Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
1.
Sci Rep ; 13(1): 19885, 2023 11 14.
Article in English | MEDLINE | ID: mdl-37963940

ABSTRACT

The dosage-dependent recruitment of RNA polymerase II (Pol II) at the promoters of genes related to neurodevelopment and stem cell maintenance is required for transcription by the fine-tuned expression of SET-domain-containing protein 5 (SETD5). Pol II O-GlcNAcylation by O-GlcNAc transferase (OGT) is critical for preinitiation complex formation and transcription cycling. SETD5 dysregulation has been linked to stem cell-like properties in some cancer types; however, the role of SETD5 in cancer cell stemness has not yet been determined. We here show that aberrant SETD5 overexpression induces stemness in colorectal cancer (CRC) cells. SETD5 overexpression causes the upregulation of PI3K-AKT pathway-related genes and cancer stem cell (CSC) markers such as CD133, Kruppel-like factor 4 (KLF4), and estrogen-related receptor beta (ESRRB), leading to the gain of stem cell-like phenotypes. Our findings also revealed a functional relationship between SETD5, OGT, and Pol II. OGT-catalyzed Pol II glycosylation depends on SETD5, and the SETD5-Pol II interaction weakens in OGT-depleted cells, suggesting a SETD5-OGT-Pol II interdependence. SETD5 deficiency reduces Pol II occupancy at PI3K-AKT pathway-related genes and CD133 promoters, suggesting a role for SETD5-mediated Pol II recruitment in gene regulation. Moreover, the SETD5 depletion nullified the SETD5-induced stemness of CRC cells and Pol II O-GlcNAcylation. These findings support the hypothesis that SETD5 mediates OGT-catalyzed O-GlcNAcylation of RNA Pol II, which is involved in cancer cell stemness gain via CSC marker gene upregulation.


Subject(s)
Colorectal Neoplasms , RNA Polymerase II , Humans , RNA Polymerase II/genetics , RNA Polymerase II/metabolism , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , N-Acetylglucosaminyltransferases/metabolism , Colorectal Neoplasms/genetics , Catalysis , Protein Processing, Post-Translational , Methyltransferases/metabolism
2.
Biochem Pharmacol ; 217: 115838, 2023 11.
Article in English | MEDLINE | ID: mdl-37778445

ABSTRACT

M2 type tumor-associated macrophages, an essential component of the tumor microenvironment (TME), have been proved to contribute to tumor metastasis. Dauricine (Dau) has recently received widespread attention due to its multiple targets and low price. However, the effect of Dau on macrophage polarization of TME remains unclear. In this study, we investigated the effect of Dau on prostate cancer (PCa) metastasis and specifically its correlation to macrophage polarization. Our results showed that Dau efficiently suppressed M2 polarization of macrophages induced by interleukin (IL) -4 and IL-13. Mechanistically, Dau inhibited the activity of PI3K/AKT signaling pathway, which subsequently suppressed macrophage differentiation to M2 type. Importantly, our study indicated that Dau decreased the release of chitinase 3-like protein 1 (CHI3L1) from M2 macrophages, which ultimately inhibited the M2 macrophage-mediated progression of PCa cells in vitro and in vivo. Taken together, our data demonstrated that Dau suppressed M2 polarization of macrophages via downregulation of the PI3K/AKT signaling pathway, in turn, preventing proliferation, epithelial-mesenchymal transition, migration, and invasion of PCa cells. Thus, this study reveals a previously unrecognized function of Dau in inhibition of PCa progression via intervention in M2 polarization of macrophages.


Subject(s)
Prostatic Neoplasms , Proto-Oncogene Proteins c-akt , Male , Humans , Proto-Oncogene Proteins c-akt/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Macrophages , Prostatic Neoplasms/metabolism , Tumor Microenvironment , Cell Line, Tumor
3.
Exp Mol Pathol ; 125: 104755, 2022 04.
Article in English | MEDLINE | ID: mdl-35278461

ABSTRACT

Several studies have demonstrated that B7-H4 is highly expressed in a variety of cancers and often affects tumor development. However, its role in cancer stemness and epithelial-to-mesenchymal transition (EMT) in non-small cell lung cancer (NSCLC) has not been reported. Here, we investigated the relationship between B7-H4 expression and cancer stemness and EMT by immunohistochemistry in 106 NSCLC tissues obtained from patients. The results confirmed that B7-H4 is highly expressed in NSCLC tissues and closely correlated with the expression of EMT-related proteins (Snail, Vimentin) and cancer stemness-related proteins (SOX2, SOX9, and CD44). Immunofluorescence assay indicated that B7-H4 colocalized with SOX2 and SOX9 in the nuclei of NSCLC cells. Additionally, upon knocking down B7-H4, the expression of SOX2, SOX9, and CD44, as well as of Snail and Vimentin was inhibited, whereas E-cadherin expression was enhanced in NSCLC cells. Meanwhile, inhibiting the expression of B7-H4 resulted in reduced invasion and migration ability of NSCLC cells. Mechanistically, silencing B7-H4 activated the adenosine monophosphate-activated protein kinase /mammalian target of rapamycin signaling, which in turn, negatively regulated cell proliferation, stemness, and migration. In conclusion, our results suggest that B7-H4 expression is high in NSCLC tissues, and it has an effect on EMT and cancer stemness. This further suggests that B7-H4 has a potential role in promoting the progression of NSCLC and thereby could be a potential therapeutic target in NSCLC treatment.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , V-Set Domain-Containing T-Cell Activation Inhibitor 1 , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/pathology , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , V-Set Domain-Containing T-Cell Activation Inhibitor 1/genetics , Vimentin/genetics
4.
Int J Urol ; 29(6): 578-585, 2022 06.
Article in English | MEDLINE | ID: mdl-35218089

