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

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (891)

Search Parameters:
Keywords = endogenous inhibitors

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
11 pages, 1349 KiB  
Article
The Effect of Intracellular Calcium Buffer Bapta on Epileptiform Activity of Hippocampal Neurons
by V. P. Zinchenko, I. Yu. Teplov, F. V. Tyurin, A. E. Malibayeva, B. K. Kairat and S. T. Tuleukhanov
Int. J. Mol. Sci. 2025, 26(15), 7596; https://doi.org/10.3390/ijms26157596 - 6 Aug 2025
Abstract
The rhythm of epileptiform activity occurs in various brain injuries (ischemia, stroke, concussion, mechanical damage, AD, PD). The epileptiform rhythm is accompanied by periodic Ca2+ pulses, which are necessary for the neurotransmitter release, the repair of damaged connections between neurons, and the [...] Read more.
The rhythm of epileptiform activity occurs in various brain injuries (ischemia, stroke, concussion, mechanical damage, AD, PD). The epileptiform rhythm is accompanied by periodic Ca2+ pulses, which are necessary for the neurotransmitter release, the repair of damaged connections between neurons, and the growth of new projections. The duration and amplitude of these pulses depend on intracellular calcium-binding proteins. The effect of the synthetic fast calcium buffer BAPTA on the epileptiform activity of neurons induced by the GABA(A)-receptor inhibitor, bicuculline, was investigated in a 14-DIV rat hippocampal culture. In the epileptiform activity mode, neurons periodically synchronously generate action potential (AP) bursts in the form of paroxysmal depolarization shift (PDS) clusters and their corresponding high-amplitude Ca2+ pulses. Changes in the paroxysmal activity and Ca2+ pulses were recorded continuously for 10–11 min as BAPTA accumulated. It was shown that during BAPTA accumulation, transformation of neuronal patch activity occurs. Moreover, GABAergic and glutamatergic neurons respond differently to the presence of calcium buffer. Experiments were performed on two populations of neurons: a population of GABAergic neurons that responded selectively to ATPA, a calcium-permeable GluK1 kainate receptor agonist, and a population of glutamatergic neurons with a large amplitude of cluster depolarization (greater than −20 mV). These neurons made up the majority of neurons. In the population of GABAergic neurons, during BAPTA accumulation, the amplitude of PDS clusters decreases, which leads to a switch from the PDS mode to the classical burst mode with an increase in the electrical activity of the neuron. In glutamatergic neurons, the duration of PDS clusters decreased during BAPTA accumulation. However, the amplitude changed little. The data obtained showed that endogenous calcium-binding proteins play a significant role in switching the epileptiform rhythm to the recovery rhythm and perform a neuroprotective function by reducing the duration of impulses in excitatory neurons and the amplitude of impulses in inhibitory neurons. Full article
(This article belongs to the Section Molecular Neurobiology)
Show Figures

Figure 1

14 pages, 548 KiB  
Review
Carboxypeptidase A4: A Biomarker for Cancer Aggressiveness and Drug Resistance
by Adeoluwa A. Adeluola, Md. Sameer Hossain and A. R. M. Ruhul Amin
Cancers 2025, 17(15), 2566; https://doi.org/10.3390/cancers17152566 - 4 Aug 2025
Viewed by 63
Abstract
Carboxypeptidase A4 (CPA4) is an exopeptidase that cleaves peptide bonds at the C-terminal domain within peptides and proteins. It preferentially cleaves peptides with terminal aromatic or branched chain amino acid residues such as phenylalanine, tryptophan, or leucine. CPA4 was first discovered in prostate [...] Read more.
Carboxypeptidase A4 (CPA4) is an exopeptidase that cleaves peptide bonds at the C-terminal domain within peptides and proteins. It preferentially cleaves peptides with terminal aromatic or branched chain amino acid residues such as phenylalanine, tryptophan, or leucine. CPA4 was first discovered in prostate cancer cells, but it is now known to be expressed in various tissues throughout the body. Its physiologic expression is governed by latexin, a noncompetitive endogenous inhibitor of CPA4. Nevertheless, the overexpression of CPA4 has been associated with the progression and aggressiveness of many malignancies, including prostate, pancreatic, breast and lung cancer, to name a few. CPA4’s role in cancer has been attributed to its disruption of many cellular signaling pathways, e.g., PI3K-AKT-mTOR, STAT3-ERK, AKT-cMyc, GPCR, and estrogen signaling. The dysregulation of these pathways by CPA4 could be responsible for inducing epithelial--mesenchymal transition (EMT), tumor invasion and drug resistance. Although CPA4 has been found to regulate cancer aggressiveness and poor prognosis, no comprehensive review summarizing the role of CPA4 in cancer is available so far. In this review, we provide a brief description of peptidases, their classification, history of CPA4, mechanism of action of CPA4 as a peptidase, its expression in various tissues, including cancers, its role in various tumor types, the associated molecular pathways and cellular processes. We further discuss the limitations of current literature linking CPA4 to cancers and challenges that prevent using CPA4 as a biomarker for cancer aggressiveness and predicting drug response and highlight a number of future strategies that can help to overcome the limitations. Full article
(This article belongs to the Special Issue Insights from the Editorial Board Member)
Show Figures

