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

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (689)

Search Parameters:
Keywords = JNK/p38MAPK/p53

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
11 pages, 231 KiB  
Article
Tempol Induces Oxidative Stress, ER Stress and Apoptosis via MAPK/Akt/mTOR Pathway Suppression in HT29 (Colon) and CRL-1739 (Gastric) Cancer Cell Lines
by Gorkem Ozdemir and Halil Mahir Kaplan
Curr. Issues Mol. Biol. 2025, 47(7), 574; https://doi.org/10.3390/cimb47070574 - 21 Jul 2025
Viewed by 219
Abstract
Tempol is a synthetic antioxidant that shows promise in preclinical cancer studies by inhibiting growth and inducing apoptosis. Given that the Mitogen-Activated Protein Kinase (MAPK) and Protein Kinase B/Mammalian Target of Rapamycin (Akt/mTOR) signaling pathways are frequently dysregulated in gastric and colon cancers [...] Read more.
Tempol is a synthetic antioxidant that shows promise in preclinical cancer studies by inhibiting growth and inducing apoptosis. Given that the Mitogen-Activated Protein Kinase (MAPK) and Protein Kinase B/Mammalian Target of Rapamycin (Akt/mTOR) signaling pathways are frequently dysregulated in gastric and colon cancers and contribute to their progression, we investigated Tempol’s anti-cancer potential in HT29 (colon) and CRL-1739 (gastric) cancer cells. Cells were treated with 2 mM Tempol for 48 h, with untreated cells as controls. We evaluated apoptosis (Bax, cleaved caspase-3, and Bcl-2), key signaling pathway activity (p-ERK, p-JNK, p-AKT, and p-mTOR), and levels of stress- and apoptosis-related proteins (WEE1, GADD153, GRP78, and AIF). Tempol significantly increased pro-apoptotic Bax and cleaved caspase-3 (p < 0.0001) and decreased anti-apoptotic Bcl-2 (p < 0.0001) in both cell lines. Furthermore, Tempol markedly reduced the activity of p-ERK, p-JNK, p-AKT, and p-mTOR (p < 0.0001) and significantly increased the protein levels of WEE1, GADD153, GRP78, and AIF (p < 0.0001). Tempol treatment also led to a significant increase in total oxidant status and a decrease in total antioxidant status. In conclusion, our findings suggest that Tempol exhibits its anti-cancer activity through multiple interconnected mechanisms, primarily inducing apoptosis and oxidative stress, while concurrently suppressing pro-survival signaling pathways. These results highlight Tempol’s potential as a therapeutic agent for gastric and colon cancers. Full article
(This article belongs to the Section Biochemistry, Molecular and Cellular Biology)
26 pages, 4733 KiB  
Article
Structural Characterization and Anti-Ultraviolet Radiation Damage Activity of Polysaccharides from Helianthus annuus (Sunflower) Receptacles
by Xiaochun Chen, Zhiying Wei, Xiaoying Mo, Yantong Lu, Guangjuan Pan, Zhenzhen Pan, Yaohua Li, Hui Tian and Xiaojiao Pan
Molecules 2025, 30(14), 2943; https://doi.org/10.3390/molecules30142943 - 11 Jul 2025
Viewed by 322
Abstract
Helianthus annuus L. (H. annuus) receptacles, a major agricultural by-product generated during seed processing, are currently underutilized. This study aimed to explore the valorization potential of this by-product by extracting H. annuus receptacles total polysaccharides (HRTP) and characterizing their potential [...] Read more.
Helianthus annuus L. (H. annuus) receptacles, a major agricultural by-product generated during seed processing, are currently underutilized. This study aimed to explore the valorization potential of this by-product by extracting H. annuus receptacles total polysaccharides (HRTP) and characterizing their potential as natural ingredients in ultraviolet (UV)-protective cosmetics. A new purified polysaccharide named H. annuus receptacles polysaccharide-1 (HRP-1) was isolated, likely exhibiting a backbone of alternating →4)-α-D-GalA-(1→ and →4)-α-D-GalA(6-OCH3)-(1→ units, with a weight-average molecular weight (Mw) of 163 kDa. HRTP demonstrated significant protective effects against UV-induced damage in human immortalized keratinocyte (HaCaT) cells by suppressing intracellular reactive oxygen species (ROS) levels and downregulating MAPK-p38/ERK/JNK pathways, thereby inhibiting inflammatory cytokines (IL-1β, IL-6, IL-8, and TNF-α) and matrix metalloproteinases (MMP-1, MMP-3, and MMP-9). Additionally, HRTP exhibited moisturizing properties. These findings highlight H. annuus receptacle polysaccharides as sustainable, bioactive ingredients for eco-friendly sunscreen formulations, providing a practical approach to converting agricultural by-products into high-value industrial biomaterials. Full article
Show Figures

