dysregulated in gliomas compared to adjacent non-tumor tissues and contributes to the regulation of glioma-related biological processes. Furthermore, numerous circRNAs function as oncogenes or tumor suppressors, mediating glioma initiation, progression, and resistance to temozolomide (TMZ). Mechanistically, circRNAs regulate glioma biology through diverse pathways, including acting as miRNA sponges, binding RNA-binding proteins (RBPs), modulating transcription, and even encoding functional peptides. These features highlight the potential of circRNAs as diagnostic and prognostic biomarkers, as well as therapeutic targets for glioma. This review summarizes the dysregulation and functions of circRNAs in glioma and explores key mechanisms through which they mediate tumor progression, including DNA damage repair, programmed cell death (PCD), angiogenesis, and metabolic reprogramming. Our aim is to provide a comprehensive perspective on the multifaceted roles of circRNAs in glioma and to highlight their potential for translational application in targeted therapy.
Collapse
Affiliation(s)
- Hongbin Wang
- Head and Neck Oncology Ward, West China Hospital of Sichuan University, Chengdu, China
| | - Chenbin Bian
- Head and Neck Oncology Ward, West China Hospital of Sichuan University, Chengdu, China
| | - Yidan Zhang
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Li Zhang
- Head and Neck Oncology Ward, West China Hospital of Sichuan University, Chengdu, China
| | - Feng Wang
- Head and Neck Oncology Ward, West China Hospital of Sichuan University, Chengdu, China.
| |
Collapse
|
20
|
Muteeb G, Kazi RNA, Aatif M, Azhar A, Oirdi ME, Farhan M. Antimicrobial resistance: Linking molecular mechanisms to public health impact. SLAS DISCOVERY : ADVANCING LIFE SCIENCES R & D 2025; 33:100232. [PMID: 40216324 DOI: 10.1016/j.slasd.2025.100232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2025] [Revised: 03/20/2025] [Accepted: 04/08/2025] [Indexed: 04/21/2025]
Abstract
BACKGROUND Antimicrobial resistance (AMR) develops into a worldwide health emergency through genetic and biochemical adaptations which enable microorganisms to resist antimicrobial treatment. β-lactamases (blaNDM, blaKPC) and efflux pumps (MexAB-OprM) working with mobile genetic elements facilitate fast proliferation of multidrug-resistant (MDR) and exttreme drug-resistant (XDR) phenotypes thus creating major concerns for healthcare systems and community health as well as the agricultural sector. OBJECTIVES The review dissimilarly unifies molecular resistance pathways with public health implications through the study of epidemiological data and monitoring approaches and innovative therapeutic solutions. Previous studies separating their attention between molecular genetics and clinical outcomes have been combined into our approach which delivers an all-encompassing analysis of AMR. KEY INSIGHTS The report investigates the resistance mechanisms which feature enzymatic degradation and efflux pump overexpression together with target modification and horizontal gene transfer because these factors represent important contributors to present-day AMR developments. This review investigates AMR effects on hospital and community environments where it affects pathogens including MRSA, carbapenem-resistant Klebsiella pneumoniae, and drug-resistant Pseudomonas aeruginosa. This document explores modern AMR management methods that comprise WHO GLASS molecular surveillance systems and three innovative strategies such as CRISPR-modified genome editing and bacteriophage treatments along with antimicrobial peptides and artificial intelligence diagnostic tools. CONCLUSION The resolution of AMR needs complete scientific and global operational methods alongside state-of-the-art therapeutic approaches. Worldwide management of drug-resistant infection burden requires both enhanced infection prevention procedures with next-generation antimicrobial strategies to reduce cases effectively.
Collapse
Affiliation(s)
- Ghazala Muteeb
- Department of Nursing, College of Applied Medical Science, King Faisal University, Al-Ahsa, Saudi Arabia.
| | - Raisa Nazir Ahmed Kazi
- Department of Respiratory Therapy, College of Applied Medical Science, King Faisal, University, Al-Ahsa, Saudi Arabia
| | - Mohammad Aatif
- Department of Public Health, College of Applied Medical Science, King Faisal University, Al-Ahsa, Saudi Arabia
| | - Asim Azhar
- NAP Life Sciences; Metropolitan Region, Maharashtra 401208, India
| | - Mohamed El Oirdi
- Department of Biological Sciences, College of Science, King Faisal University, Al Ahsa, Saudi Arabia; Department of Basic Sciences, Preparatory Year, King Faisal University, Al-Ahsa, Saudi Arabia
| | - Mohd Farhan
- Department of Basic Sciences, Preparatory Year, King Faisal University, Al-Ahsa, Saudi Arabia; Department of Chemistry, College of Science, King Faisal University, Al Ahsa, Saudi Arabia.
| |
Collapse
|
21
|
Hou D, Zhao J, Guo M, Zhang X, Yu S, Li J, Forouzanfar T, Zhang Q, Pathak JL. Data-Driven Visualization of the Dynamics of Antimicrobial Peptides in Cell Death. Probiotics Antimicrob Proteins 2025:10.1007/s12602-025-10578-3. [PMID: 40434503 DOI: 10.1007/s12602-025-10578-3] [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] [Accepted: 05/03/2025] [Indexed: 05/29/2025]
Abstract
This study explores the current status, research hotspots, and emerging trends in AMP-induced cell death through bibliometric and data-driven visual analysis. The findings aim to provide researchers and clinical professionals with new insights and potential research directions. A total of 1,897 articles and reviews published between 2006 and 2024 were retrieved from the Web of Science Core Collection. Bibliometric and visual analyses were conducted using CiteSpace, VOSviewer, Scimago Graphica, Origin 2022, and WordClouds. The analysis focused on publication trends, contributing institutions, journals, authors, cited references, and keywords. China contributed the largest share of publications (28.15%). The Chinese Academy of Sciences emerged as the most collaborative institution, demonstrating the highest centrality. The author with the highest composite index was Chen, Jyh-Yih (2,985.27). Recent research hotspots have centered on elucidating the mechanisms of AMP-induced cell death and exploring the potential applications of AMPs in cancer therapy. Keywords such as anticancer peptides, mechanism, design, and antibiotic resistance currently dominate the field, reflecting its evolving focus. Research on the application of AMPs in cancer treatment is gaining momentum. The forefront of this field involves modifying and designing AMPs to address antibiotic-resistant bacterial infections and advance cancer therapeutics. However, further investigation is needed to uncover the specific molecular mechanisms underlying AMP-induced cell death, including necrosis, pyroptosis, and ferroptosis.
Collapse
Affiliation(s)
- Dan Hou
- School and Hospital of Stomatology, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou Medical University, Guangzhou, 510182, China
- Department of Oral and Maxillofacial Surgery/Oral Pathology, UMC and Academic Centre for Dentistry Amsterdam (ACTA), Cancer Center Amsterdam, Vrije Universiteit Amsterdam, Amsterdam, 1081 HZ, The Netherlands
| | - Jiatong Zhao
- School and Hospital of Stomatology, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou Medical University, Guangzhou, 510182, China
| | - Mingshi Guo
- School and Hospital of Stomatology, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou Medical University, Guangzhou, 510182, China
| | - Xinran Zhang
- School and Hospital of Stomatology, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou Medical University, Guangzhou, 510182, China
| | - Shuiqing Yu
- School and Hospital of Stomatology, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou Medical University, Guangzhou, 510182, China
| | - Jiayue Li
- School and Hospital of Stomatology, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou Medical University, Guangzhou, 510182, China
| | - Tymour Forouzanfar
- Department of Oral and Maxillofacial Surgery/Oral Pathology, UMC and Academic Centre for Dentistry Amsterdam (ACTA), Cancer Center Amsterdam, Vrije Universiteit Amsterdam, Amsterdam, 1081 HZ, The Netherlands
- Department of Oral and Maxillofacial Surgery, Leiden University Medical Center, Leiden, 2333 ZA, The Netherlands
| | - Qing Zhang
- School and Hospital of Stomatology, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou Medical University, Guangzhou, 510182, China.
- Laboratory for Myology, Department of Human Movement Sciences, Faculty of Behavioural and Movement Sciences, Amsterdam Movement Sciences, Vrije Universiteit Amsterdam, 51081 BT, Amsterdam, The Netherlands.
| | - Janak L Pathak
- School and Hospital of Stomatology, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou Medical University, Guangzhou, 510182, China.
| |
Collapse
|
22
|
El Gendy SN, Elmotayam AK, Samir R, Ezzat MI, Rabea S, Eltanany BM, Pont L, Benavente F, El Sayed AM. Comparative LC-QTOF-MS/MS Metabolomics Applied for Valorization of Jojoba Extracts as Natural Antimicrobial and Antibiofilm Agents Aided by Molecular Networking and Chemometrics. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:12747-12764. [PMID: 40384052 DOI: 10.1021/acs.jafc.5c00890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2025]
Abstract
Jojoba (Simmondsia chinensis) seeds are broadly used worldwide for their valuable oil. The chemical composition of the other parts of the jojoba shrub remains largely unexplored. In this work, the metabolite profiles of aqueous and methanolic extracts from different jojoba plant organs were analyzed by liquid chromatography-quadrupole time-of-flight tandem mass spectrometry (LC-QTOF-MS/MS) combined with molecular networking (MN), resulting in the assignment of 104 metabolites from different classes. All methanolic extracts evinced significant antibacterial activity against the examined bacterial strains. Subminimum inhibitory concentrations (sub-MICs) of the hull and leaf extracts successfully inhibited biofilm formation of Staphylococcus aureus with inhibition rates above 90%. The seed extract at 0.5 MIC effectively eradicated S. aureus and Enterococcus faecalis biofilms (>80%). Multivariate data analysis revealed the positively correlated biomarkers with bioactivity, primarily flavonoids, phenolic compounds, and simmondsins. These results present a ground for future research to valorize jojoba seed-derived products and byproducts from other organs.
Collapse
Affiliation(s)
- Salsabeel N El Gendy
- Department of Pharmacognosy, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt
| | - Amira K Elmotayam
- Department of Pharmacognosy, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt
| | - Reham Samir
- Department of Microbiology and Immunology, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt
| | - Marwa I Ezzat
- Department of Pharmacognosy, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt
| | - Sameh Rabea
- Department of Pharmaceutical Sciences, College of Pharmacy, AlMaarefa University, Riyadh 13713, Saudi Arabia
| | - Basma M Eltanany
- Department of Pharmaceutical Analytical Chemistry, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt
| | - Laura Pont
- Department of Chemical Engineering and Analytical Chemistry, Institute for Research on Nutrition and Food Safety (INSA UB), University of Barcelona, Barcelona 08028, Spain
- Serra Hunter Program, Generalitat de Catalunya, Barcelona 08007, Spain
| | - Fernando Benavente
- Department of Chemical Engineering and Analytical Chemistry, Institute for Research on Nutrition and Food Safety (INSA UB), University of Barcelona, Barcelona 08028, Spain
| | - Aly M El Sayed
- Department of Pharmacognosy, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt
| |
Collapse
|
23
|
Sharma S, Bose A, Biswas S, Sen S, Roy I. Cyperus rotundus mediated green synthesis of silver nanoparticles for antibacterial wound dressing applications. Sci Rep 2025; 15:18394. [PMID: 40419643 DOI: 10.1038/s41598-025-03555-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2025] [Accepted: 05/21/2025] [Indexed: 05/28/2025] Open
Abstract
Wound healing is a complex biological process that can be hindered by persistent infections and inflammation, especially in the presence of multidrug-resistant (MDR) bacteria. Silver nanoparticles (AgNPs) have demonstrated significant antimicrobial efficacy; however, concerns regarding their toxicity have limited their therapeutic application. METHODS In this study, we developed a biocompatible Ag-NPs-based hydrogel using Cyperus rotundus extract via a green synthesis approach for prospective wound healing applications. The synthesized AgNPs were characterized for their physicochemical properties, confirming their stability and antibacterial potency against E. coli and S. epidermidis. The Ag-NPs-loaded hydrogel was formulated using Carbopol 974P and evaluated for its physicochemical properties, antibacterial activity, anti-inflammatory potential, and cytotoxicity. RESULTS Characterization studies confirmed the successful synthesis of AgNPs, exhibiting potent antibacterial, antioxidant, and anti-inflammatory properties. The Ag-NPs-loaded hydrogel demonstrated significant wound contraction in an excision wound model, comparable to standard treatment. Additionally, in vitro safety evaluations confirmed excellent biocompatibility, minimizing toxicity concerns associated with conventional silver formulations. CONCLUSIONS These findings suggest that the developed Ag-NPs-based hydrogel is an effective, natural, and safer alternative for advanced wound care, warranting further clinical validation.
