Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
Exp Cell Res ; 426(2): 113567, 2023 05 15.
Article in English | MEDLINE | ID: mdl-36965748

ABSTRACT

BACKGROUND: Low pleckstrin homology-like domain family A, member 3 (PHLDA3) expression has been reported to be associated with cancer specificity and disease-free survival in esophageal squamous cell carcinoma (ESCC), and was an independent predictor of postoperative recurrence. However, the specific mechanisms involved are still unclear. This paper aimed to explore the role and its mechanisms of PHLDA3 in ESCC. MATERIALS AND METHODS: PHLDA3 and BarH-like homeobox 2 (BARX2) expressions in ESCC were predicted by Gene Expression Profiling Interactive Analysis (GEPIA) analysis and determined by quantitative real-time polymerase chain reaction (qRT-PCR) and western Blot. Western blot detected the expression of proteins associated with migration, angiogenesis and phosphoinositide 3-kinase (PI3K)/protein kinase B (PKB/AKT) signaling pathway. The University of California Santa Cruz Genomics Institute (UCSC) database predicted that the relationship of BARX2 and PHLDA3 promoter and JASPAR identified the possible binding sites. Dual luciferase gene reporter verified PHLDA3 promoter activity, and the relationship of both was determined by chromatin immunoprecipitation (CHIP). Cell counting kit (CCK)-8, 5-ethynyl-2'-deoxyuridine (EDU) and colony formation were used to assess cell proliferation. Wound healing and transwell were used to detect cell migration and invasion ability. Tube formation assay was applied to assess angiogenesis. Mice were injected with transfected KYSE30 cells under the right axilla. Body weight and tumor volume and mass were recorded for each group of mice. Immunohistochemistry was performed to detect KI67 level in tumor tissues. RESULTS: Both PHLDA3 and BARX2 were downregulated in ESCC. The upregulated PHLDA3 suppressed PI3K/AKT expression. In addition, BARX2 bound to the PHLDA3 promoter and transcriptionally activated PHLDA3. PHLDA3 overexpression inhibited ESCC cell proliferation, migration, invasion and angiogenesis, but this effect was reversed by BARX2 knockdown. In addition, BARX2 overexpression inhibited ESCC cell proliferation, migration, invasion and angiogenesis, but this effect was reversed by PHLDA3 knockdown. CONCLUSION: PHLDA3 was transcriptionally activated by BARX2 and inhibited malignant progression of ESCC by downregulating PI3K/AKT levels.


Subject(s)
Esophageal Neoplasms , Esophageal Squamous Cell Carcinoma , Homeodomain Proteins , MicroRNAs , Animals , Mice , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Esophageal Neoplasms/pathology , Esophageal Squamous Cell Carcinoma/pathology , Gene Expression Regulation, Neoplastic/genetics , MicroRNAs/genetics , Phosphatidylinositol 3-Kinase/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism
2.
Biochem Biophys Res Commun ; 571: 66-73, 2021 09 24.
Article in English | MEDLINE | ID: mdl-34303965

ABSTRACT

Pleckstrin homology-like domain family A, member 3 (PHLDA3) is a novel tumor-related protein that mediates carcinogenesis of multiple cancers. However, the relevance of PHLDA3 in prostate cancer has not been explored. The purpose of this work was to illustrate the possible roles and mechanisms of PHLDA3 in prostate cancer. Our data showed strikingly lower abundance of PHLDA3 in prostate cancer, and that low levels of PHLDA3 in prostate cancer patients was associated with reduced survival. PHLDA3 was also weakly expressed in prostate cancer cells, and demethylation treatment dramatically up-regulated the expression level of PHLDA3. Up-regulation of PHLDA3 restrained proliferation, induced G1 cell cycle arrest, suppressed epithelial-mesenchymal transition of prostate cancer cells. In addition, up-regulation of PHLDA3 increased the sensitivity of prostate cancer cells to docetaxel In-depth research into the mechanism elucidated that PHLDA3 overexpression decreased the phosphorylation of Akt and suppressed the activation of Wnt/ß-catenin signaling. Overexpression of constitutively active Akt strikingly abolished PHLDA3-mediated inactivation of Wnt/ß-catenin pathway. A xenograft assay revealed that prostate cancer cells with PHLDA3 overexpression displayed reduced tumorigenicity in vivo. Collectively, these data document that PHLDA3 exerts an outstanding cancer-inhibiting role in prostate cancer by down-regulating Wnt/ß-catenin pathway via the inhibition of Akt. This work highlights PHLDA3 as a novel anticancer target for prostate cancer.


Subject(s)
Down-Regulation , Nuclear Proteins/metabolism , Prostatic Neoplasms/metabolism , Proto-Oncogene Proteins c-akt/metabolism , beta Catenin/metabolism , Animals , Apoptosis , Cell Proliferation , Humans , Male , Mice , Mice, Inbred BALB C , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Nuclear Proteins/genetics , Prostatic Neoplasms/pathology , Tumor Cells, Cultured , Wnt Signaling Pathway
3.
Pancreatology ; 21(1): 215-223, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33358592

ABSTRACT

BACKGROUND: Pancreatic neuroendocrine tumors are rare neoplasms for which few predictive and/or prognostic biomarkers have been validated. Our previous work suggested the potential of the combined expression of N-myc downstream-regulated gen-1 (NDRG-1), O6-methylguanine DNA methyltransferase (MGMT) and Pleckstrin homology-like domain family A member 3 (PHLDA-3) as prognostic factors for relapse and survival. METHODS: In this new multicenter study we evaluated immunohistochemistry expression in 76 patients with advanced PanNET who were treated with capecitabine-temozolomide or everolimus. Based on the immunohistochemistry panel, an immunohistochemistry prognostic score (IPS) was developed. RESULTS: In patients treated with capecitabine and temozolomide, low IPS was an independent prognostic factor for progression-free-survival and overall-survival. Similar findings were observed with highest IPS for overall-survival in patients treated with everolimus. CONCLUSION: From our knowledge, it is the first time that a simple IPS could be useful to predict outcome for patients with metastatic pancreatic neuroendocrine tumors treated with everolimus or capecitabine and temozolomide.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Everolimus/therapeutic use , Immunohistochemistry/methods , Immunosuppressive Agents/therapeutic use , Neuroendocrine Tumors/drug therapy , Neuroendocrine Tumors/pathology , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/pathology , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/analysis , Cell Cycle Proteins/analysis , Cell Line, Tumor , DNA Modification Methylases/analysis , DNA Repair Enzymes/analysis , Female , Humans , Intracellular Signaling Peptides and Proteins/analysis , Male , Middle Aged , Neoplasm Recurrence, Local , Neuroendocrine Tumors/mortality , Nuclear Proteins/analysis , Pancreatic Neoplasms/mortality , Prognosis , Progression-Free Survival , Survival Analysis , Tumor Suppressor Proteins/analysis , Young Adult
4.
Environ Toxicol ; 36(11): 2266-2277, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34351043

ABSTRACT

Pleckstrin homology-like domain family A, member 3 (PHLDA3) has a particularly critical role in regulating cell survival under stress conditions. However, whether PHLDA3 plays a role in myocardial ischemia/reperfusion injury has not been studied. We aimed to assess the possible role of PHLDA3 in myocardial ischemia/reperfusion (I/R) injury. PHLDA3 expression was increased in myocardial tissue from rats with myocardial I/R injury and rat cardiomyocytes with hypoxia/reoxygenation (H/R) injury. PHLDA3 knockdown protected against myocardial I/R injury in vivo and H/R injury in vitro. Inhibition of PHLDA3 increased the activation of nuclear factor erythroid-derived 2-related factor 2 (Nrf2) associated with regulation of the Akt/glycogen synthase kinase-3ß (GSK-3ß) axis. Repression of Nrf2 reversed PHLDA3-inhibition-mediated cardioprotective effects. Taken together, our work demonstrates that PHLDA3 inhibition exerts a protective role in myocardial I/R injury via regulation of the Akt/GSK-3ß/Nrf2 axis. We suggest PHLDA3 as an attractive target for developing treatments against myocardial I/R injury.


Subject(s)
Myocardial Reperfusion Injury , NF-E2-Related Factor 2 , Nuclear Proteins/genetics , Animals , Glycogen Synthase Kinase 3 beta , Myocardial Reperfusion Injury/prevention & control , Myocytes, Cardiac , NF-E2-Related Factor 2/genetics , Oxidative Stress , Proto-Oncogene Proteins c-akt/genetics , Rats
5.
Int J Mol Sci ; 21(11)2020 Jun 08.
Article in English | MEDLINE | ID: mdl-32521808

ABSTRACT

p53 is a well-known tumor suppressor gene and one of the most extensively studied genes in cancer research. p53 functions largely as a transcription factor and can trigger a variety of antiproliferative programs via induction of its target genes. We identified PHLDA3 as a p53 target gene and found that its protein product is a suppressor of pancreatic neuroendocrine tumors (PanNETs) and a repressor of Akt function. PHLDA3 is frequently inactivated by loss of heterozygosity (LOH) and methylation in human PanNETs, and LOH at the PHLDA3 gene locus correlates with PanNET progression and poor prognosis. In addition, in PHLDA3-deficient mice, pancreatic islet cells proliferate abnormally and acquire resistance to apoptosis. In this article, we briefly review the roles of p53 and Akt in human neuroendocrine tumors (NETs) and describe the relationship between the p53-PHLDA3 and Akt pathways. We also discuss the role of PHLDA3 as a tumor suppressor in various NETs and speculate on the possibility that loss of PHLDA3 function may be a useful prognostic marker for NET patients indicating particular drug therapies. These results suggest that targeting the downstream PHLDA3-Akt pathway might provide new therapies to treat NETs.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Disease Susceptibility , Neuroendocrine Tumors/etiology , Neuroendocrine Tumors/metabolism , Nuclear Proteins/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Biomarkers, Tumor , Cell Transformation, Neoplastic/genetics , Disease Models, Animal , Humans , Mutation , Neuroendocrine Tumors/pathology , Nuclear Proteins/genetics , Organ Specificity/genetics , Signal Transduction , Tumor Suppressor Protein p53/genetics
6.
Cancer Sci ; 108(6): 1101-1108, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28295876

ABSTRACT

Pancreatic neuroendocrine tumors (PanNET) are rare cancers that generally have a poor prognosis. Accurate diagnosis and proper treatment of these tumors requires a better understanding of the molecular mechanisms underlying the development of PanNET. It has been shown that the mTOR inhibitor everolimus can improve the progression-free survival of PanNET patients, suggesting that inhibition of the PI3K-Akt-mTOR pathway may suppress the progression of PanNET. PHLDA3 is a novel tumor suppressor protein that inhibits Akt activation by competition for binding to PIP3 . Our analysis of PanNET revealed frequent loss-of-heterozygosity and DNA methylation at the PHLDA3 locus, resulting in strong suppression of PHLDA3 transcription. Such alterations in the PHLDA3 gene were also frequently found in lung neuroendocrine tumors (NET), suggesting the possibility that various types of NET have in common the functional loss of the PHLDA3 gene.


Subject(s)
Neuroendocrine Tumors/genetics , Neuroendocrine Tumors/pathology , Nuclear Proteins/genetics , Proto-Oncogene Proteins c-akt/genetics , Animals , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Phosphatidylinositol 3-Kinases/genetics , Transcription, Genetic/genetics
7.
Brain Commun ; 6(4): fcae244, 2024.
Article in English | MEDLINE | ID: mdl-39144751

ABSTRACT

Pleckstrin homology-like domain family A-member 3 (PHLDA3) has recently been identified as a player in adaptive and maladaptive cellular stress pathways. The outcome of pleckstrin homology-like domain family A-member 3 signalling was shown to vary across different cell types and states. It emerges that its expression and protein level are highly increased in amyotrophic lateral sclerosis (ALS) patient-derived astrocytes. Whether it orchestrates a supportive or detrimental function remains unexplored in the context of neurodegenerative pathologies. To directly address the role of pleckstrin homology-like domain family A-member 3 in healthy and ALS astrocytes, we used overexpression and knockdown strategies. We generated cultures of primary mouse astrocytes and also human astrocytes from control and ALS patient-derived induced pluripotent stem cells harbouring the superoxide dismutase 1 mutation. Then, we assessed astrocyte viability and the impact of their secretome on oxidative stress responses in human stem cell-derived cortical and spinal neuronal cultures. Here, we show that PHLDA3 overexpression or knockdown in control astrocytes does not significantly affect astrocyte viability or reactive oxygen species production. However, PHLDA3 knockdown in ALS astrocytes diminishes reactive oxygen species concentrations in their supernatants, indicating that pleckstrin homology-like domain family A-member 3 can facilitate stress responses in cells with altered homeostasis. In support, supernatants of PHLDA3-silenced ALS and even control spinal astrocytes with a lower pleckstrin homology-like domain family A-member 3 protein content could prevent sodium arsenite-induced stress granule formation in spinal neurons. Our findings provide evidence that reducing pleckstrin homology-like domain family A-member 3 levels may transform astrocytes into a more neurosupportive state relevant to targeting non-cell autonomous ALS pathology.

8.
Bioengineered ; 13(2): 3993-4009, 2022 02.
Article in English | MEDLINE | ID: mdl-35112982

ABSTRACT

Pleckstrin homology-like domain family A, member 3 (PHLDA3), is emerging as a critical regulator for multiple cancers. Nevertheless, the expression and role of PHLDA3 in osteosarcoma remain unknown. Herein, we purposed to elucidate the role of PHLDA3 in the progression and chemoresistance of osteosarcoma. According to the bioinformatics analysis, PHLDA3 expression was low in osteosarcoma patients, and low content was linked to poor prognosis. Additionally, activation of PHLDA3 suppressed osteosarcoma cell proliferation, migration, and chemoresistance, whereas PHLDA3 inhibition caused the opposite effects. Mechanistically, our data revealed that PHLDA3 negatively regulates the Akt/GSK3ß signaling cascade in osteosarcoma. Furthermore, we found that miR-19a-3p might exert its oncogenic function by inhibiting PHLDA3 expression in osteosarcoma. These results demonstrated miR-19a-3p/ PHLDA3/ Akt/GSK3ß axis has a pivotal role in osteosarcoma, and PHLDA3 is a prospective therapeutic target for treating osteosarcoma.


Subject(s)
Bone Neoplasms , MicroRNAs/genetics , Nuclear Proteins/genetics , Osteosarcoma , Proto-Oncogene Proteins c-akt/genetics , Bone Neoplasms/genetics , Bone Neoplasms/mortality , Bone Neoplasms/pathology , Cell Line, Tumor , Computational Biology , Humans , Osteosarcoma/genetics , Osteosarcoma/mortality , Osteosarcoma/pathology , Signal Transduction/genetics
9.
Cancer Manag Res ; 12: 4085-4096, 2020.
Article in English | MEDLINE | ID: mdl-32581582

ABSTRACT

BACKGROUND: Head and neck squamous cell carcinoma (HNSCC) includes a group of heterogeneous tumors with generally invasive behavior. The PI3K/AKT pathway plays an important role in the pathogenesis of HNSCC. METHODS: In the current study, we investigated the expression of two negative feedback regulators of the PI3K pathway, namely PHLDA3 and GRHL3, in 45 paired samples of HNSCC and adjacent non-cancerous tissues (ANCTs). RESULTS: While expression of GRHL3 was down-regulated in tumoral tissues compared with ANCTs by the factor 4.21, PHLDA3 expression levels were up-regulated by 5.99-times. Gender-based analysis revealed a significant down-regulation of GRHL3 gene expression level in male patients compared with the control samples and significant up-regulation of PHLDA3 gene expression level in both sexes compared with the control samples. Differences in the expressions of both genes were significant in patients aged more than 60 years, but not in the younger patients. Expression of GRHL3 was only down-regulated in patients with positive smoking history. Expression of GRHL3 was decreased in grades 2 and 3 samples compared with controls. There was a significant increase in transcript levels of PHLDA3 in stages II and III HNSCC samples compared with the controls group. ROC curve analysis indicated that the expression level of PHLDA3 could be a promising marker for the diagnosis of HNSCC patients with a sensitivity and specificity of 0.666 and 0.688, respectively. In addition, sensitivity and specificity of GRHL3 were 0.755 and 0.577, respectively. DISCUSSION: The current study indicates dysregulation of regulators of PI3K pathway in HNSCC and their potential application as putative biomarkers for this cancer.

10.
FEBS J ; 285(21): 4071-4081, 2018 11.
Article in English | MEDLINE | ID: mdl-30188605

ABSTRACT

The phlda3 gene encodes a small, 127-amino acid protein with only a PH domain, and is involved in tumor suppression, proliferation of islet ß-cells, insulin secretion, glucose tolerance, and liver injury. However, the role of phlda3 in vascular development is unknown. Here, we show that phlda3 overexpression decreases the expression levels of hemangioblast markers scl, fli1, and etsrp and intersegmental vessel (ISV) markers flk1 and cdh5, and disrupts ISV development in tg(flk1:GFP) and tg(fli1:GFP) zebrafish. Moreover, phlda3 overexpression inhibits the activation of protein kinase B (AKT) in zebrafish embryos, and the developmental defects of ISVs by phlda3 overexpression were reversed by the expression of a constitutively active form of AKT. These data suggest that phlda3 is a negative regulator of hemangioblast specification and ISV development via AKT signaling.


Subject(s)
Blood Vessels/embryology , Embryo, Nonmammalian/pathology , Hemangioblasts/pathology , Neovascularization, Pathologic , Nuclear Proteins/metabolism , Zebrafish Proteins/metabolism , Zebrafish/embryology , Animals , Embryo, Nonmammalian/metabolism , Gene Expression Regulation, Developmental , Hemangioblasts/metabolism , Nuclear Proteins/genetics , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Zebrafish/physiology , Zebrafish Proteins/genetics
11.
Oncotarget ; 7(18): 24950-61, 2016 05 03.
Article in English | MEDLINE | ID: mdl-26894863

ABSTRACT

Pancreatic neuroendocrine tumor (PanNET) is a neoplastic entity in which few prognostic factors are well-known. Here, we aimed to evaluate the prognostic significance of N-myc downstream-regulated gen-1 (NDRG-1), O6-methylguanine DNA methyltransferase (MGMT) and Pleckstrin homology-like domain family A member 3 (PHLDA-3) by immunohistochemistry (IHC) and methylation analysis in 92 patients with resected PanNET and follow-up longer than 24 months. In multivariate analyses, ki-67 and our immunohistochemistry prognostic score (IPS-based on MGMT, NDRG-1 and PHLDA-3 IHC expression) were independent prognostic factors for disease-free-survival (DFS), while age and IPS were independent prognostic factors for overall survival (OS). Our IPS could be a useful prognostic biomarker for recurrence and survival in patients following resection for PanNET.


Subject(s)
Biomarkers, Tumor/analysis , Neoplasm Recurrence, Local/pathology , Neuroendocrine Tumors/pathology , Pancreatic Neoplasms/pathology , Cell Cycle Proteins/analysis , Cell Cycle Proteins/biosynthesis , Cell Cycle Proteins/genetics , DNA Methylation , DNA Modification Methylases/analysis , DNA Modification Methylases/biosynthesis , DNA Modification Methylases/genetics , DNA Repair Enzymes/analysis , DNA Repair Enzymes/biosynthesis , DNA Repair Enzymes/genetics , Disease-Free Survival , Female , Humans , Immunohistochemistry/methods , Intracellular Signaling Peptides and Proteins/analysis , Intracellular Signaling Peptides and Proteins/biosynthesis , Intracellular Signaling Peptides and Proteins/genetics , Kaplan-Meier Estimate , Male , Middle Aged , Neoplasm Recurrence, Local/mortality , Neuroendocrine Tumors/mortality , Nuclear Proteins/analysis , Nuclear Proteins/biosynthesis , Nuclear Proteins/genetics , Pancreatic Neoplasms/mortality , Prognosis , Tumor Suppressor Proteins/analysis , Tumor Suppressor Proteins/biosynthesis , Tumor Suppressor Proteins/genetics
12.
Anticancer Res ; 35(2): 949-54, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25667479

ABSTRACT

BACKGROUND/AIM: Patients with oesophageal squamous cell carcinoma (ESCC) have a poor prognosis. Akt has been associated with malignant potential in several cancers, including ESCC. Pleckstrin homology-like domain, family A, member 3 (PHLDA3) has been identified as a direct target gene of p53 and as a potent inhibitor of Akt activation. The present study investigated the role of PHLDA3 expression and its ability to predict prognosis in patients with ESCC who did not receive induction therapy. MATERIALS AND METHODS: The intensity of PHLDA3 expression was immunohistochemically analyzed in tumor and adjacent normal tissue samples from 84 patients with ESCC, 22 who underwent endoscopic submucosal dissection and 62 who underwent thoracic oesophagectomy. RESULTS: High expression of PHLDA3 was observed in 60 (71.4%) patients and low expression in 24 (28.6%). Cancer-specific (p=0.029) and disease-free (p<0.001) survival rates were significantly lower in the PHLDA3 low-than in the PHLDA3 high-expression group, and low PHLDA3 expression was an independent predictor of postoperative recurrence (relative risk (RR)=0.38; 95% confidence interval (CI)=0.166-0.78; p=0.0074). CONCLUSION: Low PHLDA3 expression in ESCC may be predictive of tumor recurrence suggesting that Akt activation may be a therapeutic target in ESCCs.


Subject(s)
Carcinoma, Squamous Cell/genetics , Esophageal Neoplasms/genetics , Nuclear Proteins/genetics , Aged , Female , Humans , Male , Middle Aged , Prognosis
SELECTION OF CITATIONS
SEARCH DETAIL