Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.235
Filter
1.
Zool Res ; 45(5): 1037-1047, 2024 Sep 18.
Article in English | MEDLINE | ID: mdl-39147718

ABSTRACT

Serine protease 50 (PRSS50/TSP50) is highly expressed in spermatocytes. Our study investigated its role in testicular development and spermatogenesis. Initially, PRSS50 knockdown was observed to impair DNA synthesis in spermatocytes. To further explore this, we generated PRSS50 knockout ( Prss50 -/- ) mice ( Mus musculus), which exhibited abnormal spermatid nuclear compression and reduced male fertility. Furthermore, dysplastic seminiferous tubules and decreased sex hormones were observed in 4-week-old Prss50 -/- mice, accompanied by meiotic progression defects and increased apoptosis of spermatogenic cells. Mechanistic analysis indicated that PRSS50 deletion resulted in increased phosphorylation of extracellular signal-regulated protein kinases 1 and 2 (ERK1/2) and elevated levels of MAP kinase phosphatase 3 (MKP3), a specific ERK antagonist, potentially accounting for testicular dysplasia in adolescent Prss50 -/- mice. Taken together, these findings suggest that PRSS50 plays an important role in testicular development and spermatogenesis, with the MKP3/ERK signaling pathway playing a significant role in this process.


Subject(s)
MAP Kinase Signaling System , Meiosis , Mice, Knockout , Spermatozoa , Animals , Male , Mice , Meiosis/physiology , Spermatozoa/physiology , Spermatogenesis/physiology , Dual Specificity Phosphatase 6/genetics , Dual Specificity Phosphatase 6/metabolism , Testis/metabolism , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism
2.
J Biochem Mol Toxicol ; 38(9): e23806, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39148258

ABSTRACT

Exposure to fine particulate matter with an aerodynamic diameter of less than 2.5 µm (PM2.5) can cause oxidative damage and apoptosis in the human skin. Chlorogenic acid (CGA) is a bioactive polyphenolic compound with antioxidant, antifungal, and antiviral properties. The objective of this study was to identify the ameliorating impact of CGA that might protect human HaCaT cells against PM2.5. CGA significantly scavenged the reactive oxygen species (ROS) generated by PM2.5, attenuated oxidative cellular/organelle damage, mitochondrial membrane depolarization, and suppressed cytochrome c release into the cytosol. The application of CGA led to a reduction in the expression levels of Bcl-2-associated X protein, caspase-9, and caspase-3, while simultaneously increasing the expression of B-cell lymphoma 2. In addition, CGA was able to reverse the decrease in cell viability caused by PM2.5 via the inhibition of extracellular signal-regulated kinase (ERK). This effect was further confirmed by the use of the mitogen-activated protein kinase kinase inhibitor, which acted upstream of ERK. In conclusion, CGA protected keratinocytes from mitochondrial damage and apoptosis via ameliorating PM2.5-induced oxidative stress and ERK activation.


Subject(s)
Apoptosis , Chlorogenic Acid , Keratinocytes , Oxidative Stress , Particulate Matter , Chlorogenic Acid/pharmacology , Humans , Apoptosis/drug effects , Oxidative Stress/drug effects , Particulate Matter/toxicity , Keratinocytes/drug effects , Keratinocytes/metabolism , Keratinocytes/pathology , Reactive Oxygen Species/metabolism , HaCaT Cells , Cell Survival/drug effects , Membrane Potential, Mitochondrial/drug effects , MAP Kinase Signaling System/drug effects
3.
J Cell Mol Med ; 28(16): e70016, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39175122

ABSTRACT

Natural killer (NK) cell therapy, a developing approach in cancer immunotherapy, involves isolating NK cells from peripheral blood. However, due to their limited number and activity, it is essential to significantly expand these primary NK cells and enhance their cytotoxicity. In this study, we investigated how Raddeanin A potentiate NK activity using KHYG-1 cells. The results indicated that Raddeanin A increased the expression levels of cytolytic molecules such as perforin, granzymes A and granzymes B, granulysin and FasL in KHYG-1 cells. Raddeanin A treatment increased CREB phosphorylation, p65 phosphorylation, NFAT1 and acetyl-histone H3 expression. Raddeanin A elevated caspase 3 and PARP cleavage, increased t-Bid expression, promoting apoptosis in K562 cells. Furthermore, it reduced the expression of HMGB2, SET and Ape1, impairing the DNA repair process and causing K562 cells to die caspase-independently. Additionally, Raddeanin A increased ERK, p38 and JNK phosphorylation at the molecular level, which increased granzyme B production in KHYG-1 cells. Raddeanin A treatment increased Ras, Raf phosphorylation, MEK phosphorylation, NKG2D, NKp44 and NKp30 expression in KHYG-1 cells. Collectively, our data indicate that Raddeanin A enhances the cytotoxic activity of NK cells against different cancer cells.


