Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.711
Filter
1.
Sci Rep ; 14(1): 22778, 2024 10 01.
Article in English | MEDLINE | ID: mdl-39354061

ABSTRACT

Histopathologically, oral squamous cell carcinoma (OSCC) consists of well-defined interfaces with adjacent non-cancerous epithelium. Previously, we found that SCC tissues expressed higher levels of specific proteins at this interface. Ladinin-1 (LAD1) is one of the specific molecules that has increased expressions in cancer fronts; however, its function in OSCC is unknown. Therefore, this study aimed to elucidate the function of LAD1 in human OSCC cells. LAD1 was localized on the actin arc at the distal periphery of cell clusters in the OSCC cell lines HSC-2, HSC-3, and HSC-4. When LAD1 was knocked down, cellular migration was repressed in wound scratch assays but was reversed in three-dimensional collagen gel invasion assays. Characteristic LAD1 localization along actin arcs forming the leading edge of migrating cells was diminished with loss of filopodia formation and ruffling in knockdown cells, in which the expression levels of cell motility-related genes-p21-activated kinase 1 (PAK1) and caveolin-1 (CAV1)-were upregulated and downregulated, respectively. LAD1 expression was also associated with the downregulation of vimentin and increased histological differentiation of OSCC. These results suggest that LAD1 is involved in actin dynamics during filopodia and lamellipodia formation, and in maintaining the epithelial phenotype of OSCC cells.


Subject(s)
Actins , Carcinoma, Squamous Cell , Cell Movement , Mouth Neoplasms , Humans , Actins/metabolism , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/genetics , Caveolin 1/metabolism , Caveolin 1/genetics , Cell Line, Tumor , Epithelial Cells/metabolism , Epithelial Cells/pathology , Gene Expression Regulation, Neoplastic , Mouth Neoplasms/pathology , Mouth Neoplasms/metabolism , Mouth Neoplasms/genetics , p21-Activated Kinases/metabolism , p21-Activated Kinases/genetics , Phenotype , Vimentin/metabolism , Vimentin/genetics
2.
Cells ; 13(17)2024 Aug 28.
Article in English | MEDLINE | ID: mdl-39273017

ABSTRACT

PAK4 and PD-L1 have been suggested as novel therapeutic targets in human cancers. Moreover, PAK4 has been suggested to be a molecule closely related to the immune evasion of cancers. Therefore, this study evaluated the roles of PAK4 and PD-L1 in the progression of osteosarcomas in 32 osteosarcomas and osteosarcoma cells. In human osteosarcomas, immunohistochemical positivity for the expression of PAK4 (overall survival, p = 0.028) and PD-L1 (relapse-free survival, p = 0.002) were independent indicators for the survival of patients in a multivariate analysis. In osteosarcoma cells, the overexpression of PAK4 increased proliferation and invasiveness, while the knockdown of PAK4 suppressed proliferation and invasiveness. The expression of PAK4 was associated with the expression of the molecules related to cell cycle regulation, invasion, and apoptosis. PAK4 was involved in resistance to apoptosis under a treatment regime with doxorubicin for osteosarcoma. In U2OS cells, PAK4 was involved in the stabilization of PD-L1 from ubiquitin-mediated proteasomal degradation and the in vivo infiltration of immune cells such as regulatory T cells and PD1-, CD4-, and CD8-positive cells in mice tumors. In conclusion, this study suggests that PAK4 is involved in the progression of osteosarcoma by promoting proliferation, invasion, and resistance to doxorubicin and stabilized PD-L1 from proteasomal degradation.


Subject(s)
B7-H1 Antigen , Cell Proliferation , Doxorubicin , Drug Resistance, Neoplasm , Osteosarcoma , p21-Activated Kinases , Osteosarcoma/pathology , Osteosarcoma/drug therapy , Osteosarcoma/metabolism , Osteosarcoma/genetics , Humans , B7-H1 Antigen/metabolism , Female , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Doxorubicin/pharmacology , Doxorubicin/therapeutic use , Animals , p21-Activated Kinases/metabolism , p21-Activated Kinases/genetics , Male , Cell Line, Tumor , Cell Proliferation/drug effects , Mice , Apoptosis/drug effects , Bone Neoplasms/metabolism , Bone Neoplasms/pathology , Bone Neoplasms/drug therapy , Bone Neoplasms/genetics , Adult , Adolescent , Protein Stability/drug effects , Mice, Nude , Young Adult , Gene Expression Regulation, Neoplastic/drug effects , Neoplasm Invasiveness
3.
J Cell Mol Med ; 28(17): e70065, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39233332

ABSTRACT

Glioblastoma multiforme (GBM) is a malignant tumour with a poor prognosis. Therefore, potential treatment strategies and novel therapeutic targets have gained increased attention. Our data showed that the ethanol extract of Vanilla planifolia stem (VAS) significantly decreased the viability and the colony formation of GBM cells. Moreover, VAS induced the cleavage of MAP1LC3, a marker of autophagy. Further RNA-seq and bioinformatic analysis revealed 4248 differentially expressed genes (DEGs) between VAS-treated GBM cells and the control cells. Protein-protein interactions between DEGs with fold changes less than -3 and more than 5 were further analysed, and we found that 16 and 9 hub DEGs, respectively, were correlated with other DEGs. Further qPCR experiments confirmed that 14 hub DEGs was significantly downregulated and 9 hub DEGs was significantly upregulated. In addition, another significantly downregulated DEG, p21-activated kinase 6 (PAK6), was correlated with the overall survival of GBM patients. Further validation experiments confirmed that VAS significantly reduced the mRNA and protein expression of PAK6, which led to the abolition of cell viability and colony formation. These findings demonstrated that VAS reduced cell viability, suppressed colony formation and induced autophagy and revealed PAK6 and other DEGs as potential therapeutic targets for GBM treatment.


Subject(s)
Autophagy , Gene Expression Regulation, Neoplastic , Glioblastoma , Plant Extracts , p21-Activated Kinases , Glioblastoma/metabolism , Glioblastoma/pathology , Glioblastoma/drug therapy , Glioblastoma/genetics , Humans , p21-Activated Kinases/metabolism , p21-Activated Kinases/genetics , Plant Extracts/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Cell Line, Tumor , Autophagy/drug effects , Cell Survival/drug effects , Plant Stems/chemistry , Ethanol , Cell Proliferation/drug effects , Protein Interaction Maps/drug effects , Cell Death/drug effects
4.
BMC Cancer ; 24(1): 1213, 2024 Sep 30.
Article in English | MEDLINE | ID: mdl-39350056

ABSTRACT

BACKGROUND: The main challenge in treating ovarian cancer is chemotherapy resistance. Previous studies have shown that PAK2 is highly expressed in various cancers. This research investigates whether increased PAK2 expression contributes to chemo-resistance and poor prognosis in ovarian cancer. METHODS: Initially, bioinformatics analysis was used to assess the importance of PAK2 mRNA up-regulation in ovarian cancer. This was then validated using tissue microarray to confirm PAK2 protein expression and localization in clinical samples. Univariate and multivariate logistic regression analyses were carried out to identify potential risk factors for chemo-resistance in serous epithelial ovarian cancer (EOC), while multivariate Cox regression and Kaplan-Meier analysis were conducted to ascertain prognostic factors for overall survival (OS) and disease-free survival (DFS) in patients with serous EOC. In vitro experiments were conducted to verify if inhibiting PAK2 expression could increase A2780/Taxol cells' sensitivity to paclitaxel, as shown by evaluating cell proliferation, apoptosis, transwell, and clone formation. Additionally, the interaction between PAK2, lnc-SNHG1, and miR-216b-5p was verified using RIP and luciferase reporter assays. Rescue experiments were undertaken to examine the influence of the lnc-SNHG1/miR-216b-5p/PAK2 axis on the development of paclitaxel resistance in A2780/Taxol cells. RESULTS: The bioinformatics analysis indicated a notable increase in PAK2 expression in ovarian malignant tumors compared to adjacent tissues, particularly in patients with stage III-IV disease compared to those with stage I-II disease (P = 0.0056). Elevated levels of PAK2 were linked to reduced OS in ovarian cancer patients, although no significant association was observed with DFS. Immunohistochemistry findings further supported these results, showing positive PAK2 protein expression in chemo-resistant serous EOC tissues, predominantly localized in the cytoplasm, which correlated with poorer OS and DFS outcomes. In vitro experiments demonstrated that the downregulation of PAK2 in A2780/Taxol cells led to a reduction in colony formation, an increase in apoptosis, and a diminished capacity for cell invasion. Subsequent analysis confirmed that lnc-SNHG1 functions as a competitive endogenous RNA (ceRNA) by interacting with miR-216b-5p and regulating PAK2 expression. Rescue experiments demonstrated that lnc-SNHG1 induces resistance to paclitaxel in A2780/Taxol cells by modulating the miR-216b-5p/PAK2 axis. CONCLUSIONS: PAK2 shows promise as a predictor of chemotherapy resistance and poor outcomes in ovarian cancer, indicating its potential use as a treatment target to overcome this resistance.


Subject(s)
Carcinoma, Ovarian Epithelial , Drug Resistance, Neoplasm , Gene Expression Regulation, Neoplastic , MicroRNAs , Ovarian Neoplasms , Paclitaxel , p21-Activated Kinases , Humans , p21-Activated Kinases/metabolism , p21-Activated Kinases/genetics , Female , Drug Resistance, Neoplasm/genetics , Paclitaxel/pharmacology , Paclitaxel/therapeutic use , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Ovarian Neoplasms/mortality , Prognosis , Carcinoma, Ovarian Epithelial/drug therapy , Carcinoma, Ovarian Epithelial/genetics , Carcinoma, Ovarian Epithelial/metabolism , Carcinoma, Ovarian Epithelial/pathology , Carcinoma, Ovarian Epithelial/mortality , MicroRNAs/genetics , MicroRNAs/metabolism , Cell Line, Tumor , Middle Aged , Cell Proliferation , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Apoptosis/drug effects , Up-Regulation
5.
Int J Mol Sci ; 25(18)2024 Sep 21.
Article in English | MEDLINE | ID: mdl-39337621

ABSTRACT

The serine/threonine kinase PAK4 plays a crucial role in regulating cell proliferation, survival, migration, and invasion. Overexpression of PAK4 correlates with poor prognosis in some cancers. KPT-9274, a PAK4 inhibitor, significantly reduces the growth of triple-negative breast cancer cells and mammary tumors in mouse models, and it also inhibits the growth of several other types of cancer cells. Interestingly, although it was first identified as a PAK4 inhibitor, KPT-9274 was also found to inhibit the enzyme NAMPT (nicotinamide phosphoribosyltransferase), which is crucial for NAD (nicotinamide adenine dinucleotide) synthesis and vital for cellular energy and growth. These results made us question whether growth inhibition in response to KPT-9274 was due to PAK4 inhibition, NAMPT inhibition, or both. To address this, we tested several other PAK4 inhibitors that also inhibit cell growth, to determine whether they also inhibit NAMPT activity. Our findings confirm that multiple PAK4 inhibitors also inhibit NAMPT activity. This was assessed both in cell-free assays and in a breast cancer cell line. Molecular docking studies were also used to help us better understand the mechanism by which PAK4 inhibitors block PAK4 and NAMPT activity, and we identified specific residues on the PAK4 inhibitors that interact with NAMPT and PAK4. Our results suggest that PAK4 inhibitors may have a more complex mechanism of action than previously understood, necessitating further exploration of how they influence cancer cell growth.


Subject(s)
Cytokines , Molecular Docking Simulation , Nicotinamide Phosphoribosyltransferase , p21-Activated Kinases , Nicotinamide Phosphoribosyltransferase/antagonists & inhibitors , Nicotinamide Phosphoribosyltransferase/metabolism , p21-Activated Kinases/antagonists & inhibitors , p21-Activated Kinases/metabolism , Humans , Cell Line, Tumor , Cytokines/metabolism , Cell Proliferation/drug effects , Acrylamides/pharmacology , Protein Kinase Inhibitors/pharmacology , Female , Benzeneacetamides/pharmacology , Aminopyridines
6.
J Physiol ; 602(19): 5061-5081, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39231098

ABSTRACT

Hyperreflexia associated with spasticity is a prevalent neurological condition characterized by excessive and exaggerated reflex responses to stimuli. Hyperreflexia can be caused by several diseases including multiple sclerosis, stroke and spinal cord injury (SCI). Although we have previously identified the contribution of the RAC1-PAK1 pathway underlying spinal hyperreflexia with SCI-induced spasticity, a feasible druggable target has not been validated. To assess the utility of targeting PAK1 to attenuate H-reflex hyperexcitability, we administered Romidepsin, a clinically available PAK1 inhibitor, in Thy1-YFP reporter mice. We performed longitudinal EMG studies with a study design that allowed us to assess pathological H-reflex changes and drug intervention effects over time, before and after contusive SCI. As expected, our results show a significant loss of rate-dependent depression - an indication of hyperreflexia and spasticity - 1 month following SCI as compared with baseline, uninjured controls (or before injury). Romidepsin treatment reduced signs of hyperreflexia in comparison with control cohorts and in pre- and post-drug intervention in SCI animals. Neuroanatomical study further confirmed drug response, as romidepsin treatment also reduced the presence of SCI-induced dendritic spine dysgenesis on α-motor neurons. Taken together, our findings extend previous work demonstrating the utility of targeting PAK1 activity in SCI-induced spasticity and support the novel use of romidepsin as an effective tool for managing spasticity. KEY POINTS: PAK1 plays a role in contributing to the development of spinal cord injury (SCI)-induced spasticity by contributing to dendritic spine dysgenesis. In this study, we explored the preclinical utility of inhibiting PAK1 to reduce spasticity and dendritic spine dysgenesis in an SCI mouse model. Romidepsin is a PAK1 inhibitor approved in the US in 2009 for the treatment of cutaneous T-cell lymphoma. Here we show that romidepsin treatment after SCI reduced SCI-induced H-reflex hyperexcitability and abnormal α-motor neuron spine morphology. This study provides compelling evidence that romidepsin may be a promising therapeutic approach for attenuating SCI-induced spasticity.


Subject(s)
Depsipeptides , H-Reflex , Spinal Cord Injuries , p21-Activated Kinases , Animals , Spinal Cord Injuries/drug therapy , Spinal Cord Injuries/complications , Spinal Cord Injuries/physiopathology , p21-Activated Kinases/antagonists & inhibitors , p21-Activated Kinases/metabolism , Depsipeptides/pharmacology , Mice , H-Reflex/drug effects , Female , Muscle Spasticity/drug therapy , Muscle Spasticity/etiology , Muscle Spasticity/physiopathology , Mice, Inbred C57BL , Motor Neurons/drug effects , Motor Neurons/physiology , Male
7.
CNS Neurosci Ther ; 30(9): e70055, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39328024

ABSTRACT

AIMS: Physical exercise (PE) can accelerate post-stroke recovery. This study investigated contributions of circRNAs to PE-induced improvements in post-stroke neurological function. METHODS: Rats subjected to transient middle cerebral artery occlusion were left sedentary or provided running-wheel access for 4 weeks during recovery. CircRNAs from peri-infarct cortex were identified by high-throughput sequencing, and interactions with miRNAs by immunoprecipitation, fluorescence in suit hybridization, and dual-luciferase reporter assays. In vivo circRNA knockdown was achieved using shRNA-AAVs and in vitro overexpression by plasmid transfection. Transmission electron microscopy, western blotting, and TUNEL assays were conducted to explore circRNA contributions to endoplasmic reticulum (ER) stress and neuronal apoptosis. CircRNA levels were measured in plasma from stroke patients by qRT-PCR and associations with neurological scores assessed by Pearson's correlation analysis. RESULTS: PE upregulated circAnks1b, reduced infarct volume, and mitigated neurological dysfunction, while circAnks1b knockdown exacerbated neurological dysfunction and increased infarct size despite PE. CircAnks1b sponged miR-130b-5p, thereby disinhibiting Pak2 expression. Conversely, Pak2 downregulation disrupted PE-mediated protective ER stress, leading to reduced IRE1/XBP1 and heightened apoptosis. Plasma circAnks1b was higher in stroke patients receiving PE than sedentary patients and correlated negatively with neurological scores. CONCLUSIONS: CircAnks1b upregulation may be an effective therapeutic strategy for post-stroke recovery.


Subject(s)
Endoplasmic Reticulum Stress , Ischemic Stroke , MicroRNAs , Physical Conditioning, Animal , RNA, Circular , Signal Transduction , Animals , Humans , Male , Middle Aged , Rats , Apoptosis , Disease Models, Animal , Endoplasmic Reticulum Stress/physiology , Endoplasmic Reticulum Stress/genetics , Infarction, Middle Cerebral Artery/metabolism , Infarction, Middle Cerebral Artery/genetics , Ischemic Stroke/metabolism , Ischemic Stroke/genetics , Ischemic Stroke/pathology , MicroRNAs/metabolism , MicroRNAs/genetics , p21-Activated Kinases/metabolism , p21-Activated Kinases/genetics , Physical Conditioning, Animal/physiology , Physical Conditioning, Animal/methods , Rats, Sprague-Dawley , RNA, Circular/metabolism , RNA, Circular/genetics , Signal Transduction/physiology , Up-Regulation/physiology
8.
Bioorg Med Chem ; 112: 117896, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-39214014

ABSTRACT

Triple-negative breast cancer is one of the most malignant subtypes in clinical practice, and it is urgent to find new therapies. The p21-activated kinase I (PAK1) has been considered to be an attractive therapeutic target for TNBC. In this study, we designed and synthesized a series of novel PROTAC PAK1 degraders by conjugating VHL or CRBN ligase ligands to PAK1 inhibitors which are connected by alkyl chains or PEG chains. The most promising compound, 19s, can significantly degrade PAK1 protein at concentrations as low as 0.1 µM, and achieves potent anti-proliferative activity with an IC50 value of 1.27 µM in MDA-MB-231 cells. Additionally, 19s exhibits potent anti-migration activity in vitro and induces rapid tumor regression in vivo. Collectively, these findings document that 19s is a potent and novel PAK1 degrader with promising potential for TNBC treatment.


Subject(s)
Antineoplastic Agents , Cell Proliferation , Drug Design , Triple Negative Breast Neoplasms , p21-Activated Kinases , p21-Activated Kinases/antagonists & inhibitors , p21-Activated Kinases/metabolism , Humans , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/pathology , Triple Negative Breast Neoplasms/metabolism , Cell Proliferation/drug effects , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Female , Structure-Activity Relationship , Animals , Drug Screening Assays, Antitumor , Cell Line, Tumor , Molecular Structure , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Dose-Response Relationship, Drug , Mice , Cell Movement/drug effects , Mice, Nude
9.
Nat Commun ; 15(1): 6858, 2024 Aug 10.
Article in English | MEDLINE | ID: mdl-39127697

ABSTRACT

Our recent studies have identified p21-activated kinase 4 (PAK4) as a key regulator of lipid catabolism in the liver and adipose tissue, but its role in glucose homeostasis in skeletal muscle remains to be explored. In this study, we find that PAK4 levels are highly upregulated in the skeletal muscles of diabetic humans and mice. Skeletal muscle-specific Pak4 ablation or administering the PAK4 inhibitor in diet-induced obese mice retains insulin sensitivity, accompanied by AMPK activation and GLUT4 upregulation. We demonstrate that PAK4 promotes insulin resistance by phosphorylating AMPKα2 at Ser491, thereby inhibiting AMPK activity. We additionally show that skeletal muscle-specific expression of a phospho-mimetic mutant AMPKα2S491D impairs glucose tolerance, while the phospho-inactive mutant AMPKα2S491A improves it. In summary, our findings suggest that targeting skeletal muscle PAK4 may offer a therapeutic avenue for type 2 diabetes.


Subject(s)
AMP-Activated Protein Kinases , Diabetes Mellitus, Type 2 , Glucose , Insulin Resistance , Muscle, Skeletal , p21-Activated Kinases , Animals , p21-Activated Kinases/metabolism , p21-Activated Kinases/genetics , Glucose/metabolism , Phosphorylation , Muscle, Skeletal/metabolism , AMP-Activated Protein Kinases/metabolism , Humans , Mice , Diabetes Mellitus, Type 2/metabolism , Male , Glucose Transporter Type 4/metabolism , Glucose Transporter Type 4/genetics , Mice, Inbred C57BL , Mice, Knockout , Obesity/metabolism
10.
J Exp Med ; 221(10)2024 Oct 07.
Article in English | MEDLINE | ID: mdl-39212656

ABSTRACT

A growing number of patients presenting severe combined immunodeficiencies attributed to monoallelic RAC2 variants have been identified. The expression of the RHO GTPase RAC2 is restricted to the hematopoietic lineage. RAC2 variants have been described to cause immunodeficiencies associated with high frequency of infection, leukopenia, and autoinflammatory features. Here, we show that specific RAC2 activating mutations induce the NLRP3 inflammasome activation leading to the secretion of IL-1ß and IL-18 from macrophages. This activation depends on the activation state of the RAC2 variant and is mediated by the downstream kinase PAK1. Inhibiting the RAC2-PAK1-NLRP3 inflammasome pathway might be considered as a potential treatment for these patients.


Subject(s)
Gain of Function Mutation , Inflammasomes , Interleukin-1beta , Macrophages , NLR Family, Pyrin Domain-Containing 3 Protein , RAC2 GTP-Binding Protein , p21-Activated Kinases , rac GTP-Binding Proteins , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Inflammasomes/metabolism , Inflammasomes/immunology , Humans , Macrophages/immunology , Macrophages/metabolism , rac GTP-Binding Proteins/genetics , rac GTP-Binding Proteins/metabolism , Interleukin-1beta/metabolism , Interleukin-1beta/genetics , Animals , p21-Activated Kinases/genetics , p21-Activated Kinases/metabolism , Mice , Interleukin-18/genetics , Interleukin-18/metabolism , Signal Transduction
11.
Cell Mol Biol Lett ; 29(1): 95, 2024 Jul 02.
Article in English | MEDLINE | ID: mdl-38956466

ABSTRACT

BACKGROUND: An increasing number of studies have demonstrated the association of circular RNAs (circRNAs) with the pathological processes of various diseases and their involvement in the onset and progression of multiple cancers. Nevertheless, the functional roles and underlying mechanisms of circRNAs in the autophagy regulation of gastric cancer (GC) have not been fully elucidated. METHODS: We used transmission electron microscopy and the mRFP-GFP-LC3 dual fluorescent autophagy indicator to investigate autophagy regulation. The cell counting kit-8 assay, colony formation assay, 5-ethynyl-2'-deoxyuridine incorporation assay, Transwell assay, and Western blot assay were conducted to confirm circPTPN22's influence on GC progression. Dual luciferase reporter assays validated the binding between circPTPN22 and miR-6788-5p, as well as miR-6788-5p and p21-activated kinase-1 (PAK1). Functional rescue experiments assessed whether circPTPN22 modulates PAK1 expression by competitively binding miR-6788-5p, affecting autophagy and other biological processes in GC cells. We investigated the impact of circPTPN22 on in vivo GC tumors using a nude mouse xenograft model. Bioinformatics tools predicted upstream regulatory transcription factors and binding proteins of circPTPN22, while chromatin immunoprecipitation and ribonucleoprotein immunoprecipitation assays confirmed the binding status. RESULTS: Upregulation of circPTPN22 in GC has been shown to inhibit autophagy and promote cell proliferation, migration, and invasion. Mechanistically, circPTPN22 directly binds to miR-6788-5p, subsequently regulating the expression of PAK1, which activates protein kinase B (Akt) and extracellular signal-regulated kinase (Erk) phosphorylation. This modulation ultimately affects autophagy levels in GC cells. Additionally, runt-related transcription factor 1 (RUNX1) negatively regulates circPTPN22 expression, while RNA-binding proteins such as FUS (fused in sarcoma) and ELAVL1 (recombinant ELAV-like protein 1) positively regulate its expression. Inhibition of the autophagy pathway can increase FUS expression, further upregulating circPTPN22 in GC cells, thereby exacerbating the progression of GC. CONCLUSION: Under the regulation of the transcription factor RUNX1 and RNA-binding proteins FUS and ELAVL1, circPTPN22 activates the phosphorylation of Akt and Erk through the miR-6788-5p/PAK1 axis, thereby modulating autophagy in GC cells. Inhibition of autophagy increases FUS, which in turn upregulates circPTPN22, forming a positive feedback loop that ultimately accelerates the progression of GC.


Subject(s)
Autophagy , Cell Movement , Cell Proliferation , Core Binding Factor Alpha 2 Subunit , ELAV-Like Protein 1 , MicroRNAs , RNA, Circular , RNA-Binding Protein FUS , Stomach Neoplasms , p21-Activated Kinases , Humans , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Stomach Neoplasms/metabolism , RNA, Circular/genetics , RNA, Circular/metabolism , Autophagy/genetics , MicroRNAs/genetics , MicroRNAs/metabolism , p21-Activated Kinases/metabolism , p21-Activated Kinases/genetics , Cell Proliferation/genetics , RNA-Binding Protein FUS/metabolism , RNA-Binding Protein FUS/genetics , Cell Movement/genetics , Cell Line, Tumor , Animals , ELAV-Like Protein 1/metabolism , ELAV-Like Protein 1/genetics , Core Binding Factor Alpha 2 Subunit/metabolism , Core Binding Factor Alpha 2 Subunit/genetics , Gene Expression Regulation, Neoplastic , Mice, Nude , Mice , Neoplasm Invasiveness , Mice, Inbred BALB C
12.
Int J Biol Sci ; 20(9): 3269-3284, 2024.
Article in English | MEDLINE | ID: mdl-38993552

ABSTRACT

Background: Lenvatinib is the most common multitarget receptor tyrosine kinase inhibitor for the treatment of advanced hepatocellular carcinoma (HCC). Acquired resistance to lenvatinib is one of the major factors leading to the failure of HCC treatment, but the underlying mechanism has not been fully characterized. Methods: We established lenvatinib-resistant cell lines, cell-derived xenografts (CDXs) and patient-derived xenografts (PDXs) and obtained lenvatinib-resistant HCC tumor tissues for further study. Results: We found that ubiquitin-specific protease 14 (USP14) was significantly increased in lenvatinib-resistant HCC cells and tumors. Silencing USP14 significantly attenuated lenvatinib resistance in vitro and in vivo. Mechanistically, USP14 directly interacts with and stabilizes calcium- and integrin-binding protein 1 (CIB1) by reversing K48-linked proteolytic ubiquitination at K24, thus facilitating the P21-activated kinase 1 (PAK1)-ERK1/2 signaling axis. Moreover, in vivo adeno-associated virus 9 mediated transduction of CIB1 promoted lenvatinib resistance in PDXs, whereas CIB1 knockdown resensitized the response of PDXs to lenvatinib. Conclusions: These findings provide new insights into the role of CIB1/PAK1-ERK1/2 signaling in lenvatinib resistance in HCC. Targeting CIB1 and its pathways may be a novel pharmaceutical intervention for the treatment of lenvatinib-resistant HCC.


Subject(s)
Carcinoma, Hepatocellular , Drug Resistance, Neoplasm , Liver Neoplasms , Phenylurea Compounds , Quinolines , Ubiquitin Thiolesterase , p21-Activated Kinases , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/drug therapy , Humans , Quinolines/pharmacology , Quinolines/therapeutic use , Liver Neoplasms/metabolism , Liver Neoplasms/drug therapy , Ubiquitin Thiolesterase/metabolism , Ubiquitin Thiolesterase/genetics , Phenylurea Compounds/pharmacology , Phenylurea Compounds/therapeutic use , Animals , p21-Activated Kinases/metabolism , p21-Activated Kinases/genetics , Mice , Cell Line, Tumor , MAP Kinase Signaling System , Mice, Nude , Ubiquitination
13.
Braz J Med Biol Res ; 57: e13357, 2024.
Article in English | MEDLINE | ID: mdl-38958364

ABSTRACT

The overexpression of P-glycoprotein (P-gp/ABCB1) is a leading cause of multidrug resistance (MDR). Hence, it is crucial to discover effective pharmaceuticals that counteract ABCB1-mediated multidrug resistance. FRAX486 is a p21-activated kinase (PAK) inhibitor. The objective of this study was to investigate whether FRAX486 can reverse ABCB1-mediated multidrug resistance, while also exploring its mechanism of action. The CCK8 assay demonstrated that FRAX486 significantly reversed ABCB1-mediated multidrug resistance. Furthermore, western blotting and immunofluorescence experiments revealed that FRAX486 had no impact on expression level and intracellular localization of ABCB1. Notably, FRAX486 was found to enhance intracellular drug accumulation and reduce efflux, resulting in the reversal of multidrug resistance. Docking analysis also indicated a strong affinity between FRAX486 and ABCB1. This study highlights the ability of FRAX486 to reverse ABCB1-mediated multidrug resistance and provides valuable insights for its clinical application.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B , Breast Neoplasms , Drug Resistance, Multiple , Drug Resistance, Neoplasm , Humans , Drug Resistance, Neoplasm/drug effects , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , ATP Binding Cassette Transporter, Subfamily B/metabolism , ATP Binding Cassette Transporter, Subfamily B/antagonists & inhibitors , Drug Resistance, Multiple/drug effects , Female , p21-Activated Kinases/antagonists & inhibitors , p21-Activated Kinases/metabolism , Cell Line, Tumor , Blotting, Western
14.
PLoS One ; 19(7): e0305121, 2024.
Article in English | MEDLINE | ID: mdl-39083549

ABSTRACT

Neurofibromatosis type 2 is a genetic disorder that results in the formation and progressive growth of schwannomas, ependymomas, and/or meningiomas. The NF2 gene encodes the Merlin protein, which links cell cortical elements to the actin cytoskeleton and regulates a number of key enzymes including Group I p21-activated kinases (PAKs), the Hippo-pathway kinase LATS, and mTORC. While PAK1 and PAK2 directly bind Merlin and transmit proliferation and survival signals when Merlin is mutated or absent, inhibition of Group 1 PAKs alone has not proven sufficient to completely stop the growth of NF2-deficient meningiomas or schwannomas in vivo, suggesting the need for a second pathway inhibitor. As the Hippo pathway is also activated in NF2-deficient cells, several inhibitors of the Hippo pathway have recently been developed in the form of YAP-TEAD binding inhibitors. These inhibitors prevent activation of pro-proliferation and anti-apoptotic Hippo pathway effectors. In this study, we show that PAK inhibition slows cell proliferation while TEAD inhibition promotes apoptotic cell death. Finally, we demonstrate the efficacy of PAK and TEAD inhibitor combinations in several NF2-deficient Schwannoma cell lines.


Subject(s)
Cell Proliferation , Hippo Signaling Pathway , Neurilemmoma , Neurofibromin 2 , Protein Serine-Threonine Kinases , Signal Transduction , Transcription Factors , p21-Activated Kinases , Humans , Neurilemmoma/metabolism , Neurilemmoma/genetics , Neurilemmoma/pathology , p21-Activated Kinases/metabolism , p21-Activated Kinases/antagonists & inhibitors , p21-Activated Kinases/genetics , Cell Proliferation/drug effects , Neurofibromin 2/genetics , Neurofibromin 2/deficiency , Neurofibromin 2/metabolism , Signal Transduction/drug effects , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/antagonists & inhibitors , Cell Line, Tumor , Transcription Factors/metabolism , Transcription Factors/genetics , Apoptosis/drug effects , Drug Synergism , Neurofibromatosis 2/metabolism , Neurofibromatosis 2/genetics , YAP-Signaling Proteins/metabolism , YAP-Signaling Proteins/genetics , Protein Kinase Inhibitors/pharmacology , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics
15.
J Cell Biol ; 223(10)2024 Sep 02.
Article in English | MEDLINE | ID: mdl-39012625

ABSTRACT

The GTPase Cdc42 regulates polarized growth in most eukaryotes. In the bipolar yeast Schizosaccharomyces pombe, Cdc42 activation cycles periodically at sites of polarized growth. These periodic cycles are caused by alternating positive feedback and time-delayed negative feedback loops. At each polarized end, negative feedback is established when active Cdc42 recruits the Pak1 kinase to prevent further Cdc42 activation. It is unclear how Cdc42 activation returns to each end after Pak1-dependent negative feedback. We find that disrupting branched actin-mediated endocytosis disables Cdc42 reactivation at the cell ends. Using experimental and mathematical approaches, we show that endocytosis-dependent Pak1 removal from the cell ends allows the Cdc42 activator Scd1 to return to that end to enable reactivation of Cdc42. Moreover, we show that Pak1 elicits its own removal via activation of endocytosis. These findings provide a deeper insight into the self-organization of Cdc42 regulation and reveal previously unknown feedback with endocytosis in the establishment of cell polarity.


Subject(s)
Actin-Related Protein 2-3 Complex , Cell Polarity , Endocytosis , Feedback, Physiological , Schizosaccharomyces pombe Proteins , Schizosaccharomyces , cdc42 GTP-Binding Protein , p21-Activated Kinases , Schizosaccharomyces/metabolism , Schizosaccharomyces/genetics , Schizosaccharomyces pombe Proteins/metabolism , Schizosaccharomyces pombe Proteins/genetics , p21-Activated Kinases/metabolism , p21-Activated Kinases/genetics , Actin-Related Protein 2-3 Complex/metabolism , Actin-Related Protein 2-3 Complex/genetics , cdc42 GTP-Binding Protein/metabolism , cdc42 GTP-Binding Protein/genetics , Actins/metabolism
16.
Nagoya J Med Sci ; 86(2): 216-222, 2024 May.
Article in English | MEDLINE | ID: mdl-38962417

ABSTRACT

Psychiatric disorders are highly inheritable, and most psychiatric disorders exhibit genetic overlap. Recent studies associated the 3q29 recurrent deletion with schizophrenia (SCZ) and autism spectrum disorder (ASD). In this study, we investigated the association of genes in the 3q29 region with SCZ and ASD. TM4SF19 and PAK2 were chosen as candidate genes for this study based on evidence from previous research. We sequenced TM4SF19 and PAK2 in 437 SCZ cases, 187 ASD cases and 524 controls in the Japanese population. Through targeted sequencing, we identified 6 missense variants among the cases (ASD & SCZ), 3 missense variants among controls, and 1 variant common to both cases and controls; however, no loss-of-function variants were identified. Fisher's exact test showed a significant association of variants in TM4SF19 among cases (p=0.0160). These results suggest TM4SF19 variants affect the etiology of SCZ and ASD in the Japanese population. Further research examining 3q29 region genes and their association with SCZ and ASD is thus needed.


Subject(s)
Autism Spectrum Disorder , Genetic Predisposition to Disease , Schizophrenia , Adult , Female , Humans , Male , Autism Spectrum Disorder/genetics , Case-Control Studies , Chromosomes, Human, Pair 3/genetics , East Asian People/genetics , Genetic Association Studies , Japan , Mutation, Missense , p21-Activated Kinases/genetics , Schizophrenia/genetics
17.
BMC Biol ; 22(1): 139, 2024 Jun 24.
Article in English | MEDLINE | ID: mdl-38915055

ABSTRACT

BACKGROUND: The intermediate filament protein vimentin is widely recognized as a molecular marker of epithelial-to-mesenchymal transition. Although vimentin expression is strongly associated with cancer metastatic potential, the exact role of vimentin in cancer metastasis and the underlying mechanism of its pro-metastatic functions remain unclear. RESULTS: This study revealed that vimentin can enhance integrin ß1 surface expression and induce integrin-dependent clustering of cells, shielding them against anoikis cell death. The increased integrin ß1 surface expression in suspended cells was caused by vimentin-mediated protection of the internal integrin ß1 pool against lysosomal degradation. Additionally, cell detachment was found to induce vimentin Ser38 phosphorylation, allowing the translocation of internal integrin ß1 to the plasma membrane. Furthermore, the use of an inhibitor of p21-activated kinase PAK1, one of the kinases responsible for vimentin Ser38 phosphorylation, significantly reduced cancer metastasis in animal models. CONCLUSIONS: These findings suggest that vimentin can act as an integrin buffer, storing internalized integrin ß1 and releasing it when needed. Overall, this study provides insights regarding the strong correlation between vimentin expression and cancer metastasis and a basis for blocking metastasis using this novel therapeutic mechanism.


Subject(s)
Anoikis , Integrin beta1 , Vimentin , Vimentin/metabolism , Vimentin/genetics , Integrin beta1/metabolism , Integrin beta1/genetics , Humans , Animals , Cell Survival , Mice , Cell Line, Tumor , Phosphorylation , p21-Activated Kinases/metabolism , p21-Activated Kinases/genetics
18.
Sci Rep ; 14(1): 14092, 2024 06 18.
Article in English | MEDLINE | ID: mdl-38890401

ABSTRACT

Oral squamous cell carcinoma (OSCC) is one of the most common malignant tumours, warranting novel treatments. Here, we examined the therapeutic efficacy of inhibiting p21 activated kinase 4 (PAK4) in OSCC and determined its immunomodulatory effect by focusing on the enhancement of anti-tumour effects. We examined PAK4 expression in OSCC cells and human clinical samples and analysed the proliferation and apoptosis of OSCC cells following PAK4 inhibition in vitro. We also investigated the effects of in vivo administration of a PAK4 inhibitor on immune cell distribution and T-cell immune responses in OSCC tumour-bearing mice. PAK4 was detected in all OSCC cells and OSCC tissue samples. PAK4 inhibitor reduced the proliferation of OSCC cells and induced apoptosis. PAK4 inhibitor significantly attenuated tumour growth in mouse and was associated with increased proportions of IFN-γ-producing CD8+ T-cells. Furthermore, PAK4 inhibitor increased the number of dendritic cells (DCs) and up-regulated the surface expression of various lymphocyte co-stimulatory molecules, including MHC-class I molecules, CD80, CD83, CD86, and CD40. These DCs augmented CD8+ T-cell activation upon co-culture. Our results suggest that PAK4 inhibition in OSCC can have direct anti-tumour and immunomodulatory effects, which might benefit the treatment of this malignancy.


Subject(s)
Carcinoma, Squamous Cell , Cell Proliferation , Immunomodulation , Mouth Neoplasms , p21-Activated Kinases , p21-Activated Kinases/metabolism , p21-Activated Kinases/antagonists & inhibitors , Mouth Neoplasms/drug therapy , Mouth Neoplasms/immunology , Mouth Neoplasms/pathology , Mouth Neoplasms/metabolism , Humans , Animals , Mice , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/immunology , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/metabolism , Immunomodulation/drug effects , Cell Proliferation/drug effects , Cell Line, Tumor , Apoptosis/drug effects , Dendritic Cells/immunology , Dendritic Cells/drug effects , Dendritic Cells/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/drug effects , Female , Male
19.
Reprod Biol Endocrinol ; 22(1): 74, 2024 Jun 25.
Article in English | MEDLINE | ID: mdl-38918809

ABSTRACT

BACKGROUND: Erectile dysfunction (ED) is a common male sexual dysfunction, with an increasing incidence, and the current treatment is often ineffective. METHODS: Vascular endothelial growth factor (VEGFA) was used to treat bone marrow-derived mesenchymal stem cells (BM-MSCs), and their cell migration rates were determined by Transwell assays. The expression of the von Willebrand Factor (vWF)VE-cadherin, and endothelial nitric oxide synthase(eNOS) endothelial markers was determined by qRT‒PCR and Western blot analyses. The MALAT1-induced differentiation of BM-MCs to ECs via the CDC42/PAK1/paxillin pathway was explored by transfecting VEGFA-induced BM-MSC with si-MALAT1 and overexpressing CDC42 and PAK1. The binding capacity between CDC42, PAK1, and paxillin in VEGFA-treated and non-VEGFA-treated BM-MSCs was examined by protein immunoprecipitation. MiR-206 was overexpressed in VEGFA-induced BM-MSC, and the binding sites of MALAT1, miR-206, and CDC42 were identified using a luciferase assay. Sixty male Sprague‒Dawley rats were divided into six groups (n = 10/group). DMED modelling was demonstrated by APO experiments and was assessed by measuring blood glucose levels. Erectile function was assessed by measuring the intracavernosa pressure (ICP) and mean arterial pressure (MAP). Penile erectile tissue was analysed by qRT‒PCR, Western blot analysis, and immunohistochemical staining. RESULTS: MALAT1 under VEGFA treatment conditions regulates the differentiation of BM-MSCs into ECs by modulating the CDC42/PAK1/paxillin axis. In vitro experiments demonstrated that interference with CDC42 and MALAT1 expression inhibited the differentiation of BM-MSCs to ECs. CDC42 binds to PAK1, and PAK1 binds to paxillin. In addition, CDC42 in the VEGFA group had a greater ability to bind to PAK1, whereas PAK1 in the VEGFA group had a greater ability to bind to paxillin. Overexpression of miR-206 in VEGFA-induced BM-MSCs demonstrated that MALAT1 competes with the CDC42 3'-UTR for binding to miR-206, which in turn is involved in the differentiation of BM-MSCs to ECs. Compared to the DMED model group, the ICP/MAP ratio was significantly greater in the three BM-MSCs treatment groups. CONCLUSIONS: MALAT1 facilitates BM-MSC differentiation into ECs by regulating the miR-206/CDC42/PAK1/paxillin axis to improve ED. The present findings revealed the vital role of MALAT1 in the repair of BM-MSCs for erectile function and provided new mechanistic insights into the BM-MSC-mediated repair of DMED.


Subject(s)
Cell Differentiation , Erectile Dysfunction , Mesenchymal Stem Cells , MicroRNAs , Paxillin , RNA, Long Noncoding , Rats, Sprague-Dawley , Signal Transduction , cdc42 GTP-Binding Protein , p21-Activated Kinases , Male , Animals , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Cell Differentiation/genetics , cdc42 GTP-Binding Protein/metabolism , cdc42 GTP-Binding Protein/genetics , Rats , p21-Activated Kinases/genetics , p21-Activated Kinases/metabolism , Mesenchymal Stem Cells/metabolism , Erectile Dysfunction/therapy , Erectile Dysfunction/genetics , Erectile Dysfunction/metabolism , Paxillin/metabolism , Paxillin/genetics , Endothelial Cells/metabolism , Cells, Cultured , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor A/genetics
20.
Clin Genet ; 106(4): 518-524, 2024 Oct.
Article in English | MEDLINE | ID: mdl-38894571

ABSTRACT

The p21-activated kinase (PAK) family of proteins regulates various processes requiring dynamic cytoskeleton organization such as cell adhesion, migration, proliferation, and apoptosis. Among the six members of the protein family, PAK2 is specifically involved in apoptosis, angiogenesis, or the development of endothelial cells. We report a novel de novo heterozygous missense PAK2 variant, p.(Thr406Met), found in a newborn with clinical manifestations of Knobloch syndrome. In vitro experiments indicated that this and another reported variant, p.(Asp425Asn), result in substantially impaired protein kinase activity. Similar findings were described previously for the PAK2 p.(Glu435Lys) variant found in two siblings with proposed Knobloch syndrome type 2 (KNO2). These new variants support the association of PAK2 kinase deficiency with a second, autosomal dominant form of Knobloch syndrome: KNO2.


Subject(s)
p21-Activated Kinases , Humans , p21-Activated Kinases/genetics , Retinal Detachment/genetics , Retinal Detachment/pathology , Retinal Detachment/congenital , Retinal Degeneration/genetics , Retinal Degeneration/pathology , Male , Infant, Newborn , Female , Mutation, Missense/genetics , Encephalocele
SELECTION OF CITATIONS
SEARCH DETAIL