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
remove_circle_outline

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (395)

Search Parameters:
Keywords = inflamed tissue

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
37 pages, 4312 KiB  
Review
Neutrophils and NETs in Pathophysiology and Treatment of Inflammatory Bowel Disease
by Marina Ortega-Zapero, Raquel Gomez-Bris, Ines Pascual-Laguna, Angela Saez and Jose M. Gonzalez-Granado
Int. J. Mol. Sci. 2025, 26(15), 7098; https://doi.org/10.3390/ijms26157098 - 23 Jul 2025
Viewed by 482
Abstract
Inflammatory Bowel Disease (IBD), which includes ulcerative colitis (UC) and Crohn’s disease (CD), results from dysregulated immune responses that drive chronic intestinal inflammation. Neutrophils, as key effectors of the innate immune system, contribute to IBD through multiple mechanisms, including the release of reactive [...] Read more.
Inflammatory Bowel Disease (IBD), which includes ulcerative colitis (UC) and Crohn’s disease (CD), results from dysregulated immune responses that drive chronic intestinal inflammation. Neutrophils, as key effectors of the innate immune system, contribute to IBD through multiple mechanisms, including the release of reactive oxygen species (ROS), pro-inflammatory cytokines, and neutrophil extracellular traps (NETs). NETs are web-like structures composed of DNA, histones, and associated proteins including proteolytic enzymes and antimicrobial peptides. NET formation is increased in IBD and has a context-dependent role; under controlled conditions, NETs support antimicrobial defense and tissue repair, whereas excessive or dysregulated NETosis contributes to epithelial injury, barrier disruption, microbial imbalance, and thrombotic risk. This review examines the roles of neutrophils and NETs in IBD. We summarize recent single-cell and spatial-omics studies that reveal extensive neutrophil heterogeneity in the inflamed gut. We then address the dual role of neutrophils in promoting tissue damage—through cytokine release, immune cell recruitment, ROS production, and NET formation—and in supporting microbial clearance and mucosal healing. We also analyze the molecular mechanisms regulating NETosis, as well as the pathways involved in NET degradation and clearance. Focus is given to the ways in which NETs disrupt the epithelial barrier, remodel the extracellular matrix, contribute to thrombosis, and influence the gut microbiota. Finally, we discuss emerging therapeutic strategies aimed at restoring NET homeostasis—such as PAD4 inhibitors, NADPH oxidase and ROS pathway modulators, and DNase I—while emphasizing the need to preserve antimicrobial host defenses. Understanding neutrophil heterogeneity and NET-related functions may facilitate the development of new therapies and biomarkers for IBD, requiring improved detection tools and integrated multi-omics and clinical data. Full article
Show Figures

Figure 1

27 pages, 1269 KiB  
Review
Old and New Analgesic Acetaminophen: Pharmacological Mechanisms Compared with Non-Steroidal Anti-Inflammatory Drugs
by Hironori Tsuchiya and Maki Mizogami
Future Pharmacol. 2025, 5(3), 40; https://doi.org/10.3390/futurepharmacol5030040 - 22 Jul 2025
Viewed by 441
Abstract
Although it is more than a century since it was first marketed, acetaminophen remains one of the most popular analgesic agents. In addition, acetaminophen has recently been applied to multimodal analgesia in combination with non-steroidal anti-inflammatory drugs, and its consumption significantly increased during [...] Read more.
Although it is more than a century since it was first marketed, acetaminophen remains one of the most popular analgesic agents. In addition, acetaminophen has recently been applied to multimodal analgesia in combination with non-steroidal anti-inflammatory drugs, and its consumption significantly increased during the pandemic of coronavirus disease 2019 as well as diclofenac and ibuprofen. However, the detailed mode of analgesic action of acetaminophen is still unclear. In the present study, we comprehensively discuss conventional, recognized, and postulated mechanisms of analgesic acetaminophen and highlight the current mechanistic concepts while comparing with diclofenac and ibuprofen. Acetaminophen inhibits cyclooxygenase with selectivity for cyclooxygenase-2, which is higher than that of ibuprofen but lower than that of diclofenac. In contrast to diclofenac and ibuprofen, however, anti-inflammatory effects of acetaminophen depend on the extracellular conditions of inflamed tissues. Since the discovery of cyclooxygenase-3 in the canine brain, acetaminophen had been hypothesized to inhibit such a cyclooxygenase-1 variant selectively. However, this hypothesis was abandoned because cyclooxygenase-3 was revealed not to be physiologically and clinically relevant to humans. Recent studies suggest that acetaminophen is deacetylated to 4-aminophenol in the liver and after crossing the blood–brain barrier, it is metabolically converted into N-(4-hydroxyphenyl)arachidonoylamide. This metabolite exhibits bioactivities by targeting transient receptor potential vanilloid 1 channel, cannabinoid receptor 1, Cav3.2 calcium channel, anandamide, and cyclooxygenase, mediating acetaminophen analgesia. These targets may be partly associated with diclofenac and ibuprofen. The perspective of acetaminophen as a prodrug will be crucial for a future strategy to develop analgesics with higher tolerability and activity. Full article
Show Figures