Figure 1

22 pages, 533 KiB  
Review
Modulation of Glutathione-S-Transferase by Phytochemicals: To Activate or Inhibit—That Is the Question
by Irina Anna-Maria Stoian, Adelina Vlad, Marilena Gilca and Dorin Dragos
Int. J. Mol. Sci. 2025, 26(15), 7202; https://doi.org/10.3390/ijms26157202 - 25 Jul 2025
Viewed by 253
Abstract
Glutathione S-transferases (GSTs) are phase II detoxification enzymes that display several enzymatic activities, including transferase, peroxidase, reductase, and isomerase functions, as well as non-enzymatic roles (e.g., serving as binding proteins). Their complex functionality lies in the biotransformation of xenobiotics (e.g., pesticides, drugs) and [...] Read more.
Glutathione S-transferases (GSTs) are phase II detoxification enzymes that display several enzymatic activities, including transferase, peroxidase, reductase, and isomerase functions, as well as non-enzymatic roles (e.g., serving as binding proteins). Their complex functionality lies in the biotransformation of xenobiotics (e.g., pesticides, drugs) and certain endogenous compounds, primarily metabolites produced by phase I detoxification enzymes. Several plant-derived compounds have been shown to modulate the activity and expression levels of these enzymes. Phytochemical activators of GSTs are potentially beneficial for detoxification in cases of exposure to various toxic compounds, whereas inhibitors of GSTs could have positive effects as adjuvant treatments for cancers that express high levels of GSTs associated with drug resistance. Full article
(This article belongs to the Special Issue Natural-Derived Bioactive Compounds in Disease Treatment)
Show Figures

Graphical abstract

14 pages, 7293 KiB  
Article
Components of Mineralocorticoid Receptor System in Human DRG Neurons Co-Expressing Pain-Signaling Molecules: Implications for Nociception
by Shaaban A. Mousa, Xueqi Hong, Elsayed Y. Metwally, Sascha Tafelski, Jan David Wandrey, Jörg Piontek, Sascha Treskatsch, Michael Schäfer and Mohammed Shaqura
Cells 2025, 14(15), 1142; https://doi.org/10.3390/cells14151142 - 24 Jul 2025
Viewed by 287
Abstract
The mineralocorticoid receptor (MR), traditionally associated with renal function, has also been identified in various extrarenal tissues, including the heart, brain, and dorsal root ganglion (DRG) neurons in rodents. Previous studies suggest a role for the MR in modulating peripheral nociception, with MR [...] Read more.
The mineralocorticoid receptor (MR), traditionally associated with renal function, has also been identified in various extrarenal tissues, including the heart, brain, and dorsal root ganglion (DRG) neurons in rodents. Previous studies suggest a role for the MR in modulating peripheral nociception, with MR activation in rat DRG neurons by its endogenous ligand, aldosterone. This study aimed to determine whether MR, its protective enzyme 11β-hydroxysteroid dehydrogenase type 2 (11β-HSD2), its endogenous ligand aldosterone, and the aldosterone-synthesizing enzyme CYP11B2 are expressed in human DRG neurons and whether they colocalize with key pain-associated signaling molecules as potential targets for genomic regulation. To this end, we performed mRNA transcript profiling and immunofluorescence confocal microscopy on human and rat DRG tissues. We detected mRNA transcripts for MR, 11β-HSD2, and CYP11B2 in human DRG, alongside transcripts for key thermosensitive and nociceptive markers such as TRPV1, the TTX-resistant sodium channel Nav1.8, and the neuropeptides CGRP and substance P (Tac1). Immunofluorescence analysis revealed substantial colocalization of MR with 11β-HSD2 and CGRP, a marker of unmyelinated C-fibers and thinly myelinated Aδ-fibers, in human DRG. MR immunoreactivity was primarily restricted to small- and medium-diameter neurons, with lower expression in large neurons (>70 µm). Similarly, aldosterone colocalized with CYP11B2 and MR with nociceptive markers including TRPV1, Nav1.8, and TrkA in human DRG. Importantly, functional studies demonstrated that prolonged intrathecal inhibition of aldosterone synthesis within rat DRG neurons, using an aldosterone synthase inhibitor significantly downregulated pain-associated molecules and led to sustained attenuation of inflammation-induced hyperalgesia. Together, these findings identify a conserved peripheral MR signaling axis in humans and highlight its potential as a novel target for pain modulation therapies. Full article
(This article belongs to the Section Cells of the Nervous System)
Show Figures

