1
|
Peng T, Chen Y, Luan X, Hu W, Wu W, Guo B, Lu C, Wu C, Pan X. Microneedle technology for enhanced topical treatment of skin infections. Bioact Mater 2025; 45:274-300. [PMID: 39659727 PMCID: PMC11629152 DOI: 10.1016/j.bioactmat.2024.11.027] [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: 01/20/2024] [Revised: 10/31/2024] [Accepted: 11/20/2024] [Indexed: 12/12/2024] Open
Abstract
Skin infections caused by microbes such as bacteria, fungi, and viruses often lead to aberrant skin functions and appearance, eventually evolving into a significant risk to human health. Among different drug administration paradigms for skin infections, microneedles (MNs) have demonstrated superiority mainly because of their merits in enhancing drug delivery efficiency and reducing microbial resistance. Also, integrating biosensing functionality to MNs offers point-of-care wearable medical devices for analyzing specific pathogens, disease status, and drug pharmacokinetics, thus providing personalized therapy for skin infections. Herein, we do a timely update on the development of MN technology in skin infection management, with a special focus on how to devise MNs for personalized antimicrobial therapy. Notably, the advantages of state-of-the-art MNs for treating skin infections are pointed out, which include hijacking sequential drug transport barriers to enhance drug delivery efficiency and delivering various therapeutics (e.g., antibiotics, antimicrobial peptides, photosensitizers, metals, sonosensitizers, nanoenzyme, living bacteria, poly ionic liquid, and nanomoter). In addition, the nanoenzyme-based multimodal antimicrobial therapy is highlighted in addressing intractable infectious wounds. Furthermore, the MN-based biosensors used to identify pathogen types, track disease status, and quantify antibiotic concentrations are summarized. The limitations of antimicrobial MNs toward clinical translation are offered regarding large-scale production, quality control, and policy guidance. Finally, the future development of biosensing MNs with easy-to-use and intelligent properties and MN-based wearable drug delivery for home-based therapy are prospected. We hope this review will provide valuable guidance for future development in MN-mediated topical treatment of skin infections.
Collapse
Affiliation(s)
- Tingting Peng
- State Key Laboratory of Bioactive Molecules and Druggability Assessment/ International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Ministry of Education (MOE) of China/College of Pharmacy, Jinan University, Guangzhou 511436, China
| | - Yangyan Chen
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Xuanyu Luan
- Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, London, W12 0NN, UK
| | - Wanshan Hu
- State Key Laboratory of Bioactive Molecules and Druggability Assessment/ International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Ministry of Education (MOE) of China/College of Pharmacy, Jinan University, Guangzhou 511436, China
| | - Wentao Wu
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Bing Guo
- School of Science, Harbin Institute of Technology, Shenzhen 518055, China
| | - Chao Lu
- State Key Laboratory of Bioactive Molecules and Druggability Assessment/ International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Ministry of Education (MOE) of China/College of Pharmacy, Jinan University, Guangzhou 511436, China
| | - Chuanbin Wu
- State Key Laboratory of Bioactive Molecules and Druggability Assessment/ International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Ministry of Education (MOE) of China/College of Pharmacy, Jinan University, Guangzhou 511436, China
| | - Xin Pan
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| |
Collapse
|
2
|
Yousfan A, Al Khatib AO, Salman AMH, Abu Elella MH, Barrett G, Michael N, Zariwala MG, Al-Obaidi H. Innovative Microencapsulation of Polymyxin B for Enhanced Antimicrobial Efficacy via Coated Spray Drying. Mol Pharm 2025; 22:113-130. [PMID: 39378315 PMCID: PMC11707731 DOI: 10.1021/acs.molpharmaceut.4c00594] [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: 05/29/2024] [Revised: 09/21/2024] [Accepted: 09/24/2024] [Indexed: 10/10/2024]
Abstract
This study aims to develop an innovative microencapsulation method for coated Polymyxin B, utilizing various polysaccharides such as hydroxypropyl β-cyclodextrin, alginate, and chitosan, implemented through a three-fluid nozzle (3FN) spray drying process. High-performance liquid chromatography (HPLC) analysis revealed that formulations with a high ratio of sugar cage, hydroxypropyl β-cyclodextrin (HPβCD), and sodium alginate (coded as ALGHCDHPLPM) resulted in a notable 16-fold increase in Polymyxin B recovery compared to chitosan microparticles. Morphological assessments using fluorescence labeling confirmed successful microparticle formation with core/shell structures. Alginate-based formulations exhibited distinct layers, while chitosan formulations showed uniform fluorescence throughout the microparticles. Focused beam reflectance and histograms from fluorescence microscopic measurements provided insights into physical size analysis, indicating consistent sizes of 6.8 ± 1.2 μm. Fourier-transform infrared (FTIR) spectra unveiled hydrogen bonding between Polymyxin B and other components within the microparticle structures. The drug release study showed sodium alginate's sustained release capability, reaching 26 ± 3% compared to 94 ± 3% from the free solution at the 24 h time point. Furthermore, the antimicrobial properties of the prepared microparticles against two Gram-negative bacteria, Escherichia coli and Pseudomonas aeruginosa, were investigated. The influence of various key excipients on the minimum inhibitory concentration (MIC) and minimum bactericidal concentration (MBC) values was evaluated. Results demonstrated effective bactericidal effects of ALGHCDHPLPM against both E. coli and P. aeruginosa. Additionally, the antibiofilm assay highlighted the potential efficacy of ALGHCDHPLPM against the biofilm viability of E. coli and P. aeruginosa, with concentrations ranging from 3.9 to 500 μg/m. This signifies a significant advancement in antimicrobial drug delivery systems, promising improved precision and efficacy in combating bacterial infections.
Collapse
Affiliation(s)
- Amal Yousfan
- School
of Pharmacy, University of Reading, Reading RG6 6AD, U.K.
| | | | - Afrah M. H. Salman
- School
of Biological Sciences, University of Reading, Reading RG6 6AD, U.K.
- College
of Pharmacy, Pharmacology and Toxicology Department, Mustansiriyha University, Baghdad 14132, Iraq
| | | | - Glyn Barrett
- School
of Biological Sciences, University of Reading, Reading RG6 6AD, U.K.
| | - Nicholas Michael
- Chemical
Analysis Facility, University of Reading, Reading RG6 6AD, U.K.
| | - Mohammed Gulrez Zariwala
- Centre for
Nutraceuticals, School of Life Sciences, University of Westminster, 115 New, Cavendish Street, London W1W 6UW, U.K.
| | - Hisham Al-Obaidi
- School
of Pharmacy, University of Reading, Reading RG6 6AD, U.K.
| |
Collapse
|
3
|
Guo D, Li Z, Zhang Y, Zhang W, Wang C, Zhang DX, Liu F, Gao Z, Xu B, Wang N. The effect of lambda-cyhalothrin nanocapsules on the gut microbial communities and immune response of the bee elucidates the potential environmental impact of emerging nanopesticides. JOURNAL OF HAZARDOUS MATERIALS 2024; 479:135650. [PMID: 39216249 DOI: 10.1016/j.jhazmat.2024.135650] [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: 02/14/2024] [Revised: 08/22/2024] [Accepted: 08/24/2024] [Indexed: 09/04/2024]
Abstract
Emerging nanopesticides are gradually gaining widespread application in agriculture due to their excellent properties, but their potential risks to pollinating insects are not fully understood. In this study, lambda-cyhalothrin nanocapsules (LC-NCs) were constructed by electrostatic self-assembly method with iron mineralization optimization, and their effects on bee gut microbial communities and host immune-related factors were investigated. Microbiome sequencing revealed that LC-NCs increase the diversity of gut microbial communities and reduce the complexity of network features, disrupting the overall structure of the microbial communities. In addition, LC-NCs also had systemic effects on the immune response of bees, including increased activity of SOD and CAT enzymes and expression of their genes, as well as downregulation of Defensin1. Furthermore, we noticed that the immune system of the host was activated simultaneously with a rise in the abundance of beneficial bacteria in the gut. Our research emphasizes the importance of both the host and gut microbiota of holobiont in revealing the potential risks of LC-NCs to environmental indicators of honey bees, and provides references for exploring the interactions between host-microbiota systems under exogenous stress. At the same time, we hope that more research can focus on the potential impacts of nanopesticides on the ecological environment.