Figure 1

15 pages, 1365 KiB  
Article
SUMOylation Regulates Neutrophil Phagocytosis and Migration
by Ran Zhang, Wanying Miao, Jin Zhang, Xinyuan Yu, Lihong Dang, Ata Ur Rehman, Feng Xu, Huaxin Sheng, G. Chad Hughes, Joseph P. Mathew, Jörn Karhausen and Wei Yang
Pharmaceuticals 2025, 18(7), 1070; https://doi.org/10.3390/ph18071070 - 20 Jul 2025
Viewed by 391
Abstract
Introduction: Accumulating evidence indicates that neutrophils undergo reprogramming of their effector functions as they migrate from the bloodstream into an inflamed tissue. Here, we examined the role of the small ubiquitin-like modifier (SUMO) conjugation in modulating neutrophil functional changes in the inflammatory [...] Read more.
Introduction: Accumulating evidence indicates that neutrophils undergo reprogramming of their effector functions as they migrate from the bloodstream into an inflamed tissue. Here, we examined the role of the small ubiquitin-like modifier (SUMO) conjugation in modulating neutrophil functional changes in the inflammatory microenvironment. Methods: Primary human and murine neutrophils were used to assess SUMOylation levels in vitro by Western blotting and results were validated in clinical samples from patients undergoing surgery involving hypothermic circulatory arrest. SUMOylation was inhibited with TAK-981, and its impact on neutrophil migration, NETosis, and phagocytosis was assessed in vitro. The in vivo effect of TAK-981 on neutrophil tissue infiltration was further evaluated using a sterile sponge assay in mice. Results: Our results demonstrated that neutrophil SUMOylation was induced by factors of the inflammatory microenvironment (temperature and oxidative stress) and inflammatory stimulants in vitro, and under conditions of general inflammatory activation in patients. Further, we found that blocking SUMOylation with TAK-981 in vitro blunted neutrophil migration and phagocytosis but did not affect NETosis. Notably, TAK-981 treatment reduced neutrophil accumulation in sterile sponges in mice. Conclusions: Our work identifies SUMOylation as a novel mechanism of neutrophil tissue reprogramming. Blocking SUMOylation may provide a therapeutic option to limit the contribution of neutrophils to inflammation-associated tissue damage. Full article
(This article belongs to the Section Pharmacology)
Show Figures

Figure 1

19 pages, 401 KiB  
Review
The Role of Protein Kinases in the Suppressive Phenotype of Myeloid-Derived Suppressor Cells
by Aikyn Kali, Nurshat Abdolla, Yuliya V. Perfilyeva, Yekaterina O. Ostapchuk and Raikhan Tleulieva
Int. J. Mol. Sci. 2025, 26(14), 6936; https://doi.org/10.3390/ijms26146936 - 19 Jul 2025
Viewed by 358
Abstract
Inflammation is a self-defense mechanism that controls the homeostasis of an organism, and its alteration by persistent noxious stimuli could lead to an imbalance in the regulation of inflammatory responses mediated by innate and adaptive immunity. During chronic inflammation, sustained exposure of myeloid [...] Read more.
Inflammation is a self-defense mechanism that controls the homeostasis of an organism, and its alteration by persistent noxious stimuli could lead to an imbalance in the regulation of inflammatory responses mediated by innate and adaptive immunity. During chronic inflammation, sustained exposure of myeloid cells to the various inflammatory signals derived from inflamed tissue could lead to the generation of myeloid cells with an immunosuppressive state, called myeloid-derived suppressor cells (MDSCs), which can exert protective or deleterious functions depending on the nature of signals and the specific inflammatory conditions created by different pathophysiological contexts. Initially identified in various tumor models and cancer patient samples, these cells have long been recognized as negative regulators of anti-tumor immunity. Consequently, researchers have focused on elucidating the molecular mechanisms underlying their potent immunosuppressive activity. As a key component of the signal transducing processes, protein kinases play a central role in regulating the signal transduction mechanisms of many cellular activities, including differentiation and immunosuppression. Over the past decade, at least a dozen kinases, including mechanistic target of rapamycin (mTOR), phosphoinositide 3-kinases (PI3Ks), TAM (Tyro3, Axl, Mer) family of receptor tyrosine kinases (TAM RTKs), mitogen-activated protein kinases (MAPKs), and others, have emerged as key contributors to the generation and differentiation of MDSCs. Here, we discuss the recent findings on these kinases that directly contribute to the immunosuppressive functions of MDSCs. Full article
(This article belongs to the Section Molecular Immunology)
Show Figures

Figure 1

12 pages, 3566 KiB  
Article
Differential Regulation of Angiogenesis, Lymphangiogenesis, and Neural Tissue in Normal and Inflamed Dental Pulp: Immunohistochemical Analysis
by Nooruldeen Ammar Alani and Bashar Hamid Abdullah
Diagnostics 2025, 15(14), 1819; https://doi.org/10.3390/diagnostics15141819 - 19 Jul 2025
Viewed by 378
Abstract
Background/Objectives: Pulp inflammation impairs healing, yet the underlying vascular and neural mechanisms remain poorly understood. This study investigated the differential regulation of lymphatic vessels, blood vessels, and neural tissue in pulpitis to elucidate healing limitations in inflamed dental pulp. Methods: This study evaluated [...] Read more.
Background/Objectives: Pulp inflammation impairs healing, yet the underlying vascular and neural mechanisms remain poorly understood. This study investigated the differential regulation of lymphatic vessels, blood vessels, and neural tissue in pulpitis to elucidate healing limitations in inflamed dental pulp. Methods: This study evaluated 38 pulp samples (14 symptomatic irreversible pulpitis, 13 asymptomatic irreversible pulpitis, and 11 healthy controls) via immunohistochemistry, using D2-40 to identify lymphatic vessels, CD31 to mark blood vessels, and PGP9.5 to detect neural tissue. Vessel counts and neural tissue scoring were performed by blinded examiners and analyzed using appropriate statistical tests. Results: Dental pulp with symptomatic irreversible pulpitis exhibited significantly increased blood vessel density (50.3 vs. 39.2 in asymptomatic irreversible pulpitis and 25.8 in controls, p = 0.001, Cohen’s d = 1.82), while lymphatic vessel density remained unchanged across all groups (p ≥ 0.05), indicating impaired lymphangiogenesis despite inflammation. Neural tissue density was consistent across conditions, with a significant negative correlation between PGP9.5 expression and age (r = −0.5, p = 0.001). CD31 and D2-40 expression showed a positive correlation (r = 0.389, p = 0.016), suggesting coordinated vascular development. Conclusions: Our findings reveal a critical imbalance between enhanced angiogenesis and impaired lymphangiogenesis during pulpitis, potentially explaining the compromised healing capacity of inflamed dental pulp. This vascular dysregulation, combined with persistent neural tissue density, creates an environment in which inflammatory exudates accumulate with limited clearance. These insights indicate a need for new therapeutic strategies aimed at enhancing lymphangiogenesis to improve endodontic outcomes. Full article
(This article belongs to the Section Pathology and Molecular Diagnostics)
Show Figures

Figure 1

28 pages, 5716 KiB  
Article
Novel Benzenesulfonamide Derivatives of 5′-Aminospirotriazolotriazine Exhibit Anti-Inflammatory Activity by Suppressing Pro-Inflammatory Mediators: In Vitro and In Vivo Evaluation Using a Rat Model of Carrageenan-Induced Paw Edema
by Amany M. Hamed, Souhaila S. Enaili, Walaa I. Mohammed, Azza M. A. Abouelella, Zeyad Elsayed Eldeeb Mohana, Dina M. Monir, Safaa S. Soliman, Elsayed Eldeeb Mehana Hamouda, Hytham Mahmoud Abd Elatif and Ahmed M. El-Saghier
Biomedicines 2025, 13(7), 1732; https://doi.org/10.3390/biomedicines13071732 - 15 Jul 2025
Viewed by 832
Abstract
Background/Objectives: Inflammation is a crucial and complex mechanism that protects the body against infections. In our study, we propose to provide scientific evidence for the anti-inflammatory properties of 1,3,5-triazine derivatives. Methods: Initially, we ensured the safety of the three synthesized derivatives [...] Read more.
Background/Objectives: Inflammation is a crucial and complex mechanism that protects the body against infections. In our study, we propose to provide scientific evidence for the anti-inflammatory properties of 1,3,5-triazine derivatives. Methods: Initially, we ensured the safety of the three synthesized derivatives by administering graded doses of up to 2000 mg/kg intraperitoneally in Wistar rats. Thus, the three derivatives were considered generally safe. We also evaluated their ability to reduce carrageenan-induced rat paw edema. Results: Compounds 1, 2, and 3 demonstrated stronger anti-inflammatory activity than indomethacin (10 mg/kg), achieving maximum inhibition at the fourth hour with percentages of 96.31%, 72.08%, and 99.69%, respectively, at a dose of 200 mg/kg, compared to 57.66% for the standard drug. To explore the mechanism, levels of pro-inflammatory cytokines (TNF-α, IL-1α, IL-1β, IL-6, CRP) and oxidative stress markers were measured in paw tissue. All three compounds significantly reduced these markers more effectively than indomethacin and enhanced antioxidant levels (SOD and GSH) beyond those achieved by the standard treatment. Additionally, the compounds reduced COX-1 and COX-2 levels to values comparable to those in the normal (non-inflamed) control group. Conclusions: Compounds 1, 2, and 3 at doses of 200 mg/kg significantly (p  < 0.05) inhibited the heat-induced hemolysis of red blood cell (RBC) membranes by 94.6%, 93.9%, and 95.2%, respectively, compared to 94.5% produced by indomethacin. Consequently, we concluded that 1,3,5-triazine derivatives are a safe antioxidant agent with significant anti-inflammatory activity. Full article
(This article belongs to the Section Drug Discovery, Development and Delivery)
Show Figures

