1
|
Liu Y, Song W, Dong W, Gong X, Dong C, Zhao J, Wang R, Song S, Shuang S. Preparation of mitochondrial targeted near-infrared ratio fluorescent probe and its dual response detection for viscosity and ONOO - and cell imaging. Talanta 2025; 292:127909. [PMID: 40081248 DOI: 10.1016/j.talanta.2025.127909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Revised: 03/05/2025] [Accepted: 03/06/2025] [Indexed: 03/15/2025]
Abstract
The changes in viscosity and the concentration of ONOO- in mitochondria can effectively reflect the physiological and pathological status of cells. Therefore, the development of effective fluorescent probes for the sensing of viscosity and the concentration of ONOO- in mitochondria has great significance. In this article, a mitochondrial targeted fluorescent probe named Mito-RP was synthesized for the dual responsive sensing of viscosity and ONOO- by introducing pyridine ring and phenylboronic acid ester structure into 4-dimethylamino-cinnamaldehyde with long conjugated chain structure as the parent material. Mito-RP exhibits 600 folds fluorescence enhancement of viscosity in the red-light channel at 700 nm, with pyridine cation as the mitochondrial anchoring group. Simultaneously, Mito-RP appears excellent selectivity towards ONOO- using boronic acid esters as response sites. A new ratio fluorescence analysis method was constructed based on the linear correlation between the emission intensity ratio of Mito-RP at 616 nm/700 nm and the concentration of ONOO-. The linear range is 0.05-33 μM and the detection limit is 9.2 nM. Meanwhile, Mito-RP successfully monitored the changes in viscosity during lipopolysaccharide induced inflammation and rapamycin induced mitochondrial autophagy in HeLa cells. In addition, Mito-RP has also achieved visual imaging of intracellular exogenous/endogenous ONOO-. These studies provide a novel method for in-depth investigation of mitochondrial function and its role in diseases.
Collapse
Affiliation(s)
- Yang Liu
- School of Chemistry and Chemical Engineering, and Institute of Environmental Science, Shanxi University, Taiyuan, 030006, China
| | - Wenqiang Song
- School of Chemistry and Chemical Engineering, and Institute of Environmental Science, Shanxi University, Taiyuan, 030006, China
| | - Wenjuan Dong
- School of Chemistry and Chemical Engineering, and Institute of Environmental Science, Shanxi University, Taiyuan, 030006, China
| | - Xiaojuan Gong
- School of Chemistry and Chemical Engineering, and Institute of Environmental Science, Shanxi University, Taiyuan, 030006, China
| | - Chuan Dong
- School of Chemistry and Chemical Engineering, and Institute of Environmental Science, Shanxi University, Taiyuan, 030006, China
| | - Jie Zhao
- Shanxi Provincial Key Laboratory of Classical Prescription Strengthening Yang, Shanxi Provincial Integrated TCM and WM Hospital, Taiyuan, 030013, China
| | - Ruibing Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, 999078, Macau
| | - Shengmei Song
- School of Chemistry and Chemical Engineering, and Institute of Environmental Science, Shanxi University, Taiyuan, 030006, China.
| | - Shaomin Shuang
- School of Chemistry and Chemical Engineering, and Institute of Environmental Science, Shanxi University, Taiyuan, 030006, China.
| |
Collapse
|
2
|
Song Q, Meng Q, Meng X, Wang X, Zhang Y, Zhao T, Cong J. Size- and duration-dependent toxicity of heavy vehicle tire wear particles in zebrafish. JOURNAL OF HAZARDOUS MATERIALS 2025; 493:138299. [PMID: 40253784 DOI: 10.1016/j.jhazmat.2025.138299] [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: 02/03/2025] [Revised: 04/09/2025] [Accepted: 04/14/2025] [Indexed: 04/22/2025]
Abstract
Tire wear particles (TWPs), as a pervasive environmental pollutant, pose significant risks to aquatic ecosystems. This study investigates the effects of small (HS) and large (HL) TWPs produced by heavy vehicles on zebrafish, focusing on physiological, microbial, and transcriptomic levels, as well as their intergenerational consequences, under short-term (15 days) and long-term (90 days) exposure. Short-term exposure to small particles (HS15) significantly reduced body width and triggered widespread oxidative stress, while long-term exposure to large particles (HL90) increased gut weight and decreased gill weight, reflecting respiratory and digestive disruptions. Tissue-level analyses revealed that smaller particles accumulated more readily in internal organs, whereas larger particles caused localized physiological stress. Gut microbiota profiling indicated a marked decline in microbial diversity, compositional shifts, and network simplification, with HL15 enriched in Acinetobacter and xenobiotic metabolism pathways, and HS15 exhibiting Proteobacteria-dominated dysbiosis and enrichment of LPS biosynthesis genes. Liver transcriptomics revealed group-specific responses: HL15 exposure activated innate immunity via the NOD-MAPK axis, while HS15 induced atypical PI3K-NF-κB signaling, potentially linked to microbial LPS. Notably, all TWP-exposed groups showed enrichment of the herpes simplex virus 1 (HSV-1) infection pathway, suggesting a conserved antiviral-like host response. Transgenerational effects were evidenced by impaired growth and significant downregulation of GH/IGF signaling and upregulation of apoptotic genes in offspring, despite only subtle transcriptomic changes in long-term exposed parents. These findings underscore the importance of particle size, exposure duration, and microbiota-gut-liver axis interactions in mediating TWP toxicity and highlight potential transgenerational risks associated with environmental microplastic exposure.
Collapse
Affiliation(s)
- Qianqian Song
- College of Biological Engineering, Qingdao University of Science and Technology, Qingdao 266000, China
| | - Qingxuan Meng
- College of Biological Engineering, Qingdao University of Science and Technology, Qingdao 266000, China
| | - Xinrui Meng
- College of Biological Engineering, Qingdao University of Science and Technology, Qingdao 266000, China
| | - Xiaolong Wang
- College of Biological Engineering, Qingdao University of Science and Technology, Qingdao 266000, China
| | - Yun Zhang
- College of Biological Engineering, Qingdao University of Science and Technology, Qingdao 266000, China
| | - Tianyu Zhao
- College of Biological Engineering, Qingdao University of Science and Technology, Qingdao 266000, China
| | - Jing Cong
- College of Biological Engineering, Qingdao University of Science and Technology, Qingdao 266000, China.
| |
Collapse
|
3
|
Liu F, Wu M, Shao D, Zhou X, Liu Q, Sheng X, Li D, Dai M. Exposure to DDAB disinfectants promotes antimicrobial resistance to antibiotics and collateral-sensitivity to polymyxins in Salmonella enterica. Microb Pathog 2025; 203:107428. [PMID: 40021029 DOI: 10.1016/j.micpath.2025.107428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 02/08/2025] [Accepted: 02/25/2025] [Indexed: 03/03/2025]
Abstract
SALMONELLA: as an important food-borne zoonotic pathogen, is found in soil and processing environment by human or animal feces, causing serious public health problems. Quaternary ammonium compounds (QACs) disinfectants are widely used in hospitals, livestock farms and food processing sites because of their low toxicity and broad-spectrum disinfection. However, sub-lethal levels of QACs disinfectants can induce bacteria to develop tolerance to disinfectants and cross-resistance to other antimicrobial agents. The acquired resistance will undoubtedly pose a threat to the prevention of antimicrobial resistance. In this study, Salmonella enterica SE211 was induced by the sub-inhibitory concentration and sub-lethal concentration of dodecyl dimethyl ammonium bromide (DDAB) in vitro. Following exposure to DDAB, the strains showed increased resistance to DDAB, doxycycline, amphenicols and fluoroquinolones, and increased sensitivity to colistin drugs. Phenotypic experiments showed that the induced strains exhibited changes in efflux pump activity, biofilm formation ability, motility and membrane characterization. Next-generation sequencing revealed mutations in induced strains involved in LPS-related genes (msbA, lptDE) and cationic antimicrobial peptide (CAMP) resistance-related genes (phoQ, pmrD). Transcriptome sequencing (RNA-seq) analysis revealed up-regulation of efflux pump genes and down-regulation of CAMP resistance, LPS and peptidoglycan related genes. Our study provided a theoretical basis for the potential consequences of disinfection failures and environmental residues of QACs disinfectants on the evolution of antibiotic resistance in salmonella. Furthermore, the induction of colistin sensitivity in salmonella by DDBA resulted in the emergence of collateral sensitivity, which offered a new strategy for drug combination applications to prevent the rise of colistin-resistant superbugs.
Collapse
Affiliation(s)
- Fangjia Liu
- The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, 430070, China; MOA Key Laboratory of Food Safety Evaluation/National Reference Laboratory of Veterinary Drug Residue (HZAU), Huazhong Agricultural University, Wuhan, 430070, China
| | - Menghui Wu
- The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, 430070, China; MOA Key Laboratory of Food Safety Evaluation/National Reference Laboratory of Veterinary Drug Residue (HZAU), Huazhong Agricultural University, Wuhan, 430070, China
| | - Dan Shao
- The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, 430070, China; MOA Key Laboratory of Food Safety Evaluation/National Reference Laboratory of Veterinary Drug Residue (HZAU), Huazhong Agricultural University, Wuhan, 430070, China
| | - Xueya Zhou
- The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, 430070, China; MOA Key Laboratory of Food Safety Evaluation/National Reference Laboratory of Veterinary Drug Residue (HZAU), Huazhong Agricultural University, Wuhan, 430070, China
| | - Quan Liu
- The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, 430070, China; MOA Key Laboratory of Food Safety Evaluation/National Reference Laboratory of Veterinary Drug Residue (HZAU), Huazhong Agricultural University, Wuhan, 430070, China
| | - Xijing Sheng
- The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, 430070, China; MOA Key Laboratory of Food Safety Evaluation/National Reference Laboratory of Veterinary Drug Residue (HZAU), Huazhong Agricultural University, Wuhan, 430070, China
| | - Donghua Li
- The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, 430070, China; MOA Key Laboratory of Food Safety Evaluation/National Reference Laboratory of Veterinary Drug Residue (HZAU), Huazhong Agricultural University, Wuhan, 430070, China
| | - Menghong Dai
- The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, 430070, China; MOA Key Laboratory of Food Safety Evaluation/National Reference Laboratory of Veterinary Drug Residue (HZAU), Huazhong Agricultural University, Wuhan, 430070, China.
| |
Collapse
|
4
|
Li Y, Pan L, Zhao Q, Xiong H, Fang X, Guo X, Wang Y, Su X, Liu P, Hao J, Wang L, Wang X, Dong Q, Li Z, Jin F. Tanshinone IIA + Osthole alleviates ferroptosis in LPS-induced acute lung injury by Keap1-Nrf2/HO-1 pathway. Microb Pathog 2025; 203:107347. [PMID: 40147558 DOI: 10.1016/j.micpath.2025.107347] [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: 09/08/2024] [Revised: 01/17/2025] [Accepted: 01/27/2025] [Indexed: 03/29/2025]
Abstract
BACKGROUND Acute lung injury (ALI) is associated with a high mortality rate and requires effective treatment. Tanshinone IIA (T) and Osthole (O) exhibit anti-inflammatory effects and have been used to protect against lipopolysaccharide (LPS)-induced lung injury in mice. However, the combined effects of T and O on lung injury protection and their potential protective mechanisms have not been studied. OBJECTIVE To assess the protective effects of TO on LPS-induced ALI in mice and BEAS-2B cell injury and to investigate the potential mechanisms underlying these protective effects. METHODS Models of ALI induced by LPS were established. The assessment encompassed the viability of BEAS-2B cells, cell count, myeloperoxidase (MPO) activity, protein content, as well as IL-6 and TNF-a levels in bronchoalveolar lavage fluid (BALF). Additionally, malondialdehyde (MDA), reactive oxygen species (ROS), and glutathione (GSH) levels in mouse lung tissue were measured. The effects of TO were assessed using immunofluorescence (IF), immunohistochemistry (IHC), Western Blot (WB), RT-PCR, and ELISA. Statistical analysis involved one-way ANOVA and t-test. RESULTS TO administration led to a significant reduction in lung edema (W/D), MDA, ROS, GSH, and superoxide dismutase (SOD) levels compared to the individual T or O groups, alleviating LPS-induced ALI. TO also significantly attenuated lung tissue damage, reduced inflammatory response, decreased Fe2+ and 4-HNE levels, and increased GPX4, SLC7A11, and Nrf2 gene expression in mice. Ultimately, TO alleviated ferroptosis in LPS-induced ALI by activating Nrf2 expression, and no markedly adverse reactions were observed. CONCLUSION TO alleviates LPS-induced ALI and effectively treats against LPS-induced ALI.
Collapse
Affiliation(s)
- Yujuan Li
- Department of Pulmonary and Critical Care Medicine, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi, 710038, China
| | - Lei Pan
- Department of Pulmonary and Critical Care Medicine, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi, 710038, China
| | - Qi Zhao
- Department of Pulmonary and Critical Care Medicine, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi, 710038, China
| | - Huanqing Xiong
- Department of Pulmonary and Critical Care Medicine, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi, 710038, China
| | - Xiaoan Fang
- Department of Pulmonary and Critical Care Medicine, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi, 710038, China
| | - Xian Guo
- Department of Pulmonary and Critical Care Medicine, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi, 710038, China
| | - Ying Wang
- Department of Pulmonary and Critical Care Medicine, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi, 710038, China
| | - Xiaorong Su
- Department of Endocrinology, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi, 710038, China
| | - Pan Liu
- Department of Pulmonary and Critical Care Medicine, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi, 710038, China
| | - Jing Hao
- Department of Pulmonary and Critical Care Medicine, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi, 710038, China
| | - Lei Wang
- Department of Pulmonary and Critical Care Medicine, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi, 710038, China
| | - Xinqun Wang
- Department of Pulmonary and Critical Care Medicine, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi, 710038, China
| | - Quandi Dong
- General Surgery Department, 947 Army Hospital, China.
| | - Zhichao Li
- Department of Pulmonary and Critical Care Medicine, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi, 710038, China.
| | - Faguang Jin
- Department of Pulmonary and Critical Care Medicine, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi, 710038, China.
| |
Collapse
|
5
|
Zhou K, Peng K, Lin J, Wang J, Jiang Q, Ai M, Xu L, Ouyang P, Huang X, Chen D, Geng Y. ADP-heptose-LPS heptosyltransferase I (WaaC) as a key molecule maintains morphological structure and metabolic activity, simultaneously influences pathogenicity in Vibrio mimicus. Int J Biol Macromol 2025; 313:144203. [PMID: 40373916 DOI: 10.1016/j.ijbiomac.2025.144203] [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: 03/04/2025] [Revised: 05/06/2025] [Accepted: 05/12/2025] [Indexed: 05/17/2025]
Abstract
ADP-heptose-LPS heptosyltransferase I (encoded by waaC gene) is a crucial enzyme in the lipopolysaccharide (LPS) synthesis, maintaining the stability of LPS and cell walls in numerous Gram-negative bacteria. Vibrio mimicus is an epidemic pathogen that threatens aquatic animals and human health, resulting in high morbidity and mortality after infecting the fish. Currently, the role of waaC gene in V. mimicus is still unclear. In this study, waaC gene deletion and complementation strains of V. mimicus were constructed. Our results show that ΔwaaC exhibited a rough phenotype on LB agar, with elevated exocrine protein and a disordered cell wall observed by the electron microscope. Transcriptomic analysis revealed that following the deletion of waaC gene, the expression of 224 genes was drastically upregulated, while the expression of 229 genes was significantly downregulated. These genes involve various pathways, including material transport, metabolism, and environmental adaptation. The tricarboxylic acid cycle (TCA cycle) and pyruvate metabolism are the primary pathways affected by waaC gene. Our phenotypic analysis is consistent with transcriptomic findings, indicating that the decreased pathogenicity of ΔwaaC is related to the effect of waaC gene on these genes, which negatively impacts V. mimicus growth, motility, adhesion, and biofilm formation while enhancing its self-aggregation. The virulence of the ΔwaaC was 103-fold lower than that of the wild strain. Moreover, the expression levels of inflammatory cytokine and pro-apoptotic genes in epithelioma papulosum cyprini (EPC) cells were dramatically upregulated during the ΔwaaC infection. These results provide valuable insights into revealing the pathogenic mechanism of V. mimicus, further bringing more options for the candidate deletion targets of the V. mimicus attenuated vaccine.
Collapse
Affiliation(s)
- Keyu Zhou
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Kun Peng
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Jingyi Lin
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Jiao Wang
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Qibin Jiang
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Mingqi Ai
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Le Xu
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Ping Ouyang
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Xiaoli Huang
- Department of Aquaculture, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Defang Chen
- Department of Aquaculture, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Yi Geng
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China.
| |
Collapse
|
6
|
Jiang J, Pan Z, Su Y, Dai L, Xu N, Wu H, Chen X. Carbon dots from purple sweet potato as a promising anti-inflammatory biomaterial for alleviating the LPS-induced inflammation in macrophages. J Nanobiotechnology 2025; 23:397. [PMID: 40448145 DOI: 10.1186/s12951-025-03494-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Accepted: 05/26/2025] [Indexed: 06/02/2025] Open
Abstract
This study synthesizes carbon dots derived from crude extracts of purple sweet potato (CPP-CDs) and evaluates its anti-inflammatory effects in a lipopolysaccharide (LPS) -induced acute inflammation model. Characterization revealed that CPP-CDs possess a uniform spherical structure and excellent photoluminescent properties. In vitro, CPP-CDs significantly inhibited the expression of pro-inflammatory cytokines (IL-1β, IL-6, TNF-α), reduced the accumulation of reactive oxygen species (ROS), suppressed pyroptosis, and facilitated the polarization of macrophages from the M1 phenotype to the M2 phenotype. In vivo, CPP-CDs significantly improved the survival rates of LPS-treated mice, mitigated tissue damage, and suppressed the levels of pro-inflammatory cytokines. Mechanistic studies indicated that CPP-CDs exert anti-inflammatory effects through the inhibition of the TLR4/NF-κB signaling pathway and the modulation of the NLRP3 inflammasome. Additionally, CPP-CDs exhibited excellent biocompatibility, with no significant toxicity observed in mice. This study provides strong evidence supporting the application of CPP-CDs as a novel anti-inflammatory material, highlighting their potential for acute inflammation treatment and expanding the possibilities for the development of carbon-dot-based anti-inflammatory therapies.
