Nothing Special   »   [go: up one dir, main page]

You seem to have javascript disabled. Please note that many of the page functionalities won't work as expected without javascript enabled.
 
 
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
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
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
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
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (11,741)

Search Parameters:
Keywords = clinical pathways

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
26 pages, 2259 KiB  
Article
MGST1 Protects Pancreatic Ductal Cells from Inflammatory Damage in Acute Pancreatitis by Inhibiting Ferroptosis: Bioinformatics Analysis with Experimental Validation
by Ruoyi Zhang, Xin Ling, Xianwen Guo and Zhen Ding
Int. J. Mol. Sci. 2025, 26(5), 1899; https://doi.org/10.3390/ijms26051899 (registering DOI) - 22 Feb 2025
Abstract
Numerous animal experiments have implicated ferroptosis in the pathogenesis of acute pancreatitis (AP). Nonetheless, due to sampling constraints, the precise role of ferroptosis in the human body during AP remains elusive. Method: Peripheral blood sequencing data of patients with acute pancreatitis (GSE194331) were [...] Read more.
Numerous animal experiments have implicated ferroptosis in the pathogenesis of acute pancreatitis (AP). Nonetheless, due to sampling constraints, the precise role of ferroptosis in the human body during AP remains elusive. Method: Peripheral blood sequencing data of patients with acute pancreatitis (GSE194331) were obtained from the Gene Expression Omnibus (GEO) database. We analyzed differentially expressed genes whose expression increased or decreased with increasing disease severity and intersected them with the ferroptosis gene set to identify ferroptosis-related driver genes for the disease. The hub genes were selected using machine learning algorithms, and a nomogram diagnosis model was constructed. Clinical samples, animal models, and an in vitro experiment were also used for validation. The investigation unveiled 22 ferroptosis-related driver genes, and we identified three hub genes, AQP3, TRIB2, and MGST1, by employing two machine learning algorithms. AQP3 and TRIB2 exhibit robust correlations with various immune cells. The disease diagnosis model constructed utilizing these three genes demonstrated high sensitivity and specificity (AUC = 0.889). In the in vitro experiments, we discovered for the first time that ferroptosis occurs in pancreatic duct cells during acute pancreatitis, and that MGST1 is significantly upregulated in duct cells, where it plays a crucial role in negatively regulating ferroptosis via the ACSL4/GPX4 axis. In addition, overexpression of MGST1 protects ductal cells from inflammatory damage. In our investigation, we explored the mechanisms of ferroptosis in immune cells and pancreatic duct cells in patients with AP. These results highlight a potential pathway for the early diagnosis and treatment of acute pancreatitis. Full article
(This article belongs to the Section Molecular Immunology)
38 pages, 1818 KiB  
Review
Sensory Dysfunction in ALS and Other Motor Neuron Diseases: Clinical Relevance, Histopathology, Neurophysiology, and Insights from Neuroimaging
by Jana Kleinerova, Rangariroyashe H. Chipika, Ee Ling Tan, Yana Yunusova, Véronique Marchand-Pauvert, Jan Kassubek, Pierre-Francois Pradat and Peter Bede
Biomedicines 2025, 13(3), 559; https://doi.org/10.3390/biomedicines13030559 (registering DOI) - 22 Feb 2025
Viewed by 89
Abstract
Background: The clinical profiles of MNDs are dominated by inexorable motor decline, but subclinical proprioceptive, nociceptive and somatosensory deficits may also exacerbate mobility, dexterity, and bulbar function. While extra-motor pathology and frontotemporal involvement are widely recognised in motor neuron diseases (MNDs), reports [...] Read more.
Background: The clinical profiles of MNDs are dominated by inexorable motor decline, but subclinical proprioceptive, nociceptive and somatosensory deficits may also exacerbate mobility, dexterity, and bulbar function. While extra-motor pathology and frontotemporal involvement are widely recognised in motor neuron diseases (MNDs), reports of sensory involvement are conflicting. The potential contribution of sensory deficits to clinical disability is not firmly established and the spectrum of sensory manifestations is poorly characterised. Methods: A systematic review was conducted to examine the clinical, neuroimaging, electrophysiology and neuropathology evidence for sensory dysfunction in MND phenotypes. Results: In ALS, paraesthesia, pain, proprioceptive deficits and taste alterations are sporadically reported and there is also compelling electrophysiological, histological and imaging evidence of sensory network alterations. Gait impairment, impaired dexterity, and poor balance in ALS are likely to be multifactorial, with extrapyramidal, cerebellar, proprioceptive and vestibular deficits at play. Human imaging studies and animal models also confirm dorsal column-medial lemniscus pathway involvement as part of the disease process. Sensory symptoms are relatively common in spinal and bulbar muscular atrophy (SBMA) and Hereditary Spastic Paraplegia (HSP), but are inconsistently reported in primary lateral sclerosis (PLS) and in post-poliomyelitis syndrome (PPS). Conclusions: Establishing the prevalence and nature of sensory dysfunction across the spectrum of MNDs has a dual clinical and academic relevance. From a clinical perspective, subtle sensory deficits are likely to impact the disability profile and care needs of patients with MND. From an academic standpoint, sensory networks may be ideally suited to evaluate propagation patterns and the involvement of subcortical grey matter structures. Our review suggests that sensory dysfunction is an important albeit under-recognised facet of MND. Full article
16 pages, 4810 KiB  
Article
Echinococcus granulosus-Induced Liver Damage Through Ferroptosis in Rat Model
by Shaohua Zhai, Yueqi Yang, Yang Zhou, Qianqian Lai, Kunlei Li, Songhan Liu, Weilu Li, Feng Gao and Jiyu Guan
Cells 2025, 14(5), 328; https://doi.org/10.3390/cells14050328 (registering DOI) - 22 Feb 2025
Viewed by 117
Abstract
(1) Background: Cystic echinococcosis (CE) is an Echinococcus granulosus-induced worldwide parasitic zoonosis and is a recognized public health and socio-economic concern. The liver is the major target organ for CE’s infective form protoscolex (PSC), which causes serious liver damage and endangers the [...] Read more.
(1) Background: Cystic echinococcosis (CE) is an Echinococcus granulosus-induced worldwide parasitic zoonosis and is a recognized public health and socio-economic concern. The liver is the major target organ for CE’s infective form protoscolex (PSC), which causes serious liver damage and endangers the host’s life. Reports show that PSC infection causes liver cell Fe2+ metabolism disorder and abnormal deposition of Fe2+ in liver cells and results in liver cell death. However, whether PSC-induced liver cell death is associated with ferroptosis remains to be clarified. (2) Methods: Using both an in vivo rat model and an in vitro co-culture of PSCs and the cell system, we studied the histopathological progress of PSCs infection and the cytopathogenesis of PSC-induced cell death in the liver. Hepatic-injury-related ferroptosis signaling pathways were identified by proteomics analysis at various stages of PSCs infection, and physiological and the biochemical indexes and expression of pathway proteins related to hepatic ferroptosis were studied. Ferrostatin-1, a ferroptosis inhibitor, was employed for in vivo interference with early protoscolices infection in rats, and the effects of the inhibition of hepatocyte ferroptosis on hepatocyte injury and the generation of fibrotic cysts were investigated. Additionally, PSCs were exposed to in vitro co-culture with BRL, a rat hepatocyte line, to clarify the direct influences of PSCs on BRL ferroptosis. (3) Results: The results of our in vivo studies revealed that PSCs infection induced Fe2+ enrichment in liver cells surrounding the PSCs cysts, cellular oxidation, and liver tissue damage along with the prolongation of PSCs parasitism. The results of our in vitro studies verified the ability of PSCs to directly induce ferroptosis, the formation of fibrotic cysts, and alteration of the iron metabolism of liver cells. The analysis of KEGG signaling pathways revealed that ferroptosis- and ROS-related pathways were significantly induced with PSC infection. Using Ferrostatin-1 effectively blocked ferroptosis, reversed Fe2+content, reduced liver cell oxidation, and reduced PSC-induced fibrosis cysts. (4) Conclusions: Our study reveals the histopathological progress of PSC infection and the cytopathogenesis of PSC-induced ferroptosis. Ferrostatin-1 effectively blocked PSCs infection and PSC-induced cell death in vivo and in vitro. Accordingly, the inhibition of PSC-induced hepatocyte ferroptosis may be an effective method in the control of Echinococcus granulosus infection and should be seriously considered in clinical studies. Full article
(This article belongs to the Section Cellular Pathology)
Show Figures

Figure 1

Figure 1
<p>(<b>A</b>) Establishment of rat infection model by PSCs liver lobe injection. (<b>B</b>) Morphological characteristics of rat liver cysts 1, 3, and 6 months after infection with PSCs. Cysts are indicated by white arrows (<span class="html-italic">n</span> = 6 rats/group). (<b>C</b>) Histopathological observation results of rat liver cysts at different stages of infection with PSCs; germinal layer, keratinization layer, and inflammatory cell zone are indicated by black, red, white, and blue arrows (<span class="html-italic">n</span> = 6 rats/group, scale bars represent 100 μm and 50 μm). (<b>D</b>) Statistical results of inflammatory cells in rat liver cysts in peripheral liver tissue at different stages of infection with PSCs (<span class="html-italic">n</span> = 3 rats/group). (<b>E</b>–<b>G</b>) Inflammatory cytokine contents of TGF-β1, TNF, and IFN in rat liver cysts in peripheral liver tissue at different stages of infection with PSCs (<span class="html-italic">n</span> = 3 rats/group). (<b>H</b>) Western-blotting detection of expression level of death marker proteins ((<b>H</b>(<b>a</b>)–(<b>d</b>)) Ferroptosis TFRC, Apoptotic caspase3, Autophagy GSDMD, Pyroptosis LC3 II/I) in rat hepatocytes at different stages of infection with PSCs (<span class="html-italic">n</span> = 3 rats/group); * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001, **** <span class="html-italic">p</span> &lt; 0.0001 (Student <span class="html-italic">t</span> test).</p>
Full article ">Figure 2
<p>(<b>A</b>–<b>C</b>) Rat liver differential protein statistical results at different stages of infection with PSCs; (<b>D</b>–<b>F</b>) rat liver differential KEGG signaling pathway at different stages of infection with PSCs.</p>
Full article ">Figure 3
<p><b>(A)</b> Ultra-structure of hepatocytes near cysts at different stages of infection with PSCs (0, 1, 3, and 6 months); mitochondria are indicated by white arrows,PSC are indicated by black arrows(<span class="html-italic">n</span> = 3 rats/group, scale bars represent 2 μm and 1 μm). (<b>B</b>) Fe<sup>2+</sup>, (<b>C</b>) GSH, (<b>D</b>(<b>a</b>,<b>b</b>)) ROS, (<b>E</b>) MDA, (<b>F</b>) SOD, and (<b>G</b>) LDH of hepatocytes near cysts at different stages of infection with PSCs. (<b>H</b>(<b>a</b>,<b>b</b>)) Western blotting of protein expression levels of TFRC, GPX4, FTH1, NOX1, SLC3A2, and SLC7A11 in ferroptosis signaling pathway in rat liver tissue at different stages of infection with PSCs (<span class="html-italic">n</span> = 3 rats/group)); * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001, **** <span class="html-italic">p</span> &lt; 0.0001 (Student <span class="html-italic">t</span> test).</p>
Full article ">Figure 4
<p><b>(A)</b> Ferrostatin-1 inhibitor intervenes in vivo in PSC-mediated hepatocyte ferroptosis model establishment. (<b>B</b>) Characteristics of rat liver cyst morphology in rat blank control group, 1-month infection model group, and Ferrostatin-1 (5 mg/kg) intervention group. Cysts are indicated by white arrows (<span class="html-italic">n</span> = 6 rats/group). (<b>C</b>) Histological changes in Ferrostatin-1 on liver cysts in a rat model infected with PSCs for 1 month; germinal layer, keratinization layer, and inflammatory cell zone are indicated by black, white, and blue arrows (<span class="html-italic">n</span> = 6 rats/group; scale bars represent 100 μm and 50 μm). (<b>D</b>) TEM observation of Ferrostatin-1 on cyst peripheral hepatocyte ultra-structure changes in a rat model infected with PSCs for 1 month; mitochondria are indicated by white arrows (<span class="html-italic">n</span> = 3 rats/group; scale bars represent 2 μm and 1 μm). Effect of Ferrostatin-1 on (<b>E</b>) Fe<sup>2+</sup>, (<b>F</b>) GSH, (<b>G</b>(<b>a</b>,<b>b</b>)) ROS, (<b>H</b>) SOD, (<b>I</b>) LDH, and (<b>J</b>) MDA in cyst peripheral liver tissue in rat model infected with PSCs for 1 month; (<b>K</b>(<b>a</b>,<b>b</b>)) Western-blotting assay of Ferrostatin-1 on expression levels of ferroptosis signaling pathway proteins, such as TFRC, GPX4, FTH1, and SLC7A11, in liver tissues in rat model infected with PSCs for 1 month (<span class="html-italic">n</span> = 3 rats/group); * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001, **** <span class="html-italic">p</span> &lt; 0.0001 (Student <span class="html-italic">t</span> test).</p>
Full article ">Figure 5
<p>(<b>A</b>) Normal BRL cell, PSCs + BRL cell, PSCs+ Ferrostatin-1 + BRL cell, and cell infection models were set up. (<b>B</b>) TEM observation of morphology of PSCs and BRL co-cultured cells. Shedding cell plaques are indicated by red arrows, and PSCs are indicated by black arrows (<span class="html-italic">n</span> = 6 in every group; scale bars represent 100 μm). (<b>C</b>) TEM observation of ultra-structure changes in PSCs and BRL co-cultured cells. Mitochondria are indicated by white arrows (<span class="html-italic">n</span> = 3 rats/group; scale bars represent 2 μm and 1 μm). (<b>D</b>) CCK-8 detection of effect of PSCs infection on BRL cell proliferation. (<b>E</b>(<b>a</b>,<b>b</b>)) ROS detection of co-culture of PSCs and BRL cells (<span class="html-italic">n</span> = 3 rats/group); * <span class="html-italic">p</span> &lt; 0.05, *** <span class="html-italic">p</span> &lt; 0.001 (Student <span class="html-italic">t</span> test).</p>
Full article ">Figure 6
<p>(<b>A</b>) Fe<sup>2+</sup> in normal BRL cells, PSCs and BRL co-cultured cells, and PSCs +Ferrostatin-1 and BRL co-cultured cells. (<b>B</b>) GSH, (<b>C</b>) GSSH, (<b>D</b>) GSH/GSSH, (<b>E</b>) SOD, (<b>F</b>) LDH, and (<b>G</b>) MDA concentration detection (<span class="html-italic">n</span> = 6 rats/group). (<b>H</b>(<b>a</b>,<b>b</b>)) Western-blotting detection of expression levels of cell ferroptosis signaling pathway proteins in each group, such as TFRC, GPX4, FTH1, NOX1, SLC3A2, andSLC7A11 (<span class="html-italic">n</span> = 3 rats/group); * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001, **** <span class="html-italic">p</span> &lt; 0.0001 (Student <span class="html-italic">t</span> test).</p>
Full article ">
13 pages, 1433 KiB  
Commentary
Anastasis and Other Apoptosis-Related Prosurvival Pathways Call for a Paradigm Shift in Oncology: Significance of Deintensification in Treating Solid Tumors
by Razmik Mirzayans
Int. J. Mol. Sci. 2025, 26(5), 1881; https://doi.org/10.3390/ijms26051881 (registering DOI) - 22 Feb 2025
Viewed by 190
Abstract
What is apoptosis? The Nomenclature Committee on Cell Death and numerous other pioneering cancer/p53 biologists use the terms “apoptosis” and “cell death” interchangeably, disregard the mind-numbing complexity and heterogeneity that exists within a tumor (intratumor heterogeneity), disregard the contribution of polyploid giant cancer [...] Read more.
What is apoptosis? The Nomenclature Committee on Cell Death and numerous other pioneering cancer/p53 biologists use the terms “apoptosis” and “cell death” interchangeably, disregard the mind-numbing complexity and heterogeneity that exists within a tumor (intratumor heterogeneity), disregard the contribution of polyploid giant cancer cells (PGCCs; the root causes of therapy resistance and relapse) to this heterogeneity, and then propose novel apoptosis-stimulating anticancer strategies. This is shocking for the following three reasons. First, clinical studies reported since the 1990s have revealed that increased apoptosis in solid tumors is associated with increased tumor diversity and poor prognosis. Second, we have known for years that dying (apoptotic) cancer cells release a panel of secretions (e.g., via phoenix rising and other pathways) that promote metastatic outgrowth. Third, over a decade ago, it was demonstrated that cancer cells can recover from late stages of apoptosis (after the formation of apoptotic bodies) via the homeostatic process of anastasis, resulting in the emergence of aggressive variants. The cell surface expression of CD24 has recently been reported to be preferentially enriched in recovered (anastatic) cancer cells that exhibit tumorigenic properties. These and related discoveries outlined herein call for a paradigm shift in oncology to focus on strategies that minimize the occurrence of treacherous apoptosis and other tumor-repopulating events (e.g., therapy-induced cancer cell dormancy and reactivation). They also raise an intriguing question: is deregulated anastasis (rather than evasion of apoptosis) a hallmark of cancer? Full article
(This article belongs to the Section Molecular Oncology)
Show Figures

Figure 1

Figure 1
<p>Early (canonical) and late (tumor-repopulating) events after engaging apoptosis in solid tumors. Early events: different stimuli trigger apoptosis, which involves the induction of proapoptotic genes, mitochondrial changes, caspase activation, loss of cell volume (shrinkage), and formation of membrane-bound apoptotic bodies (adapted from Kim et al. [<a href="#B41-ijms-26-01881" class="html-bibr">41</a>]). Late events: cancer cells can return from various stages of apoptosis, even after the formation of apoptotic bodies, via the homeostatic process of anastasis, often resulting in the emergence of more aggressive cancers. Several other prosurvival pathways are associated with apoptosis, as indicted. For details, see the text and [<a href="#B15-ijms-26-01881" class="html-bibr">15</a>].</p>
Full article ">Figure 2
<p>Therapy-induced responses in different subpopulations of cancer cells within a solid tumor that contribute to resistance and relapse. For details, see the text and [<a href="#B53-ijms-26-01881" class="html-bibr">53</a>].</p>
Full article ">Figure 3
<p>Representative images showing the induction of apoptosis (treated) followed by recovery (washed) in HeLa cervical carcinoma cells. The images are screenshots of a video presented online by Montell [<a href="#B66-ijms-26-01881" class="html-bibr">66</a>].</p>
Full article ">
20 pages, 2258 KiB  
Review
A Comprehensive Review of Nanoparticle-Based Drug Delivery for Modulating PI3K/AKT/mTOR-Mediated Autophagy in Cancer
by Md Ataur Rahman, Maroua Jalouli, Sujay Kumar Bhajan, Mohammed Al-Zharani and Abdel Halim Harrath
Int. J. Mol. Sci. 2025, 26(5), 1868; https://doi.org/10.3390/ijms26051868 - 21 Feb 2025
Viewed by 170
Abstract
The phosphoinositide 3-kinase (PI3K)/AKT/mammalian target of the rapamycin (mTOR) pathway plays a crucial role in the regulation of autophagy, a cellular mechanism vital for homeostasis through the degradation of damaged organelles and proteins. The dysregulation of this pathway is significantly associated with cancer [...] Read more.
The phosphoinositide 3-kinase (PI3K)/AKT/mammalian target of the rapamycin (mTOR) pathway plays a crucial role in the regulation of autophagy, a cellular mechanism vital for homeostasis through the degradation of damaged organelles and proteins. The dysregulation of this pathway is significantly associated with cancer progression, metastasis, and resistance to therapy. Targeting the PI3K/AKT/mTOR signaling pathway presents a promising strategy for cancer treatment; however, traditional therapeutics frequently encounter issues related to nonspecific distribution and systemic toxicity. Nanoparticle-based drug delivery systems represent a significant advancement in addressing these limitations. Nanoparticles enhance the bioavailability, stability, and targeted delivery of therapeutic agents, facilitating the precise modulation of autophagy in cancer cells. Functionalized nanoparticles, such as liposomes, polymeric nanoparticles, and metal-based nanocarriers, facilitate targeted drug delivery to tumor tissues, minimizing off-target effects and improving therapeutic efficacy. These systems can deliver multiple agents concurrently, enhancing the modulation of PI3K/AKT/mTOR-mediated autophagy and related oncogenic pathways. This review examines advancements in nanoparticle-mediated drug delivery that target the PI3K/AKT/mTOR pathway, emphasizing their contribution to improving precision and minimizing side effects in cancer therapy. The integration of nanotechnology with molecularly targeted therapies presents substantial potential for addressing drug resistance. Future initiatives must prioritize the optimization of these systems to enhance clinical translation and patient outcomes. Full article
Show Figures