Collapse
Affiliation(s)
- Dezheng Guo
- College of Life Sciences, Shandong Agricultural University, Taian, Shandong 271018, PR China
| | - Zhongyu Li
- College of Life Sciences, Shandong Agricultural University, Taian, Shandong 271018, PR China
| | - Yiwen Zhang
- College of Life Sciences, Shandong Agricultural University, Taian, Shandong 271018, PR China
| | - Wei Zhang
- College of Life Sciences, Shandong Agricultural University, Taian, Shandong 271018, PR China
| | - Chen Wang
- College of Life Sciences, Shandong Agricultural University, Taian, Shandong 271018, PR China
| | - Da-Xia Zhang
- College of Plant Protection, Shandong Agricultural University, Taian, Shandong 271018, PR China
| | - Feng Liu
- College of Plant Protection, Shandong Agricultural University, Taian, Shandong 271018, PR China
| | - Zheng Gao
- College of Life Sciences, Shandong Agricultural University, Taian, Shandong 271018, PR China.
| | - Baohua Xu
- College of Animal Science and Technology, Shandong Agricultural University, Taian, Shandong 271018, PR China.
| | - Ningxin Wang
- College of Plant Protection, Shandong Agricultural University, Taian, Shandong 271018, PR China.
| |
Collapse
|
4
|
Jiang C, Chen J, Xu J, Chen C, Zhu H, Xu Y, Zhao H, Chen J. Integrated analysis reveals NLRC4 as a potential biomarker in sepsis pathogenesis. Genes Immun 2024; 25:397-408. [PMID: 39181981 DOI: 10.1038/s41435-024-00293-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 08/05/2024] [Accepted: 08/13/2024] [Indexed: 08/27/2024]
Abstract
Sepsis remains a significant global health burden and contributor to mortality, yet the precise molecular mechanisms underlying the immune response are not fully elucidated. To gain insight into this issue, we performed a comprehensive analysis using a variety of techniques including bulk RNA sequencing, single-cell RNA sequencing, and enzyme-linked immunosorbent assay (ELISA). We performed enrichment analysis of differentially expressed genes in sepsis and healthy individuals by utilizing Gene Ontology (GO) analysis and indicated significant enrichment of immune-related response. Following Weighted Gene Co-Expression Network Analysis (WGCNA) and protein-protein interaction analysis (PPI) were used to identify key immune-related hub genes and validated by ELISA to show that NLRC4 is highly expressed in sepsis. Additionally, an analysis of scRNA-seq data from newly diagnosed sepsis, sepsis diagnosis at 6 hours, and healthy samples demonstrates a significant increase in both the expression levels and proportions of NLRC4 in sepsis monocytes and neutrophils. In addition, using pySCENIC we identified upstream transcription factors that regulate NLRC4. Our study provides valuable insights into the identification of NLRC4 in peripheral blood as a potential candidate gene for the diagnosis and treatment of sepsis.
Collapse
Affiliation(s)
- Chunhui Jiang
- School of Basic Medical Sciences & Forensic Medicine, Hangzhou Medical College, Hangzhou, Zhejiang, 310013, China
- Taizhou Hospital of Zhejiang, Wenzhou Medical University, Linhai, 318000, China
- Department of Laboratory Medicine, Enze Hospital, Taizhou Enze Medical Center (Group), Taizhou, 318000, China
| | - Jiani Chen
- School of Basic Medical Sciences & Forensic Medicine, Hangzhou Medical College, Hangzhou, Zhejiang, 310013, China
- Taizhou Hospital of Zhejiang, Wenzhou Medical University, Linhai, 318000, China
- Department of Laboratory Medicine, Enze Hospital, Taizhou Enze Medical Center (Group), Taizhou, 318000, China
| | - Jiaqing Xu
- Taizhou Hospital of Zhejiang, Wenzhou Medical University, Linhai, 318000, China
- Department of Laboratory Medicine, Enze Hospital, Taizhou Enze Medical Center (Group), Taizhou, 318000, China
| | - Chen Chen
- Taizhou Hospital of Zhejiang, Wenzhou Medical University, Linhai, 318000, China
- Department of Laboratory Medicine, Enze Hospital, Taizhou Enze Medical Center (Group), Taizhou, 318000, China
| | - Hongguo Zhu
- Taizhou Hospital of Zhejiang, Wenzhou Medical University, Linhai, 318000, China
- Department of Laboratory Medicine, Enze Hospital, Taizhou Enze Medical Center (Group), Taizhou, 318000, China
| | - Yinghe Xu
- Taizhou Hospital of Zhejiang, Wenzhou Medical University, Linhai, 318000, China.
- Department of Laboratory Medicine, Enze Hospital, Taizhou Enze Medical Center (Group), Taizhou, 318000, China.
| | - Hui Zhao
- Taizhou Hospital of Zhejiang, Wenzhou Medical University, Linhai, 318000, China.
- Department of Laboratory Medicine, Enze Hospital, Taizhou Enze Medical Center (Group), Taizhou, 318000, China.
| | - Jiaxi Chen
- School of Basic Medical Sciences & Forensic Medicine, Hangzhou Medical College, Hangzhou, Zhejiang, 310013, China.
- Taizhou Hospital of Zhejiang, Wenzhou Medical University, Linhai, 318000, China.
- Department of Laboratory Medicine, Enze Hospital, Taizhou Enze Medical Center (Group), Taizhou, 318000, China.
| |
Collapse
|
5
|
Cui D, Yu T. Unveiling the glycolysis in sepsis: Integrated bioinformatics and machine learning analysis identifies crucial roles for IER3, DSC2, and PPARG in disease pathogenesis. Medicine (Baltimore) 2024; 103:e39867. [PMID: 39331858 PMCID: PMC11441936 DOI: 10.1097/md.0000000000039867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/29/2024] Open
Abstract
Sepsis, a multifaceted syndrome driven by an imbalanced host response to infection, remains a significant medical challenge. At its core lies the pivotal role of glycolysis, orchestrating immune responses especially in severe sepsis. The intertwined dynamics between glycolysis, sepsis, and immunity, however, have gaps in knowledge with several Crucial genes still shrouded in ambiguity. We harvested transcriptomic profiles from the peripheral blood of 107 septic patients juxtaposed against 29 healthy controls. Delving into this dataset, differential expression analysis shed light on genes distinctly linked to glycolysis in both cohorts. Harnessing the prowess of LASSO regression and SVM-RFE, we isolated Crucial genes, paving the way for a sepsis risk prediction model, subsequently vetted via Calibration and decision curve analysis. Using the CIBERSORT algorithm, we further mapped 22 immune cell subtypes within the septic samples, establishing potential interactions with the delineated Crucial genes. Our efforts unveiled 21 genes intricately tied to glycolysis that exhibited differential expression patterns. Gene set enrichment analysis (GSEA) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses offered insights, spotlighting pathways predominantly associated with oxidative phosphorylation, PPAR signaling pathway, Glycolysis/Gluconeogenesis and HIF-1 signaling pathway. Among the myriad genes, IER3, DSC2, and PPARG emerged as linchpins, their prominence in sepsis further validated through ROC analytics. These sentinel genes demonstrated profound affiliations with various immune cell facets, bridging the complex terrain of glycolysis, sepsis, and immune responses. In line with our endeavor to "unveil the glycolysis in sepsis," the discovery of IER3, DSC2, and PPARG reinforces their cardinal roles in sepsis pathogenesis. These revelations accentuate the intricate dance between glycolysis and immunological shifts in septic conditions, offering novel avenues for therapeutic interventions.
Collapse
Affiliation(s)
- Dongqing Cui
- Emergency Department, Beijing Sixth Hospital, Beijing, China
| | - Tian Yu
- Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
6
|
Hu X, Li D, Li H, Piao Y, Wan H, Zhou T, Karimi M, Zhao X, Li Y, Shi L, Liu Y. Reaction-Induced Self-Assembly of Polymyxin Mitigates Cytotoxicity and Reverses Drug Resistance. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2406156. [PMID: 39022883 DOI: 10.1002/adma.202406156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 07/07/2024] [Indexed: 07/20/2024]
Abstract
Polymyxins have been regarded as an efficient therapeutic against many life-threatening, multidrug resistant Gram-negative bacterial infections; however, the cytotoxicity and emergence of drug resistance associated with polymyxins have greatly hindered their clinical potential. Herein, the reaction-induced self-assembly (RISA) of polymyxins and natural aldehydes in aqueous solution is presented. The resulting assemblies effectively mask the positively charged nature of polymyxins, reducing their cytotoxicity. Moreover, the representative PMBA4 (composed of polymyxin B (PMB) and (E)-2-heptenal (A4)) assemblies demonstrate enhanced binding to Gram-negative bacterial outer membranes and exhibit multiple antimicrobial mechanisms, including increased membrane permeability, elevated bacterial metabolism, suppression of quorum sensing, reduced ATP synthesis, and potential reduction of bacterial drug resistance. Remarkably, PMBA4 assemblies reverse drug resistance in clinically isolated drug-resistant strains of Gram-negative bacteria, demonstrating exceptional efficacy in preventing and eradicating bacterial biofilms. PMBA4 assemblies efficiently eradicate Gram-negative bacterial biofilm infections in vivo and alleviate inflammatory response. This RISA strategy offers a practical and clinically applicable approach to minimize side effects, reverse drug resistance, and prevent the emergence of resistance associated with free polymyxins.