Collapse
Affiliation(s)
- Jiebang Jiang
- Department of General Surgery, Affiliated Hospital of Nantong University, No.20 Xisi Road, Nantong, Jiangsu, 226001, China
| | - Zhiyuan Pan
- School of Life Sciences, Jiangsu Normal University, Xuzhou, Jiangsu, 221116, China
| | - Yiren Su
- School of Life Sciences, Jiangsu Normal University, Xuzhou, Jiangsu, 221116, China
| | - Lu Dai
- Department of General Surgery, Nantong Women's Prison Hospital, Nantong, Jiangsu, 226001, China
| | - Nana Xu
- Laboratory of Morphology, Xuzhou Medical University, No.209 Tongshan Road, Xuzhou, Jiangsu, 221004, China.
| | - Han Wu
- Department of General Surgery, Affiliated Hospital of Nantong University, No.20 Xisi Road, Nantong, Jiangsu, 226001, China.
| | - Xin Chen
- Department of General Surgery, Affiliated Hospital of Nantong University, No.20 Xisi Road, Nantong, Jiangsu, 226001, China.
- Department of Clinical and Translational Research Center, Affiliated Hospital of Nantong University, Nantong, Jiangsu, 226001, China.
- Department of Clinical Biobank & Institute of Oncology, Affiliated Hospital of Nantong University, Nantong, Jiangsu, 226001, China.
| |
Collapse
|
7
|
Portakal T, Havlíček V, Herůdková J, Pelková V, Gruntová T, Çakmakci RC, Kotasová H, Hampl A, Vaňhara P. Lipopolysaccharide induces retention of E-cadherin in the endoplasmic reticulum and promotes hybrid epithelial-to-mesenchymal transition of human embryonic stem cells-derived expandable lung epithelial cells. Inflamm Res 2025; 74:82. [PMID: 40413286 PMCID: PMC12103375 DOI: 10.1007/s00011-025-02041-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Revised: 04/09/2025] [Accepted: 04/15/2025] [Indexed: 05/27/2025] Open
Abstract
BACKGROUND Lipopolysaccharide (LPS)-induced inflammation of lung tissues triggers irreversible alterations in the lung parenchyma, leading to fibrosis and pulmonary dysfunction. While the molecular and cellular responses of immune and connective tissue cells in the lungs are well characterized, the specific epithelial response remains unclear due to the lack of representative cell models. Recently, we introduced human embryonic stem cell-derived expandable lung epithelial (ELEP) cells as a novel model for studying lung injury and regeneration. METHODS ELEPs were derived from the CCTL 14 human embryonic stem cell line through activin A-mediated endoderm specification, followed by further induction toward pulmonary epithelium using FGF2 and EGF. ELEPs exhibit a high proliferation rate and express key structural and molecular markers of alveolar progenitors, such as NKX2-1. The effects of Escherichia coli LPS serotype O55:B5 on the phenotype and molecular signaling of ELEPs were analyzed using viability and migration assays, mRNA and protein levels were determined by qRT-PCR, western blotting, and immunofluorescent microscopy. RESULTS We demonstrated that purified LPS induces features of a hybrid epithelial-to-mesenchymal transition in pluripotent stem cell-derived ELEPs, triggers the unfolded protein response, and upregulates intracellular β-catenin level through retention of E-cadherin within the endoplasmic reticulum. CONCLUSIONS Human embryonic stem cell-derived ELEPs provide a biologically relevant, non-cancerous lung cell model to investigate molecular responses to inflammatory stimuli and address epithelial plasticity. This approach offers novel insights into the fine molecular processes underlying lung injury and repair.
Collapse
Affiliation(s)
- Türkan Portakal
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Kamenice 753/5, 625 00, Brno, Czech Republic
| | - Vítězslav Havlíček
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Kamenice 753/5, 625 00, Brno, Czech Republic
| | - Jarmila Herůdková
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Kamenice 753/5, 625 00, Brno, Czech Republic
- University Hospital Brno, Jihlavská 340/20, 625 00, Brno, Czech Republic
| | - Vendula Pelková
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Kamenice 753/5, 625 00, Brno, Czech Republic
- University Hospital Brno, Jihlavská 340/20, 625 00, Brno, Czech Republic
| | - Tereza Gruntová
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Kamenice 753/5, 625 00, Brno, Czech Republic
| | - Rıza Can Çakmakci
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Kamenice 753/5, 625 00, Brno, Czech Republic
| | - Hana Kotasová
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Kamenice 753/5, 625 00, Brno, Czech Republic
| | - Aleš Hampl
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Kamenice 753/5, 625 00, Brno, Czech Republic
- International Clinical Research Center, St. Anne's University Hospital, Pekařská 664/53, 602 00, Brno, Czech Republic
- University Hospital Brno, Jihlavská 340/20, 625 00, Brno, Czech Republic
| | - Petr Vaňhara
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Kamenice 753/5, 625 00, Brno, Czech Republic.
- International Clinical Research Center, St. Anne's University Hospital, Pekařská 664/53, 602 00, Brno, Czech Republic.
- University Hospital Brno, Jihlavská 340/20, 625 00, Brno, Czech Republic.
| |
Collapse
|
8
|
Gundavarapu B, Nallamotu KC, Murapaka VV, Venkataraman B, Saisree L, Reddy M. Identification of SanA as a novel regulator of peptidoglycan biogenesis in Escherichia coli. PLoS Genet 2025; 21:e1011712. [PMID: 40402998 DOI: 10.1371/journal.pgen.1011712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Accepted: 05/06/2025] [Indexed: 05/24/2025] Open
Abstract
Gram-negative bacterial cell envelope consists of a surface-exposed lipid bilayer (outer membrane or OM) that serves as a permeability barrier to maintain the cellular integrity. Beneath the OM is the periplasmic space that harbours peptidoglycan (PG), a highly cross-linked mesh-like glycan polymer closely encasing the inner membrane (IM). During growth of a bacterium balanced synthesis of the envelope components is required to maintain the cellular integrity, of which little is known. In this study, we identify sanA, an ORF of unknown function encoding a predicted IM-anchored protein as a factor contributing to balanced synthesis of PG in E. coli. Absence of SanA increased the rate of nascent PG strand incorporation, and restored growth and viability to several mutants defective in either cell division or cell elongation. Detailed mutant analysis of sanA showed that it is defective in the envelope barrier properties. Interestingly, overexpression of the periplasmic endopeptidases that cleave the cross-links of the PG mesh was able to alleviate the phenotypes of sanA mutant implying the envelope defects are due to alterations in the PG sacculus. Additionally, a SanA variant (SSDsbA-SanA) targeted to the periplasm, complemented the SanA- phenotypes suggesting it functions in the periplasmic phase of the PG synthesis. Further, we find that SanA functions independently of its paralog, ElyC, known to regulate the synthesis of enterobacterial common antigen (ECA), a surface polysaccharide found in the cell envelopes of most enteric bacteria. Overall, our results suggest a role for SanA in the maintenance of optimal PG synthesis, providing evidence for the existence of an additional layer of regulation in Gram-negative cell envelope biogenesis.
Collapse
Affiliation(s)
- Bhargavi Gundavarapu
- CSIR-Centre for Cellular and Molecular Biology, Hyderabad, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Krishna Chaitanya Nallamotu
- CSIR-Centre for Cellular and Molecular Biology, Hyderabad, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Vishnu Vachana Murapaka
- CSIR-Centre for Cellular and Molecular Biology, Hyderabad, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | | | | | - Manjula Reddy
- CSIR-Centre for Cellular and Molecular Biology, Hyderabad, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| |
Collapse
|
9
|
Krymchenko R, Pfirrmann M, van der Leeuw S, Avila-Martinez N, Versteeg EMM, Meuwese RTC, Vlig M, Verdoes M, Boekema BKHL, van Kuppevelt TH, Daamen WF. Preparation, fractionation, and characterization of solubilized elastin and comparison of cellular response on fibroblasts and macrophages. Int J Biol Macromol 2025; 315:144548. [PMID: 40409629 DOI: 10.1016/j.ijbiomac.2025.144548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2025] [Revised: 05/09/2025] [Accepted: 05/20/2025] [Indexed: 05/25/2025]
Abstract
Elastin and elastin-derived compounds are promising biomaterials due to their biological activity, unique natural crosslinks, and ability to mimic native tissue properties. Solubilized elastin peptides retain the bioactivity of elastin and are more suitable for wound care applications than the insoluble form. Chemically solubilized elastins have shown advantageous effects in skin regeneration in humans. Here, five solubilized elastins were prepared via chemical (stepwise and continuously hydrolyzed with oxalic acid - OxA-st-ELN and OxA-ELN, or with potassium hydroxide - KOH-ELN), enzymatic (Enz-ELN), or combined (Combi-ELN) methods. OxA-st-ELN had the largest molecular weights (MWs) fragments, while Enz-ELN and Combi-ELN yielded the smallest. The effects of elastin preparations were evaluated on primary human cells - dermal fibroblasts and macrophages. In fibroblast assays, Enz-ELN induced elastin, collagen, and fibrillin-2 protein deposition, while other preparations exhibited levels comparable to the control. α-smooth muscle actin (SMA) expression remained low across all conditions. Continuous oxalic acid hydrolysis simplified the traditional stepwise approach while maintaining bioactivity. Macrophage studies showed chemical hydrolysates preserved the M0-like subtype, while Enz-ELN promoted a pro-inflammatory M1-like phenotype, and Combi-ELN had mixed effects. OxA-ELN and KOH-ELN appeared to be the most promising options for developing biomaterial dermal scaffolds that support tissue regeneration in vivo.
Collapse
Affiliation(s)
- Roman Krymchenko
- Radboud university medical center, Research Institute for Medical Innovation, Department of Medical BioSciences, Nijmegen, the Netherlands
| | - Maren Pfirrmann
- Radboud university medical center, Research Institute for Medical Innovation, Department of Medical BioSciences, Nijmegen, the Netherlands
| | - Sjoerd van der Leeuw
- Radboud university medical center, Research Institute for Medical Innovation, Department of Medical BioSciences, Nijmegen, the Netherlands
| | - Nancy Avila-Martinez
- Radboud university medical center, Research Institute for Medical Innovation, Department of Medical BioSciences, Nijmegen, the Netherlands
| | - Elly M M Versteeg
- Radboud university medical center, Research Institute for Medical Innovation, Department of Medical BioSciences, Nijmegen, the Netherlands
| | - Rob T C Meuwese
- Radboud university medical center, Research Institute for Medical Innovation, Department of Medical BioSciences, Nijmegen, the Netherlands
| | - Marcel Vlig
- Alliance of Dutch Burn Care, Burn Research Lab, Beverwijk, the Netherlands
| | - Martijn Verdoes
- Radboud university medical center, Research Institute for Medical Innovation, Department of Medical BioSciences, Nijmegen, the Netherlands; Leiden University Medical Center, Department of Immunology, Leiden, the Netherlands
| | - Bouke K H L Boekema
- Alliance of Dutch Burn Care, Burn Research Lab, Beverwijk, the Netherlands; Amsterdam University Medical Center (AUMC), Amsterdam, the Netherlands; Tissue Function and Regeneration, Amsterdam Movement Sciences Research Institute, Amsterdam, the Netherlands; Department of Plastic, Reconstructive and Hand Surgery, AUMC, location VUmc, Amsterdam, the Netherlands
| | - Toin H van Kuppevelt
- Radboud university medical center, Research Institute for Medical Innovation, Department of Medical BioSciences, Nijmegen, the Netherlands
| | - Willeke F Daamen
- Radboud university medical center, Research Institute for Medical Innovation, Department of Medical BioSciences, Nijmegen, the Netherlands.
| |
Collapse
|
10
|
Yang N, Zhang H, Zhang Y, Lin B, Huang R, Cui T, Li X. Bacterial colonization contributes to pathological scar formation via the regulation of inflammatory response. J Transl Med 2025; 23:569. [PMID: 40400009 PMCID: PMC12096502 DOI: 10.1186/s12967-025-06585-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2025] [Accepted: 05/07/2025] [Indexed: 05/23/2025] Open
Abstract
BACKGROUND It has been established that inflammatory factors are involved in the formation of pathological scars. Therefore, pathological scars are regarded to be highly associated with chronic inflammation, whereas what factors contribute to this inflammation remains unclear. OBJECTIVE To confirm that bacterial colonization is involved in the formation of pathological scars, and to reveal that the persistent inflammatory response mediated by macrophages due to bacterial colonization promotes scar formation. METHODS This study included 23 normal skin controls and 58 untreated pathological scar samples. To detect the presence of bacteria in surgically-excised scar samples and alterations of histology, as well as bacteria-associated gene levels, histological staining, immunoelectron microscopy, microbiological and cell culture and molecular biology detection methods were employed. The PICRUSt2 tool and BugBase were employed to identify pathways, genes, and phenotypic differences. RESULTS We found that in pathological scars, bacteria were widely distributed both extracellularly and intracellularly, with intracellular bacteria primarily located in the cytoplasm of macrophages and fibroblasts. A total of 2,260 bacterial species were detected in pathological scars, primarily from the Clostridiales, Burkholderiales, Actinomycetales, and Bacteroidales orders. Moreover, the pathogenicity and motility of colonizing bacteria were positively correlated with the degree of scar hyperplasia and invasiveness. The lysates of four clinically-relevant bacterial species had differential effects on the secretion of inflammatory cytokines from macrophages. When treated macrophage supernatant was added to fibroblasts, collagen secretion was dysregulated, and fibroblast differentiation into myofibroblasts prominently increased. In rat scar model, the expression of inflammatory factors and growth factors in the scar tissue was increased, which activated the TGF-β/Smad signaling pathway, resulting in the increasing of α-SMA. CONCLUSIONS Persistent activation of macrophages by tissue-colonizing bacteria may be a key factor in promoting inflammatory response and dysregulated collagen deposition in pathological scars, offering a potential new strategy for preventing and treating pathological scars.
Collapse
Affiliation(s)
- Ning Yang
- Department of Burn and Plastics Surgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China
| | - Hao Zhang
- Department of Plastic and Burn Surgery, Joint Logistics Support Force of Chinese PLA, No. 927 Hospital, Puer, 665000, China
| | - Yuheng Zhang
- Department of Burn and Plastics Surgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China
- Department of Orthopedics, Western Theater Air Force Hospital of PLA, Chengdu, 610011, China
| | - Bin Lin
- Department of Burn and Plastics Surgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China
| | - Rong Huang
- Department of Burn and Plastics Surgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China
| | - Tingting Cui
- Department of Biochemistry and Molecular Biology, Preclinical Medicine College, Xinjiang Medical University, Urumqi, 830011, China.
| | - Xueyong Li
- Department of Burn and Plastics Surgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China.
| |
Collapse
|
11
|
Wang C, Hong Z, Song M, Zheng H, Zhou Q, Yang H, Li H, Huang D. Production of astaxanthin with high purity and activity based on engineering improvement strategies. J Biotechnol 2025; 405:139-149. [PMID: 40379138 DOI: 10.1016/j.jbiotec.2025.05.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Revised: 05/06/2025] [Accepted: 05/13/2025] [Indexed: 05/19/2025]
Abstract
Here, astaxanthin production in Escherichia coli was systematically improved step by step. By introducing the additional copy of CrtZ and fusion complex of CrtZ and CrtW, astaxanthin content in cells increased from 0.10 mg/g to 0.16 mg/g and 0.63 mg/g DCW, respectively. Remolding the astaxanthin gene cluster by replacing the PanCrtE by HpGGPPS3-1 and the fusion of CrtZ and CrtW increased astaxanthin content to 1.98 mg/g DCW. Further selecting the productive host and optimizing culture conditions dramatically increased astaxanthin content to 3.61 mg/g DCW. Subsequently, the fed-batch fermentation achieved the maximum yield of astaxanthin at 509.58 mg/L with the productivity of 7.72 mg/L/h and 5.91 mg/g DCW, covering 98.17 % of detected carotenoids. The chirality analysis assigned the same isomer of astaxanthin extracted from our fermentation system and Haematococcus pluvialis. Moreover, the radical and superoxide anion scavenging activity analysis revealed that astaxanthin achieved in this study performed better than natural astaxanthin extracted from H. pluvialis and chemical synthetic astaxanthin. This study provides a step-by-step example for bioengineering improvement of natural products in E. coli with high purity and activity.
Collapse
Affiliation(s)
- Chaogang Wang
- Guangdong Technology Research Center for Marine Algal Bioengineering, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen 518060, PR China; Shenzhen Engineering Laboratory for Marine Algal Biological Development and Application, Shenzhen University, Shenzhen 518060, PR China
| | - Zeyu Hong
- Guangdong Technology Research Center for Marine Algal Bioengineering, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen 518060, PR China
| | - Mingjian Song
- Guangdong Technology Research Center for Marine Algal Bioengineering, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen 518060, PR China
| | - Hao Zheng
- Guangdong Technology Research Center for Marine Algal Bioengineering, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen 518060, PR China
| | - Qiaomian Zhou
- Guangdong Technology Research Center for Marine Algal Bioengineering, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen 518060, PR China
| | - Haihong Yang
- Guangdong Technology Research Center for Marine Algal Bioengineering, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen 518060, PR China
| | - Hui Li
- Guangdong Technology Research Center for Marine Algal Bioengineering, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen 518060, PR China; Shenzhen Engineering Laboratory for Marine Algal Biological Development and Application, Shenzhen University, Shenzhen 518060, PR China
| | - Danqiong Huang
- Guangdong Technology Research Center for Marine Algal Bioengineering, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen 518060, PR China; Shenzhen Engineering Laboratory for Marine Algal Biological Development and Application, Shenzhen University, Shenzhen 518060, PR China.
| |
Collapse
|
12
|
Fernandez JA, Han Q, Rajczewski AT, Kono T, Weirath NA, Lee AS, Rahim A, Tretyakova NY. Multi-Omics Analysis of the Epigenetic Effects of Inflammation in Murine Type II Pneumocytes. Int J Mol Sci 2025; 26:4692. [PMID: 40429836 PMCID: PMC12112469 DOI: 10.3390/ijms26104692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2025] [Revised: 05/07/2025] [Accepted: 05/07/2025] [Indexed: 05/29/2025] Open
Abstract
Chronic inflammation plays a central role in the pathogenesis of lung diseases including asthma, long COVID, chronic obstructive pulmonary disease (COPD), and lung cancer. Lipopolysaccharide (LPS) is a potent inflammatory agent produced by Gram-negative bacteria and also found in cigarette smoke. Our earlier study revealed that the intranasal exposure of A/J mice to LPS for 7 days altered gene expression levels in alveolar Type II epithelial cells (AECIIs), which serve as precursors to lung adenocarcinoma and are also preferentially targeted by SARS-CoV-2. In the present work, we employed a comprehensive multi-omics approach to characterize changes in DNA methylation/hydroxymethylation, gene expression, and global protein abundances in the AECIIs of A/J mice following the sub-chronic exposure to LPS and after a 4-week recovery period. Exposure to LPS led to hypermethylation at regulatory elements within the genome such as enhancer regions and expression changes in genes known to play a role in lung cancer tumorigenesis. Changes in protein abundance were consistent with an inflammatory phenotype and also included tumor suppressor proteins. Integration of the multi-omics data resulted in a model where LPS-driven inflammation in AECIIs triggers epigenetic changes that, along with genetic mutations, may contribute to lung cancer development.