Figure 1

Figure 1
<p>The PI3K/AKT/mTOR pathway in Cancer. After a growth factor binds to its receptor, the route activates Class I phosphoinositide 3-kinases. Receptor tyrosine kinases activate Class IA PI3K, while G-protein-coupled receptors activate Class IB. Both classes convert PIP2 into PIP3, which recruits and activates plasma membrane AKT. Activated AKT phosphorylates and suppresses TSC1/2, a negative regulator of mTORC1. This blockage activates mTORC1, a protein synthesis, cell metabolism, and growth master regulator. The mTORC1 complex boosts translation and cancer growth, proliferation, and survival. The mechanism also aids cancer migration and metastasis. The figure was created using the <a href="http://BioRender.com" target="_blank">BioRender.com</a> online commercial platform.</p>
Full article ">Figure 2
<p>PI3K/AKT/mTOR and autophagy-mediated nanoparticle signaling in cancer. Nanoparticles bind with receptor tyrosine kinases (RTKs) on cancer cells, initiating the activation of the PI3K/AKT/mTOR signaling cascade. Activated PI3K produces PIP3, which attracts and activates AKT. AKT phosphorylates and inhibits the TSC1/2 (tuberous sclerosis complex), hence obstructing the inhibition of Rheb, a small GTPase that stimulates mTORC1 and autophagy-mediated cell death. Nanoparticles play a significant role in cancer therapy by modulating reactive oxygen species (ROS) levels and inducing apoptosis in cancer cells. The interactions among nanoparticles, ROS production, and apoptosis can occur either independently or simultaneously in cancer cells, depending upon cellular context, NP characteristics, and microenvironmental factors. The figure was created using the <a href="http://BioRender.com" target="_blank">BioRender.com</a> online commercial platform.</p>
Full article ">Figure 3
<p>Key nanoparticle categories for cancer drug delivery. Many nanoparticles are used in cancer therapy and a therapeutic use. Liposomes and solid lipid nanoparticles (SLNs) are intensively explored for encapsulating hydrophilic and hydrophobic medicines. Another important group is polymer-based nanoparticles, such as PLGA, chitosan, and dendrimers. PLGA nanoparticles are biocompatible and biodegradable, making them excellent for prolonged drug release. Chitosan, a natural polymer, is mucoadhesive and ideal for targeted therapy. Dendrimers’ branching architectures enable precise drug loading and functionalization, improving targeting. Gold nanoparticles (AuNPs), silica nanoparticles, and quantum dots are recognized for their unique functions. In photothermal therapy, gold nanoparticles absorb light and generate heat to kill cancer cells. Silica nanoparticles’ porous architecture allows high drug loading, while quantum dots’ fluorescence allows imaging-guided drug delivery. Hybrid nanoparticles combine organic and inorganic benefits. Lipid-coated gold nanoparticles combine biocompatibility with photothermal characteristics, improving cancer treatment. The figure was created using the BioRender online commercial platform.</p>
Full article ">Figure 4
<p>Modulation of PI3K/AKT/mTOR-mediated autophagy by NPs in cancer therapy. The dual function of nanoparticles (NPs) in modulating the PI3K/AKT/mTOR signaling pathway to regulate autophagy in cancer cells. Nanoparticles, including gold nanoparticles (AuNPs), silica nanoparticles (SiNPs), lipid-based nanoparticles, iron oxide nanoparticles (IONPs), graphene oxide nanoparticles (GONPs), chitosan-based nanoparticles, and polymeric nanoparticles, impede the PI3K/AKT/mTOR pathway, resulting in the induction of autophagy. This pathway promotes cancer cell death by elevating autophagic flux, oxidative stress, and apoptosis. Conversely, some nanoparticles, such as cerium oxide nanoparticles (CeONPs), silica-coated quantum dots, calcium phosphate nanoparticles (CaPNPs), and albumin-based nanoparticles stimulate the PI3K/AKT/mTOR pathway, thereby suppressing autophagy. This inhibition diminishes autophagy-related cancer cell survival, hence augmenting the effectiveness of chemotherapeutic drugs. The figure was created using the <a href="http://BioRender.com" target="_blank">BioRender.com</a> online commercial platform.</p>
Full article ">
15 pages, 705 KiB  
Review
Biased Opioid Receptor Agonists: Balancing Analgesic Efficacy and Side-Effect Profiles
by Jie Ju, Zheng Li, Jie Liu, Xiaoling Peng and Feng Gao
Int. J. Mol. Sci. 2025, 26(5), 1862; https://doi.org/10.3390/ijms26051862 - 21 Feb 2025
Viewed by 52
Abstract
Opioids are the most effective option for severe pain. However, it is well documented that the side effects associated with prolonged opioid use significantly constrain dosage in the clinical setting. Recently, researchers have concentrated on the development of biased opioid receptor agonists that [...] Read more.
Opioids are the most effective option for severe pain. However, it is well documented that the side effects associated with prolonged opioid use significantly constrain dosage in the clinical setting. Recently, researchers have concentrated on the development of biased opioid receptor agonists that preferentially activate the G protein signaling pathway over β-arrestin signaling. This approach is based on the hypothesis that G protein signaling mediates analgesic effects, whereas β-arrestin signaling is implicated in adverse side effects. Although certain studies have demonstrated that the absence or inhibition of β-arrestin signaling can mitigate the incidence of side effects, recent research appears to challenge these earlier findings. In-depth investigations into biased signal transduction of opioid receptor agonists have been conducted, potentially offering novel insights for the development of biased opioid receptors. Consequently, this review elucidates the contradictory roles of β-arrestin signaling in the adverse reactions associated with opioid receptor activation. Furthermore, a comparative analysis was conducted to evaluate the efficacy of the classic G protein-biased agonists, TRV130 and PZM21, relative to the traditional non-biased agonist morphine. This review aims to inform the development of novel analgesic drugs that can optimize therapeutic efficacy and safety, while minimizing adverse reactions to the greatest extent possible. Full article
(This article belongs to the Section Molecular Pharmacology)
Show Figures

Figure 1

Figure 1
<p>Intracellular signaling mediated by opioid receptors. Activation of opioid receptors by exogenous agonists leads to dissociation of G protein heterotrimers into α and βγ subunits. Gα inhibits adenylate cyclase activity, thereby preventing adenosine ATP from producing cAMP. Gβ/γ leads to the inactivation of calcium channels and activation of potassium channels. Consequently, these effects inhibit the release of nociceptive neurotransmitters, thereby reducing the transmission of pain signals and exerting analgesic effects. The interaction of β-arrestin with phosphorylated MOR results in receptor internalization and desensitization, and activates the MAPK cascade pathways.</p>
Full article ">
28 pages, 2001 KiB  
Article
Pharmacogenomic Study of SARS-CoV-2 Treatments: Identifying Polymorphisms Associated with Treatment Response in COVID-19 Patients
by Alexandre Serra-Llovich, Natalia Cullell, Olalla Maroñas, María José Herrero, Raquel Cruz, Berta Almoguera, Carmen Ayuso, Rosario López-Rodríguez, Elena Domínguez-Garrido, Rocio Ortiz-Lopez, María Barreda-Sánchez, Marta Corton, David Dalmau, Esther Calbo, Lucía Boix-Palop, Beatriz Dietl, Anna Sangil, Almudena Gil-Rodriguez, Encarna Guillén-Navarro, Esther Mancebo, Saul Lira-Albarán, Pablo Minguez, Estela Paz-Artal, Gladys G. Olivera, Sheila Recarey-Rama, Luis Sendra, Enrique G. Zucchet, Miguel López de Heredia, Carlos Flores, José A. Riancho, Augusto Rojas-Martinez, Pablo Lapunzina, Ángel Carracedo, María J. Arranz and SCOURGE COHORT GROUPadd Show full author list remove Hide full author list
Biomedicines 2025, 13(3), 553; https://doi.org/10.3390/biomedicines13030553 - 21 Feb 2025
Viewed by 190
Abstract
Background/Objectives: The COVID-19 pandemic resulted in 675 million cases and 6.9 million deaths by 2022. Despite substantial declines in case fatalities following widespread vaccination campaigns, the threat of future coronavirus outbreaks remains a concern. Current treatments for COVID-19 have been repurposed from [...] Read more.
Background/Objectives: The COVID-19 pandemic resulted in 675 million cases and 6.9 million deaths by 2022. Despite substantial declines in case fatalities following widespread vaccination campaigns, the threat of future coronavirus outbreaks remains a concern. Current treatments for COVID-19 have been repurposed from existing therapies for other infectious and non-infectious diseases. Emerging evidence suggests a role for genetic factors in both susceptibility to SARS-CoV-2 infection and response to treatment. However, comprehensive studies correlating clinical outcomes with genetic variants are lacking. The main aim of our study is the identification of host genetic biomarkers that predict the clinical outcome of COVID-19 pharmacological treatments. Methods: In this study, we present findings from GWAS and candidate gene and pathway enrichment analyses leveraging diverse patient samples from the Spanish Coalition to Unlock Research of Host Genetics on COVID-19 (SCOURGE), representing patients treated with immunomodulators (n = 849), corticoids (n = 2202), and the combined cohort of both treatments (n = 2487) who developed different outcomes. We assessed various phenotypes as indicators of treatment response, including survival at 90 days, admission to the intensive care unit (ICU), radiological affectation, and type of ventilation. Results: We identified significant polymorphisms in 16 genes from the GWAS and candidate gene studies (TLR1, TLR6, TLR10, CYP2C19, ACE2, UGT1A1, IL-1α, ZMAT3, TLR4, MIR924HG, IFNG-AS1, ABCG1, RBFOX1, ABCB11, TLR5, and ANK3) that may modulate the response to corticoid and immunomodulator therapies in COVID-19 patients. Enrichment analyses revealed overrepresentation of genes involved in the innate immune system, drug ADME, viral infection, and the programmed cell death pathways associated with the response phenotypes. Conclusions: Our study provides an initial framework for understanding the genetic determinants of treatment response in COVID-19 patients, offering insights that could inform precision medicine approaches for future epidemics. Full article
Show Figures