Collapse
Affiliation(s)
- Xiaowen Hu
- Joint Centre of Translational Medicine, Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
- Department of Orthodontics School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang, 325001, China
| | - Dongdong Li
- Joint Centre of Translational Medicine, Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang, 325001, China
| | - Huaping Li
- Joint Centre of Translational Medicine, Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang, 325001, China
| | - Yinzi Piao
- Joint Centre of Translational Medicine, Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang, 325001, China
| | - Hongping Wan
- Center for Sustainable Antimicrobials, Department of Pharmacy, Sichuan Agricultural University, Chengdu, 611130, China
| | - Tieli Zhou
- Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, Department of Clinical Laboratory, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Mahdi Karimi
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, 1449614535, Iran
| | - Xinghong Zhao
- Center for Sustainable Antimicrobials, Department of Pharmacy, Sichuan Agricultural University, Chengdu, 611130, China
| | - Yuanfeng Li
- Joint Centre of Translational Medicine, Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Linqi Shi
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, China
| | - Yong Liu
- Joint Centre of Translational Medicine, Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang, 325001, China
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, China
| |
Collapse
|
7
|
Wang P, Xie C, Zhang Y, Li H, Lu Y, Sun L, Hu X, Nie T, Li C, Li G, Lu X, Pang J, Yang X, Yu L, Li X, Wang X, You X. Negatively charged nanodiscs for the reduction of toxicity and enhanced efficacy of polymyxin B against Acinetobacter baumannii sepsis. Acta Biomater 2024; 184:323-334. [PMID: 38901753 DOI: 10.1016/j.actbio.2024.06.017] [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: 03/14/2024] [Revised: 05/27/2024] [Accepted: 06/12/2024] [Indexed: 06/22/2024]
Abstract
The treatment of sepsis caused by multidrug-resistant (MDR) Gram-negative bacterial infections remains challenging. With these pathogens exhibiting resistance to carbapenems and new generation cephalosporins, the traditional antibiotic polymyxin B (PMB) has reemerged as a critical treatment option. However, its severe neurotoxicity and nephrotoxicity greatly limit the clinical application. Therefore, we designed negatively charged high-density lipoprotein (HDL) mimicking nanodiscs as a PMB delivery system, which can simultaneously reduce toxicity and enhance drug efficacy. The negative charge prevented the PMB release in physiological conditions and binding to cell membranes, significantly reducing toxicity in mammalian cells and mice. Notably, nanodisc-PMB exhibits superior efficacy than free PMB in sepsis induced by carbapenem-resistant Acinetobacter baumannii (CRAB) strains. Nanodisc-PMB shows promise as a treatment for carbapenem-resistant Gram-negative bacterial sepsis, especially caused by Acinetobacter baumannii, and the nanodiscs could be repurposed for other toxic antibiotics as an innovative delivery system. STATEMENT OF SIGNIFICANCE: Multidrug-resistant Gram-negative bacteria, notably carbapenem-resistant Acinetobacter baumannii, currently pose a substantial challenge due to the scarcity of effective treatments, rendering Polymyxins a last-resort antibiotic option. However, their therapeutic application is significantly limited by severe neurotoxic and nephrotoxic side effects. Prevailing polymyxin delivery systems focus on either reducing toxicity or enhancing bioavailability yet fail to simultaneously achieve both. In this scenario, we have developed a distinctive HDL-mimicking nanodisc for polymyxin B, which not only significantly reduces toxicity but also improves efficacy against Gram-negative bacteria, especially in sepsis caused by CRAB. This research offers an innovative drug delivery system for polymyxin B. Such advancement could notably improve the therapeutic landscape and make a significant contribution to the arsenal against these notorious pathogens.
Collapse
Affiliation(s)
- Penghe Wang
- Beijing Key Laboratory of Antimicrobial Agents, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China; Division for Medicinal Microorganisms Related Strains, CAMS Collection Center of Pathogenic Microorganisms, Beijing 100050, China; State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Chunyang Xie
- Beijing Key Laboratory of Antimicrobial Agents, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China; Division for Medicinal Microorganisms Related Strains, CAMS Collection Center of Pathogenic Microorganisms, Beijing 100050, China; State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Youwen Zhang
- Beijing Key Laboratory of Antimicrobial Agents, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China; Division for Medicinal Microorganisms Related Strains, CAMS Collection Center of Pathogenic Microorganisms, Beijing 100050, China; State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Haibin Li
- Beijing Key Laboratory of Antimicrobial Agents, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China; Division for Medicinal Microorganisms Related Strains, CAMS Collection Center of Pathogenic Microorganisms, Beijing 100050, China; State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Yun Lu
- Beijing Key Laboratory of Antimicrobial Agents, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China; Division for Medicinal Microorganisms Related Strains, CAMS Collection Center of Pathogenic Microorganisms, Beijing 100050, China; State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Lang Sun
- Beijing Key Laboratory of Antimicrobial Agents, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China; Division for Medicinal Microorganisms Related Strains, CAMS Collection Center of Pathogenic Microorganisms, Beijing 100050, China; State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Xinxin Hu
- Beijing Key Laboratory of Antimicrobial Agents, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China; Division for Medicinal Microorganisms Related Strains, CAMS Collection Center of Pathogenic Microorganisms, Beijing 100050, China; State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Tongying Nie
- Beijing Key Laboratory of Antimicrobial Agents, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China; Division for Medicinal Microorganisms Related Strains, CAMS Collection Center of Pathogenic Microorganisms, Beijing 100050, China; State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Congran Li
- Beijing Key Laboratory of Antimicrobial Agents, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China; Division for Medicinal Microorganisms Related Strains, CAMS Collection Center of Pathogenic Microorganisms, Beijing 100050, China; State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Guoqing Li
- Beijing Key Laboratory of Antimicrobial Agents, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China; Division for Medicinal Microorganisms Related Strains, CAMS Collection Center of Pathogenic Microorganisms, Beijing 100050, China; State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Xi Lu
- Beijing Key Laboratory of Antimicrobial Agents, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China; Division for Medicinal Microorganisms Related Strains, CAMS Collection Center of Pathogenic Microorganisms, Beijing 100050, China; State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Jing Pang
- Beijing Key Laboratory of Antimicrobial Agents, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China; Division for Medicinal Microorganisms Related Strains, CAMS Collection Center of Pathogenic Microorganisms, Beijing 100050, China; State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Xinyi Yang
- Beijing Key Laboratory of Antimicrobial Agents, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China; Division for Medicinal Microorganisms Related Strains, CAMS Collection Center of Pathogenic Microorganisms, Beijing 100050, China; State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Liyan Yu
- Beijing Key Laboratory of Antimicrobial Agents, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China; Division for Medicinal Microorganisms Related Strains, CAMS Collection Center of Pathogenic Microorganisms, Beijing 100050, China; State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Xue Li
- Beijing Key Laboratory of Antimicrobial Agents, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China; Division for Medicinal Microorganisms Related Strains, CAMS Collection Center of Pathogenic Microorganisms, Beijing 100050, China; State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China.
| | - Xiukun Wang
- Beijing Key Laboratory of Antimicrobial Agents, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China; Division for Medicinal Microorganisms Related Strains, CAMS Collection Center of Pathogenic Microorganisms, Beijing 100050, China; State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China.
| | - Xuefu You
- Beijing Key Laboratory of Antimicrobial Agents, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China; Division for Medicinal Microorganisms Related Strains, CAMS Collection Center of Pathogenic Microorganisms, Beijing 100050, China; State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China.
| |
Collapse
|
8
|
Du F, Yuk SA, Qian Y, Vincent MP, Bobbala S, Abbott TM, Kim H, Li Y, Li H, Yi S, Qiao B, Scott EA. A Biomimetic Multi-Component Subunit Vaccine via Ratiometric Loading of Hierarchical Hydrogels. RESEARCH SQUARE 2024:rs.3.rs-4177821. [PMID: 38746232 PMCID: PMC11092859 DOI: 10.21203/rs.3.rs-4177821/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
The development of subunit vaccines that mimic the molecular complexity of attenuated vaccines has been limited by the difficulty of intracellular co-delivery of multiple chemically diverse payloads at controllable concentrations. We report on hierarchical hydrogel depots employing simple poly(propylene sulfone) homopolymers to enable ratiometric loading of a protein antigen and four physicochemically distinct adjuvants in a hierarchical manner. The optimized vaccine consisted of immunostimulants either adsorbed to or encapsulated within nanogels, which were capable of noncovalent anchoring to subcutaneous tissues. These 5-component nanogel vaccines demonstrated enhanced humoral and cell-mediated immune responses compared to formulations with standard single adjuvant and antigen pairing. The use of a single simple homopolymer capable of rapid and stable loading and intracellular delivery of diverse molecular cargoes holds promise for facile development and optimization of scalable subunit vaccines and complex therapeutic formulations for a wide range of biomedical applications.
Collapse
Affiliation(s)
- Fanfan Du
- Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, USA
| | - Simseok A. Yuk
- Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, USA
| | - Yuan Qian
- Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, USA
| | - Michael P. Vincent
- Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, USA
| | - Sharan Bobbala
- Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, USA
| | - Tirzah M. Abbott
- Northwestern University Atomic and Nanoscale Characterization Experimental Center, Evanston, IL 60208, USA
| | - Hyeohn Kim
- Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, USA
| | - Yang Li
- Department of Chemical and Biological Engineering, Northwestern University, Evanston, Illinois 60201, USA
| | - Haoyu Li
- Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, USA
| | - Sijia Yi
- Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, USA
| | - Baofu Qiao
- Department of Natural Sciences, Baruch Colleg-e, City University of New York, New York, 10010, USA
| | - Evan A. Scott
- Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, USA
- Simpson Querrey Institute, Northwestern University, Chicago, IL 60611, USA
- Chemistry of Life Processes Institute, Northwestern University, Evanston, IL 60208, USA
- Interdisciplinary Biological Sciences Program, Northwestern University, Evanston, IL 60208, USA
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL 60611, USA
| |
Collapse
|
9
|
Cao H, Zhang J, Yang L, Li H, Tian R, Wu H, Li Y, Gu Z. Robust and Multifunctional Therapeutic Nanoparticles against Peritonitis-Induced Sepsis. Biomacromolecules 2024; 25:1133-1143. [PMID: 38226558 DOI: 10.1021/acs.biomac.3c01133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2024]
Abstract
Apart from bacterial growth and endotoxin generation, the excessive production of reactive radicals linked with sepsis also has a substantial impact on triggering an inflammatory response and further treatment failure. Hence, the rational design and fabrication of robust and multifunctional nanoparticles (NPs) present a viable means of overcoming this dilemma. In this study, we used antibiotic polymyxin B (PMB) and antioxidant natural polyphenolic protocatechualdehyde (PCA) to construct robust and multifunctional NPs for sepsis treatment, leveraging the rich chemistries of PCA. The PMB release profile from the NPs demonstrated pH-responsive behavior, which allowed the NPs to exhibit effective bacterial killing and radical scavenging properties. Data from in vitro cells stimulated with H2O2 and lipopolysaccharide (LPS) showed the multifunctionalities of NPs, including intracellular reactive oxygen species (ROS) scavenging, elimination of the bacterial toxin LPS, inhibiting macrophage M1 polarization, and anti-inflammation capabilities. Additionally, in vivo studies further demonstrated that NPs could increase the effectiveness of sepsis treatment by lowering the bacterial survival ratio, the expression of the oxidative marker malondialdehyde (MDA), and the expression of inflammatory cytokine TNF-α. Overall, this work provides ideas of using those robust and multifunctional therapeutic NPs toward enhanced sepsis therapy efficiency.