Figure 1

13 pages, 3325 KiB  
Article
microRNA-200c Mitigates Pulpitis and Promotes Dentin Regeneration
by Tadkamol Krongbaramee, Chawin Upara, Matthew T. Remy, Long Jiang, Jue Hu, Kittiphoj Tikkhanarak, Bruno Cavalcanti, Hongli Sun, Fabricio B. Teixeira and Liu Hong
Int. J. Mol. Sci. 2025, 26(14), 6734; https://doi.org/10.3390/ijms26146734 - 14 Jul 2025
Viewed by 260
Abstract
MicroRNA (miR)-200c enhances osteogenesis, modulates inflammation, and participates in dentin development. This study was to investigate the beneficial potential of miR-200c in vital pulp therapy (VPT) by mitigating pulpitis and promoting dentin regeneration. We explored the miR-200c variations in inflamed pulp tissues from [...] Read more.
MicroRNA (miR)-200c enhances osteogenesis, modulates inflammation, and participates in dentin development. This study was to investigate the beneficial potential of miR-200c in vital pulp therapy (VPT) by mitigating pulpitis and promoting dentin regeneration. We explored the miR-200c variations in inflamed pulp tissues from patients with symptomatic irreversible pulpitis and primary human dental pulp-derived cells (DPCs) challenged with P.g. lipopolysaccharide (Pg-LPS). We further assessed the functions of overexpression of miR-200c on odontogenic differentiation, pulpal inflammation, and dentin regeneration in vitro and in vivo. Our findings revealed a noteworthy downregulation of miR-200c expression in inflamed pulp tissues and primary human DPCs. Through the overexpression of miR-200c via transfecting plasmid DNA (pDNA), we observed a substantial downregulation of proinflammatory cytokines interleukin (IL)-6 and IL-8 in human DPCs. Furthermore, this overexpression significantly enhanced the transcript and protein levels of odontogenic differentiation markers, including Runt-related transcription factor (Runx)2, osteocalcin (OCN), dentin matrix protein (DMP)1, and dentin sialophosphoprotein (DSPP). In a rat model of pulpitis induced by Pg-LPS, we demonstrated notable benefits by local application of pDNA encoding miR-200c delivered by CaCO3-based nanoparticles to reduce pulpal inflammation and promote dentin formation. These results underscore the significant impact of locally applied miR-200c in modulating pulpal inflammation and facilitating dentin repair, showcasing its ability to improve VPT outcomes. Full article
(This article belongs to the Section Molecular Biology)
Show Figures

Figure 1

26 pages, 1980 KiB  
Review
The Destructive Cycle in Bronchopulmonary Dysplasia: The Rationale for Systems Pharmacology Therapeutics
by Mia Teng, Tzong-Jin Wu, Kirkwood A. Pritchard, Billy W. Day, Stephen Naylor and Ru-Jeng Teng
Antioxidants 2025, 14(7), 844; https://doi.org/10.3390/antiox14070844 - 10 Jul 2025
Viewed by 516
Abstract
Bronchopulmonary dysplasia (BPD) remains a significant complication of premature birth and neonatal intensive care. While much is known about the drivers of lung injury, few studies have addressed the interrelationships between oxidative stress, inflammation, and downstream events, such as endoplasmic reticulum (ER) stress. [...] Read more.
Bronchopulmonary dysplasia (BPD) remains a significant complication of premature birth and neonatal intensive care. While much is known about the drivers of lung injury, few studies have addressed the interrelationships between oxidative stress, inflammation, and downstream events, such as endoplasmic reticulum (ER) stress. In this review, we explore the concept of a “destructive cycle” in which these drivers self-amplify to push the lung into a state of maladaptive repair. Animal models, primarily the hyperoxic rat pup model, support a sequential progression from the generation of reactive oxygen species (ROS) and inflammation to endoplasmic reticulum (ER) stress and mitochondrial injury. We highlight how these intersecting pathways offer not just therapeutic targets but also opportunities for interventions that reprogram system-wide responses. Accordingly, we explore the potential of systems pharmacology therapeutics (SPTs) to address the multifactorial nature of BPD. As a prototype SPT, we describe the development of N-acetyl-L-lysyl-L-tyrosyl-L-cysteine amide (KYC), a systems chemico-pharmacology drug (SCPD), which is selectively activated in inflamed tissues and modulates key nodal targets such as high-mobility group box-1 (HMGB1) and Kelch-like ECH-associated protein-1 (Keap1). Collectively, the data suggest that future therapies may require a coordinated, network-level approach to break the destructive cycle and enable proper regeneration rather than partial repair. Full article
(This article belongs to the Special Issue Oxidative Stress in the Newborn)
Show Figures

Figure 1

13 pages, 653 KiB  
Review
Topical Percutaneous Drug Delivery for Allergic Diseases: A Novel Strategy for Site-Directed Pharmacologic Modulation
by Mitsuhiro Kamimura, Hiroaki Todo, Kenji Sugibayashi and Koichiro Asano
Pharmaceutics 2025, 17(7), 867; https://doi.org/10.3390/pharmaceutics17070867 - 2 Jul 2025
Viewed by 336
Abstract
Topical percutaneous drug delivery has recently emerged as a novel strategy for the treatment of allergic diseases, offering targeted drug delivery to mucosal tissues adjacent to the skin. Unlike conventional topical approaches that act on the skin surface or mucosal membranes, topical percutaneous [...] Read more.
Topical percutaneous drug delivery has recently emerged as a novel strategy for the treatment of allergic diseases, offering targeted drug delivery to mucosal tissues adjacent to the skin. Unlike conventional topical approaches that act on the skin surface or mucosal membranes, topical percutaneous drug delivery enables non-invasive pharmacologic modulation of deeper structures such as the conjunctiva, nasal mucosa, and trachea. This review explores the rationale, pharmacokinetic foundation, clinical data, and future prospects of transdermal therapy in allergic conjunctivitis, allergic rhinitis, and asthma-related cough. In allergic conjunctivitis, eyelid-based transdermal delivery of antihistamines such as diphenhydramine and epinastine has shown rapid and long-lasting symptom relief, with epinastine cream recently approved in Japan following a randomized controlled trial (RCT) demonstrating its efficacy. Preclinical and clinical pharmacokinetic studies support the eyelid’s unique permeability and sustained drug release profile, reinforcing its utility as a delivery site for ocular therapies. In allergic rhinitis, diphenhydramine application to the nasal ala demonstrated symptomatic improvement in patients intolerant to intranasal therapies, though anatomical separation from the inflamed turbinates may limit consistent efficacy. Similarly, cervical tracheal application of steroids and antihistamines has shown potential benefit in asthma-related cough, especially for patients refractory to inhaled treatments, despite anatomical and depth-related limitations. Overall, site-specific anatomy, skin permeability, and disease localization are critical factors in determining therapeutic outcomes. While trans-eyelid therapy is supported by robust data, studies on the nasal ala and trachea remain limited to small-scale pilot trials. No major adverse events have been reported with nasal or tracheal application, but eyelid sensitivity requires formulation caution. To validate this promising modality, further RCTs, pharmacokinetic analyses, and formulation optimization are warranted. Topical percutaneous drug delivery holds potential as a non-invasive, site-directed alternative for managing allergic diseases beyond dermatologic indications. Full article
(This article belongs to the Section Drug Delivery and Controlled Release)
Show Figures

Figure 1

18 pages, 3098 KiB  
Article
(-)-Epigallocatechin-3-Gallate Suppresses Hyperexcitability in Rat Primary Nociceptive Neurons Innervating Inflamed Tissues: A Comparison with Lidocaine
by Syogo Utugi, Yukito Sashide and Mamoru Takeda
Metabolites 2025, 15(7), 439; https://doi.org/10.3390/metabo15070439 - 1 Jul 2025
Viewed by 329
Abstract
Objective: Given the side effects and reduced efficacy of conventional local anesthetics in inflammatory conditions, there is a compelling need for complementary alternative medicine (CAM), particularly those based on phytochemicals. While a previous study showed that in vivo local injection of (-)-epigallocatechin-3-gallate (EGCG) [...] Read more.
Objective: Given the side effects and reduced efficacy of conventional local anesthetics in inflammatory conditions, there is a compelling need for complementary alternative medicine (CAM), particularly those based on phytochemicals. While a previous study showed that in vivo local injection of (-)-epigallocatechin-3-gallate (EGCG) into the peripheral receptive field suppresses the excitability of rat trigeminal ganglion (TG) neurons in the absence of inflammation, the acute effects of EGCG in vivo, especially on TG neurons under inflammatory conditions, are still unknown. We aimed to determine if acute local EGCG administration into inflamed tissue effectively attenuates the excitability of nociceptive TG neurons evoked by mechanical stimulation. Methods: The escape reflex threshold was measured to assess hyperalgesia caused by complete Freund’s adjuvant (CFA)-induced inflammation. To assess neuronal activity, extracellular single-unit recordings were performed on TG neurons in anesthetized CFA-inflamed rats in response to orofacial mechanical stimulation. Results: The mechanical escape threshold was significantly lower in CFA-inflamed rats compared to before CFA injection. EGCG (1–10 mM) reversibly and dose-dependently inhibited the mean firing frequency of TG neurons evoked by both non-noxious and noxious mechanical stimuli (p < 0.05). For comparison, 1% lidocaine (37 mM), a local anesthetic, also caused reversible inhibition of the mean firing frequency in inflamed TG neurons responding to mechanical stimuli. Importantly, 10 mM EGCG produced a significantly greater magnitude of inhibition on TG neuronal discharge frequency than 1% lidocaine (noxious, lidocaine vs. EGCG, 19.7 ± 3.3% vs. 42.3 ± 3.4%, p < 0.05). Conclusions: Local injection of EGCG into inflamed tissue effectively suppresses the excitability of nociceptive primary sensory TG neurons, as indicated by these findings. Significantly, locally administered EGCG exerted a more potent local analgesic action compared to conventional voltage-gated sodium channel blockers. This heightened efficacy originates from EGCG’s ability to inhibit both generator potentials and action potentials directly at nociceptive primary nerve terminals. As a result, EGCG stands out as a compelling candidate for novel analgesic development, holding particular relevance for CAM strategies. Full article
(This article belongs to the Special Issue Flavonoids: Novel Therapeutic Potential for Chronic Diseases)
Show Figures

Figure 1

18 pages, 996 KiB  
Review
Future Perspectives and Conclusions from Animal Models of CHI3L1-Related Inflammation-Associated Cancer
by Emiko Mizoguchi and Siyuan Wang
Cells 2025, 14(13), 982; https://doi.org/10.3390/cells14130982 - 26 Jun 2025
Viewed by 645
Abstract
Among the molecules implicated in inflammation-associated tumorigenesis, Chitinase 3-like 1 (CHI3L1/YKL-40/Brp-39) has emerged as a particularly compelling target due to its multifaced roles in immune regulation, tissue remodeling, and cancer progression. Elevated CHI3L1 expression is observed in various human cancers and corresponding animal [...] Read more.
Among the molecules implicated in inflammation-associated tumorigenesis, Chitinase 3-like 1 (CHI3L1/YKL-40/Brp-39) has emerged as a particularly compelling target due to its multifaced roles in immune regulation, tissue remodeling, and cancer progression. Elevated CHI3L1 expression is observed in various human cancers and corresponding animal models. CHI3L1 directly promotes tumor cell proliferation and angiogenesis and also contributes to immune evasion by establishing an immunosuppressive environment in inflamed tissues. Mechanistically, CHI3L1 exerts its effects through the modulation of STAT3, MAPK, and PI3K/Akt signaling pathways and by interacting with cell surface receptors, such as IL-13Rα2 and RAGE. Studies using transgenic and knockout mouse models have revealed a strong association between CHI3L1 expression and cancer progression. In models of colon and lung cancer, CHI3L1 overexpression correlates with increased tumor size and number, whereas CHI3L1 deficiency markedly suppresses tumor formation. However, its involvement appears to be context-dependent and varies among different epithelial tumor types. These findings suggest that CHI3L1 is a potential therapeutic target and diagnostic biomarker for inflammation-associated cancers. Animal studies provide valuable insights into the immunological mechanisms of CHI3L1-mediated tumorigenesis but also highlight the need for cautious interpretation due to inherent technical limitations. Full article
(This article belongs to the Special Issue Pathogenic Mechanisms of Chronic Inflammation-Associated Cancer)
Show Figures

Figure 1

25 pages, 1575 KiB  
Review
Galectin-3—Insights from Inflammatory Bowel Disease and Primary Sclerosing Cholangitis
by Thomas Grewal, Hauke Christian Tews and Christa Buechler
Int. J. Mol. Sci. 2025, 26(13), 6101; https://doi.org/10.3390/ijms26136101 - 25 Jun 2025
Viewed by 609
Abstract
Inflammatory bowel disease (IBD) and primary sclerosing cholangitis (PSC) are related diseases with poorly understood pathophysiology. While therapy options for IBD have increased, treatment options for PSC remain limited. Galectin-3 is a multifunctional lectin expressed in intestinal epithelial cells, and is abundant in [...] Read more.
Inflammatory bowel disease (IBD) and primary sclerosing cholangitis (PSC) are related diseases with poorly understood pathophysiology. While therapy options for IBD have increased, treatment options for PSC remain limited. Galectin-3 is a multifunctional lectin expressed in intestinal epithelial cells, and is abundant in immune cells such as macrophages, with roles in cell adhesion, apoptosis, inflammation and fibrosis being associated with IBD and PSC disease development and progression. In addition, galectin-3 is also a visceral fat-derived protein whose systemic levels are increased in obese individuals, the latter correlating with a poorer prognosis in IBD and PSC patients. On the other hand, decreased galectin-3 expression in the inflamed mucosal tissues of mice and patients with IBD possibly indicate a protective role of this lectin in IBD. However, galectin-3 loss or inhibition is protective in most animal models of liver fibrosis but exacerbates the severity of autoimmune liver disease. Hence, with PSC being a slowly progressing autoimmune hepatobiliary disease closely related to IBD, further studies evaluating galectin-3 as a therapeutic target or biomarker for the severity of IBD and the occurrence of PSC are still needed. This review summarizes studies that have analyzed expression patterns and functions of galectin-3 in IBD and PSC. Current evidence suggests that strategies to block galectin-3 are not advised for patients with IBD and PSC-IBD. Full article
(This article belongs to the Special Issue Galectins (Gals), 2nd Edition)
Show Figures

Figure 1

9 pages, 731 KiB  
Communication
Protective Effects of Pasireotide in LPS-Induced Acute Lung Injury
by Saikat Fakir, Md Matiur Rahman Sarker, Madan Sigdel and Nektarios Barabutis
Pharmaceuticals 2025, 18(7), 942; https://doi.org/10.3390/ph18070942 - 22 Jun 2025
Viewed by 528
Abstract
Background/Objectives: Acute lung injury (ALI) is an inflammatory condition characterized by tissue barrier damage, which leads to vascular leakage, pulmonary edema, and compromised gas exchange. Lipopolysaccharides (LPS) are a component of Gram-negative bacteria, which trigger inflammation by Toll-like receptor 4 (TLR4) activation. Herein, [...] Read more.
Background/Objectives: Acute lung injury (ALI) is an inflammatory condition characterized by tissue barrier damage, which leads to vascular leakage, pulmonary edema, and compromised gas exchange. Lipopolysaccharides (LPS) are a component of Gram-negative bacteria, which trigger inflammation by Toll-like receptor 4 (TLR4) activation. Herein, we investigated the possibility that Pasireotide (PAS) exerts protective effects in an experimental model of ALI. Methods: C57BL/6 male mice received an intratracheal injection of saline or LPS, followed by PAS or vehicle treatment. Bronchoalveolar lavage fluid (BALF) was collected via tracheal catheterization, and Western blot analysis was used to detect protein expression variations. Results: Our results suggest that PAS treatment alleviates LPS-induced mouse lung injury and inflammation. JAK/STAT and MAPK activation levels in the inflamed lungs were suppressed due to PAS treatment, as well as BALF protein concentration. Additionally, PAS counteracted LPS-induced Grp94 protein reduction, suggesting the involvement of ATF6 in PAS-triggered barrier-protective effects. Grp94 is a downstream ATF6 target. Conclusions: Our data demonstrate that PAS protects mouse lungs against LPS in an experimental model of ALI. Full article
(This article belongs to the Section Pharmacology)
Show Figures

