Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (126)

Search Parameters:
Keywords = mouse sepsis model

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
22 pages, 3921 KiB  
Article
Quantitative Proteomics Reveals Fh15 as an Antagonist of TLR4 Downregulating the Activation of NF-κB, Inducible Nitric Oxide, Phagosome Signaling Pathways, and Oxidative Stress of LPS-Stimulated Macrophages
by Albersy Armina-Rodriguez, Bianca N. Valdés Fernandez, Carlimar Ocasio-Malavé, Yadira M. Cantres Rosario, Kelvin Carrasquillo Carrión, Loyda M. Meléndez, Abiel Roche Lima, Eduardo L. Tosado Rodriguez and Ana M. Espino
Int. J. Mol. Sci. 2025, 26(14), 6914; https://doi.org/10.3390/ijms26146914 - 18 Jul 2025
Viewed by 150
Abstract
There is a present need to develop alternative biotherapeutic drugs to mitigate the exacerbated inflammatory immune responses characteristic of sepsis. The potent endotoxin lipopolysaccharide (LPS), a major component of Gram-negative bacterial outer membrane, activates the immune system via Toll-like receptor 4 (TLR4), triggering [...] Read more.
There is a present need to develop alternative biotherapeutic drugs to mitigate the exacerbated inflammatory immune responses characteristic of sepsis. The potent endotoxin lipopolysaccharide (LPS), a major component of Gram-negative bacterial outer membrane, activates the immune system via Toll-like receptor 4 (TLR4), triggering macrophages and a persistent cascade of inflammatory mediators. Our previous studies have demonstrated that Fh15, a recombinant member of the Fasciola hepatica fatty acid binding protein family, can significantly increase the survival rate by suppressing many inflammatory mediators induced by LPS in a septic shock mouse model. Although Fh15 has been proposed as a TLR4 antagonist, the specific mechanisms underlying its immunomodulatory effect remained unclear. In the present study, we employed a quantitative proteomics approach using tandem mass tag (TMT) followed by LC-MS/MS analysis to identify and quantify differentially expressed proteins that participate in signaling pathways downstream TLR4 of macrophages, which can be dysregulated by Fh15. Data are available via ProteomeXchange with identifier PXD065520. Based on significant fold change (FC) cut-off of 1.5 and p-value ≤ 0.05 criteria, we focused our attention to 114 proteins that were upregulated by LPS and downregulated by Fh15. From these proteins, TNFα, IL-1α, Lck, NOS2, SOD2 and CD36 were selected for validation by Western blot on murine bone marrow-derived macrophages due to their relevant roles in the NF-κB, iNOS, oxidative stress, and phagosome signaling pathways, which are closely associated with sepsis pathogenesis. These results suggest that Fh15 exerts a broad spectrum of action by simultaneously targeting multiple downstream pathways activated by TLR4, thereby modulating various aspects of the inflammatory responses during sepsis. Full article
(This article belongs to the Special Issue From Macrophage Biology to Cell and EV-Based Immunotherapies)
Show Figures

Figure 1

24 pages, 12400 KiB  
Article
Synthesis, Antimicrobial Activities, and Model of Action of Novel Tetralone Derivatives Containing Aminoguanidinium Moiety
by Qing-Jie Zhang, Yu-Xi Li, Wen-Bo Ge, Li-Xia Bai, Xiao Xu, Ya-Jun Yang, Xi-Wang Liu and Jian-Yong Li
Int. J. Mol. Sci. 2025, 26(13), 5980; https://doi.org/10.3390/ijms26135980 - 21 Jun 2025
Viewed by 370
Abstract
The objectives of this study were to design, synthesize, and evaluate the antibacterial activity of a series of novel aminoguanidine-tetralone derivatives. Thirty-four new compounds were effectively synthesized through nucleophilic substitution reaction and guanidinylation reaction. Chemical structures of all the desired compounds were identified [...] Read more.
The objectives of this study were to design, synthesize, and evaluate the antibacterial activity of a series of novel aminoguanidine-tetralone derivatives. Thirty-four new compounds were effectively synthesized through nucleophilic substitution reaction and guanidinylation reaction. Chemical structures of all the desired compounds were identified by NMR and HR-MS spectroscopy. Most of the synthesized compounds showed significant antibacterial activity against ESKAPE pathogens and clinically resistant Staphylococcus aureus (S. aureus) isolates. S. aureus is an important pathogen that has the capacity to cause a variety of diseases, including skin infections, pneumonia, and sepsis. The most active compound, 2D, showed rapid bactericidal activity against S. aureus ATCC 29213 and MRSA-2 with MIC/MBC values of 0.5/4 µg/mL and 1/4 µg/mL, respectively. The hemolytic activity and cytotoxicity of 2D was low, with HC50 and IC50(HEK 293-T) values of 50.65 µg/mL and 13.09 µg/mL, respectively. Compound 2D induced the depolarization of the bacterial membrane and disrupted bacterial membrane integrity, ultimately leading to death. Molecular docking revealed that dihydrofolate reductase (DHFR) may be a potential target for 2D. In the mouse skin abscess model caused by MRSA-2, 2D reduced the abscess volume, decreased bacterial load, and alleviated tissue pathological damage at doses of 5 and 10 mg/kg. Therefore, compound 2D may be a promising drug candidate for antibacterial purposes against S. aureus. Full article
(This article belongs to the Special Issue Advanced Research in Veterinary Drugs)
Show Figures