Collapse
Affiliation(s)
- Susrita Sharma
- School of Pharmaceutical Sciences, Siksha O Anusandhan (Deemed to be University), Bhubaneswar, Odisha, 751003, India
| | - Anindya Bose
- School of Pharmaceutical Sciences, Siksha O Anusandhan (Deemed to be University), Bhubaneswar, Odisha, 751003, India.
| | - Soumojit Biswas
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, S.A.S. Nagar, S.A.S. Nagar, Punjab, 160062, India
| | - Shreeja Sen
- School of Pharmaceutical Sciences, Siksha O Anusandhan (Deemed to be University), Bhubaneswar, Odisha, 751003, India
| | - Ipsita Roy
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, S.A.S. Nagar, S.A.S. Nagar, Punjab, 160062, India
| |
Collapse
|
24
|
Adhikary K, Barman S, Banerjee P, Mondal P, Dam B, Misra A, Chandra Mandal N, Mukherjee D, Bandyopadhyay B, Maiti R. Characterization and antimicrobial activity of essential oils extracted from lemongrass ( Cymbopogon flexuosus) using microwave-assisted hydro distillation. Z NATURFORSCH C 2025; 80:201-211. [PMID: 39319826 DOI: 10.1515/znc-2024-0095] [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: 02/29/2024] [Accepted: 09/05/2024] [Indexed: 09/26/2024]
Abstract
Lemongrass (Cymbopogon flexuosus) essential oil (LGEO) contains α-citral, β-citral and other phytochemicals extracted using various methods. This research extracted essential oils using steam distillation (SD) and microwave-assisted hydro distillation (MAHD) to maximize quantity and purity. LGEO was tested for antibacterial properties. LGEO was extracted using SD and compared to MAHD output based on oil production and chemical composition. We performed GCMS to characterize LGEO. Fourier transform infrared spectroscopy (FTIR) used for quantum chemical analysis. Spectroscopic analysis showed that SD extracted secondary metabolites (ethyl-linalool, isogeranial, β-citral, α-citral, geranyl acetate, and caryophyllene) yielded 9.7 %, 11.5 %, 35.4 %, 13.4 %, 6.4 %, and 6.4 %, respectively, while MAHD yielded 10.2 %, 13.4 %, 43.2 %, 17.3 %, 6.9 %, and 7.3 %. MAHD extracted α and β citral content was better than SD extraction technique. FTIR spectroscopy and quantum chemistry analysis showed extracted oil chemical composition, electronic structure of α and β citral isomers. In the disc-diffusion experiment, both extracts were effective against Gram-positive and Gram-negative bacteria and harmful fungi. LGEO from SD and MAHD extraction (30 mg/mL) demonstrated disc diffusion assay antibacterial efficacy against microorganisms. The two extracts effectively inhibited microorganisms with MIC values of 3.75 and 7.5 μg/mL. It can be concluded that, LGEO have greater antimicrobial activity in MAHD extraction.
Collapse
Affiliation(s)
- Krishnendu Adhikary
- Department of Interdisciplinary Science, Centurion University of Technology and Management, Paralakhemundi, Odisha, 761211, India
| | - Saurav Barman
- Department of Soil Science, Centurion University of Technology and Management, Paralakhemundi, Odisha, 761211, India
| | - Pradipta Banerjee
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, 15219, USA
- Department of Biochemistry and Plant Physiology, Centurion University of Technology and Management, Paralakhemundi, Odisha, 761211, India
| | - Pallav Mondal
- Department of Chemistry, National Institute of Technology, Durgapur, 713209, West Bengal, India
| | - Bomba Dam
- Microbiology Laboratory, Department of Botany, Institute of Science, Visva Bharati (A Central University), Santiniketan, West Bengal, 731235, India
| | - Arijit Misra
- Microbiology Laboratory, Department of Botany, Institute of Science, Visva Bharati (A Central University), Santiniketan, West Bengal, 731235, India
| | - Narayan Chandra Mandal
- Mycology and Plant Pathology Laboratory, Department of Botany, Institute of Science, Visva Bharati (A Central University), Santiniketan, 731235, India
| | - Debosmita Mukherjee
- Mycology and Plant Pathology Laboratory, Department of Botany, Institute of Science, Visva Bharati (A Central University), Santiniketan, 731235, India
- Plant Pathologist, Director of Cinchona and Other Medicinal Plants, Mungpoo, West Bengal, 734313, India
| | - Bidyut Bandyopadhyay
- Department of Biotechnology, Oriental Institute of Science and Technology, Dewandighi, Burdwan, West Bengal, 713102, India
| | - Rajkumar Maiti
- Department of Physiology, 326624 Bankura Christian College , Bankura, West Bengal, 722101, India
| |
Collapse
|
25
|
Baba B, Ceylani T, Teker HT, Keskin S, Genc AI, Gurbanov R, Acikgoz E. Therapeutic potential of young plasma in reversing age-related liver inflammation via modulation of NLRP3 inflammasome and necroptosis. Biogerontology 2025; 26:117. [PMID: 40418410 DOI: 10.1007/s10522-025-10260-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2025] [Accepted: 05/18/2025] [Indexed: 05/27/2025]
Abstract
The phenomenon of inflammaging, characterized by an increase in low-grade chronic inflammation, is closely associated with diseases related to liver dysfunction. This study investigated daily plasma exchange between 5-week-old and 24-month-old Sprague Dawley rats for 30 days, focusing on protein secondary structures, NLRP3 inflammasome, and necroptosis. Conformation changes in protein secondary structures were identified by infrared spectroscopy-based pattern recognition analysis. Liver biopsies with histochemical and immunohistochemical staining were used to assess molecules associated with inflammation, necroptosis and NLRP3 inflammasome complex. Expression levels of NLRP3 components were determined by qPCR. Enhanced random coils, 310 helices, β-turns, and loop structures were identified in old rats and young rats with old plasma. Young rats and old rats with young plasma displayed higher α-helices and β-sheet structures. Young rats with old plasma showed increased NLRP3, ASC, caspase-1, IL-1β, and IL-18 mRNA levels, indicating an inflammatory response. Whereas old rats with young plasma exhibited lower inflammation levels. Histological evaluations revealed that young rats receiving aged plasma showed significantly increased levels of NLRP3, ASC, caspase-1, IL-1β, TNF-α, VEGFR2, RIPK1, and MLKL immunoreactivity, whereas decreased immunoreactivity in aged rats receiving young plasma. These findings suggest that young plasma reduces NLRP3 inflammasome activation and necroptosis in aged rats.
Collapse
Affiliation(s)
- Burcu Baba
- Department of Medical Biochemistry, Faculty of Medicine, Yuksek Ihtisas University, Ankara, Turkey
| | - Taha Ceylani
- Department of Molecular Biology and Genetics, Faculty of Science and Literature, Mus Alparslan University, Mus, Turkey
- Department of Food Quality Control and Analysis, Mus Alparslan University, Mus, Turkey
| | - Hikmet Taner Teker
- Department of Medical Biology and Genetics, Faculty of Medicine, Ankara Medipol University, Ankara, Turkey.
| | - Seda Keskin
- Department of Histology and Embryology, Faculty of Medicine, Van Yuzuncu Yil University, 65090, Van, Turkey
| | - Aysun Inan Genc
- Department of Biology, Faculty of Natural Sciences, Kastamonu University, Kastamonu, Turkey
| | - Rafig Gurbanov
- Department of Bioengineering, Bilecik Seyh Edebali University, Bilecik, Turkey
- Central Research Laboratory, Bilecik Seyh Edebali University, Bilecik, Turkey
| | - Eda Acikgoz
- Department of Histology and Embryology, Faculty of Medicine, Van Yuzuncu Yil University, 65090, Van, Turkey.
- Department of Neuroscience, Faculty of Medicine, Van Yuzuncu Yil University, Van, Turkey.
| |
Collapse
|
26
|
Georgakis N, Premetis GE, Pantiora P, Varotsou C, Bodourian CS, Labrou NE. The impact of metagenomic analysis on the discovery of novel endolysins. Appl Microbiol Biotechnol 2025; 109:126. [PMID: 40411603 PMCID: PMC12103483 DOI: 10.1007/s00253-025-13513-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2025] [Revised: 05/04/2025] [Accepted: 05/05/2025] [Indexed: 05/26/2025]
Abstract
Metagenomics has revolutionized enzyme discovery by enabling the study of genetic material directly from environmental samples, bypassing the need for microbial cultivation. This approach is particularly effective for identifying novel endolysins, phage-derived enzymes with antibacterial properties suited for therapeutic and industrial applications. Diverse ecosystems, such as biofilms, human microbiome, hot springs, and geothermal areas, serve as rich reservoirs for endolysins with traits like thermostability, broad-spectrum activity, specificity and resistance to harsh conditions. Functional metagenomics, complemented by bioinformatics, enables the discovery and annotation of previously uncharacterized endolysins. Examples of endolysins discovered from metagenomics analysis are discussed. Despite the challenges of analyzing complex microbial ecosystems and isolating target genes, metagenomics holds immense potential for uncovering innovative endolysins, paving the way for developing new biotechnological applications. KEY POINTS: • Endolysins offer antibacterial potential for therapeutic and industrial use. • Metagenomics enables discovery of novel endolysins from diverse ecosystems. • Advances in tools and methods have accelerated novel endolysins discovery.
Collapse
Affiliation(s)
- Nikolaos Georgakis
- Laboratory of Enzyme Technology, Department of Biotechnology, School of Applied Biology and Biotechnology, Agricultural University of Athens, 75 Iera Odos Street, Athens, 11855, Greece
| | - Georgios E Premetis
- Laboratory of Enzyme Technology, Department of Biotechnology, School of Applied Biology and Biotechnology, Agricultural University of Athens, 75 Iera Odos Street, Athens, 11855, Greece
| | - Panagiota Pantiora
- Laboratory of Enzyme Technology, Department of Biotechnology, School of Applied Biology and Biotechnology, Agricultural University of Athens, 75 Iera Odos Street, Athens, 11855, Greece
| | - Christina Varotsou
- Laboratory of Enzyme Technology, Department of Biotechnology, School of Applied Biology and Biotechnology, Agricultural University of Athens, 75 Iera Odos Street, Athens, 11855, Greece
| | - Charoutioun S Bodourian
- Laboratory of Enzyme Technology, Department of Biotechnology, School of Applied Biology and Biotechnology, Agricultural University of Athens, 75 Iera Odos Street, Athens, 11855, Greece
| | - Nikolaos E Labrou
- Laboratory of Enzyme Technology, Department of Biotechnology, School of Applied Biology and Biotechnology, Agricultural University of Athens, 75 Iera Odos Street, Athens, 11855, Greece.
| |
Collapse
|
27
|
Canales-Huerta N, Cádiz M, Ulloa MT, Chilet LA, Palma K, Jara-Wilde J, Cuevas F, González MJ, Navarro N, Toledo J, Castañeda V, Scavone P, Härtel S. Delayed biofilm formation in non-motile uropathogenic Escherichia coli strain in static and dynamic growth conditions. Microb Pathog 2025; 206:107712. [PMID: 40412736 DOI: 10.1016/j.micpath.2025.107712] [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: 12/12/2024] [Revised: 05/04/2025] [Accepted: 05/15/2025] [Indexed: 05/27/2025]
Abstract
Urinary tract infections associated with the placement of indwelling urinary catheters are a significant concern in hospital settings, as they are linked to an increased risk of severe infections and complications due to biofilm formation. These infections are primarily caused by uropathogens such as Escherichia coli (UPEC). UPEC possesses peritrichous flagella, which facilitates its motility, adhesion to surfaces, and biofilm formation. Understanding the development of UPEC communities is essential for developing effective treatment and eradication strategies. In this study, we characterized the biofilm formation of a clinical non-motile UPEC strain under both static and dynamic culture conditions that simulate the urinary catheter environment. We developed a dynamic culture system coupled with light sheet fluorescence microscopy (LSFM) to quantify the stages of biofilm formation over time. Our results demonstrate that flagella play a crucial role in the initial phase of biofilm formation. The non-motile strain exhibited a delay in the adhesion phase compared to motile strains but ultimately formed biofilms of similar volume during subsequent stages. These findings highlight the significance of flagella in dynamic biofilm formation models and provide valuable insights for modeling the evolution of bacterial communities in nosocomial environments using LSFM.
