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
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (842)

Search Parameters:
Keywords = M1-like macrophage

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
23 pages, 11376 KB  
Article
Hyssopus cuspidatus Boriss Volatile Extract (SXC): A Dual-Action Antioxidant and Antifungal Agent Targeting Candida albicans Pathogenicity and Vulvovaginal Candidiasis via Host Oxidative Stress Modulation and Fungal Metabolic Reprogramming
by Yun-Dan Guo, Ming-Xuan Zhang, Quan-Yong Yu, Lu-Lu Wang, Yan-Xing Han, Tian-Le Gao, Yuan Lin, Cai Tie and Jian-Dong Jiang
Antioxidants 2025, 14(9), 1046; https://doi.org/10.3390/antiox14091046 - 25 Aug 2025
Viewed by 331
Abstract
Background and purpose: Vulvovaginal candidiasis (VVC), caused by Candida albicans (C. albicans), is exacerbated by oxidative stress and uncontrolled inflammation. Pathogens like C. albicans generate reactive oxygen species (ROS) to enhance virulence, while host immune responses further amplify oxidative damage. This [...] Read more.
Background and purpose: Vulvovaginal candidiasis (VVC), caused by Candida albicans (C. albicans), is exacerbated by oxidative stress and uncontrolled inflammation. Pathogens like C. albicans generate reactive oxygen species (ROS) to enhance virulence, while host immune responses further amplify oxidative damage. This study investigates the antioxidant and antifungal properties of Hyssopus cuspidatus Boriss volatile extract (SXC), a traditional Uyghur medicinal herb, against fluconazole-resistant VVC. We hypothesize that SXC’s bioactive volatiles counteract pathogen-induced oxidative stress while inhibiting fungal growth and inflammation. Methods: GC-MS identified SXC’s major bioactive components, while broth microdilution assays determined minimum inhibitory concentrations (MICs) against bacterial/fungal pathogens, and synergistic interactions with amphotericin B (AmB) or fluconazole (FLC) were assessed via time–kill kinetics. Anti-biofilm activity was quantified using crystal violet/XTT assays, and in vitro studies evaluated SXC’s effects on C. albicans-induced cytotoxicity (LDH release in A431 cells) and inflammatory responses (cytokine production in LPS-stimulated RAW264.7 macrophages). A murine VVC model, employing estrogen-mediated pathogenesis and intravaginal C. albicans challenge, confirmed SXC’s in vivo effects. Immune modulation was assessed using ELISA and RT-qPCR targeting inflammatory and antioxidative stress mediators, while UPLC-MS was employed to profile metabolic perturbations in C. albicans. Results: Gas chromatography-mass spectrometry identified 10 key volatile components contributing to SXC’s activity. SXC exhibited broad-spectrum antimicrobial activity with MIC values ranging from 0.125–16 μL/mL against bacterial and fungal pathogens, including fluconazole-resistant Candida strains. Time–kill assays revealed that combinations of AmB-SXC and FLC-SXC achieved sustained synergistic bactericidal activity across all tested strains. Mechanistic studies revealed SXC’s dual antifungal actions: inhibition of C. albicans hyphal development and biofilm formation through downregulation of the Ras1-cAMP-Efg1 signaling pathway, and attenuation of riboflavin-mediated energy metabolism crucial for fungal proliferation. In the VVC model, SXC reduced vaginal fungal burden, alleviated clinical symptoms, and preserved vaginal epithelial integrity. Mechanistically, SXC modulated host immune responses by suppressing oxidative stress and pyroptosis through TLR4/NF-κB/NLRP3 pathway inhibition, evidenced by reduced caspase-1 activation and decreased pro-inflammatory cytokines (IL-1β, IL-6, TNF-α). Conclusions: SXC shows promise as a broad-spectrum natural antimicrobial against fungal pathogens. It inhibited C. albicans hyphal growth, adhesion, biofilm formation, and invasion in vitro, while reducing oxidative and preserving vaginal mucosal integrity in vivo. By disrupting fungal metabolic pathways and modulating host immune responses, SXC offers a novel approach to treating recurrent, drug-resistant VVC. Full article
Show Figures

Graphical abstract

19 pages, 2846 KB  
Article
Synovial Fluid Biomarker Profile After Intra-Articular Administration of Neosaxitoxin in Horses: A Feasibility Study
by Cristóbal Dörner, Néstor Lagos, Lissette Oyaneder, Bruno C. Menarim and Galia Ramírez-Toloza
Animals 2025, 15(16), 2453; https://doi.org/10.3390/ani15162453 - 21 Aug 2025
Viewed by 361
Abstract
Osteoarthritis (OA) is an inflammatory joint disease and the leading cause of musculoskeletal disability affecting human and veterinary patients. New therapeutics halting inflammation while preserving joint homeostasis remain a critical need. Voltage-gated sodium (NaV) channels regulate the pro-inflammatory response of macrophages in the [...] Read more.
Osteoarthritis (OA) is an inflammatory joint disease and the leading cause of musculoskeletal disability affecting human and veterinary patients. New therapeutics halting inflammation while preserving joint homeostasis remain a critical need. Voltage-gated sodium (NaV) channels regulate the pro-inflammatory response of macrophages in the synovium, the central driver of joint homeostasis. Neosaxitoxin (NeoSTX) is a phycotoxin that blocks NaV channels, conferring a unique potential to regulate joint inflammation. This study evaluated the safety of intra-articular administration of NeoSTX in horses. Sixteen horses were allocated into two groups (n = 8/each). One group received one intraarticular dose (20 µg/2 mL of saline) of NeoSTX into one tarsocrural joint, while the control group received 2 mL of saline (0.9% NaCl). No differences were observed between groups for systemic or local signs of inflammation, including objective measures of surface temperature and joint effusion. Concentrations of synovial fluid total nucleated and differential cell counts, total protein, glucose, calcium, and 23 cytokines/chemokines measured throughout this study did not differ between treatment groups. In this short-term study, intra-articular NeoSTX injection was shown to be well tolerated and likely safe. Ongoing studies should elucidate the role of NeoSTX in modulating synovial mechanisms of inflammation and its endogenous resolution. Full article
(This article belongs to the Section Equids)
Show Figures

Figure 1

20 pages, 2328 KB  
Article
Effect of Silk Fibroin as a Sustainable Solvent on the Extraction of Bixin from Annatto Seeds (Bixa orellana L.)
by Swanny Ferreira Borges, Fabricio H. e Holanda, Kaio C. De Maria, Sônia do Socorro do C. Oliveira, David E. Q. Jimenez, Celisnolia Morais Leite, Valtencir Zucolotto and Irlon M. Ferreira
Sustainability 2025, 17(16), 7518; https://doi.org/10.3390/su17167518 - 20 Aug 2025
Viewed by 415
Abstract
Bixin, an apocarotenoid from Bixa orellana seeds, is a valuable natural pigment with industrial and pharmacological applications. Traditional extraction methods rely on organic solvents, but eco-friendly alternatives like silk fibroin solution (SFS) are emerging. This study evaluated SFS for bixin extraction from annatto [...] Read more.
Bixin, an apocarotenoid from Bixa orellana seeds, is a valuable natural pigment with industrial and pharmacological applications. Traditional extraction methods rely on organic solvents, but eco-friendly alternatives like silk fibroin solution (SFS) are emerging. This study evaluated SFS for bixin extraction from annatto seeds, optimizing conditions using Box-Behnken Design (BBD). The optimal parameters 1.5% SFS, 60 °C, and 60 min yielded 10.87 mg/mL (liquid extract of annatto seeds, LEAS + SFS) and 150.72 mg/g (solid extract of annatto seeds, SEAS + SFS). Cell viability was assessed in human dermal fibroblasts (HDFn) and RAW 264.7 murine macrophages via MTT assay. After 24 and 72 h, LEAS + SFS, SEAS + SFS, purified bixin (PB), and SFS maintained >70% viability in HDFn cells. Similarly, RAW 264.7 cells showed >70% viability after 24 h, indicating low cytotoxicity. These results highlight the biocompatibility of SFS-extracted bixin, supporting its potential in food, cosmetics, and biomedicine. The study demonstrates that SFS is an effective, sustainable alternative to traditional solvents, offering high extraction efficiency and minimal toxicity. This method aligns with green chemistry principles, providing a promising solution for bixin production. Full article
Show Figures

Graphical abstract

25 pages, 5512 KB  
Article
Unraveling the Clinical Landscape of RNA Modification Regulators with Multi-Omics Insights in Pan-Cancer
by Qingman Li, Jingjing Zhang, Zuyi Cao, Jiale Wang, Jiaxing Song and Xianfu Yi
Cancers 2025, 17(16), 2695; https://doi.org/10.3390/cancers17162695 - 19 Aug 2025
Viewed by 403
Abstract
Background/Objectives: Cancer remains a major global health challenge, with RNA modifications increasingly recognized as key regulators of tumor progression. However, integrated pan-cancer analyses across multiple modification types are limited. Methods: We performed a comprehensive analysis of 170 RNA modification-related genes across 33 cancer [...] Read more.
Background/Objectives: Cancer remains a major global health challenge, with RNA modifications increasingly recognized as key regulators of tumor progression. However, integrated pan-cancer analyses across multiple modification types are limited. Methods: We performed a comprehensive analysis of 170 RNA modification-related genes across 33 cancer types, uncovering diverse expression, mutation, and epigenetic patterns. Results: Key regulators such as IGF2BP3, CFI, and ELF3 showed cancer-specific prognostic significance. We developed an RNA Modification Score (RMS) with strong prognostic performance (AUC up to 0.92), correlating with the tumor stage, immune infiltration, and immunotherapy response. High-risk groups exhibited immune checkpoint dysregulation and enriched M1 macrophages in glioblastoma. Drug screening highlighted oncrasin-72 as a potential therapy. Validation via single-cell/spatial transcriptomics and immunohistochemistry confirmed the spatial localization of critical genes like CFI and ELF3. Conclusions: Our study reveals the multifaceted role of RNA modifications in cancer, providing a translational framework for personalized prognosis and therapy in precision oncology. Full article
(This article belongs to the Special Issue Advancements in “Cancer Biomarkers” for 2025–2026)
Show Figures

Figure 1

19 pages, 1400 KB  
Systematic Review
Targeting Macrophages in Glioblastoma: Current Therapies and Future Directions
by Giovanni Pennisi, Federico Valeri, Benedetta Burattini, Placido Bruzzaniti, Carmelo Lucio Sturiale, Andrea Talacchi, Fabio Papacci, Alessandro Olivi and Giuseppe Maria Della Pepa
Cancers 2025, 17(16), 2687; https://doi.org/10.3390/cancers17162687 - 18 Aug 2025
Viewed by 613
Abstract
Glioblastoma (GBM) is an aggressive brain tumor characterized by an immunosuppressive tumor microenvironment (TME), which contributes to treatment resistance and disease progression. Background: Tumor-associated macrophages (TAMs), comprising both resident microglia and bone marrow–derived macrophages, play a central role in supporting tumor growth, [...] Read more.
Glioblastoma (GBM) is an aggressive brain tumor characterized by an immunosuppressive tumor microenvironment (TME), which contributes to treatment resistance and disease progression. Background: Tumor-associated macrophages (TAMs), comprising both resident microglia and bone marrow–derived macrophages, play a central role in supporting tumor growth, angiogenesis, and immune evasion. Most TAMs adopt an M2-like immunosuppressive phenotype, making them a promising target for immunomodulatory strategies in GBM. Method: According to PRISMA guidelines, we conducted a systematic literature review and recruited eligible studies focused on therapeutic approaches targeting TAMs in GBM, emphasizing mechanisms of action, efficacy, and challenges. Data extraction focused on therapeutic classes, outcomes, and TAM-related biomarkers. Results: We identified 30 studies meeting the inclusion criteria. These therapies are categorized into three main strategies: inhibition of TAM recruitment, enhancement of TAM-mediated phagocytosis, and reprogramming of TAMs. Combination strategies, including TAM-targeting with checkpoint inhibitors, nanoparticles, and oncolytic viruses, show synergistic effects in preclinical models. Conclusions: Targeting TAMs represents a multifaceted strategy for GBM treatment. Current evidence underscores the need for combination approaches integrating TAM modulation with existing standard-of-care therapies. Clinical translation remains limited due to challenges such as TAM heterogeneity, plasticity, immunosuppressive therapies, and restricted drug delivery across the blood–brain barrier. Future directions should highlight personalized treatments based on detailed TME profiling. Combining TAM-targeted therapies with agents modulating metabolic or immune pathways, and leveraging advanced delivery systems and spatial transcriptomics may improve efficacy. Full article
Show Figures

Figure 1

14 pages, 2613 KB  
Article
miR-7a-5p Contributes to Suppressing NLRP3/Caspase-1 Signaling Pathway in Response to Streptococcus suis Type 2 Infection
by Ziteng Deng, Qian Sun, Shun Li, Yibo Wang, Yuxin Che, Yunfei Huang, Jiedan Liao, Honglin Xie, Xiaoshu Zhan, Qinqin Sun and Qiang Fu
Microorganisms 2025, 13(8), 1924; https://doi.org/10.3390/microorganisms13081924 - 18 Aug 2025
Viewed by 313
Abstract
Streptococcus suis type 2 (SS2) is a pathogen causing diseases like meningitis and septicaemia worldwide. While microRNAs (miRNAs) are acknowledged for their role in post-transcriptional regulation of gene expression and influence on immune responses, their exact functions in hosts during SS2 infection remain [...] Read more.
Streptococcus suis type 2 (SS2) is a pathogen causing diseases like meningitis and septicaemia worldwide. While microRNAs (miRNAs) are acknowledged for their role in post-transcriptional regulation of gene expression and influence on immune responses, their exact functions in hosts during SS2 infection remain elusive. This study aims to explore the role of miR-7a-5p in macrophages during SS2 infection. Our findings reveal that SS2 infection in J774A.1 cells triggers upregulation of the NLRP3 inflammasome signaling pathways, evidenced by enhanced mRNA expression of pro-inflammatory cytokines (IL-6, IL-18, IL-23, TNF-α) and elevated protein levels of NLRP3, caspase-1, and IL-1β. Concurrently, SS2 infection reduces miR-7a-5p expression. Dual-luciferase reporter assays confirm that miR-7a-5p directly targets the 3′UTR of NLRP3 mRNA. Notably, miR-7a-5p overexpression in SS2-infected J774A.1 cells suppresses NLRP3 inflammasome activation and downstream signaling, as demonstrated by reduced mRNA levels of inflammatory mediators and decreased protein levels of NLRP3, caspase-1, IL-1β, and IL-18. Conversely, miR-7a-5p inhibition produces effects opposite to those of overexpression. In mice, administration of miR-7a-5p mimics mitigates SS2-induced lung, liver, and spleen damage, reducing histological scores in these organs. Collectively, these results show that miR-7a-5p alleviates SS2-induced inflammation by inhibiting the NLRP3 inflammasome, underscoring its potential as a therapeutic target for SS2-associated diseases. Full article
(This article belongs to the Section Veterinary Microbiology)
Show Figures

Figure 1

19 pages, 623 KB  
Review
Decoding Pancreatic Neuroendocrine Tumors: Molecular Profiles, Biomarkers, and Pathways to Personalized Therapy
by Linda Galasso, Federica Vitale, Gabriele Giansanti, Giorgio Esposto, Raffaele Borriello, Irene Mignini, Alberto Nicoletti, Lorenzo Zileri Dal Verme, Antonio Gasbarrini, Maria Elena Ainora and Maria Assunta Zocco
Int. J. Mol. Sci. 2025, 26(16), 7814; https://doi.org/10.3390/ijms26167814 - 13 Aug 2025
Viewed by 524
Abstract
Pancreatic neuroendocrine tumors (pNETs) are rare malignancies, accounting for 1–2% of pancreatic cancers, with an incidence of ≤1 case per 100,000 individuals annually. Originating from pancreatic endocrine cells, pNETs display significant clinical and biological heterogeneity. Traditional classification based on proliferative grading does not [...] Read more.
Pancreatic neuroendocrine tumors (pNETs) are rare malignancies, accounting for 1–2% of pancreatic cancers, with an incidence of ≤1 case per 100,000 individuals annually. Originating from pancreatic endocrine cells, pNETs display significant clinical and biological heterogeneity. Traditional classification based on proliferative grading does not fully capture the complex mechanisms involved, such as oxidative stress, mitochondrial dysfunction, and tumor-associated macrophage infiltration. Recent advances in molecular profiling have revealed key oncogenic drivers, including MEN1 (menin 1), DAXX (death domain–associated protein), ATRX (alpha thalassemia/mental retardation syndrome X-linked), CDKN1B (cyclin-dependent kinase inhibitor 1B) mutations, chromatin remodeling defects, and dysregulation of the mTOR pathway. Somatostatin receptors, particularly SSTR2, play a central role in tumor biology and serve as important prognostic markers, enabling the use of advanced diagnostic imaging (e.g., Gallium-68 DOTATATE PET/CT) and targeted therapies like somatostatin analogs and peptide receptor radionuclide therapy (PRRT). Established biomarkers such as Chromogranin A and the Ki-67 proliferation index remain vital for diagnosis and prognosis, while emerging markers, like circulating tumor DNA and microRNAs, show promise for enhancing disease monitoring and diagnostic accuracy. This review summarizes the molecular landscape of pNETs and highlights genomic, transcriptomic, proteomic, and epigenomic factors that support the identification of novel diagnostic, prognostic, and therapeutic biomarkers, ultimately advancing personalized treatment strategies. Full article
Show Figures

Figure 1

22 pages, 8184 KB  
Article
Porphyromonas gingivalis GroEL Accelerates Abdominal Aortic Aneurysm Formation by Induction of M1 Polarization in Macrophages
by Yi-Wen Lin, Yi-Ting Tsai, Ming-Jen Cheng, Chun-Ming Shih, Chun-Yao Huang, Chien-Sung Tsai, Shih-Ying Sung, Ze-Hao Lai, Chen-Wei Liu and Feng-Yen Lin
Int. J. Mol. Sci. 2025, 26(16), 7781; https://doi.org/10.3390/ijms26167781 - 12 Aug 2025
Viewed by 328
Abstract
Abdominal aortic aneurysm (AAA) is a life-threatening vascular disease characterized by chronic inflammation, extracellular matrix degradation, and smooth muscle cell apoptosis. Porphyromonas gingivalis (P. gingivalis), a key periodontal pathogen, has been implicated in the progression of cardiovascular diseases, including AAA, but [...] Read more.
Abdominal aortic aneurysm (AAA) is a life-threatening vascular disease characterized by chronic inflammation, extracellular matrix degradation, and smooth muscle cell apoptosis. Porphyromonas gingivalis (P. gingivalis), a key periodontal pathogen, has been implicated in the progression of cardiovascular diseases, including AAA, but the underlying mechanisms remain unclear. In this study, we investigated the role of GroEL, a bacterial heat shock protein 60 homolog derived from P. gingivalis, in AAA development. We employed a CaCl2-induced AAA mouse model to evaluate the in vivo effects of GroEL. Mice received periaortic CaCl2 application followed by intravenous injections of recombinant GroEL. Histological analyses were performed to assess aneurysmal dilation, elastin degradation, and inflammatory cell infiltration. Flow cytometry and immunohistochemistry were used to determine macrophage phenotypes, while cytokine profiles were quantified via ELISA. In vitro, THP-1 monocytes were treated with GroEL to evaluate its impact on macrophage polarization and cytokine expression. Our results showed that GroEL administration significantly enhanced aortic diameter expansion and elastin breakdown, accompanied by increased infiltration of M1-like macrophages and elevated levels of pro-inflammatory cytokines such as TNF-α and IL-6. In vitro findings confirmed that GroEL promotes M1 polarization and inhibits M2 marker expression in THP-1-derived macrophages. These findings suggest that P. gingivalis-derived GroEL plays a pathogenic role in AAA by modulating macrophage polarization toward a pro-inflammatory phenotype. Targeting microbial components such as GroEL may offer new therapeutic strategies for AAA management. Full article
Show Figures

Figure 1

20 pages, 2284 KB  
Article
Balancing the Cellular Inflammatory-Homeostatic Axis Through Natural Ingredient Supplementation
by Valentina Bordano, Chiara Gerbino, Valentina Boscaro, Patrizia Rubiolo, Arianna Marengo, Stefania Pizzimenti, Marie Angèle Cucci, Stefania Cannito, Jessica Nurcis, Margherita Gallicchio, Simona Federica Spampinato, Luigi Cangemi, Claudia Bocca, Chiara Dianzani, Arianna Carolina Rosa and Elisa Benetti
Nutrients 2025, 17(16), 2587; https://doi.org/10.3390/nu17162587 - 8 Aug 2025
Viewed by 448
Abstract
Background/Objectives: Dietary supplements are sources of nutrients or other substances that added to a healthy lifestyle help to preserve human homeostasis. Since inflammation is one of the major contributors to the alteration of homeostasis, this work investigated the effects of a multi-ingredient dietary [...] Read more.
Background/Objectives: Dietary supplements are sources of nutrients or other substances that added to a healthy lifestyle help to preserve human homeostasis. Since inflammation is one of the major contributors to the alteration of homeostasis, this work investigated the effects of a multi-ingredient dietary supplement on human macrophages, cells involved in the inflammatory response. Methods: THP-1 cells were differentiated into macrophage-like cells and polarized in M1 or M2 phenotypes. Cell migration was evaluated by Boyden chamber assay; phenotypic markers by qRT-PCR; cytokine release by ELISA and LPS/ATP-induced pyroptosis by LDH assay. The antioxidant properties of the supplement were evaluated in human and mouse fibroblasts by DCF-DA assay. After supplement treatment, cell extracts were analyzed by HPLC-PDA-MS/MS and GC-MS to evaluate the presence of the ingredients. Results: Our results showed that the dietary supplement promoted M2 migration and polarization and significantly reduced migration of M1. In a model of LPS-induced inflammation in M0, it significantly reduced NF-κB activation, COX-2 expression, and cytokine release. The supplement was not a specific inhibitor of NLRP-3, but it was able to modulate LPS priming. In addition, the supplement decreased granulocyte adhesion to HUVEC and reduced the oxidative stress in fibroblasts. The analysis of cell extracts showed the presence of the following ingredients of the formulation inside the cells: CoQ10, spermidine, resveratrol, 5-hydroxytryptophan from Griffonia simplicifolia (Vahl ex DC.) Baill., bacosides from Bacopa monnieri (L.) Wettst, vit B2, B5, E acetate. Conclusions: Our results demonstrate how a combination of natural active ingredients may contribute to the maintenance of homeostasis in human cells. Full article
Show Figures

Figure 1

31 pages, 721 KB  
Review
The Epigenetics of Sepsis: How Gene Modulation Shapes Outcomes
by Giulia Pignataro, Cristina Triunfo, Andrea Piccioni, Simona Racco, Mariella Fuorlo, Evelina Forte, Francesco Franceschi and Marcello Candelli
Biomedicines 2025, 13(8), 1936; https://doi.org/10.3390/biomedicines13081936 - 8 Aug 2025
Viewed by 619
Abstract
Sepsis is a complex and heterogeneous condition, arising from a disrupted immune response to infection that can progress to organ failure and carries a high risk of death. In recent years, growing attention has been paid to the role of epigenetic mechanisms—including DNA [...] Read more.
Sepsis is a complex and heterogeneous condition, arising from a disrupted immune response to infection that can progress to organ failure and carries a high risk of death. In recent years, growing attention has been paid to the role of epigenetic mechanisms—including DNA methylation, histone modifications, non-coding RNAs, and RNA methylation—in shaping immune activity during sepsis. These processes affect immune functions such as macrophage polarization, cytokine release, and the exhaustion of immune cells, and they help explain the shift from an initial phase of overwhelming inflammation to a later state of immune suppression. Epigenetic alterations also contribute to tissue-specific damage, notably in the lungs, kidneys, and heart, and have been linked to disease severity and clinical prognosis. Advances in transcriptomic and epigenetic profiling have made it possible to distinguish molecular subtypes of septic patients, each with distinct immune features and varied responses to treatments such as corticosteroids and metabolic therapies. Emerging biomarkers—like AQP5 methylation, histone lactylation (H3K18la), and m6A RNA methylation—are opening new options for patient classification and more tailored therapeutic strategies. This review examines the current understanding of how epigenetic regulation contributes to the pathophysiology of sepsis and considers its implications for developing more individualized approaches to care. Full article
Show Figures

Figure 1

4 pages, 5595 KB  
Correction
Correction: Zhu et al. HIF-1α-Overexpressing Mesenchymal Stem Cells Attenuate Colitis by Regulating M1-like Macrophages Polarization toward M2-like Macrophages. Biomedicines 2023, 11, 825
by Wenya Zhu, Qianqian Chen, Yi Li, Jun Wan, Jia Li and Shuai Tang
Biomedicines 2025, 13(8), 1903; https://doi.org/10.3390/biomedicines13081903 - 5 Aug 2025
Viewed by 183
Abstract
In the original publication [...] Full article
(This article belongs to the Section Cell Biology and Pathology)
Show Figures

Figure 4

23 pages, 3297 KB  
Article
Phenotypic Changes and Oxidative Stress in THP-1 Macrophages in Response to Vanilloids Following Stimulation with Allergen Act d 1 and LPS
by Milena Zlatanova, Jovana Grubač, Jovana Trbojević-Ivić and Marija Gavrović-Jankulović
Antioxidants 2025, 14(8), 949; https://doi.org/10.3390/antiox14080949 - 1 Aug 2025
Viewed by 609
Abstract
Activation of macrophages plays a key role in both inflammation and oxidative stress, key features of many chronic diseases. Pro-inflammatory M1-like macrophages, in particular, contribute to pro-oxidative environments and are a frequent focus of immunological research. This research examined the effects of kiwifruit [...] Read more.
Activation of macrophages plays a key role in both inflammation and oxidative stress, key features of many chronic diseases. Pro-inflammatory M1-like macrophages, in particular, contribute to pro-oxidative environments and are a frequent focus of immunological research. This research examined the effects of kiwifruit allergen Act d 1, in comparison to LPS, on THP-1 macrophages in vitro differentiated under optimized conditions, both in the presence and in the absence of selected vanilloids. THP-1 monocyte differentiation was optimized by varying PMA exposure and resting time. Act d 1 induced M1-like phenotypic changes comparable to LPS, including upregulation of CD80, IL-1β and IL-6 secretion, gene expression of iNOS and NF-κB activation, in addition to increased reactive oxygen species (ROS) and catalase activity. Treatment with specific vanilloids mitigated these responses, primarily through reduced oxidative stress and NF-κB activation. Notably, vanillin (VN) was the most effective, also reducing CD80 expression and IL-1β levels. These results suggest that vanilloids can affect pro-inflammatory signaling and oxidative stress in THP-1 macrophages and highlight their potential to alter inflammatory conditions characterized by similar immune responses. Full article
(This article belongs to the Section Natural and Synthetic Antioxidants)
Show Figures

Figure 1

19 pages, 10625 KB  
Article
SZC-6 Promotes Diabetic Wound Healing in Mice by Modulating the M1/M2 Macrophage Ratio and Inhibiting the MyD88/NF-χB Pathway
by Ang Xuan, Meng Liu, Lingli Zhang, Guoqing Lu, Hao Liu, Lishan Zheng, Juan Shen, Yong Zou and Shengyao Zhi
Pharmaceuticals 2025, 18(8), 1143; https://doi.org/10.3390/ph18081143 - 31 Jul 2025
Viewed by 577
Abstract
Background/Objectives: The prolonged M1-like pro-inflammatory polarization of macrophages is a key factor in the delayed healing of diabetic ulcers (DU). SIRT3, a primary mitochondrial deacetylase, has been identified as a regulator of inflammation and represents a promising new therapeutic target for DU [...] Read more.
Background/Objectives: The prolonged M1-like pro-inflammatory polarization of macrophages is a key factor in the delayed healing of diabetic ulcers (DU). SIRT3, a primary mitochondrial deacetylase, has been identified as a regulator of inflammation and represents a promising new therapeutic target for DU treatment. Nonetheless, the efficacy of existing SIRT3 agonists remains suboptimal. Methods: Here, we introduce a novel compound, SZC-6, demonstrating promising activity levels. Results: SZC-6 treatment down-regulated the expression of inflammatory factors in LPS-treated RAW264.7 cells and reduced the proportion of M1 macrophages. Mitosox, IF, and JC-1 staining revealed that SZC-6 preserved cellular mitochondrial homeostasis and reduced the accumulation of reactive oxygen species. In vivo experiments demonstrated that SZC-6 treatment accelerated wound healing in diabetic mice. Furthermore, HE and Masson staining revealed increased neovascularization at the wound site with SZC-6 treatment. Tissue immunofluorescence results indicated that SZC-6 effectively decreased the proportion of M1-like cells and increased the proportion of M2-like cells at the wound site. We also found that SZC-6 significantly reduced MyD88, p-IκBα, and NF-χB p65 protein levels and inhibited the nuclear translocation of P65 in LPS-treated cells. Conclusions: The study concluded that SZC-6 inhibited the activation of the NF-χB pathway, thereby reducing the inflammatory response and promoting skin healing in diabetic ulcers. SZC-6 shows promise as a small-molecule compound for promoting diabetic wound healing. Full article
(This article belongs to the Section Pharmacology)
Show Figures

Graphical abstract

18 pages, 2125 KB  
Article
A Replication-Defective Myxoma Virus Inducing Pro-Inflammatory Responses as Monotherapy and an Adjuvant to Chemo- and DC Immuno-Therapy for Ovarian Cancer
by Martin J. Cannon and Jia Liu
Viruses 2025, 17(8), 1058; https://doi.org/10.3390/v17081058 - 29 Jul 2025
Viewed by 542
Abstract
Myxoma virus (MYXV), a rabbit-specific poxvirus and non-pathogenic in humans and mice, is an excellent candidate oncolytic virus for cancer therapy. MYXV also has immunotherapeutic benefits. In ovarian cancer (OC), immunosuppressive tumor-associated macrophages (TAMs) are key to inhibiting antitumor immunity while hindering therapeutic [...] Read more.
Myxoma virus (MYXV), a rabbit-specific poxvirus and non-pathogenic in humans and mice, is an excellent candidate oncolytic virus for cancer therapy. MYXV also has immunotherapeutic benefits. In ovarian cancer (OC), immunosuppressive tumor-associated macrophages (TAMs) are key to inhibiting antitumor immunity while hindering therapeutic benefit by chemotherapy and dendritic cell (DC) vaccine. Because MYXV favors binding/entry of macrophages/monocytes, we examined the therapeutic potential of MYXV against TAMs. We found previously that a replication-defective MYXV with targeted deletion of an essential gene, M062R, designated ΔM062R MYXV, activated both the host DNA sensing pathway and the SAMD9 pathway. Treatment with ΔM062R confers therapeutic benefit comparable to that of wild-type replicating MYXV in preclinical models. Here we found that ΔM062R MYXV, when integrated with cisplatin and DC immunotherapy, further improved treatment benefit, likely through promoting tumor antigen-specific T cell function. Moreover, we also tested ΔM062R MYXV in targeting human immunosuppressive TAMs from OC patient ascites in a co-culture system. We found that ΔM062R treatment subverted the immunosuppressive properties of TAMs and elevated the avidity of cytokine production in tumor antigen-specific CD4+ T cells. Overall, ΔM062R presents a promising immunotherapeutic platform as a beneficial adjuvant to chemotherapy and DC vaccine. Full article
(This article belongs to the Special Issue Women in Virology 2025)
Show Figures

Figure 1

13 pages, 1842 KB  
Article
Pro-Inflammatory and Lipid Metabolism Dysregulating Effects of ANGPTL3 in THP-1 Macrophages
by Ilenia Milani, Ilaria Rossi, Giorgia Marodin, Maria Giovanna Lupo, Maria Pia Adorni, Francesca Zimetti and Nicola Ferri
Lipidology 2025, 2(3), 14; https://doi.org/10.3390/lipidology2030014 - 26 Jul 2025
Viewed by 432
Abstract
Background and aim: ANGPTL3 is a hepatokine acting as a negative regulator of lipoprotein lipase (LPL) through its N-terminal domain. Besides this activity, the C-terminal domain of ANGPTL3 interacts with integrin αVβ3. Since integrins are involved in inflammation and in the initiation of [...] Read more.
Background and aim: ANGPTL3 is a hepatokine acting as a negative regulator of lipoprotein lipase (LPL) through its N-terminal domain. Besides this activity, the C-terminal domain of ANGPTL3 interacts with integrin αVβ3. Since integrins are involved in inflammation and in the initiation of atherosclerotic plaque, the aim of our study was to evaluate the potential direct pro-inflammatory action of ANGPTL3 through the interaction of the fibrinogen-like domain and integrin αVβ3. Methods: We utilized cultured THP-1 human-derived macrophages and evaluated their pro-inflammatory phenotype in response to treatment with human recombinant ANGPTL3 (hANGPTL3). By Western blot, RT-qPCR, biochemical analysis, and ELISA assays, we determined the expression of genes and proteins involved in lipid metabolism and inflammatory response as well as intracellular cholesterol and triglyceride levels. In addition, we evaluated the effect of hANGPTL3 on the cellular cholesterol efflux process. Results: Incubation of THP-1-derived macrophages with 100 ng/mL of hANGPTL3 increased the mRNA expression of the pro-inflammatory cytokines IL-1β, IL-6, and TNFα (respectively, 1.87 ± 0.08-fold, 1.35 ± 0.11-fold, and 2.49 ± 0.43-fold vs. control). The secretion of TNFα, determined by an ELISA assay, was also induced by hANGPTL3 (1.98 ± 0.4-fold vs. control). The pro-inflammatory effect of hANGPTL3 was partially counteracted by co-treatment with the integrin αVβ3 inhibitor RGD peptide, reducing the mRNA levels of IL-1β (3.35 ± 0.35-fold vs. 2.54 ± 0.25-fold for hANGPTL3 vs. hANGPTL3 + RGD, respectively). Moreover, hANGPTL3 reduced cholesterol efflux to apoA-I, with a parallel increase in the intracellular triglyceride and cholesterol contents by 31.2 ± 2.8% and 20.0 ± 4.1%, respectively, compared to the control. Conclusions: ANGPTL3 is an important liver-derived regulator of plasma lipoprotein metabolism, and overall, our results add a new important pro-inflammatory activity of this circulating protein. This new function of ANGPTL3 could also be related to triglyceride and cholesterol accumulation into macrophages. Full article
(This article belongs to the Special Issue Lipid Metabolism and Inflammation-Related Diseases)
Show Figures

Figure 1

Back to TopTop