Subject(s)
Apoptosis , Killer Cells, Natural , Leukemia, Myelogenous, Chronic, BCR-ABL Positive , Humans , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , K562 Cells , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , ras Proteins/metabolism , Cytotoxicity, Immunologic , Signal Transduction , raf Kinases/metabolism , MAP Kinase Signaling System/drug effects , DNA Repair , Granzymes/metabolism
4.
Commun Biol ; 7(1): 1022, 2024 Aug 20.
Article in English | MEDLINE | ID: mdl-39164588

ABSTRACT

IL-33 belongs to the inflammatory factor family and is closely associated with the inflammatory response. However, its role in the development of intrauterine adhesions (IUAs) remains unclear. In this study, the role of IL-33 in the formation of IUAs after endometrial injury was identified via RNA sequencing after mouse endometrial organoids were transplanted into an IUA mouse model. Major pathological changes in the mouse uterus, consistent with the expression of fibrotic markers, such as TGF-ß, were observed in response to treatment with IL-33. This finding may be attributed to activation of the phosphorylation of downstream MAPK signaling pathway components, which are activated by the release of IL-33 in macrophages. Our study provides a novel mechanism for elucidating IUA formation, suggesting a new therapeutic strategy for the prevention and clinical treatment of IUAs.


Subject(s)
Interleukin-33 , MAP Kinase Signaling System , Animals , Interleukin-33/metabolism , Interleukin-33/genetics , Female , Mice , Tissue Adhesions/metabolism , Tissue Adhesions/pathology , Uterine Diseases/pathology , Uterine Diseases/metabolism , Uterine Diseases/genetics , Mice, Inbred C57BL , Disease Models, Animal , Signal Transduction , Uterus/metabolism , Uterus/pathology , Endometrium/metabolism , Endometrium/pathology , Mitogen-Activated Protein Kinases/metabolism , Mitogen-Activated Protein Kinases/genetics
5.
J Cell Mol Med ; 28(16): e70015, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39159189

ABSTRACT

Pyrocurzerenone is a natural compound found in Curcuma zedoaria and Chloranthus serratus. However, the anticancer effect of pyrocurzerenone in oral cancer remains unclear. Using the MTT assay, wound healing assay, transwell assay and western blot analysis, we investigated the impact of pyrocurzerenone on antimetastatic activity, as well as the critical signalling pathways that underlie the processes of oral cancer cell lines SCC-9, SCC-1 and SAS in this work. Our findings suggested that pyrocurzerenone inhibits cell migration and invasion ability in oral cancer cell lines. Furthermore, phosphorylation of ERK1/2 had significant inhibitory effects in SCC-9 and SCC-1 cell lines. Combining ERK1/2 inhibitors with pyrocurzerenone decreased the migration and invasion activity of SCC-9 and SCC-1 cell lines. We also found that the expressed level of cathepsin S decreased under pyrocurzerenone treatment. This study showed that pyrocurzerenone reduced ERK1/2 expression of the proteins and cathepsin S, suggesting that it could be a valuable treatment to inhibit human oral cancer cell metastasis.


Subject(s)
Cathepsins , Cell Movement , Mouth Neoplasms , Humans , Mouth Neoplasms/pathology , Mouth Neoplasms/drug therapy , Mouth Neoplasms/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Cathepsins/metabolism , Cathepsins/antagonists & inhibitors , Phosphorylation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Neoplasm Metastasis , MAP Kinase Signaling System/drug effects , Mitogen-Activated Protein Kinase 3/metabolism , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 1/genetics , Neoplasm Invasiveness , Cell Proliferation/drug effects , Signal Transduction/drug effects
6.
PLoS One ; 19(8): e0308977, 2024.
Article in English | MEDLINE | ID: mdl-39159207

ABSTRACT

Perioperative acute kidney injury (AKI), which is mainly mediated by renal ischemia‒reperfusion (I/R) injury, is commonly observed in clinical practice. However, effective measures for preventing and treating this perioperative complication are still lacking in the clinic. Thus, we designed this study to examine whether remote liver ischemic preconditioning (RLIPC) has a protective effect on damage caused by renal I/R injury. In a rodent model, 30 mice were divided into five groups to assess the effects of RLIPC and ERK1/2 inhibition on AKI. The groups included the sham-operated (sham), kidney ischemia and reperfusion (CON), remote liver ischemic preconditioning (RLIPC), CON with the ERK1/2 inhibitor U0126 (CON+U0126), and RLIPC with U0126 (RLIPC+U0126). RLIPC consisted of 4 liver ischemia cycles before renal ischemia. Renal function and injury were assessed through biochemical assays, histology, cell apoptosis and protein phosphorylation analysis. RLIPC significantly mitigated renal dysfunction, tissue damage, inflammation, and apoptosis caused by I/R, which was associated with ERK1/2 phosphorylation. Furthermore, ERK1/2 inhibition with U0126 negated the protective effects of RLIPC and exacerbated renal injury. To summarize, we demonstrated that RLIPC has a strong renoprotective effect on kidneys post I/R injury and that this effect may be mediated by phosphorylation of ERK1/2.