Collapse
Affiliation(s)
- Nicole Canales-Huerta
- Laboratory for Scientific Image Analysis SCIAN-Lab, Integrative Biology Program, Institute of Biomedical Sciences ICBM, Faculty of Medicine, University of Chile, Santiago, Chile; Biomedical Neuroscience Institute BNI, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Martín Cádiz
- Laboratory for Scientific Image Analysis SCIAN-Lab, Integrative Biology Program, Institute of Biomedical Sciences ICBM, Faculty of Medicine, University of Chile, Santiago, Chile; Biomedical Neuroscience Institute BNI, Faculty of Medicine, University of Chile, Santiago, Chile; Centro de Informática Médica y Telemedicina CIMT, Faculty of Medicine, University of Chile, Santiago, Chile
| | | | - Lucas Alé Chilet
- Laboratory for Scientific Image Analysis SCIAN-Lab, Integrative Biology Program, Institute of Biomedical Sciences ICBM, Faculty of Medicine, University of Chile, Santiago, Chile; Biomedical Neuroscience Institute BNI, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Karina Palma
- Laboratory for Scientific Image Analysis SCIAN-Lab, Integrative Biology Program, Institute of Biomedical Sciences ICBM, Faculty of Medicine, University of Chile, Santiago, Chile; Biomedical Neuroscience Institute BNI, Faculty of Medicine, University of Chile, Santiago, Chile; Centro de Informática Médica y Telemedicina CIMT, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Jorge Jara-Wilde
- Laboratory for Scientific Image Analysis SCIAN-Lab, Integrative Biology Program, Institute of Biomedical Sciences ICBM, Faculty of Medicine, University of Chile, Santiago, Chile; Biomedical Neuroscience Institute BNI, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Fabrizio Cuevas
- Laboratory for Scientific Image Analysis SCIAN-Lab, Integrative Biology Program, Institute of Biomedical Sciences ICBM, Faculty of Medicine, University of Chile, Santiago, Chile
| | - María José González
- Laboratorio de Biofilms Microbianos, Departamento de Microbiología, Instituto de Investigaciones Biológicas Clemente Estable, Montevideo, Uruguay
| | - Nicolás Navarro
- Advanced Center for Chronic Diseases ACCDiS, Santiago, Chile; Laboratorio de Biofilms Microbianos, Departamento de Microbiología, Instituto de Investigaciones Biológicas Clemente Estable, Montevideo, Uruguay
| | - Jorge Toledo
- Red de Equipamiento Científico Avanzado REDECA, Institute of Biomedical Sciences ICBM, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Victor Castañeda
- Departamento de Tecnología Médica, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Paola Scavone
- Laboratorio de Biofilms Microbianos, Departamento de Microbiología, Instituto de Investigaciones Biológicas Clemente Estable, Montevideo, Uruguay
| | - Steffen Härtel
- Laboratory for Scientific Image Analysis SCIAN-Lab, Integrative Biology Program, Institute of Biomedical Sciences ICBM, Faculty of Medicine, University of Chile, Santiago, Chile; Biomedical Neuroscience Institute BNI, Faculty of Medicine, University of Chile, Santiago, Chile; Centro de Informática Médica y Telemedicina CIMT, Faculty of Medicine, University of Chile, Santiago, Chile; Centro Nacional en Sistemas de Información en Salud CENS, Santiago, Chile; Centro de Modelamiento Matemático CMM, Universidad de Chile, Santiago, Beauchef 851, Casilla 170-3, Santiago, Chile.
| |
Collapse
|
28
|
Yan Q, Liu X, Liu Y, Cai K, Huang J, Zhang B, Wang Z, Han Z, Wen J, Li H. Decoy EPS layers for trapping and killing bacteria. Int J Biol Macromol 2025:144479. [PMID: 40412689 DOI: 10.1016/j.ijbiomac.2025.144479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 04/04/2025] [Accepted: 05/20/2025] [Indexed: 05/27/2025]
Abstract
Here we report a novel strategy using bacterial extracellular polymeric substances (EPS) as decoys to enhance bacterial adhesion and contact-based antimicrobial activity. EPS extracted from Staphylococcus aureus and Bacillus subtilis was used to coat wafers as a conditioning layer to alter surface properties and facilitate bacterial aggregation. Results show that EPS downregulates quorum sensing-related genes (agr and atl in Staphylococcus aureus by 80.5 % and 86.6 %, respectively; fliC in Escherichia coli by ~58.3 %), suggesting that EPS facilitates energy-efficient adhesion independent of quorum sensing signals. Loading antibiotics (erythromycin, linezolid, levofloxacin) into the EPS layer further enhances adhesion and contact killing. Especially, the surfaces loaded with a levofloxacin concentration of 2 μg/mL exhibit a significant antimicrobial effect. For Staphylococcus aureus, the antimicrobial rate reaches 83.66 % after 4 h incubation but drops to 39.9 % after 8 h incubation. In contract, Escherichia coli exhibits greater sensitivity, with antibacterial activity increasing to 92.97 % after 8 h incubation. Laser confocal microscopy characterization further reveals that the antibiotic-loaded EPS surfaces possess remarkable contact bacteria-killing activity. Our results show the promising recruiting-killing efficacy of the antibiotics-loaded EPS against bacteria, which would give insight into exploring new antibacterial strategies for enhanced contact-antibacterial performances.
Collapse
Affiliation(s)
- Qi Yan
- Zhejiang-Japan Joint Laboratory for Antibacterial and Antifouling Technology, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo 315201, China; Zhejiang Engineering Research Center for Biomedical Materials Laboratory of Advanced Theranostic Materials and Technology, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo 315201, China; Cixi Biomedical Research Institute, Wenzhou Medical University, Ningbo 315300, China
| | - Xiaomei Liu
- Zhejiang-Japan Joint Laboratory for Antibacterial and Antifouling Technology, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo 315201, China; Zhejiang Engineering Research Center for Biomedical Materials Laboratory of Advanced Theranostic Materials and Technology, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo 315201, China.
| | - Yi Liu
- Zhejiang-Japan Joint Laboratory for Antibacterial and Antifouling Technology, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo 315201, China; Zhejiang Engineering Research Center for Biomedical Materials Laboratory of Advanced Theranostic Materials and Technology, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo 315201, China
| | - Kedan Cai
- The Department of Nephrology, Ningbo No.2Hospital, Ningbo 315000, China
| | - Jing Huang
- Zhejiang-Japan Joint Laboratory for Antibacterial and Antifouling Technology, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo 315201, China; Zhejiang Engineering Research Center for Biomedical Materials Laboratory of Advanced Theranostic Materials and Technology, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo 315201, China
| | - Botao Zhang
- Zhejiang-Japan Joint Laboratory for Antibacterial and Antifouling Technology, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo 315201, China; Zhejiang Engineering Research Center for Biomedical Materials Laboratory of Advanced Theranostic Materials and Technology, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo 315201, China
| | - Zhijuan Wang
- Zhejiang-Japan Joint Laboratory for Antibacterial and Antifouling Technology, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo 315201, China; Zhejiang Engineering Research Center for Biomedical Materials Laboratory of Advanced Theranostic Materials and Technology, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo 315201, China
| | - Zhuoyue Han
- Zhejiang-Japan Joint Laboratory for Antibacterial and Antifouling Technology, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo 315201, China; Zhejiang Engineering Research Center for Biomedical Materials Laboratory of Advanced Theranostic Materials and Technology, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo 315201, China
| | - Jianxin Wen
- Zhejiang-Japan Joint Laboratory for Antibacterial and Antifouling Technology, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo 315201, China; Zhejiang Engineering Research Center for Biomedical Materials Laboratory of Advanced Theranostic Materials and Technology, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo 315201, China
| | - Hua Li
- Zhejiang-Japan Joint Laboratory for Antibacterial and Antifouling Technology, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo 315201, China; Zhejiang Engineering Research Center for Biomedical Materials Laboratory of Advanced Theranostic Materials and Technology, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo 315201, China.
| |
Collapse
|
29
|
Garcia-Maset R, Chu V, Yuen N, Blumgart D, Yoon J, Murray BO, Joseph AA, Rohn JL. Effect of host microenvironment and bacterial lifestyles on antimicrobial sensitivity and implications for susceptibility testing. NPJ ANTIMICROBIALS AND RESISTANCE 2025; 3:42. [PMID: 40399473 PMCID: PMC12095824 DOI: 10.1038/s44259-025-00113-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Accepted: 05/01/2025] [Indexed: 05/23/2025]
Abstract
Bacterial infections remain a major global health issue, with antimicrobial resistance (AMR) worsening the crisis. However, treatment failure can occur even when bacteria show antibiotic susceptibility in diagnostic tests. We explore factors such as phenotypic resilience, bacterial lifestyles such as biofilms, and differences between laboratory tests and real infection sites, highlighting the need for improved platforms to better predict treatment outcomes, and reviewing emerging technologies aimed at improving susceptibility testing.
Collapse
Affiliation(s)
- Ramon Garcia-Maset
- Centre for Urological Biology, Department of Renal Medicine, Division of Medicine, University College London, London, WC1E 6BT, UK.
| | - Victoria Chu
- Centre for Urological Biology, Department of Renal Medicine, Division of Medicine, University College London, London, WC1E 6BT, UK
| | - Nicholas Yuen
- Centre for Urological Biology, Department of Renal Medicine, Division of Medicine, University College London, London, WC1E 6BT, UK
| | - Dalia Blumgart
- Centre for Urological Biology, Department of Renal Medicine, Division of Medicine, University College London, London, WC1E 6BT, UK
| | - Jenny Yoon
- Centre for Urological Biology, Department of Renal Medicine, Division of Medicine, University College London, London, WC1E 6BT, UK
| | - Benjamin O Murray
- Centre for Urological Biology, Department of Renal Medicine, Division of Medicine, University College London, London, WC1E 6BT, UK
| | - Amelia A Joseph
- Nottingham University Hospitals NHS Trust, Nottingham, NG5 1PB, UK
| | - Jennifer L Rohn
- Centre for Urological Biology, Department of Renal Medicine, Division of Medicine, University College London, London, WC1E 6BT, UK.
| |
Collapse
|
30
|
Alhosani F, Islayem D, Almansoori S, Zaka A, Nayfeh L, Rezk A, Yousef AF, Pappa AM, Nayfeh A. Antibiofilm activity of ZnO-Ag nanoparticles against Pseudomonas aeruginosa. Sci Rep 2025; 15:17321. [PMID: 40389571 PMCID: PMC12089420 DOI: 10.1038/s41598-025-02372-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Accepted: 05/13/2025] [Indexed: 05/21/2025] Open
Abstract
Biofilm-related infections remain a major concern in clinical settings due to the increasing challenge of antimicrobial resistance to conventional antimicrobial treatments. Surface coatings of nanomaterials that can effectively prevent biofilm formation and disrupt established biofilms are essential to addressing this challenge. In this study, a ZnO-Ag nanocomposite was synthesized via a dry chemical method and characterized using XRD, XPS, TEM, SEM-EDX, and AFM, confirming the presence of highly crystalline and pure ZnO and Ag nanoparticles with sharp nanoscale features. The nanocomposite demonstrated potent antibiofilm activity against Pseudomonas aeruginosa, a common Gram-negative biofilm-forming pathogen. Surface-coated glass slides prevented initial biofilm formation, while treatment with higher nanocomposite concentrations (≥ 0.25 g/L) significantly disrupted pre-formed biofilms and altered biofilm architecture, as shown by SEM and crystal violet assays. Mechanistic investigations suggested that nanoparticle surface sharpness may contribute to membrane disruption, and EPR analysis confirmed the generation of reactive oxygen species (ROS), particularly superoxide and methyl radicals, under light exposure. These results highlight the composite's strong potential for integration into surfaces prone to bacterial colonization, offering a practical approach for reducing biofilm-related complications.
Collapse
Affiliation(s)
- Fatima Alhosani
- Department of Electrical Engineering, Khalifa University, Abu Dhabi, United Arab Emirates
| | - Deema Islayem
- Department of Biomedical Engineering, Khalifa University, Abu Dhabi, United Arab Emirates
- Biotechnology Center (BTC), Khalifa University, Abu Dhabi, United Arab Emirates
| | - Shamma Almansoori
- Department of Biomedical Engineering, Khalifa University, Abu Dhabi, United Arab Emirates
| | - Awais Zaka
- Department of Chemical Engineering, Khalifa University, Abu Dhabi, United Arab Emirates
- Research and Innovation Center for Graphene and 2D Materials (RIC-2D), Khalifa University, Abu Dhabi, United Arab Emirates
| | - Laith Nayfeh
- Department of Electrical Engineering, Khalifa University, Abu Dhabi, United Arab Emirates
| | - Ayman Rezk
- Department of Biomedical Engineering, Khalifa University, Abu Dhabi, United Arab Emirates
- Research and Innovation Center for Graphene and 2D Materials (RIC-2D), Khalifa University, Abu Dhabi, United Arab Emirates
| | - Ahmed F Yousef
- Department of Biological Sciences, Khalifa University, Abu Dhabi, United Arab Emirates
- Biotechnology Center (BTC), Khalifa University, Abu Dhabi, United Arab Emirates
- Center for Membranes and Advanced Water Technology (CMAT), Khalifa University, Abu Dhabi, United Arab Emirates
| | - Anna Maria Pappa
- Department of Biomedical Engineering, Khalifa University, Abu Dhabi, United Arab Emirates
| | - Ammar Nayfeh
- Department of Electrical Engineering, Khalifa University, Abu Dhabi, United Arab Emirates.
- Research and Innovation Center for Graphene and 2D Materials (RIC-2D), Khalifa University, Abu Dhabi, United Arab Emirates.
| |
Collapse
|
31
|
Stachura P, Lu Z, Kronberg RM, Xu HC, Liu W, Tu JW, Schaal K, Kameri E, Picard D, von Karstedt S, Fischer U, Bhatia S, Lang PA, Borkhardt A, Pandyra AA. Deep transfer learning approach for automated cell death classification reveals novel ferroptosis-inducing agents in subsets of B-ALL. Cell Death Dis 2025; 16:396. [PMID: 40382332 DOI: 10.1038/s41419-025-07704-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 04/23/2025] [Accepted: 04/29/2025] [Indexed: 05/20/2025]
Abstract
Ferroptosis is a recently described type of regulated necrotic cell death whose induction has anti-cancer therapeutic potential, especially in hematological malignancies. However, efforts to uncover novel ferroptosis-inducing therapeutics have been largely unsuccessful. In the current investigation, we classified brightfield microscopy images of tumor cells undergoing defined modes of cell death using deep transfer learning (DTL). The trained DTL network was subsequently combined with high-throughput pharmacological screening approaches using automated live cell imaging to identify novel ferroptosis-inducing functions of the polo-like kinase inhibitor volasertib. Secondary validation showed that subsets of B-cell acute lymphoblastic leukemia (B-ALL) cell lines, namely 697, NALM6, HAL01, REH and primary patient B-ALL samples were sensitive to ferroptosis induction by volasertib. This was accompanied by an upregulation of ferroptosis-related genes post-volasertib treatment in cell lines and patient samples. Importantly, using several leukemia models, we determined that volasertib delayed tumor growth and induced ferroptosis in vivo. Taken together, we have applied DTL to automated live-cell imaging in pharmacological screening to identify novel ferroptosis-inducing functions of a clinically relevant anti-cancer therapeutic.
Collapse
Affiliation(s)
- Paweł Stachura
- Department of Pediatric Oncology, Hematology and Clinical Immunology, Medical Faculty, Heinrich-Heine-University, Moorenstrasse 5, 40225, Düsseldorf, Germany
- Department of Molecular Medicine II, Medical Faculty, Heinrich-Heine-University, Universitätsstraße 1, 40225, Düsseldorf, Germany
- Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Düsseldorf, Germany
- German Consortium for Translational Cancer Research (DKTK), partner site Essen/Düsseldorf, Düsseldorf, Germany
| | - Zhe Lu
- Department of Pediatric Oncology, Hematology and Clinical Immunology, Medical Faculty, Heinrich-Heine-University, Moorenstrasse 5, 40225, Düsseldorf, Germany
- Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Düsseldorf, Germany
- German Consortium for Translational Cancer Research (DKTK), partner site Essen/Düsseldorf, Düsseldorf, Germany
| | - Raphael M Kronberg
- Department of Molecular Medicine II, Medical Faculty, Heinrich-Heine-University, Universitätsstraße 1, 40225, Düsseldorf, Germany
- Mathematical Modelling of Biological Systems, Heinrich Heine University, Düsseldorf, North Rhine-Westphalia, Germany
- Deep-Sea Ecology and Technology, Alfred Wegener Institute Helmholtz Centre for Polar and Marine Research, Bremerhaven, Germany
| | - Haifeng C Xu
- Department of Molecular Medicine II, Medical Faculty, Heinrich-Heine-University, Universitätsstraße 1, 40225, Düsseldorf, Germany
| | - Wei Liu
- Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Düsseldorf, Germany
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
- German Center for Infection Research (DZIF), Partner Site Bonn-Cologne, Bonn, Germany
| | - Jia-Wey Tu
- Department of Pediatric Oncology, Hematology and Clinical Immunology, Medical Faculty, Heinrich-Heine-University, Moorenstrasse 5, 40225, Düsseldorf, Germany
- Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Düsseldorf, Germany
- German Consortium for Translational Cancer Research (DKTK), partner site Essen/Düsseldorf, Düsseldorf, Germany
| | - Katerina Schaal
- Department of Pediatric Oncology, Hematology and Clinical Immunology, Medical Faculty, Heinrich-Heine-University, Moorenstrasse 5, 40225, Düsseldorf, Germany
- Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Düsseldorf, Germany
- German Consortium for Translational Cancer Research (DKTK), partner site Essen/Düsseldorf, Düsseldorf, Germany
| | - Ersen Kameri
- Department of Pediatric Oncology, Hematology and Clinical Immunology, Medical Faculty, Heinrich-Heine-University, Moorenstrasse 5, 40225, Düsseldorf, Germany
- Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Düsseldorf, Germany
- German Consortium for Translational Cancer Research (DKTK), partner site Essen/Düsseldorf, Düsseldorf, Germany
- Cancer Prevention Graduate School (CPGS), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Daniel Picard
- Department of Pediatric Oncology, Hematology and Clinical Immunology, Medical Faculty, Heinrich-Heine-University, Moorenstrasse 5, 40225, Düsseldorf, Germany
- German Consortium for Translational Cancer Research (DKTK), partner site Essen/Düsseldorf, Düsseldorf, Germany
- Division of Pediatric Neuro-Oncogenomics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Silvia von Karstedt
- Department of Translational Genomics, Faculty of Medicine and University Hospital Cologne, University of Cologne, Weyertal 115b, Cologne, 50931, Germany
- CECAD Cluster of Excellence, Faculty of Medicine and University Hospital Cologne, University of Cologne, Joseph-Stelzmann-Straße 26, Cologne, 50931, Germany
- Center for Molecular Medicine Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Robert-Koch-Straße 21, Cologne, 50931, Germany
| | - Ute Fischer
- Department of Pediatric Oncology, Hematology and Clinical Immunology, Medical Faculty, Heinrich-Heine-University, Moorenstrasse 5, 40225, Düsseldorf, Germany
- Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Düsseldorf, Germany
- German Consortium for Translational Cancer Research (DKTK), partner site Essen/Düsseldorf, Düsseldorf, Germany
- Cancer Prevention Graduate School (CPGS), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Sanil Bhatia
- Department of Pediatric Oncology, Hematology and Clinical Immunology, Medical Faculty, Heinrich-Heine-University, Moorenstrasse 5, 40225, Düsseldorf, Germany
- Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Düsseldorf, Germany
- German Consortium for Translational Cancer Research (DKTK), partner site Essen/Düsseldorf, Düsseldorf, Germany
| | - Philipp A Lang
- Department of Molecular Medicine II, Medical Faculty, Heinrich-Heine-University, Universitätsstraße 1, 40225, Düsseldorf, Germany
| | - Arndt Borkhardt
- Department of Pediatric Oncology, Hematology and Clinical Immunology, Medical Faculty, Heinrich-Heine-University, Moorenstrasse 5, 40225, Düsseldorf, Germany
- Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Düsseldorf, Germany
- German Consortium for Translational Cancer Research (DKTK), partner site Essen/Düsseldorf, Düsseldorf, Germany
| | - Aleksandra A Pandyra
- Department of Pediatric Oncology, Hematology and Clinical Immunology, Medical Faculty, Heinrich-Heine-University, Moorenstrasse 5, 40225, Düsseldorf, Germany.
- Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Düsseldorf, Germany.
- German Consortium for Translational Cancer Research (DKTK), partner site Essen/Düsseldorf, Düsseldorf, Germany.
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Venusberg-Campus 1, 53127, Bonn, Germany.
- German Center for Infection Research (DZIF), Partner Site Bonn-Cologne, Bonn, Germany.
| |
Collapse
|
32
|
Gao C, Li R, Li Y, Wu Y, Qu Y, Ampomah-Wireko M, Zheng J, Wang Z, Wang YN, Zhang E. Design, synthesis and evaluation of quinolone quaternary ammonium antibacterial agent with killing ability to biofilm. Bioorg Chem 2025; 162:108579. [PMID: 40383012 DOI: 10.1016/j.bioorg.2025.108579] [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: 12/26/2024] [Revised: 05/08/2025] [Accepted: 05/09/2025] [Indexed: 05/20/2025]
Abstract
Staphylococcus aureus (S. aureus) is the most common and widely distributed pathogenic bacterium. The problem of methicillin-resistant Staphylococcus aureus (MRSA) caused by the widespread use of antibiotics is particularly severe. In addition, S. aureus can resist antibiotics by forming biofilms, making clinical treatment difficult. A series of antimicrobial quinolone-based quaternary ammonium compounds were designed and synthesized. Among them, the optimal compound 3e showed the strongest activity against S. aureus, and it had relatively low hemolytic toxicity and cytotoxicity. Compound 3e has excellent bactericidal performance, capable of quickly and thoroughly sterilizing. In continuous sub-lethal concentration bacterial passage culture, no bacterial resistance tendency caused by 3e was found. Moreover, 3e can exert a significant level of activity in blood components and still has a period of suppression on bacteria after the drug is removed. Encouragingly, 3e has a certain bactericidal potential against bacteria with high concentration and high tolerance. It has shown strong bactericidal effects when fighting against persister bacteria and biofilms in vitro. Mechanism research indicates that 3e exerts its antimicrobial action through related membrane activity and is related to membrane components phosphatidylglycerol (PG) and cardiolipin (CL). In addition, 3e can also bind to bacterial DNA.