ABSTRACT

OBJECTIVES: Tenascin C is a potential biomarker of cancer-associated fibroblasts and has been significantly associated with poor prognosis in patients with prostate cancer. However, the effects of Tenascin C in prostate cancer cell glycolysis largely remain unclear. Thus, this study aimed to investigate the Tenascin C expression in prostate cancer and its correlation to glycolysis-related protein and gene expression, clinicopathological parameters, and survival of patients. METHODS: We performed immunohistochemical staining for Tenascin C in 141 cases of primary prostate cancer. Based on public data sets, we explored the association of Tenascin C with angiogenesis-related genes, M2 macrophage-related gene, androgen receptor levels, PI3K/AKT/NF-κB pathway genes, and glycolytic enzyme expression. The glucose uptake, lactate production, and glycolytic enzyme levels were detected by glycolysis assay and western blotting. RESULTS: Our results showed that Tenascin C expression is upregulated in prostate cancer tissues compared with benign prostatic hyperplasia tissues. High Tenascin C expression in prostate cancer cells was positively associated with lymph node metastasis, advanced clinical stage, the expression of CD105, CD206, and androgen receptor levels. The Kaplan-Meier curves showed a significant association of Tenascin C expression with the patient's overall survival. Tenascin C expression was positively associated with PI3K p85, pAKT-ser308, and NF-κB p65 protein expression in prostate cancer samples. Moreover, siRNA-mediated knockdown of Tenascin C expression inhibited cell glucose uptake, lactate production, and glycolytic-enzyme expression in prostate cancer cells in vitro. CONCLUSIONS: Together, our findings suggest that Tenascin C is a prognostic marker for patients with prostate cancer and that its effects might be mediated via regulation of the glycolysis process of prostate cancer cells.


Subject(s)
Prostatic Neoplasms , Tenascin/metabolism , Cell Line, Tumor , Glucose , Glycolysis , Humans , Lactates , Male , NF-kappa B/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Prostatic Neoplasms/pathology , Receptors, Androgen/metabolism , Tenascin/genetics
5.
Am J Pathol ; 192(4): 712-721, 2022 04.
Article in English | MEDLINE | ID: mdl-35063407

ABSTRACT

Although glycolysis plays a pivotal role in breast cancer stem-like cell (BCSC) reprogramming, the molecular mechanisms that couple glycolysis to cancer stem-like cells remain unclear. SETD5 is a previously uncharacterized member of the histone lysine methyltransferase family. The goal of this study was to explore the mechanisms underlying the promotion of stem-like and glycolysis activation traits by SETD5. Previous studies have shown that overexpression of SETD5 in breast cancer tissues is associated positively with progression. The present study showed that SETD5 expression was enriched in BCSCs. Down-regulation of SETD5 significantly decreased BCSC properties and glycolysis in vitro and in vivo. Interestingly, SETD5 and glycolytic enzymes were accumulated in the central hypoxic regions of subcutaneous tumor tissues. Bioinformatic analysis predicted SETD5 binding to E1A binding protein p300 (EP300), and subsequently to hypoxia-inducible factor 1α (HIF-1α). The mechanistic study found that SETD5 is an upstream effector of EP300/HIF-1α. SETD5 knockdown reduced the expression of HIF-1α, hexokinase-2, and 6-phosphofructo-2-kinase in the nucleus after treatment with cobalt chloride, a chemical hypoxia mimetic agent that activates HIF-1α to accumulate in the nucleus. Therefore, SETD5 is required for glycolysis in BCSCs through binding to EP300/HIF-1α and could be a potential therapeutic target for breast cancer patients.


Subject(s)
Breast Neoplasms , Methyltransferases , Neoplastic Stem Cells , Breast Neoplasms/pathology , Cell Line, Tumor , Down-Regulation , Female , Glycolysis/physiology , Humans , Hypoxia/pathology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Methyltransferases/metabolism , Neoplastic Stem Cells/pathology
6.
Exp Mol Pathol ; 121: 104665, 2021 08.
Article in English | MEDLINE | ID: mdl-34216584

ABSTRACT

Abnormal metabolism and uncontrolled angiogenesis are two important characteristics of malignant tumors. Although HBXIP is known to be associated with a poor prognosis for bladder cancer (BC), its effects on glycolysis and angiogenesis in BC have not been investigated. BC prognosis and relative gene expression of HBXIP were analyzed using the GEPIA, UALCAN, and STRING databases. BC cell angiogenesis and glycolysis were assessed by vasculogenic mimicry and glycolysis assay. Human umbilical vein endothelial cell (HUVEC) viability, migration, and angiogenesis were assessed by CCK8, transwell, wound healing, and tube formation assays. The results showed that HBXIP was highly expressed in BC tissues and cells. Knockdown of HBXIP expression decreased the levels of glucose uptake, lactate production, and glycolytic enzyme expression in BC cells, and decreased cell viability and migration of HUVECs. Additionally, silencing HBXIP reduced the total length of tubes and number of intersections, and EPO and VEGF protein expression in BC cells and HUVECs. Furthermore, knockdown of HBXIP expression reversed cell viability, migration, tube formation, and vasculogenic mimicry under high glucose and lactate conditions. Mechanistically, silencing of HBXIP reduced the protein expression levels of pAKT-ser473 and pmTOR, and inhibition of HBXIP, AKT, and mTOR expression decreased glycolytic enzyme protein expression. Our findings suggest that HBXIP reduces glycolysis in BC cells via regulation of AKT/mTOR signaling, thereby blocking BC angiogenesis. Collectively, this study provides a potential strategy to target HBXIP and AKT/mTOR for regulating glycolysis progression concurrently with anti-angiogenesis effects, and thereby develop novel therapeutics for the treatment of BC.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Glycolysis , Neovascularization, Pathologic/pathology , Proto-Oncogene Proteins c-akt/metabolism , TOR Serine-Threonine Kinases/metabolism , Urinary Bladder Neoplasms/blood supply , Adaptor Proteins, Signal Transducing/genetics , Cell Movement , Cell Proliferation , Cell Survival , Human Umbilical Vein Endothelial Cells , Humans , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/genetics , Signal Transduction , TOR Serine-Threonine Kinases/genetics , Tumor Cells, Cultured , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/metabolism , Urinary Bladder Neoplasms/pathology
7.
Pathol Res Pract ; 218: 153323, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33429327

ABSTRACT

B7-H4 is a unique negative regulator of T cells that is typically significantly overexpressed in various carcinomas and is associated with poor prognosis. However, the effects of B7-H4 expression on epithelial-mesenchymal transition (EMT) and cancer stemness of colorectal cancer (CRC) are not entirely clear. In the present study, we used tissue samples from 98 patients with CRC and CRC cell lines to determine the clinicopathological significance of B7-H4 in CRC and its effects on CRC stemness. We performed immunohistochemical staining; immunofluorescence imaging; western blotting; and tumor sphere formation, wound healing, transwell migration, and in vivo tumorigenesis assays. B7-H4 expression was upregulated in CRC tissues and was associated with lymph node metastasis, distant metastasis, clinical stage, a shorter overall survival rate, and disease-free survival rate. Cox regression analyses indicated that B7-H4 is an independent poor prognostic factor for CRC. In addition, B7-H4 expression was correlated with the expression of EMT-related proteins and cancer stemness-related proteins. Moreover, immunohistochemical and immunofluorescence analyses revealed that B7-H4 was correlated with CD133 and CD44 expression levels in both CRC tissues and HT29 and HCT116 cell lines. Conversely, B7-H4 knockdown downregulated the expression of EMT- and cancer stemness-related proteins, while inhibiting tumor spheroid formation, cell migration, and invasion of CRC cell lines. These results indicate that B7-H4 can promote EMT and may be a novel stem cell marker, suggesting its potential as a prognostic biomarker for CRC.


Subject(s)
Biomarkers, Tumor/metabolism , Colorectal Neoplasms/metabolism , Epithelial-Mesenchymal Transition , Neoplastic Stem Cells/metabolism , V-Set Domain-Containing T-Cell Activation Inhibitor 1/metabolism , Animals , Biomarkers, Tumor/genetics , Colorectal Neoplasms/genetics , Colorectal Neoplasms/mortality , Colorectal Neoplasms/pathology , Disease-Free Survival , Gene Expression Regulation, Neoplastic , HCT116 Cells , HT29 Cells , Humans , Mice, Inbred BALB C , Mice, Nude , Neoplasm Metastasis , Neoplasm Staging , Neoplastic Stem Cells/pathology , Retrospective Studies , Signal Transduction , V-Set Domain-Containing T-Cell Activation Inhibitor 1/genetics
8.
J Cell Mol Med ; 25(4): 2110-2120, 2021 02.
Article in English | MEDLINE | ID: mdl-33314691

ABSTRACT

Leucine zipper-EF-hand-containing transmembrane protein 1 (LETM1) is a mitochondrial inner membrane protein that is highly expressed in various cancers. Although LETM1 is known to be associated with poor prognosis in colorectal cancer (CRC), its roles in autophagic cell death in CRC have not been explored. In this study, we examined the mechanisms through which LETM1 mediates autophagy in CRC. Our results showed that LETM1 was highly expressed in CRC tissues and that down-regulation of LETM1 inhibited cell proliferation and induced S-phase arrest. LETM1 silencing also suppressed cancer stem cell-like properties and induced autophagy in CRC cells. Additionally, the autophagy inhibitor 3-methyladenine reversed the inhibitory effects of LETM1 silencing on proliferation and stemness, whereas the autophagy activator rapamycin had the opposite effects. Mechanistically, suppression of LETM1 increased the levels of reactive oxygen species (ROS) and mitochondrial ROS by regulation of SOD2, which in turn activated AMP-activated protein kinase (AMPK)/mammalian target of rapamycin (mTOR), initiated autophagy, and inhibited proliferation and stemness. Our findings suggest that silencing LETM1 induced autophagy in CRC cells by triggering ROS-mediated AMPK/mTOR signalling, thus blocking CRC progression, which will enhance our understanding of the molecular mechanism of LETM1 in CRC.


Subject(s)
Autophagy , Calcium-Binding Proteins/genetics , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , Membrane Proteins/genetics , Neoplastic Stem Cells/metabolism , Reactive Oxygen Species/metabolism , Biomarkers , Calcium-Binding Proteins/metabolism , Cell Line, Tumor , Cell Proliferation , Colorectal Neoplasms/pathology , Gene Silencing , Humans , Immunophenotyping , Membrane Proteins/metabolism , Mitochondria/genetics , Mitochondria/metabolism , Models, Biological , Signal Transduction , TOR Serine-Threonine Kinases/metabolism
9.
Am J Pathol ; 191(1): 180-193, 2021 01.
Article in English | MEDLINE | ID: mdl-33129761

ABSTRACT

Several studies have confirmed the function of Su(var)3-9, Enhancer of zeste, and Trithorax (SET) domain-containing 5 (SETD5) in post-translational modifications of nonhistone proteins. Mutation of the SETD5 gene has been implicated in the progression of many human cancers, such as breast cancer (BC), but its functional role in BC progression is still unknown. The current article investigates the clinical significance and the functional role of SETD5 in BC. Our studies show that SETD5 expression in BC was related to poor clinical outcomes, including lymph node metastasis and advanced clinical stage. SETD5 expression positively correlated with tumor-associated macrophages. SETD5 was an independent predictor of poor overall survival in BC. Furthermore, these studies show that down-regulation of SETD5 significantly decreased BC cell proliferation, metastasis, and angiogenesis, and increased apoptosis of BC cells. The mechanistic analysis showed that SETD5 contributes BC progression by interacting with AKT1 pathway. Also, in vivo experiments show that blocking of SETD5 expression significantly inhibited tumor growth and pulmonary metastasis of BC cells. These findings indicate that SETD5 is a potential prognosis marker and facilitates tumor progression of BC.


Subject(s)
Breast Neoplasms/pathology , Carcinoma, Ductal, Breast/pathology , Gene Expression Regulation, Neoplastic/physiology , Methyltransferases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Animals , Biomarkers, Tumor/metabolism , Breast Neoplasms/metabolism , Carcinoma, Ductal, Breast/metabolism , Cell Proliferation/physiology , Disease Progression , Female , Heterografts , Humans , Lung Neoplasms/secondary , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Invasiveness/pathology , Signal Transduction/physiology , Up-Regulation
10.
Pathol Res Pract ; 216(12): 153258, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33130499

ABSTRACT

SETD8 is a lysine methyltransferase containing an SET domain and has been reported to regulate various biological processes, including carcinogenesis. However, its prognostic value and mechanisms of action in non-small cell lung cancer (NSCLC) have not been extensively studied. Here, we assessed SETD8 expression and its relationship with clinicopathological parameters, cancer stemness proteins, and cell cycle-regulating proteins in NSCLC. SETD8 expression in NSCLC tissues was correlated with primary tumor stage, lymph node metastases, and clinical stage. Moreover, SETD8 was an independent predictor of poor overall survival in NSCLC. A Cox regression analysis showed that SETD8 was a potential biomarker of unfavorable clinical outcomes in patients with NSCLC. SETD8 overexpression was associated with cancer stemness-related genes and cell cycle-related genes in NSCLC tissue samples. SETD8 silencing significantly reduced the expression of cancer stemness-associated genes (CD44, LGR5, and SOX2) and inhibited NSCLC cell proliferation, spheroid formation, invasion, and migration. Our findings demonstrate that SETD8 may be a novel cancer stemness-associated protein and a potential prognostic biomarker in NSCLC.


Subject(s)
Biomarkers, Tumor/metabolism , Carcinoma, Non-Small-Cell Lung/enzymology , Histone-Lysine N-Methyltransferase/metabolism , Lung Neoplasms/enzymology , Neoplastic Stem Cells/enzymology , A549 Cells , Aged , Biomarkers, Tumor/genetics , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Cell Movement , Cell Proliferation , Female , Gene Expression Regulation, Neoplastic , Histone-Lysine N-Methyltransferase/genetics , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Male , Middle Aged , Neoplasm Invasiveness , Neoplastic Stem Cells/pathology , Phenotype , Signal Transduction
11.
Exp Mol Pathol ; 117: 104560, 2020 12.
Article in English | MEDLINE | ID: mdl-33127342

ABSTRACT

SETD8 is a lysine methyltransferase containing an SET domain, which is involved in the carcinogenesis of many cancer types through monomethylation of the histone H4 lysine 20. However, its prognostic value and underlying mechanisms in gastric adenocarcinoma (GA) have not been extensively studied. Here, we assessed SETD8 expression and its relationship with clinicopathological parameters, cancer stemness-related proteins, cell cycle-related proteins, and PI3K/Akt pathway proteins in GA. SETD8 expression in GA tissues was correlated with the primary tumor stage, lymph node metastasis, tumor size, gross type, and clinical stage. SETD8 was an independent predictor of poor overall survival of patients with GA. Cox regression analysis showed that SETD8 is a potential biomarker of unfavorable clinical outcomes in patients with GA. Moreover, SETD8 overexpression was associated with cancer stemness-related genes, cell cycle-related genes, and PI3K/Akt/NF-κB pathway genes in clinical GA tissue samples. SETD8 silencing downregulated the expression of cancer stemness-associated genes (LSD1 and SOX2) and inhibited GA cell proliferation, spheroid formation, invasion, and migration. Additionally, LY294002 significantly reduced the expression of SETD8, pAkt-Ser473, pPI3K-p85, and NFκB-p65 in MKN74 and MKN28 cells. SETD8 may be a novel cancer stemness-associated protein and potential prognostic biomarker in GA.


Subject(s)
Adenocarcinoma/genetics , Histone Demethylases/genetics , Histone-Lysine N-Methyltransferase/genetics , SOXB1 Transcription Factors/genetics , Stomach Neoplasms/genetics , Adenocarcinoma/pathology , Aged , Biomarkers, Tumor/genetics , Carcinogenesis/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Chromones/pharmacology , Female , Gene Expression Regulation, Neoplastic/genetics , Histone-Lysine N-Methyltransferase/antagonists & inhibitors , Humans , Male , Middle Aged , Morpholines/pharmacology , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Oncogene Protein v-akt/genetics , Prognosis , Stomach Neoplasms/pathology
12.
J Tissue Eng ; 11: 2041731420933407, 2020.
Article in English | MEDLINE | ID: mdl-32637062

ABSTRACT

Cancer stem cells have been shown to be important in tumorigenesis processes, such as tumor growth, metastasis, and recurrence. As such, many three-dimensional models have been developed to establish an ex vivo microenvironment that cancer stem cells experience under in vivo conditions. Cancer stem cells propagating in three-dimensional culture systems show physiologically related signaling pathway profiles, gene expression, cell-matrix and cell-cell interactions, and drug resistance that reflect at least some of the tumor properties seen in vivo. Herein, we discussed the presently available Cancer stem cell three-dimensional culture models that use biomaterials and engineering tools and the biological implications of these models compared to the conventional ones.

13.
Diagn Pathol ; 15(1): 60, 2020 May 19.
Article in English | MEDLINE | ID: mdl-32430068

ABSTRACT

BACKGROUND: Glioma-associated oncogene homolog 1 (Gli1), affects the progression and the stemness characteristics of malignant carcinoma. The aim of the present study was to identify the relation between Glioma-associated oncogene homolog 1 (Gli1) and stemness and determine its clinical significance in gastric adenocarcinoma (GA). We investigated Gli1 expression and its correlation with other stemness-associated proteins in 169 GA samples and 5 GA cell lines. METHODS: To elucidate the role of Gli1 in the clinicopathological significance and stemness of GA, tissues samples from 169 GA patients were collected for immunohistochemistry (IHC). Additionally, MKN74, MKN28, NCI-N87, SNU638, AGS cells were collected for western blotting, MKN28 cells were collected for spheroid formation assay. RESULTS: Results showed that Gli1 expression was closely related to tumor grade, primary tumor (pT) stage, distant metastasis, clinical stage, gross type, microvessel density, and shorter overall survival (OS). Cox regression analysis verified that Gli1 was an independent prognostic factor for OS. Furthermore, Gli1 expression correlated with the expression of stemness-related genes, CD44, LSD1, and Sox9. Gli1 inhibitor GANT61 significantly decreased the expression of CD44 and LSD1, and spheroid formation ability of the MKN28 cells. CONCLUSIONS: In conclusion, Gli1 may be a poor prognostic indicator and a potential cancer stemness-related protein in GA.


Subject(s)
Adenocarcinoma/pathology , Neoplastic Stem Cells/pathology , Stomach Neoplasms/pathology , Zinc Finger Protein GLI1/metabolism , Aged , Biomarkers, Tumor/metabolism , Female , Humans , Male , Middle Aged , Prognosis
14.
Cancer Cell Int ; 20: 122, 2020.
Article in English | MEDLINE | ID: mdl-32322169

ABSTRACT

BACKGROUND: Tenascin-C (TNC) is an extracellular matrix protein that is widely expressed in the stromal fibroblasts of various cancers. However, the roles of TNC in colorectal cancer (CRC) cells remain unclear. METHODS: The expression of TNC, cancer stem cell-like (CSC) and cell cycle markers, and Hedgehog (HH) signaling pathway genes were assessed in 100 paraffin embedded clinical CRC patient tissues using immunohistochemistry. The interaction between TNC and CSC marker or HH related genes in CRC cells were detected by immunofluorescence. Cell cycle distribution was measured by flow cytometry. Migration and invasion were evaluated by transwell assays. The expressions of TNC, CSC marker, and HH related proteins were analyzed by western blot. RESULTS: TNC expression was markedly upregulated in CRC tissues, and was associated with worse clinical outcomes. TNC overexpression was positively associated with CSC marker LSD1, cell cycle markers CDK4 and p16, and HH signaling pathway related genes SMO and GLI1 in clinical CRC tissue samples. TNC silencing downregulated the expression of the CSC marker LSD1, and the proliferation, migration, and invasion of CRC cells. Interestingly, the GLI1 inhibitor GANT61 strongly inhibited the expression of TNC in CRC cells. CONCLUSIONS: TNC may drive tumor progression and is involved in CSC properties via the HH signaling pathway. TNC has potential value in the evaluation of poor prognosis in CRC.

15.
Cancer Manag Res ; 12: 1649-1660, 2020.
Article in English | MEDLINE | ID: mdl-32184668

ABSTRACT

PURPOSE: The leucine zipper-EF-hand containing transmembrane protein 1 (LETM1) is a mitochondrial protein that has been associated with the occurrence and development of malignant tumors. Previous studies have shown that LETM1 expression is increased in several types of human cancer and is associated with a poor clinical outcome. However, the role of LETM1 in prostate cancer (PCa) has not yet been determined. In this study, we investigated the clinicopathological significance of LETM1 expression and its role in PCa progression. METHODS: We assessed the expression of LETM1 and genes related to cancer stemness, epithelial-mesenchymal transition (EMT), cell cycle, and PI3K/Akt signaling in 133 paraffin-embedded PCa tissue samples and cancer cells by using immunohistochemistry, immunofluorescence, and Western blotting. RESULTS: LETM1 expression was significantly increased in PCa, and it was positively correlated with Gleason score, pathologic tumor (pT) stage, clinical stage, and high microvessel density. Survival analysis showed that patients with PCa with a high level of LETM1 expression exhibited a low overall survival. Cox regression analysis indicated that LETM1 is an independent poor prognostic PCa factor. Additionally, the expression of LETM1 was correlated with cancer cell stemness-associated genes, EMT-related genes, cell cycle regulatory genes, and PI3K/Akt signaling gene expression in PCa. Furthermore, knocking down LETM1 expression down-regulated the expression of stemness-related proteins, while inhibiting tumor spheroid formation, EMT-like changes, cell proliferation, migration, and invasion in PCa cells. Importantly, the PI3K inhibitor LY294002 strongly inhibited the expression of LETM1, pPI3K-p85, and pAkt (Thr308, Ser473) in PCa cells. CONCLUSION: These results indicate that LETM1 expression is associated with cancer cell stemness, promotes EMT-like changes and cell proliferation and is a potential prognostic biomarker for PCa.

16.
Exp Mol Pathol ; 114: 104406, 2020 06.
Article in English | MEDLINE | ID: mdl-32088189

ABSTRACT

B7-H4 is a member of B7 family which regulates immune responses by delivering costimulatory signals. However, it negatively regulates T cell-mediated immunity and may play an important role in tumor immune evasion. Although several studies have been reported that expression of B7-H4 is elevated in the several types of human cancer with a poor clinical outcome, its clinical significance in the prostate cancer (PCa) has not been well studied. In this study, we investigated the clinical significance of B7-H4 in human PCa and determined if B7-H4 expression is associated with the cancer cell stemness in PCa. Our studies show that expression of B7-H4 is correlated with the pathologic tumor (pT) stage and the clinical stage of PCa. The Kaplan-Meier survival analysis revealed that PCa patients with high expression of B7-H4 exhibits a shorter overall survival (OS) rate. Univariate and multivariate Cox regression analysis indicated that B7-H4 is an independent poor prognostic factor of PCa. In addition, the expression of B7-H4 is correlated with the cancer cell stemness associated genes expression in PCa. Further, our studies show that B7-H4 regulates cancer cell stemness associated genes expression and effects on the cell cycle and PI3K/Akt signaling related genes expression in PCa. These results indicate that B7-H4 expression is associated with cancer cell stemness, and B7-H4 is a potential prognostic biomarker and a therapeutic target of PCa.


Subject(s)
Biomarkers, Tumor/genetics , Prognosis , Prostatic Neoplasms/genetics , V-Set Domain-Containing T-Cell Activation Inhibitor 1/genetics , Aged , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Progression-Free Survival , Prostatic Neoplasms/epidemiology , Prostatic Neoplasms/pathology
17.
Exp Cell Res ; 389(1): 111861, 2020 04 01.
Article in English | MEDLINE | ID: mdl-31981592

ABSTRACT

SET domain-containing 5 (SETD5) is an uncharacterized member of the protein lysine methyltransferase family. Although it was reported that SETD5 gene mutations are associated with the several types of human cancer, its functional role in esophageal squamous cell carcinoma (ESCC) progression has not been fully elucidated. In the present study, we used tissue samples from 147 patients with ESCC and ESCC cell lines to determine the clinicopathological significance of SETD5 in ESCC and its effects on ESCC stemness. We performed immunohistochemical staining, immunofluorescence imaging, and tumor sphere formation, colony formation, flow cytometry, wound healing, Transwell, and western blotting assays. SETD5 expression was upregulated in ESCC tissue and associated with primary tumor (pT) stage, clinical stage, lymph node metastasis, shorter overall survival rate, and disease-free survival rate. Cox regression analyses indicated that SETD5 is an independent poor prognostic factor of ESCC. In addition, SETD5 expression was correlated with cancer stemness-related protein, hypoxia-inducible factor-1α (HIF-1α), and CD68 expression. Moreover, immunofluorescence analysis revealed that SETD5 was co-localized with CD44 and SOX2 in TE10 and TE11 cells and that exposing cells to cobalt chloride increased HIF-1α, SETD5, and stemness-related protein expression in a time-dependent manner. Furthermore, SETD5 expression was significantly correlated with the expression of cell cycle-related genes and PI3K/Akt signaling pathway-related proteins. Finally, knocking down SETD5 downregulated the expression of stemness-related and PI3K/Akt signaling pathway proteins, while inhibiting tumor spheroid formation, cell proliferation, migration, and invasion in ESCC cells. These results indicate that SETD5 expression is associated with cancer stemness and that SETD5 is a potential prognostic biomarker and therapeutic target for ESCC.


Subject(s)
Esophageal Neoplasms/diagnosis , Esophageal Neoplasms/pathology , Esophageal Squamous Cell Carcinoma/diagnosis , Esophageal Squamous Cell Carcinoma/pathology , Methyltransferases/physiology , Neoplastic Stem Cells/pathology , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/physiology , Disease Progression , Esophageal Neoplasms/genetics , Esophageal Squamous Cell Carcinoma/genetics , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Male , Middle Aged , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Prognosis , RNA, Small Interfering/pharmacology , Tumor Cells, Cultured
18.
J Neuroimmunol ; 339: 577123, 2020 02 15.
Article in English | MEDLINE | ID: mdl-31838278

ABSTRACT

Neuroinflammation is an important pathological process of neurodegenerative diseases, and microglial contributes to chronic inflammation and neuronal loss in progressive neurodegenerative. Therefore, regulating the inflammatory response of microglia could lead to the discovery of promising treatments for neurodegenerative diseases. In this study, we investigated the effects of the nutmeg plant seed extract, macelignan, on the inflammatory response of microglia and neuronal cell survival. We detected NO and iNOS using the Griess test and Western blotting. We measured phosphoinositide 3 kinase (PI3K)/Akt expression by Western blotting. The release of NO and inflammatory cytokines and the expression of iNOS decreased in a concentration-dependent manner, with an increase in macelignan concentration. PI3K/Akt phosphorylation levels decreased in a dose-dependent manner in lipopolysaccharide (LPS)-activated microglial cells after exposure to macelignan. We also demonstrated that macelignan improved HT22 cell viability, following exposure to a microglial-conditioned medium. Furthermore, macelignan inhibited microglial cell near neurons treated with a hypoxic conditioned medium. Finally, macelignan treatment reduced the expression of p27 and cyclin D1 in neurons cultured in an LPS-activated microglia-conditioned medium. Therefore, these results imply that macelignan can inhibit the inflammatory response of microglia and regulate neuronal survival through the PI3K/Akt pathway.


Subject(s)
Inflammation Mediators/antagonists & inhibitors , Lignans/pharmacology , Microglia/drug effects , Neurons/drug effects , Animals , Cell Line , Cell Survival/drug effects , Cell Survival/physiology , Dose-Response Relationship, Drug , Humans , Inflammation Mediators/metabolism , Mice , Microglia/metabolism , Microglia/pathology , Neurons/metabolism , Neurons/pathology
19.
Exp Mol Pathol ; 112: 104333, 2020 02.
Article in English | MEDLINE | ID: mdl-31705880

ABSTRACT

Leucine zipper-EF-hand containing transmembrane protein 1 (LETM1) is closely linked to the occurrence and development of many malignant tumors. Many studies have reported that enhanced expression of LETM1 in several types of human cancers was associated with poor clinical outcomes; however, its clinical significance in gastric adenocarcinoma (GA) has not been elucidated. In this study, we assessed the expression of LETM1 along with the genes related to cancer stemness, cell cycle, and PI3K/Akt signaling in 189 paraffin-embedded GA tissue samples and GA-derived cell lines using immunohistochemistry (IHC), western blotting, and immunofluorescence. Our results showed that the expression of LETM1 was strongly correlated with the tumor grade, primary tumor (pT) stage, lymph node metastasis, clinical stage, and tumor gross type of GA. The Kaplan-Meier survival analysis revealed that patients with GA with high expression of LETM1 exhibit a shorter overall survival (OS) rate. Univariate and multivariate Cox regression analysis indicated that LETM1 is an independent poor prognostic factor of GA. Significantly, LETM1 expression was positively correlated with the expression of cancer stemness-related genes such as CD44 and LGR5 and expression of cell-cycle related gene, cyclin D1. Further, the expression of proteins involved in PI3K/Akt signaling pathway, such as pPI3K-p85 and pAkt-Thr308, was also increased. Additionally, small interfering RNA (esiRNA)-mediated silencing of LETM1 expression in GA-derived cell lines MKN28 and MKN74 strongly inhibited the expression of stemness and cell cycle-related proteins, suppressed cancer cell spheroid formation, migration and invasion ability, and affected cell cycle distribution. Furthermore, the GA-derived cell line AGS exhibited enhanced expression of LETM1, CD44, LGR5, and HIF1-α under hypoxic conditions. Lastly, we blocked PI3K expression using an inhibitor LY294002, which downregulated the expression of LETM1, pPI3K-p85, and pAkt-Thr308. Taken together, our results demonstrate that LETM1 regulates cell proliferation and promotes tumorigenicity of GA, and its overexpression is associated with the poor progression of GA. Therefore, LETM1 could serve as a potential prognostic biomarker and a therapeutic target for the better clinical management of GA.


Subject(s)
Adenocarcinoma/genetics , Biomarkers, Tumor/genetics , Calcium-Binding Proteins/genetics , Membrane Proteins/genetics , Stomach Neoplasms/genetics , Adenocarcinoma/pathology , Aged , Cell Proliferation/genetics , Cyclin D1/genetics , Disease-Free Survival , Female , Gene Expression Regulation, Neoplastic/genetics , Humans , Hyaluronan Receptors/genetics , Kaplan-Meier Estimate , Male , Middle Aged , Oncogene Protein v-akt/genetics , Phosphatidylinositol 3-Kinases/genetics , Prognosis , Stomach Neoplasms/pathology
20.
BMC Cancer ; 19(1): 898, 2019 Sep 09.
Article in English | MEDLINE | ID: mdl-31500591

ABSTRACT

BACKGROUND: Although the leucine zipper-EF-hand-containing transmembrane protein 1 (LETM1) is one of the mitochondrial inner membrane proteins that is involved in cancer prognosis in various tumors, LETM1 as a biomarker for prognostic evaluation of non-small cell lung carcinoma (NSCLC) has not been well studied. METHODS: To address this issue, we used 75 cases NSCLC, 20 cases adjacent normal lung tissues and NSCLC cell lines. We performed immunohistochemistry staining and western blot analysis as well as immunofluorescence imaging. RESULTS: Our studies show that expression of LETM1 is significantly correlated with the lymph node metastasis (p = 0.003) and the clinical stage (p = 0.005) of NSCLC. The Kaplan-Meier survival analysis revealed that NSCLC patients with positive expression of LETM1 exhibits a shorter overall survival (OS) rate (p = 0.005). The univariate and multivariate Cox regression analysis indicated that LETM1 is a independent poor prognostic marker of NSCLC. In addition, the LETM1 expression is correlated with cancer stemness-related gene LGR5 (p < 0.001) and HIF1α expression (p < 0.001), but not with others. Moreover, LETM1 expression was associated with the expression of cyclin D1 (p = 0.003), p27 (p = 0.001), pPI3K(p85) (p = 0.025), and pAkt-Thr308 (p = 0.004). Further, our studies show in LETM1-positive NSCLC tissues the microvessel density was significantly higher than in the negative ones (p = 0.024). CONCLUSION: These results indicate that LETM1 is a potential prognostic biomarker of NSCLC.


Subject(s)
Calcium-Binding Proteins/analysis , Carcinoma, Non-Small-Cell Lung/metabolism , Lung Neoplasms/metabolism , Membrane Proteins/analysis , Aged , Biomarkers, Tumor/analysis , Calcium-Binding Proteins/genetics , Carcinoma, Non-Small-Cell Lung/diagnosis , Carcinoma, Non-Small-Cell Lung/genetics , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/analysis , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Immunohistochemistry , Kaplan-Meier Estimate , Lung Neoplasms/diagnosis , Lung Neoplasms/genetics , Male , Membrane Proteins/genetics , Middle Aged , Prognosis , Receptors, G-Protein-Coupled/analysis , Receptors, G-Protein-Coupled/genetics
SELECTION OF CITATIONS
SEARCH DETAIL