Collapse
Affiliation(s)
- Huan Cao
- Department of Nuclear Medicine & Laboratory of Clinical Nuclear Medicine, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jianhua Zhang
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610041, China
| | - Lei Yang
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610041, China
| | - Haotian Li
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610041, China
| | - Rong Tian
- Department of Nuclear Medicine & Laboratory of Clinical Nuclear Medicine, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Haoxing Wu
- Department of Radiology and Huaxi MR Research Center, Functional and Molecular Imaging Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yiwen Li
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610041, China
| | - Zhipeng Gu
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610041, China
| |
Collapse
|
10
|
Shen Y, Gwak H, Han B. Advanced manufacturing of nanoparticle formulations of drugs and biologics using microfluidics. Analyst 2024; 149:614-637. [PMID: 38083968 PMCID: PMC10842755 DOI: 10.1039/d3an01739g] [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] [Indexed: 01/04/2024]
Abstract
Numerous innovative nanoparticle formulations of drugs and biologics, named nano-formulations, have been developed in the last two decades. However, methods for their scaled-up production are still lagging, as the amount needed for large animal tests and clinical trials is typically orders of magnitude larger. This manufacturing challenge poses a critical barrier to successfully translating various nano-formulations. This review focuses on how microfluidics technology has become a powerful tool to overcome this challenge by synthesizing various nano-formulations with improved particle properties and product purity in large quantities. This microfluidic-based manufacturing is enabled by microfluidic mixing, which is capable of the precise and continuous control of the synthesis of nano-formulations. We further discuss the specific applications of hydrodynamic flow focusing, a staggered herringbone micromixer, a T-junction mixer, a micro-droplet generator, and a glass capillary on various types of nano-formulations of polymeric, lipid, inorganic, and nanocrystals. Various separation and purification microfluidic methods to enhance the product purity are reviewed, including acoustofluidics, hydrodynamics, and dielectrophoresis. We further discuss the challenges of microfluidics being used by broader research and industrial communities. We also provide future outlooks of its enormous potential as a decentralized approach for manufacturing nano-formulations.
Collapse
Affiliation(s)
- Yingnan Shen
- School of Mechanical Engineering, Purdue University, West Lafayette, IN 47907, USA.
| | - Hogyeong Gwak
- School of Mechanical Engineering, Purdue University, West Lafayette, IN 47907, USA.
| | - Bumsoo Han
- School of Mechanical Engineering, Purdue University, West Lafayette, IN 47907, USA.
- Purdue University Institute for Cancer Research, West Lafayette, IN, 47907, USA
| |
Collapse
|
11
|
Cui Q, Li J, Li Y, Tang L, Li K, Li T, Chen X, Zhang Z, Zhang GJ. Sensitive and rapid detection of bacterial endotoxin with a functional carbon nanotube field-effect transistor biosensor. Talanta 2024; 266:125035. [PMID: 37573682 DOI: 10.1016/j.talanta.2023.125035] [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: 03/10/2023] [Revised: 07/31/2023] [Accepted: 08/03/2023] [Indexed: 08/15/2023]
Abstract
Endotoxin in the blood can cause unexplained fever, and even death due to endotoxemia and bacteremia. Rapid and sensitive detection of endotoxin has become a priority event to intervene in the occurrence of dangerous diseases in time. In this context, a carbon nanotubes-based field-effect transistor (CNTs FET) nanosensor is developed to realize the rapid, label-free and sensitive detection of endotoxin. The CNTs FET was fabricated by assembling the polymer-sorted high-purity semiconductor CNT films onto the sensing channel. In order to improve the detection sensitivity, carboxylated graphene quantum dots (cGQDs) were coupled to the CNT surface via the poly-l-lysine (PLL). After that, polymyxin B (PMB), which is highly specific for endotoxin, was covalently combined with cGQDs, thus enabling the capture and detection of endotoxin. The method not only displayed an extremely low level of limitation of detection in PBS (4.6 fg/mL) and serum (30.3 fg/mL), respectively, with superior resistance to interference, but also enabled the analysis of Gram-negative bacterial infections in blood samples in a short duration of time. Meanwhile, the receiver operating characteristic curve (ROC) proved the superior diagnostic accuracy of this method (AUC = 0.990). Considering its excellent performance, the constructed CNTs FET biosensor is a promising tool to provide early warning of disease for clinical Gram-negative bacteremia and endotoxemia.
Collapse
Affiliation(s)
- Qing Cui
- School of Laboratory Medicine, Hubei University of Chinese Medicine, 16 Huangjia Lake West Road, Wuhan, 430065, PR China
| | - Jiahao Li
- School of Laboratory Medicine, Hubei University of Chinese Medicine, 16 Huangjia Lake West Road, Wuhan, 430065, PR China
| | - Yutao Li
- School of Laboratory Medicine, Hubei University of Chinese Medicine, 16 Huangjia Lake West Road, Wuhan, 430065, PR China
| | - Lina Tang
- School of Laboratory Medicine, Hubei University of Chinese Medicine, 16 Huangjia Lake West Road, Wuhan, 430065, PR China
| | - Kun Li
- The Central Hospital of Wuhan, Wuhan, 430061, China
| | - Tingxian Li
- Key Laboratory for the Physics and Chemistry of Nanodevices and Center for Carbon-based Electronics, School of Electronics, Peking University, Beijing, 100871, PR China.
| | - Xiaohua Chen
- Department of Laboratory Medicine, General Hospital of Central Theater Command, Wuhan, 430070, China.
| | - Zhiyong Zhang
- Key Laboratory for the Physics and Chemistry of Nanodevices and Center for Carbon-based Electronics, School of Electronics, Peking University, Beijing, 100871, PR China.
| | - Guo-Jun Zhang
- School of Laboratory Medicine, Hubei University of Chinese Medicine, 16 Huangjia Lake West Road, Wuhan, 430065, PR China.
| |
Collapse
|
12
|
Li M, Guo Q, Zhong C, Zhang Z. Multifunctional cell membranes-based nano-carriers for targeted therapies: a review of recent trends and future perspective. Drug Deliv 2023; 30:2288797. [PMID: 38069500 PMCID: PMC10987056 DOI: 10.1080/10717544.2023.2288797] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 11/05/2023] [Indexed: 12/18/2023] Open
Abstract
Nanotechnology has ignited a transformative revolution in disease detection, prevention, management, and treatment. Central to this paradigm shift is the innovative realm of cell membrane-based nanocarriers, a burgeoning class of biomimetic nanoparticles (NPs) that redefine the boundaries of biomedical applications. These remarkable nanocarriers, designed through a top-down approach, harness the intrinsic properties of cell-derived materials as their fundamental building blocks. Through shrouding themselves in natural cell membranes, these nanocarriers extend their circulation longevity and empower themselves to intricately navigate and modulate the multifaceted microenvironments associated with various diseases. This comprehensive review provides a panoramic view of recent breakthroughs in biomimetic nanomaterials, emphasizing their diverse applications in cancer treatment, cardiovascular therapy, viral infections, COVID-19 management, and autoimmune diseases. In this exposition, we deliver a concise yet illuminating overview of the distinctive properties underpinning biomimetic nanomaterials, elucidating their pivotal role in biomedical innovation. We subsequently delve into the exceptional advantages these nanomaterials offer, shedding light on the unique attributes that position them at the forefront of cutting-edge research. Moreover, we briefly explore the intricate synthesis processes employed in creating these biomimetic nanocarriers, shedding light on the methodologies that drive their development.