Collapse
Affiliation(s)
- Jenna A. Fernandez
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, MN 55455, USA; (J.A.F.); (N.A.W.); (A.R.)
| | - Qiyuan Han
- Department of Biochemistry, Biophysics, and Molecular Biology, University of Minnesota, Minneapolis, MN 55455, USA; (Q.H.); (A.T.R.)
| | - Andrew T. Rajczewski
- Department of Biochemistry, Biophysics, and Molecular Biology, University of Minnesota, Minneapolis, MN 55455, USA; (Q.H.); (A.T.R.)
| | - Thomas Kono
- Research Informatics Services, University of Minnesota, Minneapolis, MN 55455, USA
| | - Nicholas A. Weirath
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, MN 55455, USA; (J.A.F.); (N.A.W.); (A.R.)
| | - Alexander S. Lee
- Department of Chemistry, University of Minnesota, Minneapolis, MN 55455, USA;
| | - Abdur Rahim
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, MN 55455, USA; (J.A.F.); (N.A.W.); (A.R.)
| | - Natalia Y. Tretyakova
- Department of Medicinal Chemistry, College of Pharmacy, and the Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
| |
Collapse
|
13
|
Kuttoth H, Pathak T, Sandhyarani N. A Point-of-Care Aptasensor for the Real-Time Detection of Sepsis Biomarker. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2025. [PMID: 40359382 DOI: 10.1021/acs.langmuir.5c00784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/15/2025]
Abstract
The rising demand for noninvasive portable point-of-care (POC) sensors for on-site detection of biomarkers reflects a shift toward personalized healthcare and real-time diagnostics. Lipopolysaccharide (LPS) is a key sepsis biomarker, a bacteria-borne endotoxin that can induce fatal conditions, such as septic shock, if left undetected. Timely bedside detection of LPS is critical for the appropriate intervention and treatment of sepsis. In this work, we report a highly sensitive electrochemical sensor chip designed for the selective detection of LPS, which is compatible with a portable analyzer for on-site detection. The sensor is fabricated using functionalized CNT (fCNT) and copper(I) oxide nanoparticles (Cu2O). Functionalized with the LPS-specific aptamer, the sensor demonstrated remarkable selectivity and sensitivity toward LPS. It achieved a limit of detection (LOD) of 10 ag mL-1 and exhibited a linear detection range from 10 ag mL-1 to 10 ng mL-1. The device detected LPS across various samples, including food samples such as mayonnaise and fruit juices, as well as a clinical sample, whole blood. These results underscore its potential for practical application in food quality assurance and clinical diagnostics, offering real-time analysis capabilities.
Collapse
Affiliation(s)
- Haritha Kuttoth
- Nanoscience Research Laboratory, Department of Materials Science and Engineering, National Institute of Technology Calicut, Calicut 673601, Kerala, India
| | - Tushar Pathak
- Nanoscience Research Laboratory, Department of Materials Science and Engineering, National Institute of Technology Calicut, Calicut 673601, Kerala, India
| | - N Sandhyarani
- Nanoscience Research Laboratory, Department of Materials Science and Engineering, National Institute of Technology Calicut, Calicut 673601, Kerala, India
| |
Collapse
|
14
|
Lin ZT, Jana B, Korupolu S, Kong Y, Liu G, Dong Y, Li Y, Zhang Q, Shou W, Upadhyay P, Wang Z, Ran Z, Wu MX. Wearable Photonic Device for Multiple Biomarker Sampling and Detection without Blood Draws. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025:e2416240. [PMID: 40326959 DOI: 10.1002/adma.202416240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 03/25/2025] [Indexed: 05/07/2025]
Abstract
Needle-based blood draws or phlebotomy practice in clinics for centuries, often causing pain, discomfort, and inconvenience. Here, a wearable photonic device is presented by integrating a microlens array (MLA) and an optic microneedle array (OMNA) functionalized with immunobinding for safe and needle-free biomarker sampling and detection. The MLA-integrated OMNA amplifies and transmits LED light at 595 nm into skin through the OMNA, bypassing the light-absorbing melanin in the epidermal layer, and evenly distributing it in the capillary-enriched dermis independent of the skin colors. The 595 nm light is absorbed by hemoglobin (Hb) and oxygen-Hb within the capillaries, triggering thermal dilation of capillaries without damaging them or causing petechiae. The light illumination remarkably increases in the concentrations of various blood biomarkers in the skin through biomarker extravasation. These biomarkers bound specifically to the capture antibodies on OMNA with each microneedle covalently immobilized with one specific antibody. The OMNA is extensively modified to amplify the immunobinding signals and achieve sensitivity superior to that of enzyme-linked immunosorbent assay (ELISA) kits. As proof of concept, the functionality of the prototype for minimally invasive sampling and precise multiplexed blood biomarker detection in two mouse models is validated to quantify acute inflammation and specific antibody production.
Collapse
Affiliation(s)
- Zuan-Tao Lin
- Wellman Center for Photomedicine, Massachusetts General Hospital, Department of Dermatology, Harvard Medical School, Boston, MA, 02114, USA
| | - Biswabandhu Jana
- Wellman Center for Photomedicine, Massachusetts General Hospital, Department of Dermatology, Harvard Medical School, Boston, MA, 02114, USA
| | - Sandeep Korupolu
- Wellman Center for Photomedicine, Massachusetts General Hospital, Department of Dermatology, Harvard Medical School, Boston, MA, 02114, USA
| | - Yifei Kong
- Wellman Center for Photomedicine, Massachusetts General Hospital, Department of Dermatology, Harvard Medical School, Boston, MA, 02114, USA
| | - Guishi Liu
- Wellman Center for Photomedicine, Massachusetts General Hospital, Department of Dermatology, Harvard Medical School, Boston, MA, 02114, USA
| | - Yan Dong
- Wellman Center for Photomedicine, Massachusetts General Hospital, Department of Dermatology, Harvard Medical School, Boston, MA, 02114, USA
| | - Yongli Li
- Wellman Center for Photomedicine, Massachusetts General Hospital, Department of Dermatology, Harvard Medical School, Boston, MA, 02114, USA
| | - Quanwei Zhang
- Wellman Center for Photomedicine, Massachusetts General Hospital, Department of Dermatology, Harvard Medical School, Boston, MA, 02114, USA
| | - Wan Shou
- Department of Mechanical Engineering, University of Arkansas, Fayetteville, AR, 72701, USA
| | - Prabhat Upadhyay
- Wellman Center for Photomedicine, Massachusetts General Hospital, Department of Dermatology, Harvard Medical School, Boston, MA, 02114, USA
| | - Zhilong Wang
- Wellman Center for Photomedicine, Massachusetts General Hospital, Department of Dermatology, Harvard Medical School, Boston, MA, 02114, USA
| | - Zihan Ran
- Wellman Center for Photomedicine, Massachusetts General Hospital, Department of Dermatology, Harvard Medical School, Boston, MA, 02114, USA
| | - Mei X Wu
- Wellman Center for Photomedicine, Massachusetts General Hospital, Department of Dermatology, Harvard Medical School, Boston, MA, 02114, USA
| |
Collapse
|
15
|
Gao Y, Liu P, Peng L, Li M, Ni B. α-GalCer regulates acute stress-induced steroidogenesis by modulating lipid metabolism in female BALB/c mice. Psychoneuroendocrinology 2025; 178:107481. [PMID: 40328043 DOI: 10.1016/j.psyneuen.2025.107481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 04/26/2025] [Accepted: 05/01/2025] [Indexed: 05/08/2025]
Abstract
The immune system orchestrates the hypothalamus-pituitary-adrenal (HPA) axis response to stress. However, the impact of invariant natural killer T (iNKT) cell activation on stress-induced glucocorticoid levels remains poorly understood. Alpha-galactosylceramide (α-GalCer), a specific agonist for iNKT cells, activates iNKT cells to produce inflammatory cytokines including interleukin (IL)-4 and interferon (IFN)-γ. Our findings indicate that treatment with α-GalCer 3 hours before acute restraint stress suppressed the elevation of adrenocorticotropic hormone (ACTH) but did not affect the increase in corticosterone (CORT) in mice. However, treatment with α-GalCer 24 hours prior to restraint stress did not alter the rise in ACTH but reduced the increase in CORT by about half. This dissociation between stress-induced ACTH and CORT levels suggests an intra-adrenal regulation of HPA axis responses to acute stress following α-GalCer treatment. We further found that administration of α-GalCer enhances lipid utilization within adrenocortical cells and elicits a hyperresponsive reaction to ACTH stimulation. Mechanistically, IL-4 elevates the expression of type II 3β-hydroxysteroid dehydrogenase/isomerase (HSD3B2) and scavenger receptor class B type I (SRBI) protein in adrenocortical cells, thereby facilitating ACTH-induced glucocorticoid release. Additionally, we observed that acute stress amplifies both α-GalCer-induced IL-4 and IFN-γ production as well as liver injury. Our findings not only elucidate the mechanistic basis underlying interactions between immunity and stress but also highlight potential targets for therapeutic intervention.
Collapse
Affiliation(s)
- Yafei Gao
- Department of Military Psychology, School of Psychology, Army Medical University, Chongqing 400038, China
| | - Paiyu Liu
- Department of Pathophysiology, College of High Altitude Military Medicine, Army Medical University, Chongqing 400038, China
| | - Li Peng
- Department of Military Psychology, School of Psychology, Army Medical University, Chongqing 400038, China
| | - Min Li
- Department of Military Psychology, School of Psychology, Army Medical University, Chongqing 400038, China.
| | - Bing Ni
- Department of Pathophysiology, College of High Altitude Military Medicine, Army Medical University, Chongqing 400038, China.
| |
Collapse
|
16
|
Guerra FE, Karlinsey JE, Libby SJ, Fang FC. Evasion of serum antibodies and complement by Salmonella Typhi and Paratyphi A. PLoS Pathog 2025; 21:e1012917. [PMID: 40315236 PMCID: PMC12068720 DOI: 10.1371/journal.ppat.1012917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Revised: 05/12/2025] [Accepted: 04/18/2025] [Indexed: 05/04/2025] Open
Abstract
Nontyphoidal and enteric fever serovars of Salmonella enterica display distinctive interactions with serum antibodies and the complement system, which initiate the host immune response to invading microbes. This study examines the contributions of lipopolysaccharide O-antigen (O-ag) and the S. Typhi Vi polysaccharide capsule to serum resistance, complement activation and deposition, and immunoglobulin (Ig) binding in nontyphoidal S. enterica serovar Typhimurium and the enteric fever serovars S. Typhi and S. Paratyphi A. Although all three serovars are resistant to serum killing, S. Typhi and S. Paratyphi A exhibit lower levels of Ig binding, complement binding and complement activation compared to S. Typhimurium. In S. Typhimurium, WzzB-dependent long O-antigen (L O-ag) production with 16-to-35 repeating O-ag units, and FepE-dependent very long O-antigen (VL O-ag) production with over 100 repeating O-ag units, are required for serum resistance but do not prevent IgM binding or complement deposition. S. Typhi lacks VL O-ag, but its production of Vi capsule inhibits IgM binding and complement deposition, while acting in concert with L O-ag to resist serum killing. In S. Paratyphi A, L O-ag production is deficient due to a hypofunctional WzzB protein, but this is compensated by greater quantities of VL O-ag, which are required for serum resistance. Restoration of WzzB function by exchange with the S. Typhimurium or S. Typhi wzzB alleles can restore L O-ag production in S. Paratyphi A but decreases VL O-ag production, resulting in increased IgM binding. Replacement of the S. Paratyphi A O2-type polysaccharide with the S. Typhi O9 polysaccharide further increases IgM binding of S. Paratyphi A, which enhances complement activation but not complement deposition. Lastly, a gene duplication of rfbV in S. Paratyphi A is necessary for higher levels of VL O-ag and resistance to complement deposition and antibody binding. Collectively, these observations demonstrate fundamental differences between nontyphoidal and enteric fever Salmonella serovars in their interactions with innate immune effectors. Whereas nontyphoidal S. Typhimurium elicits, exploits and withstands the host acute inflammatory response, the enteric fever serovars S. Typhi and S. Paratyphi A evade it by limiting antibody recognition and complement activation and deposition.
Collapse
Affiliation(s)
- Fermin E. Guerra
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, United States of America
| | - Joyce E. Karlinsey
- Department of Microbiology, University of Washington, Seattle, Washington, United States of America
| | - Stephen J. Libby
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, United States of America
| | - Ferric C. Fang
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, United States of America
- Department of Microbiology, University of Washington, Seattle, Washington, United States of America
| |
Collapse
|
17
|
Krouch D, Vreeke GJC, America AHP, Mes JJ, Wierenga PA, Vincken JP, Bastiaan-Net S, Weegels PL. Amylase trypsin inhibitors activation of toll-like receptor 4 revisited: The dominance of lipopolysaccharides contamination. Int J Biol Macromol 2025; 310:143378. [PMID: 40288707 DOI: 10.1016/j.ijbiomac.2025.143378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 04/12/2025] [Accepted: 04/18/2025] [Indexed: 04/29/2025]
Abstract
Amylase trypsin inhibitors (ATIs) potentially play a role in irritable bowel syndrome (IBS) and non-celiac wheat sensitivity (NCWS). These cereal-derived inhibitors are suspected to bind to the TLR4-MD2-CD14 complex and trigger intestinal pro-inflammatory responses, but confirmation through more extensive cell line studies is required. In this study, an amylase trypsin inhibitors enriched fraction (AEF) was prepared and characterized. Then, AEF binding potential to TLR4-MD2-CD14 was investigated using the human TLR4 reporter cell line HEK-BlueTM. The method took into account the presence of lipopolysaccharides (LPS) using Polymyxin B (PMB) to block LPS binding to TLR4. Proteinase K was also used to hydrolyze AEF proteins and eliminate their induced response. The cell line experiments showed that PMB treatment of AEF reduced the binding signal by 92 %. Complete hydrolysis of the protein by Proteinase K doubled the TLR4 activation signal and might indicate that protein-LPS complexation reduced LPS's ability to activate the TLR4-receptor. These finding underline the need for future work to consider the non-protein part in cell assays, especially the LPS bias. Altogether, these results indicated that LPS activates TLR4-MD2-CD14 and challenges ATIs' intestinal inflammation capacity contributing in irritable bowel syndrome and non-celiac wheat sensitivity.
Collapse
Affiliation(s)
- Dounia Krouch
- Laboratory of Food Chemistry, Wageningen University & Research, P.O. Box 17, 6700 AA Wageningen, the Netherlands
| | - Gijs J C Vreeke
- Laboratory of Food Chemistry, Wageningen University & Research, P.O. Box 17, 6700 AA Wageningen, the Netherlands
| | - Antoine H P America
- Wageningen Plant Research, Wageningen University & Research, The Netherlands, P.O. Box 16, 6700 AA Wageningen, the Netherlands
| | - Jurriaan J Mes
- Wageningen Food & Biobased Research, Wageningen University & Research, P.O. Box 17, 6700 AA Wageningen, the Netherlands
| | - Peter A Wierenga
- Laboratory of Food Chemistry, Wageningen University & Research, P.O. Box 17, 6700 AA Wageningen, the Netherlands
| | - Jean-Paul Vincken
- Laboratory of Food Chemistry, Wageningen University & Research, P.O. Box 17, 6700 AA Wageningen, the Netherlands
| | - Shanna Bastiaan-Net
- Wageningen Food & Biobased Research, Wageningen University & Research, P.O. Box 17, 6700 AA Wageningen, the Netherlands.
| | - Peter L Weegels
- Laboratory of Food Chemistry, Wageningen University & Research, P.O. Box 17, 6700 AA Wageningen, the Netherlands.
| |
Collapse
|
18
|
Charity OJD, Thilliez G, Al-Khanaq H, Acton L, Kolenda R, Bawn M, Petrovska L, Kingsley RA. Reversible excision of the wzy locus in Salmonella Typhimurium may aid recovery following phage predation. PLoS Genet 2025; 21:e1011688. [PMID: 40315401 PMCID: PMC12074656 DOI: 10.1371/journal.pgen.1011688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 05/13/2025] [Accepted: 04/11/2025] [Indexed: 05/04/2025] Open
Abstract
Bacteriophage (phage) are promising novel antimicrobials but a key challenge to their effective implementation is the rapid emergence of phage resistance. An improved understanding of phage-host interactions is therefore needed. The Anderson phage typing scheme differentiates closely related strains of Salmonella enterica serovar Typhimurium (S. Typhimurium) based on sensitivity to a panel of phage preparations. Switches in phage type are indicative of changes in phage sensitivity and inform on the dynamics of phage interaction with their host bacteria. We investigated the molecular basis of switches between the relatively phage sensitive S. Typhimurium DT8 and phage resistant DT30 strains that are present in the same phylogenetic clade. DT30 strains emerged from DT8 strains predominantly by deletion of a genomic region affecting the wzy locus encoding an O-antigen polymerase. The deletion site was flanked by two perfect direct repeats designated attL and attR. During broth culture in the presence of a typing phage that used O-antigen as primary receptor the Δwzy genotype increased in frequency compared with culture in the absence of phage and removal of attL prevented deletion of the wzy locus. Co-culture of S. Typhimurium DT8 with a strain lacking wzy resulted in reversion of the latter to wild type. We propose a model in which reversible deletion of the wzy locus enables recovery of S. Typhimurium DT8 following predation by phage that use O-antigen as their primary receptor. This was consistent with ancestral state reconstruction of DT8 and DT30 phylogeny that supported a model of reversible transition from DT8 to DT30 in natural populations.