Graphical abstract

23 pages, 4624 KiB  
Review
Farnesoid X Receptor (FXR) Agonists and Protein Kinase Regulation in NAFLD and NASH: Mechanisms and Therapeutic Potential
by Ayan Saha, Emily Wood, Luna Omeragic, Maya Minkara, Kethain Marma, Shipan Das Gupta and Jannatul Ferdoush
Kinases Phosphatases 2025, 3(3), 16; https://doi.org/10.3390/kinasesphosphatases3030016 - 11 Jul 2025
Viewed by 712
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a common metabolic condition characterized by hepatic lipid deposits, insulin resistance, and inflammation which may progress to non-alcoholic steatohepatitis (NASH) and fibrosis. Protein kinases play an important role in NAFLD development by regulating metabolic and inflammatory pathways. [...] Read more.
Non-alcoholic fatty liver disease (NAFLD) is a common metabolic condition characterized by hepatic lipid deposits, insulin resistance, and inflammation which may progress to non-alcoholic steatohepatitis (NASH) and fibrosis. Protein kinases play an important role in NAFLD development by regulating metabolic and inflammatory pathways. Mitogen-activated protein kinases (MAPKs), protein kinase C (PKC), AMP-activated protein kinase (AMPK), phosphoinositide 3-kinase (PI3K)/AKT, and mechanistic target of rapamycin (mTOR) are all involved in NAFLD and NASH progression. Emerging evidence indicates that Farnesoid X Receptor (FXR) agonists have therapeutic potential by modulating bile acid metabolism, lipid balance, and inflammatory responses. This review examines the mechanistic interplay between FXR agonists and important protein kinases in NAFLD and NASH. FXR agonists activate AMPK, which promotes fatty acid oxidation and reduces hepatic steatosis. They also regulate MAPK signaling, which reduces c-Jun NH2-terminal kinase (JNK)- and p38 MAPK-mediated inflammation. Furthermore, FXR agonists activate the PI3K/AKT pathway, enhancing insulin sensitivity and modulating mTOR signaling to reduce hepatic fibrosis. Clinical studies in NAFLD/NASH indicate that FXR agonists confer metabolic and anti-inflammatory benefits, although optimizing efficacy and minimizing adverse effects remain challenging. Future studies should focus on combination therapies targeting FXR alongside specific kinases to improve therapeutic outcomes. This review highlights the potential of FXR agonists to modulate protein kinase signaling, opening new avenues for targeted NAFLD/NASH therapy. Full article
Show Figures

Figure 1

20 pages, 2891 KiB  
Review
MAPK, PI3K/Akt Pathways, and GSK-3β Activity in Severe Acute Heart Failure in Intensive Care Patients: An Updated Review
by Massimo Meco, Enrico Giustiniano, Fulvio Nisi, Pierluigi Zulli and Emiliano Agosteo
J. Cardiovasc. Dev. Dis. 2025, 12(7), 266; https://doi.org/10.3390/jcdd12070266 - 10 Jul 2025
Viewed by 595
Abstract
Acute heart failure (AHF) is a clinical syndrome characterized by the sudden onset or rapid worsening of heart failure signs and symptoms, frequently triggered by myocardial ischemia, pressure overload, or cardiotoxic injury. A central component of its pathophysiology is the activation of intracellular [...] Read more.
Acute heart failure (AHF) is a clinical syndrome characterized by the sudden onset or rapid worsening of heart failure signs and symptoms, frequently triggered by myocardial ischemia, pressure overload, or cardiotoxic injury. A central component of its pathophysiology is the activation of intracellular signal transduction cascades that translate extracellular stress into cellular responses. Among these, the mitogen-activated protein kinase (MAPK) pathways have received considerable attention due to their roles in mediating inflammation, apoptosis, hypertrophy, and adverse cardiac remodeling. The canonical MAPK cascades—including extracellular signal-regulated kinases (ERK1/2), p38 MAPK, and c-Jun N-terminal kinases (JNK)—are activated by upstream stimuli such as angiotensin II (Ang II), aldosterone, endothelin-1 (ET-1), and sustained catecholamine release. Additionally, emerging evidence highlights the role of receptor-mediated signaling, cellular stress, and myeloid cell-driven coagulation events in linking MAPK activation to fibrotic remodeling following myocardial infarction. The phosphatidylinositol 3-kinase (PI3K)/Akt signaling cascade plays a central role in regulating cardiomyocyte survival, hypertrophy, energy metabolism, and inflammation. Activation of the PI3K/Akt pathway has been shown to confer cardioprotective effects by enhancing anti-apoptotic and pro-survival signaling; however, aberrant or sustained activation may contribute to maladaptive remodeling and progressive cardiac dysfunction. In the context of AHF, understanding the dual role of this pathway is crucial, as it functions both as a marker of compensatory adaptation and as a potential therapeutic target. Recent reviews and preclinical studies have linked PI3K/Akt activation with reduced myocardial apoptosis and attenuation of pro-inflammatory cascades that exacerbate heart failure. Among the multiple signaling pathways involved, glycogen synthase kinase-3β (GSK-3β) has emerged as a key regulator of apoptosis, inflammation, metabolic homeostasis, and cardiac remodeling. Recent studies underscore its dual function as both a negative regulator of pathological hypertrophy and a modulator of cell survival, making it a compelling therapeutic candidate in acute cardiac settings. While earlier investigations focused primarily on chronic heart failure and long-term remodeling, growing evidence now supports a critical role for GSK-3β dysregulation in acute myocardial stress and injury. This comprehensive review discusses recent advances in our understanding of the MAPK signaling pathway, the PI3K/Akt cascade, and GSK-3β activity in AHF, with a particular emphasis on mechanistic insights, preclinical models, and emerging therapeutic targets. Full article
(This article belongs to the Topic Molecular and Cellular Mechanisms of Heart Disease)
Show Figures

Figure 1

22 pages, 5021 KiB  
Article
Luteolin-Rich Extract from Harrisonia perforata (Blanco) Merr. Root Alleviates SARS-CoV-2 Spike Protein-Stimulated Lung Inflammation via Inhibition of MAPK/NLRP3 Inflammasome Signaling Pathways
by Warathit Semmarath, Punnida Arjsri, Kamonwan Srisawad, Sonthaya Umsumarng and Pornngarm Dejkriengkraikul
Life 2025, 15(7), 1077; https://doi.org/10.3390/life15071077 - 5 Jul 2025
Viewed by 377
Abstract
The COVID-19-related long-standing effect or Post-Acute Sequelae of COVID-19 (PASC) is often associated with NLRP3 inflammasome activation in pulmonary inflammation elicited by SARS-CoV-2 spike proteins. Spike proteins engage toll-like receptors (TLRs) in respiratory epithelial cells, leading to excessive cytokine production. Given the need [...] Read more.
The COVID-19-related long-standing effect or Post-Acute Sequelae of COVID-19 (PASC) is often associated with NLRP3 inflammasome activation in pulmonary inflammation elicited by SARS-CoV-2 spike proteins. Spike proteins engage toll-like receptors (TLRs) in respiratory epithelial cells, leading to excessive cytokine production. Given the need for effective therapeutic strategies to mitigate spike protein-stimulated lung inflammation, we examined the anti-inflammatory properties of luteolin and ethanolic extract from Harrisonia perforata (Blanco) Merr. root. The ethanolic extract of H. perforata root (HPEE) contained a high concentration of luteolin flavonoid (143.53 ± 1.58 mg/g extract). Both HPEE (25–100 μg/mL) and luteolin (4.5–36 μM) significantly inhibited inflammation stimulated by the Wuhan (W) and Omicron (O) spike protein S1, as evidenced by a dose-dependent significant decrease in IL-6, IL-1β, and IL-18 secretion in A549 lung epithelial cells (p < 0.05). Furthermore, pretreatment with HPEE or luteolin prior to spike protein exposure (100 ng/mL) significantly, in a dose-dependent manner, repressed the inflammatory mRNA expression (p < 0.05). Mechanistic study revealed that HPEE and luteolin suppressed NLRP3 inflammasome signaling activation by reducing their machinery protein expressions. Additionally, they inhibited the ERK/JNK/p38 MAPK signaling activation, resulting in decreased inflammatory mRNA expression and cytokine release. These findings suggest that H. perforata root extract and its major flavonoid luteolin exert potent anti-inflammatory effects and may offer therapeutic potential against spike protein-induced lung inflammation. Full article
Show Figures

Figure 1

34 pages, 765 KiB  
Review
Transcription Factors and Methods for the Pharmacological Correction of Their Activity
by Svetlana V. Guryanova, Tatiana V. Maksimova and Madina M. Azova
Int. J. Mol. Sci. 2025, 26(13), 6394; https://doi.org/10.3390/ijms26136394 - 2 Jul 2025
Viewed by 731
Abstract
Transcription factors (TFs) are proteins that control gene expression by binding to specific DNA sequences and are essential for cell development, differentiation, and homeostasis. Dysregulation of TFs is implicated in numerous diseases, including cancer, autoimmune disorders, and neurodegeneration. While TFs were traditionally considered [...] Read more.
Transcription factors (TFs) are proteins that control gene expression by binding to specific DNA sequences and are essential for cell development, differentiation, and homeostasis. Dysregulation of TFs is implicated in numerous diseases, including cancer, autoimmune disorders, and neurodegeneration. While TFs were traditionally considered “undruggable” due to their lack of well-defined binding pockets, recent advances have made it possible to modulate their activity using diverse pharmacological strategies. Major TF families include NF-κB, p53, STATs, HIF-1α, AP-1, Nrf2, and nuclear hormone receptors, which take part in the regulation of inflammation, tumor suppression, cytokine signaling, hypoxia and stress response, oxidative stress, and hormonal response, respectively. TFs can perform multiple functions, participating in the regulation of opposing processes depending on the context. NF-κB, for instance, plays dual roles in immunity and cancer, and is targeted by proteasome and IKKβ inhibitors. p53, often mutated in cancer, is reactivated using MDM2 antagonist Nutlin-3, refunctionalizing compound APR-246, or stapled peptides. HIF-1α, which regulates hypoxic responses and angiogenesis, is inhibited by agents like acriflavine or stabilized in anemia therapies by HIF-PHD inhibitor roxadustat. STATs, especially STAT3 and STAT5, are oncogenic and targeted via JAK inhibitors or novel PROTAC degraders, for instance SD-36. AP-1, implicated in cancer and arthritis, can be inhibited by T-5224 or kinase inhibitors JNK and p38 MAPK. Nrf2, a key antioxidant regulator, can be activated by agents like DMF or inhibited in chemoresistant tumors. Pharmacological strategies include direct inhibitors, activators, PROTACs, molecular glues, and epigenetic modulators. Challenges remain, including the structural inaccessibility of TFs, functional redundancy, off-target effects, and delivery barriers. Despite these challenges, transcription factor modulation is emerging as a viable and promising therapeutic approach, with ongoing research focusing on specificity, safety, and efficient delivery methods to realize its full clinical potential. Full article
(This article belongs to the Topic Research in Pharmacological Therapies, 2nd Edition)
Show Figures

Figure 1

13 pages, 4302 KiB  
Article
Analysis of Processing Impact on Raspberries Based on Broad-Spectrum Metabolomics
by Xiaoge Wang, Qiyuan Liao, Fan Wang, Xuelin Rui, Yushan Liu and Rui Wang
Metabolites 2025, 15(7), 435; https://doi.org/10.3390/metabo15070435 - 26 Jun 2025
Viewed by 361
Abstract
Objective: Our objective was to explore the regulatory mechanism of salt processing on the metabolome of the raspberry and its potential efficacy against diabetic nephropathy (DN), providing metabolomic and network pharmacological evidence for the scientific connotation of traditional Chinese medicine processing. Methods: Ultra-high-performance [...] Read more.
Objective: Our objective was to explore the regulatory mechanism of salt processing on the metabolome of the raspberry and its potential efficacy against diabetic nephropathy (DN), providing metabolomic and network pharmacological evidence for the scientific connotation of traditional Chinese medicine processing. Methods: Ultra-high-performance liquid chromatography–tandem mass spectrometry (UHPLC-MS/MS)-based metabolomics was used to compare the metabolic profiles between raw and salt-processed raspberries. Network pharmacology was applied to screen the common targets of the active components in the salt-processed raspberry and DN-related pathways, followed by in vitro cell experiments to validate the regulation of the MAPK signaling pathway. Results: The metabolomic analysis identified 80 differentially expressed metabolites, among which 13 key components (VIP ≥ 1, FC ≥ 2) were significantly altered, including enriched flavonoids (e.g., luteolin-7-O-glucoside), triterpenoid saponins (Raspberryides H/F), and phenolic acids (ellagic acid). The network pharmacology revealed that the salt-processed raspberries regulated the DN-related pathways through 122 common targets, with the core nodes focusing on the signaling molecules (e.g., AKT1, EGFR) involved in the MAPK signaling pathway and apoptosis regulation. The in vitro experiments confirmed that the salt-processed raspberry extract (160–640 μg/mL) significantly inhibited the phosphorylation levels of p38/ERK/JNK in high-glucose-induced renal cells. Conclusions: This study firstly combines metabolomics and network pharmacology to reveal the regulatory mechanism of salt processing on the active components of raspberries. The salt-processing technology enhanced the inhibitory effect of raspberries on the MAPK signaling pathway, thereby ameliorating the progression of DN. These findings provide scientific support for establishing a metabolomics-based quality control system for traditional Chinese medicine processing. The current findings are primarily based on in vitro models, and in vivo validation using DN animal models is essential to confirm the therapeutic efficacy and safety of salt-processed raspberries. Full article
Show Figures

Figure 1

22 pages, 3867 KiB  
Article
Neuroprotective Mechanisms of Porcine Brain Enzyme Hydrolysate in Memory Impairment: Multi-Target Strategy Against Amyloid-β-Induced Neurotoxicity
by Sun Myung Yoon, Ye-Won Lee, Min Ju Kim, Jae-Joon Shin, Gun Won Bae and Sunmin Park
Int. J. Mol. Sci. 2025, 26(13), 6030; https://doi.org/10.3390/ijms26136030 - 24 Jun 2025
Viewed by 474
Abstract
This study investigated the potential neuroprotective mechanisms of porcine brain enzyme hydrolysate (PBEH) against Alzheimer’s disease pathology using differentiated SH-SY5Y cells. Differentiated neuronal cells were treated with 40 μM amyloid-β(1-42; Aβ) to induce neurotoxicity, followed by PBEH treatment (12.5–400 μg/mL), Com-A (peptide-based neuroprotective [...] Read more.
This study investigated the potential neuroprotective mechanisms of porcine brain enzyme hydrolysate (PBEH) against Alzheimer’s disease pathology using differentiated SH-SY5Y cells. Differentiated neuronal cells were treated with 40 μM amyloid-β(1-42; Aβ) to induce neurotoxicity, followed by PBEH treatment (12.5–400 μg/mL), Com-A (peptide-based neuroprotective supplement; 200 μg/mL) treatment, and Com-B (herbal extract known for improving memory function; 100 μg/mL) treatment. Key assessments included cell viability, Aβ aggregation in adding 10 μM Aβ, amyloidogenic proteins (APP, BACE), synaptic markers (BDNF, ERK), apoptotic markers (BAX/BCL-2, caspase-3), oxidative stress (reactive oxygen species (ROS)), cholinergic function (ChAT, AChE), MAPK signaling (JNK, p38), and neuroinflammation (IL-1β). PBEH contained high concentrations of amino acids, including L-lysine (32.3 mg/g), L-leucine (42.4 mg/g), L-phenylalanine (30.0 mg/g) and the PSIS peptide (86.9 μg/g). Treatment up to 400 μg/mL showed no cytotoxicity and had cognitive protection effects up to 152% under Aβ stress (p < 0.05). PBEH significantly attenuated Aβ aggregation, decreased APP (28%) and BACE (51%) expression, enhanced synaptic function through increased BDNF, and restored ERK phosphorylation (p < 0.05). Anti-apoptotic effects included a 76% reduction in the BAX/BCL-2 ratio, a 47% decrease in caspase-3, and a 56% reduction in ROS levels. Cholinergic function showed restoration via increased ChAT activity (p < 0.01) and decreased AChE activity (p < 0.05). PBEH reduced IL-1β levels by 70% and suppressed JNK/p38 phosphorylation (p < 0.05). While Com-A enhanced BDNF and Com-B showed anti-inflammatory effects, PBEH demonstrated activity across multiple pathway markers. In conclusion, these findings suggest that PBEH may enable neuronal preservation through multi-pathway modulation, establishing foundational evidence for further mechanistic investigation in cognitive enhancement applications. Full article
(This article belongs to the Special Issue Molecular Mechanisms of Alzheimer’s Disease)
Show Figures

Figure 1

15 pages, 2958 KiB  
Article
Isostrictiniin Alleviates LPS-Induced Acute Lung Injury via the Regulation of the Keap1-Nrf2/HO-1 and MAPK/NF-κB Signaling Pathways
by Wanting Ding, Yuan Sun, Wulipan Tuohudaali, Chenyang Li, Yuhan Yao and Jun Zhao
Int. J. Mol. Sci. 2025, 26(12), 5912; https://doi.org/10.3390/ijms26125912 - 19 Jun 2025
Viewed by 564
Abstract
This study aimed to investigate the preventive effects of isostrictiniin (ITN) from Nymphaea candida against acute lung injury (ALI) through lipopolysaccharide (LPS)-induced ALI mice and LPS-induced A549 cells. Compared with the model group, ITN (50 and 100 mg/kg) significantly reduced the lung indexes, [...] Read more.
This study aimed to investigate the preventive effects of isostrictiniin (ITN) from Nymphaea candida against acute lung injury (ALI) through lipopolysaccharide (LPS)-induced ALI mice and LPS-induced A549 cells. Compared with the model group, ITN (50 and 100 mg/kg) significantly reduced the lung indexes, W/D rates, BALF WBC counts, and total protein contents in ALI mice (p < 0.05), as well as the blood neu counts (p < 0.01), while increasing the blood lym counts (p < 0.01). ITN (50 and 100 mg/kg) also markedly decreased the lung tissue TNF-α, IL-6, IL-1β, MDA, and MPO activities in ALI mice (p < 0.01) and enhanced the SOD and GSH levels (p < 0.01). Additionally, ITN (50 and 100 mg/kg) significantly improved lung histopathological damage in ALI mice. Moreover, ITN (10 and 25 µM) significantly reduced the NO, PGE2, IL-1β, IL-6, TNF-α, and MDA levels in LPS-induced A549 cells (p < 0.01) while significantly increasing the SOD and GSH activities (p < 0.01). After LPS-induced A549 cells, the Keap1, p-JNK/JNK, p-ERK1/2/ERK1/2, p-P38/P38, p-IκBα/IκBα, and p-NF-κBp65/NF-κB p65 levels were significantly upregulated (p < 0.05), whereas the Nrf2 and HO-1 protein expressions were downregulated (p < 0.05). After treatment with ITN (25 μM), the changes in these relative protein expressions in LPS-induced A549 cells were significantly reversed (p < 0.05). The above results indicate that ITN has a better preventive effect against ALI, and its mechanisms are related to the regulation of the Keap1-Nrf2/HO-1 and MAPK/NF-κB signaling pathways. Full article
(This article belongs to the Special Issue Antioxidants: The Molecular Guardians Against Oxidative Stress)
Show Figures

Figure 1

19 pages, 3569 KiB  
Article
Bladder p75NTR-Mediated Anti-Inflammatory Response via the TLR4/TRAF6/NF-κB Axis
by Claudia Covarrubias, Abubakr H. Mossa, Laura R. Yan, Benjamin Desormeau, Philippe G. Cammisotto, H. Uri Saragovi and Lysanne Campeau
Life 2025, 15(6), 957; https://doi.org/10.3390/life15060957 - 14 Jun 2025
Viewed by 2391
Abstract
Recurrent bacterial cystitis in women can lead to interstitial cystitis or bladder pain syndrome (IC/BPS). Activation of Toll-like receptor 4 (TLR4) by LPS can upregulate signaling of the pro-inflammatory receptor p75NTR. The aim of the presented study was to assess whether [...] Read more.
Recurrent bacterial cystitis in women can lead to interstitial cystitis or bladder pain syndrome (IC/BPS). Activation of Toll-like receptor 4 (TLR4) by LPS can upregulate signaling of the pro-inflammatory receptor p75NTR. The aim of the presented study was to assess whether p75NTR antagonist THX-B can modulate LPS-mediated inflammation in bladder cells. In vitro expression and LPS-activation of p75NTR were confirmed in urothelial (URO) and smooth muscle (SMC) cells. In UROs, p75NTR antagonism abolished the LPS-elicited rise in membrane-bound and soluble TNF-α. However, it could not prevent LPS-induced rise in phosphorylated ERK nor decrease in phosphorylated p38MAPK, nor the increase in iNOS and nitric oxide (NO) content. On the other hand, in SMCs, LPS increased phosphorylation of JNK, nuclear translocation of NF-κB, and association of TRAF6 to p75NTR, outcomes prevented by p75NTR antagonism. In UROs, LPS decreased the expression of tight junction proteins, ZO-1 and occludin, with the latter rescued by p75NTR antagonism. Intraurethral instillation of LPS increased inflammation in the lamina propria, activation of JNK, and contractile activity of bladder tissue. Alternatively, intraperitoneal THX-B injections prevented LPS-induced inflammation but not enhanced muscle contraction. Our results suggest that inhibition of p75NTR could help in reducing bladder symptoms during cystitis. Full article
(This article belongs to the Section Microbiology)
Show Figures

Figure 1

20 pages, 5381 KiB  
Article
Role of Central Inflammatory and Oxidative Pathways in the Morphine Exacerbation of Cardiovascular Effects of Sepsis in Rats
by Mohamed Abdelnaby, Marwa Y. Sallam, Mai M. Helmy, Hanan M. El-Gowelli and Mahmoud M. El-Mas
Pharmaceuticals 2025, 18(6), 882; https://doi.org/10.3390/ph18060882 - 12 Jun 2025
Viewed by 579
Abstract
Background/Objectives: Sepsis remains one of the most serious and possibly fatal complications encountered in intensive care units. Considering the frequent use of narcotic analgesics in this setting, we investigated whether the cardiovascular and peripheral and central inflammatory features of sepsis could be modified [...] Read more.
Background/Objectives: Sepsis remains one of the most serious and possibly fatal complications encountered in intensive care units. Considering the frequent use of narcotic analgesics in this setting, we investigated whether the cardiovascular and peripheral and central inflammatory features of sepsis could be modified by morphine. Methods: Rats were instrumented with femoral and intracisternal (i.c.) indwelling catheters and sepsis was induced by cecal ligation and puncture (CLP). Results: The i.v. administration of morphine (3 and 10 mg/kg) significantly and dose-dependently aggravated septic manifestations of hypotension and impaired cardiac autonomic activity, as reflected by the reductions in indices of heart rate variability (HRV). Cardiac contractility (dP/dtmax) was also reduced by morphine in septic rats. The morphine effects were mostly eliminated following (i) blockade of μ-opioid receptors by i.v. naloxone and (ii) inhibition of central PI3K, MAPK-ERK, MAPK-JNK, NADPH oxidase (NADPHox), or Rho-kinase (ROCK) by i.c. wortmannin, PD98059, SP600125, diphenyleneiodonium, and fasudil, respectively. Further, these pharmacologic interventions significantly reduced the heightened protein expression of toll-like receptor 4 (TLR4) and monocyte chemoattractant protein-1 (MCP1) in brainstem rostral ventrolateral medullary (RVLM), but not cardiac, tissues of CLP/morphine-treated rats. Conclusions: Morphine worsens cardiovascular and autonomic disturbances caused by sepsis through a mechanism mediated via μ-opioid receptors and upregulated central inflammatory, chemotactic, and oxidative signals. Clinical studies are warranted to re-affirm the adverse cardiovascular interaction between opioids and the septic challenge. Full article
(This article belongs to the Special Issue Pharmacology and Toxicology of Opioids)
Show Figures

Graphical abstract

17 pages, 4963 KiB  
Article
Characterization and Functional Analysis of a Novel Fungal Immunomodulatory Protein Gene from Ganoderma leucocontextum in B16-F10 Mouse Melanoma Cells
by Jiayi Yang, Mengyuan Jin, Lida Zhang, Yingying Wu and Xuanwei Zhou
Int. J. Mol. Sci. 2025, 26(11), 5063; https://doi.org/10.3390/ijms26115063 - 24 May 2025
Viewed by 515
Abstract
Ganoderma leucocontextum, a newly identified species from the Tibetan Plateau, has been mainly studied for its polysaccharides and triterpenoids, with no prior reports on fungal immunomodulatory proteins (FIPs). This study explores the biological activity of FIP-gle2, cloned from G. leucocontextum and expressed [...] Read more.
Ganoderma leucocontextum, a newly identified species from the Tibetan Plateau, has been mainly studied for its polysaccharides and triterpenoids, with no prior reports on fungal immunomodulatory proteins (FIPs). This study explores the biological activity of FIP-gle2, cloned from G. leucocontextum and expressed in Pichia pastoris. The effects and mechanisms of recombinant FIP-gle2 (rFIP-gle2) on cell activity and melanin synthesis in mouse melanoma B16-F10 cells were investigated in vitro. The results showed that the FIP-gle2 gene, with an open reading frame (ORF) of 333 bp, encodes a 111-amino acid polypeptide with a molecular weight of 12.60 kDa and an isoelectric point of 4.48. We achieved a yield of 184.18 mg/L of rFIP-gle2. In vitro functional experiments showed that rFIP-gle2 significantly inhibited the proliferation of B16-F10 melanoma cells and induced apoptosis in a dose-dependent manner, particularly at concentrations above 1 μg/mL. At 3 μg/mL, rFIP-gle2 effectively inhibited tyrosinase activity and reduced melanin content, downregulating microphthalmia-associated transcription factor (MITF), tyrosinase (TYR), and tyrosinase-related proteins (TRP-1 and TRP-2). Furthermore, RNA-seq analysis indicated that differentially expressed genes in treated cells were enriched in the mitogen-activated protein kinase (MAPK) signaling pathway, with Western blotting confirming enhanced phosphorylation of JNK, ERK, and p38 proteins. Thus, P. pastoris is an effective host for rFIP-gle2 production, which shows potential for applications in pharmaceuticals, cosmeceuticals, and food fields. Full article
(This article belongs to the Special Issue Anticancer Drug Discovery Based on Natural Products)
Show Figures

Graphical abstract

19 pages, 3614 KiB  
Article
Immunoenhancement Function of the Novel Hexapeptide (LVVLGH) from Thick-Shelled Mussel (Mytilus coruscus) on Immunodeficient Mice by Activating the NF-κB/MAPK Pathway
by Xu Yang, Yu Zeng, Fandi Que, Shiqing Fu, Li Xu, Fangmiao Yu and Bin Wang
Foods 2025, 14(11), 1865; https://doi.org/10.3390/foods14111865 - 24 May 2025
Viewed by 551
Abstract
A novel hexapeptide LVVLGH (LH-6) from the thick-shelled mussel (Mytilus coruscus) demonstrated potent immune-enhancing effects in RAW264.7 cells in vitro, but its immunological activity in vivo is unclear. As a result, the present study was designed to investigate the in vivo [...] Read more.
A novel hexapeptide LVVLGH (LH-6) from the thick-shelled mussel (Mytilus coruscus) demonstrated potent immune-enhancing effects in RAW264.7 cells in vitro, but its immunological activity in vivo is unclear. As a result, the present study was designed to investigate the in vivo effects of LH-6 on cyclophosphamide-induced immunodeficient mice. The results demonstrate that LH-6 promoted the growth and development of immunodeficient mice in a concentration-dependent manner, remarkably elevated the immune organ index, and relieved the pathological characteristics of the spleen and thymus. Additional experiments also revealed that LH-6 effectively promoted the multiplication of splenic lymphocytes and natural killer activity, enhanced the function of abdominal macrophages, and apparently recovered delayed-type hypersensitivity in immunodeficient mice. The secretion of IgA, IgG, IgM, TNF-α, IL-1β, IL-6, and serum hemolysin were remarkably improved by LH-6, suggesting that LH-6 can synergistically strengthen cellular and humoral immunity. In addition, LH-6 promoted the phosphorylation of IκBα and nuclear translocation of p65, which correspondingly increased the phosphorylation levels of p38, JNK, and ERK; activated the NF-κB and MAPK pathways; and exerted in vivo immunomodulatory activities. Docking results show that LH-6 has favorable binding energies to candidate proteins in the NF-κB and MAPK pathways. To summarize, this research further demonstrated that LH-6 possesses in vivo immunomodulatory activity, which provides a possibility for the subsequent development of immune-enhancing functional foods. Full article
Show Figures

Figure 1

13 pages, 1682 KiB  
Article
Dihydrocapsaicin Enhances Tumor Necrosis Factor-α-Induced Apoptosis and G1 Cell Cycle Arrest in Human Cervical Cancer Cells Through TAK1-Mediated NF-κB and EGFR Pathways
by Chantana Boonyarat, Hiroaki Sakurai, Yoshihiro Hayakawa, Suchada Chaiwiwatrakul, Rawiwun Kaewamatawong, Teeraporn Sadira Supapaan, Sureewan Duangjit, Benjabhorn Sethabouppha and Pornthip Waiwut
Int. J. Mol. Sci. 2025, 26(11), 5011; https://doi.org/10.3390/ijms26115011 - 22 May 2025
Viewed by 619
Abstract
Dihydrocapsaicin (DHC), a prominent capsaicinoid derived from red chili peppers, has shown cytotoxic effects against various cancer cell types. However, its role in modulating cytokine-induced survival and apoptotic signaling in cancer cells remains unclear. In this study, we investigated the effects of DHC [...] Read more.
Dihydrocapsaicin (DHC), a prominent capsaicinoid derived from red chili peppers, has shown cytotoxic effects against various cancer cell types. However, its role in modulating cytokine-induced survival and apoptotic signaling in cancer cells remains unclear. In this study, we investigated the effects of DHC on tumor necrosis factor-α (TNF-α)-induced cell cycle arrest and apoptosis in HeLa human cervical cancer cells. Our results demonstrate that DHC significantly enhances TNF-α-induced G1 phase cell cycle arrest and apoptosis by targeting the transforming growth factor-β-activated kinase 1 (TAK1)-mediated prosurvival pathways. DHC inhibited the phosphorylation of TAK1 and downstream effectors including IKKα, NF-κB p65, MAPKs (p38, JNK, ERK), Akt, and EGFR, thereby disrupting key signaling networks that typically confer resistance to TNF-α-induced cytotoxicity. Additionally, DHC suppressed the TNF-α-induced phosphorylation of EGFR at Ser-1046/1047 and Thr-669, sites critical for survival signaling. Co-treatment with DHC and TNF-α led to enhanced apoptotic features, including increased PARP-1 cleavage. These findings suggest that DHC sensitizes cervical cancer cells to cytokine-induced cell death by interfering with TAK1/NF-κB and EGFR signaling axes. Our study positions DHC as a promising candidate for combination therapies aimed at overcoming resistance in cancers with aberrant inflammatory and survival signaling. Full article
(This article belongs to the Special Issue The Role of Natural Compounds in Cancer and Inflammation)
Show Figures

Figure 1

17 pages, 2112 KiB  
Article
Mitigation of 3.5 GHz Electromagnetic Field-Induced BV2 Microglial Cytotoxicity by Polydeoxyribonucleotide
by Shailashree Pachhapure, Amila Mufida, Qun Wei, Jong-Soon Choi and Byeong-Churl Jang
Curr. Issues Mol. Biol. 2025, 47(6), 386; https://doi.org/10.3390/cimb47060386 - 22 May 2025
Viewed by 706
Abstract
Emerging evidence highlights the biological risks associated with electromagnetic fields (EMFs) generated by electronic devices. The toxic effects and mechanisms induced by exposure to EMFs on microglial cells and natural substances that inhibit them are limited to date. Here, we investigated whether exposure [...] Read more.
Emerging evidence highlights the biological risks associated with electromagnetic fields (EMFs) generated by electronic devices. The toxic effects and mechanisms induced by exposure to EMFs on microglial cells and natural substances that inhibit them are limited to date. Here, we investigated whether exposure to 3.5 GHz EMF radiation, potentially generated by smartphones working in 5G communication or cooking using microwave ovens, affects the growth of BV2 mouse microglial cells and polydeoxyribonucleotide (PDRN), a DNA preparation derived from salmon sperm, inhibits it. Of note, exposure to 3.5 GHz EMF radiation for 2 h markedly inhibited the growth and triggered apoptosis in BV2 cells, characterized by the reduced number of surviving cells, increased genomic DNA fragmentation, increased reactive oxygen species (ROS) levels, and altered phosphorylation and expression levels of JNK-1/2, p38 MAPK, ERK-1/2, eIF-2α, and procaspase-9. Pharmacological inhibition studies revealed that JNK-1/2 and p38 MAPK activation and ROS generation were crucial for 3.5 GHz EMF-induced BV2 cytotoxicity. Of interest, PDRN effectively countered these effects by inhibiting the activation of JNK-1/2, p38 MAPK, and caspase-9, and the production of ROS, although it did not affect eIF-2 phosphorylation. In conclusion, this study is the first to report that PDRN protects against 3.5 GHz EMF-induced toxicities in BV2 microglial cells, and PDRN’s protective effects on 3.5 GHz EMF-induced BV2 cytotoxicity are mediated primarily by modulating ROS, JNK-1/2, p38 MAPK, and caspase-9. Full article
(This article belongs to the Special Issue Radiation-Induced Cellular and Molecular Responses)
Show Figures

Graphical abstract

Back to TopTop