Collapse
Affiliation(s)
- Chen Gao
- School of Pharmaceutical Sciences, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou 450001, PR China
| | - Ruirui Li
- School of Pharmaceutical Sciences, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou 450001, PR China
| | - Yuanbo Li
- School of Pharmaceutical Sciences, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou 450001, PR China
| | - Yuequan Wu
- School of Pharmaceutical Sciences, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou 450001, PR China
| | - Ye Qu
- School of Pharmaceutical Sciences, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou 450001, PR China
| | - Maxwell Ampomah-Wireko
- School of Pharmaceutical Sciences, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou 450001, PR China
| | - Jiangbo Zheng
- School of Pharmaceutical Sciences, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou 450001, PR China
| | - Zhenya Wang
- School of Pharmaceutical Sciences, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou 450001, PR China
| | - Ya-Na Wang
- School of Pharmaceutical Sciences, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou 450001, PR China.
| | - En Zhang
- School of Pharmaceutical Sciences, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou 450001, PR China; Pingyuan Laboratory (Zhengzhou University), PR China.
| |
Collapse
|
33
|
Nangpal P, Nagpal NL, Angrish N, Khare G. Model systems to study Mycobacterium tuberculosis infections: an overview of scientific potential and impediments. Front Cell Infect Microbiol 2025; 15:1572547. [PMID: 40406522 PMCID: PMC12095297 DOI: 10.3389/fcimb.2025.1572547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Accepted: 04/04/2025] [Indexed: 05/26/2025] Open
Abstract
Despite years of global efforts to combat tuberculosis (TB), Mycobacterium tuberculosis (Mtb), the causative agent of this disease, continues to haunt the humankind making TB elimination a distant task. To comprehend the pathogenic nuances of this organism, various in vitro, ex vivo and in vivo experimental models have been employed by researchers. This review focuses on the salient features as well as pros and cons of various model systems employed for TB research. In vitro and ex vivo macrophage infection models have been extensively used for studying Mtb physiology. Animal models have provided us with great wealth of information and have immensely contributed to the understanding of TB pathogenesis and host responses during infection. Additionally, they have been used for evaluation of anti-mycobacterial drug therapy as well as for determining the efficacy of potential vaccine candidates. Advancements in various 'omics' based approaches have enhanced our understanding about the host-pathogen interface. Although animal models have been the cornerstone to TB research, none of them is ideal that gives us a complete picture of human infection, disease and progression. Further, the review also discusses about the newer systems including three dimensional (3D)-tissue models, lung-on-chip infection model, in vitro TB granuloma model and their limitations for studying TB. Thus, converging information gained from various in vitro and ex vivo models in tandem with in vivo experiments will ultimately bridge the gap that exists in understanding human TB.
Collapse
Affiliation(s)
| | | | | | - Garima Khare
- Department of Biochemistry, University of Delhi, New Delhi, India
| |
Collapse
|
34
|
Yu H, Xue T, Mao X. Chinese herbal extracts mediated programmed cell death in cancer and inflammation therapy. J Leukoc Biol 2025; 117:qiaf051. [PMID: 40313183 DOI: 10.1093/jleuko/qiaf051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 12/13/2024] [Accepted: 04/30/2025] [Indexed: 05/03/2025] Open
Abstract
Programmed cell death is a common phenomenon in the development of organisms. It is an active and orderly mode of cell death determined by genes. Programmed cell death is usually classified into 3 different types according to the cell morphological changes, stimulus, and biochemical pathways involved, namely, apoptosis, programmed necrosis, and autophagy. Chinese herbal extracts, mainly obtained from traditional Chinese medicine and their primary plants through the physicochemical extraction and separation process, are concentrated with 1 or more effective ingredients from the herbal materials. Recently, studies focused on the influence of traditional Chinese medicine on programmed cell death are increasing, involving the protection of the nervous system and cardio-cerebrovascular system, the prevention of gastrointestinal and immune function damage, the treatment against tumors, and so on. This review mainly focuses on the effects of Chinese herbal extracts on various types of programmed cell death. In addition, the therapeutic approaches and prospects of CHEs are also discussed. Although there are promising clinical applications of Chinese herbal extracts, some challenges are still waiting to be overcome by further research for the wider use of Chinese herbal extracts in clinical practice.
Collapse
Affiliation(s)
- Haihong Yu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, 999078
| | - Tingmao Xue
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, 999078
| | - Xiaowen Mao
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, 999078
- Department of Pharmaceutical Sciences, Faculty of Health Science, University of Macau, Macao SAR, 999078, China
| |
Collapse
|
35
|
Huang H, Chen Y, Zhang L, Wan P, Chen Y, Li Y, Zeng Z. Identification and characterization of a novel major facilitator superfamily (MFS) efflux pump conferring multidrug resistance in Staphylococcus aureus and Staphylococcus epidermidis. Antimicrob Agents Chemother 2025; 69:e0173924. [PMID: 40192467 PMCID: PMC12057375 DOI: 10.1128/aac.01739-24] [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: 11/21/2024] [Accepted: 03/06/2025] [Indexed: 05/08/2025] Open
Abstract
A novel major facilitator superfamily (MFS) efflux pump in Staphylococcus, designated Nms, was identified via topology prediction. The secondary structure indicated the presence of 12 transmembrane segments (TMSs) and characteristic motif A of MFS efflux pumps. Experimental verification of efflux activity was conducted using ethidium bromide accumulation and efflux assays and biofilm formation assays. Antimicrobial susceptibility testing and efflux pump inhibition confirmed that Nms effectively effluxed various antimicrobial agents to confer multidrug resistance. Comprehensive genomic analyses were used to assess the prevalence and possible origins of the nms gene. The results revealed that the nms gene was present in Staphylococcus aureus ST398/ST541 and Staphylococcus epidermidis ST570/ST1166 strains from global isolates. The transmission of nms was associated with the prevalence of S. aureus ST398-t571 in swine-derived samples from China. Phylogenetic analysis revealed that nms-positive strains formed a distinct clade separate from other S. aureus ST398 strains. Genetic analysis of the nms gene revealed a significant presence of plasmid-related mobile genetic elements, with extended nucleotide sequences containing circular intermediates exhibiting high homology with those found in an S. aureus plasmid. These findings suggested that the nms gene likely initially originated from plasmids and subsequently integrated into chromosomes. In conclusion, Nms is a novel MFS efflux pump that confers multidrug resistance to S. aureus and has been carried predominantly by ST398-t571 isolates in recent years. Ongoing surveillance is essential to elucidate the origin of nms in S. aureus, particularly MRSA ST398-t571, and to understand the transmission among humans, animals, and the environment.
Collapse
Affiliation(s)
- Honghao Huang
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Yiyi Chen
- Department of Infectious Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Lingxuan Zhang
- Institute of Quality Standard and Monitoring Technology for Agro-products, Guangdong Academy of Agricultural Sciences, Guangzhou, Guangdong, China
| | - Peng Wan
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Yan Chen
- Department of Infectious Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yafei Li
- Institute of Quality Standard and Monitoring Technology for Agro-products, Guangdong Academy of Agricultural Sciences, Guangzhou, Guangdong, China
| | - Zhenling Zeng
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| |
Collapse
|
36
|
Reynolds J, Yoon JY. Fluorescence-based spectrometric and imaging methods and machine learning analyses for microbiota analysis. Mikrochim Acta 2025; 192:334. [PMID: 40323435 DOI: 10.1007/s00604-025-07159-0] [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] [Accepted: 04/06/2025] [Indexed: 06/11/2025]
Abstract
Most microbiota determination (skin, gut, soil, etc.) are currently conducted in a laboratory using expensive equipment and lengthy procedures, including culture-dependent methods, nucleic acid amplifications (including quantitative PCR), DNA microarray, immunoassays, 16S rRNA sequencing, shotgun metagenomics, and sophisticated mass spectrometric methods. In situ and rapid analysis methods are desirable for fast turnaround time and low assay cost. Fluorescence identification of bacteria and their mixtures is emerging to meet this demand, thanks to the recent development in various machine learning methods. High-dimensional spectroscopic or microscopic imaging data can be obtained to identify the bacterial makeup and its implications for human health and the environment. For example, we can classify healthy versus non-healthy skin microbiome, inflammatory versus non-inflammatory gut microbiome, degraded versus non-degraded soil microbiome, etc. This tutorial summarizes the various machine-learning algorithms used in bacteria identification and microbiota determinations. It also summarizes the various fluorescence spectroscopic methods used to identify bacteria and their mixtures, including fluorescence lifetime spectroscopy, fluorescence resonance energy transfer (FRET), and synchronous fluorescence (SF) spectroscopy. Finally, various fluorescence microscopic imaging methods were summarized that have been used to identify bacteria and their mixtures, including epi-fluorescence microscopy, confocal microscopy, two-photon/multi-photon microscopy, and super-resolution imaging methods (STED, SIM, PALM, and STORM). Finally, it discusses how these methods can be applied to microbiota determinations, what can be demonstrated in the future, opportunities and challenges, and future directions.
Collapse
Affiliation(s)
- Jocelyn Reynolds
- Department of Biomedical Engineering, The University of Arizona, Tucson, AZ, 85721, USA
| | - Jeong-Yeol Yoon
- Department of Biomedical Engineering, The University of Arizona, Tucson, AZ, 85721, USA.
| |
Collapse
|
37
|
Banerjee R. Tiny but Mighty: Small RNAs-The Micromanagers of Bacterial Survival, Virulence, and Host-Pathogen Interactions. Noncoding RNA 2025; 11:36. [PMID: 40407594 PMCID: PMC12101431 DOI: 10.3390/ncrna11030036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2025] [Revised: 04/07/2025] [Accepted: 04/28/2025] [Indexed: 05/26/2025] Open
Abstract
Bacterial pathogens have evolved diverse strategies to infect hosts, evade immune responses, and establish successful infections. While the role of transcription factors in bacterial virulence is well documented, emerging evidence highlights the significant contribution of small regulatory RNAs (sRNAs) in bacterial pathogenesis. These sRNAs function as posttranscriptional regulators that fine-tune gene expression, enabling bacteria to adapt rapidly to challenging environments. This review explores the multifaceted roles of bacterial sRNAs in host-pathogen interactions. Firstly, it examines how sRNAs regulate pathogenicity by modulating the expression of key virulence factors, including fimbriae, toxins, and secretion systems, followed by discussing the role of sRNAs in bacterial stress response mechanisms that counteract host immune defenses, such as oxidative and envelope stress. Additionally, this review investigates the involvement of sRNAs in antibiotic resistance by regulating efflux pumps, biofilm formation, and membrane modifications, which contribute to multi-drug resistance phenotypes. Lastly, this review highlights how sRNAs contribute to intra- and interspecies communication through quorum sensing, thereby coordinating bacterial behavior in response to environmental cues. Understanding these regulatory networks governed by sRNAs is essential for the development of innovative antimicrobial strategies. This review highlights the growing significance of sRNAs in bacterial pathogenicity and explores their potential as therapeutic targets for the treatment of bacterial infections.
Collapse
Affiliation(s)
- Rajdeep Banerjee
- Department of Biomolecular Chemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| |
Collapse
|
38
|
Verma N, Bajiya M, Dolhey R, Surabhi, Yadav AS, Chaudhary C, Meena D, Arya H, Bhatt TK, Yadav JK, Shukla JN, Swaroop S, Pandey J. Mechanistic Insights into the Antibiofilm Activity of Simvastatin and Lovastatin against Bacillus subtilis. Mol Pharm 2025; 22:2703-2722. [PMID: 40100146 DOI: 10.1021/acs.molpharmaceut.5c00191] [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: 03/20/2025]
Abstract
Statins have been reported for diverse pleiotropic activities, including antimicrobial and antibiofilm. However, due to the limited understanding of their mode of action, none of the statins have gained approval for antimicrobial or antibiofilm applications. In a recent drug repurposing study, we observed that two statins (i.e., Simvastatin and Lovastatin) interact stably with TasA(28-261), the principal extracellular matrix protein of Bacillus subtilis, and also induce inhibition of biofilm formation. Nevertheless, the underlying mechanism remained elusive. In the present study, we examined the impact of these statins on the physiological activity of TasA(28-261), specifically its interaction with TapA(33-253) and aggregation into the amyloid-like structure using purified recombinant TasA(28-261) and TapA(33-253) in amyloid detection-specific in vitro assays (i.e., CR binding and ThT staining assays). Results revealed that both statins interfered with amyloid formation by the TasA(28-261)-TapA(33-253) complex, while neither statin inhibited amyloid formation by lysozyme, a model amyloid-forming protein. Moreover, neither statin significantly altered the expressions of terminal regulatory genes (viz, sinR, sinI) and terminal effector genes (viz, tasA, tapA, and bslA) involved in biofilm formation by B. subtilis. While the intricate interplay between Simvastatin and Lovastatin with the diverse molecular constituents of B. subtilis biofilm remains to be elucidated conclusively, the findings obtained during the present study suggest that the underlying mechanism for Simvastatin- and Lovastatin-mediated inhibition of B. subtilis biofilm formation is manifested by interfering with the aggregation and amyloid formation by TasA(28-261)-TapA(33-253). These results represent one of the first experimental evidence for the underlying mechanism of antibiofilm activity of statins and offer valuable directions for future research to harness statins as antibiofilm therapeutics.
Collapse
Affiliation(s)
- Nidhi Verma
- Department of Biotechnology, Central University of Rajasthan, Kishangarh, Ajmer 305801, Rajasthan, India
| | - Mamta Bajiya
- Department of Biotechnology, Central University of Rajasthan, Kishangarh, Ajmer 305801, Rajasthan, India
| | - Ragini Dolhey
- Department of Biotechnology, Central University of Rajasthan, Kishangarh, Ajmer 305801, Rajasthan, India
| | - Surabhi
- Department of Biotechnology, Central University of Rajasthan, Kishangarh, Ajmer 305801, Rajasthan, India
| | - Abhishek Singh Yadav
- Department of Biotechnology, Central University of Rajasthan, Kishangarh, Ajmer 305801, Rajasthan, India
| | - Chhavi Chaudhary
- Department of Biotechnology, Central University of Rajasthan, Kishangarh, Ajmer 305801, Rajasthan, India
| | - Dhankesh Meena
- Department of Biochemistry, Central University of Rajasthan, Kishangarh, Ajmer 305801, Rajasthan, India
| | - Hemant Arya
- Department of Biotechnology, Central University of Rajasthan, Kishangarh, Ajmer 305801, Rajasthan, India
| | - Tarun K Bhatt
- Department of Biotechnology, Central University of Rajasthan, Kishangarh, Ajmer 305801, Rajasthan, India
| | - Jay Kant Yadav
- Department of Biotechnology, Central University of Rajasthan, Kishangarh, Ajmer 305801, Rajasthan, India
| | - Jayendra Nath Shukla
- Department of Biotechnology, Central University of Rajasthan, Kishangarh, Ajmer 305801, Rajasthan, India
| | - Shiv Swaroop
- Department of Biochemistry, Central University of Rajasthan, Kishangarh, Ajmer 305801, Rajasthan, India
| | - Janmejay Pandey
- Department of Biotechnology, Central University of Rajasthan, Kishangarh, Ajmer 305801, Rajasthan, India
| |
Collapse
|
39
|
Bell I PJ, Muniyan R. Synergistic pathogenesis: exploring biofilms, efflux pumps and secretion systems in Acinetobacter baumannii and Staphylococcus aureus. Arch Microbiol 2025; 207:134. [PMID: 40314822 DOI: 10.1007/s00203-025-04336-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2025] [Revised: 04/03/2025] [Accepted: 04/17/2025] [Indexed: 05/03/2025]
Abstract
Antimicrobial resistance (AMR) is a growing global health crisis, particularly among ESKAPE pathogens: Enterococcus faecium, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa, and Enterobacter species. Among them, A. baumannii and S. aureus are major contributors to nosocomial infections, with high prevalence in intensive care units and immunocompromised patients. Their ability to resist multiple antibiotic classes complicates treatment strategies, leading to increased morbidity and mortality. Key resistance mechanisms, including biofilm formation, efflux pump activity, and horizontal gene transfer, enhance their survival and persistence. Furthermore, interactions during polymicrobial infections intensify disease severity through synergistic effects that promote both virulence and resistance. The epidemiological burden of these pathogens highlights the urgent need for novel antimicrobial strategies and targeted interventions. This review explores their virulence factors, resistance mechanisms, pathogenic interactions, and clinical implications, emphasizing the necessity of innovative therapeutic approaches to combat their growing threat.
Collapse
Affiliation(s)
- Praisy Joy Bell I
- School of Bio Sciences and Technology (SBST), Vellore Institute of Technology, Vellore, 632014, Tamil Nadu, India
| | - Rajiniraja Muniyan
- School of Bio Sciences and Technology (SBST), Vellore Institute of Technology, Vellore, 632014, Tamil Nadu, India.
| |
Collapse
|
40
|
Ampomah-Wireko M, Qu Y, Li D, Wu Y, Li R, Li Y, Kong H, Li ZH, Wang YN, Zhang E. Design, synthesis and antibacterial evaluation of oxazolidinone derivatives containing N-methylglycyl or quaternary ammonium salts. Bioorg Med Chem 2025; 122:118144. [PMID: 40056888 DOI: 10.1016/j.bmc.2025.118144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 02/20/2025] [Accepted: 02/28/2025] [Indexed: 03/10/2025]
Abstract
The continuous evolution of multidrug-resistant (MDR) bacteria to existing antibiotic treatment regimens poses a serious threat to human health, so the discovery of new and potent antimicrobial drugs that are less likely to develop resistance is of great clinical significance. As a result, oxazolidinone antibiotics have emerged as a significant class of bacterial protein synthesis inhibitors, with particular success in the treatment of MDR Gram-positive infections. Herein, a series of novel C-ring modified oxazolidinone derivatives with the introduction of N-methylglycyl groups or quaternary ammonium salts were synthesized and evaluated for their antibacterial efficacy, among which most of the N-methylglycyl derivatives showed significant activity against E. faecalis. Notably, compounds 11g-11i showed good activity against E. faecalis and S. aureus with MICs of 2-8 μg/mL. The selected compound 11g exhibited rapid bactericidal properties, good biofilm disruption capacity, low tendency to induce bacterial resistance, and low cytotoxicity against mammalian cells (HeLa). Furthermore, compound 11g showed relatively good stability in mammalian body fluids and exhibited a longer post-antibiotic effect (PAE). Mechanistic studies showed that compound 11g exerted its antibacterial effect by inhibiting glutathione (GSH) activity and inducing reactive oxygen species (ROS) accumulation, leading to bacterial death. These findings suggest that 11g is a promising candidate for the exploitation of N-methylglycyl oxazolidinones as novel antibacterial agents.
Collapse
Affiliation(s)
- Maxwell Ampomah-Wireko
- School of Pharmaceutical Sciences, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou 450001, PR China
| | - Ye Qu
- School of Pharmaceutical Sciences, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou 450001, PR China
| | - Daran Li
- School of Pharmaceutical Sciences, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou 450001, PR China
| | - Yuequan Wu
- School of Pharmaceutical Sciences, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou 450001, PR China
| | - Ruirui Li
- School of Pharmaceutical Sciences, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou 450001, PR China
| | - Yuanbo Li
- School of Pharmaceutical Sciences, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou 450001, PR China
| | - Hongtao Kong
- School of Pharmaceutical Sciences, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou 450001, PR China
| | - Zhi-Hao Li
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, PR China
| | - Ya-Na Wang
- School of Pharmaceutical Sciences, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou 450001, PR China.
| | - En Zhang
- School of Pharmaceutical Sciences, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou 450001, PR China; Pingyuan Laboratory (Zhengzhou University), PR China.
| |
Collapse
|
41
|
Shah T, Zhu C, Shah C, Upadhyaya I, Upadhyay A. Trans-cinnamaldehyde nanoemulsion reduces Salmonella Enteritidis biofilm on steel and plastic surfaces and downregulates expression of biofilm associated genes. Poult Sci 2025; 104:105086. [PMID: 40168703 PMCID: PMC11997393 DOI: 10.1016/j.psj.2025.105086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 03/20/2025] [Accepted: 03/21/2025] [Indexed: 04/03/2025] Open
Abstract
Salmonella Enteritidis is a major poultry-associated foodborne pathogen that can form sanitizer-tolerant biofilms on various surfaces. The biofilm-forming capability of S. Enteritidis facilitates its survival on farm and food processing equipment. Conventional sanitization methods are not completely effective in killing S. Enteritidis biofilms. This study investigated the efficacy of a Generally Recognized as Safe phytochemical Trans-cinnamaldehyde (TC), and in its nanoemulsion form (TCNE), for inhibiting S. Enteritidis biofilm formation and inactivating mature biofilms developed on polystyrene and stainless-steel surfaces. Moreover, the effect of TC on Salmonella genes critical for biofilm formation was studied. TCNE was prepared using a high energy sonication method with Tween 80. For biofilm inhibition assay, S. Enteritidis was allowed to form biofilms either in the presence or absence of sub-inhibitory concentration (SIC; 0.01 %) of TCNE at 25°C and the biofilm formed was quantified at 24-h intervals for 48 h. For the inactivation assay, S. Enteritidis biofilms developed at 25°C for 48 h were exposed to TCNE (0.5, 1 %) for 1, 5, and 15 min, and surviving S. Enteritidis in the biofilm were enumerated. SIC of TCNE inhibited S. Enteritidis biofilm by 45 % on polystyrene and 75 % on steel surface after 48 h at 25°C compared to control (P < 0.05). All TCNE treatments rapidly inactivated S. Enteritidis mature biofilm on polystyrene and steel surfaces (P < 0.05). The lower concentration of TCNE (0.5 %) reduced S. Enteritidis counts by 1.5 log CFU/ml as early as 1 min of exposure on both polystyrene and stainless-steel surfaces. After 15 min of exposure, TCNE at concentration of 0.5 or 1 % reduced S. Enteritidis count significantly by 4.5 log CFU or 6 log CFU/ml on polystyrene or stainless-steel surfaces. TC downregulated the expression of S. Enteritidis genes (hilA, hilC, flhD, csgA, csgD, sdiA) responsible for biofilm formation (P < 0.05). Results suggest that TCNE has potential as a natural disinfectant for controlling S. Enteritidis biofilms on common farm and food processing surfaces, such as plastic and steel.
Collapse
Affiliation(s)
- Trushenkumar Shah
- Department of Animal Science, University of Connecticut, Storrs, Connecticut, USA
| | - Chen Zhu
- Department of Animal Science, University of Connecticut, Storrs, Connecticut, USA
| | - Chetna Shah
- Department of Animal Science, University of Connecticut, Storrs, Connecticut, USA
| | - Indu Upadhyaya
- Department of Extension, University of Connecticut, Storrs, Connecticut, USA
| | - Abhinav Upadhyay
- Department of Animal Science, University of Connecticut, Storrs, Connecticut, USA.
| |
Collapse
|
42
|
Valença-Pereira F, Sheridan RM, Riemondy KA, Thornton T, Fang Q, Barret B, Paludo G, Thompson C, Collins P, Santiago M, Oltz E, Rincon M. Inactivation of GSK3β by Ser 389 phosphorylation prevents thymocyte necroptosis and impacts Tcr repertoire diversity. Cell Death Differ 2025; 32:880-898. [PMID: 39779909 DOI: 10.1038/s41418-024-01441-z] [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: 06/18/2024] [Revised: 12/11/2024] [Accepted: 12/27/2024] [Indexed: 01/11/2025] Open
Abstract
The assembly of Tcrb and Tcra genes require double negative (DN) thymocytes to undergo multiple rounds of programmed DNA double-strand breaks (DSBs), followed by their efficient repair. However, mechanisms governing cell cycle checkpoints and specific survival pathways during the repair process remain unclear. Here, we report high-resolution scRNA-seq analyses of individually sorted mouse DN3 and DN4 thymocytes, which reveals a G2M cell cycle checkpoint, in addition to the known G1 checkpoint, during Tcrb and Tcra recombination. We also show that inactivation of GSK3β by phosphorylation on Ser389 is essential for DN3/DN4 thymocytes to survive while being stalled at the G1 and G2/M checkpoints. GSK3β promotes death by necroptosis, but not by apoptosis, of DN3/DN4 thymocytes during V(D)J recombination. Failure to inactivate GSK3β in DN3 thymocytes alters the Tcrb gene repertoire primarily through Trbv segment utilization. In addition, preferential recombination of proximal V segments in Tcra depends on GSK3β inactivation. Our study identifies a unique thymocyte survival pathway, enabling them to undergo cell cycle checkpoints for DNA repair during V(D)J recombination of Tcrb and Tcra genes. Thymocyte survival during cell cycle checkpoints for V(D)J recombination DNA repair determines TCRα/β repertoire.