Collapse
Affiliation(s)
- Oliver JD Charity
- Quadram Institute Bioscience, Norwich, United Kingdom
- University of East Anglia, Norwich Research Park, Norwich, United Kingdom
| | - Gaetan Thilliez
- Quadram Institute Bioscience, Norwich, United Kingdom
- School of Biotechnology, Dublin City University, Dublin, Ireland
| | | | - Luke Acton
- Quadram Institute Bioscience, Norwich, United Kingdom
- University of East Anglia, Norwich Research Park, Norwich, United Kingdom
| | - Rafał Kolenda
- Quadram Institute Bioscience, Norwich, United Kingdom
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Wrocław University of Environmental and Life Sciences, Wrocław, Poland
| | - Matt Bawn
- Quadram Institute Bioscience, Norwich, United Kingdom
- Earlham Institute, Norwich, United Kingdom
- School of Natural and Environmental Sciences, Newcastle University, Newcastle, United Kingdom
| | - Liljana Petrovska
- Animal & Plant Health Agency (APHA), Weybridge, London, United Kingdom
| | - Robert A. Kingsley
- Quadram Institute Bioscience, Norwich, United Kingdom
- University of East Anglia, Norwich Research Park, Norwich, United Kingdom
| |
Collapse
|
19
|
Germon P, Foucras G, Smith DGE, Rainard P. Invited review: Mastitis Escherichia coli strains-Mastitis-associated or mammo-pathogenic? J Dairy Sci 2025; 108:4485-4507. [PMID: 40139360 DOI: 10.3168/jds.2024-26109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Accepted: 02/11/2025] [Indexed: 03/29/2025]
Abstract
Bovine mastitis remains a major concern for dairy farmers, mainly because of its effect on the economy of their activity and on animal welfare. Because Escherichia coli is considered a major mastitis pathogen, the diversity of E. coli strains isolated from mastitis cases has been studied for decades, with the aim to discover new ways to fight this infection. With the recent advances in whole-genome sequencing, a detailed view of the peculiarities of mastitis E. coli strains has emerged. This review aims to bring together the knowledge garnered over the years with the more recent results of whole-genome analyses. Whereas the concept of a mammary pathogenic E. coli has been proposed, because a common set of virulence genes cannot be identified among mastitis E. coli strains, we prefer the use of mastitis-associated E. coli (MAEC), with MAEC being more an "ecotype" rather than a "pathotype." Indeed, data available so far suggest that a common feature of MAEC would rather be an enrichment in fitness capabilities that makes them well-suited for survival and rapid adaptation to changing biotopes in the mammary gland, which we qualify as intramammary ecotopes.
Collapse
Affiliation(s)
- Pierre Germon
- INRAE, ISP UMR 1282, Université François Rabelais de Tours, 37380 Nouzilly, France.
| | - Gilles Foucras
- IHAP, Université de Toulouse, INRAE, ENVT, 31076 Toulouse, France
| | | | - Pascal Rainard
- INRAE, ISP UMR 1282, Université François Rabelais de Tours, 37380 Nouzilly, France
| |
Collapse
|
20
|
Tahmasebi A, Beheshti R, Mahmoudi M, Jalilzadeh M, Salehi-Pourmehr H. Alterations in gut microbial community structure in obstructive sleep apnea /hypopnea syndrome (OSAHS): A systematic review and meta-analysis. Respir Med 2025; 241:108077. [PMID: 40158663 DOI: 10.1016/j.rmed.2025.108077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Revised: 03/27/2025] [Accepted: 03/28/2025] [Indexed: 04/02/2025]
Abstract
OBJECTIVES This systematic review investigates gut bacterial diversity and composition in patients with Obstructive Sleep Apnea-Hypopnea Syndrome (OSAHS) and examines how these changes may contribute to cardiovascular complications. METHODS A comprehensive search was conducted in PubMed, Web of Science, and Scopus up to March 2025. After removing duplicates, titles and abstracts were screened by two reviewers, and full texts were assessed for inclusion. Data extraction on study characteristics and outcomes was performed. Methodological quality was evaluated using the Joanna Briggs Institute checklist. α-diversity was assessed using richness and diversity indices, while β-diversity examined community structure differences. Meta-analysis was conducted using standardized mean differences (SMD) and confidence intervals (CIs), and heterogeneity was assessed with the Cochrane I2 test. RESULTS The review included 18 studies (16 adults, 2 pediatrics) examining the gut microbiome in OSAHS. Meta-analysis revealed significant reductions in α-diversity indices (Shannon, Chao1, observed species, ACE) in OSAHS patients, while Simpson's index showed no difference. β-diversity analyses showed distinct gut microbiome differences in OSA. Key differential bacteria included Bacteroides, Proteobacteria, Faecalibacterium, Ruminococcaceae, Megamonas, Oscillibacter, Dialister, Roseburia, and Lachnospira. Study quality was medium to high. CONCLUSION OSAHS is associated with significant gut microbiome alterations, including a reduction in beneficial bacteria and an increase in LPS-producing bacteria, leading to intestinal barrier dysfunction. These changes may contribute to systemic inflammation and elevate the risk of cardiovascular diseases.
Collapse
Affiliation(s)
- Ali Tahmasebi
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran; Research Center for Evidence-Based Medicine, Iranian EBM Center: A Joanna Briggs Institute Center of Excellence, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Rasa Beheshti
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran; Research Center for Evidence-Based Medicine, Iranian EBM Center: A Joanna Briggs Institute Center of Excellence, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammadsina Mahmoudi
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran; Research Center for Evidence-Based Medicine, Iranian EBM Center: A Joanna Briggs Institute Center of Excellence, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mahan Jalilzadeh
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran; Research Center for Evidence-Based Medicine, Iranian EBM Center: A Joanna Briggs Institute Center of Excellence, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hanieh Salehi-Pourmehr
- Research Center for Evidence-Based Medicine, Iranian EBM Center: A Joanna Briggs Institute Center of Excellence, Tabriz University of Medical Sciences, Tabriz, Iran; Medical Philosophy and History Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
21
|
Cina NP, Klug CS. The LptC transmembrane helix undergoes a rigid body movement upon LptB 2FG cavity collapse. Protein Sci 2025; 34:e70133. [PMID: 40260908 PMCID: PMC12012751 DOI: 10.1002/pro.70133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2025] [Revised: 04/04/2025] [Accepted: 04/08/2025] [Indexed: 04/24/2025]
Abstract
Lipopolysaccharide (LPS) is an essential component of the cellular envelope of Gram-negative bacteria and contributes to antibiotic resistance and pathogenesis. Proper localization of LPS at the outer membrane is facilitated via seven distinct LPS transport (Lpt) proteins that bridge the inner and outer membranes. Mature LPS diffuses into the membrane cavity of the inner membrane ABC transporter LptB2FGC through a lateral gate formed by the LptF and LptG transmembrane (TM) helices. The TM helix of LptC intercalates within the LPS entry point and has been shown to regulate the ATPase activity of LptB2FG and contribute to thermal stability. Determination of the LptB2FGC open state structure revealed the location of the LptC TM helix within the membrane complex. However, in the closed state structure, the LptC TM helix is unresolved, suggesting the helix may be displaced from the lateral gate prior to or upon closure of the cavity. To determine the conformational states of the LptC TM helix in the open and closed LptB2FGC conformations, we utilized site-directed spin labeling in combination with both continuous wave electron paramagnetic resonance (EPR) and double electron electron resonance (DEER) spectroscopies to investigate the LptC TM helix and linker region. These data indicate that the LptC TM helix undergoes a rigid body movement away from the central LptB2FG cavity upon cavity closure. The findings presented here will support structure-based drug design optimization of recently discovered antibiotics that bind LptB2FG and occlude the LptC TM helix from the lateral gate.
Collapse
Affiliation(s)
- Nicholas P. Cina
- Department of BiophysicsMedical College of WisconsinMilwaukeeWisconsinUSA
| | - Candice S. Klug
- Department of BiophysicsMedical College of WisconsinMilwaukeeWisconsinUSA
| |
Collapse
|
22
|
Zeng Z, Wei N, Cai X, Xiao J. A magnetic bifunctional endotoxin removal nano-agent for the efficient elimination of endotoxins in recombinant protein preparation. Int J Biol Macromol 2025; 311:143663. [PMID: 40311983 DOI: 10.1016/j.ijbiomac.2025.143663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Revised: 04/21/2025] [Accepted: 04/28/2025] [Indexed: 05/03/2025]
Abstract
Endotoxin contamination, as one of the most significant challenges in recombinant protein production by Escherichia coli, represents a critical biosafety concern and greatly hinders the biomedical application of recombinant proteins. Conventional methods, such as extreme-condition inactivation and chromatography-based separation, are plagued by issues including protein denaturation, low efficiency, and operational complexity in endotoxin removal. In this work, we developed a novel magnetic bifunctional endotoxin removal nano-agent (MagBER) with a multi-layered structure, consisting of a superparamagnetic Fe3O4 core, a mesoporous TiO2 intermediary layer, and an outer shell functionalized with boronic acid groups. This dual-functional design significantly enhances endotoxin removal efficiency through the TiO2 layer and boronic acid groups, ensuring stable endotoxin clearance performance even in high-salt environments and complex biological matrices. MagBER exhibits reusability while maintaining protein structural integrity. Moreover, MagBER has been successfully employed for endotoxin removal in various proteins, establishing it as a promising and sustainable solution for endotoxin clearance in biopharmaceutical applications.
Collapse
Affiliation(s)
- Zibing Zeng
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, PR China; Gansu Engineering Research Center of Medical Collagen, Lanzhou, Gansu 730000, PR China
| | - Nannan Wei
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, PR China; Gansu Engineering Research Center of Medical Collagen, Lanzhou, Gansu 730000, PR China
| | - Xiangdong Cai
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, PR China; Gansu Engineering Research Center of Medical Collagen, Lanzhou, Gansu 730000, PR China.
| | - Jianxi Xiao
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, PR China; Gansu Engineering Research Center of Medical Collagen, Lanzhou, Gansu 730000, PR China.
| |
Collapse
|
23
|
Zhou Z, Zhang P, Chen D, Kong N, Liu H, Liang J, Huang K, Wang H. Cecropin A-Derived Peptide for the Treatment of Osteomyelitis by Inhibiting the Growth of Multidrug-Resistant Bacteria and Eliminating Inflammation. ACS NANO 2025; 19:15733-15750. [PMID: 40231707 DOI: 10.1021/acsnano.4c18858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/16/2025]
Abstract
Osteomyelitis poses substantial therapeutic challenges due to the prevalence of multidrug-resistant bacterial infections and associated inflammation. Current treatment regimens often rely on a combination of corticosteroids and antibiotics, which can lead to complications and impede effective bacterial clearance. In this study, we present CADP-10, a Cecropin A-derived peptide, designed to target methicillin-resistant Staphylococcus aureus (MRSA) and multidrug-resistant Escherichia coli (MRE), while simultaneously addressing inflammatory responses. CADP-10 self-assembles into nanobacterial net (NBacN) that selectively identify and bind to bacterial endotoxins (LPS and LTA), disrupting membrane integrity and depolarizing membrane potential, which culminates in bacterial death. Importantly, these NBacN are bound to LPS and LTA from dead bacteria, preventing their engagement with TLR receptors and effectively blocking downstream inflammatory pathways. Our assessments of CADP-10 demonstrate good biosafety in both in vitro and in vivo models. Notably, in a rabbit osteomyelitis model, CADP-10 eliminated MRSA-induced bone infections, mitigated inflammation, and promoted bone tissue regeneration. This research highlights the potential of CADP-10 as a multifunctional antimicrobial agent for the management of infectious inflammatory diseases.
Collapse
Affiliation(s)
- Ziao Zhou
- Department of Chemistry, Zhejiang University, Hangzhou 310027, Zhejiang Province, China
- Department of Chemistry, School of Science, Institute of Natural Sciences, Westlake Institute for Advanced Study, Westlake University, 18 Shilongshan Road, Hangzhou 310024, Zhejiang Province, China
| | - Peng Zhang
- Department of Chemistry, School of Science, Institute of Natural Sciences, Westlake Institute for Advanced Study, Westlake University, 18 Shilongshan Road, Hangzhou 310024, Zhejiang Province, China
| | - Dinghao Chen
- Department of Chemistry, School of Science, Institute of Natural Sciences, Westlake Institute for Advanced Study, Westlake University, 18 Shilongshan Road, Hangzhou 310024, Zhejiang Province, China
| | - Nan Kong
- Department of Chemistry, School of Science, Institute of Natural Sciences, Westlake Institute for Advanced Study, Westlake University, 18 Shilongshan Road, Hangzhou 310024, Zhejiang Province, China
| | - Huayang Liu
- Department of Chemistry, School of Science, Institute of Natural Sciences, Westlake Institute for Advanced Study, Westlake University, 18 Shilongshan Road, Hangzhou 310024, Zhejiang Province, China
| | - Juan Liang
- Department of Chemistry, School of Science, Institute of Natural Sciences, Westlake Institute for Advanced Study, Westlake University, 18 Shilongshan Road, Hangzhou 310024, Zhejiang Province, China
| | - Kai Huang
- Department of Orthopedics, Tongde Hospital of Zhejiang Province, No. 234 Gucui Road, Hangzhou 310012, China
| | - Huaimin Wang
- Department of Chemistry, School of Science, Institute of Natural Sciences, Westlake Institute for Advanced Study, Westlake University, 18 Shilongshan Road, Hangzhou 310024, Zhejiang Province, China
| |
Collapse
|
24
|
Pharkjaksu S, Cai H, Walter PJ, Chang YC, Kwon-Chung KJ. Elevated UDP-glucuronic acid levels mend drug resistance and stress responses via a protease and a transporter in Cryptococcus gattii. Proc Natl Acad Sci U S A 2025; 122:e2503960122. [PMID: 40267138 PMCID: PMC12054807 DOI: 10.1073/pnas.2503960122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Accepted: 03/18/2025] [Indexed: 04/25/2025] Open
Abstract
UDP-glucuronic acid (UDP-GlcUA) is a nucleotide sugar essential for various biological processes in many organisms, and its excess within the cell can disrupt cellular functions. In Cryptococcus, mutations in the UXS1 gene which encodes an enzyme responsible for converting UDP-GlcUA into UDP-xylose, result in excessive accumulation of UDP-GlcUA and confer resistance to the antifungal drug 5-fluorocytosine. Here, we demonstrate that elevation of UDP-GlcUA affects several cellular processes in Cryptococcus gattii, including growth rate, ability to grow under various stress conditions and resistance to fluorinated pyrimidine analogs. RNA-seq analyses of the uxs1Δ mutant identify three acid protease genes, notably PEP401, that are differentially expressed. The absence of PEP401 in the uxs1Δ background significantly reduces UDP-GlcUA levels and reverts all the phenotypes of the uxs1Δ mutant to the wild-type characteristics. High levels of UDP-GlcUA not only regulate expression of PEP401 at RNA and protein levels but also enhance the proteolytic activity of total protein extracts in a PEP401-dependent manner, establishing a functional link between nucleotide sugar metabolism and proteolytic regulation. Moreover, the UDP-GlcUA transporter gene, UUT1, can further modulate the levels of UDP-GlcUA in the uxs1Δ pep401Δ double mutant and manifests drug resistance phenotypes observed in the uxs1Δ mutant. Collectively, these findings reveal a previously unrecognized regulatory network that links UDP-GlcUA metabolism to protease-mediated cellular processes and the transport of UDP-GlcUA. This interaction provides a foundation for targeting nucleotide sugar metabolism and protease regulation in the development of enhanced therapeutic strategies against cryptococcosis.
Collapse
Affiliation(s)
- Sujiraphong Pharkjaksu
- Molecular Microbiology Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD20892
| | - Hongyi Cai
- Clinical Mass Spectrometry Core, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, MD20892
| | - Peter J. Walter
- Clinical Mass Spectrometry Core, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, MD20892
| | - Yun C. Chang
- Molecular Microbiology Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD20892
| | - Kyung J. Kwon-Chung
- Molecular Microbiology Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD20892
| |
Collapse
|
25
|
Chancharoenthana W, Kamolratanakul S, Udompornpitak K, Wannigama DL, Schultz MJ, Leelahavanichkul A. Alcohol-induced gut permeability defect through dysbiosis and enterocytic mitochondrial interference causing pro-inflammatory macrophages in a dose dependent manner. Sci Rep 2025; 15:14710. [PMID: 40289168 PMCID: PMC12034794 DOI: 10.1038/s41598-025-97593-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Accepted: 04/07/2025] [Indexed: 04/30/2025] Open
Abstract
Although toxicity of alcohol toward the intestines and immunity is mentioned, there might be different effect of alcohol in a low and a high dose and the rodent model development using a simple SHIRPA binary score night be useful. Hence, a low and high dose of alcohol (6.30 and 1.26 g/kg/day) were administered in might for 16 weeks before determination of several parameters. As such, the peak blood alcohol concentration (BAC) of low and high dose of alcohol were approximately at 0.05 and 0.15%, respectively, at 1 h post-administration, which correlated with SHIRPA score at 1.8 ± 0.8 and 7.2 ± 0.6, respectively. After 16 wk of administration, a significant liver injury in high-dose alcohol was indicated by liver enzymes, liver weight, histology score, apoptosis, and hepatic accumulation of triglyceride (TG) and oxidative stress (malondialdehyde; MDA) with reduced anti-oxidant (glutathione). Meanwhile, low-dose alcohol demonstrated only elevated apoptosis with increased TG and MDA in liver tissue. Leaky gut from both dose of alcohol was also demonstrated by FITC-dextran, endotoxemia, serum beta glucan, and reduced occludin. However, bacterial abundance (microbiome analysis) of the feces from small bowel of high-dose alcohol, but not the low dose, was different from the control (increased Alitipes spp. with reduced Lachnospiraceae). In conclusion, both low- and high-dose alcohol induced leaky gut, while only the high-dose caused gut dysbiosis and alcohol damaged mitochondria but enhanced glycolysis in enterocytes and macrophages. Leaky gut might be more sensitive than dysbiosis to determine alcohol-induced intestinal injury.
Collapse
Affiliation(s)
- Wiwat Chancharoenthana
- Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, 16/F Ratchanakarin Building 420/6 Rajvithi Rd., Ratchathewi, Bangkok, 10400, Thailand.
- Tropical Immunology and Translational Research Unit (TITRU), Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok, 10400, Thailand.
| | - Supitcha Kamolratanakul
- Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, 16/F Ratchanakarin Building 420/6 Rajvithi Rd., Ratchathewi, Bangkok, 10400, Thailand
- Tropical Immunology and Translational Research Unit (TITRU), Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok, 10400, Thailand
| | - Kanyarat Udompornpitak
- Center of Excellence on Translational Research in Inflammation and Immunology (CETRII), Department of Microbiology, Chulalongkorn University, Bangkok, 10500, Thailand
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, 10500, Thailand
| | - Dhammika Leshan Wannigama
- Department of Infectious Diseases, Faculty of Medicine Yamagata University and Yamagata University Hospital, Yamagata, Japan
- Department of Infectious Diseases and Infection Control, Yamagata Prefectural Central Hospital, Yamagata, Japan
- Pathogen Hunter's Research Collaborative Team, Department of Infectious Diseases and Infection Control, Yamagata Prefectural Central Hospital, Yamagata, Japan
- Yamagata Prefectural University of Health Sciences, Kamiyanagi, Yamagata, 990-2212, Japan
- Biofilms and Antimicrobial Resistance Consortium of ODA receiving countries, The University of Sheffield, Sheffield, UK
| | - Marcus J Schultz
- Department of Intensive Care and Laboratory of Experimental Intensive Care and Anesthesiology (L.E.I.C.A), Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, Oxford University, Oxford, UK
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, 10400, Thailand
| | - Asada Leelahavanichkul
- Center of Excellence on Translational Research in Inflammation and Immunology (CETRII), Department of Microbiology, Chulalongkorn University, Bangkok, 10500, Thailand.
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, 10500, Thailand.
- Immunology Unit, Department of Microbiology, Chulalongkorn University, Bangkok, 10330, Thailand.
| |
Collapse
|
26
|
Rahman MM, Talukder A, Rahi MS, Das PK, Grice ID, Ulett GC, Wei MQ. Evaluation of Immunostimulatory Effects of Bacterial Lysate Proteins on THP-1 Macrophages: Pro-inflammatory Cytokine Response and Proteomic Profiling. J Immunol Res 2025; 2025:2289241. [PMID: 40322557 PMCID: PMC12048194 DOI: 10.1155/jimr/2289241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Accepted: 04/02/2025] [Indexed: 05/08/2025] Open
Abstract
Bacterial lysate proteins (BLPs) serve as potential immunostimulants, recognized by pattern recognition receptors (PRRs) on immune cells, eliciting a robust immune response. In this study, THP-1 macrophages were treated with varying doses of BLPs derived from Streptococcus pyogenes (SP), Streptococcus agalactiae (SA), and Serratia marcescens (SM). The results showed significant increases (p < 0.05) in pro-inflammatory cytokines such as TNF-α, IL-1β, IL-6, IL-12, granulocyte macrophage-colony stimulating factor (GM-CSF), eotaxin, and macrophage inflammatory protein (MIP)-1α, except for 5 µg of all BLPs for TNF-α and eotaxin, and 5 µg of SP for IL-12 production. No significant differences were found between the corresponding doses of SP and SA or SP and SM, except for GM-CSF in all doses, while SA and SM only showed a difference at the 5 µg dose for GM-CSF. Furthermore, there were no significant differences between the 10 and 20 µg doses of all BLPs, indicating that doses higher than 10 µg do not significantly enhance the pro-inflammatory response. Combination doses of SP + SM and SA + SM did not show significant differences, except for IL-1β, suggesting no synergistic effect. Cytotoxicity was observed to increase with higher BLP concentrations in a dose-dependent manner, with combinations of SP + SM and SA + SM exhibiting greater cytotoxicity than the individual BLPs. Proteomic analysis of BLPs identified immunostimulatory proteins, including heat shock proteins (HSPs; ClpB, DnaK, and GroEL), metabolic enzymes (glyceraldehyde 3-phosphate dehydrogenase (GAPDH), enolase, and arginine deiminase (ADI)), and surface and secreted proteins (ESAT-6-like protein, CRISPR-associated endonuclease Cas9, OmpA, porin OmpC, and serralysin), which are involved in immune modulation, bacterial clearance, and immune evasion. This study underscores the potential of bacterial proteins as vaccine adjuvants or supplementary therapies; however, further research is essential to find a balance between immune activation and inflammation reduction to develop safer and more effective immunostimulants.
Collapse
Affiliation(s)
- Md. Mijanur Rahman
- School of Pharmacy and Medical Sciences, Griffith University, Gold Coast, Queensland, Australia
- Department of Microbiology, Noakhali Science and Technology University, Noakhali, Chittagong, Bangladesh
| | - Asma Talukder
- School of Pharmacy and Medical Sciences, Griffith University, Gold Coast, Queensland, Australia
| | - Md. Sifat Rahi
- School of Pharmacy and Medical Sciences, Griffith University, Gold Coast, Queensland, Australia
| | - Plabon Kumar Das
- Institute for Biomedicine and Glycomics, Griffith University, Gold Coast, Queensland, Australia
| | - I. Darren Grice
- School of Pharmacy and Medical Sciences, Griffith University, Gold Coast, Queensland, Australia
- Institute for Biomedicine and Glycomics, Griffith University, Gold Coast, Queensland, Australia
| | - Glen C. Ulett
- School of Pharmacy and Medical Sciences, Griffith University, Gold Coast, Queensland, Australia
- Institute for Biomedicine and Glycomics, Griffith University, Gold Coast, Queensland, Australia
| | - Ming Q. Wei
- School of Pharmacy and Medical Sciences, Griffith University, Gold Coast, Queensland, Australia
| |
Collapse
|
27
|
Sohail S, Wisal A, Rana S, Abid Khan RM, Ullah A, Khan FA, Irfan M, Imran M, Carvalho PVSD, Rozental R, Shaheen F, Hassan SS. Druggable proteins of Alistipes reveal promising antimicrobial targets against chronic intestinal inflammation. Front Pharmacol 2025; 16:1555062. [PMID: 40342998 PMCID: PMC12058783 DOI: 10.3389/fphar.2025.1555062] [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: 01/12/2025] [Accepted: 03/31/2025] [Indexed: 05/11/2025] Open
Abstract
Introduction The genus Alistipes consists of anaerobic, Gram-negative bacteria with 13 species that colonize the entire gastrointestinal tract and are a serious health concern. They contribute to gut dysbiosis, intestinal inflammation, colorectal cancer, and depression. Methods To explore potential therapeutic targets and inhibitors, we filtered the core genome of Alistipes strains through subtractive genomics for non-host homology, gene essentiality, PPI, KEGG pathways, virulence, cellular localization, and druggability. The potential targets were docked against two drug-like libraries (ZINC, n = 11,993) and TCM (n = 36,043). ADMET profiling for best hits and MD simulation for apo/complex structures were performed, followed by physicochemical and pharmacokinetic evaluation and complex stabilities. Results and Discussion A set of 39 potential proteins was drastically reduced to only two targets after sequential data mining. The 3D structures of the selected targets (LpxA and KdsB) revealed good druggability scores. The top hits (ZINC85530940, ZINC05161112, ZINC95911713, and ZINC05566415) for both targets showed maximum H-bond interactions. The RMSD and RMSF values exhibited compactness with minimum fluctuation in ligand-bound complexes. The β-factor of ZINC05161112 at 327th residue and 352nd residue exhibited higher thermal instability, consistent with the RMSF results. The globularity of the complexes and apo structures remained consistent, whereas the LpxA complexes exhibited lower solvent-accessible surface area. For the KdsB, the surface area for ZINC5566415 increased significantly, with a steep decrease for ZINC95911713, establishing rather stable protein-ligand complexes. The results highlight the importance of identifying novel inhibitors and therapeutic targets. They are crucial for establishing better treatment regimes for human health and to aid in controlling the pathogenicity of Alistipes species.
Collapse
Affiliation(s)
- Saman Sohail
- Department of Chemistry, Islamia College Peshawar, Peshawar, Pakistan
| | - Ayesha Wisal
- Department of Chemistry, Islamia College Peshawar, Peshawar, Pakistan
| | - Sobia Rana
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences (PCMD-ICCBS), University of Karachi, Karachi, Pakistan
| | - Rao Muhammad Abid Khan
- Department of Clinical Microbiology, Sindh Institute of Urology and Transplantation (SIUT), Karachi, Pakistan
| | - Asad Ullah
- Department of Chemistry, Islamia College Peshawar, Peshawar, Pakistan
| | | | - Muhammad Irfan
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences (PCMD-ICCBS), University of Karachi, Karachi, Pakistan
| | - Muhammad Imran
- Research Center for Advanced Materials Science (RCAMS), Department of Chemistry, Faculty of Science, King Khalid University, Abha Aseer, Saudi Arabia
| | - Paulo V. S. D. Carvalho
- Centre for Technological Development in Health (CDTS), Oswaldo Cruz Foundation (Fiocruz), Rio de Janeiro, Brazil
| | - Renato Rozental
- Centre for Technological Development in Health (CDTS), Oswaldo Cruz Foundation (Fiocruz), Rio de Janeiro, Brazil
| | - Farzana Shaheen
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences (PCMD-ICCBS), University of Karachi, Karachi, Pakistan
- TWC, HEJ-ICCBS, University of Karachi, Karachi, Pakistan
| | - Syed Shah Hassan
- Department of Chemistry, Islamia College Peshawar, Peshawar, Pakistan
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences (PCMD-ICCBS), University of Karachi, Karachi, Pakistan
| |
Collapse
|
28
|
Ascari A, Morona R. Recent insights into Wzy polymerases and lipopolysaccharide O-antigen biosynthesis. J Bacteriol 2025; 207:e0041724. [PMID: 40066993 PMCID: PMC12004945 DOI: 10.1128/jb.00417-24] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/18/2025] Open
Abstract
Bacteria synthesize a plethora of complex surface-associated polysaccharides which enable them to persist and thrive in distinct niches. These glycans serve an array of purposes pertaining to virulence, colonization, antimicrobial resistance, stealth, and biofilm formation. The Wzx/Wzy-dependent pathway is universally the predominant system for bacterial polysaccharide synthesis. This system is responsible for the production of lipopolysaccharide (LPS) O-antigen (Oag), enterobacterial common antigen, capsule, and exopolysaccharides, with orthologs present in both Gram-negative and Gram-positive microbes. Studies focusing principally on Pseudomonas, Shigella, and Salmonella LPS Oag synthesis have provided much of the framework underpinning the biochemical and molecular mechanism behind polysaccharide synthesis via this pathway. LPS Oag production via the Wzx/Wzy-dependent pathway occurs through the stepwise activity of multiple key biosynthetic enzymes, including primarily the polymerase, Wzy, which is responsible for the Oag assembly, and the polysaccharide co-polymerase, Wzz, which effectively modulates the length of the glycan produced. In this review, we provide a comprehensive summary of the latest genetic, structural, and mechanistic data for the main protein candidates of the Wzx/Wzy-dependent pathway, in addition to an examination of their substrate specificities. Furthermore, we have reviewed recent insights pertaining to the dynamics/kinetics of glycan synthesis by this mechanism, including the interplay of the key proteins among themselves and in complex with their substrate. Lastly, we outline key gaps in the literature and suggest future research avenues, with the aim to stimulate ongoing research into this critical pathway responsible for the production of key virulence factors for numerous debilitating and lethal pathogens.
Collapse
Affiliation(s)
- Alice Ascari
- Institute for Biomedicine and Glycomics, Griffith University, Gold Coast, Queensland, Australia
| | - Renato Morona
- School of Biological Sciences, Department of Molecular and Biomedical Science, Research Centre for Infectious Diseases, University of Adelaide, Adelaide, Australia
| |
Collapse
|
29
|
Magri M, Eljaoudi R, Belyamani L, Ibrahimi A, Bouricha EM. In silico analysis of zosurabalpin-LptB2FG binding in Acinetobacter spp., Klebsiella pneumoniae, and Shigella flexneri: mechanisms underlying its differential efficacy. In Silico Pharmacol 2025; 13:62. [PMID: 40255254 PMCID: PMC12003254 DOI: 10.1007/s40203-025-00343-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Accepted: 03/18/2025] [Indexed: 04/22/2025] Open
Abstract
Zosurabalpin, a novel tethered macrocyclic peptide antibiotic, exhibits potent activity against Acinetobacter spp., particularly carbapenem-resistant Acinetobacter baumannii (CRAB). Zosurabalpin inhibits lipopolysaccharide (LPS) transport by targeting the LptB2FG protein complex, resulting in toxic LPS accumulation and bacterialdeath. This study investigates zosurabalpin's molecular specificity against Acinetobacter spp., its ineffectiveness against Klebsiella pneumoniae, and its potential efficacy against Shigella flexneri. Comparative analysis of LptB2FG sequences and structures, revealed significant differences in LptB2FG protein conformations, pocket geometry and electrostatic surface surrounding the binding pocket among the three species, which may influence zosurabalpin binding. Docking results for zosurabalpin showed lower binding affinities for K. pneumoniae and S. flexneri compared to Acinetobacter baylyi. Additionally, other zosurabalpin derivatives were tested showing improved binding affinities for K. pneumoniae but not for S. flexneri. These findings underscore the need for tailored zosurabalpin derivatives to enhance efficacy against a broader spectrum of Gram-negative bacteria. Supplementary Information The online version contains supplementary material available at 10.1007/s40203-025-00343-3.
Collapse
Affiliation(s)
- Meryam Magri
- Medical Biotechnology Laboratory (MedBiotech), Faculty of Medicine and Pharmacy of Rabat, Mohammed Vth University in Rabat, Rabat, Morocco
| | - Rachid Eljaoudi
- Medical Biotechnology Laboratory (MedBiotech), Faculty of Medicine and Pharmacy of Rabat, Mohammed Vth University in Rabat, Rabat, Morocco
| | - Lahcen Belyamani
- Mohammed VI University of Sciences and Health (UM6SS), Casablanca, Morocco
- Mohammed VI Center for Research and Innovation (CM6RI), Rabat, Morocco
- Emergency Department, Military Hospital Mohammed V, Rabat, Morocco
| | - Azeddine Ibrahimi
- Medical Biotechnology Laboratory (MedBiotech), Faculty of Medicine and Pharmacy of Rabat, Mohammed Vth University in Rabat, Rabat, Morocco
| | - El Mehdi Bouricha
- Mohammed VI University of Sciences and Health (UM6SS), Casablanca, Morocco
- Mohammed VI Center for Research and Innovation (CM6RI), Rabat, Morocco
| |
Collapse
|
30
|
Ahmad B, Abbas Z, Xubiao W, Hussain K, Rehman A, Dayong S, Abbas A, Hölzel C, Rijun Z. Synergistic effects of recombinant expressed Fowlicidin and Thymosin α1 hybrid peptides in modulating inflammation and infection in avian macrophages. Front Microbiol 2025; 16:1568451. [PMID: 40309114 PMCID: PMC12042281 DOI: 10.3389/fmicb.2025.1568451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2025] [Accepted: 03/26/2025] [Indexed: 05/02/2025] Open
Abstract
The hybrid FowlTα1 peptide represents a promising biomolecule synthesized from two naturally occurring peptides, namely Fowlicidins (Fowl) and Thymosin α1 (Tα1). This particular peptide exhibits remarkable anti-inflammatory and antimicrobial properties and demonstrates the capacity to effectively interact with lipopolysaccharide (LPS), while simultaneously inducing minimal cytotoxicity and hemolytic repercussions. Despite its potential, the high cost of this peptide has limited its use. To overcome this limitation, the present study developed a cost-effective and biocompatible method for expressing the FowlTα1 peptide in Pichia pastoris (P. pastoris). We obtained a transgenic strain of the hybrid FowlTα1 peptide with a predicted molecular weight of 3.1 kDa. The FowlTα1 peptide was purified followed by reverse-phase high-performance liquid chromatography (RP-HPLC), yielding 7.2 mg with a purity of 98.2%. Furthermore, physiochemical and structural analysis revealed an amphipathic helical configuration that enhances bioactivity. Moreover, in LPS-stimulated HD11 macrophages, the hybrid FowlTα1 peptide significantly reduced the release of nitric oxide (NO), TNF-α, IL-6, and IL-1β in a dose-dependent manner (p < 0.05) and displayed robust antimicrobial activity against Escherichia coli (E. coli) compared to conventional antibiotic. Overall, the results of this study highlighted the production method and potential of the FowlTα1 peptide as a novel therapeutic agent for antimicrobial, anti-inflammatory, and anti-endotoxin applications.
Collapse
Affiliation(s)
- Baseer Ahmad
- Laboratory of Feed Biotechnology, State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
- Faculty of Animal and Veterinary Sciences, Muhammad Nawaz Shareef University of Agriculture, Multan, Pakistan
| | - Zaheer Abbas
- Laboratory of Feed Biotechnology, State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Wei Xubiao
- Laboratory of Feed Biotechnology, State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Kashif Hussain
- Faculty of Animal and Veterinary Sciences, Muhammad Nawaz Shareef University of Agriculture, Multan, Pakistan
| | - Atif Rehman
- Faculty of Animal and Veterinary Sciences, Muhammad Nawaz Shareef University of Agriculture, Multan, Pakistan
| | - Si Dayong
- Laboratory of Feed Biotechnology, State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Asghar Abbas
- Faculty of Animal and Veterinary Sciences, Muhammad Nawaz Shareef University of Agriculture, Multan, Pakistan
| | - Christina Hölzel
- Institute for Animal Breeding and Animal Husbandry, Kiel University, Kiel, Germany
| | - Zhang Rijun
- Laboratory of Feed Biotechnology, State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| |
Collapse
|
31
|
Hu M, Chua SL. Antibiotic-Resistant Pseudomonas aeruginosa: Current Challenges and Emerging Alternative Therapies. Microorganisms 2025; 13:913. [PMID: 40284749 PMCID: PMC12029751 DOI: 10.3390/microorganisms13040913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2025] [Revised: 04/04/2025] [Accepted: 04/10/2025] [Indexed: 04/29/2025] Open
Abstract
Antibiotic-resistant Pseudomonas aeruginosa is a pathogen notorious for its resilience in clinical settings due to biofilm formation, efflux pumps, and the rapid acquisition of resistance genes. With traditional antibiotic therapy rendered ineffective against Pseudomonas aeruginosa infections, we explore alternative therapies that have shown promise, including antimicrobial peptides, nanoparticles and quorum sensing inhibitors. While these approaches offer potential, they each face challenges, such as specificity, stability, and delivery, which require careful consideration and further study. We also delve into emerging alternative strategies, such as bacteriophage therapy and CRISPR-Cas gene editing that could enhance targeted treatment for personalized medicine. As most of them are currently in experimental stages, we highlight the need for clinical trials and additional research to confirm their feasibility. Hence, we offer insights into new therapeutic avenues that could help address the pressing issue of antibiotic-resistant Pseudomonas aeruginosa, with an eye toward practical applications in future healthcare.
Collapse
Affiliation(s)
- Minqi Hu
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China
| | - Song Lin Chua
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China
- State Key Laboratory of Chemical Biology and Drug Discovery, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China
- Research Centre for Deep Space Explorations (RCDSE), The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China
- Research Institute for Future Food (RiFood), The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China
| |
Collapse
|
32
|
Cheon M, Kim W, Chung C. Distinct Impact of Inflammatory Versus Psychophysiological Stress on Brain-Wide Activation of Melanocortin Receptor 4-Expressing Neurons. FASEB J 2025; 39:e70527. [PMID: 40203074 PMCID: PMC11981239 DOI: 10.1096/fj.202403158r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 03/11/2025] [Accepted: 03/27/2025] [Indexed: 04/11/2025]
Abstract
Central melanocortin signaling plays a critical role in maintaining energy homeostasis by regulating energy intake and expenditure, with impairment of this system closely related to metabolic diseases such as obesity. Among melanocortin receptor subtypes, melanocortin receptor 4 (MC4R) is the primary mediator of these effects within the central nervous system. Accumulating evidence suggests that MC4R contributes to stress-induced disruptions in feeding behavior and energy homeostasis. However, the precise neural mechanisms by which stress alters MC4R activity remain incompletely understood. In this study, we compared brain-wide c-Fos expression patterns induced by two distinct stress paradigms: lipopolysaccharide (LPS)-induced inflammatory stress and restraint stress in male mice, and further examined the involvement of MC4R-expressing (MC4R+) neurons in these stress conditions. We found that both stressors elicited c-Fos activation in brain areas associated with stress responses as well as feeding regulation. Notably, LPS-induced stress, but not restraint stress, selectively activated MC4R+ neurons in the central amygdala (CeA) and oval nucleus of the bed nucleus of stria terminalis (ovBNST). These results highlight the distinct recruitment of MC4R+ neurons during acute inflammatory stress in male mice, offering novel insights into the role of MC4R in the stress-induced imbalance of energy homeostasis depending on stressor types.