Collapse
Affiliation(s)
- Mo Li
- Department of Endocrinology, the Second Hospital of Jilin University, Changchun, China
| | - Qiushi Guo
- Pharmacy Department, First Hospital of Jilin University—the Eastern Division, Changchun, China
| | - Chongli Zhong
- Department of Endocrinology, the Second Hospital of Jilin University, Changchun, China
| | - Ziyan Zhang
- Department of Orthopedics, the Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
13
|
He W, Fu D, Gai Y, Liu X, Yang C, Ye Z, Chen X, Liu J, Chang B. An infection-microenvironment-targeted and responsive peptide-drug nanosystem for sepsis emergency by suppressing infection and inflammation. Asian J Pharm Sci 2023; 18:100869. [PMID: 38161786 PMCID: PMC10755722 DOI: 10.1016/j.ajps.2023.100869] [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: 07/25/2023] [Revised: 10/13/2023] [Accepted: 11/19/2023] [Indexed: 01/03/2024] Open
Abstract
Sepsis is a life-threatening emergency that causes millions of deaths every year due to severe infection and inflammation. Nevertheless, current therapeutic regimens are inadequate to promptly address the vast diversity of potential pathogens. Omiganan, an antimicrobial peptide, has shown promise for neutralizing endotoxins and eliminating diverse pathogens. However, its clinical application is hindered by safety and stability concerns. Herein, we present a nanoscale drug delivery system (Omi-hyd-Dex@HA NPs) that selectively targets infectious microenvironments (IMEs) and responds to specific stimuli for efficient intervention in sepsis. The system consists of omiganan-dexamethasone conjugates linked by hydrazone bonds which self-assemble into nanoparticles coated with a hyaluronic acid (HA). The HA coating not only facilitates IMEs-targeting through interaction with intercellular-adhesion-molecule-1 on inflamed endotheliocytes, but also improves the biosafety of the nanosystem and enhances drug accumulation in primary infection sites triggered by hyaluronidase. The nanoparticles release dual drugs in IMEs through pH-sensitive cleavage of hydrazone bonds to eradicate pathogens and suppress inflammation. In multiple tissue infection and sepsis animal models, Omi-hyd-Dex@HA NPs exhibited rapid source control and comprehensive inflammation reduction, thereby preventing subsequent fatal complications and significantly improving survival outcomes. The bio-responsive and self-delivering nanosystem offers a promising strategy for systemic sepsis treatment in emergencies.
Collapse
Affiliation(s)
- Wei He
- The Second Clinical College, The Second Affiliated Hospital, Guizhou University of Traditional Chinese Medicine, Guiyang 550003, China
| | - Daan Fu
- Department of Anesthesiology, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yongkang Gai
- Department of Nuclear Medicine, Hubei Province Key Laboratory of Molecular Imaging, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Xingxin Liu
- Department of Laboratory Medicine, West China Second University Hospital, Sichuan University, Chengdu 610065, China
| | - Chang Yang
- Guizhou Provincial Key Laboratory of Pharmaceutics, Guizhou Medical University, Guiyang 550004, China
| | - Zhilan Ye
- Department of Geriatrics, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Xu Chen
- The Second Clinical College, The Second Affiliated Hospital, Guizhou University of Traditional Chinese Medicine, Guiyang 550003, China
| | - Jia Liu
- Research Center for Tissue Engineering and Regenerative Medicine, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Bingcheng Chang
- The Second Clinical College, The Second Affiliated Hospital, Guizhou University of Traditional Chinese Medicine, Guiyang 550003, China
| |
Collapse
|
14
|
Chen L, Zhao R, Shen J, Liu N, Zheng Z, Miao Y, Zhu J, Zhang L, Wang Y, Fang H, Zhou J, Li M, Yang Y, Liu Z, Chen Q. Antibacterial Fusobacterium nucleatum-Mimicking Nanomedicine to Selectively Eliminate Tumor-Colonized Bacteria and Enhance Immunotherapy Against Colorectal Cancer. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2306281. [PMID: 37722134 DOI: 10.1002/adma.202306281] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 08/28/2023] [Indexed: 09/20/2023]
Abstract
Clinical evidence indicates that tumor-colonizing bacteria can be closely related to the tumor development and therapeutic responses. Selectively eliminating bacteria within tumors may be an attractive approach to enhance cancer treatment without additional side effects. Herein, it is found that, owing to the high affinity between the membrane protein Fap-2 on Fusobacterium nucleatum and d-galactose-β (1-3)-N-acetyl-d-galactosamine (Gal-GalNAc) overexpressed on colorectal tumor cells, F. nucleatum can colonize in colorectal tumors, as evidenced by both clinical samples and animal tumor models. Notably, F. nucleatum colonized in colorectal tumors can lead to an immunosuppressive tumor microenvironment, greatly reducing their responses to immune checkpoint blockade (ICB) therapy. Inspired by this finding, an F. nucleatum-mimetic nanomedicine is designed by fusing F. nucleatum cytoplasmic membrane (FM) with Colistin-loaded liposomes to achieve selective killing of tumor-colonizing F. nucleatum without affecting gut microbes. As a result, the therapeutic responses of F. nucleatum-colonized tumors to ICB therapies can be successfully restored, as demonstrated in an F. nucleatum-infected subcutaneous CT-26 tumor model, chemically induced spontaneous colorectal cancer models, and MC-38 tumor model. In summary, this work presents an F. nucleatum-mimicking nanomedicine that can selectively eliminate tumor-colonized bacteria, which is promising for enhancing the responses of cancer immunotherapy against F. nucleatum-colonized colorectal cancer.
Collapse
Affiliation(s)
- Linfu Chen
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, 215123, P.R. China
| | - Rui Zhao
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, 215123, P.R. China
| | - Jingjing Shen
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, 215123, P.R. China
| | - Nanhui Liu
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, 215123, P.R. China
| | - Zixuan Zheng
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, P.R. China
| | - Yu Miao
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, 215123, P.R. China
| | - Jiafei Zhu
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, 215123, P.R. China
| | - Lin Zhang
- Department of Gynecologic Oncology, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200030, P.R. China
| | - Yingyao Wang
- Department of Gynecology, Kunshan Maternity and Children's Health Care Hospital, Suzhou, Jiangsu, 215300, P.R. China
| | - Huapan Fang
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, 215123, P.R. China
| | - Jun Zhou
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, 215123, P.R. China
| | - Maoyi Li
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, 215123, P.R. China
| | - Yang Yang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, P.R. China
| | - Zhuang Liu
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, 215123, P.R. China
| | - Qian Chen
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, 215123, P.R. China
| |
Collapse
|
15
|
Li Y, Piao YZ, Chen H, Shi K, Dai J, Wang S, Zhou T, Le AT, Wang Y, Wu F, Ma R, Shi L, Liu Y. Dynamic covalent nano-networks comprising antibiotics and polyphenols orchestrate bacterial drug resistance reversal and inflammation alleviation. Bioact Mater 2023; 27:288-302. [PMID: 37113688 PMCID: PMC10126917 DOI: 10.1016/j.bioactmat.2023.04.014] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 03/29/2023] [Accepted: 04/14/2023] [Indexed: 04/29/2023] Open
Abstract
New antimicrobial strategies are urgently needed to meet the challenges posed by the emergence of drug-resistant bacteria and bacterial biofilms. This work reports the facile synthesis of antimicrobial dynamic covalent nano-networks (aDCNs) composing antibiotics bearing multiple primary amines, polyphenols, and a cross-linker acylphenylboronic acid. Mechanistically, the iminoboronate bond drives the formation of aDCNs, facilitates their stability, and renders them highly responsive to stimuli, such as low pH and high H2O2 levels. Besides, the representative A1B1C1 networks, composed of polymyxin B1(A1), 2-formylphenylboronic acid (B1), and quercetin (C1), inhibit biofilm formation of drug-resistant Escherichia coli, eliminate the mature biofilms, alleviate macrophage inflammation, and minimize the side effects of free polymyxins. Excellent bacterial eradication and inflammation amelioration efficiency of A1B1C1 networks are also observed in a peritoneal infection model. The facile synthesis, excellent antimicrobial performance, and biocompatibility of these aDCNs potentiate them as a much-needed alternative in current antimicrobial pipelines.
Collapse
Affiliation(s)
- Yuanfeng Li
- Translational Medicine Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Yin-Zi Piao
- Wenzhou Institute, University of Chinese Academy of Sciences, Oujiang Laboratory, Zhejiang Lab for Regenerative Medicine, Vision and Brain Health, Wenzhou, Zhejiang, 325001, China
| | - Hua Chen
- Wenzhou Institute, University of Chinese Academy of Sciences, Oujiang Laboratory, Zhejiang Lab for Regenerative Medicine, Vision and Brain Health, Wenzhou, Zhejiang, 325001, China
- Key Laboratory of Functional Polymer Materials of Ministry of Education, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin, 300071, China
| | - Keqing Shi
- Translational Medicine Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Juqin Dai
- Translational Medicine Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Siran Wang
- Wenzhou Institute, University of Chinese Academy of Sciences, Oujiang Laboratory, Zhejiang Lab for Regenerative Medicine, Vision and Brain Health, Wenzhou, Zhejiang, 325001, China
| | - Tieli Zhou
- Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
- Corresponding author.
| | - Anh-Tuan Le
- Nano Institute, Phenikaa University, Yen Nghia, Ha Dong, Ha Noi, Viet Nam
| | - Yaran Wang
- Wenzhou Institute, University of Chinese Academy of Sciences, Oujiang Laboratory, Zhejiang Lab for Regenerative Medicine, Vision and Brain Health, Wenzhou, Zhejiang, 325001, China
| | - Fan Wu
- Key Laboratory of Functional Polymer Materials of Ministry of Education, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin, 300071, China
| | - Rujiang Ma
- Key Laboratory of Functional Polymer Materials of Ministry of Education, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin, 300071, China
- Corresponding author.
| | - Linqi Shi
- Key Laboratory of Functional Polymer Materials of Ministry of Education, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin, 300071, China
| | - Yong Liu
- Translational Medicine Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
- Wenzhou Institute, University of Chinese Academy of Sciences, Oujiang Laboratory, Zhejiang Lab for Regenerative Medicine, Vision and Brain Health, Wenzhou, Zhejiang, 325001, China
- Corresponding author. Translational Medicine Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China.
| |
Collapse
|
16
|
Shi Z, Zhang X, Yang X, Zhang X, Ma F, Gan H, Chen J, Wang D, Sun W, Wang J, Wang C, Lyu L, Yang K, Deng L, Qing G. Specific Clearance of Lipopolysaccharide from Blood Based on Peptide Bottlebrush Polymer for Sepsis Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2302560. [PMID: 37247257 DOI: 10.1002/adma.202302560] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 05/16/2023] [Indexed: 05/31/2023]
Abstract
Lipopolysaccharide (LPS) is the primary bacterial toxin that is vital to the pathogenesis and progression of sepsis associated with extremely high morbidity and mortality worldwide. However, specific clearance of LPS from circulating blood is highly challenging because of the structural complexity and its variation between/within bacterial species. Herein, a robust strategy based on phage display screening and hemocompatible peptide bottlebrush polymer design for specific clearance of targeted LPS from circulating blood is proposed. Using LPS extracted from Escherichia coli as an example, a novel peptide (HWKAVNWLKPWT) with high affinity (KD < 1.0 nм), specificity, and neutralization activity (95.9 ± 0.1%) against the targeted LPS is discovered via iterative affinity selection coupled with endotoxin detoxification screening. A hemocompatible bottlebrush polymer bearing the short peptide [poly(PEGMEA-co-PEP-1)] exhibits high LPS selectivity to reduce circulating LPS level from 2.63 ± 0.01 to 0.78 ± 0.05 EU mL-1 in sepsis rabbits via extracorporeal hemoperfusion (LPS clearance ratio > 70%), reversing the LPS-induced leukocytopenia and multiple organ damages significantly. This work provides a universal paradigm for developing a highly selective hemoadsorbent library fully covering the LPS family, which is promising to create a new era of precision medicine in sepsis therapy.
Collapse
Affiliation(s)
- Zhenqiang Shi
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, P.R. China
| | - Xiancheng Zhang
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, P.R. China
| | - Xijing Yang
- Animal Experiment Center, West China Hospital, Sichuan University, Chengdu, 610041, P.R. China
| | - Xiaoyu Zhang
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, P.R. China
| | - Fei Ma
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, 100850, P.R. China
| | - Hui Gan
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, 100850, P.R. China
| | - Junjun Chen
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, P.R. China
| | - Dongdong Wang
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, P.R. China
| | - Wenjing Sun
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, P.R. China
| | - Jingxia Wang
- Radiation Chemistry Department, Sichuan Institute of Atomic Energy, Chengdu, 610101, P.R. China
| | - Cunli Wang
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, P.R. China
| | - Liting Lyu
- Dalian Key Laboratory of Energy Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, P.R. China
| | - Kaiguang Yang
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, P.R. China
| | - Lijing Deng
- Pediatric Intensive Care Unit, Department of Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, 610041, P.R. China
| | - Guangyan Qing
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, P.R. China
| |
Collapse
|
17
|
Vasconcelos I, Santos T. Nanotechnology Applications in Sepsis: Essential Knowledge for Clinicians. Pharmaceutics 2023; 15:1682. [PMID: 37376129 DOI: 10.3390/pharmaceutics15061682] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 05/29/2023] [Accepted: 06/06/2023] [Indexed: 06/29/2023] Open
Abstract
Sepsis is a life-threatening condition caused by a dysregulated host response to an invading pathogen such as multidrug-resistant bacteria. Despite recent advancements, sepsis is a leading cause of morbidity and mortality, resulting in a significant global impact and burden. This condition affects all age groups, with clinical outcomes mainly depending on a timely diagnosis and appropriate early therapeutic intervention. Because of the unique features of nanosized systems, there is a growing interest in developing and designing novel solutions. Nanoscale-engineered materials allow a targeted and controlled release of bioactive agents, resulting in improved efficacy with minimal side effects. Additionally, nanoparticle-based sensors provide a quicker and more reliable alternative to conventional diagnostic methods for identifying infection and organ dysfunction. Despite recent advancements, fundamental nanotechnology principles are often presented in technical formats that presuppose advanced chemistry, physics, and engineering knowledge. Consequently, clinicians may not grasp the underlying science, hindering interdisciplinary collaborations and successful translation from bench to bedside. In this review, we abridge some of the most recent and most promising nanotechnology-based solutions for sepsis diagnosis and management using an intelligible format to stimulate a seamless collaboration between engineers, scientists, and clinicians.
Collapse
Affiliation(s)
- Inês Vasconcelos
- School of Medicine, University of Minho, 4710-057 Braga, Portugal
- Department of Surgery and Physiology, Cardiovascular Research and Development Center-UnIC, Faculty of Medicine, University of Porto, Al. Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
| | - Tiago Santos
- School of Medicine, University of Minho, 4710-057 Braga, Portugal
| |
Collapse
|
18
|
Shen Y, Yuk SA, Kwon S, Tamam H, Yeo Y, Han B. A timescale-guided microfluidic synthesis of tannic acid-Fe III network nanocapsules of hydrophobic drugs. J Control Release 2023; 357:484-497. [PMID: 37068522 PMCID: PMC10225907 DOI: 10.1016/j.jconrel.2023.04.024] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 04/07/2023] [Accepted: 04/13/2023] [Indexed: 04/19/2023]
Abstract
Many drugs are poorly water-soluble and suffer from low bioavailability. Metal-phenolic network (MPN), a hydrophilic thin layer such as tannic acid (TA)-FeIII network, has been recently used to encapsulate hydrophobic drugs to improve their bioavailability. However, it remains challenging to synthesize nanocapsules of a wide variety of hydrophobic drugs and to scale up the production in a continuous manner. Here, we present a microfluidic synthesis method to continuously produce TA-FeIII network nanocapsules of hydrophobic drugs. We hypothesize that nanocapsules can continuously be formed only when the microfluidic mixing timescale is shorter than the drug's nucleation timescale. The hypothesis was tested on three hydrophobic drugs - paclitaxel, curcumin, and vitamin D with varying solubility and nucleation timescale. The proposed mechanism was validated by successfully predicting the synthesis outcomes. The microfluidically-synthesized nanocapsules had well-controlled sizes of 100-200 nm, high drug loadings of 40-70%, and a throughput of up to 70 mg hr-1 per channel. The release kinetics, cellular uptake, and cytotoxicity were further evaluated. The effect of coating constituents on nanocapsule properties were characterized. Fe content of nanocapsules was reported. The stability of nanocapsules at different temperatures and pHs were also tested. The results suggest that the present method can provide a quantitative guideline to predictively design a continuous synthesis scheme for hydrophobic drug encapsulation via MPN nanocapsules with scaled-up capability.
Collapse
Affiliation(s)
- Yingnan Shen
- School of Mechanical Engineering, Purdue University, West Lafayette, IN 47907, USA
| | - Simseok A Yuk
- Department of Industrial and Physical Pharmacy, Purdue University, West Lafayette, IN 47907, USA
| | - Soonbum Kwon
- Department of Industrial and Physical Pharmacy, Purdue University, West Lafayette, IN 47907, USA
| | - Hassan Tamam
- Department of Industrial and Physical Pharmacy, Purdue University, West Lafayette, IN 47907, USA; Department of industrial pharmacy, Faculty of Pharmacy, Assiut University, Assiut 71526, Egypt
| | - Yoon Yeo
- Department of Industrial and Physical Pharmacy, Purdue University, West Lafayette, IN 47907, USA; Purdue University Institute for Cancer Research, West Lafayette, IN 47907, USA
| | - Bumsoo Han
- School of Mechanical Engineering, Purdue University, West Lafayette, IN 47907, USA; Purdue University Institute for Cancer Research, West Lafayette, IN 47907, USA.
| |
Collapse
|
19
|
Dokla EME, Abutaleb NS, Milik SN, Kandil EAEA, Qassem OM, Elgammal Y, Nasr M, McPhillie MJ, Abouzid KAM, Seleem MN, Imming P, Adel M. SAR investigation and optimization of benzimidazole-based derivatives as antimicrobial agents against Gram-negative bacteria. Eur J Med Chem 2023; 247:115040. [PMID: 36584632 DOI: 10.1016/j.ejmech.2022.115040] [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: 09/09/2022] [Revised: 12/03/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022]
Abstract
Antibiotic-resistant bacteria represent a serious threat to modern medicine and human life. Only a minority of antibacterial agents are active against Gram-negative bacteria. Hence, the development of novel antimicrobial agents will always be a vital need. In an effort to discover new therapeutics against Gram-negative bacteria, we previously reported a structure-activity-relationship (SAR) study on 1,2-disubstituted benzimidazole derivatives. Compound III showed a potent activity against tolC-mutant Escherichia coli with an MIC value of 2 μg/mL, representing a promising lead for further optimization. Building upon this study, herein, 49 novel benzimidazole compounds were synthesized to investigate their antibacterial activity against Gram-negative bacteria. Our design focused on three main goals, to address the low permeability of our compounds and improve their cellular accumulation, to expand the SAR study to the unexplored ring C, and to optimize the lead compound (III) by modification of the methanesulfonamide moiety. Compounds (25a-d, 25f-h, 25k, 25l, 25p, 25r, 25s, and 26b) exhibited potent activity against tolC-mutant E. coli with MIC values ranging from 0.125 to 4 μg/mL, with compound 25d displaying the highest potency among the tested compounds with an MIC value of 0.125 μg/mL. As its predecessor, III, compound 25d exhibited an excellent safety profile without any significant cytotoxicity to mammalian cells. Time-kill kinetics assay indicated that 25d exhibited a bacteriostatic activity and significantly reduced E. coli JW55031 burden as compared to DMSO. Additionally, combination of 25d with colistin partially restored its antibacterial activity against Gram-negative bacterial strains (MIC values ranging from 4 to 16 μg/mL against E. coli BW25113, K. pneumoniae, A. baumannii, and P. aeruginosa). Furthermore, formulation of III and 25d as lipidic nanoparticles (nanocapsules) resulted in moderate enhancement of their antibacterial activity against Gram-negative bacterial strains (A. Baumannii, N. gonorrhoeae) and compound 25d demonstrated superior activity to the lead compound III. These findings establish compound 25d as a promising candidate for treatment of Gram-negative bacterial infections and emphasize the potential of nano-formulations in overcoming poor cellular accumulation in Gram-negative bacteria where further optimization and investigation are warranted to improve the potency and broaden the spectrum of our compounds.
Collapse
Affiliation(s)
- Eman M E Dokla
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Ain Shams University, Abbassia, Cairo, 11566, Egypt; Institute für Pharmazie, Martin-Luther-Universität Halle-Wittenberg, Wolfgang-Langenbeck-Str. 4, Halle (Saale), 06120, Germany.
| | - Nader S Abutaleb
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA, 24061, USA; Department of Microbiology and Immunology, Zagazig University, Zagazig, 44519, Egypt
| | - Sandra N Milik
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Ain Shams University, Abbassia, Cairo, 11566, Egypt; School of Chemistry, University of Leeds, Leeds, LS2 9JT, United Kingdom
| | - Ezzat A E A Kandil
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Ain Shams University, Abbassia, Cairo, 11566, Egypt
| | - Omar M Qassem
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Ain Shams University, Abbassia, Cairo, 11566, Egypt; Purdue University Institute of Drug Discovery, Purdue University, West Lafayette, IN, 47907, USA
| | - Yehia Elgammal
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA, 24061, USA
| | - Maha Nasr
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Ain Shams University, Abbassia, Cairo, 11566, Egypt
| | - Martin J McPhillie
- School of Chemistry, University of Leeds, Leeds, LS2 9JT, United Kingdom
| | - Khaled A M Abouzid
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Ain Shams University, Abbassia, Cairo, 11566, Egypt
| | - Mohamed N Seleem
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA, 24061, USA; Center for Emerging, Zoonotic and Arthropod-borne Pathogens, Virginia Polytechnic Institute and State University, Blacksburg, VA, 24061, USA
| | - Peter Imming
- Institute für Pharmazie, Martin-Luther-Universität Halle-Wittenberg, Wolfgang-Langenbeck-Str. 4, Halle (Saale), 06120, Germany
| | - Mai Adel
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Ain Shams University, Abbassia, Cairo, 11566, Egypt.
| |
Collapse
|
20
|
Saha S, Mishra A. Protein-directed synthesis of ZIF-8 functionalized with a polymer as core-shell drug coatings with antibacterial and anti-inflammatory properties. Biomater Sci 2023; 11:481-488. [PMID: 36193822 DOI: 10.1039/d2bm01295b] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
We developed a strategy to use lysozyme (Lys) as a template to produce mesoporous zeolitic imidazolate framework (ZIF-8) structures under physiological conditions. Thereafter, an amphiphilic triblock copolymer, PEG-PPG-PEG, was used to form protective core-shell ZIF-8 nanocomposite coatings to protect the encapsulated drug epigallocatechin-3-gallate (EGCG), to achieve notable antibacterial properties against E. coli, S. aureus and MRSA strains. Moreover, nanocomposites exhibited anti-inflammatory activity by counteracting the secretion of cytokines in THP-1 macrophages.
Collapse
Affiliation(s)
- Sarmistha Saha
- Materials Engineering, Indian Institute of Technology Gandhinagar, Palaj, Gandhinagar, India.
| | - Abhijit Mishra
- Materials Engineering, Indian Institute of Technology Gandhinagar, Palaj, Gandhinagar, India.
| |
Collapse
|
21
|
Xu J, Gao C, He Y, Fang X, Sun D, Peng Z, Xiao H, Sun M, Zhang P, Zhou T, Yang X, Yu Y, Li R, Zou X, Shu H, Qiu Y, Zhou X, Yuan S, Yao S, Shang Y. NLRC3 expression in macrophage impairs glycolysis and host immune defense by modulating the NF-κB-NFAT5 complex during septic immunosuppression. Mol Ther 2023; 31:154-173. [PMID: 36068919 PMCID: PMC9840117 DOI: 10.1016/j.ymthe.2022.08.023] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 06/23/2022] [Accepted: 08/30/2022] [Indexed: 01/28/2023] Open
Abstract
Impairment of innate immune cell function and metabolism underlies immunosuppression in sepsis; however, a promising therapy to orchestrate this impairment is currently lacking. In this study, high levels of NOD-like receptor family CARD domain containing-3 (NLRC3) correlated with the glycolytic defects of monocytes/macrophages from septic patients and mice that developed immunosuppression. Myeloid-specific NLRC3 deletion improved macrophage glycolysis and sepsis-induced immunosuppression. Mechanistically, NLRC3 inhibits nuclear factor (NF)-κB p65 binding to nuclear factor of activated T cells 5 (NFAT5), which further controls the expression of glycolytic genes and proinflammatory cytokines of immunosuppressive macrophages. This is achieved by decreasing NF-κB activation-co-induced by TNF-receptor-associated factor 6 (TRAF6) or mammalian target of rapamycin (mTOR)-and decreasing transcriptional co-activator p300 activity by inducing NLRC3 sequestration of mTOR and p300. Genetic inhibition of NLRC3 disrupted the NLRC3-mTOR-p300 complex and enhanced NF-κB binding to the NFAT5 promoter in concert with p300. Furthermore, intrapulmonary delivery of recombinant adeno-associated virus harboring a macrophage-specific NLRC3 deletion vector significantly improved the defense of septic mice that developed immunosuppression upon secondary intratracheal bacterial challenge. Collectively, these findings indicate that NLRC3 mediates critical aspects of innate immunity that contribute to an immunocompromised state during sepsis and identify potential therapeutic targets.
Collapse
Affiliation(s)
- Jiqian Xu
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Chenggang Gao
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yajun He
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Xiangzhi Fang
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Deyi Sun
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Zhekang Peng
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Hairong Xiao
- Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Miaomiao Sun
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Pei Zhang
- Department of Paediatrics, Jinling Hospital, School of Medicine, Nanjing University, Nanjing 210016, China
| | - Ting Zhou
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Xiaobo Yang
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yuan Yu
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Ruiting Li
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Xiaojing Zou
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Huaqing Shu
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yang Qiu
- State Key Laboratory of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan Institute of Virology, Wuhan 43007, China
| | - Xi Zhou
- State Key Laboratory of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan Institute of Virology, Wuhan 43007, China
| | - Shiying Yuan
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Shanglong Yao
- Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - You Shang
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| |
Collapse
|
22
|
Zhang P, Ouyang Q, Zhai T, Sun J, Wu J, Qin F, Zhang N, Yue S, Yang X, Zhang H, Hou Y, Deng L, Wang F, Zhan Q, Yu Q, Qin M, Gan Z. An inflammation-targeted nanoparticle with bacteria forced release of polymyxin B for pneumonia therapy. NANOSCALE 2022; 14:15291-15304. [PMID: 36039653 DOI: 10.1039/d2nr02026b] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
The epidemic of multidrug-resistant Gram-negative bacteria is an ever-growing global concern. Polymyxin B (PMB), a kind of "old fashioned" antibiotic, has been revived in clinical practice and mainly used as last-line antibiotics for otherwise untreatable serious infections because the incidence of the resistance to PMB is currently relatively low in comparison with other antibiotics in vivo owing to the unique bactericidal mechanism of PMB. However, serious adverse side effects, including nephrotoxicity and neurotoxicity, hamper its clinical application. Herein, we describe the development of a nanoparticle that can target sites of inflammation and forcedly release PMB specifically in the area of Gram-negative bacteria. This particle was constructed through the electrostatic self-assembly of hyaluronic acid (HA) and PMB molecules in order to realize the safe and effective treatment of pneumonia. After systemic administration, PMB-HA nanoparticles were found to actively accumulate in the lungs, precisely target the CD44 receptors over-expressed on the membrane of activated endothelial cells in inflammatory sites, and then come into contact with the bacteria resident in the damaged alveolar-capillary membrane. Due to the electrostatic and hydrophobic interactions between PMB and the lipopolysaccharide (LPS) in the outer membranes of bacteria, the PMB molecules in the PMB-HA nanoparticles are expected to escape from the nanoparticles to insert into the bacteria via competitive binding with LPS. Through shielding the cationic nature of PMB, PMB-HA nanoparticles also possess outstanding biosafety performance in comparison to free PMB. It is thus believed that this smart delivery system may pave a new way for the resurrection of PMB in the future clinical treatment of bacterial inflammatory diseases.