Figure 1

Figure 1
<p>Manhattan plot of results for the type of ventilation in the subgroup of patients treated with immunomodulators. The results of the candidate gene analyses are highlighted in green and the GWAS results in grey and black. The blue line indicates the threshold <span class="html-italic">p</span>-value &lt; 1 × 10<sup>−5</sup> and the red line the threshold <span class="html-italic">p</span>-value &lt; 5 × 10<sup>−8</sup>. The corresponding QQ plot is in the top left corner.</p>
Full article ">Figure 2
<p>Manhattan plot of the results for radiological affectation in the subgroup of patients treated with corticoids. The results of the candidate gene analyses are highlighted in green and the GWAS results in grey and black. The blue line indicates the threshold <span class="html-italic">p</span>-value &lt; 1 × 10<sup>−5</sup> and the red line the threshold <span class="html-italic">p</span>-value &lt; 5 × 10<sup>−8</sup>. The corresponding QQ plot is in the top left corner.</p>
Full article ">Figure 3
<p>Manhattan plot of the results for radiological affectation in the total combined sample. The results of the candidate gene analyses are highlighted in green and the GWAS results in grey and black. The blue line indicates the threshold <span class="html-italic">p</span>-value &lt; 1 × 10<sup>−5</sup> and the red line the threshold <span class="html-italic">p</span>-value &lt; 5 × 10<sup>−8</sup>. The corresponding QQ plot is in the top left corner.</p>
Full article ">Figure 4
<p>Interactions between the significative results of the GWAS and candidate genes analysis and the pro-inflammatory molecule Nuclear Factor kappa-B. Created with BioRender.com.</p>
Full article ">
19 pages, 907 KiB  
Review
Fatty Degenerative Osteonecrosis of the Jaw: Bridging Molecular Insights and Clinical Practice—A Scoping Review
by Marzena Dominiak, Wojciech Niemczyk, Artur Pitułaj, Witold Świenc and Jacek Matys
Int. J. Mol. Sci. 2025, 26(5), 1853; https://doi.org/10.3390/ijms26051853 - 21 Feb 2025
Viewed by 56
Abstract
Fatty degenerative osteonecrosis of the jaw (FDOJ) is a chronic, aseptic inflammatory condition that is characterized by molecular disruptions in bone metabolism and necrotic bone marrow within the jawbone cavities. In contrast to the overt clinical signs typically observed in osteopathies, FDOJ frequently [...] Read more.
Fatty degenerative osteonecrosis of the jaw (FDOJ) is a chronic, aseptic inflammatory condition that is characterized by molecular disruptions in bone metabolism and necrotic bone marrow within the jawbone cavities. In contrast to the overt clinical signs typically observed in osteopathies, FDOJ frequently presents with a “silent inflammation” phenotype. The electronic databases PubMed, Scopus, and Embase were searched using appropriate search terms, and the methodology was performed according to PRISMA-ScR guidelines. The elevated expression of inflammatory mediators, particularly C-C motif Chemokine Ligand-5/Regulated on Activation, Normal T Cell Expressed and Secreted (CCL5/RANTES), fibroblast growth factor-2, and interleukin-1 receptor antagonist, distinguishes FDOJ at the molecular level and links it to systemic inflammatory and autoimmune diseases. These immunohistochemical markers play a pivotal role in the pathogenesis of chronic inflammation, immune response regulation, and abnormal bone remodeling. Advanced diagnostic tools, such as conebeam computed tomography and trans-alveolar ultrasonography, facilitate the detection of pathological changes that are not easily discernible with conventional radiography. Surgical intervention remains the primary treatment modality, often complemented by therapies that target these molecular pathways to modulate chronic inflammation. This article underscores the importance of integrating molecular diagnostics, advanced imaging, and clinical data for effective FDOJ detection and management. Full article
(This article belongs to the Special Issue Oral Soft Tissue Repair and Oral Diseases: 2nd Edition)
Show Figures

Figure 1

Figure 1
<p>PRISMA 2020 flow diagram.</p>
Full article ">Figure 2
<p>Principal signaling pathways that are disrupted by ROS in osteoblasts, osteoclasts, and MSCs.</p>
Full article ">Figure 3
<p>A diagram illustrating the causal sequence of FDOJ formation subsequent to tooth extractions.</p>
Full article ">
20 pages, 10576 KiB  
Article
Clinical Research on Positron Emission Tomography Imaging of the Neuro-Stimulation System in Patients with Cochleo-Vestibular Implants: Is There a Response Beyond the Peripheral Organ?
by Joan Lorente-Piera, Elena Prieto, Ángel Ramos de Miguel, Manuel Manrique, Nicolás Pérez-Fernández, Ángel Ramos Macías, Jaime Monedero Afonso, Alina Sanfiel Delgado, Jorge Miranda Ramos, Paula Alonso Alonso, Javier Arbizu and Raquel Manrique-Huarte
J. Clin. Med. 2025, 14(5), 1445; https://doi.org/10.3390/jcm14051445 - 21 Feb 2025
Viewed by 131
Abstract
Introduction: In patients refractory to vestibular rehabilitation in the management of bilateral vestibulopathy, the cochleo-vestibular implant has emerged as a viable alternative to enhance both audiovestibular function and quality of life. The main objective of this study is to pioneer the use of [...] Read more.
Introduction: In patients refractory to vestibular rehabilitation in the management of bilateral vestibulopathy, the cochleo-vestibular implant has emerged as a viable alternative to enhance both audiovestibular function and quality of life. The main objective of this study is to pioneer the use of PET to assess cortical modifications in patients with cochleo-vestibular implants, aiming to evaluate the safety and functional improvements in individuals with bilateral vestibulopathy and severe to profound hearing loss. Methods: A phase I pilot clinical trial was conducted with participants who received a BIONIC-VEST CI24RE cochleo-vestibular implant, with pre- and post-implantation assessments conducted for twelve months. Audiovestibular testing and two PET studies with 18F-FDG under baseline conditions and with active stimulus to observe cortical-level differences were performed. Results: Five patients were included in the study, all of them treated with a cochleo-vestibular implant, none of whom presented postoperative adverse effects. Audiologically, the mean post-implant gain was 56.63 ± 14.53 dB and 50.40 ± 35.54% in terms of speech intelligibility. From a vestibular perspective, the most remarkable findings were observed at the graviceptive pathway level, where a mean posturographic improvement was observed, with a sensory organization test score of 24.20 ± 13.74 and a subjective visual vertical of 1.57° ± 0.79°, achieving, in most cases, results within the normal range (<2.3°) by the end of the follow-up. PET images confirmed that with the electrical stimulus active (implant ON), there was a supratentorial activation pattern, particularly in areas related to somatosensory integration, emotional regulation, and autonomic control. Conclusions: The BIONIC-VEST implant significantly improved the vestibular system, particularly the graviceptive pathway, enhancing balance and SVV and reducing fall risk. PET revealed distinct uptake patterns in baseline and activated conditions, highlighting a cortical-level response with the use of the cochleo-vestibular implant. Full article
(This article belongs to the Special Issue Current Updates on the Inner Ear)
Show Figures

Figure 1

Figure 1
<p>Summary of the postoperative follow-up conducted on the patients included in the study.</p>
Full article ">Figure 2
<p>Summary of the PET subtraction algorithm applied in our study. Subtraction maps are presented over a standard MRI image.</p>
Full article ">Figure 3
<p>Cortical representation and summary of the different areas studied in the clinical trial using PET-CT imaging.</p>
Full article ">Figure 4
<p>Progression of auditory performance recorded in the PTA (<b>left</b> panel) and rate of discrimination (<b>right</b> panel) in the ipsilateral ears of each patient included in the study.</p>
Full article ">Figure 5
<p>(<b>A</b>). Summary of the different gains recorded in the vHIT in patients from the trial, analyzing the three different canals separately. The blue colors represent the ear ipsilateral to the cochleo-vestibular implant, while the purple colors represent the contralateral ears. (<b>B</b>). Example of vHIT results for the lateral semicircular canals of one of the patients in the trial. It shows two scenarios: the top image corresponds to the pre-implantation phase, and the bottom image shows the post-implantation phase with the cochleo-vestibular implant (CVI). Despite the lack of improvement in gain, clear refixation saccade phenomena can be observed. (<b>C</b>). Representation of the evolution of the different quotients included in the SOT. The dark blue color corresponds to the pre-implantation moment and the light blue color corresponds to post-implantation. SOMATO: somatosensorial; VESTIB: vestibular; PREF: visual preference.</p>
Full article ">Figure 5 Cont.
<p>(<b>A</b>). Summary of the different gains recorded in the vHIT in patients from the trial, analyzing the three different canals separately. The blue colors represent the ear ipsilateral to the cochleo-vestibular implant, while the purple colors represent the contralateral ears. (<b>B</b>). Example of vHIT results for the lateral semicircular canals of one of the patients in the trial. It shows two scenarios: the top image corresponds to the pre-implantation phase, and the bottom image shows the post-implantation phase with the cochleo-vestibular implant (CVI). Despite the lack of improvement in gain, clear refixation saccade phenomena can be observed. (<b>C</b>). Representation of the evolution of the different quotients included in the SOT. The dark blue color corresponds to the pre-implantation moment and the light blue color corresponds to post-implantation. SOMATO: somatosensorial; VESTIB: vestibular; PREF: visual preference.</p>
Full article ">Figure 6
<p>Representation of the evolution of SVV in the different subjects. The dark blue color corresponds to the pre-implantation moment and the light blue color corresponds to post-implantation.</p>
Full article ">Figure 7
<p>Subtraction PET images illustrating the metabolic changes between baseline conditions and electrical stimulation. The yellow circle in patient 3 represents notable uptake at the ipsilateral frontal operculum related to the implant, while the blue circle in the fourth patient shows a decrease in uptake in the ipsilateral precentral gyrus, and the yellow circle in the fifth subject indicates an increase in uptake with the presence of electrical stimulation in the ipsilateral inferior frontal gyrus.</p>
Full article ">
20 pages, 16775 KiB  
Article
Galangin’s Neuroprotective Role: Targeting Oxidative Stress, Inflammation, and Apoptosis in Ischemic Stroke in a Rat Model of Permanent Middle Cerebral Artery Occlusion
by Nut Palachai, Araya Supawat, Ratchaniporn Kongsui, Lars Klimaschewski and Jinatta Jittiwat
Int. J. Mol. Sci. 2025, 26(5), 1847; https://doi.org/10.3390/ijms26051847 - 21 Feb 2025
Viewed by 118
Abstract
The rising incidence of ischemic stroke poses significant health and healthcare burdens. Given the limitations of current therapeutic options, there is increasing interest in exploring the potential of galangin, a natural flavonoid compound, as a treatment for ischemic stroke. This study aimed to [...] Read more.
The rising incidence of ischemic stroke poses significant health and healthcare burdens. Given the limitations of current therapeutic options, there is increasing interest in exploring the potential of galangin, a natural flavonoid compound, as a treatment for ischemic stroke. This study aimed to evaluate the neuroprotective effects and underlying mechanisms of galangin in mitigating oxidative stress, inflammation, and apoptosis in a rat model of permanent cerebral ischemia. Sixty male Wistar rats were divided into six groups: control; right middle cerebral artery occlusion (Rt.MCAO) with vehicle; Rt.MCAO with piracetam, a synthetic compound known as a cognitive enhancer; and Rt.MCAO with galangin administered at doses of 25, 50, and 100 mg/kg body weight. Neurological deficit scores, brain edema, neuronal density, and microglial morphology were assessed along with the activity of myeloperoxidase (MPO), a marker of inflammation, and superoxide dismutase (SOD). Additionally, the expression of key markers for inflammation and apoptosis, cyclooxygenase-2 (COX-2), interleukin-6 (IL-6), Bcl-2-associated X protein (Bax), B-cell lymphoma-extra large (Bcl-XL), and caspase-3, was analyzed to elucidate potential mechanisms. The results demonstrated that galangin treatment significantly improved neurological deficit scores, reduced brain edema, enhanced neuronal density, attenuated microglial activation, decreased MPO activity, and increased SOD activity in both the cortex and hippocampus, highlighting its neuroprotective potential. These effects were linked to the modulation of inflammatory and apoptotic pathways. Specifically, galangin significantly reduced the expression of IL-6, COX-2, Bax, and caspase-3 while increasing the levels of the anti-apoptotic protein Bcl-XL. In conclusion, galangin demonstrates significant promise as a neuroprotective agent for ischemic stroke by suppressing inflammation and apoptosis, thereby improving neurological outcomes. However, clinical trials are required to validate these preclinical findings and confirm galangin’s therapeutic efficacy in humans. Full article
(This article belongs to the Special Issue Natural Bioactives and Inflammation, 2nd Edition)
Show Figures

Figure 1

Figure 1
<p>Effects of galangin on neurological deficit score. Data are expressed as the mean ± SEM (n = 5). * <span class="html-italic">p</span> &lt; 0.05 when compared to the control group; # <span class="html-italic">p</span> &lt; 0.05 when compared to the Rt.MCAO + vehicle group. Rt.MCAO, right middle cerebral artery occlusion; BW, body weight.</p>
Full article ">Figure 2
<p>Effects of galangin on brain edema. Data are expressed as the mean ± SEM (n = 5). * <span class="html-italic">p</span> &lt; 0.05 when compared to the control group; # <span class="html-italic">p</span> &lt; 0.05 when compared to the Rt.MCAO + vehicle group. Rt.MCAO, right middle cerebral artery occlusion; BW, body weight.</p>
Full article ">Figure 3
<p>Effect of galangin on neuronal density in the cortex and hippocampus. (<b>A</b>,<b>B</b>) Representative images of Nissl-stained sections from the CA1 and CA3 regions of the rat brains, captured at 40× magnification. (<b>C</b>) A graph illustrates neuronal density in the cortex and the CA1 and CA3 regions of the hippocampus. Data are expressed as the mean ± SEM (n = 5). * <span class="html-italic">p</span> &lt; 0.05 when compared to the control group; # <span class="html-italic">p</span> &lt; 0.05 when compared to the Rt.MCAO + vehicle group. Rt.MCAO, right middle cerebral artery occlusion; BW, body weight.</p>
Full article ">Figure 3 Cont.
<p>Effect of galangin on neuronal density in the cortex and hippocampus. (<b>A</b>,<b>B</b>) Representative images of Nissl-stained sections from the CA1 and CA3 regions of the rat brains, captured at 40× magnification. (<b>C</b>) A graph illustrates neuronal density in the cortex and the CA1 and CA3 regions of the hippocampus. Data are expressed as the mean ± SEM (n = 5). * <span class="html-italic">p</span> &lt; 0.05 when compared to the control group; # <span class="html-italic">p</span> &lt; 0.05 when compared to the Rt.MCAO + vehicle group. Rt.MCAO, right middle cerebral artery occlusion; BW, body weight.</p>
Full article ">Figure 4
<p>Effect of galangin on microglial activation following ischemic stroke. Iba-1 immunohistochemical staining was performed to determine microglial activation in the cortex (<b>A</b>) and the hippocampus (<b>B</b>). Representative images of ramified microglia in control group and activated microglia in the Rt.MCAO + vehicle group. Treatments with piracetam and galangin demonstrated a reduction in microglial activation. All images were taken at 40× magnification. Scale bar = 50 µm.</p>
Full article ">Figure 5
<p>Effect of galangin on MPO activity in the cortex and hippocampus of Rt.MCAO-induced rats. Data are expressed as the mean ± SEM (n = 5). * <span class="html-italic">p</span> &lt; 0.05 when compared to the control group; # <span class="html-italic">p</span> &lt; 0.05 when compared to the Rt.MCAO + vehicle group. MPO, myeloperoxidase; Rt.MCAO, right middle cerebral artery occlusion; BW, body weight.</p>
Full article ">Figure 6
<p>Effect of galangin on SOD activity in the cortex and hippocampus of Rt.MCAO-induced rats. Data are expressed as the mean ± SEM (n = 5). * <span class="html-italic">p</span> &lt; 0.05 when compared to the control group; # <span class="html-italic">p</span> &lt; 0.05 when compared to the Rt.MCAO + vehicle group. SOD, superoxide dismutase; Rt.MCAO, right middle cerebral artery occlusion; BW, body weight.</p>
Full article ">Figure 7
<p>Anti-inflammatory effects of galangin in Rt.MCAO-induced rats. (<b>A</b>) Image of an immunoblot of COX-2 (74 kDa) and IL-6 (25 kDa) from the cerebral cortex. β-actin (41 kDa) was used as a loading control. (<b>B</b>,<b>C</b>) Quantitative analysis of (<b>B</b>) COX-2 and (<b>C</b>) IL-6 band density normalized with beta-actin. Data are expressed as the mean ± SEM (n = 5). * <span class="html-italic">p</span> &lt; 0.05 vs. the control; # <span class="html-italic">p</span> &lt; 0.05 vs. the Rt.MCAO+ vehicle group. COX-2, cyclooxygenase-2; IL-6, interleukin-6; Rt.MCAO, right middle cerebral artery occlusion; BW, body weight.</p>
Full article ">Figure 7 Cont.
<p>Anti-inflammatory effects of galangin in Rt.MCAO-induced rats. (<b>A</b>) Image of an immunoblot of COX-2 (74 kDa) and IL-6 (25 kDa) from the cerebral cortex. β-actin (41 kDa) was used as a loading control. (<b>B</b>,<b>C</b>) Quantitative analysis of (<b>B</b>) COX-2 and (<b>C</b>) IL-6 band density normalized with beta-actin. Data are expressed as the mean ± SEM (n = 5). * <span class="html-italic">p</span> &lt; 0.05 vs. the control; # <span class="html-italic">p</span> &lt; 0.05 vs. the Rt.MCAO+ vehicle group. COX-2, cyclooxygenase-2; IL-6, interleukin-6; Rt.MCAO, right middle cerebral artery occlusion; BW, body weight.</p>
Full article ">Figure 8
<p>Neuroprotective role of galangin in rats with Rt.MCAO-induced injury via activation of anti-apoptotic pathways. (<b>A</b>) Image of an immunoblot of Bax (21 kDa), Bcl-xL (26 kDa), and caspase-3 (32 kDa) from the cerebral cortex. β-actin (41 kDa) was used as a loading control. (<b>B</b>–<b>D</b>) Quantitative analysis of (<b>B</b>) Bcl-xL, (<b>C</b>) Bax, and (<b>D</b>) caspase-3 band density normalized with beta-actin. Data are expressed as the mean ± SEM (n = 5). * <span class="html-italic">p</span> &lt; 0.05 vs. the control; # <span class="html-italic">p</span> &lt; 0.05 vs. the Rt.MCAO+ vehicle group.</p>
Full article ">Figure 8 Cont.
<p>Neuroprotective role of galangin in rats with Rt.MCAO-induced injury via activation of anti-apoptotic pathways. (<b>A</b>) Image of an immunoblot of Bax (21 kDa), Bcl-xL (26 kDa), and caspase-3 (32 kDa) from the cerebral cortex. β-actin (41 kDa) was used as a loading control. (<b>B</b>–<b>D</b>) Quantitative analysis of (<b>B</b>) Bcl-xL, (<b>C</b>) Bax, and (<b>D</b>) caspase-3 band density normalized with beta-actin. Data are expressed as the mean ± SEM (n = 5). * <span class="html-italic">p</span> &lt; 0.05 vs. the control; # <span class="html-italic">p</span> &lt; 0.05 vs. the Rt.MCAO+ vehicle group.</p>
Full article ">
12 pages, 522 KiB  
Review
A New Hope for Woman with Vasomotor Symptoms: Neurokinin B Antagonists
by Blazej Meczekalski, Anna Kostrzak, Christian Unogu, Stefania Bochynska, Marzena Maciejewska-Jeske, Gregory Bala and Anna Szeliga
J. Clin. Med. 2025, 14(5), 1438; https://doi.org/10.3390/jcm14051438 - 21 Feb 2025
Viewed by 133
Abstract
KNDy (kisspeptine, neurokinin B, dynorphin) neurons, located in the hypothalamus, play a crucial role in the development of vasomotor symptoms (VSM) in menopausal women. Estrogen withdrawal during menopause leads to the hyperactivation of kisspeptin and neurokinin B (NKB) secretion, contributing to the onset [...] Read more.
KNDy (kisspeptine, neurokinin B, dynorphin) neurons, located in the hypothalamus, play a crucial role in the development of vasomotor symptoms (VSM) in menopausal women. Estrogen withdrawal during menopause leads to the hyperactivation of kisspeptin and neurokinin B (NKB) secretion, contributing to the onset of these symptoms. The identification of NKB/neurokinin B receptor (NK3R) signaling as a key mechanism in menopausal hot flashes has driven the development of NK3R antagonists. These antagonists restore the disrupted balance in KNDy neuron activity caused by estrogen deficiency, thereby reducing the frequency and severity of VMS. In 2023, the FDA approved fezolinetant, the first selective NK3R antagonist, for the treatment of moderate to severe VMS associated with menopause. Additionally, elinzanetant, a dual neurokinin-1 and neurokinin-3 receptor antagonist, has demonstrated promising results. The approval application for elinzanetant was supported by positive findings from the OASIS 1, 2, and 3 Phase III clinical studies. The dual antagonism of NK-1 and NK-3 receptors enhances its efficacy by alleviating menopause-related sleep disturbances and modulating peripheral vasodilatation. In this regard, elinzanetant represents a promising non-hormonal treatment that targets the underlying causes of VMS through NK-1 and NK-3 receptor pathways. The development of neurokinin B antagonist for VMS treatment exemplifies the impact of advanced pharmacological research on gynecological endocrinology. Full article
(This article belongs to the Section Obstetrics & Gynecology)
Show Figures

Figure 1

Figure 1
<p>Anatomy of KNDy neurons. Neurokinin B (NKB) synthesized in KNDy neurons stimulates GnRH neurons, promoting GnRH production, which in turn stimulates the secretion of FSH and LH in response to low estradiol concentrations. NKB antagonists inhibit the hyperactivation of KNDy neurons in postmenopausal patients, restoring normal signaling in the hypothalamus and preventing dysregulated vasodilation. Abbreviations: NKB—Neurokinin B, Dyn—dynorphin, DynR—Dynorphin receptor, NKBR—Neurokinin B receptor, PG—progesterone receptor, ERα—Estradiol receptor α.</p>
Full article ">
24 pages, 1074 KiB  
Review
The Impact of Immunomodulatory Components Used in Clinical Nutrition—A Narrative Review
by Aleksandra Raczyńska, Teresa Leszczyńska, Piotr Skotnicki and Aneta Koronowicz
Nutrients 2025, 17(5), 752; https://doi.org/10.3390/nu17050752 - 21 Feb 2025
Viewed by 209
Abstract
Background: Malnutrition is a clinical condition that leads to unfavourable changes in health. It affects 35–55% of hospitalized patients, and in the case of cancer, this prevalence rises to 40–90% of patients. Screening nutritional status is essential for preventing undernutrition, which is crucial [...] Read more.
Background: Malnutrition is a clinical condition that leads to unfavourable changes in health. It affects 35–55% of hospitalized patients, and in the case of cancer, this prevalence rises to 40–90% of patients. Screening nutritional status is essential for preventing undernutrition, which is crucial as its treatment. Undernutrition in patients after severe injuries significantly increases catabolic changes. Cytokines and hormones, such as epinephrine, glucagon, and cortisol, are released, which can increase energy expenditure by 50%. Properly conducted nutritional treatment aims to maintain or improve the nutritional status of patients whose nutrition with a natural diet is insufficient, moreover, in some cases, treatment of the underlying disease. Methods: This study is a narrative review focused on immunonutrition. The search for source articles, mainly from the last 10 years, was conducted in the PubMed and Google Schoolar databases, as well as in printed books. The key words used were “malnutrition”, “inflammation”, “clinical nutrition”, “immunomodulatory components”, “nutritional status assessment”, “enteral nutrition”, “parenteral nutrition”, and their combinations. Results: Providing substances such as omega-3 fatty acids, glutamine, arginine, nucleotides, antioxidants, and prebiotic fiber has a beneficial impact on immunological and anti-inflammatory pathways. The above-mentioned ingredients may inhibit the secretion of pro-inflammatory cytokines, activate anti-inflammatory cytokines, stimulate immune cells, and have a beneficial effect in allergic diseases, respiratory infections, or wound healing. Conslusion: Immunonutrition can be administrated via oral, enteral, and parenteral routes. It is crucial to highlight the importance of proper nutritional status in patients. The relationship between inflammation and malnutrition creates a vicious cycle, where one negatively affects the other due to increased metabolic demand, loss of appetite, weakened immune system, and gut dysbiosis. Full article
(This article belongs to the Special Issue The Effect of Bioactive Compounds in Anti-inflammation)
Show Figures

Figure 1

Figure 1
<p>Algorithm of action in clinical nutrition [<a href="#B3-nutrients-17-00752" class="html-bibr">3</a>].</p>
Full article ">Figure 2
<p>Simplified impact of immunonutrition. Immunonutrition plays a key role in modulating the immune response and reducing inflammation, which translates into improved clinical parameters for patients. Nutrients such as glutamine, arginine, omega-3 fatty acids, vitamins (E, D, and C), nucleotides, and microelements (selenium, zinc, and magnesium) have significant immunomodulatory effects, influencing the functioning of immune cells and inflammatory processes. Omega-3 fatty acids and vitamins E, D, C have the ability to reduce (as indicated by arrows) the activity of pro-inflammatory cytokines. Reducing inflammation through appropriate nutritional intervention translates into numerous clinical benefits, including reduced infection rates, improved immunological parameters in surgical patients, increased efficacy of anticancer therapies (radiotherapy and chemotherapy), and shortened hospitalization time. Consequently, immunonutrition is an important element of supportive therapy that can significantly improve treatment outcomes and quality of life of patients [<a href="#B68-nutrients-17-00752" class="html-bibr">68</a>,<a href="#B69-nutrients-17-00752" class="html-bibr">69</a>,<a href="#B70-nutrients-17-00752" class="html-bibr">70</a>,<a href="#B71-nutrients-17-00752" class="html-bibr">71</a>,<a href="#B72-nutrients-17-00752" class="html-bibr">72</a>,<a href="#B73-nutrients-17-00752" class="html-bibr">73</a>,<a href="#B74-nutrients-17-00752" class="html-bibr">74</a>,<a href="#B75-nutrients-17-00752" class="html-bibr">75</a>,<a href="#B76-nutrients-17-00752" class="html-bibr">76</a>].</p>
Full article ">
21 pages, 4317 KiB  
Article
Topical Application of VitB6 Ameliorates PM2.5-Induced Dry Eye via NFκB Pathway in a Murine Model
by Jinyu Hu, Yanmei Zeng, Liying Tang, Lei Ye, Cheng Chen, Qian Ling, Xiaoyu Wang, Liangqi He, Xu Chen, Yixin Wang, Qianmin Ge and Yi Shao
Biomedicines 2025, 13(3), 541; https://doi.org/10.3390/biomedicines13030541 - 21 Feb 2025
Viewed by 172
Abstract
Background/Objectives: Dry eye (DE) is mainly characterized by dryness, foreign body sensation, eye pain and visual impairment. Their possible causes are mainly inflammation, tissue damage and neurosensory abnormalities, and vitamin B6 (VitB6) attenuates the inflammatory response by modulating the NF-κB pathway to quench [...] Read more.
Background/Objectives: Dry eye (DE) is mainly characterized by dryness, foreign body sensation, eye pain and visual impairment. Their possible causes are mainly inflammation, tissue damage and neurosensory abnormalities, and vitamin B6 (VitB6) attenuates the inflammatory response by modulating the NF-κB pathway to quench reactive oxygen species (ROS). The aim of this experiment was to investigate the therapeutic effect of VitB6 eye drops on particulate matter 2.5 (PM2.5)-induced dry eye in mice. Methods: Mice induced with the dry eye group were first induced using PM2.5 eye drops in a standard environment for 14 days, and then treated with different concentrations of VitB6 eye drops for 14 consecutive days. The phenol red cotton test was used to measure tear production. Ocular inflammation index and tear film function were evaluated by slim microscopy. Hematoxylin–eosin (HE) staining was used to observe conjunctival and corneal structure. Periodate–Schiff (PAS) staining was used to quantify conjunctival goblet cells. Corneal cell apoptosis was determined by TUNEL assay. The expression of keratin 10 (K10) and p-NF-κB p65 was detected by immunofluorescent staining and Western blot analysis. Results: Mice using only the PM2.5 model all exhibited varying degrees of dry eye symptoms. VitB6 treatment increased tear secretion and reduced inflammatory indices in mice with increased nerve density and number of branches in the basement membrane of the corneal epithelium. Conclusions: We found that administering VitB6 eye drops has a therapeutic effect in PM2.5-induced DE. This observation suggests that VitB6 may be useful in the clinical therapy of DE. Full article
(This article belongs to the Special Issue Wound Healing: From Mechanisms to Therapeutic Approaches)
Show Figures

Figure 1

Figure 1
<p>Representative pictures of the changes in the ocular surface and the degree of inflammation in different groups of mice before and after dry eye treatment. (<b>A</b>) FL staining. At the beginning of the experiment, the DE, PBS, 0.02%, and 0.05% groups showed patchy staining of corneal FL, and after 14 days of treatment, the 0.02% and 0.05% groups showed a significant decrease in corneal staining. (<b>B</b>) RB staining. At the beginning of the experiment, the DE group, PBS group, 0.02% group and 0.05% group showed large patchy staining of corneal RB staining, and after 14 days of treatment, the 0.02% group and 0.05% group showed a significant decrease in corneal staining. (<b>C</b>) LG staining. At the beginning of the experiment, the DE group, PBS group, 0.02% group and 0.05% group showed large staining of corneal LG staining, and after 14 days of treatment, the 0.05% group showed almost disappearance of corneal staining. Slit lamp (40×). Image taken by EOS 200D (Canon, Tokyo, Japan). (n = 10 mice/group). Abbreviations: D, Day; DE, dry eye; PBS, phosphate-buffered saline; VitB6, vitamin B6; FL, fluorescein; RB, Rose bengal; LG, Lissamine Green.</p>
Full article ">Figure 2
<p>Comparative analysis graphs of different test results in dry eyes. (<b>A</b>) FL staining results before treatment, on the 7th day of treatment, and on the 14th day of treatment. The corneal staining was reduced in the 0.02% VitB6 treatment group and the 0.05% VitB6 treatment group starting from the 7th day of treatment. (<b>B</b>) RB staining results before treatment, on day 7 of treatment, and on day 14 of treatment. Corneal staining was reduced in the 0.02% VitB6 treatment group relative to the DE group at day 7; corneal staining was reduced in the 0.05% VitB6 treatment group relative to the DE and PBS treatment groups. At day 14 of treatment, corneal staining was reduced in the 0.02% VitB6 treatment group and the 0.05% VitB6 treatment group. (<b>C</b>) LG staining results before treatment, on day 7 of treatment, and on day 14 of treatment. Corneal staining was reduced in the 0.02% VitB6-treated group and the 0.05% VitB6-treated group starting from the 7th day of treatment. (<b>D</b>) SIT results before treatment, on day 7 of treatment, and on day 14 of treatment. Tear secretion increased in both the 0.02% VitB6-treated group and the 0.05% VitB6-treated group starting on day 7 of treatment, and the increase was statistically significant in the 0.05% VitB6-treated group relative to the 0.02% VitB6-treated group by day 7. (<b>E</b>) TBUT test results before treatment, on day 7 of treatment, and on day 14 of treatment. At day 7 of treatment, there was no significant difference in BUT assay results between the four groups. On day 14 of treatment, the BUT time was significantly prolonged in both the 0.02% VitB6 treatment group and the 0.05% VitB6 treatment group. Note: <sup>a</sup> <span class="html-italic">p</span> &lt; 0.05, vs. DE; <sup>b</sup> <span class="html-italic">p</span> &lt; 0.05, vs. PBS; <sup>c</sup> <span class="html-italic">p</span> &lt; 0.05, vs. 0.02% VitB6. Abbreviations: D, Day; DE, dry eye; PBS, phosphate-buffered saline; VitB6, vitamin B6; FL, fluorescein staining; RB, Rose bengal staining; LG, Lissamine Green staining; SIT, Schirmer I test; TBUT, Tear break-up time.</p>
Full article ">Figure 3
<p>Representative pictures of nerve fibers in different groups of mice after 14 days of treatment. The distribution of nerves under the basement membrane of corneal epithelium was observed under confocal microscope after 14 days of treatment, and the corneal stromal nerve fibers were clear and straight in the control group. In the DE group, PBS treatment group, 0.02% VitB6 and 0.05% VitB6 treatment groups, the nerve fibers in the corneal stroma were curved, while the curvature of the nerve fibers in the 0.02% VitB6 and 0.05% VitB6 groups improved and the nerve fiber density increased (n = 10 mice/group). Scale bars: 100 µm. Abbreviations: D, Day; DE, dry eye; PBS, phosphate-buffered saline; VitB6, vitamin B6.</p>
Full article ">Figure 4
<p>Representative pictures of HE staining of the corneal conjunctiva of mice in different groups after 14 days of treatment. (<b>A</b>) The central corneal and conjunctival epithelial cells in the DE and PBS-treated groups were disorganized, with thickened layers and inflammatory cell infiltration in the stroma. In contrast, the central corneal and conjunctival epithelium of 0.02% VitB6 and 0.05% VitB6 treatment groups were smooth and the cell morphology gradually normalized. (<b>B</b>) Comparison of changes in the number of layers of epithelial cells after 14 days of treatment with two different concentrations of VitB6 eye drops. (<b>C</b>) No significant difference in the number of layers of conjunctival epithelial cell layers was observed in the four groups of mice. Note: * <span class="html-italic">p</span> &lt; 0.05, vs. dry eye, # <span class="html-italic">p</span> &lt; 0.05, vs. PBS. (n = 10 mice/group). Scale bars: 20 µm. Abbreviations: DE, dry eye; PBS, phosphate-buffered saline; VitB6, vitamin B6.</p>
Full article ">Figure 5
<p>Comparison of TUNEL staining results of different groups of mice after 14 days of treatment. (<b>A</b>) Corneal sections of mice were stained with TUNEL and then nuclei (blue) were re-stained with DAPI, and the number of apoptotic cells was indicated in green. The number of apoptotic cells in the control group was minimal, and after 14 days of treatment, the number of apoptotic cells in the 0.02% VitB6 and 0.05% VitB6 treatment groups was reduced compared with the dry eye group and the PBS treatment group. (<b>B</b>) The IOD value was used to determine the number of TUNEL-positive cells in the central cornea of mice, and the IOD value in the 0.02% VitB6 and 0.05% VitB6 treatment groups was lower than the dry eye group and the PBS treatment group. (n = 10 mice/group). Scale bars: 10 µm. Note: * <span class="html-italic">p</span> &lt; 0.05, vs. dry eye, # <span class="html-italic">p</span> &lt; 0.05, vs. PBS. Abbreviations: DE, dry eye; PBS, phosphate-buffered saline; VitB6, vitamin B6; TUNEL, terminal deoxynucleotidyl transferase-mediated end-of-cut labeling.</p>
Full article ">Figure 6
<p>Comparison of PAS staining of the domed conjunctiva in different groups of mice after 14 days of treatment. (<b>A</b>,<b>B</b>) There was no significant difference in the number of conjunctival cupped cells in different groups of mice (n = 10 mice/group). Scale bars: 50 µm. Abbreviations: DE, dry eye; PBS, phosphate-buffered saline; VitB6, vitamin B6; PAS, Periodic acid–Schiff.</p>
Full article ">Figure 7
<p>Comparison of corneal K10 expression in different groups of mice after 14 days of treatment. (<b>A</b>) The nuclei (blue) were re-stained with DAPI after immunofluorescence staining for K10, and the expression of K10 protein in the central cornea was highlighted in green. Control group mice did not express K10, and the central corneal expression of K10 in mice treated with VitB6 for 14 days was lower than in the dry eye group and the PBS-treated group. (<b>B</b>) The IOD values of K10 in the central cornea of mice treated with 0.02 percent VitB6 and 0.05 percent VitB6 were considerably lower than those in the PBS treatment group and the dry eye group after 14 days of therapy (n = 10 mice/group). Scale bars: 10 µm. Note: * <span class="html-italic">p</span> &lt; 0.05, vs. dry eye, # <span class="html-italic">p</span> &lt; 0.05, vs. PBS. Abbreviations: DE, dry eye; PBS, phosphate-buffered saline; VitB6, vitamin B6; K10, keratin 10.</p>
Full article ">Figure 8
<p>Activation of NF-κB in the corneas of different groups of mice was detected by WB using β-actin as an internal reference. (<b>A</b>) WB assay: After 14 days of therapy, mice in the 0.02% VitB6 and 0.05% VitB6 treatment groups had significantly lower levels of p-NF-κB p65 expression in their corneas than mice in the dry eye and PBS treatment groups. (<b>B</b>) IOD results are statistically analyzed. The central corneal p-NF-κB/Total NF-κB values were considerably lower after 14 days of treatment than in the PBS-treated group and the dry eye group (n = 10 mice/group). Note: * <span class="html-italic">p</span> &lt; 0.05, vs. dry eye, # <span class="html-italic">p</span> &lt; 0.05, vs. PBS. Abbreviations: DE, dry eye; PBS, phosphate-buffered saline; VitB6, vitamin B6; WB, Western blot.</p>
Full article ">
11 pages, 985 KiB  
Article
Disease Acceptance and Control from the Subjective Health Experience Model as Health Perception Predictors in Immune-Mediated Inflammatory Diseases
by Tessa S. Folkertsma, Sjaak Bloem, Robert M. Vodegel, Reinhard Bos and Greetje J. Tack
Biomedicines 2025, 13(3), 538; https://doi.org/10.3390/biomedicines13030538 - 20 Feb 2025
Viewed by 117
Abstract
Background/Objectives: The multifactorial nature of immune-mediated inflammatory diseases (IMIDs) requires integrating pathophysiological understanding with subjective patient experiences. Patient-reported outcome measures (PROMs) are a useful tool for incorporating this in routine clinical practice. However, current PROMs do not fully encompass the complete subjective health [...] Read more.
Background/Objectives: The multifactorial nature of immune-mediated inflammatory diseases (IMIDs) requires integrating pathophysiological understanding with subjective patient experiences. Patient-reported outcome measures (PROMs) are a useful tool for incorporating this in routine clinical practice. However, current PROMs do not fully encompass the complete subjective health experiences (SHE) of patients and are thus insufficient for guiding truly personalised care. The SHE model provides insights into SHE through the determinants of disease acceptance and perceived control. While validated across various demographics, its predictive power in IMIDs cohorts remains unexplored. This study aims to assess whether acceptance and perceived control in the SHE model predict health experiences in patients with IMID and how these immunological conditions compare. Methods: Questionnaires regarding health perception, acceptance, control, and subjective health experiences were distributed to 450 Dutch citizens. Descriptive statistics, reliability checks, and partial least squares structural equation modelling were used to examine relationships between variables. Results: Health perception strongly predicts SHE through acceptance and control. Across all conditions, the pathway moves from health perception to control, then to acceptance, and finally to SHE. However, the roles of acceptance and control differ by condition. In burdensome diseases like inflammatory bowel disease and rheumatoid arthritis, acceptance plays a greater role, while control has a stronger influence in conditions like psoriasis. Conclusions: This study supports the predictive validity of the SHE model for IMIDs, showing that disease acceptance and control affect health experiences differently across conditions. These insights improve the understanding of psychological factors in health experiences and call for tailored interventions for patients with IMID. Full article
Show Figures

Figure 1

Figure 1
<p>Analytical process chart.</p>
Full article ">Figure 2
<p>Characteristics of the SHE model for the total study population and for disease-specific data. Health perception was measured via the EQ-5D-5L questionnaire, and acceptance, control, and SHE were measured via the SHE questionnaire. Non-significant <span class="html-italic">p</span>-values (<span class="html-italic">p</span> &gt; 0.05) are indicated in red.</p>
Full article ">
26 pages, 6815 KiB  
Article
Identifying Baicalein as a Key Bioactive Compound in XueBiJing Targeting KEAP1: Implications for Antioxidant Effects
by Ting-Syuan Lin, Xiao-Xuan Cai, Yi-Bing Wang, Jia-Tong Xu, Ji-Han Xiao, Hsi-Yuan Huang, Shang-Fu Li, Kun-Meng Liu, Ji-Hang Chen, Li-Ping Li, Jie Ni, Yi-Gang Chen, Zi-Hao Zhu, Jing Li, Yuan-Jia Hu, Hsien-Da Huang, Hua-Li Zuo and Yang-Chi-Dung Lin
Antioxidants 2025, 14(3), 248; https://doi.org/10.3390/antiox14030248 - 20 Feb 2025
Viewed by 125
Abstract
Background: XueBiJing injection (XBJ) is renowned for its multi-target pharmacological effects, including immunomodulatory, antithrombotic, and antioxidant activities, offering potential therapeutic benefits for patients with severe infections such as sepsis and Coronavirus disease 2019 (COVID-19). Despite its clinical effectiveness, the molecular targets and mechanisms [...] Read more.
Background: XueBiJing injection (XBJ) is renowned for its multi-target pharmacological effects, including immunomodulatory, antithrombotic, and antioxidant activities, offering potential therapeutic benefits for patients with severe infections such as sepsis and Coronavirus disease 2019 (COVID-19). Despite its clinical effectiveness, the molecular targets and mechanisms of XBJ remain unclear, warranting further investigation. Purpose: This study aimed to identify the key bioactive compounds in XBJ and elucidate their molecular targets and mechanisms. Methods: The zebrafish model was first used to evaluate the anti-inflammatory and antioxidant effects of XBJ, and the differentially expressed genes (DEGs) were identified by RNA sequencing and network analysis. Network pharmacology was used to analyze the relationship between bioactive compounds and molecular targets, and molecular docking and kinetic simulation were used to explore the target binding ability of key compounds. Cellular Thermal Shift Assay-Western Blot (CETSA-WB) and Surface Plasmon Resonance (SPR) further verified the interaction between compounds and targets; finally, the key pathways were confirmed by gene silencing experiments. Results: The zebrafish model results reveal that XBJ significantly reduced neutrophil and macrophage counts in a dose-dependent manner, emphasizing its potent anti-inflammatory effects. A transcriptomic analysis highlighted the differential expression of key genes in the KEAP1/NRF2 pathway, including HMOX1, SLC7A11, NQO1, and TXNRD1. A network analysis further pinpointed KEAP1 as a central molecular target, with tanshinone IIA, baicalein, and luteolin identified as key active compounds modulating this pathway. Among these, tanshinone IIA and baicalein exhibited strong binding interactions with KEAP1, which were confirmed through molecular docking and kinetic simulations. Further validation showed that baicalein directly targets KEAP1, as demonstrated by CETSA-WB and SPR analysis. Additionally, the gene silencing experiments of KEAP1 and NRF2 reinforced their crucial roles in activating the KEAP1/NRF2 pathway. Conclusion: These findings collectively establish baicalein as a critical bioactive compound in XBJ, driving its antioxidant and anti-inflammatory effects via KEAP1/NRF2 pathway activation through direct binding to KEAP1, providing new insights into the mechanism of action of XBJ. Full article
Show Figures

Figure 1

Figure 1
<p>Workflow of investigation on XueBiJing (XBJ) injection.</p>
Full article ">Figure 2
<p>The effect of XBJ on neutrophils and macrophages. (<b>A</b>) A representative image showing fluorescence detection on neutrophils, captured using a fluorescent microscope (Nikon AZ100, Minato City, Japan). (<b>B</b>) The number of neutrophils in zebrafish swim bladder after treatment. (<b>C</b>) A representative image showing fluorescence detection on macrophages, captured using a fluorescent microscope (Nikon AZ100, Minato City, Japan). (<b>D</b>) The fluorescence intensity of macrophages in zebrafish swim bladder after treatment. In all experimental trials, a minimum of 30 larvae were utilized for each condition. Statistical significance is denoted by *** for <span class="html-italic">p</span> &lt; 0.001 and ** for <span class="html-italic">p</span> &lt; 0.01 when compared to the Dexamethasone group.</p>
Full article ">Figure 3
<p>Results from RNA sequencing data analysis of samples treated with XBJ. (<b>A</b>) Differential expression analysis of low, medium, and high concentrations. (<b>B</b>) Heatmap visually represents expression patterns of differentially expressed genes. (<b>C</b>) Functional enrichment analysis indicates significant correlation between these differentially expressed genes and specific biological pathways. (<b>D</b>) Activation pathways of Nrf2 and its role in various cellular processes.</p>
Full article ">Figure 4
<p>The CS network of XBJ. (<b>A</b>) CS network of XBJ was established based on 701 compounds. (<b>B</b>) Primary constituents of XBJ within each module exhibit substantial structural similarities.</p>
Full article ">Figure 5
<p>The TPT network of XBJ. (<b>A</b>) In total, 847 human targets are associated with 701 compounds, and 683 protein targets were enriched to pathways with <span class="html-italic">p</span> &lt; 0.05. (<b>B</b>) The TPT network decomposed into four major modules.</p>
Full article ">Figure 6
<p>The calculated association strength (<b><span class="html-italic">e<sub>uv</sub></span></b>) of each compound module with each target module. The purple and white refer to the highest and lowest <b><span class="html-italic">e<sub>uv</sub></span></b> values, respectively. The values highlighted with an orange frame denote the locations of the targets that participated in the pathway (NRF2 regulation of oxidative stress response) we are interested in within the TPT network and the locations of the compounds that are associated with these targets in the CS network.</p>
Full article ">Figure 7
<p>In silico evaluation assessing binding potential of predicted compounds with KEAP1. (<b>A</b>) Binding mode and interaction of compounds and activity center of KEAP1. (<b>B</b>) Molecular dynamics simulation of positive control and anticipated active compounds. Left to right: RMSD of carbon α of protein compared with initial equilibrium structure; RMSD of ligands to KEAP1 protein; number of hydrogen bonds formed with standard hydrogen bond length and bond angle; MMPB(GB)SA binding energy distribution (ΔGGas (van der Waals and electrostatic energies), ΔGSolid (potential energy of polar solvent and non-polar solvent), and ΔG<sub>Total</sub> (ΔGGas<sub>Total</sub> and ΔGSolid<sub>Total</sub>); energy contribution of each residue. Top to bottom: ML334, deoxynyboquinone, baicalein, tanshinone II A, and luteolin.</p>
Full article ">Figure 8
<p>The on-target validation of binding affinity of baicalein with KEAP1. (<b>A</b>) The indicated protein specificity was analyzed by Western blotting using KEAP1 antibody. The protein extracted from the MCF-7 cell line lysate was used for CETSA-WB in vitro. (<b>B</b>) An SPR analysis of baicalein and KEAP1 protein.</p>
Full article ">Figure 9
<p>QPCR and Western blot results. (<b>A</b>) Gene and protein expression in MCF-7 cell lines treated by baicalein. * <span class="html-italic">p</span> &lt; 0.05 vs. CTRL; ** <span class="html-italic">p</span> &lt; 0.01 vs. CTRL; *** <span class="html-italic">p</span> &lt; 0.001 vs. CTRL; **** <span class="html-italic">p</span> &lt; 0.0001 vs. CTRL. MCF-7 cell lines were transfected with ncr, siKEAP1, or siNRF2. Cells were then treated with baicalein (8.772 mg/mL) for 24 h. (<b>B</b>) siKEAP1 and siNRF2 knockdown silencing efficiency. (<b>C</b>) Control groups with no drug or baicalein. (<b>D</b>) siKEAP1 groups with drug and untreated drug. (<b>E</b>) siNRF2 groups with drug and untreated drug. * <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01; *** <span class="html-italic">p</span> &lt; 0.001; **** <span class="html-italic">p</span> &lt; 0.0001. (<b>F</b>) ROS production. Control group (red), baicalein group (blue), NCR group (dark green), positive control (light green), siKEAP1 with no drug (purple), siKEAP1 with baicalein (dark blue), sNRF2 with no drug (yellow), siNRF2 with baicalein (gray).</p>
Full article ">Figure 10
<p>The molecular mechanism of XBJ’s active compound baicalein in the NRF2/KEAP1 pathway.</p>
Full article ">
Back to TopTop