Figure 1

21 pages, 2670 KiB  
Article
Regulatory Effect of PGE2-EP2/EP4 Receptor Pathway on Staphylococcus aureus-Induced Inflammatory Factors in Dairy Cow Neutrophils
by Yi Zhao, Chao Wang, Bo Liu, Shuangyi Zhang, Yongfei Wang, Yinghong Qian, Zhiguo Gong, Jiamin Zhao, Xiaolin Yang, Yuting Bai and Wei Mao
Biomolecules 2025, 15(8), 1062; https://doi.org/10.3390/biom15081062 - 22 Jul 2025
Viewed by 265
Abstract
Naturally occurring prostaglandin E2 (PGE2) influences cytokine production regulation in bovine neutrophils exposed to Staphylococcus aureus Rosenbach. Here, we employed bovine neutrophils as the primary experimental system, and administered specific inhibitors targeting various receptors, which were subsequently exposed to S. [...] Read more.
Naturally occurring prostaglandin E2 (PGE2) influences cytokine production regulation in bovine neutrophils exposed to Staphylococcus aureus Rosenbach. Here, we employed bovine neutrophils as the primary experimental system, and administered specific inhibitors targeting various receptors, which were subsequently exposed to S. aureus. Cytokine expression levels in dairy cow neutrophils induced by S. aureus via the endogenous PGE2-EP2/4 receptor pathway were investigated, and its effects on P38, extracellular signal-regulated kinase (ERK), P65 activation, and phagocytic function in Staphylococcus aureus Rosenbach-induced dairy cow neutrophils, were examined. Blocking cyclooxygenase-2 (COX-2) and microsomal prostaglandin E synthase-1 (mPGES-1) enzymes substantially decreased PGE2 production and release in S. aureus-exposed bovine neutrophils. Cytokine output showed significant reduction compared to that in SA113-infected controls. Phosphorylation of P38, ERK, and P65 signaling molecules was depressed in the infected group. Pharmacological interference with EP2/EP4 receptors similarly diminished cytokine secretion and phosphorylation patterns of P38, ERK, and P65, with preserved cellular phagocytic function. During S. aureus infection of bovine neutrophils, COX-2 and mPGES-1 participated in controlling PGE2 biosynthesis, and internally produced PGE2 molecules triggered NF-κB and MAPK inflammatory pathways via EP2/EP4 receptor activation, later adjusting the equilibrium between cytokine types that promote or suppress inflammation. This signaling mechanism coordinated inflammatory phases through receptor-mediated processes. Full article
(This article belongs to the Section Molecular Biology)
Show Figures

Figure 1

15 pages, 2174 KiB  
Article
Weak Acids as Endogenous Inhibitors of the Proton-Activated Chloride Channel
by Inês C. A. Pombeiro Stein, Maren Schulz, Daniel Rudolf, Christine Herzog, Frank Echtermeyer, Nils Kriedemann, Robert Zweigerdt and Andreas Leffler
Cells 2025, 14(14), 1110; https://doi.org/10.3390/cells14141110 - 19 Jul 2025
Viewed by 338
Abstract
The recently identified proton-activated chloride (PAC) channel is ubiquitously expressed, and it regulates several proton-sensitive physiological and pathophysiological processes. While the PAC channel is activated by strong acids due to the binding of protons to extracellular binding sites, here, we describe the way [...] Read more.
The recently identified proton-activated chloride (PAC) channel is ubiquitously expressed, and it regulates several proton-sensitive physiological and pathophysiological processes. While the PAC channel is activated by strong acids due to the binding of protons to extracellular binding sites, here, we describe the way in which weak acids inhibit the PAC channel by a mechanism involving a distinct extracellular binding site. Whole-cell patch clamp was performed on wildtype HEK293T cells, PAC-knockout HEK293 cells expressing human (h)PAC mutant constructs, and on hiPSC-derived cardiomyocytes. Proton-induced cytotoxicity was examined in HEK293T cells. Acetic acid inhibited endogenous PAC channels in HEK 293T cells in a reversible, concentration-dependent, and pH-dependent manner. The inhibition of PAC channels was also induced by lactic acid, propionic acid, itaconic acid, and β-hydroxybutyrate. Weak acids also inhibited recombinant wildtype hPAC channels and PAC-like currents in hiPSC-derived cardiomyocytes. Replacement of the extracellular arginine 93 by an alanine (hPAC–Arg93Ala) strongly reduced the inhibition by some weak acids, including arachidonic acid. Although lactic acid inhibited PAC, it did not reduce the proton-induced cytotoxicity examined in wildtype HEK 293 cells. To conclude, weak acids inhibit PAC via an extracellular mechanism involving Arg93. These data warrant further investigations into the regulation of the PAC channel by endogenous weak acids. Full article
(This article belongs to the Special Issue pH Sensing, Signaling, and Regulation in Cellular Processes)
Show Figures