Figure 1

20 pages, 3957 KiB  
Article
Selective Blockade of Two Aquaporin Channels, AQP3 and AQP9, Impairs Human Leukocyte Migration
by Sabino Garra, Charlotte Mejlstrup Hymøller, Daria Di Molfetta, Nicola Zagaria, Patrizia Gena, Rosa Angela Cardone, Michael Rützler, Svend Birkelund and Giuseppe Calamita
Cells 2025, 14(12), 880; https://doi.org/10.3390/cells14120880 - 11 Jun 2025
Cited by 1 | Viewed by 534
Abstract
Peripheral blood leukocytes are able to migrate to the inflamed tissue, and to engulf and kill invading microbes. This requires rapid modifications of cell morphology and volume through fast movements of osmotic water into or out of the cell. In this process, membrane [...] Read more.
Peripheral blood leukocytes are able to migrate to the inflamed tissue, and to engulf and kill invading microbes. This requires rapid modifications of cell morphology and volume through fast movements of osmotic water into or out of the cell. In this process, membrane water channels, aquaporins (AQPs), are critical for cell shape changes as AQP-mediated water movement indirectly affects the cell cytoskeleton and, thereby, the signaling cascades. Recent studies have shown that the deletion or gating of two immune cell AQPs, AQP3 and AQP9, impairs inflammation and improves survival in microbial sepsis. Here, we assessed the expression and distribution of AQP3 and AQP9 in human leukocytes and investigated their involvement in the phagocytosis and killing of the Gram-negative pathogenic bacterium Klebsiella pneumoniae, and their role in lipopolysaccharide (LPS)-induced cell migration. By RT-qPCR, AQP3 mRNA was found in peripheral blood mononuclear cells (PBMCs) but it was undetectable in polymorphonuclear white blood cells (PMNs). AQP9 was found both in PBMCs and PMNs, particularly in neutrophil granulocytes. Immunofluorescence confirmed the AQP3 expression in monocytes and, to a lesser degree, in lymphocytes. AQP9 was expressed both in PBMCs and neutrophils. Specific inhibitors of AQP3 (DFP00173) and AQP9 (HTS13286 and RG100204) were used for bacterial phagocytosis and killing studies. No apparent involvement of individually blocked AQP3 or AQP9 was observed in the phagocytosis of K. pneumoniae by neutrophils or monocytes after 10, 30, or 60 min of bacterial infection. A significant impairment in the phagocytic capacity of monocytes but not neutrophils was observed only when both AQPs were inhibited simultaneously and when the infection lasted for 60 min. No impairment in bacterial clearance was found when AQP3 and AQP9 were individually or simultaneously blocked. PBMC migration was significantly impaired after exposure to the AQP9 blocker RG100204 in the presence or absence of LPS. The AQP3 inhibitor DFP00173 reduced PBMC migration only under LPS exposure. Neutrophil migration was considerably reduced in the presence of RG100204 regardless of whether there was an LPS challenge or not. Taken together, these results indicate critical but distinct involvements for AQP3 and AQP9 in leukocyte motility, while no roles are played in bacterial killing. Further studies are needed in order to understand the precise ways in which these two AQPs intervene during bacterial infections. Full article
Show Figures

Figure 1

23 pages, 8153 KiB  
Article
SARS-Cov-2 Replication in a Blood–Brain Barrier Model Established with Human Brain Microvascular Endothelial Cells Induces Permeability and Disables ACE2-Dependent Regulation of Bradykinin B1 Receptor
by Sharton Vinicius Antunes Coelho, Gabriela Lisboa e Souza, Bruno Braz Bezerra, Luan Rocha Lima, Isadora Alonso Correa, Dalziza Victalina de Almeida, Rodrigo Pacheco da Silva-Aguiar, Ana Acácia S. Pinheiro, Pierre Sirois, Celso Caruso-Neves, Luciana Jesus da Costa, Julio Scharfstein and Luciana Barros de Arruda
Int. J. Mol. Sci. 2025, 26(12), 5540; https://doi.org/10.3390/ijms26125540 - 10 Jun 2025
Viewed by 764
Abstract
Endothelial dysfunction plays a central role in COVID-19 pathogenesis, by affecting vascular homeostasis and worsening thromboinflammation. This imbalance may contribute to blood–brain barrier (BBB) disruption, which has been reported in long COVID-19 patients with neurological sequelae. The kallikrein–kinin system (KKS) generates bradykinin (BK), [...] Read more.
Endothelial dysfunction plays a central role in COVID-19 pathogenesis, by affecting vascular homeostasis and worsening thromboinflammation. This imbalance may contribute to blood–brain barrier (BBB) disruption, which has been reported in long COVID-19 patients with neurological sequelae. The kallikrein–kinin system (KKS) generates bradykinin (BK), a proinflammatory peptide that induces microvascular leakage via B2R. Under inflammatory conditions, BK is converted to Des-Arg-BK (DABK), which activates B1R, a receptor upregulated in inflamed tissues. DABK is degraded by ACE2, the main SARS-CoV-2 receptor; thus, viral binding and ACE2 downregulation may lead to DABK/B1R imbalance. Here, we investigated these interactions using human brain microvascular endothelial cells (HBMECs), as a model of the BBB. Since endothelial cell lines express low levels of ACE2, HBMECs were modified with an ACE2-carrying pseudovirus. SARS-CoV-2 replication was confirmed by RNA, protein expression, and infectious particles release. Infection upregulated cytokines and endothelial permeability, enhancing viral and leukocyte transmigration. Additionally, viral replication impaired ACE2 function in HBMECs, amplifying the response to DABK, increasing nitric oxide (NO) production, and further disrupting endothelial integrity. Our findings reveal a mechanism by which SARS-CoV-2 impacts the BBB and highlights the ACE2/KKS/B1R axis as a potential contributor to long COVID-19 neurological symptoms. Full article
Show Figures

Figure 1

Back to TopTop