Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Publication year range
1.
Cell Death Dis ; 13(10): 858, 2022 10 08.
Article in English | MEDLINE | ID: mdl-36209201

ABSTRACT

PLEKHH2 is an important FERM domain containing-protein. However, the role of PLEKHH2 in human solid tumors has not been reported yet. We report that PLEKHH2 showed enhanced cytoplasmic expression in non-small cell lung cancer (NSCLC). Its overexpression was positively correlated with high TNM stage, low differentiation, lymphatic node metastasis, and poor prognosis. In A549 and H1299 cells, high expression of PLEKHH2 significantly promoted cell proliferation, migration, invasion, and increased the expression of proliferation- and invasion-related proteins. It also enhanced the phosphorylation of FAK and promoted the activity of the PI3K/AKT pathway. Immunofluorescence and co-immunoprecipitation analyses were performed to elucidate the molecular mechanism underlying PLEKHH2-mediated regulation of proliferation and invasion in lung cancer cells. Upon transfection of full length PLEKHH2 or its FERM domain, we observed enhanced binding of PLEKHH2 to ß-arrestin1, whereas FAK- ß-arrestin1 binding was diminished and this led to an increase in FAK phosphorylation. PLEKHH2-mutant plasmids without the FERM domain could not effectively promote its binding to ß-arrestin1, activation of FAK phosphorylation, PI3K/AKT activation, or the malignant phenotype. Our findings suggested that PLEKHH2 is an important oncogene in NSCLC. PLEKHH2 binding to ß-arrestin1 through the FERM domain competitively inhibits ß-arrestin1 binding to FAK, which causes the dissociation of FAK from the FAK-ß-arrestin1 complex. Furthermore, the dissociation of FAK promotes its autophosphorylation, activates the PI3K/AKT signaling pathway, and subsequently promotes lung cancer cell proliferation, migration, and invasion. These results provide evidence for the potential use of PLEKHH2 inhibition as an anticancer therapy.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , beta-Arrestins/metabolism , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Movement , Cell Proliferation , Cytoskeletal Proteins/metabolism , Focal Adhesion Kinase 1/genetics , Focal Adhesion Kinase 1/metabolism , Humans , Lung Neoplasms/pathology , Phenotype , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism
2.
Front Immunol ; 13: 907309, 2022.
Article in English | MEDLINE | ID: mdl-35769488

ABSTRACT

Identifying biomarkers for abdominal aortic aneurysms (AAA) is key to understanding their pathogenesis, developing novel targeted therapeutics, and possibly improving patients outcomes and risk of rupture. Here, we identified AAA biomarkers from public databases using single-cell RNA-sequencing, weighted co-expression network (WGCNA), and differential expression analyses. Additionally, we used the multiple machine learning methods to identify biomarkers that differentiated large AAA from small AAA. Biomarkers were validated using GEO datasets. CIBERSORT was used to assess immune cell infiltration into AAA tissues and investigate the relationship between biomarkers and infiltrating immune cells. Therefore, 288 differentially expressed genes (DEGs) were screened for AAA and normal samples. The identified DEGs were mostly related to inflammatory responses, lipids, and atherosclerosis. For the large and small AAA samples, 17 DEGs, mostly related to necroptosis, were screened. As biomarkers for AAA, G0/G1 switch 2 (G0S2) (Area under the curve [AUC] = 0.861, 0.875, and 0.911, in GSE57691, GSE47472, and GSE7284, respectively) and for large AAA, heparinase (HPSE) (AUC = 0.669 and 0.754, in GSE57691 and GSE98278, respectively) were identified and further verified by qRT-PCR. Immune cell infiltration analysis revealed that the AAA process may be mediated by T follicular helper (Tfh) cells and the large AAA process may also be mediated by Tfh cells, M1, and M2 macrophages. Additionally, G0S2 expression was associated with neutrophils, activated and resting mast cells, M0 and M1 macrophages, regulatory T cells (Tregs), resting dendritic cells, and resting CD4 memory T cells. Moreover, HPSE expression was associated with M0 and M1 macrophages, activated and resting mast cells, Tregs, and resting CD4 memory T cells. Additional, G0S2 may be an effective diagnostic biomarker for AAA, whereas HPSE may be used to confer risk of rupture in large AAAs. Immune cells play a role in the onset and progression of AAA, which may improve its diagnosis and treatment.


Subject(s)
Aortic Aneurysm, Abdominal , Cell Cycle Proteins , Glucuronidase , Machine Learning , Aortic Aneurysm, Abdominal/diagnosis , Aortic Aneurysm, Abdominal/metabolism , Biomarkers/metabolism , Cell Cycle Proteins/metabolism , Glucuronidase/metabolism , Heparin Lyase/metabolism , Humans , Macrophages/metabolism
3.
Aging (Albany NY) ; 14(10): 4402-4424, 2022 05 17.
Article in English | MEDLINE | ID: mdl-35579998

ABSTRACT

Gliomas are the most common malignant tumor in the brain. As with other tumors, the progression of glioma depends on intra-tumoral angiogenesis. However, the effect of angiogenesis on gliomas is still not fully understood. In this study, we developed an angiogenesis pathway score using Gene Set Variation Analysis (GSAV) in R to assess the status of intra-glioma angiogenesis in The Cancer Genome Atlas (TCGA), Chinese Glioma Genome Atlas (CGGA mRNAseq_325, CGGA mRNA-array), and GSE16011 datasets. We found that the angiogenesis pathway score not only accurately predicted the prognosis of glioma patients, but also accurately distinguished the malignant phenotype and immune characteristics of gliomas. In addition, as an independent prognostic factor, the score could predict glioma sensitivity to radiotherapy and chemotherapy. In summary, we used the angiogenesis pathway score to reveal the relationship between glioma angiogenesis and the malignant phenotype, immune characteristics, and prognosis of glioma.


Subject(s)
Brain Neoplasms , Glioma , Biomarkers, Tumor/genetics , Brain Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Glioma/pathology , Humans , Prognosis
4.
Article in Chinese | MEDLINE | ID: mdl-12508431

ABSTRACT

OBJECTIVE: To provide anatomical bases for dorso-ulnar aspect of mid-hand reverse flap. METHODS: After red latex was infused into the arteries of 40 sides of adult cadava upper limbs, the origin, course, branches, distribution and distal anastomosis on the dorsal carpal branch of ulnar arteries were observed. And the mid-hand flap transfer was used to repair two cases of soft tissue defect (ranged 4.5-5.0 cm x 2.0-3.5 cm on ring and little fingers). RESULTS: The dorsal carpal branch begins with ulnar artery (3.9 +/- 1.2) cm above the pisiform with diameter of (1.3 +/- 0.2) mm, and branches off into ascending and descending branches. The descending one is the continuing of dorsal branch, it crosses the ulnar edge of the fifth metecarpal bone and anastomizes with the digital artery of little finger or hypothenar branch of deep palmar (accounted for 70%). While the other ascending branch with the former two branches formed anastomosis accounts for 30%. The two cases got healed in one-stage. The function of fingers recovered after 3-4 month follow-up. CONCLUSION: The reverse flap of dorso-ulnar aspect of mid-hand is available to repair the soft tissue defect on dorsum of hand with neighbor finger.


Subject(s)
Finger Injuries/surgery , Surgical Flaps , Ulnar Artery/anatomy & histology , Ulnar Nerve/anatomy & histology , Adult , Anastomosis, Surgical , Female , Hand/blood supply , Hand/innervation , Hand/surgery , Humans , Male , Microsurgery , Soft Tissue Injuries/surgery , Surgical Flaps/blood supply , Surgical Flaps/innervation , Ulnar Artery/innervation
SELECTION OF CITATIONS
SEARCH DETAIL
...