Figure 1

14 pages, 1649 KiB  
Article
LC-MS-Based Untargeted Metabolic Profiling in Plasma Following Dapagliflozin Administration in Healthy Volunteers
by Hyeon Ji Kim, Jae Hwa Lee, Ji Seo Park, Jin Ju Park, Hae Won Lee, Heeyoun Bunch, Sook Jin Seong, Mi-Ri Gwon and Young-Ran Yoon
Metabolites 2025, 15(7), 484; https://doi.org/10.3390/metabo15070484 - 17 Jul 2025
Viewed by 484
Abstract
Dapagliflozin, a sodium-glucose cotransporter 2 inhibitor, treats type 2 diabetes by blocking renal glucose reabsorption and promoting urinary glucose excretion. This mechanism lowers blood glucose concentrations independently of insulin. The resulting caloric loss also contributes to weight reduction. Although these effects are well [...] Read more.
Dapagliflozin, a sodium-glucose cotransporter 2 inhibitor, treats type 2 diabetes by blocking renal glucose reabsorption and promoting urinary glucose excretion. This mechanism lowers blood glucose concentrations independently of insulin. The resulting caloric loss also contributes to weight reduction. Although these effects are well documented in patients with diabetes, their magnitude and underlying mechanisms in healthy individuals remain poorly understood. Background/Objectives: We investigated metabolic alterations after a single 10 mg dose of dapagliflozin in healthy adults with normal body-mass indices (BMIs) using untargeted metabolomics. Methods: Thirteen healthy volunteers completed this study. Plasma was collected before and 24 h after dosing. Untargeted metabolic profiling was performed with ultra-high-performance liquid chromatography–quadrupole time-of-flight/mass spectrometry. Results: Twenty-five endogenous metabolites were annotated; ten were putatively identified. Eight metabolites increased significantly, whereas two decreased. Up-regulated metabolites included phosphatidylcholine (PC) species (PC O-36:5, PC 36:3), phosphatidylserine (PS) species (PS 40:2, PS 40:3, PS 36:1, PS 40:4), lysophosphatidylserine 22:1, and uridine. Dehydroepiandrosterone sulfate and bilirubin were down-regulated. According to the Human Metabolome Database, these metabolites participate in glycerophospholipid, branched-chain amino acid, pyrimidine, and steroid-hormone metabolism. Conclusions: Dapagliflozin may affect pathways related to energy metabolism and homeostasis beyond glucose regulation. These data provide a reference for future investigations into energy balance and metabolic flexibility in metabolic disorders. Full article
(This article belongs to the Section Pharmacology and Drug Metabolism)
Show Figures

Figure 1

27 pages, 3370 KiB  
Review
Sourdough Fermentation and Gluten Reduction: A Biotechnological Approach for Gluten-Related Disorders
by Ricardo H. Hernández-Figueroa, Aurelio López-Malo and Emma Mani-López
Microbiol. Res. 2025, 16(7), 161; https://doi.org/10.3390/microbiolres16070161 - 17 Jul 2025
Viewed by 597
Abstract
Sourdough fermentation has emerged as a promising biotechnological approach to reducing gluten content and modifying gluten proteins in wheat-based products. This review assesses the current scientific literature on the enzymatic degradation and hydrolysis of gluten during lactic acid bacteria (LAB) sourdough fermentation. It [...] Read more.
Sourdough fermentation has emerged as a promising biotechnological approach to reducing gluten content and modifying gluten proteins in wheat-based products. This review assesses the current scientific literature on the enzymatic degradation and hydrolysis of gluten during lactic acid bacteria (LAB) sourdough fermentation. It explores implications for individuals with gluten-related disorders, including celiac disease, non-celiac gluten sensitivity and intolerance, as well as irritable bowel syndrome (IBS). In addition, LAB sourdough effect on fermentable oligo-, di-, monosaccharides and polyols (FODMAPs), amylase-trypsin inhibitors (ATIs), and phytate are revised. Selected homo- and heterofermentative LAB are capable of degrading gluten proteins, especially the polypeptides derived from the action of native cereal proteases. Mixed cultures of LAB degrade gluten peptides more effectively than monocultures. However, LAB sourdough is not sufficient to remove the toxic peptides to the minimal level (<20 ppm). This goal is achieved only if sourdough is combined with fungal proteases during sourdough fermentation. LAB sourdough directly contributes to lower FODMAPs but not ATIs and phytate. Phytate is reduced by the endogenous cereal phytases activated at acidic pHs (pH < 5.0), conditions generated during sourdough fermentation. ATIs are also lowered by endogenous cereal proteases instead of LAB proteases/peptidases. Despite LAB sourdough not fully degrading the gluten or directly reducing the ATIs and phytate, it participates through peptidases activity and acidic pH that trigger the action of endogenous cereal proteases and phytases. Full article
Show Figures

Figure 1

40 pages, 2429 KiB  
Review
Hepatocytes as Model for Investigating Natural Senotherapeutic Compounds and Their Effects on Cell Cycle Dynamics and Genome Stability
by Anastasia Fizikova, Anna Prokhorova, Daria Churikova, Zahar Konstantinov, Roman Ivanov, Alexander Karabelsky and Stanislav Rybtsov
Int. J. Mol. Sci. 2025, 26(14), 6794; https://doi.org/10.3390/ijms26146794 - 16 Jul 2025
Viewed by 692
Abstract
DNA is inherently unstable and is susceptible to damage from both endogenous sources (such as reactive oxygen species) and exogenous factors (including UV, ionizing radiation, and chemicals). The accumulation of DNA damage manifests as genetic mutations, chromosomal instability, and the stalling of DNA [...] Read more.
DNA is inherently unstable and is susceptible to damage from both endogenous sources (such as reactive oxygen species) and exogenous factors (including UV, ionizing radiation, and chemicals). The accumulation of DNA damage manifests as genetic mutations, chromosomal instability, and the stalling of DNA replication and transcription processes. Accumulated DNA damage influences apoptosis and cell cycle checkpoints, serving as one of the key triggers for the manifestation of the senescent phenotype. Both aging and cancer are associated with the accumulation of mutations in somatic cells. Disruption of cell cycle control and uncontrolled proliferation are fundamental characteristics of any cancer cell, with the majority of anticancer drugs acting as inhibitors of cyclin-dependent kinases, thereby inducing a transition of cells into a senescent state. Consequently, disturbances in the dynamics and regulation of inflammatory responses, oxidative stress, cell proliferation, DNA damage repair, and epigenetic anomalies, along with the influence of retroviruses and transposons, lead to the accumulation of senescent cells within the human body, characterized by blocked replication and cell cycle, as well as a distinct secretory phenotype. The age-related or disease-associated accumulation of these senescent cells significantly alters the physiology of tissues and the organism as a whole. Many secondary metabolites of higher plants exhibit senolytic and senomorphic activities, although most of them are not fully characterized. In this review, we will explore the principal signaling pathways in mammalian cells that govern the cell cycle and cellular senescence, with a particular emphasis on how their dynamics, expression, and regulation have been modified through the application of senotherapeutic compounds. The second section of the review will identify key target genes for the metabolic engineering, primarily aimed at enhancing the accumulation of plant secondary metabolites with potential therapeutic benefits. Lastly, we will discuss the rationale for utilizing liver cells as a model system to investigate the effects of senolytic compounds on human physiology and health, as well as how senotherapeutic substances can be leveraged to improve gene therapy approaches based on CRISPR/Cas9 and prime-editing technologies. Full article
(This article belongs to the Collection State-of-the-Art Macromolecules in Russia)
Show Figures

Figure 1

18 pages, 8672 KiB  
Article
Under Blue Light Treatment, OsCSN2 Regulates the Phenotype of Rice Seedlings Through the GA Signaling Pathway
by Xinhai Yu, Tongtong Jiao, Changfeng Liu, Hexin Zhang, Yanxi Liu, Chunyu Zhang, Ming Wu and Liquan Guo
Plants 2025, 14(13), 2015; https://doi.org/10.3390/plants14132015 - 1 Jul 2025
Viewed by 347
Abstract
Blue light is a significant environmental cue influencing plant photomorphogenesis and regulating plant growth and development. The COP9 signaling complex (CSN), a multi-subunit protein complex, plays a pivotal role in regulating photomorphogenesis, with CSN2 being identified as a key subunit essential for the [...] Read more.
Blue light is a significant environmental cue influencing plant photomorphogenesis and regulating plant growth and development. The COP9 signaling complex (CSN), a multi-subunit protein complex, plays a pivotal role in regulating photomorphogenesis, with CSN2 being identified as a key subunit essential for the assembly and function of the CSN. This study investigated the role of OsCSN2 in rice under blue-light conditions. Utilizing OsCSN2 knockout (KO) mutant plants and transgenic overexpression (OE) lines for wild-type (WT) and mutated versions of OsCSN2, we observed significant suppression of the overall seedling phenotype under blue light, indicating that OsCSN2 acts as a negative regulator of blue light-mediated morphogenesis. Further analysis revealed that exogenous application of gibberellin (GA3) and the GA synthesis inhibitor paclobutrazol (PAC) modulated seedling elongation in response to blue light, particularly affecting plant height, coleoptile, and first incomplete leaf length without altering root growth. This suggests that OsCSN2 mediates the inhibitory effects of blue light on aboveground development through the gibberellin signaling pathway. On day 9, the analyses of endogenous GA3 levels combined with Western blotting (WB) and quantitative real-time PCR (qRT-PCR) revealed that OsCSN2 senses blue light signals through cryptochrome 2 (CRY2), influences the expression of COP1 and BBX14, and highlights its role in the photoreceptive signaling pathway. This regulation ultimately influences the degradation of SLR1 within the GA signaling pathway, affecting rice seedling growth and development. Our findings also highlight the differential roles of OsCSN1 and OsCSN2 within the CSN in modulating rice seedling photomorphogenesis, thereby providing new insights into the intricate regulatory mechanisms governing plant responses to blue light. Full article
(This article belongs to the Section Plant Physiology and Metabolism)
Show Figures

Figure 1

20 pages, 5004 KiB  
Article
Maresin1 Alleviates Ischemia Reperfusion Injury After Lung Transplantation by Inhibiting Ferroptosis via the PKA-Hippo-YAP Signaling Pathway
by Peng Deng, You Wu, Li Wan, Xiangfu Sun and Quanchao Sun
Biomedicines 2025, 13(7), 1594; https://doi.org/10.3390/biomedicines13071594 - 30 Jun 2025
Viewed by 424
Abstract
Background: Lung ischemia reperfusion injury (LIRI) is a severe complication after lung transplantation (LT). Ferroptosis contributes to the pathogenesis of LIRI. Maresin1 (MaR1) is an endogenous pro-resolving lipid mediator that exerts protective effects against multiorgan diseases. However, the role and mechanism of [...] Read more.
Background: Lung ischemia reperfusion injury (LIRI) is a severe complication after lung transplantation (LT). Ferroptosis contributes to the pathogenesis of LIRI. Maresin1 (MaR1) is an endogenous pro-resolving lipid mediator that exerts protective effects against multiorgan diseases. However, the role and mechanism of MaR1 in the ferroptosis of LIRI after LT need to be further investigated. Methods: A mouse LT model and a pulmonary vascular endothelial cell line after hypoxia reoxygenation (H/R) culture were established in our study. Histological morphology and inflammatory cytokine levels predicted the severity of LIRI. Cell viability and cell injury were determined by CCK-8 and LDH assays. Ferroptosis biomarkers, including Fe2+, MDA, 4-HNE, and GSH, were assessed by relevant assay kits. Transferrin receptor (TFRC) and Acyl-CoA Synthetase Long Chain Family Member 4 (ACSL4) protein levels were examined by western blotting. In vitro, lipid peroxide levels were detected by DCFH-DA staining and flow cytometry analysis. The ultrastructure of mitochondria was imaged using transmission electron microscopy. Furthermore, the potential mechanism by which MaR1 regulates ferroptosis was explored and verified with signaling pathway inhibitors using Western blotting. Results: MaR1 protected mice from LIRI after LTx, which was reversed by the ferroptosis agonist Sorafenib in vivo. MaR1 administration decreased Fe2+, MDA, 4-HNE, TFRC, and ACSL4 contents, increased GSH levels, and ameliorated mitochondrial ultrastructural injury after LTx. In vitro, Sorafenib resulted in lower cell viability and worsened cell injury and enhanced the hallmarks of ferroptosis after H/R culture, which was rescued by MaR1 treatment. Mechanistically, the protein kinase A and YAP inhibitors partly blocked the effects of MaR1 on ferroptosis inhibition and LIRI protection. Conclusions: This study revealed that MaR1 alleviates LIRI and represses ischemia reperfusion-induced ferroptosis via the PKA-Hippo-YAP signaling pathway, which may offer a promising theoretical basis for the clinical application of organ protection after LTx. Full article
(This article belongs to the Special Issue The Role of Iron in Human Diseases)
Show Figures

Figure 1

35 pages, 1877 KiB  
Review
Dysregulation of the Cannabinoid System in Childhood Epilepsy: From Mechanisms to Therapy
by Gloria Montebello and Giuseppe Di Giovanni
Int. J. Mol. Sci. 2025, 26(13), 6234; https://doi.org/10.3390/ijms26136234 - 27 Jun 2025
Viewed by 1937
Abstract
Epilepsy affects over 12 million children worldwide, with approximately 30% classified as having drug-resistant epilepsy (DRE), often accompanied by neuropsychiatric comorbidities that severely impact quality of life. The endocannabinoid system (ECS) functions as a multifaceted neuromodulatory network regulating neuronal excitability, synaptic plasticity, and [...] Read more.
Epilepsy affects over 12 million children worldwide, with approximately 30% classified as having drug-resistant epilepsy (DRE), often accompanied by neuropsychiatric comorbidities that severely impact quality of life. The endocannabinoid system (ECS) functions as a multifaceted neuromodulatory network regulating neuronal excitability, synaptic plasticity, and immune homeostasis from early life through adolescence and into aging. In pediatric epilepsies, alterations in ECS components, particularly CB1 receptor expression and endocannabinoid levels, reveal disorder-specific vulnerabilities and therapeutic opportunities. Cannabidiol (CBD), a non-psychoactive compound from Cannabis sativa, has shown strong preclinical and clinical efficacy in treating DRE and is approved for Dravet syndrome, Lennox–Gastaut syndrome, and Tuberous Sclerosis Complex. Other ECS-based strategies, such as the use of CB1 receptor-positive allosteric modulators, can selectively enhance endogenous cannabinoid signaling where and when it is active, potentially reducing seizures in conditions like Dravet and absence epilepsy. Similarly, FAAH and MAGL inhibitors may help restore ECS tone without directly activating CB1 receptors. Precision targeting of ECS components based on regional expression and syndrome-specific pathophysiology may optimize seizure control and associated comorbidities. Nonetheless, long-term pediatric use must be approached with caution, given the critical role of the ECS in brain development. Full article
Show Figures

Figure 1

21 pages, 4447 KiB  
Article
The Construction of ceRNA Regulatory Network Unraveled Prognostic Biomarkers and Repositioned Drug Candidates for the Management of Pancreatic Ductal Adenocarcinoma
by Busra Aydin, Keziban Okutan, Ozge Onluturk Aydogan, Raghu Sinha and Beste Turanli
Curr. Issues Mol. Biol. 2025, 47(7), 496; https://doi.org/10.3390/cimb47070496 - 27 Jun 2025
Viewed by 442
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal cancer types due to its late diagnosis, low survival rates, and high frequency of metastasis. Considering the molecular mechanism of PDAC development has not been fully elucidated, this study aimed to shed more [...] Read more.
Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal cancer types due to its late diagnosis, low survival rates, and high frequency of metastasis. Considering the molecular mechanism of PDAC development has not been fully elucidated, this study aimed to shed more light on the molecular regulatory signatures of circular RNAs (circRNAs) in PDAC progression and provide a different perspective to identify potential biomarkers as well as discover candidate repositioned drug molecules for the prevention or treatment of PDAC with network-based integrative analysis. The mRNA, miRNA, and circRNA expression profiles of PDAC were obtained from nine microarray datasets. Differentially expressed genes (DEGs), microRNAs (DEmiRNAs), and circular RNAs (DEcircRNAs) were identified. The competing endogenous RNA (ceRNA; DEG–DEmiRNA–DEcircRNA) regulatory network was constructed, which included 12 DEcircRNAs, 64 DEGs, and 6 miRNAs specific to PDAC. The ADAM12, MET, QKI, SEC23A, and ZEB2 were identified as hub genes and demonstrated significant survival probability for PDAC. In addition to providing novel biomarkers for diagnosis that can be detected non-invasively, the secretion levels of hub genes-associated proteins were found in plasma, serum, and oral epithelium. The drug repositioning analysis revealed vorinostat, meclocycline sulfosalicylate, and trichostatin A, which exhibited significant binding affinities to the hub genes compared to their inhibitors via molecular docking analysis. Full article
Show Figures

Figure 1

14 pages, 3162 KiB  
Article
Palmitoylation Transduces the Regulation of Epidermal Growth Factor to Organic Anion Transporter 3
by Zhou Yu, Jinghui Zhang, Jiaxu Feng and Guofeng You
Pharmaceutics 2025, 17(7), 825; https://doi.org/10.3390/pharmaceutics17070825 - 25 Jun 2025
Viewed by 397
Abstract
Background: Organic anion transporter 3 (OAT3) in the kidney proximal tubule cells plays a critical role in renal clearance of numerous endogenous metabolites and exogenous drugs and toxins. In this study, we discovered that epidermal growth factor (EGF) regulates the expression and activity [...] Read more.
Background: Organic anion transporter 3 (OAT3) in the kidney proximal tubule cells plays a critical role in renal clearance of numerous endogenous metabolites and exogenous drugs and toxins. In this study, we discovered that epidermal growth factor (EGF) regulates the expression and activity of OAT3 through palmitoylation, a novel mechanism that has never been described in the OAT field. Methods/Results: Our results showed that treatment of OAT3-expressing cells with EGF led to a ~40% increase in OAT3 expression and OAT3-mediated transport of estrone sulfate, a prototypical substrate for OAT3. EGF-stimulated OAT3 transport activity was abrogated by H-89, a protein kinase A (PKA) inhibitor, indicating that an EGF-PKA signaling pathway is involved in the regulation of OAT3. We also showed that treatment of OAT3-expressing cells with EGF resulted in an enhancement of OAT3 palmitoylation, a novel type of post-translational modification for OATs, and such an enhancement was blocked by H-89, suggesting that the EGF-PKA signaling pathway participated in the modulation of OAT3 palmitoylation. Palmitoylation was catalyzed by a group of palmitoyltransfereases, and we showed that OAT3 palmitoylation and expression were inhibited by 2-BP, a general inhibitor for palmitoyltransfereases. We also explored the relationship among EGF/PKA signaling, OAT palmitoylation, and OAT transport activity. We treated OAT3-expressing cells with EGF or Bt2-cAMP, a PKA activator, in the presence and absence of 2-BP, followed by the measurement of OAT3-mediated transport of estrone sulfate. We showed that both EGF- and Bt2-cAMP-stimulated OAT3 transport activity were abolished by 2-BP, suggesting that palmitoylation mediates the regulation of EGF/PKA on OAT3. Finally, we showed that osimertinib, an anti-cancer drug/EGFR inhibitor, blocked EGF-stimulated OAT3 transport activity. Conclusions: In summary, we provided the first evidence that palmitoylation transduces the EGF/PKA signaling pathway to the modulation of OAT3 expression and function. Our study also provided an important implication that during comorbidity therapies, EGFR inhibitor drugs could potentially decrease the transport activity of renal OAT3, which would subsequently alter the therapeutic efficacy and toxicity of many co-medications that are OAT3 substrates. Full article
(This article belongs to the Section Biopharmaceutics)
Show Figures

Figure 1

17 pages, 1171 KiB  
Review
Ketone Body Induction: Insights into Metabolic Disease Management
by Byung Min Yoo, So Ra Kim and Byung-Wan Lee
Biomedicines 2025, 13(6), 1484; https://doi.org/10.3390/biomedicines13061484 - 16 Jun 2025
Cited by 1 | Viewed by 1397
Abstract
Ketone bodies (KBs), particularly β-hydroxybutyrate, are crucial metabolites that provide clean and efficient energy, especially during periods of low glucose availability. Ketogenesis is a promising therapeutic avenue for conditions such as obesity, metabolic syndrome, and diabetes. This review aims to summarize the current [...] Read more.
Ketone bodies (KBs), particularly β-hydroxybutyrate, are crucial metabolites that provide clean and efficient energy, especially during periods of low glucose availability. Ketogenesis is a promising therapeutic avenue for conditions such as obesity, metabolic syndrome, and diabetes. This review aims to summarize the current evidence on ketogenesis across different health conditions and therapeutic modalities, highlighting the potential to mitigate metabolic disorders and diabetes-related complications. By reducing inflammation and oxidative stress, increased KB production provides cardiovascular and neuroprotective benefits. Ketogenesis is enhanced under physiological conditions like pregnancy and fasting, as well as in pathophysiological states such as diabetes and heart failure. Various interventions, including the promotion of endogenous ketogenesis through diet and exercise, drug-induced ketogenesis via sodium-glucose cotransporter 2 inhibitors, and exogenous ketone supplementation, have demonstrated favorable effects on metabolic health. However, challenges remain, including risks such as pathological ketoacidosis and dyslipidemia. In specific populations, such as lean mass hyper-responders, laboratory lipid profiles might reflect the metabolic privilege. This review will assist in the future clarification of individual differences and optimized therapeutic approaches leveraging ketogenesis for the personalized management of metabolic disorders. Full article
(This article belongs to the Section Molecular and Translational Medicine)
Show Figures

Figure 1

Back to TopTop