Subject(s)
Acute Kidney Injury , Ischemic Preconditioning , Liver , Mitogen-Activated Protein Kinase 3 , Nitriles , Reperfusion Injury , Animals , Reperfusion Injury/prevention & control , Reperfusion Injury/metabolism , Ischemic Preconditioning/methods , Liver/metabolism , Liver/pathology , Liver/blood supply , Phosphorylation , Mice , Male , Mitogen-Activated Protein Kinase 3/metabolism , Acute Kidney Injury/prevention & control , Acute Kidney Injury/metabolism , Acute Kidney Injury/pathology , Nitriles/pharmacology , Kidney/pathology , Kidney/blood supply , Kidney/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Butadienes/pharmacology , Apoptosis/drug effects , Mice, Inbred C57BL , MAP Kinase Signaling System/drug effects
7.
BMC Cancer ; 24(1): 991, 2024 Aug 12.
Article in English | MEDLINE | ID: mdl-39128984

ABSTRACT

BACKGROUND: Our previous study demonstrated that ß2-microglobulin (ß2M) promoted ER+/HER2- breast cancer survival via the SGK1/Bcl-2 signaling pathway. However, the role of ß2M has not been investigated in ER-/HER2+ breast cancer. Here, we aimed to determine the role of ß2M in ER-/HER2+ breast cancer. METHODS: The interaction between ß2M and HFE was confirmed by co-immunoprecipitation, mass spectrometry, yeast two-hybrid screening, and His pull-down. The knockdown and overexpression of ß2M or HFE were performed in MDA-MB-453 cells, and ERK signaling pathway was subsequently analyzed via western blotting. Apoptotic cells were detected using flow cytometer. ß2M, HFE, and p-ERK1/2 were examined in tumor and paired adjacent tissues via immunohistochemistry. RESULTS: HFE was found to be an interacting protein of ß2M in ER-/HER2+ breast cancer cells MDA-MB-453 by co-immunoprecipitation and mass spectrometry. A yeast two-hybrid system and His-pull down experiments verified that ß2M directly interacted with HFE. ß2M and HFE as a complex were mainly located in the cytoplasm, with some on the cytomembrane of MDA-MB-453 cells. In addition to breast cancer cells BT474, endogenous ß2M directly interacted with HFE in breast cancer cells MDA-MB-453, MDA-MB-231, and MCF-7. ß2M activated the ERK signaling pathway by interacting with HFE and induced apoptosis of MDA-MB-453 cells. The expression of HFE and p-ERK1/2 showed significantly high levels in HER2-overexpressing breast cancer tumor tissue compared with adjacent normal tissue, consistent with the results obtained from the cell experiments. CONCLUSIONS: ß2M induced apoptosis of tumor cells via activation of the ERK signal pathway by directly interacting with HFE in HER2-overexpressing breast cancer.


Subject(s)
Apoptosis , Breast Neoplasms , Hemochromatosis Protein , MAP Kinase Signaling System , Receptor, ErbB-2 , beta 2-Microglobulin , Humans , beta 2-Microglobulin/metabolism , beta 2-Microglobulin/genetics , Breast Neoplasms/pathology , Breast Neoplasms/metabolism , Breast Neoplasms/genetics , Female , Receptor, ErbB-2/metabolism , Receptor, ErbB-2/genetics , Cell Line, Tumor , Hemochromatosis Protein/genetics , Hemochromatosis Protein/metabolism , Protein Binding , Gene Expression Regulation, Neoplastic
8.
Mol Med ; 30(1): 119, 2024 Aug 11.
Article in English | MEDLINE | ID: mdl-39129004

ABSTRACT

BACKGROUND: AGTPBP1 is a cytosolic carboxypeptidase that cleaves poly-glutamic acids from the C terminus or side chains of α/ß tubulins. Although its dysregulated expression has been linked to the development of non-small cell lung cancer, the specific roles and mechanisms of AGTPBP1 in pancreatic cancer (PC) have yet to be fully understood. In this study, we examined the role of AGTPBP1 on PC in vitro and in vivo. METHODS: Immunohistochemistry was used to examine the expression of AGTPBP1 in PC and non-cancerous tissues. Additionally, we assessed the malignant behaviors of PC cells following siRNA-mediated AGTPBP1 knockdown both in vitro and in vivo. RNA sequencing and bioinformatics analysis were performed to identify the differentially expressed genes regulated by AGTPBP1. RESULTS: We determined that AGTPBP1 was overexpressed in PC tissues and the higher expression of AGTPBP1 was closely related to the location of tumors. AGTPBP1 inhibition can significantly decrease cell progression in vivo and in vitro. Moreover, the knockdown of AGTPBP1 inhibited the expression of ERK1/2, P-ERK1/2, MYLK, and TUBB4B proteins via the ERK signaling pathway. CONCLUSION: Our research indicates that AGTPBP1 may be a putative therapeutic target for PC.