Collapse
Affiliation(s)
- Myunghyun Cheon
- Department of Biological SciencesKonkuk UniversitySeoulSouth Korea
| | - Woonhee Kim
- Department of Biological SciencesKonkuk UniversitySeoulSouth Korea
| | - ChiHye Chung
- Department of Biological SciencesKonkuk UniversitySeoulSouth Korea
| |
Collapse
|
33
|
Goto T, Blaukopf M, Stöger B, Pantophlet R, Kerner L, Kosma P. Glycosylation of an N-Acetylated Glucosamine Disaccharide Using an Orthogonally Protected 3-Iodo-Kdo Fluoride Donor. ChemistryOpen 2025:e2500141. [PMID: 40223430 DOI: 10.1002/open.202500141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2025] [Revised: 03/31/2025] [Indexed: 04/15/2025] Open
Abstract
Kdo (3-Deoxy-d-manno-oct-2-ulosonic acid) is an essential sugar found in bacterial lipopolysaccharides with significant biomedical relevance. This study introduces an orthogonally protected 3-iodo-Kdo fluoride donor and demonstrates its coupling to a pre-synthesized β-(1→6)-linked N-acetylglucosamine disaccharide acceptor as an example. Nuclear magnetic resonance data indicates the presence of an intraresidue hydrogen bond in the distal glucosamine unit. Two complementary glycosylation approaches are explored with an emphasis on achieving high stereoselectivity and minimizing protecting-group manipulation. The orthogonal protection of 3-iodo Kdo fluoride donor offers insights into tailoring Kdo-based donors for specific biomedical applications. While yields vary depending on the approach, they are sufficient to demonstrate the donor's applicability. These findings enable the design of advanced glycomimetic constructs for therapeutic and vaccine research.
Collapse
Affiliation(s)
- Takaaki Goto
- Institute of Organic Chemistry, University of Natural Resources and Life Sciences-Vienna, Muthgasse 18, A-1190, Vienna, Austria
| | - Markus Blaukopf
- Institute of Organic Chemistry, University of Natural Resources and Life Sciences-Vienna, Muthgasse 18, A-1190, Vienna, Austria
| | - Berthold Stöger
- X-Ray Center (XRC), University of Technology Vienna, Lehargasse 4, A-1060, Vienna, Austria
| | - Ralph Pantophlet
- Faculty of Health Sciences, Simon Fraser University, 8888 University Drive, Burnaby, V5A1S6, Canada
| | - Lukáš Kerner
- Institute of Organic Chemistry, University of Natural Resources and Life Sciences-Vienna, Muthgasse 18, A-1190, Vienna, Austria
- Department of Organic Chemistry, Faculty of Natural Sciences, Comenius University, Bratislava, SK, 842 15, Slovakia
| | - Paul Kosma
- Institute of Organic Chemistry, University of Natural Resources and Life Sciences-Vienna, Muthgasse 18, A-1190, Vienna, Austria
| |
Collapse
|
34
|
Zhang Q, Su T, Pan Y, Wang X, Zhang C, Qin H, Li M, Li Q, Li X, Guo J, Wu L, Qin L, Liu T. Malus hupehensis leaves: a functional beverage for alleviating hepatic inflammation and modulating gut microbiota in diabetic mice. Food Funct 2025; 16:2972-2990. [PMID: 40126388 DOI: 10.1039/d4fo05325g] [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: 03/25/2025]
Abstract
Malus hupehensis leaves (MHL), consumed as a daily beverage in Chinese folk tradition and recently recognized as a new food material, are abundant in polyphenols and bioactive compounds that demonstrate hypoglycemic, lipid-lowering, and anti-inflammatory effects. However, the antidiabetic mechanisms have not been fully elucidated. This study aimed to investigate the protective mechanisms of Malus hupehensis leaves' extract (MHLE) against type 2 diabetes mellitus (T2DM). The results showed that MHLE effectively ameliorated glucose and lipid metabolic abnormalities in db/db mice, and attenuated hepatic macrophage activation. Transcriptomic analysis of the liver revealed that MHLE primarily affects genes involved in inflammatory responses and inhibited the TLR4/MAPK pathway to reduce hepatic inflammation. Metagenomic sequencing identified changes in gut microbiota composition and showed that MHLE restored the abundance of Lachnospiraceae bacterium, Oscillospiraceae bacterium, and Clostridia bacterium while reducing the abundance of Escherichia coli, thereby ameliorating gut dysbiosis. The integrated regulation of metabolism, immune response, and the microbial environment by MHLE significantly alleviated symptoms of T2DM. This study offers strong scientific evidence for the potential use of MHL as a functional food.
Collapse
Affiliation(s)
- Qiue Zhang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 102488, China
- Key Laboratory of Health Cultivation of the Ministry of Education, Beijing University of Chinese Medicine, Beijing, 102488, China.
| | - Tong Su
- Key Laboratory of Health Cultivation of the Ministry of Education, Beijing University of Chinese Medicine, Beijing, 102488, China.
- Dongfang Hospital of Beijing University of Chinese Medicine, Beijing, 100078, China
| | - Yajing Pan
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Xiaomeng Wang
- Key Laboratory of Health Cultivation of the Ministry of Education, Beijing University of Chinese Medicine, Beijing, 102488, China.
| | - Chengfei Zhang
- Dongfang Hospital of Beijing University of Chinese Medicine, Beijing, 100078, China
| | - Huizhao Qin
- Key Laboratory of Health Cultivation of the Ministry of Education, Beijing University of Chinese Medicine, Beijing, 102488, China.
- Dongfang Hospital of Beijing University of Chinese Medicine, Beijing, 100078, China
| | - Mingxiu Li
- Key Laboratory of Health Cultivation of the Ministry of Education, Beijing University of Chinese Medicine, Beijing, 102488, China.
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
| | - Qingsong Li
- Key Laboratory of Health Cultivation of the Ministry of Education, Beijing University of Chinese Medicine, Beijing, 102488, China.
- Shaanxi University of Chinese Medicine, Xianyang, 712046, China
| | - Xiaochen Li
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 102488, China
- Key Laboratory of Health Cultivation of the Ministry of Education, Beijing University of Chinese Medicine, Beijing, 102488, China.
| | - Jiangfan Guo
- Key Laboratory of Health Cultivation of the Ministry of Education, Beijing University of Chinese Medicine, Beijing, 102488, China.
- Shaanxi University of Chinese Medicine, Xianyang, 712046, China
| | - Lili Wu
- Key Laboratory of Health Cultivation of the Ministry of Education, Beijing University of Chinese Medicine, Beijing, 102488, China.
| | - Lingling Qin
- Key Laboratory of Health Cultivation of the Ministry of Education, Beijing University of Chinese Medicine, Beijing, 102488, China.
| | - Tonghua Liu
- Key Laboratory of Health Cultivation of the Ministry of Education, Beijing University of Chinese Medicine, Beijing, 102488, China.
| |
Collapse
|
35
|
Zhang J, Zhao L, He J, Wu H, Guo M, Yu Z, Ma X, Yong Y, Li Y, Ju X, Liu X. Protect Effects of Perilla Seed Extract and Its Active Ingredient Luteolin Against Inflammatory Bowel Disease Model via the PI3K/AKT Signal Pathway In Vivo and In Vitro. Int J Mol Sci 2025; 26:3564. [PMID: 40332054 PMCID: PMC12026851 DOI: 10.3390/ijms26083564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Revised: 03/19/2025] [Accepted: 04/06/2025] [Indexed: 05/08/2025] Open
Abstract
The purpose of this study was to investigate the anti-inflammatory effects of Perilla Seed Extract (PSE) and its active ingredient on Inflammatory Bowel Disease (IBD) in vitro and in vivo. Thirty-two C57/BL mice were randomly divided into four groups (n = 8): control group (CON), PBS group, LPS group (LPS 3.5 mg/kg given intraperitoneally [ip] on day 7 of the study only), and PSE group (100 mg/kg orally daily + LPS ip at 3.5 mg/kg on day 7). Mice were euthanized 24 h after LPS administration. MODE-K cells were divided into five groups: control group (CON), LPS group (50 μg/mL LPS for 2 h), and PSE group (low dose, 25 μg/mL PSE + LPS; middle dose, 50 μg/mL PSE + LPS; high dose, 100 μg/mL PSE + LPS). In vivo, compared with the CON group, LPS revealed a significant decrease in the villus length-to-crypt depth ratio (p < 0.01) and goblet cell density per unit area (p < 0.01). Conversely, PSE administration resulted in a significant increase in the villus length-to-crypt depth ratio (p < 0.01) and goblet cell density (p < 0.01). LPS significantly increased the ROS content (p < 0.01), the secretion of inflammatory cytokines of IL-6 (p < 0.01), TNF-α (p < 0.01), and the mRNA expressions of HO-1 (p < 0.01). LPS significantly decreased the mRNA expressions of Occludin (p < 0.01) and Claudin1 (p < 0.01). In contrast, PSE treatment led to a marked decrease in ROS levels (p < 0.01), along with a reduction in the secretion of inflammatory factors IL-6 (p < 0.01) and TNF-α(p < 0.05), as well as the mRNA expressions of HO-1 (p < 0.01). Concurrently, PSE significantly increased the mRNA expressions of Occludin (p < 0.05) and Claudin1 (p < 0.01). In vitro, PSE treatment also significantly reversed LPS-induced inflammation, oxidation and tight junction-related factors. Network pharmacology identified 97 potential targets for PSE in treating IBD, while transcriptomics analysis revealed 342 differentially expressed genes (DEGs). Network pharmacology and transcriptomics analysis indicated that significant pathways included the PI3K-Akt signaling pathway, MAPK signaling pathway, and TNF signaling pathway, of which the PI3K-AKT pathway may represent the primary mechanism. In an in vivo setting, compared with the CON group, LPS led to a significant increase in the protein expression of p-PI3K/PI3K (p < 0.01) and p-AKT1/AKT1 (p < 0.01). Conversely, PSE resulted in a significant decrease in the protein expression of p-PI3K/PI3K (p < 0.01) and p-AKT1/AKT1 (p < 0.01). In vitro, compared with the LPS group, PSE also significantly blocked the protein expression of p-PI3K/PI3K (p < 0.01) and p-AKT1/AKT1 (p < 0.01). The chemical composition of PSE was analyzed using UPLC-MS/MS, which identified six components including luteolin (content 0.41%), rosmarinic acid (content 0.27%), α-linolenic acid (content 1.2%), and oleic acid (content 0.2%). Molecular docking found that luteolin could establish stable binding with eight targets, and luteolin significantly decreased the p-AKT1/AKT1 ratio (p < 0.01) compared to the LPS group in MODE-K cells. In summary, PSE demonstrates efficacy against IBD progression by enhancing intestinal barrier function and inhibiting inflammatory responses and oxidative stress via the PI3K/AKT signaling pathway, and luteolin's inhibition of AKT1 protein phosphorylation appears to play a particularly crucial role in this therapeutic mechanism.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Xiaoxi Liu
- Department of Veterinary Medicine, College of Coastal Agricultural Sciences, Guangdong Ocean University, Zhanjiang 524091, China; (J.Z.)
| |
Collapse
|
36
|
Li S, Zhang Y, Ding S, Chang J, Liu G, Hu S. Curcumin Ameliorated Glucocorticoid-Induced Osteoporosis While Modulating the Gut Microbiota and Serum Metabolome. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:8254-8276. [PMID: 40139762 DOI: 10.1021/acs.jafc.4c06689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/29/2025]
Abstract
Glucocorticoid-induced osteoporosis (GIOP) is the leading cause of secondary osteoporosis. Recently, the "bone-gut axis" theory has linked bone development with gut microbial diversity, community composition, and metabolites. Curcumin, a well-studied polyphenol, shows potential in mitigating bone loss and osteoporosis. Alendronate, a standard therapeutic agent for osteoporosis, serves as a positive control in this investigation. The study demonstrates the potency of curcumin in reducing bone loss and restoring bone mineral density, enhancing trabecular parameters notably through increased trabecular number, volume, and thickness and reduced bone marrow cavity size. Gut microbiome sequencing revealed that both curcumin and alendronate treatments similarly enhanced gut microbial diversity and altered microbiota composition, increasing beneficial bacteria (Akkermansia_muciniphila, Dubosiella_sp910585105, and Ruminococcus_sp910584195) while reducing harmful bacteria (Treponema_D_sp910584475 and Duncaniella_sp910584825). Furthermore, significant changes in serum levels of metabolites including raffinose, ursolic acid, spermidine, inosine, hypoxanthine, thiamine, and pantothenic acid were observed post-treatment with curcumin or alendronate. Importantly, these beneficial metabolites and microorganisms were negatively correlated with inflammatory cytokines. In conclusion, curcumin holds promise for use against GIOP by modulating the gut microbiome and serum metabolome as well as reducing systemic inflammation.
Collapse
Affiliation(s)
- Siying Li
- The Orthopaedic Center, The First People' s Hospital of Wenling, Wenling Hospital of Wenzhou Medical University, Wenling 317500, Zhejiang Province, China
- College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha, Hunan 410128, China
| | - Yating Zhang
- College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha, Hunan 410128, China
| | - Sujuan Ding
- College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha, Hunan 410128, China
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang 330047, China
| | - Jiang Chang
- The Orthopaedic Center, The First People' s Hospital of Wenling, Wenling Hospital of Wenzhou Medical University, Wenling 317500, Zhejiang Province, China
| | - Gang Liu
- College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha, Hunan 410128, China
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang 330047, China
| | - Siwang Hu
- The Orthopaedic Center, The First People' s Hospital of Wenling, Wenling Hospital of Wenzhou Medical University, Wenling 317500, Zhejiang Province, China
| |
Collapse
|
37
|
Kim H, Lupoli TJ. Defined Glycan Ligands for Detecting Rare l-Sugar-Binding Proteins. J Am Chem Soc 2025; 147:11693-11699. [PMID: 40167164 PMCID: PMC11987014 DOI: 10.1021/jacs.5c03251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Revised: 03/17/2025] [Accepted: 03/20/2025] [Indexed: 04/02/2025]
Abstract
Most cells are decorated with distinct sugar sequences that can be recognized by carbohydrate-binding proteins (CBPs), such as antibodies and lectins. While humans utilize ten monosaccharide building blocks, bacteria biosynthesize hundreds of activated sugars to assemble diverse glycans. Monosaccharides absent in mammals are termed "rare" and are enriched in deoxy l-sugars beyond the "common" sugar l-fucose (l-Fuc) found across species. While immune proteins recognize microbial surfaces, there are limited probes to identify CBPs for the many rare sugars that may mediate these interactions. Here, we devise chemoenzymatic strategies to defined glycoconjugates containing l-Fuc and its structural analog l-colitose (l-Col), a bacterial dideoxysugar believed to bind immune proteins. We report a concise synthesis of l-Col and semisynthetic routes to several activated l-sugars. Incorporation of these sugars into glycans is evaluated using bacterial and mammalian glycosyltransferases (GTs) annotated to transfer l-Col or l-Fuc, respectively. We find that each GT can transfer both l-sugars, along with the rare hexose l-galactose (l-Gal), onto various glycan acceptors. Incorporation of these l-sugars into the resulting glycoconjugates is confirmed using known CBPs. Finally, these glycan ligands are employed to detect rare sugar-binding proteins in human serum. Overall, this work reveals similarities between bacterial and mammalian GTs that may be exploited for in vitro glycoconjugate construction to unveil novel mediators of host-pathogen interactions.
Collapse
Affiliation(s)
- Hanee Kim
- Department of Chemistry, New
York University, New York, New York 10003, United States
| | - Tania J. Lupoli
- Department of Chemistry, New
York University, New York, New York 10003, United States
| |
Collapse
|
38
|
Lee S, Kim H, Kim M, Kang R, Lim I, Jang Y. Rapid and simple on-site salmonella detection in food via direct sample loading using a lipopolysaccharide-imprinted polymer. J Nanobiotechnology 2025; 23:279. [PMID: 40189550 PMCID: PMC11974074 DOI: 10.1186/s12951-025-03341-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2024] [Accepted: 03/20/2025] [Indexed: 04/09/2025] Open
Abstract
Salmonella is a major foodborne pathogen that causes salmonellosis, which is characterized by symptoms such as diarrhea, fever, and abdominal cramps. Existing methods for detecting Salmonella, such as culture plating, ELISA, and PCR, are accurate but time-consuming and unsuitable for on-site applications. In this study, we developed a rapid and sensitive electrochemical sensor using a molecularly imprinted polymer (MIP) to detect Salmonella typhimurium (S. typhimurium) by targeting lipopolysaccharides (LPS). Polydopamine (PDA) was used as the polymer matrix because of its cost-efficiency and functional versatility. The sensor demonstrated high sensitivity and selectivity, with a detection limit of 10 CFU/mL and a linear response over the 10²-10⁸ CFU/mL range. The specificity of the sensor was validated against other gram-positive and gram-negative bacteria and showed no significant cross-reactivity. Furthermore, the sensor performed effectively in real food samples, including tap water, milk, and pork, without complex preprocessing. These results highlight the potential of the LPS-imprinted MIP sensor for practical on-site detection of S. typhimurium, improving food safety monitoring and preventing outbreaks in food-handling environments.
Collapse
Affiliation(s)
- Solpa Lee
- Department of Medical and Digital Engineering, College of Engineering, Hanyang University, Seoul, 04763, Korea
| | - Hyunsoo Kim
- DRB Research, DRB Industrial, 28, Gongdandong-ro 55beon-gil, Busan, 46329, Republic of Korea
| | - Minwoo Kim
- Department of Medical and Digital Engineering, College of Engineering, Hanyang University, Seoul, 04763, Korea
| | - Ryun Kang
- Department of Medical and Digital Engineering, College of Engineering, Hanyang University, Seoul, 04763, Korea
| | - Inje Lim
- Department of Medical and Digital Engineering, College of Engineering, Hanyang University, Seoul, 04763, Korea
| | - Yongwoo Jang
- Department of Medical and Digital Engineering, College of Engineering, Hanyang University, Seoul, 04763, Korea.