Collapse
Affiliation(s)
- Peisen Zhang
- Beijing Advanced Innovation Centre for Soft Matter Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, PR China.
| | - Qiuhong Ouyang
- Beijing Advanced Innovation Centre for Soft Matter Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, PR China.
| | - Tianshu Zhai
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing 100029, PR China.
| | - Jing Sun
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Chaoyang District, Beijing 100029, PR China
| | - Jun Wu
- Beijing Advanced Innovation Centre for Soft Matter Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, PR China.
| | - Feng Qin
- National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, Department of Psychiatry, West China Hospital of Sichuan University, Chengdu, Sichuan, 610041, PR China
| | - Ni Zhang
- National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, Department of Psychiatry, West China Hospital of Sichuan University, Chengdu, Sichuan, 610041, PR China
| | - Saisai Yue
- Beijing Advanced Innovation Centre for Soft Matter Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, PR China.
| | - Xinchen Yang
- Beijing Advanced Innovation Centre for Soft Matter Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, PR China.
| | - Hanyi Zhang
- Beijing Advanced Innovation Centre for Soft Matter Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, PR China.
| | - Yi Hou
- Beijing Advanced Innovation Centre for Soft Matter Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, PR China.
| | - Li Deng
- Beijing Advanced Innovation Centre for Soft Matter Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, PR China.
| | - Fang Wang
- Beijing Advanced Innovation Centre for Soft Matter Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, PR China.
| | - Qingyuan Zhan
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing 100029, PR China.
| | - Qingsong Yu
- Beijing Advanced Innovation Centre for Soft Matter Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, PR China.
| | - Meng Qin
- Beijing Advanced Innovation Centre for Soft Matter Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, PR China.
| | - Zhihua Gan
- Beijing Advanced Innovation Centre for Soft Matter Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, PR China.
| |
Collapse
|
23
|
Ma J, Jiang L, Liu G. Cell membrane-coated nanoparticles for the treatment of bacterial infection. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2022; 14:e1825. [PMID: 35725897 DOI: 10.1002/wnan.1825] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 04/20/2022] [Accepted: 04/22/2022] [Indexed: 06/15/2023]
Abstract
Despite the enormous success of antibiotics in antimicrobial therapy, the rapid emergence of antibiotic resistance and the complexity of the bacterial infection microenvironment make traditional antibiotic therapy face critical challenges against resistant bacteria, antitoxin, and intracellular infections. Consequently, there is a critical need to design antimicrobial agents that target infection microenvironment and alleviate antibiotic resistance. Cell membrane-coated nanoparticles (CMCNPs) are biomimetic materials that can be obtained by wrapping the cell membrane vesicles directly onto the surface of the nanoparticles (NPs) through physical means. Incorporating the biological functions of cell membrane vesicles and the superior physicochemical properties of NPs, CMCNPs have shown great promise in recent years for targeting infections, neutralizing bacterial toxins, and designing bacterial infection vaccines. This review highlights topics where CMCNPs present great value in advancing the treatment of bacterial infections, including drug delivery, detoxification, and vaccination. Lastly, we discuss the future hurdles and prospects of translating this technique into clinical practice, providing a comprehensive review of the technological developments of CMCNPs in the treatment of bacterial infections. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Infectious Disease Therapeutic Approaches and Drug Discovery > Emerging Technologies Nanotechnology Approaches to Biology > Nanoscale Systems in Biology.
Collapse
Affiliation(s)
- Jiaxin Ma
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, China
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, China
| | - Lai Jiang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, China
| | - Gang Liu
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, China
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, China
| |
Collapse
|
24
|
Cao H, Yang L, Tian R, Wu H, Gu Z, Li Y. Versatile polyphenolic platforms in regulating cell biology. Chem Soc Rev 2022; 51:4175-4198. [PMID: 35535743 DOI: 10.1039/d1cs01165k] [Citation(s) in RCA: 63] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Polyphenolic materials are a class of fascinating and versatile bioinspired materials for biointerfacial engineering. In particular, due to the presence of active chemical groups, a series of unique physicochemical properties become accessible and tunable of the as-prepared polyphenolic platforms, which could delicately regulate the cell activities via cell-material contact-dependent interactions. More interestingly, polyphenols could also affect the cell behaviors via cell-material contact-independent manner, which arise due to their intrinsically functional characteristics (e.g., antioxidant and photothermal behaviors). As such, a comprehensive understanding on the relationship between material properties and desired biomedical applications, as well as the underlying mechanism at the cellular and molecular level would provide material design principles and accelerate the lab-to-clinic translation of polyphenolic platforms. In this review, we firstly give a brief overview of cell hallmarks governed by surrounding cues, followed by the introduction of polyphenolic material engineering strategies. Subsequently, a detailed discussion on cell-polyphenols contact-dependent interfacial interaction and contact-independent interaction was also carefully provided. Lastly, their biomedical applications were elaborated. We believe that this review could provide guidances for the rational material design of multifunctional polyphenols and extend their application window.
Collapse
Affiliation(s)
- Huan Cao
- Laboratory of Clinical Nuclear Medicine, Department of Nuclear Medicine, National Clinical Research Center for Geriatrics, West China Hospital, College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, 610041, China.
| | - Lei Yang
- Laboratory of Clinical Nuclear Medicine, Department of Nuclear Medicine, National Clinical Research Center for Geriatrics, West China Hospital, College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, 610041, China.
| | - Rong Tian
- Laboratory of Clinical Nuclear Medicine, Department of Nuclear Medicine, National Clinical Research Center for Geriatrics, West China Hospital, College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, 610041, China.
| | - Haoxing Wu
- Huaxi MR Research Center, Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Zhipeng Gu
- Laboratory of Clinical Nuclear Medicine, Department of Nuclear Medicine, National Clinical Research Center for Geriatrics, West China Hospital, College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, 610041, China.
| | - Yiwen Li
- Laboratory of Clinical Nuclear Medicine, Department of Nuclear Medicine, National Clinical Research Center for Geriatrics, West China Hospital, College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
25
|
Ledger EVK, Sabnis A, Edwards AM. Polymyxin and lipopeptide antibiotics: membrane-targeting drugs of last resort. MICROBIOLOGY (READING, ENGLAND) 2022; 168:001136. [PMID: 35118938 PMCID: PMC8941995 DOI: 10.1099/mic.0.001136] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 01/06/2022] [Indexed: 12/15/2022]
Abstract
The polymyxin and lipopeptide classes of antibiotics are membrane-targeting drugs of last resort used to treat infections caused by multi-drug-resistant pathogens. Despite similar structures, these two antibiotic classes have distinct modes of action and clinical uses. The polymyxins target lipopolysaccharide in the membranes of most Gram-negative species and are often used to treat infections caused by carbapenem-resistant species such as Escherichia coli, Acinetobacter baumannii and Pseudomonas aeruginosa. By contrast, the lipopeptide daptomycin requires membrane phosphatidylglycerol for activity and is only used to treat infections caused by drug-resistant Gram-positive bacteria such as methicillin-resistant Staphylococcus aureus and vancomycin-resistant enterococci. However, despite having distinct targets, both antibiotic classes cause membrane disruption, are potently bactericidal in vitro and share similarities in resistance mechanisms. Furthermore, there are concerns about the efficacy of these antibiotics, and there is increasing interest in using both polymyxins and daptomycin in combination therapies to improve patient outcomes. In this review article, we will explore what is known about these distinct but structurally similar classes of antibiotics, discuss recent advances in the field and highlight remaining gaps in our knowledge.
Collapse
Affiliation(s)
- Elizabeth V. K. Ledger
- MRC Centre for Molecular Bacteriology and Infection, Imperial College London, Armstrong Rd, London, SW7 2AZ, UK
| | - Akshay Sabnis
- MRC Centre for Molecular Bacteriology and Infection, Imperial College London, Armstrong Rd, London, SW7 2AZ, UK
| | - Andrew M. Edwards
- MRC Centre for Molecular Bacteriology and Infection, Imperial College London, Armstrong Rd, London, SW7 2AZ, UK
| |
Collapse
|
26
|
Brannon ER, Guevara MV, Pacifici NJ, Lee JK, Lewis JS, Eniola-Adefeso O. Polymeric particle-based therapies for acute inflammatory diseases. NATURE REVIEWS. MATERIALS 2022; 7:796-813. [PMID: 35874960 PMCID: PMC9295115 DOI: 10.1038/s41578-022-00458-5] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 06/09/2022] [Indexed: 05/02/2023]
Abstract
Acute inflammation is essential for initiating and coordinating the body's response to injuries and infections. However, in acute inflammatory diseases, inflammation is not resolved but propagates further, which can ultimately lead to tissue damage such as in sepsis, acute respiratory distress syndrome and deep vein thrombosis. Currently, clinical protocols are limited to systemic steroidal treatments, fluids and antibiotics that focus on eradicating inflammation rather than modulating it. Strategies based on stem cell therapeutics and selective blocking of inflammatory molecules, despite showing great promise, still lack the scalability and specificity required to treat acute inflammation. By contrast, polymeric particle systems benefit from uniform manufacturing at large scales while preserving biocompatibility and versatility, thus providing an ideal platform for immune modulation. Here, we outline design aspects of polymeric particles including material, size, shape, deformability and surface modifications, providing a strategy for optimizing the targeting of acute inflammation.
Collapse
Affiliation(s)
- Emma R. Brannon
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI USA
| | | | - Noah J. Pacifici
- Department of Biomedical Engineering, University of California, Davis, CA USA
| | - Jonathan K. Lee
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI USA
| | - Jamal S. Lewis
- Department of Biomedical Engineering, University of California, Davis, CA USA
| | | |
Collapse
|
27
|
Li Z, Chen J, Tian H, Chen X. Sepsis Treatment Strategies Based on Nanomaterials ※. ACTA CHIMICA SINICA 2022. [DOI: 10.6023/a21120615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
28
|
Wang M. Nanocapsule system tackles polymyxin B toxicity. Nat Rev Nephrol 2021; 17:707. [PMID: 34545227 DOI: 10.1038/s41581-021-00493-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|