Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 38.509
1.
Cell Death Dis ; 15(4): 295, 2024 Apr 25.
Article En | MEDLINE | ID: mdl-38664392

Abnormal Transmembrane protein 9 (TMEM9) expression has been identified in various human tumors. However, the prognostic potential and mechanistic role of TMEM9 in lung adenocarcinoma (LUAD) remain unclear. Here, we first found a significant upregulation of TMEM9 in LUAD tissues, and TMEM9 expression was positively correlated with microvessel density (MVD), T stage, and clinical stage. Survival analysis demonstrated TMEM9 was an independent indicator of poor prognosis in LUAD patients. In addition, downregulation of TMEM9 suppressed tumor growth and metastasis in vitro and in vivo models, and reduced HUVEC proliferation, migration, and tube formation in a cancer cell/HUVEC coculture model. Furthermore, TMEM9 upregulated VEGF expression, and VEGF-neutralizing antibodies reversed HUVEC angiogenesis and cancer cell migration ability caused by overexpression of TMEM9. In contrast, recombinant VEGF (rVEGF) abolished the inhibitory effect of TMEM9-knockdown LUAD cells on HUVEC angiogenesis and tumor cell migration. Moreover, we showed that TMEM9 upregulated VEGF expression by activating the mitogen-activated protein kinase/extracellular signal-regulated kinase/STAT3 (MEK/ERK/STAT3) pathway. Together, our study provides mechanistic insights into the role of TMEM9 in LUAD and highlights the potential of targeting the TMEM9/MEK/ERK/STAT3/VEGF pathway as a novel therapy for preventing LUAD progression.


Adenocarcinoma of Lung , Disease Progression , Lung Neoplasms , MAP Kinase Signaling System , Membrane Proteins , STAT3 Transcription Factor , Vascular Endothelial Growth Factor A , Humans , STAT3 Transcription Factor/metabolism , Vascular Endothelial Growth Factor A/metabolism , Membrane Proteins/metabolism , Membrane Proteins/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Lung Neoplasms/genetics , Animals , Male , Female , Adenocarcinoma of Lung/pathology , Adenocarcinoma of Lung/metabolism , Adenocarcinoma of Lung/genetics , Cell Movement , Human Umbilical Vein Endothelial Cells/metabolism , Cell Proliferation , Mice, Nude , Mice , Cell Line, Tumor , Middle Aged , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/genetics , Gene Expression Regulation, Neoplastic , Mice, Inbred BALB C , A549 Cells
2.
Discov Med ; 36(183): 778-787, 2024 Apr.
Article En | MEDLINE | ID: mdl-38665026

BACKGROUND: Tropomyosin 2 (TPM2) has been linked to the advancement of various tumor types, exhibiting distinct impacts on tumor progression. In our investigation, the primary objective was to identify the potential involvement of TPM2 in the development of colitis-associated cancer (CAC) using a mice model. METHODS: This study used lentiviral vector complex for TPM2 knockdown (sh-TPM2) and the corresponding negative control lentiviral vector complex (sh-NC) for genetic interference in mice. CAC was induced in mice using azoxymethane (AOM) and dextran sulfate sodium salt (DSS). This study included 6 groups of mice models: Control, Control+sh-NC, Control+sh-TPM2, CAC, CAC+sh-NC, and CAC+sh-TPM2. Subsequently, colon tissues were collected and assessed using quantitative reverse transcription-polymerase chain reaction (qRT-PCR) for TPM2 mRNA levels and flow cytometry for infiltrating immune cells. Tumor number, size, and weight within colon tissues from CAC mice were measured and recorded. The hematoxylin-eosin staining was used for observing tissue pathology changes. The intestinal epithelial cells (IECs) were isolated and analyzed for cell proliferation. This analysis included examining the levels of 5-bromo-2-deoxyuridine (BrdU) and Ki-67 using immunohistochemistry. Additionally, the mRNA levels of proliferating cell nuclear antigen (PCNA) and Ki-67 were detected by qRT-PCR. This study also investigated the activation of the c-Jun N-terminal kinase (JNK) pathway using western blot analysis. Immunogenicity analyses were conducted using immunohistochemistry for F4/80 and flow cytometry. RESULTS: In 8-week-old mice, AOM injections and three cycles of DSS treatment induced TPM2 upregulation in tumor tissues compared to normal tissues (p < 0.05). Fluorescence-activated cell sorting (FACS)-isolated lamina CAC adenomas revealed macrophages and dendritic cells as primary TPM2 contributors (p < 0.001). Lentiviral TPM2 gene knockdown significantly reduced tumor numbers and sizes in CAC mice (p < 0.01, and p < 0.001), without invasive cancer cells. TPM2 suppression resulted in decreased IEC proliferation (p < 0.001) and reduced PCNA and Ki-67 expression (p < 0.05). Western blot analysis indicated reduced JNK pathway activation in TPM2-knockdown CAC mice (p < 0.05, p < 0.001). TPM2 knockdown decreased tumor-associated macrophage infiltration (p < 0.01) and increased CD3+ and CD8+ T cells (p < 0.01, and p < 0.001), with increased levels of regulator of inflammatory cytokines (CD44+, CD107a+) (p < 0.01, and p < 0.001), decreased levels of PD-1+ and anti-inflammatory factor (IL10+) (p < 0.01, and p < 0.001). CONCLUSIONS: Our results demonstrated that TPM2 knockdown suppressed the proliferation of CAC IECs, enhanced immune suppression on CAC IECs, and inhibited the JNK signaling pathway within the framework of CAC. These findings suggest TPM2 can serve as a potential therapeutic target for CAC treatment.


Cell Proliferation , Colitis-Associated Neoplasms , MAP Kinase Signaling System , Tropomyosin , Animals , Tropomyosin/metabolism , Tropomyosin/immunology , Tropomyosin/genetics , Mice , MAP Kinase Signaling System/immunology , Colitis-Associated Neoplasms/pathology , Colitis-Associated Neoplasms/immunology , Colitis-Associated Neoplasms/metabolism , Male , Dextran Sulfate/toxicity , Humans , Azoxymethane/toxicity , Mice, Inbred C57BL , Disease Models, Animal , Colitis/chemically induced , Colitis/pathology , Colitis/complications , Colitis/immunology
3.
Discov Med ; 36(183): 836-841, 2024 Apr.
Article En | MEDLINE | ID: mdl-38665031

BACKGROUND: Over 80% of lung cancer cases constitute non-small cell lung cancer (NSCLC), making it the most prevalent type of lung cancer globally and the leading cause of cancer-related deaths. The treatment of NSCLC patients with gefitinib has demonstrated promising initial efficacy. However, the underlying mechanism remains unclear. This study aims to investigate how gefitinib affects the mitogen-activated protein kinase kinase (MEK)/extracellular regulated protein kinases (ERK) signaling pathway-mediated growth and death of NSCLC cells. METHODS: In this study, the NSCLC cell line A549 was cultured in vitro and divided into a control group and a gefitinib group. The viability of the A549 cells was assessed using the methylthiazolyldiphenyl-tetrazolium bromide (MTT) assay. Flow cytometry was employed to detect apoptosis in A549 cells, and the expression of glutamate dehydrogenase (GDH1) mRNA in these cells was determined using real-time quantitative PCR (RT-PCR). Western blotting was utilized to evaluate the protein expression levels of key components in the MEK/ERK signaling pathway, including phospho-MEK1/2, MEK1/2, phospho-ERK1/2, and ERK1/2. Additionally, intracellular glutamine content in A549 cells was measured using a colorimetric method. RESULTS: In contrast to the control group, the proliferation of A549 cells, the transcription level of glutamate dehydrogenase (GDH1), the intracellular glutamine content, and the protein expression levels of phospho-MEK1/2 and phospho-ERK1/2 were significantly lower in the gefitinib group. Moreover, apoptosis markedly increased. CONCLUSIONS: Gefitinib expedites apoptosis and diminishes proliferation in the NSCLC cell line A549 by downregulating the epidermal growth factor receptor (EGFR)/MEK/ERK signaling pathway. This effect is accomplished by fostering the expression of GDH1 to augment glutaminolysis in A549 cells.


Apoptosis , Carcinoma, Non-Small-Cell Lung , Gefitinib , Glutamine , Lung Neoplasms , MAP Kinase Signaling System , Humans , Gefitinib/pharmacology , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/metabolism , Apoptosis/drug effects , MAP Kinase Signaling System/drug effects , Lung Neoplasms/pathology , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , A549 Cells , Glutamine/metabolism , Quinazolines/pharmacology , Quinazolines/therapeutic use , Glutamate Dehydrogenase/metabolism , Antineoplastic Agents/pharmacology , Cell Proliferation/drug effects , Cell Line, Tumor
4.
Aging (Albany NY) ; 16(7): 6510-6520, 2024 Apr 04.
Article En | MEDLINE | ID: mdl-38579169

Our research investigated the effects of hsa-miR-134-5p on glioma progression, focusing on its interaction with the BDNF/ERK signaling pathway. U251 and U87 cell lines were analyzed post-transfection with hsa-miR-134-5p mimics and inhibitors, confirming the miRNA's binding to BDNF using dual luciferase assays. Q-PCR was employed to measure expression changes, revealing that hsa-miR-134-5p markedly inhibited glioma cell proliferation, migration, and invasion, as evidenced by CCK8, monoclonal formation, and Transwell assays. Scratch tests and Western blotting demonstrated hsa-miR-134-5p's modulation of the BDNF/ERK pathway and associated decrease in MMP2/9 protein levels. Flow cytometry suggested that hsa-miR-134-5p might also block the G0/S phase transition. In vivo studies using nude mice corroborated the tumor-suppressing effects of hsa-miR-134-5p, which were negated by elevated BDNF levels. Comparative protein analysis across groups confirmed the pathway's significance in tumorigenesis. Our findings identify hsa-miR-134-5p as a key molecule impeding glioma cell growth by curtailing the BDNF/ERK pathway, with the reversal by BDNF upregulation pointing to the potential of therapeutically exploiting the hsa-miR-134-5p/BDNF axis in glioma care.


Brain-Derived Neurotrophic Factor , Cell Movement , Cell Proliferation , Glioma , MAP Kinase Signaling System , Mice, Nude , MicroRNAs , MicroRNAs/genetics , MicroRNAs/metabolism , Glioma/pathology , Glioma/metabolism , Glioma/genetics , Humans , Brain-Derived Neurotrophic Factor/metabolism , Brain-Derived Neurotrophic Factor/genetics , Animals , Cell Line, Tumor , Mice , Gene Expression Regulation, Neoplastic , Brain Neoplasms/pathology , Brain Neoplasms/metabolism , Brain Neoplasms/genetics
5.
Mol Nutr Food Res ; 68(8): e2300643, 2024 Apr.
Article En | MEDLINE | ID: mdl-38600887

SCOPE: Polyphenols are the major active substances in red jujube fruit, and their anti-inflammatory and antioxidant activities suggest their potential utility in the prevention of ulcerative colitis (UC). METHODS AND RESULTS: In this study, the effect of polyphenol extracts from red jujube (Ziziphus jujuba Mill. "Junzao") (PERJ) on the dextron sulfate sodium (DSS)-induced UC mice is investigated. The result shows that PERJ effectively improves clinical symptoms, including food and water intake, the disease activity insex (DAI) and spleen index, and routine blood levels, and alleviates the shortening of the colon, in mice with DSS-induced UC. Meanwhile, PERJ remarkably decreases the expression of proinflammatory factors. Moreover, PERJ repairs intestinal barrier damage by increasing the expression level of mucin 2 and mucin 3, and the result is also confirmed in the histological assessment. Besides, the expression levels of Nod-like receptor family pyrin domain-containing 3 (NLRP3) and mitogen-activated protein kinase cascade (MAPKs) signaling pathway-related proteins are inhibited by the PERJ administration. Finally, 16S rRNA sequencing analyses reveal that PERJ reverses intestinal microbiota dysbiosis by enhancing the abundance of Firmicutes and decreasing that of Proteobacteria and Bacteroidetes. CONCLUSION: PERJ probably inhibits the development of UC by suppressing the NLRP3 and MAPKs signaling pathways and regulating gut microbiota homeostasis, and can be considered as a potential resource for preventing UC.


Colitis, Ulcerative , Gastrointestinal Microbiome , Homeostasis , MAP Kinase Signaling System , NLR Family, Pyrin Domain-Containing 3 Protein , Plant Extracts , Polyphenols , Ziziphus , Animals , Ziziphus/chemistry , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Gastrointestinal Microbiome/drug effects , Colitis, Ulcerative/drug therapy , Colitis, Ulcerative/chemically induced , Polyphenols/pharmacology , Plant Extracts/pharmacology , Male , MAP Kinase Signaling System/drug effects , Homeostasis/drug effects , Mice , Mice, Inbred C57BL , Dextran Sulfate , Colon/drug effects , Colon/metabolism , Colon/pathology
6.
Aging (Albany NY) ; 16(7): 6588-6612, 2024 Apr 05.
Article En | MEDLINE | ID: mdl-38604156

BACKGROUND: Liver progenitor cells (LPCs) are a subpopulation of cells that contribute to liver regeneration, fibrosis and liver cancer initiation under different circumstances. RESULTS: By performing adenoviral-mediated transfection, CCK-8 analyses, F-actin staining, transwell analyses, luciferase reporter analyses and Western blotting, we observed that TGF-ß promoted cytostasis and partial epithelial-mesenchymal transition (EMT) in LPCs. In addition, we confirmed that TGF-ß activated the Smad and MAPK pathways, including the Erk, JNK and p38 MAPK signaling pathways, and revealed that TGFß-Smad signaling induced growth inhibition and partial EMT, whereas TGFß-MAPK signaling had the opposite effects on LPCs. We further found that the activity of Smad and MAPK signaling downstream of TGF-ß was mutually restricted in LPCs. Mechanistically, we found that TGF-ß activated Smad signaling through serine phosphorylation of both the C-terminal and linker regions of Smad2 and 3 in LPCs. Additionally, TGFß-MAPK signaling inhibited the phosphorylation of Smad3 but not Smad2 at the C-terminus, and it reinforced the linker phosphorylation of Smad3 at T179 and S213. We then found that overexpression of mutated Smad3 at linker phosphorylation sites intensifies TGF-ß-induced cytostasis and EMT, mimicking the effects of MAPK inhibition in LPCs, whereas mutation of Smad3 at the C-terminus caused LPCs to blunt TGF-ß-induced cytostasis and partial EMT. CONCLUSION: These results suggested that TGF-ß downstream of Smad3 and MAPK signaling were mutually antagonistic in regulating the viability and partial EMT of LPCs. This antagonism may help LPCs overcome the cytostatic effect of TGF-ß under fibrotic conditions and maintain partial EMT and progenitor phenotypes.


Epithelial-Mesenchymal Transition , Liver , MAP Kinase Signaling System , Smad3 Protein , Stem Cells , Transforming Growth Factor beta , Smad3 Protein/metabolism , Stem Cells/metabolism , Animals , Transforming Growth Factor beta/metabolism , MAP Kinase Signaling System/physiology , Liver/metabolism , Cell Survival/drug effects , Phosphorylation , Mice , Signal Transduction
7.
Aging (Albany NY) ; 16(7): 6566-6587, 2024 Apr 05.
Article En | MEDLINE | ID: mdl-38604164

Traumatic brain injury (TBI) and its resulting complications pose a major challenge to global public health, resulting in increased rates of disability and mortality. Cerebrovascular dysfunction is nearly universal in TBI cases and is closely associated with secondary injury after TBI. Transcranial direct current stimulation (tDCS) shows great potential in the treatment of TBI; however, the exact mechanism remains elusive. In this study, we performed in vivo and in vitro experiments to explore the effects and mechanisms of tDCS in a controlled cortical impact (CCI) rat model simulating TBI. In vivo experiments show that tDCS can effectively reduce brain tissue damage, cerebral edema and neurological deficits. The potential mechanism may be that tDCS improves the neurological function of rats by increasing orexin A (OXA) secretion, upregulating the TF-AKT/ERK signaling pathway, and promoting angiogenesis at the injury site. Cellular experiments showed that OXA promoted HUVEC migration and angiogenesis, and these effects were counteracted by the ERK1/2 inhibitor LY3214996. The results of Matrigel experiment in vivo showed that TNF-a significantly reduced the ability of HUVEC to form blood vessels, but OXA could rescue the effect of TNF-a on the ability of HUVEC to form blood vessels. However, LY3214996 could inhibit the therapeutic effect of OXA. In summary, our preliminary study demonstrates that tDCS can induce angiogenesis through the OXA-TF-AKT/ERK signaling pathway, thereby improving neurological function in rats with TBI.


Brain Injuries, Traumatic , MAP Kinase Signaling System , Neovascularization, Physiologic , Proto-Oncogene Proteins c-akt , Transcranial Direct Current Stimulation , Animals , Brain Injuries, Traumatic/metabolism , Brain Injuries, Traumatic/therapy , Proto-Oncogene Proteins c-akt/metabolism , Rats , Male , Neovascularization, Physiologic/drug effects , Rats, Sprague-Dawley , Humans , Human Umbilical Vein Endothelial Cells , Disease Models, Animal , Signal Transduction , 60489
8.
Sci Rep ; 14(1): 9411, 2024 04 24.
Article En | MEDLINE | ID: mdl-38658579

Matrix Metalloproteinases (MMPs) have been demonstrated to be essential in facilitating the migration and metastasis of clear cell renal cell carcinoma (ccRCC). However, the ability of the MMP family to predict clinical outcomes and guide optimal therapeutic strategies for ccRCC patients remains incompletely understood. In this investigation, we initially conducted a thorough examination of the MMP family in pan-cancer. Notably, MMPs exhibited distinctive significance in ccRCC. Following this, we undertook an extensive analysis to evaluate the clinical value of MMPs and potential mechanisms by which MMPs contribute to the progression of ccRCC. A novel stratification method and prognostic model were developed based on MMPs in order to enhance the accuracy of prognosis prediction for ccRCC patients and facilitate personalized treatment. By conducting multi-omics analysis and transcriptional regulation analysis, it was hypothesized that SAA1 plays a crucial role in promoting ccRCC migration through MMPs. Subsequently, in vitro experiments confirmed that SAA1 regulates ccRCC cell migration via the ERK-AP1-MMPs axis. In conclusion, our study has explored the potential value of the MMP family as prognostic markers for ccRCC and as guides for medication regimens. Additionally, we have identified SAA1 as a crucial factor in the migration of ccRCC.


Carcinoma, Renal Cell , Cell Movement , Kidney Neoplasms , Matrix Metalloproteinases , Serum Amyloid A Protein , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/pathology , Carcinoma, Renal Cell/metabolism , Humans , Cell Movement/genetics , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , Kidney Neoplasms/metabolism , Matrix Metalloproteinases/metabolism , Matrix Metalloproteinases/genetics , Prognosis , Cell Line, Tumor , Serum Amyloid A Protein/metabolism , Serum Amyloid A Protein/genetics , Gene Expression Regulation, Neoplastic , MAP Kinase Signaling System , Female , Extracellular Signal-Regulated MAP Kinases/metabolism , Male , Signal Transduction
9.
J Neuroinflammation ; 21(1): 105, 2024 Apr 22.
Article En | MEDLINE | ID: mdl-38649885

BACKGROUND: NADPH oxidase (NOX), a primary source of endothelial reactive oxygen species (ROS), is considered a key event in disrupting the integrity of the blood-retinal barrier. Abnormalities in neurovascular-coupled immune signaling herald the loss of ganglion cells in glaucoma. Persistent microglia-driven inflammation and cellular innate immune system dysregulation often lead to deteriorating retinal degeneration. However, the crosstalk between NOX and the retinal immune environment remains unresolved. Here, we investigate the interaction between oxidative stress and neuroinflammation in glaucoma by genetic defects of NOX2 or its regulation via gp91ds-tat. METHODS: Ex vivo cultures of retinal explants from wildtype C57BL/6J and Nox2 -/- mice were subjected to normal and high hydrostatic pressure (Pressure 60 mmHg) for 24 h. In vivo, high intraocular pressure (H-IOP) was induced in C57BL/6J mice for two weeks. Both Pressure 60 mmHg retinas and H-IOP mice were treated with either gp91ds-tat (a NOX2-specific inhibitor). Proteomic analysis was performed on control, H-IOP, and treatment with gp91ds-tat retinas to identify differentially expressed proteins (DEPs). The study also evaluated various glaucoma phenotypes, including IOP, retinal ganglion cell (RGC) functionality, and optic nerve (ON) degeneration. The superoxide (O2-) levels assay, blood-retinal barrier degradation, gliosis, neuroinflammation, enzyme-linked immunosorbent assay (ELISA), western blotting, and quantitative PCR were performed in this study. RESULTS: We found that NOX2-specific deletion or activity inhibition effectively attenuated retinal oxidative stress, immune dysregulation, the internal blood-retinal barrier (iBRB) injury, neurovascular unit (NVU) dysfunction, RGC loss, and ON axonal degeneration following H-IOP. Mechanistically, we unveiled for the first time that NOX2-dependent ROS-driven pro-inflammatory signaling, where NOX2/ROS induces endothelium-derived endothelin-1 (ET-1) overexpression, which activates the ERK1/2 signaling pathway and mediates the shift of microglia activation to a pro-inflammatory M1 phenotype, thereby triggering a neuroinflammatory outburst. CONCLUSIONS: Collectively, we demonstrate for the first time that NOX2 deletion or gp91ds-tat inhibition attenuates iBRB injury and NVU dysfunction to rescue glaucomatous RGC loss and ON axon degeneration, which is associated with inhibition of the ET-1/ERK1/2-transduced shift of microglial cell activation toward a pro-inflammatory M1 phenotype, highlighting NOX2 as a potential target for novel neuroprotective therapies in glaucoma management.


Blood-Retinal Barrier , Intraocular Pressure , Mice, Inbred C57BL , NADPH Oxidase 2 , Neuroinflammatory Diseases , Animals , NADPH Oxidase 2/metabolism , NADPH Oxidase 2/genetics , Mice , Blood-Retinal Barrier/pathology , Blood-Retinal Barrier/metabolism , Intraocular Pressure/physiology , Neuroinflammatory Diseases/metabolism , Neuroinflammatory Diseases/pathology , Mice, Knockout , Cell Proliferation/physiology , MAP Kinase Signaling System/physiology , Neuroglia/metabolism , Neuroglia/pathology , Ocular Hypertension/pathology , Ocular Hypertension/metabolism , Glaucoma/pathology , Glaucoma/metabolism , Oxidative Stress/physiology
10.
Sci Rep ; 14(1): 7747, 2024 04 02.
Article En | MEDLINE | ID: mdl-38565569

6-Gingerol, the main bioactive compound of ginger, has antioxidant, anti-inflammatory, anti-cancer and neuroprotective effects. However, it is unclear whether 6-Gingerol has protective effects against hepatic ischemia/reperfusion (I/R) injury. In this study, the mouse liver I/R injury model and the mouse AML12 cell hypoxia/reoxygenation (H/R) model were established by pretreatment with 6-Gingerol at different concentrations to explore the potential effects of 6-Gingerol. Serum transaminase levels, liver necrotic area, cell viability, inflammatory response, and cell apoptosis were used to assess the effect of 6-Gingerol on hepatic I/R or cell H/R injury. Quantitative polymerase chain reaction (qPCR) and Western blotting were used to detect the mRNA and protein expression. The results show that 6-Gingerol decreased serum alanine aminotransferase (ALT), aspartate aminotransferase (AST) levels, liver necrosis, inflammatory cytokines IL-1ß, IL-6, MCP-1, TNF-α expression, Ly6g+ inflammatory cell infiltration, protein phosphorylation of NF-κB signaling pathway, Terminal deoxynucleotidyl transferase dUTP nick-end labeling (TUNEL) positive cells, cell apoptosis rate, the protein expression of pro-apoptotic protein BAX and C-Caspase3, increased cell viability, and expression of anti-apoptotic protein BCL-2. Moreover, 6-Gingerol could increase the mRNA and protein expression of mitogen activated protein kinase phosphatase 5 (MKP5) and inhibit the activation of P38/JNK signaling pathway. In MKP5 knockout (KO) mice, the protective effect of 6-gingerol and the inhibition of P38/JNK pathway were significantly weakened. Therefore, our results suggest that 6-Gingerol exerts anti-inflammatory and anti-apoptotic effects to attenuate hepatic I/R injury by regulating the MKP5-mediated P38/JNK signaling pathway.


Catechols , Fatty Alcohols , MAP Kinase Signaling System , Reperfusion Injury , Mice , Animals , Reperfusion Injury/drug therapy , Liver , Ischemia , Anti-Inflammatory Agents/pharmacology , Apoptosis Regulatory Proteins/pharmacology , Apoptosis , RNA, Messenger/pharmacology
11.
Cell Rep ; 43(4): 114033, 2024 Apr 23.
Article En | MEDLINE | ID: mdl-38568811

Small GTPases of the Ras subfamily are best known for their role as proto-oncoproteins, while their function during microbial infection has remained elusive. Here, we show that Legionella pneumophila hijacks the small GTPase NRas to the Legionella-containing vacuole (LCV) surface. A CRISPR interference screen identifies a single L. pneumophila effector, DenR (Lpg1909), required for this process. Recruitment is specific for NRas, while its homologs KRas and HRas are excluded from LCVs. The C-terminal hypervariable tail of NRas is sufficient for recruitment, and interference with either NRas farnesylation or S-acylation sites abrogates recruitment. Intriguingly, we detect markers of active NRas signaling on the LCV, suggesting it acts as a signaling platform. Subsequent phosphoproteomics analyses show that DenR rewires the host NRas signaling landscape, including dampening of the canonical mitogen-activated protein kinase pathway. These results provide evidence for L. pneumophila targeting NRas and suggest a link between NRas GTPase signaling and microbial infection.


Bacterial Proteins , GTP Phosphohydrolases , Legionella pneumophila , MAP Kinase Signaling System , Membrane Proteins , Legionella pneumophila/metabolism , Legionella pneumophila/pathogenicity , Humans , Membrane Proteins/metabolism , Membrane Proteins/genetics , GTP Phosphohydrolases/metabolism , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Down-Regulation , HEK293 Cells , Legionnaires' Disease/microbiology , Legionnaires' Disease/metabolism , Vacuoles/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , Proto-Oncogene Proteins p21(ras)/genetics
12.
Cell Commun Signal ; 22(1): 214, 2024 Apr 03.
Article En | MEDLINE | ID: mdl-38570838

Smoking is the major cause of cardiovascular diseases and cancer. It induces oxidative stress, leading to DNA damage and cellular senescence. Senescent cells increase the expression and release of pro-inflammatory molecules and matrix metalloproteinase, which are known to play a vital role in the initiation and progression of cardiovascular diseases and metastasis in cancer. The current study investigated the smoking induced cellular senescence and employed colchicine that blocked senescence in endothelial cells exposed to tobacco smoke condensate. Colchicine prevented oxidative stress and DNA damage in tobacco smoke-condensate-treated endothelial cells. Colchicin reduced ß-gal activity, improved Lamin B1, and attenuated cell growth arrest markers P21 and P53. Colchicine also ameliorated the expression of SASP factors and inhibited the activation of NF-kB and MAPKs P38 and ERK. In summary, colchicine inhibited tobacco smoke condensate-induced senescence in endothelial cells by blocking the activation of NF-kB and MAPKs P38 and ERK.


Cardiovascular Diseases , Neoplasms , Tobacco Smoke Pollution , Humans , NF-kappa B/metabolism , Endothelial Cells/metabolism , MAP Kinase Signaling System , Smoke/adverse effects , Cellular Senescence
13.
Biochemistry (Mosc) ; 89(2): 201-211, 2024 Feb.
Article En | MEDLINE | ID: mdl-38622090

Visomitin eye drops are the first and, so far, the only drug based on SkQ1 - the mitochondria-targeted antioxidant 10-(6'-plastoquinonyl) decyltriphenylphosphonium, developed in the laboratories of Moscow State University under the leadership of Academician V. P. Skulachev. SkQ1 is considered as a potential tool to combat the aging program. We have previously shown that it is able to prevent and/or suppress development of all manifestations of accelerated senescence in OXYS rats, including retinopathy, similar to the age-related macular degeneration (AMD). Here, we assessed the effect of Visomitin instillations on progression of the AMD-like pathology and p38 MAPK and ERK1/2 activity in the OXYS rat retina (from the age of 9 to 12 months). Wistar and OXYS rats treated with placebo (composition identical to Visomitin with the exception of SkQ1) were used as controls. Ophthalmological examination showed that in the OXYS rats receiving placebo, retinopathy progressed and severity of clinical manifestations did not differ from the intact OXYS rats. Visomitin suppressed progression of the AMD-like pathology in the OXYS rats and significantly improved structural and functional parameters of the retinal pigment epithelium cells and state of microcirculation in the choroid, which, presumably, contributed to preservation of photoreceptors, associative and ganglion neurons. It was found that the activity of p38 MAPK and ERK1/2 in the retina of 12-month-old OXYS rats is higher than that of the Wistar rats of the same age, as indicated by the increased content of phosphorylated forms of p38 MAPK and ERK1/2 and their target protein tau (at position T181 and S396). Visomitin decreased phosphorylation of p38 MAPK, ERK1/2, and tau indicating suppression of activity of these MAPK signaling cascades. Thus, Visomitin eye drops are able to suppress progression of the AMD-like pathology in the OXYS rats and their effect is associated with the decrease in activity of the MAPK signaling cascades.


Benzalkonium Compounds , MAP Kinase Signaling System , Macular Degeneration , Methylcellulose , Plastoquinone , Humans , Rats , Animals , Infant , Rats, Wistar , Ophthalmic Solutions/pharmacology , p38 Mitogen-Activated Protein Kinases/metabolism , Macular Degeneration/drug therapy , Macular Degeneration/metabolism , Aging/metabolism , Signal Transduction , Drug Combinations
14.
PeerJ ; 12: e17123, 2024.
Article En | MEDLINE | ID: mdl-38560469

Background: The purpose of the present study was to investigate the effect of erythropoietin (EPO) on lung ischemia-reperfusion injury (LIRI). Methods: Sprague Dawley rats and BEAS-2B cells were employed to construct an ischemia-reperfusion (I/R)-induced model in vivo and in vitro, respectively. Afterward, I/R rats and tert-butyl hydroperoxide (TBHP)-induced cells were treated with different concentrations of EPO. Furthermore, 40 patients with LIRI and healthy controls were enrolled in the study. Results: It was observed that lung tissue damage, cell apoptosis and the expression of BAX and caspase-3 were higher in the LIRI model in vivo and in vitro than in the control group, nevertheless, the Bcl-2, FGF23 and FGFR4 expression level was lower than in the control group. EPO administration significantly reduced lung tissue damage and cell apoptosis while also up-regulating the expression of FGF23 and FGFR4. Rescue experiments indicated that EPO exerted a protective role associated with the FGF23/FGFR4/p-ERK1/2 signal pathway. Notably, the expression of serum EPO, FGF23, FGFR4 and Bcl-2 was decreased in patients with LIRI, while the expression of caspase-3 and BAX was higher. Conclusion: EPO could effectively improve LIRI, which might be related to the activation of the FGF23/FGFR4/p-ERK1/2 signaling pathway.


Erythropoietin , Reperfusion Injury , Animals , Humans , Rats , bcl-2-Associated X Protein/metabolism , Caspase 3/genetics , Epoetin Alfa/metabolism , Erythropoietin/pharmacology , Ischemia , Lung/metabolism , MAP Kinase Signaling System , Proto-Oncogene Proteins c-bcl-2/metabolism , Rats, Sprague-Dawley , Receptor, Fibroblast Growth Factor, Type 4/genetics , Reperfusion Injury/drug therapy , Signal Transduction
15.
Elife ; 122024 Apr 02.
Article En | MEDLINE | ID: mdl-38564252

Currently, the identification of patient-specific therapies in cancer is mainly informed by personalized genomic analysis. In the setting of acute myeloid leukemia (AML), patient-drug treatment matching fails in a subset of patients harboring atypical internal tandem duplications (ITDs) in the tyrosine kinase domain of the FLT3 gene. To address this unmet medical need, here we develop a systems-based strategy that integrates multiparametric analysis of crucial signaling pathways, and patient-specific genomic and transcriptomic data with a prior knowledge signaling network using a Boolean-based formalism. By this approach, we derive personalized predictive models describing the signaling landscape of AML FLT3-ITD positive cell lines and patients. These models enable us to derive mechanistic insight into drug resistance mechanisms and suggest novel opportunities for combinatorial treatments. Interestingly, our analysis reveals that the JNK kinase pathway plays a crucial role in the tyrosine kinase inhibitor response of FLT3-ITD cells through cell cycle regulation. Finally, our work shows that patient-specific logic models have the potential to inform precision medicine approaches.


Leukemia, Myeloid, Acute , Signal Transduction , Humans , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , MAP Kinase Signaling System , Cell Line , Drug Resistance , fms-Like Tyrosine Kinase 3/genetics
16.
Int J Hyperthermia ; 41(1): 2335199, 2024.
Article En | MEDLINE | ID: mdl-38565204

PURPOSE: c-Jun N-terminal kinases (JNKs) comprise a subfamily of mitogen-activated protein kinases (MAPKs). The JNK group is known to be activated by a variety of stimuli. However, the molecular mechanism underlying heat-induced JNK activation is largely unknown. The aim of this study was to clarify how JNK activity is stimulated by heat. METHODS AND MATERIALS: The expression levels of various MAPK members in HeLa cells, with or without hyperthermia treatment, were evaluated via western blotting. The kinase activity of MAPK members was assessed through in vitro kinase assays. Cell death was assessed in the absence or presence of siRNAs targeting MAPK-related members. RESULTS: Hyperthermia decreased the levels of MAP3Ks, such as ASK1 and MLK3 which are JNK kinase kinase members, but not those of the downstream MAP2K/SEK1 and MAPK/JNK. Despite the reduced or transient phosphorylation of ASK1, MLK3, or SEK1, downstream JNK was phosphorylated in a temperature-dependent manner. In vitro kinase assays demonstrated that heat did not directly stimulate SEK1 or JNK. However, the expression levels of DUSP16, a JNK phosphatase, were decreased upon hyperthermia treatment. DUSP16 knockdown enhanced the heat-induced activation of ASK1-SEK1-JNK pathway and apoptosis. CONCLUSION: JNK was activated in a temperature-dependent manner despite reduced or transient phosphorylation of the upstream MAP3K and MAP2K. Hyperthermia-induced degradation of DUSP16 may induce activation of the ASK1-SEK1-JNK pathway and subsequent apoptosis.


Hyperthermia, Induced , MAP Kinase Signaling System , Humans , HeLa Cells , Mitogen-Activated Protein Kinases/metabolism , Mitogen-Activated Protein Kinase Kinases/metabolism , Apoptosis/physiology
17.
Chem Biol Drug Des ; 103(4): e14519, 2024 Apr.
Article En | MEDLINE | ID: mdl-38570708

Kaempferol (KPR), a flavonoid compound found in various plants and foods, has garnered attention for its anti-inflammatory, antioxidant, and anticancer properties. In preliminary studies, KPR can modulate several signaling pathways involved in inflammation, making it a candidate for treating cholecystitis. This study aimed to explore the effects and mechanisms of KPR on lipopolysaccharide (LPS)-induced human gallbladder epithelial cells (HGBECs). To assess the impact of KPR on HGBECs, the HGBECs were divided into control, KPR, LPS, LPS + KPR, and LPS + UDCA groups. Cell viability and cytotoxicity were evaluated by MTT assay and lactate dehydrogenase (LDH) assay, respectively, and concentrations of KPR (10-200 µM) were tested. LPS-induced inflammatory responses in HGBECs were to create an in vitro model of cholecystitis. The key inflammatory markers (IL-1ß, IL-6, and TNF-α) levels were quantified using ELISA, The modulation of the MAPK/NF-κB signaling pathway was measured by western blot using specific antibodies against pathway components (p-IκBα, IκBα, p-p65, p65, p-JNK, JNK, p-ERK, ERK, p-p38, and p38). The cell viability and LDH levels in HGBECs were not significantly affected by 50 µM KPR, thus it was selected as the optimal KPR intervention concentration. KPR increased the viability of LPS-induced HGBECs. Additionally, KPR inhibited the inflammatory factors level (IL-1ß, IL-6, and TNF-α) and protein expression (iNOS and COX-2) in LPS-induced HGBECs. Furthermore, KPR reversed LPS-induced elevation of p-IκBα/IκBα, p-p65/p65, p-JNK/JNK, p-ERK/ERK, and p-p38/p38 ratios. KPR attenuates the LPS-induced inflammatory response in HGBECs, possibly by inhibiting MAPK/NF-κB signaling.


Cholecystitis , NF-kappa B , Humans , NF-kappa B/metabolism , Lipopolysaccharides/toxicity , NF-KappaB Inhibitor alpha/metabolism , Tumor Necrosis Factor-alpha/metabolism , Interleukin-6/metabolism , Kaempferols/pharmacology , Signal Transduction , Inflammation/chemically induced , Inflammation/drug therapy , Epithelial Cells/metabolism , MAP Kinase Signaling System
18.
Curr Pharm Biotechnol ; 25(4): 499-509, 2024.
Article En | MEDLINE | ID: mdl-38572608

Background: Salpingitis obstructive infertility (SOI) refers to infertility caused by abnormal conditions such as tubal adhesion and blockage caused by acute and chronic salpingitis. SOI has a serious impact on women's physical and mental health and family harmony, and it is a clinical problem that needs to be solved urgently.

Objective: The purpose of the present study was to explore the potential pharmacological mechanisms of the Yinjia tablets (Yin Jia Pian, YJP) on tubal inflammation.

Methods: Networks of YJP-associated targets and tubal inflammation-related genes were constructed through the STRING database. Potential targets and pathway enrichment analysis related to the therapeutic efficacy of YJP were identified using Cytoscape and Database for Annotation, Visualization, and Integrated Discovery (metascape). E. coli was used to establish a rat model of tubal inflammation and to validate the predictions of network pharmacology and the therapeutic efficacy of YJP. H&E staining was used to observe the pathological changes in fallopian tubes. TEM observation of the ultrastructure of the fallopian tubes. ELISA was used to detect the changes of IL-6 and TNF-α in fallopian tubes. Immunohistochemistry was used to detect the expression of ESR1. The changes of Bcl-2, ERK1/2, p-ERK1/2, MEK, p-MEK, EGFR, and p-EGFR were detected by western blot.

Results: Through database analysis, it was found that YJP shared 105 identical targets with the disease. Network pharmacology analysis showed that IL-6, TNF, and EGFR belong to the top 5 core proteins associated with salpingitis, and EGFR/MEK/ERK may be the main pathway involved. The E. coli-induced disease rat model of fallopian tube tissue showed damage, mitochondrial disruption, and increased levels of the inflammatory factors IL-6 and TNF-α. Tubal inflammatory infertility rats have increased expression of Bcl-2, p-ERK1/2, p-MEK, and p-EGFR, and decreased expression of ESR1. In vivo, experiments showed that YJP improved damage of tissue, inhibited shedding of tubal cilia, and suppressed the inflammatory response of the body. Furthermore, YJP inhibited EGFR/MEK/ERK signaling, inhibited the apoptotic protein Bcl-2, and upregulated ESR1.

Conclusion: This study revealed that YJP Reducing tubal inflammation and promoting tissue repair may be associated with inhibition of the EGFR/MEK/ERK signaling pathway.

.


Drugs, Chinese Herbal , Infertility , Salpingitis , Humans , Female , Rats , Animals , Salpingitis/complications , Salpingitis/metabolism , Salpingitis/pathology , MAP Kinase Signaling System , Tumor Necrosis Factor-alpha/metabolism , Interleukin-6/metabolism , Escherichia coli/metabolism , Network Pharmacology , Infertility/complications , Signal Transduction , Inflammation/drug therapy , ErbB Receptors/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Mitogen-Activated Protein Kinase Kinases/metabolism
19.
J Vet Sci ; 25(2): e21, 2024 Mar.
Article En | MEDLINE | ID: mdl-38568823

BACKGROUND: Peste des petits ruminants (PPR) is a contagious and fatal disease of sheep and goats. PPR virus (PPRV) infection induces endoplasmic reticulum (ER) stress-mediated unfolded protein response (UPR). The activation of UPR signaling pathways and their impact on apoptosis and virus replication remains controversial. OBJECTIVES: To investigate the role of PPRV-induced ER stress and the IRE1-XBP1 and IRE1-JNK pathways and their impact on apoptosis and virus replication. METHODS: The cell viability and virus replication were assessed by 3-(4,5-Dimethyl-2-thiazolyl)-2,5-diphenyl-2H-tetrazolium bromide assay, immunofluorescence assay, and Western blot. The expression of ER stress biomarker GRP78, IRE1, and its downstream molecules, PPRV-N protein, and apoptosis-related proteins was detected by Western blot and quantitative reverse transcription-polymerase chain reaction, respectively. 4-Phenylbutyric acid (4-PBA) and STF-083010 were respectively used to inhibit ER stress and IRE1 signaling pathway. RESULTS: The expression of GRP78, IRE1α, p-IRE1α, XBP1s, JNK, p-JNK, caspase-3, caspase-9, Bax and PPRV-N were significantly up-regulated in PPRV-infected cells, the expression of Bcl-2 was significantly down-regulated. Due to 4-PBA treatment, the expression of GRP78, p-IRE1α, XBP1s, p-JNK, caspase-3, caspase-9, Bax, and PPRV-N were significantly down-regulated, the expression of Bcl-2 was significantly up-regulated. Moreover, in PPRV-infected cells, the expression of p-IRE1α, p-JNK, Bax, and PPRV-N was significantly decreased, and the expression of Bcl-2 was increased in the presence of STF-083010. CONCLUSIONS: PPRV infection induces ER stress and IRE1 activation, resulting in apoptosis and enhancement of virus replication through IRE1-XBP1s and IRE1-JNK pathways.


Butylamines , Goat Diseases , Peste-des-Petits-Ruminants , Peste-des-petits-ruminants virus , Sheep Diseases , Sulfonamides , Thiophenes , Sheep , Animals , MAP Kinase Signaling System , Caspase 3/metabolism , Caspase 9/metabolism , Endoplasmic Reticulum Chaperone BiP , Endoribonucleases/metabolism , bcl-2-Associated X Protein/metabolism , Protein Serine-Threonine Kinases , Goats/metabolism , Apoptosis , Endoplasmic Reticulum Stress
20.
PLoS One ; 19(4): e0297695, 2024.
Article En | MEDLINE | ID: mdl-38568917

BACKGROUND: This study aims to study the possible action mechanism of T-cell immunoglobulin and mucin domain 3 (TIM3) on the migratory and invasive abilities of thyroid carcinoma (TC) cells. METHODS: GSE104005 and GSE138198 datasets were downloaded from the GEO database for identifying differentially expressed genes (DEGs). Functional enrichment analysis and protein-protein interaction (PPI) analysis were performed on the common DEGs in GSE104005 and GSE138198 datasets. Subsequently, in order to understand the effect of a common DEG (TIM3) on TC cells, we performed in vitro experiments using FRO cells. The migratory and invasive abilities of FRO cells were detected by wound scratch assay and Transwell assay. Proteins expression levels of the phosphorylated (p)-extracellular signal-regulated kinase (ERK)1/2, matrix metalloproteinase-2 (MMP-2) and MMP-9 were determined via Western blotting after ERK1/2 inhibition in TIM3-NC group and TIM3-mimic group. RESULTS: 316 common DEGs were identified in GSE104005 and GSE138198 datasets. These DEGs were involved in the biological process of ERK1 and ERK2 cascade. TIM3 was significantly up-regulated in TC. In vitro cell experiments showed that TIM3 could promote migration and invasion of TC cells. Moreover, TIM3 may affect the migration, invasive abilities of TC cells by activating the ERK1/2 pathway. CONCLUSION: The above results indicate that TIM3 may affect the migratory and invasive of TC cells by activating the ERK1/2 pathway.


MAP Kinase Signaling System , Thyroid Neoplasms , Humans , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 2/metabolism , Hepatitis A Virus Cellular Receptor 2/metabolism , Cell Line, Tumor , Neoplastic Processes , Thyroid Neoplasms/genetics , Cell Movement/genetics
...