Subject(s)
Carboxypeptidases , Gene Expression Regulation, Neoplastic , MAP Kinase Signaling System , Microtubules , Pancreatic Neoplasms , Humans , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Carboxypeptidases/metabolism , Carboxypeptidases/genetics , Cell Line, Tumor , Microtubules/metabolism , Animals , Mice , Male , Female , Cell Proliferation , Disease Progression , Middle Aged , Cell Movement/genetics
9.
Sci Rep ; 14(1): 18418, 2024 08 08.
Article in English | MEDLINE | ID: mdl-39117695

ABSTRACT

Bacterial testicular inflammation is one of the important causes of male infertility. Using plant-derived compounds to overcome the side effects of antibiotics is an alternative treatment strategy for many diseases. Schizandrin B (SchB) is a bioactive compound of herbal medicine Schisandra chinensis which has multiple pharmacological effects. However its effect and the mechanism against testicular inflammation are unknown. Here we tackled these questions using models of lipopolysaccharide (LPS)-induced mice and -Sertoli cells (SCs). Histologically, SchB ameliorated the LPS-induced damages of the seminiferous epithelium and blood-testicular barrier, and reduced the production of pro-inflammatory mediators in mouse testes. Furthermore, SchB decreased the levels of pro-inflammatory mediators and inhibited the nuclear factor kB (NF-κB) and MAPK (especially JNK) signaling pathway phosphorylation in LPS-induced mSCs. The bioinformatics analysis based on receptor prediction and the molecular docking was further conducted. We targeted androgen receptor (AR) and illustrated that AR might bind with SchB in its function. Further experiments indicate that the AR expression was upregulated by LPS stimulation, while SchB treatment reversed this phenomenon; similarly, the expression of the JNK-related proteins and apoptotic-related protein were also reversed after AR activator treatment. Together, SchB mitigates LPS-induced inflammation and apoptosis by inhibiting the AR-JNK pathway.


Subject(s)
Apoptosis , Cyclooctanes , Lignans , Lipopolysaccharides , Polycyclic Compounds , Sertoli Cells , Animals , Male , Cyclooctanes/pharmacology , Polycyclic Compounds/pharmacology , Polycyclic Compounds/therapeutic use , Lignans/pharmacology , Lignans/therapeutic use , Apoptosis/drug effects , Mice , Sertoli Cells/drug effects , Sertoli Cells/metabolism , Receptors, Androgen/metabolism , MAP Kinase Signaling System/drug effects , Inflammation/drug therapy , Inflammation/metabolism , Inflammation/pathology , Molecular Docking Simulation , Testis/drug effects , Testis/metabolism , Testis/pathology , NF-kappa B/metabolism
10.
Sci Rep ; 14(1): 18757, 2024 08 13.
Article in English | MEDLINE | ID: mdl-39138279

ABSTRACT

Hepatocellular carcinoma (HCC) still presents poor prognosis with low overall survival rates and limited therapeutic options available. Recently, attention has been drawn to peptidomic analysis, an emerging field of proteomics for the exploration of new potential peptide drugs for the treatment of various diseases. However, research on the potential function of HCC peptides is lacking. Here, we analyzed the peptide spectrum in HCC tissues using peptidomic techniques and explored the potentially beneficial peptides involved in HCC. Changes in peptide profiles in HCC were examined using liquid chromatography-mass spectrometry (LC-MS/MS). Analyze the physicochemical properties and function of differently expressed peptides using bioinformatics. The effect of candidate functional peptides on HCC cell growth and migration was evaluated using the CCK-8, colony formation, and transwell assays. Transcriptome sequencing analysis and western blot were employed to delve into the mode of action of potential peptide on HCC. Peptidomic analysis of HCC tissue yielded a total of 8683 peptides, of which 452 exhibited up-regulation and 362 showed down-regulation. The peptides that were differentially expressed, according to bioinformatic analysis, were closely linked to carbon metabolism and the mitochondrial inner membrane. The peptide functional validation identified a novel peptide, PDLC (peptide derived from liver cancer), which was found to dramatically boost HCC cell proliferation through the Ras/Raf/MEK/ERK signaling cascade. Our research defined the peptide's properties and pattern of expression in HCC and identified a novel peptide, PDLC, with a function in encouraging HCC progression, offering an entirely new potential therapeutic target the disease.


Subject(s)
Carcinoma, Hepatocellular , Cell Proliferation , Liver Neoplasms , MAP Kinase Signaling System , Proteomics , raf Kinases , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Liver Neoplasms/genetics , Humans , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/genetics , Cell Proliferation/drug effects , MAP Kinase Signaling System/drug effects , raf Kinases/metabolism , Proteomics/methods , Cell Line, Tumor , ras Proteins/metabolism , ras Proteins/genetics , Peptides/metabolism , Peptides/pharmacology , Cell Movement/drug effects , Tandem Mass Spectrometry , Gene Expression Regulation, Neoplastic
11.
Mol Genet Genomic Med ; 12(8): e2503, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39140707

ABSTRACT

BACKGROUND: The causes of migraine remain unclear. Evidence suggests that the MAPK and PI3K/Akt signaling pathways play a role in migraine pathogenesis. However, studies on genetic polymorphisms in the two pathways associated with migraine are still limited. METHODS: This study included 226 migraineurs and 452 age- and sex-matched nonmigraine control individuals. Genotyping of 31 Single Nucleotide Polymorphisms (SNPs) in 21 genes was performed. The relationship between migraine and gene polymorphisms was analyzed by using logistic regression. SNP-SNP interactions were examined by a generalized multifactor dimension reduction (GMDR) approach. The possible role of SNPs was evaluated with gene expression data from the GTEx database. RESULTS: The RASGRP2-rs2230414 GT genotype was associated with decreased migraine risk compared with the wild-type GG genotype [ORadj (95% CI): 0.674(0.458-0.989)]. PIK3R1-rs3730089 was associated with migraine in the recessive model [ORadj (95% CI): 1.446(1.004-2.083)]. The CACNA1H-rs61734410 CT genotype was associated with migraine risk [ORadj (95% CI): 1.561(1.068-2.281)]. One significant two-way SNP-SNP interaction was found (PRKCA rs2228945-BDNF rs6265) (p = 0.0107). Significant eQTL and sQTL signals were observed for the SNP rs2230414. CONCLUSIONS: This is the first study to systematically reveal significant associations between MAPK and PI3K/Akt signaling pathway-related gene polymorphisms and migraine risk.


Subject(s)
Migraine Disorders , Polymorphism, Single Nucleotide , Proto-Oncogene Proteins c-akt , Humans , Migraine Disorders/genetics , Female , Male , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Adult , Signal Transduction/genetics , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , MAP Kinase Signaling System/genetics , Class Ia Phosphatidylinositol 3-Kinase/genetics , Middle Aged
12.
Sci Adv ; 10(33): eadn5993, 2024 Aug 16.
Article in English | MEDLINE | ID: mdl-39141732

ABSTRACT

Skeletal muscle has gained recognition as an endocrine organ releasing myokines upon contraction during physical exercise. These myokines exert both local and pleiotropic health benefits, underscoring the crucial role of muscle function in countering obesity and contributing to the overall positive effects of exercise on health. Here, we found that exercise activates muscle p38γ, increasing locomotor activity through the secretion of interleukin-15 (IL-15). IL-15 signals in the motor cortex, stimulating locomotor activity. This activation of muscle p38γ, leading to an increase locomotor activity, plays a crucial role in reducing the risk of diabetes and liver steatosis, unveiling a vital muscle-brain communication pathway with profound clinical implications. The correlation between p38γ activation in human muscle during acute exercise and increased blood IL-15 levels highlights the potential therapeutic relevance of this pathway in treating obesity and metabolic diseases. These findings provide valuable insights into the molecular basis of exercise-induced myokine responses promoting physical activity.


Subject(s)
Exercise , Interleukin-15 , Muscle, Skeletal , Interleukin-15/metabolism , Muscle, Skeletal/metabolism , Humans , Animals , Exercise/physiology , Locomotion , Mice , p38 Mitogen-Activated Protein Kinases/metabolism , Signal Transduction , Male , MAP Kinase Signaling System , Obesity/metabolism
13.
CNS Neurosci Ther ; 30(8): e14924, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39143678

ABSTRACT

AIMS: Postoperative cognitive dysfunction (POCD) is prevalent among the elderly, characterized primarily by cognitive decline after surgery. This study aims to explore how extracellular vesicles (EVs) derived from BV2 microglial cells, with and without the C-C chemokine receptor type 5 (CCR5), affect neuroinflammation, neuronal integrity, and cognitive function in a POCD mouse model. METHODS: We collected EVs from LPS-stimulated BV2 cells expressing CCR5 (EVsM1) and from BV2 cells with CCR5 knockdown (EVsM1-CCR5). These were administered to POCD-induced mice. Protein interactions between CCR5, G-protein-coupled receptors (GPCRs), and Ras were analyzed using structure-based docking and co-immunoprecipitation (Co-IP). We assessed the phosphorylation of p38 and Erk, the expression of synaptic proteins PSD95 and MAP2, and conducted Morris Water Maze tests to evaluate cognitive function. RESULTS: Structure-based docking and Co-IP confirmed interactions between CCR5, GPR, and Ras, suggesting a CCR5-GPCRs-Ras-MAPK pathway involvement in neuroinflammation. EVsM1 heightened neuroinflammation, reduced synaptic integrity, and impaired cognitive function in POCD mice. In contrast, EVsM1-CCR5 reduced neuroinflammatory markers, preserved synaptic proteins, enhanced dendritic spine structure, and improved cognitive outcomes. CONCLUSION: EVsM1 induced neuroinflammation via the CCR5-GPCRs-Ras-MAPK pathway, with EVsM1-CCR5 showing protective effects on POCD progression, suggesting a new therapeutic strategy for POCD management via targeted modification of microglial EVs.


Subject(s)
Mice, Inbred C57BL , Microglia , Neuroinflammatory Diseases , Postoperative Cognitive Complications , Receptors, CCR5 , Animals , Microglia/metabolism , Mice , Receptors, CCR5/metabolism , Neuroinflammatory Diseases/metabolism , Postoperative Cognitive Complications/metabolism , Male , Extracellular Vesicles/metabolism , Receptors, G-Protein-Coupled/metabolism , ras Proteins/metabolism , Cognition/physiology , Cognition/drug effects , MAP Kinase Signaling System/physiology , MAP Kinase Signaling System/drug effects , Cognitive Dysfunction/metabolism
14.
Nat Commun ; 15(1): 7037, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-39147750

ABSTRACT

The quest for targeted therapies is critical in the battle against cancer. The RAS/MAP kinase pathway is frequently implicated in neoplasia, with ERK playing a crucial role as the most distal kinase in the RAS signaling cascade. Our previous research demonstrated that the interaction between ERK and MYD88, an adaptor protein in innate immunity, is crucial for RAS-dependent transformation and cancer cell survival. In this study, we examine the biological consequences of disrupting the ERK-MYD88 interaction through the ERK D-recruitment site (DRS), while preserving ERK's kinase activity. Our results indicate that EI-52, a small-molecule benzimidazole targeting ERK-MYD88 interaction induces an HRI-mediated integrated stress response (ISR), resulting in immunogenic apoptosis specific to cancer cells. Additionally, EI-52 exhibits anti-tumor efficacy in patient-derived tumors and induces an anti-tumor T cell response in mice in vivo. These findings suggest that inhibiting the ERK-MYD88 interaction may be a promising therapeutic approach in cancer treatment.


Subject(s)
Benzimidazoles , Extracellular Signal-Regulated MAP Kinases , Myeloid Differentiation Factor 88 , Myeloid Differentiation Factor 88/metabolism , Myeloid Differentiation Factor 88/genetics , Humans , Animals , Mice , Extracellular Signal-Regulated MAP Kinases/metabolism , Cell Line, Tumor , Benzimidazoles/pharmacology , Apoptosis/drug effects , Immunogenic Cell Death/drug effects , Neoplasms/immunology , Neoplasms/drug therapy , Neoplasms/pathology , Neoplasms/metabolism , Female , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/immunology , Xenograft Model Antitumor Assays , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use
15.
J Hazard Mater ; 477: 135093, 2024 Sep 15.
Article in English | MEDLINE | ID: mdl-39088948

ABSTRACT

Exposure to particulate matter (PM) can cause airway inflammation and worsen various airway diseases. However, the underlying molecular mechanism by which PM triggers airway inflammation has not been completely elucidated, and effective interventions are lacking. Our study revealed that PM exposure increased the expression of histone deacetylase 9 (HDAC9) in human bronchial epithelial cells and mouse airway epithelium through the METTL3/m6A methylation/IGF2BP3 pathway. Functional assays showed that HDAC9 upregulation promoted PM-induced airway inflammation and activation of MAPK signaling pathway in vitro and in vivo. Mechanistically, HDAC9 modulated the deacetylation of histone 4 acetylation at K12 (H4K12) in the promoter region of dual specificity phosphatase 9 (DUSP9) to repress the expression of DUSP9 and resulting in the activation of MAPK signaling pathway, thereby promoting PM-induced airway inflammation. Additionally, HDAC9 bound to MEF2A to weaken its anti-inflammatory effect on PM-induced airway inflammation. Then, we developed a novel inhaled lipid nanoparticle system for delivering HDAC9 siRNA to the airway, offering an effective treatment for PM-induced airway inflammation. Collectively, we elucidated the crucial regulatory mechanism of HDAC9 in PM-induced airway inflammation and introduced an inhaled therapeutic approach targeting HDAC9. These findings contribute to alleviating the burden of various airway diseases caused by PM exposure.


Subject(s)
Epigenesis, Genetic , Histone Deacetylases , Particulate Matter , Up-Regulation , Animals , Particulate Matter/toxicity , Humans , Histone Deacetylases/metabolism , Histone Deacetylases/genetics , Epigenesis, Genetic/drug effects , Up-Regulation/drug effects , Mice , Dual-Specificity Phosphatases/genetics , Dual-Specificity Phosphatases/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Inflammation , Nanoparticles/chemistry , Nanoparticles/toxicity , Mice, Inbred C57BL , Cell Line , MAP Kinase Signaling System/drug effects , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Male
16.
J Diabetes Complications ; 38(9): 108805, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39089052

ABSTRACT

PURPOSE: Type 1 diabetes (T1DM) is a chronic metabolic disorder that can cause damage to multiple organs including the spleen. Sole insulin therapy is not satisfactory. This study aims to investigate the effects and mechanisms of combined treatment with insulin and N-acetylcysteine (NAC) on spleen damage in T1DM canines, in order to identify drugs that may better assist patients in the management of diabetes and its complications. METHODS: The canine model of T1DM was established by intravenous injection of alloxan (ALX) and streptozotocin (STZ). The therapeutic effects of insulin and NAC were evaluated by clinical manifestations, spleen protein and mRNA expression. RESULTS: The results indicate that the combined treatment of insulin and NAC can alleviate hyperglycemia and hematologic abnormalities, improve splenic histopathological changes, prevent fibrous tissue proliferation, and glycogen deposition. In addition, we observed that this combination treatment significantly suppressed the protein expression of p-P65/P65 (17.6 %, P < 0.05), NLRP3 (46.8 %, P < 0.05), and p-P38/P38 (37.1 %, P < 0.05) induced by T1DM when compared to insulin treatment alone. Moreover, it also significantly decreased the mRNA expression of TLR4 (45.0 %, P < 0.01), TNF-α (30.3 %, P < 0.05), and NLRP3 (43.3 %, P < 0.05). CONCLUSIONS: This combination has the potential to mitigate splenic inflammatory injury in T1DM canines by suppressing the activation of MAPKs-NF-κB pathway and pyroptosis. These findings provide a reference for the treatment strategies of diabetes and its complications.


Subject(s)
Acetylcysteine , Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 1 , Drug Therapy, Combination , Insulin , NF-kappa B , Pyroptosis , Signal Transduction , Spleen , Animals , Dogs , Acetylcysteine/pharmacology , Acetylcysteine/therapeutic use , Diabetes Mellitus, Type 1/complications , Diabetes Mellitus, Type 1/drug therapy , NF-kappa B/metabolism , Pyroptosis/drug effects , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/drug therapy , Signal Transduction/drug effects , Male , Spleen/drug effects , Spleen/pathology , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/therapeutic use , Splenic Diseases/drug therapy , Splenic Diseases/etiology , Splenic Diseases/complications , MAP Kinase Signaling System/drug effects
17.
Cell Death Dis ; 15(8): 555, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39090096

ABSTRACT

Resistance to epidermal growth factor receptor (EGFR)-tyrosine kinase inhibitors (TKIs) is a significant cause of treatment failure and cancer recurrence in non-small cell lung cancer (NSCLC). Approximately 30% of patients with EGFR-activating mutations exhibit primary resistance to EGFR-TKIs. However, the potential mechanisms of primary resistance to EGFR-TKIs remain poorly understood. Recent studies have shown that increased expression of programmed death ligand-1 (PD-L1) is associated with EGFR-TKIs resistance. Therefore, the present study aimed to investigate the mechanism of PD-L1 in primary resistance to EGFR-TKIs in EGFR-mutant lung adenocarcinoma (LUAD) cells. We found that PD-L1 was associated with poor prognosis in patients with EGFR-mutant LUAD, while the combination of EGFR-TKIs with chemotherapy could improve its therapeutic efficacy. In vitro and in vivo experiments revealed that PD-L1 promoted the proliferation and autophagy and inhibited the apoptosis of LUAD cells. Mechanistic studies demonstrated that upregulation of PD-L1 was critical in inducing autophagy through the mitogen-activated protein kinase (MAPK) signaling pathway, which was beneficial for tumor progression and the development of gefitinib resistance. Furthermore, we found that gefitinib combined with pemetrexed could synergistically enhance antitumor efficacy in PD-L1-overexpression LUAD cells. Overall, our study demonstrated that PD-L1 contributed to primary resistance to EGFR-TKIs in EGFR-mutant LUAD cells, which may be mediated by inducing autophagy via the MAPK signaling pathway. These findings not only help improve the prognosis of patients with EGFR-mutant LUAD but also provide a reference for the research of other cancer types.


Subject(s)
Adenocarcinoma of Lung , Autophagy , B7-H1 Antigen , Drug Resistance, Neoplasm , ErbB Receptors , Lung Neoplasms , MAP Kinase Signaling System , Mutation , Protein Kinase Inhibitors , Humans , Autophagy/drug effects , Autophagy/genetics , ErbB Receptors/metabolism , ErbB Receptors/genetics , B7-H1 Antigen/metabolism , B7-H1 Antigen/genetics , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/drug therapy , Adenocarcinoma of Lung/pathology , Adenocarcinoma of Lung/metabolism , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Drug Resistance, Neoplasm/genetics , Drug Resistance, Neoplasm/drug effects , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Lung Neoplasms/metabolism , Animals , MAP Kinase Signaling System/drug effects , Cell Line, Tumor , Mutation/genetics , Mice , Mice, Nude , Female , Male , Gefitinib/pharmacology , Gefitinib/therapeutic use , Cell Proliferation/drug effects , Apoptosis/drug effects , Mice, Inbred BALB C
18.
PLoS Pathog ; 20(8): e1012437, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39102432

ABSTRACT

The ability of Staphylococcus aureus (S. aureus) to survive within macrophages is a critical strategy for immune evasion, contributing to the pathogenesis and progression of osteomyelitis. However, the underlying mechanisms remain poorly characterized. This study discovered that inhibiting the MEK1/2 pathway reduced bacterial load and mitigated bone destruction in a mouse model of S. aureus osteomyelitis. Histological staining revealed increased phosphorylated MEK1/2 levels in bone marrow macrophages surrounding abscess in the mouse model of S. aureus osteomyelitis. Activation of MEK1/2 pathway and its roles in impairing macrophage bactericidal function were confirmed in primary mouse bone marrow-derived macrophages (BMDMs). Transcriptome analysis and in vitro experiments demonstrated that S. aureus activates the MEK1/2 pathway through EGFR signaling. Moreover, we found that excessive activation of EGFR-MEK1/2 cascade downregulates mitochondrial reactive oxygen species (mtROS) levels by suppressing Chek2 expression, thereby impairing macrophage bactericidal function. Furthermore, pharmacological inhibition of EGFR signaling prevented upregulation of phosphorylated MEK1/2 and restored Chek2 expression in macrophages, significantly enhancing S. aureus clearance and improving bone microstructure in vivo. These findings highlight the critical role of the EGFR-MEK1/2 cascade in host immune defense against S. aureus, suggesting that S. aureus may reduce mtROS levels by overactivating the EGFR-MEK1/2 cascade, thereby suppressing macrophage bactericidal function. Therefore, combining EGFR-MEK1/2 pathway blockade with antibiotics could represent an effective therapeutic approach for the treatment of S. aureus osteomyelitis.