Figure 1

17 pages, 5473 KiB  
Article
Sivelestat-Loaded Neutrophil-Membrane-Coated Antioxidative Nanoparticles for Targeted Endothelial Protection in Sepsis
by Juexian Wei, Aijia Zhong, Yuting Zhang, Ehua Deng, Hengzong Mo, Hongyu Zhao, Jiayu Huang, Huaidong Peng, Kaiyin Zhang, Xiaohui Chen, Haifeng Mao, Yixin Chen and Yongcheng Zhu
Pharmaceutics 2025, 17(6), 766; https://doi.org/10.3390/pharmaceutics17060766 - 10 Jun 2025
Viewed by 648
Abstract
Background/Objectives: This study aims to develop and evaluate neutrophil-membrane-coated nanoparticles (Siv@NMs) encapsulating sivelestat for the treatment of sepsis-induced endothelial injury. Leveraging the intrinsic chemotactic properties of neutrophil membranes, Siv@NMs are engineered to achieve site-specific delivery of sivelestat to damaged endothelia, thereby overcoming [...] Read more.
Background/Objectives: This study aims to develop and evaluate neutrophil-membrane-coated nanoparticles (Siv@NMs) encapsulating sivelestat for the treatment of sepsis-induced endothelial injury. Leveraging the intrinsic chemotactic properties of neutrophil membranes, Siv@NMs are engineered to achieve site-specific delivery of sivelestat to damaged endothelia, thereby overcoming the limitations of conventional therapies in mitigating endothelial dysfunction and multiorgan failure associated with sepsis. Methods: Siv@NMs were synthesized through a combination of ultrasonication and extrusion techniques to encapsulate sivelestat within neutrophil-membrane-derived vesicles. Comprehensive physicochemical characterization included analysis of particle size distribution, zeta potential, and encapsulation efficiency. Stability profiles and controlled release kinetics were systematically evaluated under simulated conditions. In vitro investigations encompassed (1) endothelial cell biocompatibility assessment via cytotoxicity assays, (2) investigation of the targeting efficiency in suppressing endothelial neutrophil extracellular trap generation during inflammation, and (3) ROS-scavenging capacity quantification using flow cytometry with DCFH-DA fluorescent probes. In vivo therapeutic efficacy was validated using a cecal ligation and puncture (CLP) sepsis mouse model, with multiparametric monitoring of endothelial function, inflammatory markers, ROS levels, and survival outcomes. Results: The optimized Siv@NMs exhibited an average particle size of approximately 150 nm, and a zeta potential of −10 mV was achieved. Cellular studies revealed that (1) Siv@NMs selectively bound to inflammatory endothelial cells with minimal cytotoxicity, and (2) Siv@NMs significantly reduced ROS accumulation in endothelial cells subjected to septic stimuli. In vitro experiments demonstrated that Siv@NMs treatment markedly attenuated endothelial injury biomarkers’ expression (ICAM-1 and iNOS), suppressed formation of neutrophil extracellular traps, and improved survival rates compared to treatment with free sivelestat. Conclusions: The neutrophil-membrane-coated nanoparticles loaded with sivelestat present a breakthrough strategy for precision therapy of sepsis-associated endothelial injury. This bioengineered system synergistically combines targeted drug delivery with multimodal therapeutic effects, including ROS mitigation, anti-inflammatory action, and endothelial protection. These findings substantiate the clinical translation potential of Siv@NMs as a next-generation nanotherapeutic for sepsis management. Full article
(This article belongs to the Special Issue ROS-Mediated Nano Drug Delivery for Antitumor Therapy)
Show Figures

Figure 1

13 pages, 1080 KiB  
Article
3-Deoxysappanchalcone Inhibited High Mobility Group Box Protein 1-Mediated Severe Inflammatory Responses
by Jinhee Lee, Gyuri Han and Jong-Sup Bae
Pharmaceuticals 2025, 18(5), 731; https://doi.org/10.3390/ph18050731 - 16 May 2025
Cited by 1 | Viewed by 418
Abstract
Background/Objectives: Phytochemicals are increasingly recognized for their therapeutic potential in treating various diseases, including vascular disorders. High mobility group box 1 (HMGB1), a key mediator of late-stage sepsis, triggers the release of proinflammatory cytokines, leading to inflammation and systemic complications. Elevated plasma levels [...] Read more.
Background/Objectives: Phytochemicals are increasingly recognized for their therapeutic potential in treating various diseases, including vascular disorders. High mobility group box 1 (HMGB1), a key mediator of late-stage sepsis, triggers the release of proinflammatory cytokines, leading to inflammation and systemic complications. Elevated plasma levels of HMGB1 impair diagnosis and prognosis while worsening outcomes in inflammatory conditions. 3-deoxysappanchalcone (3-DSC), a compound derived from Biancaea sappan (L.) Tod., has demonstrated anti-influenza and anti-allergic effects, though its role in HMGB1-mediated severe vascular inflammation remains unclear. This study hypothesized that 3-DSC could modulate lipopolysaccharide-induced HMGB1 activity and its downstream inflammatory pathways in human umbilical vein endothelial cells (HUVECs). Methods: In vitro and in vivo permeability; cell viability, adhesion, and excavation of leukocytes; the development of cell adhesion molecules; and lastly, the production of proinflammatory substances were investigated on human endothelial cells and mouse disease models to investigate the efficacy of 3-DSC in inflammatory conditions. Results: Experiments revealed that 3-DSC inhibited HMGB1 translocation from HUVECs, reduced neutrophil adhesion and extravasation, suppressed HMGB1 receptor formation, and blocked nuclear factor-κB (NF-κB) activation and tumor necrosis factor-α (TNF-α) synthesis. Conclusions: These findings suggest that 3-DSC effectively mitigates HMGB1-driven inflammation, offering promise as a therapeutic candidate for inflammatory diseases. Full article
(This article belongs to the Section Natural Products)
Show Figures

Graphical abstract

15 pages, 8400 KiB  
Article
Hydrogen Sulfide (H2S) Mitigates Sepsis-Induced Adrenal Dysfunction via Inhibition of TNFα-Mediated Necroptosis
by Kai Ma, Jingwen Huang, Jin Zhang, Yuan Tian, Jing Hu, Linhao Ma and Changnan Wang
Pathogens 2025, 14(5), 439; https://doi.org/10.3390/pathogens14050439 - 30 Apr 2025
Viewed by 528
Abstract
Background: Sepsis is a life-threatening condition that is characterized by systemic inflammation and organ dysfunction, with adrenal dysfunction being a significant complication. This study aimed to investigate the role of necroptosis and hydrogen sulfide (H2S) in sepsis-induced adrenal dysfunction. Methods: A [...] Read more.
Background: Sepsis is a life-threatening condition that is characterized by systemic inflammation and organ dysfunction, with adrenal dysfunction being a significant complication. This study aimed to investigate the role of necroptosis and hydrogen sulfide (H2S) in sepsis-induced adrenal dysfunction. Methods: A cecal ligation and puncture (CLP)-induced sepsis mouse model was employed. Adrenocortical-specific mixed lineage kinase domain-like pseudokinase (MLKL) knockout (MLKL-KO) and cystathioneine β-synthase (CBS) knockout (CBS-KO) mice were generated using Cre-loxP technology and adrenocortical-specific Cre tool mice. In vitro experiments utilized TNFα-stimulated Y1 adrenocortical cells. The treatments included the H2S donor NaHS, TNFα inhibitor R-7050, necroptosis inhibitor NSA and CBS inhibitor AOAA. Pathological assessment involved hematoxylin–eosin (H&E) staining and a Western blot analysis of necroptosis markers (the phosphorylation of MLKL (p-MLKL) and phosphorylation of receptor-interacting protein kinases 1 (p-RIPK1)). Results: Sepsis induced adrenal congestion, elevated TNFα levels, and activated necroptosis (increased p-MLKL/p-RIPK1) in wild-type mice. H2S treatment attenuated adrenal damage, reduced TNFα, and suppressed necroptosis. MLKL knockout reduced septic adrenal dysfunction, whereas CBS knockout exacerbated septic adrenal dysfunction. In vitro, TNFα induced Y1 cell necroptosis, which was reversed by H2S or NSA. AOAA exacerbated TNFα-induced necroptosis in Y1 cells. Conclusions: H2S inhibits TNFα-mediated necroptosis, thereby preserving adrenal integrity in sepsis. Targeting the TNFα–necroptosis axis and enhancing endogenous H2S production may represent novel therapeutic strategies for sepsis-associated adrenal dysfunction. Full article
(This article belongs to the Special Issue Sepsis and Group A Streptococcus)
Show Figures

Figure 1

15 pages, 4600 KiB  
Article
Synergistic Antibacterial Activity of Amorolfine Combined with Colistin Against Acinetobacter baumannii
by Ting Lin, Shuaiyuan Liu, Xuan Chen, Fei Gao, Lu Liu, Daijie Chen and Yu Yin
Int. J. Mol. Sci. 2025, 26(7), 3312; https://doi.org/10.3390/ijms26073312 - 2 Apr 2025
Cited by 1 | Viewed by 596
Abstract
Emerging resistance to colistin in Acinetobacter baumannii is concerning because of the limited therapeutic options for this important clinical pathogen. Given the shortage of new antibiotics, one strategy that has been proven to be therapeutically effective is to overcome antibiotic-resistant pathogens by combining [...] Read more.
Emerging resistance to colistin in Acinetobacter baumannii is concerning because of the limited therapeutic options for this important clinical pathogen. Given the shortage of new antibiotics, one strategy that has been proven to be therapeutically effective is to overcome antibiotic-resistant pathogens by combining existing antibiotics with another antibiotic or non-antibiotic. This study was designed to investigate the potential synergistic antibacterial activity of amorolfine, a morpholine antifungal drug, in combination with colistin against A. baumannii. In this work, antibiotic susceptibility testing, checkerboard assays, and time-kill curves were used to investigate the synergistic efficacy of colistin combined with amorolfine. The molecular mechanisms of combination therapy were analyzed using fluorometric assays, UV-vis spectroscopy, and molecular docking. Finally, we evaluated the in vivo efficacy of combination therapy against A. baumannii. In brief, the combination therapy showed significant synergistic activity against A. baumannii (FICI = 0.094). In addition, the combination of amorolfine improved the membrane disruption of colistin, and amorolfine exhibited the capacity of binding to DNA. Moreover, in a mouse sepsis model, this combination therapy increased survival compared to colistin monotherapy. Our findings demonstrated that amorolfine serves as a potential colistin adjuvant against Acinetobacter baumannii. Full article
(This article belongs to the Section Molecular Microbiology)
Show Figures

Figure 1

15 pages, 2216 KiB  
Article
Computational Design and In Vitro and In Vivo Characterization of an ApoE-Based Synthetic High-Density Lipoprotein for Sepsis Therapy
by Ling Guo, Yaxia Yuan, Fang Zheng, Changguo Zhan and Xiangan Li
Biomolecules 2025, 15(3), 397; https://doi.org/10.3390/biom15030397 - 11 Mar 2025
Viewed by 713
Abstract
Introduction: Septic patients have low levels of high-density lipoproteins (HDLs), which is a risk factor. Replenishing HDLs with synthetic HDLs (sHDLs) has shown promise as a therapy for sepsis. This study aimed to develop a computational approach to design and test new types [...] Read more.
Introduction: Septic patients have low levels of high-density lipoproteins (HDLs), which is a risk factor. Replenishing HDLs with synthetic HDLs (sHDLs) has shown promise as a therapy for sepsis. This study aimed to develop a computational approach to design and test new types of sHDLs for sepsis treatment. Methods: We used a three-step computational approach to design sHDL nanoparticles based on the structure of HDLs and their binding to endotoxins. We tested the efficacy of these sHDLs in two sepsis mouse models—cecal ligation and puncture (CLP)-induced and P. aeruginosa-induced sepsis models—and assessed their impact on inflammatory signaling in cells. Results: We designed four sHDL nanoparticles: two based on the ApoA-I sequence (YGZL1 and YGZL2) and two based on the ApoE sequence (YGZL3 and YGZL4). We demonstrated that an ApoE-based sHDL nanoparticle, YGZL3, provides effective protection against CLP- and P. aeruginosa-induced sepsis. The sHDLs effectively suppressed inflammatory signaling in HEK-blue or RAW264 cells. Conclusions: Unlike earlier approaches, we developed a new approach that employs computational simulations to design a new type of sHDL based on HDL’s structure and function. We found that YGZL3, an ApoE sequence-based sHDL, provides effective protection against sepsis in two mouse models. Full article
(This article belongs to the Section Molecular Medicine)
Show Figures

Figure 1

18 pages, 1466 KiB  
Article
The Novel Role of the Expression of Toll-like Receptors TLR-5, TLR-6, and TLR-9 and Associated Up-Regulation of Programmed Cell Death 1 Receptor (PD-1) and Its Ligand (PD-L1) in Lung Sepsis
by Georgios Sinos, Dimitrios Schizas, Alkistis Kapelouzou, Maximos Frountzas, Michalis Katsimpoulas, Konstantinos S. Mylonas, Emmanouil I. Kapetanakis, Alexandros Papalampros, Theodore Liakakos and Andreas Alexandrou
Int. J. Mol. Sci. 2025, 26(5), 2274; https://doi.org/10.3390/ijms26052274 - 4 Mar 2025
Viewed by 795
Abstract
Sepsis is a leading cause of death in hospitalized patients. The underlying pathophysiologic mechanisms of sepsis have not been fully elucidated thus far. The receptor of programmed cell death 1 (PD-1) and its ligand (PD-L1), in combination with the Toll-like receptors (TLRs), seem [...] Read more.
Sepsis is a leading cause of death in hospitalized patients. The underlying pathophysiologic mechanisms of sepsis have not been fully elucidated thus far. The receptor of programmed cell death 1 (PD-1) and its ligand (PD-L1), in combination with the Toll-like receptors (TLRs), seem to contribute considerably in systematic responses during sepsis. Investigating the relationship between them and identifying potential target pathways is important in the future management of sepsis, especially in relation to acute lung injury. This study investigated the interactions between TLR-5, -6, and -9 and PD-1/PD-L1 expression in a septic mouse model. Sixty C57BL/6J mice were included and categorized in six study groups. Three sepsis (S) groups (24 h, 48 h, and 72 h) and three sham (Sh) groups (24 h, 48 h, and 72 h) were created. Cecal ligation and puncture (CLP) was utilized to simulate sepsis in the S groups. Hematological analysis and lung tissue histopathological analysis were performed after 24 h, 48 h, and 72 h. Significant decreases in S groups compared to Sh groups in WBC and lymphocyte counts at 24, 48, and 72 h were observed. Significant increases in S groups compared to Sh groups in RBC and monocyte counts, IL-6 and IL-10 levels, alveolar flooding, and alveolar collapse were demonstrated by histopathological analysis. This study suggested a strong correlation between TLR expression and PD-1/PD-L1 up-regulation in lung tissue during sepsis. These molecules, also, seem to contribute to the histopathological changes in lung tissue during sepsis, leading to acute lung injury. Full article
Show Figures

Figure 1

12 pages, 2906 KiB  
Article
Malocclusion Worsens Survival Following Sepsis Due to the Disruption of Innate and Acquired Immunity
by Yoshihisa Fujinami, Masafumi Saito, Yuko Ono, Masaya Akashi, Shigeaki Inoue and Joji Kotani
Int. J. Mol. Sci. 2025, 26(5), 1894; https://doi.org/10.3390/ijms26051894 - 22 Feb 2025
Cited by 1 | Viewed by 822
Abstract
Sepsis is a severe condition with high mortality, in which immune dysfunction plays a critical role. Poor oral health has been linked to frailty, but its impact on sepsis outcomes remains unclear. Therefore, we used a mouse model of malocclusion and sepsis to [...] Read more.
Sepsis is a severe condition with high mortality, in which immune dysfunction plays a critical role. Poor oral health has been linked to frailty, but its impact on sepsis outcomes remains unclear. Therefore, we used a mouse model of malocclusion and sepsis to investigate how tooth loss affects immune responses during sepsis. Adult male C57BL/6 mice were divided into four groups: Control, Malocclusion (Mal), Sepsis (CS), and Malocclusion with Sepsis (Mal + CS). Malocclusion was induced by tooth extraction, and sepsis was induced using cecal slurry injection. We assessed survival rates, immune cell counts, and biochemical markers. The Mal + CS group exhibited significantly lower survival rates and greater weight loss compared to the CS group. The flow cytometry showed reduced neutrophils, monocytes, and T cells in the Mal + CS group. Elevated ALT and AST levels indicated liver damage. No significant differences in bacterial loads were observed, but immune suppression was exacerbated in the Mal + CS group. Malocclusion worsens sepsis outcomes by impairing both innate and adaptive immune responses. These findings emphasize the importance of oral health in improving sepsis prognosis and immune function during critical illnesses. Full article
Show Figures

Figure 1

21 pages, 1594 KiB  
Article
Comparative Evaluation of the Antibacterial and Antitumor Activities of Marine Alkaloid 3,10-Dibromofascaplysin
by Maxim E. Zhidkov, Polina A. Smirnova, Natalia E. Grammatikova, Elena B. Isakova, Andrey E. Shchekotikhin, Olga N. Styshova, Anna A. Klimovich and Aleksandr M. Popov
Mar. Drugs 2025, 23(2), 68; https://doi.org/10.3390/md23020068 - 6 Feb 2025
Viewed by 1047
Abstract
Fascaplysins form a group of marine natural products with unique cationic five-ring coplanar backbone. Native fascaplysin exhibits a broad spectrum of bioactivities, among which the cytotoxic activity has been the most investigated. Several fascaplysin derivatives have more selective biological effects and are promising [...] Read more.
Fascaplysins form a group of marine natural products with unique cationic five-ring coplanar backbone. Native fascaplysin exhibits a broad spectrum of bioactivities, among which the cytotoxic activity has been the most investigated. Several fascaplysin derivatives have more selective biological effects and are promising as lead compounds. Thus, the introduction of a substituent at C-9 of fascaplysin leads to a strong increase in its antimicrobial properties. Here, a comparative assessment of the antimicrobial activity of synthetic analogs of the marine alkaloids 3-bromofascaplysin, 10-bromofascaplysin, and 3,10-dibromofascaplysin, along with some of their isomers and analogs, was carried out against a panel of Gram-positive bacteria in vitro. For the first time, a significant increase in the antimicrobial activity of fascaplysin was observed when a substituent was introduced at C-3. The introduction of two bromine atoms at C-2 and C-9 enhances the antimicrobial properties by 4 to 16 times, depending on the tested strain. Evaluation of the antimicrobial potential in vivo showed that fascaplysin and 3,10-dibromofascaplysin had comparable efficacy in the mouse staphylococcal sepsis model. Additionally, 3,10-dibromofascaplysin demonstrated a strong and reliable antitumor effect in vivo on the Ehrlich carcinoma inoculated subcutaneously, with a value of tumor growth inhibition by 49.2% 20 days after treatment. However, further studies on alternative chemical modifications of fascaplysin are needed to improve its chemotherapeutic properties. Full article
(This article belongs to the Special Issue Marine Natural Products as Anticancer Agents, 4th Edition)
Show Figures

Figure 1

25 pages, 3697 KiB  
Review
Recent Advances in Studies of Serum Amyloid A: Implications in Inflammation, Immunity and Tumor Metastasis
by Yixin Chang, Yezhou Liu, Yuanrui Zou and Richard D. Ye
Int. J. Mol. Sci. 2025, 26(3), 987; https://doi.org/10.3390/ijms26030987 - 24 Jan 2025
Cited by 1 | Viewed by 3373
Abstract
Research on serum amyloid A (SAA) has seen major advancement in recent years with combined approaches of structural analysis and genetically altered mice. Initially identified as an acute-phase reactant, SAA is now recognized as a major player in host defense, inflammation, lipid metabolism [...] Read more.
Research on serum amyloid A (SAA) has seen major advancement in recent years with combined approaches of structural analysis and genetically altered mice. Initially identified as an acute-phase reactant, SAA is now recognized as a major player in host defense, inflammation, lipid metabolism and tumor metastasis. SAA binding and the neutralization of LPS attenuate sepsis in mouse models. SAA also displays immunomodulatory functions in Th17 differentiation and macrophage polarization, contributing to a pro-metastatic tumor microenvironment. In spite of the progress, the regulatory mechanisms for these diverse functions of SAA remain unclear. This review provides a brief summary of recent advances in SAA research on immunity, inflammation, tumor microenvironment and in vivo models. Full article
Show Figures