- Department of Pharmacology, College of Medicine, Hanyang University, Seoul, 04736, Korea.
| |
Collapse
|
39
|
Luo X, Zhang X, Wen X, Wang R, Zhang Q, Luo P, Yu F, Cao H. Valence State Hydrogen Channel Enhances Sustained and Controllable Electrocatalytic Hydrogen Evolution in Diabetic Skin Wound Healing. Angew Chem Int Ed Engl 2025; 64:e202422091. [PMID: 39846755 DOI: 10.1002/anie.202422091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 01/22/2025] [Accepted: 01/23/2025] [Indexed: 01/24/2025]
Abstract
Diabetes significantly increases the risk of serious health issues, including prolonged skin inflammation and delayed wound healing, owing to inferior glucose control and suppression of the immune system. Although traditional hydrogen (H2) therapy is slightly effective, its ability to tailor the release of H2 on the skin is limited. Accordingly, this study proposed a novel strategy for electrocatalytic H2 release under neutral conditions to promote wound healing in diabetic mice and rabbit. Herein, a defect-engineered cobalt phosphide (CoP) catalyst was designed by introducing a neutral single-metal electrocatalytic Hydrogen valence state channel into CoP. By effectively regulating the formation and transfer of *H active species during the CoP catalytic process, a considerable enhancement in neutral electrocatalytic H2 evolution performance was achieved (-78.0 mV@-10.0 mA cm-2). Based on this superior catalytic performance, we developed a flexible electrode (namely, CoP/flexible gold electrode made by screen printing (FGSP) by combining a convenient electrolysis platform with continuous electrolyte supply and FGSP, enabling customized H2 release and accelerating wound healing in diabetic mice and rabbits. Notably, the designed flexible electrode features adjustable dimensions, interchangeable substrates, and material adaptability, meeting the diverse needs of clinical and basic research and demonstrating significant potential for applications in clinical medicine.
Collapse
Affiliation(s)
- Xianzhu Luo
- Key Laboratory of Emergency and Trauma of Ministry of Education, The First Affiliated Hospital of Hainan Medical University, Key Laboratory of Hainan Trauma and Disaster Rescue, Key Laboratory of Haikou Trauma, Engineering Research Center for Hainan Bio-Smart Materials and Bio-Medical Devices, School of Pharmacy, College of Emergency and Trauma, Hainan Medical University, Haikou, 571199, China
| | - Xiangcheng Zhang
- Key Laboratory of Advanced Technologies of Materials (Ministry of Education), School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China
| | - Xue Wen
- School of Chemistry and Chemical Engineering, School of Electronics, Information and Electrical Engineering, Instrumental Analysis Center, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Rui Wang
- Key Laboratory of Emergency and Trauma of Ministry of Education, The First Affiliated Hospital of Hainan Medical University, Key Laboratory of Hainan Trauma and Disaster Rescue, Key Laboratory of Haikou Trauma, Engineering Research Center for Hainan Bio-Smart Materials and Bio-Medical Devices, School of Pharmacy, College of Emergency and Trauma, Hainan Medical University, Haikou, 571199, China
- Department of Chemistry and Nanoscience, Ewha Womans University, Seoul, 03706, Republic of Korea
| | - Qingguo Zhang
- Department of General Plastic Surgery, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100006, China
| | - Pan Luo
- Department of General Plastic Surgery, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100006, China
| | - Fabiao Yu
- Key Laboratory of Emergency and Trauma of Ministry of Education, The First Affiliated Hospital of Hainan Medical University, Key Laboratory of Hainan Trauma and Disaster Rescue, Key Laboratory of Haikou Trauma, Engineering Research Center for Hainan Bio-Smart Materials and Bio-Medical Devices, School of Pharmacy, College of Emergency and Trauma, Hainan Medical University, Haikou, 571199, China
| | - Hongshuai Cao
- Key Laboratory of Emergency and Trauma of Ministry of Education, The First Affiliated Hospital of Hainan Medical University, Key Laboratory of Hainan Trauma and Disaster Rescue, Key Laboratory of Haikou Trauma, Engineering Research Center for Hainan Bio-Smart Materials and Bio-Medical Devices, School of Pharmacy, College of Emergency and Trauma, Hainan Medical University, Haikou, 571199, China
- School of Chemistry and Chemical Engineering, School of Electronics, Information and Electrical Engineering, Instrumental Analysis Center, Shanghai Jiao Tong University, Shanghai, 200240, China
| |
Collapse
|
40
|
Sim M, Hong S, Jung MH, Choi EY, Hwang GS, Shin DM, Kim CS. Gut microbiota links vitamin C supplementation to enhanced mental vitality in healthy young adults with suboptimal vitamin C status: A randomized, double-blind, placebo-controlled trial. Brain Behav Immun 2025; 128:179-191. [PMID: 40187667 DOI: 10.1016/j.bbi.2025.03.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 03/07/2025] [Accepted: 03/25/2025] [Indexed: 04/07/2025] Open
Abstract
The intricate relationship between nutrition, gut microbiome, and mental health has gained increasing attention. We aimed to determine how vitamin C supplementation improves mental vitality through the gut microbiome and associated neurological and immunological changes. We used 16S rRNA sequencing to analyze gut microbiota profiles of participants from our previous trial, in which healthy young adults (20-39 years) with inadequate serum vitamin C levels (< 50 μM) received 500 mg vitamin C or a placebo twice daily for 4 weeks (vitamin C, n = 21; placebo, n = 19). We examined whether changes in gut microbiota correlated with previously determined mental vitality indices, including Stroop test performance, work engagement, and serum brain-derived neurotrophic factor (BDNF) levels. Serum concentrations of microbial-derived molecules, cytokines, and neurotransmitters were analyzed using enzyme-linked immunosorbent assay, electrochemiluminescence-based immunoassay, or ultra-high-performance liquid chromatography-mass spectrometry. Monocyte subpopulations in peripheral blood were quantified using fluorescence-activated cell sorting analysis. Vitamin C supplementation increased the relative abundance of Bacillaceae and Anaerotruncus, while decreasing Desulfovibrio, with the Desulfovibrio reduction correlating with Stroop test performance. Moreover, participants showing a substantial Desulfovibrio reduction ("responders") demonstrated greater BDNF increases and stronger correlations between serum L-DOPA levels and work engagement scores than did non-responders. In addition, vitamin C supplementation suppressed inflammatory responses with concurrent reduction in serum lipopolysaccharide levels, and responders showed greater decreases in IL-10 levels and classical monocyte frequencies than non-responders. In conclusion, vitamin C supplementation modulates gut microbiota composition, particularly by reducing Desulfovibrio abundance, with the extent of reduction correlating with mental vitality improvements and decreased inflammation. This study provides insights into vitamin C supplementation as a critical dietary intervention, as it may modulate mental health through its influence on the gut-brain-immune axis.
Collapse
Affiliation(s)
- Minju Sim
- Department of Food and Nutrition, Seoul National University, Seoul 08826, Republic of Korea
| | - Sehwa Hong
- Department of Food and Nutrition, Seoul National University, Seoul 08826, Republic of Korea
| | - Min Ho Jung
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Eun Young Choi
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Geum-Sook Hwang
- Integrated Metabolomics Research Group, Western Seoul Center, Korea Basic Science Institute, Seoul 03759, Republic of Korea; College of Pharmacy, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Dong-Mi Shin
- Department of Food and Nutrition, Seoul National University, Seoul 08826, Republic of Korea
| | - Chong-Su Kim
- Department of Food and Nutrition, Seowon University, Cheongju 28674, Republic of Korea.
| |
Collapse
|
41
|
Lee S, Wischmeyer PE, Mintz CD, Serbanescu MA. Recent Insights into the Evolving Role of the Gut Microbiome in Critical Care. Crit Care Clin 2025; 41:379-396. [PMID: 40021286 DOI: 10.1016/j.ccc.2024.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/03/2025]
Abstract
This review explores the evolving understanding of gut microbiota's role in critical illness, focusing on how acute illness and exposures in intensive care unit (ICU) environment negatively impact the gut microbiota and the implications of these changes on host responses in critically-ill patients. Focusing on recent findings from clinical and preclinical studies, we discuss the effects of inflammation, enteral nutrient deprivation, and antibiotics on gut microbial dynamics. This review aims to enhance comprehension of microbial dynamics in the ICU and their implications for clinical outcomes and therapeutic strategies.
Collapse
Affiliation(s)
- Seoho Lee
- Department of Anesthesiology and Critical Care, Johns Hopkins University School of Medicine, Phipps 455 1800 Orleans Street, Baltimore, MD 21212, USA
| | - Paul E Wischmeyer
- Department of Anesthesiology, Duke University School of Medicine, 5692 HAFS Box 3094, 2301 Erwin Road, Durham, NC 27710, USA
| | - Cyrus D Mintz
- Department of Anesthesiology and Critical Care, Johns Hopkins University School of Medicine, Phipps 455 1800 Orleans Street, Baltimore, MD 21212, USA
| | - Mara A Serbanescu
- Department of Anesthesiology, Duke University School of Medicine, 5692 HAFS Box 3094, 2301 Erwin Road, Durham, NC 27710, USA.
| |
Collapse
|
42
|
Ameho S, Klutstein M. The effect of chronic inflammation on female fertility. Reproduction 2025; 169:e240197. [PMID: 39932461 PMCID: PMC11896653 DOI: 10.1530/rep-24-0197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 02/02/2025] [Accepted: 02/11/2025] [Indexed: 02/13/2025]
Abstract
In brief Chronic inflammation causes serious medical conditions in many organs and tissues, including female fertility. Here we review the current literature, showing that chronic inflammation has a negative impact on oocyte quality, folliculogenesis, hormone production, immune signaling and other processes that affect fertility in females. Abstract Inflammation has key biological roles in the battle against pathogens and additional key processes in development and tissue homeostasis. However, when inflammation becomes chronic, it can become a serious medical concern. Chronic inflammation has been shown to contribute to the etiology and symptoms of serious medical conditions such as ulcerative colitis, cardiovascular diseases, endometriosis and various cancers. One of the less recognized symptoms associated with chronic inflammation is its effect on reproduction, specifically on female fertility. Here we review the current literature, showing that chronic inflammation has a negative impact on oocyte quality, folliculogenesis, hormone production, immune signaling and other processes that affect fertility in females. We discuss several factors involved in the etiology of chronic inflammation and its effect on female fertility. We also discuss possible mechanisms by which these effects may be mediated and how interventions may mitigate the effect of chronic inflammation. Finally, we discuss the notion that in many cases, the effect of chronic inflammation is tightly correlated with and resembles the effect of aging, drawing interesting parallels between these processes, possibly through the effect of aging-associated inflammaging.
Collapse
Affiliation(s)
| | - Michael Klutstein
- Institute of Biomedical and Oral Research, Faculty of Dental Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| |
Collapse
|
43
|
Wang Y, Xu X, Zhang P, Hu S, Zhang L, Chen H. E3 Ubiquitin Ligase TRIM7 Alleviates Lipopolysaccharide-Induced Acute Lung Injury via Inhibiting NLRP3 Inflammasome Activation. THE AMERICAN JOURNAL OF PATHOLOGY 2025; 195:639-651. [PMID: 39864619 DOI: 10.1016/j.ajpath.2024.12.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 11/25/2024] [Accepted: 12/27/2024] [Indexed: 01/28/2025]
Abstract
Acute lung injury (ALI) is a common clinical disease with high mortality, characterized by tissue damage caused by excessive activation of inflammation. TRIM7 is an E3 ligase that plays an important role in regulating viral infection, tumor progression, and innate immune response. However, its function in ALI is unclear. In this study, lipopolysaccharide (LPS) was used to stimulate C57BL/6j mice and HULEC-5a cells to establish ALI models in vivo and in vitro. TRIM7 expression was down-regulated during ALI. Furthermore, overexpressing TRIM7 in HULEC-5a cells relieved cell damage and inflammatory activation induced by LPS stimulation. TRIM7 knockdown had the opposite effect. Trim7-overexpressing mice were established by endotracheal injection of adeno-associated virus 6-Trim7 virus in vivo; the ALI model was then induced by LPS stimulation. Overexpression of TRIM7 could alleviate lung tissue injury, pulmonary interstitial hemorrhage, increased alveolar and vascular permeability, inflammatory cell infiltration, and secretion of inflammatory factors induced by LPS stimulation. Mechanistically, TRIM7 inhibited the expression of NOD-, LRR- and pyrin domain-containing 3 (NLRP3) and the activation of the NLRP3 inflammasome. The regulatory effect of TRIM7 on ALI depended on the NLRP3 inflammasome. This investigation, for the first time, showed the inhibitory effect of TRIM7 on ALI and activation of the NLRP3 inflammasome, providing new targets and ideas for the research on the mechanism and treatment of ALI.
Collapse
Affiliation(s)
- Youna Wang
- Department of Pulmonary and Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xiaohong Xu
- Department of Pulmonary and Critical Care Medicine, Hanchuan People's Hospital, Hanchuan, China
| | - Peng Zhang
- Taikang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, China
| | - Sha Hu
- Taikang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, China
| | - Li Zhang
- Center for Animal Experiment, Wuhan University, Wuhan, China.
| | - Hongbin Chen
- Department of Pulmonary and Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, China.
| |
Collapse
|
44
|
Kulkarni R, Kumari S, Dhapola R, Sharma P, Singh SK, Medhi B, HariKrishnaReddy D. Association Between the Gut Microbiota and Alzheimer's Disease: An Update on Signaling Pathways and Translational Therapeutics. Mol Neurobiol 2025; 62:4499-4519. [PMID: 39460901 DOI: 10.1007/s12035-024-04545-2] [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: 05/06/2024] [Accepted: 10/10/2024] [Indexed: 10/28/2024]
Abstract
Alzheimer's disease (AD) is a cognitive disease with high morbidity and mortality. In AD patients, the diversity of the gut microbiota is altered, which influences pathology through the gut-brain axis. Probiotic therapy alleviates pathological and psychological consequences by restoring the diversity of the gut microbial flora. This study addresses the role of altered gut microbiota in the progression of neuroinflammation, which is a major hallmark of AD. This process begins with the activation of glial cells, leading to the release of proinflammatory cytokines and the modulation of cholinergic anti-inflammatory pathways. Short-chain fatty acids, which are bacterial metabolites, provide neuroprotective effects and maintain blood‒brain barrier integrity. Furthermore, the gut microbiota stimulates oxidative stress and mitochondrial dysfunction, which promote AD progression. The signaling pathways involved in gut dysbiosis-mediated neuroinflammation-mediated promotion of AD include cGAS-STING, C/EBPβ/AEP, RAGE, TLR4 Myd88, and the NLRP3 inflammasome. Preclinical studies have shown that natural extracts such as Ganmaidazao extract, isoorentin, camelia oil, Sparassis crispa-1, and xanthocerasides improve gut health and can delay the worsening of AD. Clinical studies using probiotics such as Bifidobacterium spp., yeast beta-glucan, and drugs such as sodium oligomannate and rifaximine have shown improvements in gut health, resulting in the amelioration of AD symptoms. This study incorporates the most current research on the pathophysiology of AD involving the gut microbiota and highlights the knowledge gaps that need to be filled to develop potent therapeutics against AD.
Collapse
Affiliation(s)
- Rutweek Kulkarni
- Advanced Pharmacology and Neuroscience Laboratory, Department of Pharmacology, School of Health Sciences, Central University of Punjab, Bathinda, 151401, Punjab, India
| | - Sneha Kumari
- Advanced Pharmacology and Neuroscience Laboratory, Department of Pharmacology, School of Health Sciences, Central University of Punjab, Bathinda, 151401, Punjab, India
| | - Rishika Dhapola
- Advanced Pharmacology and Neuroscience Laboratory, Department of Pharmacology, School of Health Sciences, Central University of Punjab, Bathinda, 151401, Punjab, India
| | - Prajjwal Sharma
- Advanced Pharmacology and Neuroscience Laboratory, Department of Pharmacology, School of Health Sciences, Central University of Punjab, Bathinda, 151401, Punjab, India
| | - Sunil K Singh
- Department of Biochemistry, School of Basic Sciences, Central University of Punjab, Ghudda, Bathinda, Punjab, India
| | - Bikash Medhi
- Department of Pharmacology, Post Graduate Institute of Medical Education and Research, Chandigarh, 160012, India
| | - Dibbanti HariKrishnaReddy
- Advanced Pharmacology and Neuroscience Laboratory, Department of Pharmacology, School of Health Sciences, Central University of Punjab, Bathinda, 151401, Punjab, India.
| |
Collapse
|
45
|
Lesbats J, Brillac A, Reisz JA, Mukherjee P, Lhuissier C, Fernández-Monreal M, Dupuy JW, Sequeira A, Tioli G, De La Calle Arregui C, Pinson B, Wendisch D, Rousseau B, Efeyan A, Sander LE, D'Alessandro A, Garaude J. Macrophages recycle phagocytosed bacteria to fuel immunometabolic responses. Nature 2025; 640:524-533. [PMID: 40011782 DOI: 10.1038/s41586-025-08629-4] [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: 01/22/2024] [Accepted: 01/10/2025] [Indexed: 02/28/2025]
Abstract
Macrophages specialize in phagocytosis, a cellular process that eliminates extracellular matter, including microorganisms, through internalization and degradation1,2. Despite the critical role of phagocytosis during bacterial infection, the fate of phagocytosed microbial cargo and its impact on the host cell are poorly understood. In this study, we show that ingested bacteria constitute an alternative nutrient source that skews immunometabolic host responses. By tracing stable isotope-labelled bacteria, we found that phagolysosomal degradation of bacteria provides carbon atoms and amino acids that are recycled into various metabolic pathways, including glutathione and itaconate biosynthesis, and satisfies the bioenergetic needs of macrophages. Metabolic recycling of microbially derived nutrients is regulated by the nutrient-sensing mechanistic target of rapamycin complex C1 and is intricately tied to microbial viability. Dead bacteria, as opposed to live bacteria, are enriched in cyclic adenosine monophosphate, sustain the cellular adenosine monophosphate pool and subsequently activate adenosine monophosphate protein kinase to inhibit the mechanistic target of rapamycin complex C1. Consequently, killed bacteria strongly fuel metabolic recycling and support macrophage survival but elicit decreased reactive oxygen species production and reduced interleukin-1β secretion compared to viable bacteria. These results provide a new insight into the fate of engulfed microorganisms and highlight a microbial viability-associated metabolite that triggers host metabolic and immune responses. Our findings hold promise for shaping immunometabolic intervention for various immune-related pathologies.