Collapse
Affiliation(s)
- Felipe Valença-Pereira
- Department of Immunology and Microbiology, University of Colorado, Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Ryan M Sheridan
- Department of Biochemistry and Molecular Genetics, RNA Bioscience Initiative, University of Colorado, Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Kent A Riemondy
- Department of Biochemistry and Molecular Genetics, RNA Bioscience Initiative, University of Colorado, Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Tina Thornton
- Department of Medicine, Immunobiology Division, University of Vermont, Burlington, VT, 05405, USA
| | - Qian Fang
- Department of Immunology and Microbiology, University of Colorado, Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Brad Barret
- Division of Infectious Diseases, Department of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Gabriela Paludo
- Department of Pharmacosciences, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, RS, 90050-170, Brazil
| | - Claudia Thompson
- Department of Pharmacosciences, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, RS, 90050-170, Brazil
| | - Patrick Collins
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, 43210, USA
| | - Mario Santiago
- Division of Infectious Diseases, Department of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Eugene Oltz
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, 43210, USA
| | - Mercedes Rincon
- Department of Immunology and Microbiology, University of Colorado, Anschutz Medical Campus, Aurora, CO, 80045, USA.
- Department of Medicine, Immunobiology Division, University of Vermont, Burlington, VT, 05405, USA.
| |
Collapse
|
43
|
Muttiah B, Hanafiah A. Snake Venom Compounds: A New Frontier in the Battle Against Antibiotic-Resistant Infections. Toxins (Basel) 2025; 17:221. [PMID: 40423304 DOI: 10.3390/toxins17050221] [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: 03/18/2025] [Revised: 04/20/2025] [Accepted: 04/30/2025] [Indexed: 05/28/2025] Open
Abstract
The occurrence of antibiotic-resistant bacteria is a serious global health issue, and it emphasizes the need for novel antimicrobial agents. This review explores the potential of snake venom as another alternative strategy against antimicrobial resistance. Snake venoms are complex combinations of bioactive peptides and proteins, including metalloproteases (MPs), serine proteases (SPs), phospholipase A2 (PLA2) enzymes, three-finger toxins (3FTXs), cysteine-rich secretory proteins (CRISPs), L-amino acid oxidases (LAAOs), and antimicrobial peptides (AMPs). The antibacterial products possess wide-spectrum antibacterial activity against resistant microbes via diverse mechanisms such as cell membrane disruption, enzymatic hydrolysis of microbial structures, generation of oxidative stress, inhibition of biofilms, and immunomodulation. Strong antimicrobial activity is reported by most studies, but these are mostly restricted to in vitro testing with low translational use. Although preliminary insights into molecular targets and physiological effects exist, further studies are needed to clarify long-term safety and therapeutic potential. Special attention is given to snake venom-derived extracellular vesicles (SVEVs), which enhance the therapeutic potential of venom toxins by protecting them from degradation, improving bioavailability, and facilitating targeted delivery. Furthermore, innovative delivery strategies such as PEGylation, liposomes, hydrogels, microneedle patches, biopolymer films, and nanoparticles are discussed for their role in reducing systemic toxicity and enhancing antimicrobial efficacy. The rational modification of venom-derived peptides further expands their therapeutic utility by improving pharmacokinetics and minimizing off-target effects. Together, these approaches highlight the translational potential of snake venom-based therapies as next-generation antimicrobials in the fight against resistant infections. By outlining these challenges and directions, this review positions snake venom as an overlooked but fertile resource in the battle against antibiotic resistance.
Collapse
Affiliation(s)
- Barathan Muttiah
- Department of Medical Microbiology and Immunology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia
| | - Alfizah Hanafiah
- Department of Medical Microbiology and Immunology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia
- GUT Research Group, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
| |
Collapse
|
44
|
Lucena RPS, Silva-Junior AG, Frías IAM, Gil LHV, Cordeiro MT, El Salhi AE, Andrade CAS, Oliveira MDL. Microcontact printing of lectin self-assembled monolayers for arbovirus detection. Biotechnol Prog 2025; 41:e70008. [PMID: 39968671 DOI: 10.1002/btpr.70008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 12/11/2024] [Accepted: 01/29/2025] [Indexed: 02/20/2025]
Abstract
Arboviruses significantly burden public health in Brazil, constituting a constant challenge for health authorities. The diagnosis and, consequently, clinical management and the reporting of arbovirus infections in regions where multiple arboviruses coexist are complex processes. Herein, we report the development of a new electrochemical biosensor based on Concanavalin A (ConA) to identify carbohydrate patterns in the viral structure of Dengue 3 (DENV-3), Zika (ZIKV) and Chikungunya (CHIKV) viruses. The biorecognition of arboviruses was carried out through functionalization with 4-aminophenylacetic acid (CMA) on poly (ethylene terephthalate) (PET) substrate coated with a gold layer combining microcontact printing (μCP). Bovine serum albumin (BSA) was used after ConA immobilization to block binding to nonspecific sites. Subsequently, the interaction between ConA and arbovirus was characterized by standard atomic force microscopy (AFM), fluorescence microscopy, cyclic voltammetry (CV), and electrochemical impedance spectroscopy (EIS). Fluorescent imaging was conducted to confirm the occurrence of the DENV-3, ZIKV, and CHIKV detection processes. The obtained results demonstrated the success of the biosensor (CMA-ConA-BSA) manufactured on a PET substrate using μCP for detecting medically significant arboviruses. RCT values showed an increase in impedimetric response total of the system after exposition to DENV-3 (RCT = 68.82 kΩ) and a lower recognition to CHIKV (RCT = 44.44 kΩ). The present biosensor platform reveals the applicability of the ConA lectin in the viral biorecognition process based on flexible biosensors for differential detection of DENV-3, ZIKV, and CHIKV. ConA-based electrochemical biosensor provide high selectivity, real-time detection, and low volumes of analytes.
Collapse
Affiliation(s)
- Raiza P S Lucena
- Programa de Pós-Graduação em Ciências Biológicas, Universidade Federal de Pernambuco, Recife, Brazil
- Laboratório de Biodispositivos Nanoestruturados, Departamento de Bioquímica, Universidade Federal de Pernambuco, Recife, Brazil
| | - Alberto G Silva-Junior
- Laboratório de Biodispositivos Nanoestruturados, Departamento de Bioquímica, Universidade Federal de Pernambuco, Recife, Brazil
| | - Isaac A M Frías
- Institut des Sciences Analytiques, Claude Bernard University Lyon 1, Lyon, France
| | - Laura H V Gil
- Departamento de Virologiam, Instituto Aggeu Magalhães-Fiocruz, Recife, Brazil
| | - Marli T Cordeiro
- Departamento de Virologiam, Instituto Aggeu Magalhães-Fiocruz, Recife, Brazil
| | | | - Cesar A S Andrade
- Programa de Pós-Graduação em Ciências Biológicas, Universidade Federal de Pernambuco, Recife, Brazil
| | - Maria D L Oliveira
- Programa de Pós-Graduação em Ciências Biológicas, Universidade Federal de Pernambuco, Recife, Brazil
- Laboratório de Biodispositivos Nanoestruturados, Departamento de Bioquímica, Universidade Federal de Pernambuco, Recife, Brazil
| |
Collapse
|
45
|
Sharma N, Joshi B, Sharma B, Kumar S, Mohanty KK, Prakash H. Customized MHC Class I & II restricted peptides from clinical isolates of Mycobacterium tuberculosis tweak strong cellular immune response in Healthy individuals and Pulmonary Tuberculosis patients: A potential candidate in vaccine design. Tuberculosis (Edinb) 2025; 152:102640. [PMID: 40262464 DOI: 10.1016/j.tube.2025.102640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 03/27/2025] [Accepted: 04/07/2025] [Indexed: 04/24/2025]
Abstract
Tuberculosis (TB) remains a global health challenge as annual mortality rate due to drug resistant TB is increasing exponentially. This is mostly associated with the delayed diagnosis of Multidrug-resistant (MDR) or latent TB. Effective management of TB demands development of novel immunological strategies, such as peptide-based/subunit vaccines that can stimulate specific immune responses. In this context, we evaluated the immunogenic potential of two Major Histocompatibility Complex (MHC) Class I/II-restricted peptides from Mycobacterium tuberculosis (M. tuberculosis): Rv2588c and Rv0148. The peptides were tested on T and monocyte populations from healthy donors and pulmonary TB (PTB) patients. Flow cytometry analysis revealed significant T cell activation in peripheral blood mononuclear cells (PBMC) from both groups. Enzyme-linked immunosorbent assay (ELISA) demonstrated a strong IFN-γ response, confirming effective T cell activation. Additionally, these peptides induced increased nitric oxide (NO) production in macrophages, indicating their role in activating the innate immune system. Overall, Rv2588c and Rv0148 peptides exhibited robust immunogenicity, stimulating both adaptive and innate immune responses in PBMCs from healthy and PTB individuals. These findings highlight their potential as promising TB vaccine candidates, paving the way for improved TB treatment and prevention strategies.
Collapse
Affiliation(s)
- Niharika Sharma
- Department of Immunology, ICMR- National JALMA Institute for Leprosy and Other Mycobacterial Diseases, Agra, Uttar Pradesh, India; Amity Centre for Translational Research, Amity University, Noida, Uttar Pradesh, India
| | - Beenu Joshi
- Department of Immunology, ICMR- National JALMA Institute for Leprosy and Other Mycobacterial Diseases, Agra, Uttar Pradesh, India
| | - Bhawna Sharma
- Department of Immunology, ICMR- National JALMA Institute for Leprosy and Other Mycobacterial Diseases, Agra, Uttar Pradesh, India
| | - Santosh Kumar
- TB and Chest Department, Sarojini Naidu Medial College, Agra, Uttar Pradesh, India
| | - Keshar Kunja Mohanty
- Department of Immunology, ICMR- National JALMA Institute for Leprosy and Other Mycobacterial Diseases, Agra, Uttar Pradesh, India
| | - Hridayesh Prakash
- Amity Centre for Translational Research, Amity University, Noida, Uttar Pradesh, India.
| |
Collapse
|
46
|
S Y, K L M, Harithpriya K, Zong C, Sahabudeen S, Ichihara G, Ramkumar KM. Disruptive multiple cell death pathways of bisphenol-A. Toxicol Mech Methods 2025; 35:430-443. [PMID: 39815394 DOI: 10.1080/15376516.2024.2449423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 12/26/2024] [Accepted: 12/30/2024] [Indexed: 01/18/2025]
Abstract
Endocrine-disrupting chemicals (EDCs) significantly contribute to health issues by interfering with hormonal functions. Bisphenol A (BPA), a prominent EDC, is extensively utilized as a monomer and plasticizer in producing polycarbonate plastic and epoxy resins, making it one of the highest-demanded chemicals in commercial use. This is the major component used in plastic products, including bottles, containers, storage items, and food serving ware. Exposure of BPA happens through oral, respiratory, transdermal routes and eye contact. As an EDC, BPA disrupts hormonal binding, leading to various health problems, such as cancers, reproductive abnormalities, metabolic syndrome, immune dysfunction, neurological effects, cardiovascular problems, respiratory issues, and obesity. BPA mimics the hormone estrogen but exhibits a weak affinity for estrogen receptors. This weak binding affinity triggers multiple cell death pathways, including necroptosis, pyroptosis, apoptosis, ferroptosis, and autophagy, across different cell types. Numerous clinical, in-vitro, and in-vivo experiments have demonstrated that BPA exposure results in unfavorable health effects. This review highlights the mechanisms of cell death pathways initiated through BPA exposure and the associated negative health consequences. The extensive use of BPA and its frequent detection in environmental and biological models underscore the urgent need for further investigation into its effects and the development of safe alternatives. Addressing the health risks posed by BPA involves a comprehensive approach that includes reducing exposure and finding novel substitutes to lessen its detrimental impact on humans.