Figure 1

11 pages, 1215 KiB  
Article
Bacteriophage and Phage-Encoded Depolymerase Exhibit Antibacterial Activity Against K9-Type Acinetobacter baumannii in Mouse Sepsis and Burn Skin Infection Models
by Alexander I. Borzilov, Nikolay V. Volozhantsev, Olga V. Korobova, Lyubov V. Kolupaeva, Evgenia S. Pereskokova, Tatiana I. Kombarova, Mikhail M. Shneider, Konstantin A. Miroshnikov, Ivan A. Dyatlov and Anastasia V. Popova
Viruses 2025, 17(1), 70; https://doi.org/10.3390/v17010070 - 6 Jan 2025
Cited by 3 | Viewed by 1171
Abstract
Acinetobacter baumannii is a widely distributed nosocomial pathogen that causes various acute and chronic infections, particularly in immunocompromised patients. In this study, the activities of the K9-specific virulent phage AM24 and phage-encoded depolymerase DepAPK09 were assessed using in vivo mouse sepsis and burn [...] Read more.
Acinetobacter baumannii is a widely distributed nosocomial pathogen that causes various acute and chronic infections, particularly in immunocompromised patients. In this study, the activities of the K9-specific virulent phage AM24 and phage-encoded depolymerase DepAPK09 were assessed using in vivo mouse sepsis and burn skin infection models. In the mouse sepsis model, in the case of prevention or early treatment, a single K9-specific phage or recombinant depolymerase injection was able to protect 100% of the mice after parenteral infection with a lethal dose of A. baumannii of the K9-type, with complete eradication of the pathogen. In the case of delayed treatment, mouse survival decreased to 70% when injected with the phage and to 40% when treated with the recombinant enzyme. In the mouse burn skin infection model, the number of A. baumannii cells on the surface of the wound and in the deep layers of the skin decreased by several-fold after treatment with both the K9-specific phage and the recombinant depolymerase. The phage and recombinant depolymerase were highly stable and retained activity under a wide range of temperatures and pH values. The results obtained contribute to expanding our understanding of the in vivo therapeutic potential of specific phages and phage-derived depolymerases interacting with A. baumannii of different capsular types. Full article
(This article belongs to the Special Issue Phage-Bacteria Interplay in Health and Disease, Second Edition)
Show Figures

Figure 1

23 pages, 9986 KiB  
Article
Multi-Omics and Network-Based Drug Repurposing for Septic Cardiomyopathy
by Pei-Pei Liu, Xin-Yue Yu, Qing-Qing Pan, Jia-Jun Ren, Yu-Xuan Han, Kai Zhang, Yan Wang, Yin Huang and Tao Ban
Pharmaceuticals 2025, 18(1), 43; https://doi.org/10.3390/ph18010043 - 2 Jan 2025
Cited by 3 | Viewed by 1387 | Correction
Abstract
Background/Objectives: Septic cardiomyopathy (SCM) is a severe cardiac complication of sepsis, characterized by cardiac dysfunction with limited effective treatments. This study aimed to identify repurposable drugs for SCM by integrated multi-omics and network analyses. Methods: We generated a mouse model of SCM induced [...] Read more.
Background/Objectives: Septic cardiomyopathy (SCM) is a severe cardiac complication of sepsis, characterized by cardiac dysfunction with limited effective treatments. This study aimed to identify repurposable drugs for SCM by integrated multi-omics and network analyses. Methods: We generated a mouse model of SCM induced by lipopolysaccharide (LPS) and then obtained comprehensive metabolic and genetic data from SCM mouse hearts using ultra-performance liquid chromatography–tandem mass spectrometry (UPLC–MS/MS) and RNA sequencing (RNA-seq). Using network proximity analysis, we screened for FDA-approved drugs that interact with SCM-associated pathways. Additionally, we tested the cardioprotective effects of two drug candidates in the SCM mouse model and explored their mechanism-of-action in H9c2 cells. Results: Network analysis identified 129 drugs associated with SCM, which were refined to 14 drug candidates based on strong network predictions, proven anti-infective effects, suitability for ICU use, and minimal side effects. Among them, acetaminophen and pyridoxal phosphate significantly improved cardiac function in SCM moues, as demonstrated by the increased ejection fraction (EF) and fractional shortening (FS), and the reduced levels of cardiac injury biomarkers: B-type natriuretic peptide (BNP) and cardiac troponin I (cTn-I). In vitro assays revealed that acetaminophen inhibited prostaglandin synthesis, reducing inflammation, while pyridoxal phosphate restored amino acid balance, supporting cellular function. These findings suggest that both drugs possess protective effects against SCM. Conclusions: This study provides a robust platform for drug repurposing in SCM, identifying acetaminophen and pyridoxal phosphate as promising candidates for clinical translation, with the potential to improve treatment outcomes in septic patients with cardiac complications. Full article
(This article belongs to the Special Issue Data-Driven Biomarker and Drug Discovery for Complex Disease)
Show Figures

Figure 1

16 pages, 4966 KiB  
Article
Quercetin Ameliorates Acute Lethal Sepsis in Mice by Inhibiting Caspase-11 Noncanonical Inflammasome in Macrophages
by Eojin Kim, Deok-Hyeong Choi and Young-Su Yi
Molecules 2024, 29(24), 5900; https://doi.org/10.3390/molecules29245900 - 13 Dec 2024
Cited by 3 | Viewed by 1194
Abstract
Quercetin is a natural polyphenolic flavonoid widely found in plants, fruits, and vegetables, and has been reported to play pharmacological roles in numerous pathogenic conditions. The anti-inflammatory effects of quercetin in various inflammatory conditions and diseases have been well-documented. However, its regulatory role [...] Read more.
Quercetin is a natural polyphenolic flavonoid widely found in plants, fruits, and vegetables, and has been reported to play pharmacological roles in numerous pathogenic conditions. The anti-inflammatory effects of quercetin in various inflammatory conditions and diseases have been well-documented. However, its regulatory role in noncanonical inflammasome activation has not yet been demonstrated. This study investigated the anti-inflammatory effects of quercetin in caspase-11 noncanonical inflammasome-activated inflammatory responses in macrophages and a mouse model of acute lethal sepsis. Quercetin protected J774A.1 macrophages from lipopolysaccharide (LPS)-induced cell death and caspase-11 noncanonical inflammasome-induced pyroptosis. It significantly decreased the production and mRNA expression of pro-inflammatory cytokines, such as interleukin (IL)-1β, IL-18, and IL-6, but not tumor necrosis factor (TNF)-α, and inflammatory molecules, such as nitric oxide (NO) and inducible NO synthase in caspase-11 noncanonical inflammasome-activated J774A.1 cells. Mechanistically, quercetin strongly suppressed the autoproteolysis and secretion of caspase-11 and the proteolysis of gasdermin D in caspase-11 noncanonical inflammasome-activated J774A.1 cells. However, quercetin did not inhibit the direct binding of caspase-11 to LPS. In vivo, the study revealed that quercetin increased the survival rate of mice with acute lethal sepsis and decreased serum levels of pro-inflammatory cytokines without causing significant toxicity. In conclusion, this study highlights quercetin-mediated anti-inflammatory action in inflammatory responses and acute lethal sepsis through a novel mechanism that targets the caspase-11 noncanonical inflammasome in macrophages, suggesting quercetin as a promising anti-inflammatory agent in natural medicine. Full article
(This article belongs to the Special Issue Natural Polyphenols in Human Health (Volume II))
Show Figures

Graphical abstract

16 pages, 4666 KiB  
Article
Platelet Glycoprotein Ibα Cytoplasmic Tail Exacerbates Thrombosis During Bacterial Sepsis
by Yue Xia, Chenglin Sun, Kangxi Zhou, Jie Shen, Jiaojiao Li, Qiuxia Huang, Jiahao Du, Sai Zhang, Kang Sun, Renping Hu, Rong Yan and Kesheng Dai
Int. J. Mol. Sci. 2024, 25(21), 11548; https://doi.org/10.3390/ijms252111548 - 27 Oct 2024
Cited by 2 | Viewed by 1617
Abstract
Septic patients, coupling severe disseminated intravascular coagulation (DIC) and thrombocytopenia, have poor prognoses and higher mortality. The platelet glycoprotein Ibα (GPIbα) is involved in thrombosis, hemostasis, and inflammation response. However, it remains unclear whether the GPIbα cytoplasmic tail regulates sepsis-mediated platelet activation and [...] Read more.
Septic patients, coupling severe disseminated intravascular coagulation (DIC) and thrombocytopenia, have poor prognoses and higher mortality. The platelet glycoprotein Ibα (GPIbα) is involved in thrombosis, hemostasis, and inflammation response. However, it remains unclear whether the GPIbα cytoplasmic tail regulates sepsis-mediated platelet activation and inflammation, especially in Staphylococcus aureus (S. aureus) and Escherichia coli (E. coli) infections. Using a mouse model of S. aureus-induced bacteremia, we found that both 10 amino acids of GPIbα C-terminal sequence deficiency and pharmacologic inhibition of protein kinase C (PKC) alleviated pathogenesis by diminishing platelet activation and aggregate formation. Furthermore, the GPIbα cytoplasmic tail promoted the phagocytosis of platelets by Kupffer cells in vivo. The genetically deficient GPIbα cytoplasmic tail also downregulated inflammatory cytokines and reduced liver damage, ultimately improving the survival rate of the septic mice. Our results illustrate that the platelet GPIbα cytoplasmic domain exacerbates excessive platelet activation and inflammation associated with sepsis through a PKC-dependent pathway. Thus, our findings provide insights for the development of effective therapeutic strategies using PKC inhibitor treatment against bacterial infection. Full article
(This article belongs to the Section Molecular Pathology, Diagnostics, and Therapeutics)
Show Figures

Figure 1

Back to TopTop