Collapse
Affiliation(s)
| | - Aurélia Brillac
- University of Bordeaux, INSERM, MRGM, U1211, Bordeaux, France
| | - Julie A Reisz
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Parnika Mukherjee
- Department of Infectious Diseases, Respiratory Medicine, and Critical Care, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Charlène Lhuissier
- ImmunoConcEpT, CNRS UMR 5164, INSERM ERL 1303, University of Bordeaux, Bordeaux, France
| | | | - Jean-William Dupuy
- University of Bordeaux, CNRS, INSERM, TBM-Core, US5, UAR3421, OncoProt, Bordeaux, France
- University of Bordeaux, Bordeaux Protéome, Bordeaux, France
| | - Angèle Sequeira
- ImmunoConcEpT, CNRS UMR 5164, INSERM ERL 1303, University of Bordeaux, Bordeaux, France
| | - Gaia Tioli
- University of Bordeaux, INSERM, MRGM, U1211, Bordeaux, France
- Biomedical and Neuromotor Sciences, Alma Mater University of Bologna, Bologna, Italy
| | - Celia De La Calle Arregui
- Metabolism and Cell Signalling Laboratory, Spanish National Cancer Research Center (CNIO), Madrid, Spain
| | - Benoît Pinson
- Service Analyses Métabolomiques, TBMCore, CNRS UAR 3427, INSERM US005, Université Bordeaux, Bordeaux, France
| | - Daniel Wendisch
- Department of Infectious Diseases, Respiratory Medicine, and Critical Care, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Benoît Rousseau
- University of Bordeaux, Animal Facility A2, Service Commun des Animaleries, Bordeaux, France
| | - Alejo Efeyan
- Metabolism and Cell Signalling Laboratory, Spanish National Cancer Research Center (CNIO), Madrid, Spain
| | - Leif Erik Sander
- Department of Infectious Diseases, Respiratory Medicine, and Critical Care, Charité-Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
| | - Angelo D'Alessandro
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Johan Garaude
- University of Bordeaux, INSERM, MRGM, U1211, Bordeaux, France.
- ImmunoConcEpT, CNRS UMR 5164, INSERM ERL 1303, University of Bordeaux, Bordeaux, France.
| |
Collapse
|
46
|
Song W, Chen J, Ai G, Xiong P, Song Q, Wei Q, Zou Z, Chen X. Mechanisms of the effects of turpiniae folium extract on growth performance, immunity, antioxidant activity and intestinal barrier function in LPS-challenged broilers. Poult Sci 2025; 104:104903. [PMID: 39985896 PMCID: PMC11904579 DOI: 10.1016/j.psj.2025.104903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Revised: 01/24/2025] [Accepted: 02/16/2025] [Indexed: 02/24/2025] Open
Abstract
Turpiniae folium extract (TFE) has shown anti-inflammatory and immunomodulatory effects in broilers. However, its mechanisms remain unclear. The aim of this study is to investigate the underlying mechanisms by which TFE influences growth performance, jejunal morphology, immune function, antioxidant capacity and barrier integrity in broilers challenged with Lipopolysaccharide (LPS). A total of 240 one-day-old female broilers were randomly divided into four groups with six replicates of ten birds each. A 2 × 2 factorial design with TFE (basal diets supplemented with 0 or 500 mg/kg TFE) and LPS challenge (intraperitoneal injection of 1 mg/kg body weight of sterile saline or LPS at 21, 23 and 25 days of age). The trial lasted for 26 days. The results showed that: Prior to the LPS challenge, dietary supplementation with TFE for 21 days increased both average daily gain (ADG) (P = 0.037) and average daily feed intake (ADFI) (P = 0.045) in broilers. During the LPS challenge period, LPS challenge led to a decline in growth performance and a negative impact on intestinal morphology, while TFE supplementation significantly reversed these adverse effects, as evidenced by increases in ADG (P = 0.004), ADFI (P = 0.046), jejunal villus height (VH) (P = 0.035), the villus height to crypt depth ratio (VH/CD) (P = 0.007) and decreases in the feed-to-gain ratio (F/G) (P = 0.025), jejunal crypt depth (CD) (P = 0.049). LPS induced inflammatory responses and oxidative stress in the jejunum, leading to a significant upregulation of pro-inflammatory factor gene and protein expression, and a marked downregulation of anti-inflammatory and antioxidant gene and protein expression. TFE supplementation mitigated these effects by yielding completely opposite results except for the expression of toll-like receptor 4 (TLR4) protein (P = 0.916). LPS negatively regulates the expression of genes and proteins involved in intestinal mucosal barrier function. In contrast, TFE supplementation significantly upregulated the expression of zonula occludens-1 (ZO-1) (P < 0.001) gene and ZO-1 (P < 0.001), occludin (OCLN) (P < 0.001), claudin (CLDN) (P < 0.001) proteins. In conclusion, dietary supplementation with TFE effectively counteracts the intestinal immune and oxidative stress induced by LPS challenge in broilers, improves intestinal mucosal barrier integrity and tissue morphology, and ultimately mitigates the negative impact of LPS on broiler growth performance. This effect may involve the modulation of the Nrf2 and nuclear factor kappa B (NF-κB) signaling pathways.
Collapse
Affiliation(s)
- Wenjing Song
- Institute of Animal Husbandry and Veterinary Science, Jiangxi Academy of Agricultural Sciences, Nanchang 330200, PR China; Jiangxi Province Key Laboratory of Animal Green and Healthy Breeding, Nanchang 330200, PR China
| | - Jiang Chen
- Institute of Animal Husbandry and Veterinary Science, Jiangxi Academy of Agricultural Sciences, Nanchang 330200, PR China; Jiangxi Province Key Laboratory of Animal Green and Healthy Breeding, Nanchang 330200, PR China
| | - Gaoxiang Ai
- Institute of Animal Husbandry and Veterinary Science, Jiangxi Academy of Agricultural Sciences, Nanchang 330200, PR China; Jiangxi Province Key Laboratory of Animal Green and Healthy Breeding, Nanchang 330200, PR China
| | - Pingwen Xiong
- Institute of Animal Husbandry and Veterinary Science, Jiangxi Academy of Agricultural Sciences, Nanchang 330200, PR China; Jiangxi Province Key Laboratory of Animal Green and Healthy Breeding, Nanchang 330200, PR China
| | - Qiongli Song
- Institute of Animal Husbandry and Veterinary Science, Jiangxi Academy of Agricultural Sciences, Nanchang 330200, PR China; Jiangxi Province Key Laboratory of Animal Green and Healthy Breeding, Nanchang 330200, PR China
| | - Qipeng Wei
- Institute of Animal Husbandry and Veterinary Science, Jiangxi Academy of Agricultural Sciences, Nanchang 330200, PR China; Jiangxi Province Key Laboratory of Animal Green and Healthy Breeding, Nanchang 330200, PR China
| | - Zhiheng Zou
- Institute of Animal Husbandry and Veterinary Science, Jiangxi Academy of Agricultural Sciences, Nanchang 330200, PR China; Jiangxi Province Key Laboratory of Animal Green and Healthy Breeding, Nanchang 330200, PR China
| | - Xiaolian Chen
- Institute of Animal Husbandry and Veterinary Science, Jiangxi Academy of Agricultural Sciences, Nanchang 330200, PR China; Jiangxi Province Key Laboratory of Animal Green and Healthy Breeding, Nanchang 330200, PR China.
| |
Collapse
|
47
|
Xu X, Yan X, Jin X, Li J, Hu Q, Ahn DH, Zhong C. Expression and characterization of colanic acid-degrading enzyme from Escherichia phage phi92 and analysis of its hydrolysate composition and structure. Int J Biol Macromol 2025; 303:140646. [PMID: 39909247 DOI: 10.1016/j.ijbiomac.2025.140646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Revised: 01/13/2025] [Accepted: 02/02/2025] [Indexed: 02/07/2025]
Abstract
Colanic acid (CA), an exopolysaccharide synthesized by bacteria, is known for its potential to extend the lifespan of nematodes. However, its high viscosity poses practical application challenges, which can be mitigated by employing colanic acid-degrading enzymes (CAEs). This study presents the expression and characterization of a novel CAE from Escherichia phage phi92, used for the production of CA oligosaccharides (CAOSs). Optimal CAE expression conditions were identified as induction at 20 °C for 24 h with 0.1 mM IPTG. The enzyme showed maximal activity at 55 °C and pH 6.0, with stability in the range of 4-50 °C and pH 4.0-7.0. Structural characterization of CAOSs was performed using GC-MS, LC-MS and NMR spectroscopy, revealing the composition of hexasaccharide and dodecasaccharide units, with CAE cleaving the β-1,4 glycosidic linkage between glucose and fucose. CAOSs were found to mitigate oxidative stress and inflammation induced by H2O2 in RAW 264.7 macrophages, inhibiting NO and MDA production while enhancing CAT activity. Additionally, CAOSs modulated the mRNA expression of pro-inflammatory (IL-1β, COX-2) and anti-inflammatory (IL-10, HO-1) factors. This study deepens the understanding of CAE and facilitates the preparation of CA oligomers for applications in food, cosmetics, and pharmaceuticals.
Collapse
Affiliation(s)
- Xiaotong Xu
- Department of Food Science and Technology, Pukyong National University, Busan, Republic of Korea; Key Laboratory of Quantitative Synthetic Biology, Center for Materials Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Xiaoshuang Yan
- Key Laboratory of Quantitative Synthetic Biology, Center for Materials Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | | | - Juyi Li
- PAM(2)L Biotechnologies, Shenzhen, China
| | - Qiaoxia Hu
- PAM(2)L Biotechnologies, Shenzhen, China
| | - Dong-Hyun Ahn
- Department of Food Science and Technology, Pukyong National University, Busan, Republic of Korea.
| | - Chao Zhong
- Key Laboratory of Quantitative Synthetic Biology, Center for Materials Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China; PAM(2)L Biotechnologies, Shenzhen, China.
| |
Collapse
|
48
|
Alamri MA, Prinsa, Kawsar SMA, Saha S. Exploring marine-derived bioactive compounds for dual inhibition of Pseudomonas aeruginosa LpxA and LpxD: integrated bioinformatics and cheminformatics approaches. Mol Divers 2025; 29:1033-1047. [PMID: 38780832 DOI: 10.1007/s11030-024-10888-8] [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: 03/28/2024] [Accepted: 04/27/2024] [Indexed: 05/25/2024]
Abstract
Pseudomonas aeruginosa can cause serious nosocomial infections. Targeting the biosynthesis of Lipid A, a major structural domain of lipopolysaccharide (LPS) in P. aeruginosa has emerged as a valuable strategy for developing novel therapeutic agents. The biosynthesis of Lipid A involves the activation of homolog enzymes including LpxA and LpxD. LpxA enzyme facilitates the transfer of R-3-hydroxydecanoic fatty acid to uridine diphosphate N-acetylglucosamine in the first step. While LPxD is accountable in third step, wherein R-3-hydroxydodecanoate is transferred to the 2' amine of UDP-3-O-(3-hydroxydecanoyl) utilizing an ACP donor. The exploration of LpxA and LpxD has been largely neglected, as no specific small-molecule inhibitors have been identified, thus far, except for peptide inhibitors. Here, we report the identification of potential dual inhibitors of the lipid A biosynthesis pathway that target both the LpxA and LpxD enzymes as novel antibiotic agents. Among the virtually screened 32,000 marine bioactive compounds Oscillatoxin A, NCI60_041046, and LTS0192263 exhibited optimal docking interactions with LpxA and LpxD, respectively. MD simulation and MMPBSA data showcased stable interactions between selected marine products and LpxA/LpxD. FMO analysis showed that Oscillatoxin A and NCI60_041046 are the most chemically active molecules. MEP analysis data highlighted the possible electrophilic and nucleophilic distribution zones present in the structure. In addition, these bioactive molecules showed acceptable ADMET profiles. These data confirmed that Oscillatoxin A, NCI60_041046, and LTS0192263 could serve as seeds for the development of potential therapeutics to combat P. aeruginosa infection.
Collapse
Affiliation(s)
- Mubarak A Alamri
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam Bin Abdulaziz University, P.O. Box 173, 11942, Al-Kharj, Saudi Arabia
| | - Prinsa
- Siddhartha Institute of Pharmacy, Near IT-Park, Sahastradhara Road, Dehradun, 248001, Uttarakhand, India
| | - Sarkar M A Kawsar
- Laboratory of Carbohydrate and Nucleoside Chemistry, Department of Chemistry, Faculty of Science, University of Chittagong, Chittagong, 4331, Bangladesh
| | - Supriyo Saha
- Department of Pharmaceutical Chemistry, Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, 248001, Uttarakhand, India.
| |
Collapse
|
49
|
Yang Y, Hyeon H, Joo M, Lee K, Shin E. Small regulatory RNAs as key modulators of antibiotic resistance in pathogenic bacteria. J Microbiol 2025; 63:e2501027. [PMID: 40313153 DOI: 10.71150/jm.2501027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Accepted: 02/18/2025] [Indexed: 05/03/2025]
Abstract
The escalating antibiotic resistance crisis poses a significant challenge to global public health, threatening the efficacy of current treatments and driving the emergence of multidrug-resistant pathogens. Among the various factors associated with bacterial antibiotic resistance, small regulatory RNAs (sRNAs) have emerged as pivotal post-transcriptional regulators which orchestrate bacterial adaptation to antibiotic pressure via diverse mechanisms. This review consolidates the current knowledge on sRNA-mediated mechanisms, focusing on drug uptake, drug efflux systems, lipopolysaccharides, cell wall modification, biofilm formation, and mutagenesis. Recent advances in transcriptomics and functional analyses have revealed novel sRNAs and their regulatory networks, expanding our understanding of resistance mechanisms. These findings highlight the potential of targeting sRNA-mediated pathways as an innovative therapeutic strategy to combat antibiotic resistance, and offer promising avenues for managing challenging bacterial infections.
Collapse
Affiliation(s)
- Yubin Yang
- Department of Life Science, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Hana Hyeon
- Department of Life Science, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Minju Joo
- Department of Life Science, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Kangseok Lee
- Department of Life Science, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Eunkyoung Shin
- Department of Microbiology, Catholic University of Daegu School of Medicine, Daegu 42472, Republic of Korea
| |
Collapse
|
50
|
Cirella R, Andretta E, De Simone Carone L, Olmeo F, Sun ML, Zhang YZ, Mercogliano M, Molinaro A, Silipo A, Di Lorenzo F. Cold-Adapted Lipid A from Polaribacter sp. SM1127: A Study of Structural Heterogeneity and Immunostimulatory Properties. Chembiochem 2025:e2500100. [PMID: 40163369 DOI: 10.1002/cbic.202500100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 03/27/2025] [Accepted: 03/31/2025] [Indexed: 04/02/2025]
Abstract
Polaribacter sp. SM1127, a cold-adapted marine Gram-negative bacterium isolated from Laminaria in Arctic waters, plays a crucial role in nutrient cycling and biopolymer degradation in cold environments. Additionally, its exopolysaccharide (EPS) exhibits promising biotechnological potential, including antioxidant and wound-healing properties. This study focuses on the isolation and characterization of lipid A, the glycolipid component of Polaribacter sp. SM1127 lipopolysaccharide (LPS), by bypassing full LPS extraction and working directly with the ethanol precipitation product containing both EPS and bacterial cells. Mass spectrometry analysis reveals significant structural heterogeneity in the lipid A, with variations in fatty acid chain length, branching, saturation, and hydroxylation. These features likely enable the bacterium to fine-tune its response to fluctuating temperatures or other cold-related environmental stresses, contributing to resilience in the Arctic Ocean ecosystem. Furthermore, immunological assays demonstrate that both LPS and EPS produced by Polaribacter sp. SM1127 induce weak Toll-like receptor 4 activation and, in general, poorly stimulate the nuclear factor kappa-light-chain-enhancer of activated B cells pathway, compared to Escherichia coli LPS. These findings suggest their potential as immunomodulatory agents, like vaccine adjuvants.
Collapse
Affiliation(s)
- Roberta Cirella
- Department of Chemical Sciences, University of Naples Federico II, Via Cinthia 4, 80126, Naples, Italy
| | - Emanuela Andretta
- Department of Chemical Sciences, University of Naples Federico II, Via Cinthia 4, 80126, Naples, Italy
| | - Luca De Simone Carone
- Department of Chemical Sciences, University of Naples Federico II, Via Cinthia 4, 80126, Naples, Italy
| | - Francesca Olmeo
- Department of Chemical Sciences, University of Naples Federico II, Via Cinthia 4, 80126, Naples, Italy
| | - Mei-Ling Sun
- MOE Key Laboratory of Evolution and Marine Biodiversity, Frontiers Science Center for Deep Ocean Multispheres and Earth System & College of Marine Life Sciences, Ocean University of China, 5 Yushan Rd, Shinan District, Shandong, Qingdao, 266005, China
- Joint Research Center for Marine Microbial Science and Technology, Shandong University, Jinan, Licheng District, Jinan, Shanda S Rd, 27, Shandong, Qingdao, 250100, China
| | - Yu-Zhong Zhang
- MOE Key Laboratory of Evolution and Marine Biodiversity, Frontiers Science Center for Deep Ocean Multispheres and Earth System & College of Marine Life Sciences, Ocean University of China, 5 Yushan Rd, Shinan District, Shandong, Qingdao, 266005, China
- Joint Research Center for Marine Microbial Science and Technology, Shandong University, Jinan, Licheng District, Jinan, Shanda S Rd, 27, Shandong, Qingdao, 250100, China
- State Key Laboratory of Microbial Technology, Marine Biotechnology Research Center, Shandong University, Jinan, Licheng District, Jinan, Shanda S Rd, 27, Shandong, Qingdao, 250100, China
| | - Marcello Mercogliano
- Department of Chemical Sciences, University of Naples Federico II, Via Cinthia 4, 80126, Naples, Italy
| | - Antonio Molinaro
- Department of Chemical Sciences, University of Naples Federico II, Via Cinthia 4, 80126, Naples, Italy
- CEINGE, Istituto di Biotecnologie avanzate, Via Gaetano Salvatore 486, 80131, Naples, Italy
- Department of Chemistry, School of Science, Osaka University, 1-1 Osaka University Machikaneyama, Toyonaka, Osaka, 560-0043, Japan
| | - Alba Silipo
- Department of Chemical Sciences, University of Naples Federico II, Via Cinthia 4, 80126, Naples, Italy
- CEINGE, Istituto di Biotecnologie avanzate, Via Gaetano Salvatore 486, 80131, Naples, Italy
| | - Flaviana Di Lorenzo
- Department of Chemical Sciences, University of Naples Federico II, Via Cinthia 4, 80126, Naples, Italy
- CEINGE, Istituto di Biotecnologie avanzate, Via Gaetano Salvatore 486, 80131, Naples, Italy
| |
Collapse
|