Collapse
Affiliation(s)
- Yukta S
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, India
| | - Milan K L
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, India
| | - Kannan Harithpriya
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, India
| | - Cai Zong
- Department of Occupational and Environmental Health, Faculty of Pharmaceutical Science, Tokyo University of Science, Tokyo, Japan
| | - S Sahabudeen
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, India
| | - Gaku Ichihara
- Department of Occupational and Environmental Health, Faculty of Pharmaceutical Science, Tokyo University of Science, Tokyo, Japan
| | - K M Ramkumar
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, India
| |
Collapse
|
47
|
Liu H, Nan Z, Zhao C, Bai L, Shi L, He C, Wu D, Wan M, Feng Y. Emerging synergistic strategies for enhanced antibacterial sonodynamic therapy: Advances and prospects. ULTRASONICS SONOCHEMISTRY 2025; 116:107288. [PMID: 40038013 PMCID: PMC11986242 DOI: 10.1016/j.ultsonch.2025.107288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 12/29/2024] [Accepted: 02/22/2025] [Indexed: 03/06/2025]
Abstract
Antibacterial therapy has been extensively applied in medical field to alleviate the severity and mortality of infection. However, it still exists some issues such as drug side effects, limited efficacy and bacterial resistance. Among the alternative therapies, antibacterial sonodynamic therapy (aSDT) has been explored as a promising approach to tackle those crises. It is meaningful to investigate superior strategy to augment the therapeutic efficacy of aSDT. This review summarizes the potential aSDT-based antibacterial mechanisms and comprehensively discusses the prevailing synergistic strategies, such as nanomaterials-based aSDT antibacterial strategy, aSDT + strategy with physical, chemical and biological methods. Moreover, we also reviewed the medical applications of aSDT strategies. Finally, the perspectives on the current challenges that need be resolved in aSDT are proposed. We expect that this review could provide robust support to expedite the clinical applications of aSDT.
Collapse
Affiliation(s)
- Hengyu Liu
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Department of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Zhezhu Nan
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Department of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Chen Zhao
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Department of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Liang Bai
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Department of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Linrong Shi
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Department of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Chenhui He
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Department of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Daocheng Wu
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Department of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Mingxi Wan
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Department of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Yi Feng
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Department of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, People's Republic of China.
| |
Collapse
|
48
|
Liu Z, Pan C, Huang H. The role of axon guidance molecules in the pathogenesis of epilepsy. Neural Regen Res 2025; 20:1244-1257. [PMID: 39075893 PMCID: PMC11624883 DOI: 10.4103/nrr.nrr-d-23-01620] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 12/06/2023] [Accepted: 02/21/2024] [Indexed: 07/31/2024] Open
Abstract
Current treatments for epilepsy can only manage the symptoms of the condition but cannot alter the initial onset or halt the progression of the disease. Consequently, it is crucial to identify drugs that can target novel cellular and molecular mechanisms and mechanisms of action. Increasing evidence suggests that axon guidance molecules play a role in the structural and functional modifications of neural networks and that the dysregulation of these molecules is associated with epilepsy susceptibility. In this review, we discuss the essential role of axon guidance molecules in neuronal activity in patients with epilepsy as well as the impact of these molecules on synaptic plasticity and brain tissue remodeling. Furthermore, we examine the relationship between axon guidance molecules and neuroinflammation, as well as the structural changes in specific brain regions that contribute to the development of epilepsy. Ample evidence indicates that axon guidance molecules, including semaphorins and ephrins, play a fundamental role in guiding axon growth and the establishment of synaptic connections. Deviations in their expression or function can disrupt neuronal connections, ultimately leading to epileptic seizures. The remodeling of neural networks is a significant characteristic of epilepsy, with axon guidance molecules playing a role in the dynamic reorganization of neural circuits. This, in turn, affects synapse formation and elimination. Dysregulation of these molecules can upset the delicate balance between excitation and inhibition within a neural network, thereby increasing the risk of overexcitation and the development of epilepsy. Inflammatory signals can regulate the expression and function of axon guidance molecules, thus influencing axonal growth, axon orientation, and synaptic plasticity. The dysregulation of neuroinflammation can intensify neuronal dysfunction and contribute to the occurrence of epilepsy. This review delves into the mechanisms associated with the pathogenicity of axon guidance molecules in epilepsy, offering a valuable reference for the exploration of therapeutic targets and presenting a fresh perspective on treatment strategies for this condition.
Collapse
Affiliation(s)
- Zheng Liu
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou Province, China
| | - Chunhua Pan
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou Province, China
| | - Hao Huang
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou Province, China
| |
Collapse
|
49
|
Cernicchi G, Di Gregorio A, Felicetti T, Rampacci E, Casari G, Armeni T, Romaldi B, Zefaj E, Passamonti F, Massari S, Manfroni G, Barreca ML, Tabarrini O, Vignaroli C, Sabatini S. NorA Efflux Pump Inhibitors: Expanding SAR Knowledge of Pyrazolo[4,3-c][1,2]benzothiazine 5,5-Dioxide Derivatives. Arch Pharm (Weinheim) 2025; 358:e70000. [PMID: 40390184 PMCID: PMC12089509 DOI: 10.1002/ardp.70000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 03/19/2025] [Accepted: 04/07/2025] [Indexed: 05/21/2025]
Abstract
Antimicrobial resistance (AMR) represents a significant global concern, driven by the overuse of antibiotics. One of the principal mechanisms contributing to AMR is the activity of microbial efflux pumps (EPs), which expel diverse antibiotics out of bacterial cells, thereby rendering them ineffective. Our research aimed to expand the range of molecular classes that inhibit the Staphylococcus aureus EP NorA. In this study, starting from the hit compound pyrazolo[4,3-c][1,2]benzothiazine 5,5-dioxide 1, previously reported as a NorA efflux pump inhibitor (EPI), we undertook medicinal chemistry efforts, which involved the iterative combination of the design and synthesis of new analogues with data obtained through ethidium bromide efflux inhibition assays. Subsequent synergistic assays with ciprofloxacin (CPX) against the resistant strain SA-1199B led to the identification of three potent compounds (3, 10, and 19). The evaluation of these compounds in combination with CPX against NorA-overexpressing and NorA-knockout strains (SA-K2378 and SA-K1902, respectively) confirmed that the observed synergy with CPX is dependent on the presence of NorA. Additionally, the combination of NorA EPIs with CPX reduced biofilm production in NorA-overexpressing strains. These findings enhance our understanding of the structure-activity relationship of pyrazolobenzothiazine derivatives and support the use of EtBr efflux assays for rapid NorA inhibitors' identification.
Collapse
Affiliation(s)
- Giada Cernicchi
- Department of Pharmaceutical SciencesUniversità degli Studi di PerugiaPerugiaItaly
| | - Alessandra Di Gregorio
- Department of Life and Environmental ScienceUniversità Politecnica delle MarcheAnconaItaly
| | - Tommaso Felicetti
- Department of Pharmaceutical SciencesUniversità degli Studi di PerugiaPerugiaItaly
| | - Elisa Rampacci
- Department of Veterinary MedicineUniversità degli Studi di PerugiaPerugiaItaly
| | - Giulia Casari
- Department of Specialized Clinical Sciences and OdontostomatologyUniversità Politecnica delle MarcheTorrette di AnconaItaly
| | - Tatiana Armeni
- Department of Specialized Clinical Sciences and OdontostomatologyUniversità Politecnica delle MarcheTorrette di AnconaItaly
| | - Brenda Romaldi
- Department of Specialized Clinical Sciences and OdontostomatologyUniversità Politecnica delle MarcheTorrette di AnconaItaly
| | - Ermelinda Zefaj
- Department of Pharmaceutical SciencesUniversità degli Studi di PerugiaPerugiaItaly
| | - Fabrizio Passamonti
- Department of Veterinary MedicineUniversità degli Studi di PerugiaPerugiaItaly
| | - Serena Massari
- Department of Pharmaceutical SciencesUniversità degli Studi di PerugiaPerugiaItaly
| | - Giuseppe Manfroni
- Department of Pharmaceutical SciencesUniversità degli Studi di PerugiaPerugiaItaly
| | | | - Oriana Tabarrini
- Department of Pharmaceutical SciencesUniversità degli Studi di PerugiaPerugiaItaly
| | - Carla Vignaroli
- Department of Life and Environmental ScienceUniversità Politecnica delle MarcheAnconaItaly
| | - Stefano Sabatini
- Department of Pharmaceutical SciencesUniversità degli Studi di PerugiaPerugiaItaly
| |
Collapse
|
50
|
Nabawy A, Chattopadhyay AN, Makabenta JMV, Hassan MA, Yang J, Park J, Jiang M, Jeon T, Im J, Rotello VM. Cationic conjugated polymers with tunable hydrophobicity for efficient treatment of multidrug-resistant wound biofilm infections. Biomaterials 2025; 316:123015. [PMID: 39705926 PMCID: PMC11755787 DOI: 10.1016/j.biomaterials.2024.123015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 11/25/2024] [Accepted: 12/13/2024] [Indexed: 12/23/2024]
Abstract
Biofilm-associated infections arising from antibiotic-resistant bacteria pose a critical challenge to global health. We report the generation of a library of cationic conjugated poly(phenylene ethynylene) (PPE) polymers featuring trimethylammonium terminated sidechains with tunable hydrophobicity. Screening of the library identified an amphiphilic polymer with a C11 hydrophobic spacer as the polymer with the highest antimicrobial efficacy against biofilms in the dark with excellent selectivity. These polymers are highly fluorescent, allowing label-free monitoring of polymer-bacteria/biofilm interactions. The amphiphilic conjugated polymer penetrated the biofilm matrix in vitro and eradicated resident bacteria through membrane disruption. This C11 polymer was likewise effective in an in vivo murine model of antibiotic-resistant wound biofilm infections, clearing >99.9 % of biofilm colonies and efficient alleviation of biofilm-associated inflammation. The results demonstrate the therapeutic potential of the fluorescent conjugated polymer platform as a multi-modal antimicrobial and imaging tool, surpassing conventional antimicrobial strategies against resilient biofilm infection.
Collapse
Affiliation(s)
- Ahmed Nabawy
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, MA, 01003, USA
| | - Aritra Nath Chattopadhyay
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, MA, 01003, USA
| | - Jessa Marie V Makabenta
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, MA, 01003, USA
| | - Muhammad Aamir Hassan
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, MA, 01003, USA
| | - Junwhee Yang
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, MA, 01003, USA
| | - Jungmi Park
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, MA, 01003, USA
| | - Mingdi Jiang
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, MA, 01003, USA
| | - Taewon Jeon
- Molecular and Cellular Biology Graduate Program, University of Massachusetts Amherst, 230 Stockbridge Road, Amherst, MA, 01003, USA
| | - Jungkyun Im
- Department of Chemical Engineering, and Department of Electronic Materials, Devices, and Equipment Engineering, Soonchunhyang University, 22 Soonchunhyangro, Asan, 31538, Republic of Korea
| | - Vincent M Rotello
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, MA, 01003, USA.
| |
Collapse
|