Subject(s)
ErbB Receptors , MAP Kinase Kinase 1 , Macrophages , Osteomyelitis , Staphylococcal Infections , Staphylococcus aureus , Animals , Osteomyelitis/microbiology , Osteomyelitis/immunology , Osteomyelitis/metabolism , Staphylococcal Infections/immunology , Staphylococcal Infections/metabolism , Staphylococcal Infections/microbiology , Mice , Staphylococcus aureus/immunology , ErbB Receptors/metabolism , Macrophages/immunology , Macrophages/metabolism , Macrophages/microbiology , MAP Kinase Kinase 1/metabolism , MAP Kinase Kinase 2/metabolism , MAP Kinase Signaling System/physiology , Mice, Inbred C57BL , Disease Models, Animal , Signal Transduction
19.
Int J Mol Sci ; 25(15)2024 Jul 27.
Article in English | MEDLINE | ID: mdl-39125778

ABSTRACT

Neonatal hypoxic-ischemic encephalopathy is the most common cause of long-term disability in term neonates, and white matter injury is the primary cause of cerebral palsy. Therapies that focus on the neuroprotection of myelination and oligodendrocyte proliferation could potentially ameliorate long-lasting neurological impairments after hypoxic-ischemic encephalopathy. Clemastine, a histamine H1 antagonist, has been shown to exert neuroprotective effects in multiple sclerosis and spinal cord injury by promoting oligodendrogenesis and re-myelination. In this study, we demonstrated the neuroprotective effects of clemastine in our rat model of neonatal hypoxic-ischemic brain injury. Animals received a single intraperitoneal injection of either vehicle or clemastine (10 mg/kg) for 6 consecutive days. Our results showed a significant reduction in white matter loss after treatment, with a clear effect of clemastine on oligodendrocytes, showing a significant increase in the number of Olig2+ cells. We characterized the MAPK/ERK pathway as a potential mechanistic pathway underlying the neuroprotective effects of clemastine. Altogether, our results demonstrate that clemastine is a potential compound for the treatment of hypoxic-ischemic encephalopathy, with a clear neuroprotective effect on white matter injury by promoting oligodendrogenesis.


Subject(s)
Animals, Newborn , Cell Proliferation , Clemastine , Disease Models, Animal , Hypoxia-Ischemia, Brain , MAP Kinase Signaling System , Neuroprotective Agents , Oligodendroglia , Animals , Oligodendroglia/drug effects , Oligodendroglia/metabolism , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Hypoxia-Ischemia, Brain/drug therapy , Hypoxia-Ischemia, Brain/metabolism , Hypoxia-Ischemia, Brain/pathology , Rats , Clemastine/pharmacology , MAP Kinase Signaling System/drug effects , Cell Proliferation/drug effects , Rats, Sprague-Dawley
20.
Int J Mol Sci ; 25(15)2024 Jul 28.
Article in English | MEDLINE | ID: mdl-39125805

ABSTRACT

A previous study showed that high-glucose (HG) conditions induce mitochondria fragmentation through the calcium-mediated activation of extracellular signal-regulated kinase 1/2 (ERK 1/2) in H9C2 cells. This study tested whether empagliflozin could prevent HG-induced mitochondria fragmentation through this pathway. We found that exposing H9C2 cells to an HG concentration decreased cell viability and increased cell apoptosis and caspase-3. Empagliflozin could reverse the apoptosis effect of HG stimulation on H9C2 cells. In addition, the HG condition caused mitochondria fragmentation, which was reduced by empagliflozin. The expression of mitochondria fission protein was upregulated, and fusion proteins were downregulated under HG stimulation. The expression of fission proteins was decreased under empagliflozin treatment. Increased calcium accumulation was observed under the HG condition, which was decreased by empagliflozin. The increased expression of ERK 1/2 under HG stimulation was also reversed by empagliflozin. Our study shows that empagliflozin could reverse the HG condition, causing a calcium-dependent activation of the ERK 1/2 pathway, which caused mitochondria fragmentation in H9C2 cells.


Subject(s)
Apoptosis , Benzhydryl Compounds , Calcium , Glucose , Glucosides , MAP Kinase Signaling System , Mitochondria , Apoptosis/drug effects , Benzhydryl Compounds/pharmacology , Glucosides/pharmacology , Glucose/metabolism , Mitochondria/metabolism , Mitochondria/drug effects , Calcium/metabolism , Animals , Rats , Cell Line , MAP Kinase Signaling System/drug effects , Cell Survival/drug effects , Mitochondrial Dynamics/drug effects , Caspase 3/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Mitogen-Activated Protein Kinase 1/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL