Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.979
Filter
1.
J Clin Invest ; 134(19)2024 Oct 01.
Article in English | MEDLINE | ID: mdl-39352385

ABSTRACT

Severe viral pneumonia can induce rapid expansion of KRT5+ basal-like cells in small airways and alveoli; this forms a scar-like structure that persists in the injured alveoli and impedes normal alveolar epithelium regeneration. In this study, we investigated the mechanism by which viral infection induced this remodeling response. Through comparing different lung-injury models, we demonstrated that infection induced strong IFN-γ signal-stimulated dysplastic KRT5+ cell formation. Inactivation of interferon receptor 1 (Ifngr1) reduced dysplastic cell formation, ameliorated lung fibrosis, and improved lung-function recovery. Mechanistically, IFN-γ regulated dysplastic cell formation via the focal adhesion kinase (FAK)/Yes-associated protein 1 (YAP) pathway. Inhibiting FAK/Src diminished IFN-γ-induced YAP nuclear translocation and dysplastic cell formation. Inhibiting YAP during viral infection prevented dysplastic cell formation, whereas inhibiting YAP in persistent KRT5+ cells led to their conversion into distal club cells. Importantly, human dysplastic cells exhibited elevated FAK and YAP activity, and IFN-γ treatment promoted the transformation of human alveolar progenitor cells into dysplastic cells. These findings uncover the role of infection-induced inflammatory response in alveolar remodeling and may provide potential therapeutic avenues for the treatment of alveolar remodeling in patients with severe viral pneumonia.


Subject(s)
Adaptor Proteins, Signal Transducing , Focal Adhesion Kinase 1 , Interferon-gamma , Pulmonary Alveoli , YAP-Signaling Proteins , YAP-Signaling Proteins/metabolism , Animals , Mice , Humans , Interferon-gamma/metabolism , Focal Adhesion Kinase 1/metabolism , Focal Adhesion Kinase 1/genetics , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , Pulmonary Alveoli/pathology , Pulmonary Alveoli/metabolism , Pulmonary Alveoli/virology , Transcription Factors/metabolism , Transcription Factors/genetics , Signal Transduction , Mice, Knockout , Inflammation/pathology , Inflammation/metabolism , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/genetics
2.
BMC Res Notes ; 17(1): 282, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-39354559

ABSTRACT

OBJECTIVE: In highly aggressive malignant cancers including breast cancer, vasculogenic mimicry (VM) is the potential of tumor cells to generate a vascular channel network for delivering blood to tumor cells. Detection of genes involved in this process is critical to designing targeted therapy against breast cancer metastasis. In this study, we evaluated the roles of FAK and ILK in the progression of VM in metastatic breast tumor cells. RESULTS: Primary (4T1T), and highly metastatic (4T1B and 4T1L) breast tumor cells were isolated from cancerous mice. The potential of cancer cells to organize themselves into vascular-like structures (VM) has been evaluated with in vitro assessment. The expression of ILK and FAK were examined using real-time polymerase chain reaction. We confirmed the high ability of metastatic tumor cells in vascular-like structure formation. In molecular analysis, our data showed that ILK and FAK expression was significantly elevated in metastatic breast tumor cells. These results indicated that the higher potential of metastatic tumor cells in vascular-like structure formation may be related to higher expression of ILK and FAK. Analysis of molecular features of metastatic tumor cells could be utilized to create a targeted therapeutic strategy against metastasis in breast cancer.


Subject(s)
Breast Neoplasms , Neoplasm Metastasis , Protein Serine-Threonine Kinases , Signal Transduction , Animals , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Female , Mice , Cell Line, Tumor , Breast Neoplasms/pathology , Breast Neoplasms/genetics , Breast Neoplasms/blood supply , Breast Neoplasms/metabolism , Neovascularization, Pathologic/pathology , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Mice, Inbred BALB C , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Focal Adhesion Kinase 1/metabolism , Focal Adhesion Kinase 1/genetics , Gene Expression Regulation, Neoplastic
3.
Cell Mol Life Sci ; 81(1): 421, 2024 Oct 05.
Article in English | MEDLINE | ID: mdl-39367995

ABSTRACT

Cullin-RING ubiquitin ligase 4 (CRL4) is closely correlated with the incidence and progression of ovarian cancer. DDB1- and CUL4-associated factor 13 (DCAF13), a substrate-recognition protein in the CRL4 E3 ubiquitin ligase complex, is involved in the occurrence and development of ovarian cancer. However, its precise function and the underlying molecular mechanism in this disease remain unclear. In this study, we confirmed that DCAF13 is highly expressed in human ovarian cancer and its expression is negatively correlated with the overall survival rate of patients with ovarian cancer. We then used CRISPR/Cas9 to knockout DCAF13 and found that its deletion significantly inhibited the proliferation, colony formation, and migration of human ovarian cancer cells. In addition, DCAF13 deficiency inhibited tumor proliferation in nude mice. Mechanistically, CRL4-DCAF13 targeted Fraser extracellular matrix complex subunit 1 (FRAS1) for polyubiquitination and proteasomal degradation. FRAS1 influenced the proliferation and migration of ovarian cancer cell through induction of the focal adhesion kinase (FAK) signaling pathway. These findings collectively show that DCAF13 is an important oncogene that promotes tumorigenesis in ovarian cancer cells by mediating FRAS1/FAK signaling. Our findings provide a foundation for the development of targeted therapeutics for ovarian cancer.


Subject(s)
Cell Movement , Cell Proliferation , Extracellular Matrix Proteins , Focal Adhesion Kinase 1 , Mice, Nude , Ovarian Neoplasms , RNA-Binding Proteins , Animals , Female , Humans , Mice , Cell Line, Tumor , Cell Movement/genetics , Disease Progression , Focal Adhesion Kinase 1/metabolism , Focal Adhesion Kinase 1/genetics , Gene Expression Regulation, Neoplastic , Mice, Inbred BALB C , Ovarian Neoplasms/pathology , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/genetics , Signal Transduction , Ubiquitination , RNA-Binding Proteins/metabolism , Extracellular Matrix Proteins/metabolism
4.
FASEB J ; 38(17): e70050, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39259535

ABSTRACT

Tendons enable locomotion by transmitting high tensile mechanical forces between muscle and bone via their dense extracellular matrix (ECM). The application of extrinsic mechanical stimuli via muscle contraction is necessary to regulate healthy tendon function. Specifically, applied physiological levels of mechanical loading elicit an anabolic tendon cell response, while decreased mechanical loading evokes a degradative tendon state. Although the tendon response to mechanical stimuli has implications in disease pathogenesis and clinical treatment strategies, the cell signaling mechanisms by which tendon cells sense and respond to mechanical stimuli within the native tendon ECM remain largely unknown. Therefore, we explored the role of cell-ECM adhesions in regulating tendon cell mechanotransduction by perturbing the genetic expression and signaling activity of focal adhesion kinase (FAK) through both in vitro and in vivo approaches. We determined that FAK regulates tendon cell spreading behavior and focal adhesion morphology, nuclear deformation in response to applied mechanical strain, and mechanosensitive gene expression. In addition, our data reveal that FAK signaling plays an essential role in in vivo tendon development and postnatal growth, as FAK-knockout mouse tendons demonstrated reduced tendon size, altered mechanical properties, differences in cellular composition, and reduced maturity of the deposited ECM. These data provide a foundational understanding of the role of FAK signaling as a critical regulator of in situ tendon cell mechanotransduction. Importantly, an increased understanding of tendon cell mechanotransductive mechanisms may inform clinical practice as well as lead to the discovery of diagnostic and/or therapeutic molecular targets.


Subject(s)
Mechanotransduction, Cellular , Mice, Knockout , Tendons , Animals , Male , Mice , Cells, Cultured , Extracellular Matrix/metabolism , Focal Adhesion Kinase 1/metabolism , Focal Adhesion Kinase 1/genetics , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Focal Adhesion Protein-Tyrosine Kinases/genetics , Focal Adhesions/metabolism , Mechanotransduction, Cellular/physiology , Mice, Inbred C57BL , Signal Transduction/physiology , Tendons/metabolism , Tendons/physiology , Tendons/cytology , Female
5.
Int J Biol Macromol ; 279(Pt 4): 135376, 2024 Nov.
Article in English | MEDLINE | ID: mdl-39244119

ABSTRACT

Adipocyte-secreted factors intricately regulate adipose tissue function, and the underlying molecular mechanisms are only partially understood. However, the function of PRELP, which is a key component of the extracellular matrix (ECM) in adipocytes, remains largely unknown. In this study, we demonstrate that PRELP was upregulated in both obese humans and mice, which exhibited a positive correlation with metabolic disorders. PRELP knockout could resist HFD-induced obesity and inhibit adipocyte differentiation. PRELP knockout improved glucose tolerance, insulin sensitivity and alleviated adipose tissue fibrosis. Mechanistically, PRELP was secreted into the ECM and bound to the extracellular domain of its receptor p75NTR in adipocytes, which further activated the FAK/MAPK (JNK, p38 MAPK, ERK1/2) signaling pathway, promoting adipocyte differentiation and exacerbating adipocyte fibrosis. Adipocyte PRELP plays a pivotal role in regulating obesity and adipose tissue fibrosis through an autocrine manner, and PRELP may be a therapeutic target for obesity and its related metabolic disorders.


Subject(s)
Adipocytes , Adipose Tissue , Fibrosis , MAP Kinase Signaling System , Animals , Mice , Humans , Adipose Tissue/metabolism , Adipocytes/metabolism , Cell Differentiation , Obesity/metabolism , Obesity/pathology , Receptors, Nerve Growth Factor/metabolism , Receptors, Nerve Growth Factor/genetics , Male , Mice, Knockout , Focal Adhesion Kinase 1/metabolism , Focal Adhesion Kinase 1/genetics , Protein Binding , Adipogenesis , Mice, Inbred C57BL , Nerve Tissue Proteins
6.
EMBO Mol Med ; 16(10): 2402-2426, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39271958

ABSTRACT

Mutated KRAS serves as the oncogenic driver in 30% of non-small cell lung cancers (NSCLCs) and is associated with metastatic and therapy-resistant tumors. Focal Adhesion Kinase (FAK) acts as a mediator in sustaining KRAS-driven lung tumors, and although FAK inhibitors are currently undergoing clinical development, clinical data indicated that their efficacy in producing long-term anti-tumor responses is limited. Here we revealed two FAK interactors, extracellular-signal-regulated kinase 5 (ERK5) and cyclin-dependent kinase 5 (CDK5), as key players underlying FAK-mediated maintenance of KRAS mutant NSCLC. Inhibition of ERK5 and CDK5 synergistically suppressed FAK function, decreased proliferation and induced apoptosis owing to exacerbated ROS-induced DNA damage. Accordingly, concomitant pharmacological inhibition of ERK5 and CDK5 in a mouse model of KrasG12D-driven lung adenocarcinoma suppressed tumor progression and promoted cancer cell death. Cancer cells resistant to FAK inhibitors showed enhanced ERK5-FAK signaling dampening DNA damage. Notably, ERK5 inhibition prevented the development of resistance to FAK inhibitors, significantly enhancing the efficacy of anti-tumor responses. Therefore, we propose ERK5 inhibition as a potential co-targeting strategy to counteract FAK inhibitor resistance in NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Focal Adhesion Kinase 1 , Lung Neoplasms , Mitogen-Activated Protein Kinase 7 , Proto-Oncogene Proteins p21(ras) , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Animals , Mitogen-Activated Protein Kinase 7/metabolism , Mitogen-Activated Protein Kinase 7/genetics , Mitogen-Activated Protein Kinase 7/antagonists & inhibitors , Humans , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Mice , Focal Adhesion Kinase 1/metabolism , Focal Adhesion Kinase 1/antagonists & inhibitors , Focal Adhesion Kinase 1/genetics , Drug Resistance, Neoplasm/genetics , Drug Resistance, Neoplasm/drug effects , Cell Line, Tumor , Cyclin-Dependent Kinase 5/metabolism , Cyclin-Dependent Kinase 5/antagonists & inhibitors , Cyclin-Dependent Kinase 5/genetics , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Apoptosis/drug effects , Mutation , Cell Proliferation/drug effects
7.
Neoplasia ; 57: 101059, 2024 Nov.
Article in English | MEDLINE | ID: mdl-39326322

ABSTRACT

BACKGROUND: Breast cancer is a malignancy with a generally poor prognosis. With the advancement of molecular research, we have gained deeper insights into the cellular processes that drive breast cancer development. However, the precise mechanisms remain elusive. RESULTS: Based on the CPTAC database, we found that NEDD9 expression is up-regulated in breast cancer tissues and is associated with poor prognosis in breast cancer patients. Functional experiments showed that NEDD9 promotes tumor growth and metastasis both in vitro and in vivo. Overexpression of NEDD9 disrupts mammary epithelial acinus formation and triggers epithelial-mesenchymal transition in breast cancer cells, effects that are reversed upon NEDD9 gene silencing. Mechanistically, NEDD9 upregulates its expression by inhibiting HDAC4 activity, leading to enhanced H3K9 acetylation of the NEDD9 gene promoter and activation of the FAK/NF-κB signaling pathway. Furthermore, NEDD9 overexpression promotes IL-6 secretion, which further drives breast cancer progression. Notably, NEDD9 activation fosters the pro-tumoral M2 macrophage polarization in the tumor microenvironment. NEDD9 stimulates IL-6 secretion, polarizes monocytes towards an M2-like phenotype, and enhances BC cell invasiveness. CONCLUSIONS: These findings suggest that NEDD9 upregulation plays a pivotal role in breast cancer metastasis and macrophage M2 polarization via the FAK/NF-κB signaling axis. Targeting NEDD9 may offer a promising therapeutic approach for breast cancer treatment.


Subject(s)
Adaptor Proteins, Signal Transducing , Breast Neoplasms , Focal Adhesion Kinase 1 , Gene Expression Regulation, Neoplastic , Histone Deacetylases , NF-kappa B , Repressor Proteins , Animals , Female , Humans , Mice , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , Breast Neoplasms/pathology , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Line, Tumor , Disease Models, Animal , Epithelial-Mesenchymal Transition/genetics , Focal Adhesion Kinase 1/metabolism , Focal Adhesion Kinase 1/genetics , Histone Deacetylases/metabolism , Histone Deacetylases/genetics , Macrophages/metabolism , Macrophages/pathology , Neoplasm Metastasis , NF-kappa B/metabolism , Phosphoproteins/metabolism , Phosphoproteins/genetics , Prognosis , Repressor Proteins/genetics , Repressor Proteins/metabolism , Signal Transduction , Tumor Microenvironment/genetics
8.
Eur J Pharmacol ; 983: 176983, 2024 Nov 15.
Article in English | MEDLINE | ID: mdl-39243926

ABSTRACT

Chronic kidney disease (CKD) is a global public health problem, involving about 10% of the global population. Unfortunately, there are currently no effective drugs. Kidney fibrosis is the main pathology of CKD, where integrins play crucial roles in renal fibrogenesis. Recently, Bexotegrast (PLN-74809) as a dual integrin αvß1/αvß6 inhibitor could reduce the degree of lung fibrosis in patients with idiopathic pulmonary fibrosis. However, the role of PLN-74809 remains unclear in fibrotic kidney disease. Here, we have revealed that PLN-74809 administration dose-dependently delayed the progression of renal fibrosis in both adenine diet- and unilateral ureteral obstruction (UUO)-induced mice. Mechanistically, PLN-74809 targeted integrin αvß1/αvß6 to inhibit FAK/Src/Akt/ß-catenin cascade in fibrotic kidneys. In summary, our results for the first time highlighted the αvß1/αvß6 inhibitor PLN-74809 exerted potential therapeutic against kidney fibrosis.


Subject(s)
Fibrosis , Integrins , Animals , Male , Mice , Adenine/analogs & derivatives , Adenine/pharmacology , Antigens, Neoplasm/metabolism , beta Catenin/metabolism , beta Catenin/antagonists & inhibitors , Disease Models, Animal , Focal Adhesion Kinase 1/metabolism , Focal Adhesion Kinase 1/antagonists & inhibitors , Integrins/antagonists & inhibitors , Integrins/metabolism , Kidney/pathology , Kidney/drug effects , Kidney/metabolism , Kidney Diseases/pathology , Kidney Diseases/drug therapy , Kidney Diseases/metabolism , Mice, Inbred C57BL , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Vitronectin/antagonists & inhibitors , Receptors, Vitronectin/metabolism , Signal Transduction/drug effects , src-Family Kinases/antagonists & inhibitors , src-Family Kinases/metabolism , Ureteral Obstruction/pathology , Ureteral Obstruction/complications , Ureteral Obstruction/drug therapy
9.
Mol Med ; 30(1): 163, 2024 Sep 27.
Article in English | MEDLINE | ID: mdl-39333897

ABSTRACT

BACKGROUND: Fibrosis cataract occurs in patients receiving cataract extraction. Still, no medication that can cure the disease exists in clinical. This study aims to investigate the effects and mechanisms of Entrectinib on fibrotic cataract in vitro and in vivo. METHODS: The human lens cells line SRA 01/04 and C57BL/6J mice were applied in the study. Entrectinib was used in animals and cells. Cataract severity was assessed by slit lamp and Hematoxylin and Eosin staining. Expression of alpha-smooth muscle actin, fibronectin, and collagen I were examined by real-time quantitative PCR, western blotting, and immunofluorescence. Cell proliferation was evaluated by Cell Counting Kit-8. Cell migration was measured by wound healing and transwell assays. Molecular docking, Drug Affinity Responsive Target Stability, and Cellular Thermal Shift Assay were applied to seek and certify the target of Entrectinib treating fibrosis cataract. RESULTS: Entrectinib can ameliorate fibrotic cataract in vitro and in vivo. At the RNA and the protein levels, the expression of alpha-smooth muscle actin, collagen I, and fibronectin can be downgraded by Entrectinib, while E-cadherin can be upregulated. The migration and proliferation of cells were inhibited by Entrectinib. Mechanistically, Entrectinib obstructs TGFß2/Smad and TGFß2/non-Smad signaling pathways to hinder the fibrosis cataract by targeting PYK2 protein. CONCLUSIONS: Targeting with PYK2, Entrectinib can block TGF-ß2/Smad and TGF-ß2/non-Smad signaling pathways, lessen the activation of EMT, and alleviate fibrosis cataract. Entrectinib may be a potential treatment for fibrosis cataract in clinic.


Subject(s)
Cataract , Focal Adhesion Kinase 2 , Signal Transduction , Transforming Growth Factor beta2 , Animals , Mice , Signal Transduction/drug effects , Cataract/etiology , Cataract/drug therapy , Cataract/metabolism , Cataract/pathology , Humans , Transforming Growth Factor beta2/metabolism , Focal Adhesion Kinase 2/metabolism , Cell Proliferation/drug effects , Disease Models, Animal , Cell Movement/drug effects , Cell Line , Mice, Inbred C57BL , Molecular Docking Simulation , Indazoles/pharmacology , Indazoles/therapeutic use , Male , Focal Adhesion Kinase 1
10.
Cell Death Dis ; 15(9): 678, 2024 Sep 16.
Article in English | MEDLINE | ID: mdl-39284838

ABSTRACT

SPHK1 (sphingosine kinase type 1) is characterized as a rate-limiting enzyme in sphingolipid metabolism to phosphorylate sphingosine into sphingosine-1-phosphate (S1P) that can bind to S1P receptors (S1PRs) to initiate several signal transductions leading to cell proliferation and survival of normal cell. Many studies have indicated that SPHK1 is involved in several types of cancer development, however, a little is known in bladder cancer. The TCGA database analysis was utilized for analyzing the clinical relevance of SPHK1 in bladder cancer. Through CRISPR/Cas9 knockout (KO) and constitutive activation (CA) strategies on SPHK1 in the bladder cancer cells, we demonstrated the potential downstream target could be programmed cell death 1 ligand 2 (PD-L2). On the other hand, we demonstrated that FDA-approved SPHK1 inhibitor Gilenya® (FTY720) can successfully suppress bladder cancer metastasis by in vitro and in vivo approaches. This finding indicated that SPHK1 as a potent therapeutic target for metastatic bladder cancer by dissecting the mechanism of action, SPHK1/S1P-elicited Akt/ß-catenin activation promoted the induction of PD-L2 that is a downstream effector in facilitating bladder cancer invasion and migration. Notably, PD-L2 interacted with c-Src that further activates FAK. Here, we unveil the clinical relevance of SPHK1 in bladder cancer progression and the driver role in bladder cancer metastasis. Moreover, we demonstrated the inhibitory effect of FDA-approved SPHK1 inhibitor FTY720 on bladder cancer metastasis from both in vitro and in vivo models.


Subject(s)
Phosphotransferases (Alcohol Group Acceptor) , Signal Transduction , Urinary Bladder Neoplasms , Urinary Bladder Neoplasms/pathology , Urinary Bladder Neoplasms/metabolism , Urinary Bladder Neoplasms/genetics , Humans , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Animals , Cell Line, Tumor , Focal Adhesion Kinase 1/metabolism , Focal Adhesion Kinase 1/genetics , Neoplasm Metastasis , Mice , Sphingosine/analogs & derivatives , Sphingosine/metabolism , src-Family Kinases/metabolism , Cell Movement , Mice, Nude , Lysophospholipids/metabolism , CSK Tyrosine-Protein Kinase/metabolism , Fingolimod Hydrochloride/pharmacology , Cell Proliferation
11.
Sci Rep ; 14(1): 21451, 2024 09 13.
Article in English | MEDLINE | ID: mdl-39271782

ABSTRACT

Based on the joint analysis of multi-omic data and the biological experiments, we demonstrate that FOXF1 inhibits invasion and metastasis of lung adenocarcinoma cells and enhances anti-tumor immunity via regulating MFAP4/FAK signal axis in this study. The levels of FOXF1 and MFAP4 are significantly down-regulated in LUAD, and the increased levels of two genes can improve the clinical prognosis of LUAD patients. Fluorescein reporter gene determination, chromatin immunoprecipitation and gene co-expression analysis indicate that MFAP4 level is positively regulated by transcription factor FOXF1. The function enrichment analysis shows that the levels of FOXF1 and MFAP4 are closely associated with an enrichment of tumor metastasis signatures. FOXF1 can inhibit the migration and invasion of LAUD cells by transcriptionally activating MFAP4 expression. And the overexpression of FOXF1/MFAP4 can reduce focal adhesion kinase (FAK) phosphorylation, while their knockdown result in the opposite effects. The increased levels of FOXF1/MFAP4 enhance the antitumor immunity by increasing the infiltration of dendritic cells and CD4+ T cells, and the interactions between LUAD cells and immune cells, and activating multiple anti-tumor immunity-related pathways. In conclusion, our study reveals the potential function of FOXF1/MFAP4/FAK signal axis in inhibiting metastasis of LUAD cells and modulating anti-tumor immunity of LUAD patients.


Subject(s)
Adenocarcinoma of Lung , Forkhead Transcription Factors , Lung Neoplasms , Neoplasm Invasiveness , Signal Transduction , Forkhead Transcription Factors/metabolism , Forkhead Transcription Factors/genetics , Humans , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/pathology , Adenocarcinoma of Lung/immunology , Adenocarcinoma of Lung/metabolism , Lung Neoplasms/pathology , Lung Neoplasms/immunology , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/secondary , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Neoplasm Metastasis , Focal Adhesion Kinase 1/metabolism , Focal Adhesion Kinase 1/genetics , Cell Movement , Mice , Animals , Focal Adhesion Protein-Tyrosine Kinases/metabolism
12.
Int J Mol Sci ; 25(17)2024 Aug 26.
Article in English | MEDLINE | ID: mdl-39273200

ABSTRACT

Thrombosis is a key process that determines acute coronary syndrome and ischemic stroke and is the leading cause of morbidity and mortality in the world, together with cancer. Platelet adhesion and subsequent activation and aggregation are critical processes that cause thrombus formation after endothelial damage. To date, high hopes are associated with compounds of natural origin, which show anticoagulant action without undesirable effects and can be proposed as supportive therapies. We investigated the effect of the new combination of four natural compounds, escin-bromelain-ginkgo biloba-sage miltiorrhiza (EBGS), on the initial process of the coagulation cascade, which is the adhesion of platelets to activated vascular endothelium. Our results demonstrated that EBGS pretreatment of endothelial cells reduces platelet adhesion even in the presence of the monocyte-lymphocyte population. Our data indicate that EBGS exerts its effects by inhibiting the transcription of adhesion molecules, including P-selectin, platelet membrane glycoprotein GP1b, integrins αV and ß3, and reducing the secretion of the pro-inflammatory cytokines interleukin 6, interleukin 8, and the metalloproteinases MMP-2 and MMP-9. Furthermore, we demonstrated that EBGS inhibited the expression of focal adhesion kinase (FAK), strictly involved in platelet adhesion, and whose activity is correlated with that of integrin ß3. The results shown in this manuscript suggest a possible inhibitory role of the new combination EBGS in the reduction in platelet adhesion to activated endothelium, thus possibly preventing coagulation cascade initiation.


Subject(s)
Endothelium, Vascular , Platelet Adhesiveness , Signal Transduction , Tumor Necrosis Factor-alpha , Humans , Platelet Adhesiveness/drug effects , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/metabolism , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Blood Platelets/metabolism , Blood Platelets/drug effects , Salvia miltiorrhiza/chemistry , Focal Adhesion Kinase 1/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/drug effects , Plant Extracts/pharmacology
13.
Cancer Med ; 13(17): e70210, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39240189

ABSTRACT

BACKGROUND: High-grade endometrial cancers (EAC) are aggressive tumors with a high risk of progression after treatment. As EAC may harbor mutations in the RAS/MAPK pathways, we evaluated the preclinical in vitro and in vivo efficacy of avutometinib, a RAF/MEK clamp, in combination with the focal adhesion kinase (FAK) inhibitors defactinib or VS-4718, against multiple primary EAC cell lines and xenografts. METHODS: Whole-exome sequencing (WES) was used to evaluate the genetic landscape of five primary EAC cell lines. The in vitro activity of avutometinib and defactinib as single agents and in combination was evaluated using cell viability, cell cycle, and cytotoxicity assays. Mechanistic studies were performed using Western blot assays while in vivo experiments were completed in UTE10 engrafted mice treated with either vehicle, avutometinib, VS-4718, or their combination through oral gavage. RESULTS: WES results demonstrated multiple EAC cell lines to harbor genetic derangements in the RAS/MAPK pathway including KRAS/PTEN/PIK3CA/BRAF/ARID1A, potentially sensitizing to FAK and RAF/MEK inhibition. Five out of five of the EAC cell lines demonstrated in vitro sensitivity to FAK and/or RAF/MEK inhibition. By Western blot assays, exposure of EAC cell lines to defactinib, avutometinib, and their combination demonstrated decreased phosphorylated FAK (p-FAK) as well as decreased p-MEK and p-ERK. In vivo the combination of avutometinib/VS-4718 demonstrated superior tumor growth inhibition compared to single-agent treatment and controls starting at Day 9 (p < 0.02 and p < 0.04) in UTE10 xenografts. CONCLUSIONS: Avutometinib, defactinib, and to a larger extent their combinations, demonstrated promising in vitro and in vivo activity against EAC cell lines and xenografts. These preclinical data support the potential clinical evaluation of this combination in high-grade EAC patients.


Subject(s)
Endometrial Neoplasms , Xenograft Model Antitumor Assays , Female , Humans , Animals , Mice , Endometrial Neoplasms/drug therapy , Endometrial Neoplasms/pathology , Endometrial Neoplasms/genetics , Cell Line, Tumor , Carcinoma, Endometrioid/drug therapy , Carcinoma, Endometrioid/pathology , Carcinoma, Endometrioid/genetics , Carcinoma, Endometrioid/metabolism , Exome Sequencing , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cell Proliferation/drug effects , Neoplasm Grading , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Focal Adhesion Kinase 1/metabolism , Focal Adhesion Kinase 1/antagonists & inhibitors , Oxazepines , Sulfonamides , Pyrazines , Benzamides , Imidazoles
14.
Oncogene ; 43(39): 2951-2969, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39174859

ABSTRACT

High invasive capacity and acquired tyrosine kinase inhibitors (TKI) resistance of kidney renal clear cell carcinoma (KIRC) cells remain obstacles to prolonging the survival time of patients with advanced KIRC. In the present study, we reported that sine oculis homeobox 1 (SIX1) was upregulated in sunitinib-resistant KIRC cells and metastatic KIRC tissues. Subsequently, we found that SIX1 mediated metastasis and sunitinib resistance via Focal adhesion (FA) signaling, and knockdown of SIX1 enhanced the antitumor efficiency of sunitinib in KIRC. Mechanistically, Integrin subunit beta 1 (ITGB1), an upstream gene of FA signaling, was a direct transcriptional target of SIX1. In addition, we showed that DExH-box helicase 9 (DHX9) was an important mediator for SIX1-induced ITGB1 transcription, and silencing the subunits of SIX1/DHX9 complex significantly reduced transcription of ITGB1. Downregulation of SIX1 attenuated nuclear translocation of DHX9 and abrogated the binding of DHX9 to ITGB1 promoter. Collectively, our results unveiled a new signal axis SIX1/ITGB1/FAK in KIRC and identified a novel therapeutic strategy for metastatic KIRC patients.


Subject(s)
Carcinoma, Renal Cell , DEAD-box RNA Helicases , Drug Resistance, Neoplasm , Focal Adhesions , Gene Expression Regulation, Neoplastic , Homeodomain Proteins , Integrin beta1 , Kidney Neoplasms , Neoplasm Metastasis , Signal Transduction , Sunitinib , Humans , Drug Resistance, Neoplasm/genetics , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/pathology , Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/metabolism , Sunitinib/pharmacology , Sunitinib/therapeutic use , Kidney Neoplasms/pathology , Kidney Neoplasms/genetics , Kidney Neoplasms/drug therapy , Kidney Neoplasms/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Cell Line, Tumor , Integrin beta1/genetics , Integrin beta1/metabolism , Animals , Focal Adhesions/genetics , Focal Adhesions/metabolism , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/metabolism , Mice , Transcription, Genetic , Integrins/metabolism , Integrins/genetics , Focal Adhesion Kinase 1
15.
Oncogene ; 43(40): 2995-3002, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39209965

ABSTRACT

Neurofibromatosis type 2 (NF2) is a rare disorder that causes vestibular schwannomas (VS), meningiomas and ependymomas. To date, there is no FDA approved drug-based treatment for NF2. We have previously identified that BET inhibition can selectively reduce growth of the NF2-null schwannoma and Schwann cells in vitro and tumorigenesis in vivo and, separately, reported that inhibition of Focal Adhesion Kinase 1 (FAK1) via crizotinib has antiproliferative effects in NF2-null Schwann cells. The current study was aimed at determining whether combined BET and FAK inhibition can synergize and to identify the mechanisms of action. A panel of normal and NF2-null Schwann and schwannoma cell lines were used to characterize the effects of combined BET and FAK inhibition in vitro and in vivo using pharmacological and genetic approaches. The mechanism of action was explored by chromatin immunoprecipitation, ChIP-PCR, western blotting, and functional approaches. We find that combined BET and FAK inhibition are synergistic and inhibit the proliferation of NF2-null schwannoma and Schwann cell lines in vitro and in vivo, by arresting cells in the G1/S and G2/M phases of the cell cycle. Further, we identify the mechanism of action through the downregulation of FAK1 transcription by BET inhibition, which potentiates inhibition of FAK by 100-fold. Our findings suggest that combined targeting of BET and FAK1 may offer a potential therapeutic option for the treatment of NF2-related schwannomas.


Subject(s)
Cell Proliferation , Focal Adhesion Kinase 1 , Neurilemmoma , Neurofibromin 2 , Neuroma, Acoustic , Animals , Humans , Mice , Focal Adhesion Kinase 1/metabolism , Focal Adhesion Kinase 1/antagonists & inhibitors , Focal Adhesion Kinase 1/genetics , Neuroma, Acoustic/pathology , Neuroma, Acoustic/genetics , Neuroma, Acoustic/drug therapy , Neuroma, Acoustic/metabolism , Neurilemmoma/pathology , Neurilemmoma/genetics , Neurilemmoma/drug therapy , Neurilemmoma/metabolism , Neurofibromin 2/genetics , Neurofibromin 2/metabolism , Cell Proliferation/drug effects , Cell Line, Tumor , Drug Synergism , Neurofibromatoses/drug therapy , Neurofibromatoses/genetics , Neurofibromatoses/pathology , Neurofibromatosis 2/genetics , Neurofibromatosis 2/drug therapy , Neurofibromatosis 2/pathology , Neurofibromatosis 2/metabolism , Schwann Cells/drug effects , Schwann Cells/metabolism , Schwann Cells/pathology , Skin Neoplasms/pathology , Skin Neoplasms/drug therapy , Skin Neoplasms/genetics , Skin Neoplasms/metabolism , Xenograft Model Antitumor Assays
16.
Cell Biol Int ; 48(11): 1766-1778, 2024 Nov.
Article in English | MEDLINE | ID: mdl-39192576

ABSTRACT

Cholangiocarcinoma (CCA) is a hepatobiliary carcinoma with uncontrolled cell proliferation, poor prognosis, and high mortality. The ovarian tumor structural domain (OTU) containing protein 6B (OTUD6B) belongs to the OTU deubiquitin family and is vital in tumor development. However, its expression and biological function in CCA remain unknown. The expression of OTUD6B in CCA was analyzed using TIMER2.0, UALCAN, and GEO databases. MTT, clonal formation assay, immunofluorescence staining, immunohistochemistry staining, and flow cytometry examined the regulation of OTUD6B on cell proliferation, cycle, and apoptosis. The effects of OTUD6B on tumor volume and weight were assessed using the xenograft tumor model. The activities of PTK2 and STAT3 were detected by western blot and CO-IP. The biological database identified that OTUD6B was upregulated in CCA. In CCA cells, OTUD6B knockdown reduced CCA cell proliferation and promoted apoptosis. Cell cycle analysis indicated that the cycle stopped at the G0/G1 phase after OTU6B downregulation. Furthermore, OTUD6B knockdown resulted in a decrease in tumor volume and weight in xenograft tumor models. Mechanistically, OTUD6B is involved in the deubiquitination of PTK2. PTK2 further affected the phosphorylation of STAT3 thereby regulating the CCA process. Our study demonstrates that OTUD6B knockdown participates in the ubiquitination of PTK2 and phosphorylation of STAT3 to alleviate the process of CCA. These results suggest that OTUD6B may be a potential new strategy for CCA treatment.


Subject(s)
Apoptosis , Cell Proliferation , Cholangiocarcinoma , Endopeptidases , Focal Adhesion Kinase 1 , STAT3 Transcription Factor , Animals , Humans , Mice , Bile Duct Neoplasms/metabolism , Bile Duct Neoplasms/pathology , Cell Line, Tumor , Cholangiocarcinoma/metabolism , Cholangiocarcinoma/pathology , Endopeptidases/genetics , Endopeptidases/metabolism , Focal Adhesion Kinase 1/metabolism , Focal Adhesion Kinase 1/genetics , Mice, Inbred BALB C , Mice, Nude , Phosphorylation , STAT3 Transcription Factor/metabolism , Ubiquitin-Specific Proteases/metabolism , Ubiquitin-Specific Proteases/genetics , Ubiquitination
17.
PLoS One ; 19(8): e0304010, 2024.
Article in English | MEDLINE | ID: mdl-39150901

ABSTRACT

M64HCl, which has drug-like properties, is a water-soluble Focal Adhesion Kinase (FAK) activator that promotes murine mucosal healing after ischemic or NSAID-induced injury. Since M64HCl has a short plasma half-life in vivo (less than two hours), it has been administered as a continuous infusion with osmotic minipumps in previous animal studies. However, the effects of more transient exposure to M64HCl on monolayer wound closure remained unclear. Herein, we compared the effects of shorter M64HCl treatment in vitro to continuous treatment for 24 hours on monolayer wound closure. We then investigated how long FAK activation and downstream ERK1/2 activation persist after two hours of M64HCl treatment in Caco-2 cells. M64HCl concentrations immediately after washing measured by mass spectrometry confirmed that M64HCl had been completely removed from the medium while intracellular concentrations had been reduced by 95%. Three-hour and four-hour M64HCl (100 nM) treatment promoted epithelial sheet migration over 24 hours similar to continuous 24-hour exposure. 100nM M64HCl did not increase cell number. Exposing cells twice with 2-hr exposures of M64HCl during a 24-hour period had a similar effect. Both FAK inhibitor PF-573228 (10 µM) and ERK kinase (MEK) inhibitor PD98059 (20 µM) reduced basal wound closure in the absence of M64HCl, and each completely prevented any stimulation of wound closure by M64HCl. Rho kinase inhibitor Y-27632 (20 µM) stimulated Caco-2 monolayer wound closure but no further increase was seen with M64HCl in the presence of Y-27632. M64HCl (100 nM) treatment for 3 hours stimulated Rho kinase activity. M64HCl decreased F-actin in Caco-2 cells. Furthermore, a two-hour treatment with M64HCl (100 nM) stimulated sustained FAK activation and ERK1/2 activation for up to 16 and hours 24 hours, respectively. These results suggest that transient M64HCl treatment promotes prolonged intestinal epithelial monolayer wound closure by stimulating sustained activation of the FAK/ERK1/2 pathway. Such molecules may be useful to promote gastrointestinal mucosal repair even with a relatively short half-life.


Subject(s)
Intestinal Mucosa , Wound Healing , Humans , Wound Healing/drug effects , Caco-2 Cells , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Focal Adhesion Kinase 1/metabolism , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Cell Movement/drug effects , Pyridines/pharmacology , Animals , Amides/pharmacology
18.
Biomed Pharmacother ; 179: 117335, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39191020

ABSTRACT

Cancer cells mechanically interact with the tumor microenvironment during cancer development. Mechano-reciprocity has emerged as a crucial factor affecting anti-cancer drug resistance during adjuvant therapy. Here, we investigated the focal adhesion kinase (FAK)/Yes-associated protein (YAP) signaling axis as a prospective strategy for circumventing cisplatin resistance in ovarian cancer (OC). The Cancer Genome Atlas (TCGA) data analysis revealed that FAK overexpression significantly correlated with unfavorable clinical outcomes in patients with ovarian cancer. AFM indentation experiments showed that cell elasticity depends on FAK activity. Notably, the combination of FAK inhibition and cisplatin treatment led to a 69 % reduction in the IC50 of cisplatin. This combined treatment also increased apoptosis compared to the individual treatments, along with the upregulation of the pro-apoptotic factor BAX and cleaved PARP. Suppressing FAK expression sequestered YAP in the cytosol, potentially reducing cellular proliferation and promoting apoptosis. Moreover, reduced FAK expression sensitized drug-resistant OC cells to cisplatin treatment owing to a decrease in nuclear tension, allowing the relocation of YAP to the cytosol. In a mouse model, the co-administration of an FAK inhibitor and cisplatin significantly suppressed tumor growth and increased apoptotic events and DNA fragmentation. Our findings suggest that drug resistance can be attributed to the perturbation of mechanosensing signaling pathways, which drive the mechanical reinforcement of cancer cells. OC cells can restore their sensitivity to cisplatin treatment by strategically reducing YAP localization in the nucleus through FAK downregulation.


Subject(s)
Apoptosis , Cisplatin , Drug Resistance, Neoplasm , Focal Adhesion Kinase 1 , Mice, Nude , Ovarian Neoplasms , Transcription Factors , YAP-Signaling Proteins , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/pathology , Ovarian Neoplasms/metabolism , Female , Drug Resistance, Neoplasm/drug effects , Humans , Animals , Cisplatin/pharmacology , Cell Line, Tumor , YAP-Signaling Proteins/metabolism , Apoptosis/drug effects , Focal Adhesion Kinase 1/metabolism , Transcription Factors/metabolism , Antineoplastic Agents/pharmacology , Adaptor Proteins, Signal Transducing/metabolism , Mice , Xenograft Model Antitumor Assays , Mice, Inbred BALB C , Mechanotransduction, Cellular/drug effects , Cell Proliferation/drug effects , Signal Transduction/drug effects
19.
J Transl Med ; 22(1): 800, 2024 Aug 29.
Article in English | MEDLINE | ID: mdl-39210440

ABSTRACT

BACKGROUND: Recycling of integrin via endosomal vesicles is critical for the migration of cancer cells, which leads to the metastasis of pancreatic cancer and devastating cancer-related death. So, new diagnostic and therapeutic molecules which target the recycling of endosomal vesicles need to be developed. METHODS: Public databases including TCGA, ICGC, GSE21501, GSE28735, and GENT are analyzed to derive diagnostic and therapeutic targets. To reveal biological roles and underlying mechanisms of molecular targets, various molecular biological experiments were conducted. RESULTS: First, we identified UNC13D's overexpression in patients with pancreatic cancer (n = 824) and its prognostic significance and high hazard ratio (HR) in four independent pancreatic cancer cohorts (TCGA, n = 178, p = 0.014, HR = 3.629; ICGC, n = 91, p = 0.000, HR = 4.362; GSE21501, n = 102, p = 0.002, HR = 2.339; GSE28735, n = 45, p = 0.022, HR = 2.681). Additionally, its expression is associated with the clinicopathological progression of pancreatic cancer. Further biological studies have shown that UNC13D regulates the migration of pancreatic cancer cells by coupling the exocytosis of recycling endosomes with focal adhesion turnover via the regulation of FAK phosphorylation. Immunoprecipitation and immunocytochemistry showed the formation of the RAB11-UNC13D-FAK axis in endosomes during integrin recycling. We observed that UNC13D directly interacted with the FERM domain of FAK and regulated FAK phosphorylation in a calcium-dependent manner. Finally, we found co-expression of UNC13D and FAK showed the poorest survival (TCGA, p = 0.000; ICGC, p = 0.036; GSE28735, p = 0.006). CONCLUSIONS: We highlight that UNC13D, a novel prognostic factor, promotes pancreatic cancer progression by coupling integrin recycling with focal adhesion turnover via the RAB11-UNC13D-FAK axis for the migration of pancreatic cancer cells.


Subject(s)
Cell Movement , Focal Adhesions , Integrins , Pancreatic Neoplasms , rab GTP-Binding Proteins , Humans , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/genetics , rab GTP-Binding Proteins/metabolism , Cell Line, Tumor , Focal Adhesions/metabolism , Integrins/metabolism , Focal Adhesion Kinase 1/metabolism , Female , Male , Signal Transduction , Middle Aged , Prognosis , Gene Expression Regulation, Neoplastic , Endosomes/metabolism , Disease Progression
20.
Neuropeptides ; 107: 102460, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39142164

ABSTRACT

The destruction of the blood-brain barrier and damage to the gastrointestinal mucosa after intracerebral hemorrhage (ICH) are important reasons for its high disability and mortality rates. However, the exact etiology is not yet clear. In addition, there are currently no effective treatments for improving cerebral edema and gastric mucosal damage after ICH. Trefoil factor 1 (TFF1) is a secretory protein that plays a crucial role in maintaining the integrity and barrier function of the gastric mucosa, and it has been reported to have a protective effect on brain damage induced by various causes. This study utilized a rat model of ICH induced by type IV collagenase was utilized, and intervened with recombinant TFF1 protein from an external institute to investigate the protective mechanisms of TFF1 against brain edema and gastric mucosal damage after ICH. The results demonstrated that TFF1 alleviated the neurological function and gastric mucosal damage in the rat model of ICH induced by type IV collagenase. TFF1 may ensure the integrity of the blood-brain and gastric mucosal barriers by regulating the EGFR (epidermal growth factor receptor)/Src (non-receptor tyrosine kinase)/FAK (focal adhesion kinase) pathway. Clearly, the disruption of the blood-brain barrier and the destruction of the gastric mucosal barrier are key pathological features of ICH, and TFF1 can improve the progression of blood-brain barrier and gastric mucosal barrier disruption in ICH by regulating the EGFR/Src/FAK pathway. Therefore, TFF1 may be a potential target for the treatment of ICH.


Subject(s)
Brain Edema , Cerebral Hemorrhage , Disease Models, Animal , ErbB Receptors , Gastric Mucosa , Trefoil Factor-1 , src-Family Kinases , Animals , Male , Rats , Blood-Brain Barrier/metabolism , Brain Edema/metabolism , Cerebral Hemorrhage/metabolism , ErbB Receptors/metabolism , Focal Adhesion Kinase 1/metabolism , Gastric Mucosa/metabolism , Gastric Mucosa/injuries , Rats, Sprague-Dawley , Signal Transduction , src-Family Kinases/metabolism , Trefoil Factor-1/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL