1
|
Singh B, Cui K, Eisa-Beygi S, Zhu B, Cowan DB, Shi J, Wang DZ, Liu Z, Bischoff J, Chen H. Elucidating the crosstalk between endothelial-to-mesenchymal transition (EndoMT) and endothelial autophagy in the pathogenesis of atherosclerosis. Vascul Pharmacol 2024; 155:107368. [PMID: 38548093 PMCID: PMC11303600 DOI: 10.1016/j.vph.2024.107368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 03/07/2024] [Accepted: 03/25/2024] [Indexed: 04/26/2024]
Abstract
Atherosclerosis, a chronic systemic inflammatory condition, is implicated in most cardiovascular ischemic events. The pathophysiology of atherosclerosis involves various cell types and associated processes, including endothelial cell activation, monocyte recruitment, smooth muscle cell migration, involvement of macrophages and foam cells, and instability of the extracellular matrix. The process of endothelial-to-mesenchymal transition (EndoMT) has recently emerged as a pivotal process in mediating vascular inflammation associated with atherosclerosis. This transition occurs gradually, with a significant portion of endothelial cells adopting an intermediate state, characterized by a partial loss of endothelial-specific gene expression and the acquisition of "mesenchymal" traits. Consequently, this shift disrupts endothelial cell junctions, increases vascular permeability, and exacerbates inflammation, creating a self-perpetuating cycle that drives atherosclerotic progression. While endothelial cell dysfunction initiates the development of atherosclerosis, autophagy, a cellular catabolic process designed to safeguard cells by recycling intracellular molecules, is believed to exert a significant role in plaque development. Identifying the pathological mechanisms and molecular mediators of EndoMT underpinning endothelial autophagy, may be of clinical relevance. Here, we offer new insights into the underlying biology of atherosclerosis and present potential molecular mechanisms of atherosclerotic resistance and highlight potential therapeutic targets.
Collapse
Affiliation(s)
- Bandana Singh
- Vascular Biology Program, Department of Surgery, Harvard Medical School, Boston Children's Hospital, Boston, MA, USA
| | - Kui Cui
- Vascular Biology Program, Department of Surgery, Harvard Medical School, Boston Children's Hospital, Boston, MA, USA
| | - Shahram Eisa-Beygi
- Vascular Biology Program, Department of Surgery, Harvard Medical School, Boston Children's Hospital, Boston, MA, USA
| | - Bo Zhu
- Vascular Biology Program, Department of Surgery, Harvard Medical School, Boston Children's Hospital, Boston, MA, USA
| | - Douglas B Cowan
- Vascular Biology Program, Department of Surgery, Harvard Medical School, Boston Children's Hospital, Boston, MA, USA
| | - Jinjun Shi
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Da-Zhi Wang
- Center for Regenerative Medicine, University of South Florida Health Heart Institute, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Zhenguo Liu
- Division of Cardiovascular Medicine, Department of Medicine, University of Missouri School of Medicine, Columbia, MO, USA
| | - Joyce Bischoff
- Vascular Biology Program, Department of Surgery, Harvard Medical School, Boston Children's Hospital, Boston, MA, USA
| | - Hong Chen
- Vascular Biology Program, Department of Surgery, Harvard Medical School, Boston Children's Hospital, Boston, MA, USA.
| |
Collapse
|
2
|
Golgi Phosphoprotein 3 Represents a Novel Tumor Marker for Gastric and Colorectal Cancers. DISEASE MARKERS 2021; 2021:8880282. [PMID: 33680216 PMCID: PMC7929655 DOI: 10.1155/2021/8880282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 02/11/2021] [Accepted: 02/14/2021] [Indexed: 12/24/2022]
Abstract
Background Early diagnosis is very important for the clinical treatment of gastric cancer (GC) and colorectal cancer (CRC). We aimed to detect Golgi phosphoprotein 3 (GOLPH3) and evaluate its diagnostic value. Materials and Methods Serum concentrations of GOLPH3 were detected by ELISA in 136 CRC patients, 102 GC patients, and 50 healthy controls at the Second Affiliated Hospital of Fujian Medical University from June 2016 to December 2019. Serum concentrations of CEA and CA19-9 were detected by ECLIA. Results Serum concentrations of GOLPH3, CEA, and CA19-9 were higher in GC and CRC patients than in healthy controls (P < 0.001). Serum GOLPH3 concentrations were increased in GC and CRC patients with tumors greater than 5 cm, poor differentiation, greater depth of tumor invasion, and increased lymphatic and distant metastases (P < 0.05). In the GC and CRC groups, the AUCs of GOLPH3 were higher than those of CEA and CA19-9 (P < 0.05), while the AUCs of the marker combination were higher than those of GOLPH3 (P < 0.05), and postoperative serum GOLPH3 levels were lower than preoperative levels (P < 0.001). Serum GOLPH3 concentrations in CRC patients correlated positively with CEA and CA19-9 concentrations (P < 0.05). Conclusion Serum GOLPH3 concentrations in GC and CRC patients are related to TNM stage. GOLPH3 may represent a novel biomarker for the diagnosis of GC and CRC. The combination of serum GOLPH3, CEA, and CA19-9 concentrations can improve diagnostic efficiency for GC and CRC. GOLPH3 is expected to become an indicator for the early diagnosis and evaluation of surgical effects.
Collapse
|
3
|
Arriagada C, Cavieres VA, Luchsinger C, González AE, Muñoz VC, Cancino J, Burgos PV, Mardones GA. GOLPH3 Regulates EGFR in T98G Glioblastoma Cells by Modulating Its Glycosylation and Ubiquitylation. Int J Mol Sci 2020; 21:E8880. [PMID: 33238647 PMCID: PMC7700535 DOI: 10.3390/ijms21228880] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 11/02/2020] [Accepted: 11/06/2020] [Indexed: 12/13/2022] Open
Abstract
Protein trafficking is altered when normal cells acquire a tumor phenotype. A key subcellular compartment in regulating protein trafficking is the Golgi apparatus, but its role in carcinogenesis is still not well defined. Golgi phosphoprotein 3 (GOLPH3), a peripheral membrane protein mostly localized at the trans-Golgi network, is overexpressed in several tumor types including glioblastoma multiforme (GBM), the most lethal primary brain tumor. Moreover, GOLPH3 is currently considered an oncoprotein, however its precise function in GBM is not fully understood. Here, we analyzed in T98G cells of GBM, which express high levels of epidermal growth factor receptor (EGFR), the effect of stable RNAi-mediated knockdown of GOLPH3. We found that silencing GOLPH3 caused a significant reduction in the proliferation of T98G cells and an unexpected increase in total EGFR levels, even at the cell surface, which was however less prone to ligand-induced autophosphorylation. Furthermore, silencing GOLPH3 decreased EGFR sialylation and fucosylation, which correlated with delayed ligand-induced EGFR downregulation and its accumulation at endo-lysosomal compartments. Finally, we found that EGF failed at promoting EGFR ubiquitylation when the levels of GOLPH3 were reduced. Altogether, our results show that GOLPH3 in T98G cells regulates the endocytic trafficking and activation of EGFR likely by affecting its extent of glycosylation and ubiquitylation.
Collapse
Affiliation(s)
- Cecilia Arriagada
- Department of Physiology, School of Medicine and Center for Interdisciplinary Studies of the Nervous System (CISNe), Universidad Austral de Chile, Valdivia 5090000, Chile; (C.A.); (V.A.C.); (C.L.); (A.E.G.); (V.C.M.)
| | - Viviana A. Cavieres
- Department of Physiology, School of Medicine and Center for Interdisciplinary Studies of the Nervous System (CISNe), Universidad Austral de Chile, Valdivia 5090000, Chile; (C.A.); (V.A.C.); (C.L.); (A.E.G.); (V.C.M.)
| | - Charlotte Luchsinger
- Department of Physiology, School of Medicine and Center for Interdisciplinary Studies of the Nervous System (CISNe), Universidad Austral de Chile, Valdivia 5090000, Chile; (C.A.); (V.A.C.); (C.L.); (A.E.G.); (V.C.M.)
| | - Alexis E. González
- Department of Physiology, School of Medicine and Center for Interdisciplinary Studies of the Nervous System (CISNe), Universidad Austral de Chile, Valdivia 5090000, Chile; (C.A.); (V.A.C.); (C.L.); (A.E.G.); (V.C.M.)
| | - Vanessa C. Muñoz
- Department of Physiology, School of Medicine and Center for Interdisciplinary Studies of the Nervous System (CISNe), Universidad Austral de Chile, Valdivia 5090000, Chile; (C.A.); (V.A.C.); (C.L.); (A.E.G.); (V.C.M.)
| | - Jorge Cancino
- Center for Cell Biology and Biomedicine, School of Science and Medicine, Universidad San Sebastián, Santiago 7510235, Chile; (J.C.); (P.V.B.)
| | - Patricia V. Burgos
- Center for Cell Biology and Biomedicine, School of Science and Medicine, Universidad San Sebastián, Santiago 7510235, Chile; (J.C.); (P.V.B.)
- Center for Aging and Regeneration (CARE), Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile
| | - Gonzalo A. Mardones
- Department of Physiology, School of Medicine and Center for Interdisciplinary Studies of the Nervous System (CISNe), Universidad Austral de Chile, Valdivia 5090000, Chile; (C.A.); (V.A.C.); (C.L.); (A.E.G.); (V.C.M.)
| |
Collapse
|
4
|
Nan P, Wang T, Li C, Li H, Wang J, Zhang J, Dou N, Zhan Q, Ma F, Wang H, Qian H. MTA1 promotes tumorigenesis and development of esophageal squamous cell carcinoma via activating the MEK/ERK/p90RSK signaling pathway. Carcinogenesis 2020; 41:1263-1272. [PMID: 31783401 DOI: 10.1093/carcin/bgz200] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Revised: 11/27/2019] [Accepted: 11/28/2019] [Indexed: 01/01/2023] Open
Abstract
Metastasis-associated protein 1 (MTA1) is upregulated in multiple malignancies and promotes cancer proliferation and metastasis, but whether and how MTA1 promotes esophageal squamous cell carcinoma (ESCC) tumorigenesis remain unanswered. Here, we established an ESCC model in MTA1 transgenic mice induced by the chemical carcinogen 4-nitroquinoline 1-oxide (4-NQO) and found that MTA1 promotes ESCC tumorigenesis in mice. MTA1 overexpression was observed in ESCC cells and clinical ESCC samples. Overexpressed MTA1 increased colony formation and the invasiveness and migration of ESCC cells, whereas knock down of MTA1 in ESCC cells significantly decreased colony formation, invasion and migration in vitro and inhibited the growth of xenograft tumors in vivo. RNA sequencing (RNA-seq) analysis combined with western blot assays revealed that MTA1 promotes carcinogenesis by enhancing MEK/ERK/p90RSK signaling. The phosphorylation of MEK, ERK and their downstream target p90RSK was significantly decreased after MTA1 knockdown in ESCC cells and was increased in MTA1-overexpressing cells. Moreover, colony formation, invasion and migration potential were dramatically suppressed when cells overexpressing MTA1 were treated with MEK (PD0325901) or ERK (SCH772948) inhibitors. In conclusion, MTA1 plays a pivotal oncogenic role in ESCC tumorigenesis and development through activating the MEK/ERK/p90RSK pathway.
Collapse
MESH Headings
- Animals
- Apoptosis
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Cell Proliferation
- Disease Models, Animal
- Esophageal Neoplasms/genetics
- Esophageal Neoplasms/metabolism
- Esophageal Neoplasms/pathology
- Esophageal Squamous Cell Carcinoma/genetics
- Esophageal Squamous Cell Carcinoma/metabolism
- Esophageal Squamous Cell Carcinoma/pathology
- Gene Expression Regulation, Neoplastic
- Humans
- MAP Kinase Signaling System
- Male
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Nude
- Repressor Proteins/genetics
- Repressor Proteins/metabolism
- Ribosomal Protein S6 Kinases, 90-kDa/genetics
- Ribosomal Protein S6 Kinases, 90-kDa/metabolism
- Signal Transduction
- Trans-Activators/genetics
- Trans-Activators/metabolism
- Tumor Cells, Cultured
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Peng Nan
- National Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ting Wang
- National Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chunxiao Li
- National Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hui Li
- National Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jinsong Wang
- National Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jingyao Zhang
- National Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Na Dou
- National Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qimin Zhan
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital and Institute, Beijing, China
| | - Fei Ma
- Department of Medical Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Haijuan Wang
- National Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Haili Qian
- National Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
5
|
Tenacissoside H Induces Apoptosis and Inhibits Migration of Colon Cancer Cells by Downregulating Expression of GOLPH3 Gene. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2020; 2020:2824984. [PMID: 32454851 PMCID: PMC7229548 DOI: 10.1155/2020/2824984] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Revised: 04/07/2020] [Accepted: 04/17/2020] [Indexed: 12/24/2022]
Abstract
Objective Tenacissoside H (TDH) is a Chinese medicine monomer extracted from Marsdenia tenacissima extract (MTE), which has been confirmed to have antitumor effects, but its mechanism is still unclear. The aim of this study was to investigate the effect and mechanism of TDH on human colon cancer LoVo cell proliferation and migration and explore the correlation of TDH treatment with the expression of GOLPH3 and cell signaling pathways in LoVo cells. Methods LoVo cells were treated with TDH at 0.1, 1, 10, and 100 μg/mL for 24, 48, and 72 h. The proliferation rate of LoVo cells was evaluated by MTT assay. Recombinant plasmid p-CMV-2-GOLPH3 was constructed, and p-CMV-2-GOLPH3 and p-CMV-2 empty plasmids were transfected into LoVo cells by lipofection. Western blotting was used to detect the transfection efficiency and the expression of p-p70S6K, p70S6K, β-catenin, and GOLPH3. The apoptosis rate was analyzed with Annexin V-FITC/PI double-staining method, and cell migration assessed by transwell assay. Results TDH inhibited the proliferation of LoVo cells in a concentration-dependent manner. The IC50 of TDH treatment in LoVo cells at 24, 48, and 72 h was 40.24, 13.00, and 5.73 μg/mL, respectively. TDH treatment significantly induced apoptosis and suppressed the viability and migration of human colon cancer LoVo cells. The effect of TDH on induction of apoptosis and inhibition of migration in LoVo cells decreased significantly after activating the PI3K/AKT/mTOR and Wnt/β-catenin signaling pathways with agonists. Additionally, the expression of GOLPH3 protein downregulated significantly in LoVo cells under TDH treatment. Overexpression of the GOLPH3 gene increased the expression of key proteins in PI3K/AKT/mTOR and Wnt/β-catenin signaling pathways and blocked the antitumor activity of TDH. Conclusion Collectively, the present results indicated that TDH can inhibit the proliferation vitality of colon cancer LoVo cells through downregulating GOLPH3 expression and activity of PI3K/AKT/mTOR and Wnt/β-catenin signaling pathways.
Collapse
|
6
|
Chen Q, Zheng Y, Wu B, Chen X, Sun F, Ge P, Wang P. BANCR Regulates The Cell Invasion And Migration In Esophageal Squamous Cell Carcinoma Through Wnt/β-Catenin Signaling Pathway. Onco Targets Ther 2019; 12:9319-9327. [PMID: 31807012 PMCID: PMC6847997 DOI: 10.2147/ott.s227220] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Accepted: 10/24/2019] [Indexed: 12/19/2022] Open
Abstract
Objective To explore the regulation of long-chain noncoding BANCR on cell invasion and migration of esophageal squamous carcinoma cells and related mechanisms. Method The mRNA expression of BANCR in esophageal squamous carcinoma cells and esophageal squamous cells was detected by quantitative PCR . The relationship between the expression of BANCR and the survival rate of patients with esophageal squamous cell carcinoma (ESCC) was analyzed by Kaplan–Meier method. The BANCR pair was detected by Transwell invasion and scratch test. In ESCC cell lines, the cells had invasion and migration ability; Western blot was applied to detect the expression of proteins involved in the Wnt/β-catenin signaling pathway. Results BANCR revealed relatively high expression in esophageal squamous carcinoma cells, and the higher the expression of BANCR was, the lower the survival rate of patients with ESCC was. Inhibition of BANCR expression could effectively reduce the invasion and migration ability of esophageal squamous cell carcinoma. After silencing BANCR, the expression of wnt3a, survivin, β-catenin and c-myc protein was downregulated compared with the negative control group (p<0.05). Conclusion Long-chain noncoding BANCR was highly expressed in patients with ESCC and was negatively correlated with patients' survival time. It was of the capability to modulate the cell migration and invasion of ESCC cells through inducing Wnt/β-catenin signaling pathway.
Collapse
Affiliation(s)
- Quan Chen
- Department of Thoracic Surgery, Taizhou People's Hospital, Taizhou, Jiangsu Province 225300, People's Republic of China
| | - Yiming Zheng
- Department of Thoracic Surgery, Taizhou People's Hospital, Taizhou, Jiangsu Province 225300, People's Republic of China
| | - Bingbing Wu
- Department of Thoracic Surgery, Taizhou People's Hospital, Taizhou, Jiangsu Province 225300, People's Republic of China
| | - Xia Chen
- Department of Thoracic Surgery, Taizhou People's Hospital, Taizhou, Jiangsu Province 225300, People's Republic of China
| | - Fei Sun
- Department of Thoracic Surgery, Taizhou People's Hospital, Taizhou, Jiangsu Province 225300, People's Republic of China
| | - Pengfei Ge
- Department of Thoracic Surgery, Taizhou People's Hospital, Taizhou, Jiangsu Province 225300, People's Republic of China
| | - Pengcheng Wang
- Department of Thoracic Surgery, Taizhou People's Hospital, Taizhou, Jiangsu Province 225300, People's Republic of China
| |
Collapse
|
7
|
Loss of FBXO9 Enhances Proteasome Activity and Promotes Aggressiveness in Acute Myeloid Leukemia. Cancers (Basel) 2019; 11:cancers11111717. [PMID: 31684170 PMCID: PMC6895989 DOI: 10.3390/cancers11111717] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 10/30/2019] [Accepted: 11/01/2019] [Indexed: 12/11/2022] Open
Abstract
The hematopoietic system is maintained throughout life by stem cells that are capable of differentiating into all hematopoietic lineages. An intimate balance between self-renewal, differentiation, and quiescence is required to maintain hematopoiesis and disruption of this balance can result in malignant transformation. FBXO9, the substrate recognition component from the SCF E3 ubiquitin ligase family, is downregulated in patients with acute myeloid leukemia (AML) compared to healthy bone marrow, and this downregulation is particularly evident in patients with inv(16) AML. To study FBXO9 in malignant hematopoiesis, we generated a conditional knockout mouse model using a novel CRISPR/Cas9 strategy. Deletion of Fbxo9 in the murine hematopoietic system showed no adverse effects on stem and progenitor cell function but in AML lead to markedly accelerated and aggressive leukemia development in mice with inv(16). Not only did Fbxo9 play a role in leukemia initiation but it also functioned to maintain AML activity and promote disease progression. Quantitative mass spectrometry from primary tumors reveals tumors lacking Fbxo9 highly express proteins associated with metastasis and invasion as well as components of the ubiquitin proteasome system. We confirmed that the loss of FBXO9 leads to increased proteasome activity and tumors cells were more sensitive to in vitro proteasome inhibition with bortezomib, suggesting that FBXO9 expression may predict patients’ response to bortezomib.
Collapse
|
8
|
Lu J, Zhong F, Sun B, Wang C. Diagnostic Utility of Serum Golgi Phosphoprotein 3 in Bladder Cancer Patients. Med Sci Monit 2019; 25:6736-6741. [PMID: 31494662 PMCID: PMC6752096 DOI: 10.12659/msm.915950] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Background This study assessed whether serum Golgi phosphoprotein 3 (GOLPH3) could be used as a biomarker for detecting bladder cancer. Material/Methods Enzyme-linked immunosorbent assay (ELISA) and immunohistochemistry (IHC) assay were performed to measure GOLPH3 expression in serum and tissue samples, respectively, of bladder cancer patients. The associations of serum GOLPH3 expression with clinicopathological factors and the diagnostic accuracy were statistically evaluated using the chi-square test and receiver operating characteristic (ROC) curve analysis. Results Compared with the healthy control group, serum GOLPH3 level was distinctly enhanced in bladder cancer patients (P<0.001). Moreover, compared to the non-malignant tissues, GOLPH3 showed positive expression in bladder cancer tissues. The abnormal GOLPH3 levels were tightly related to grade (P=0.018), tumor stage (P=0.000), lymph node status (P=0.030), and muscle invasion (P=0.012). ROC analysis showed that serum GOLPH3 exhibited a high diagnostic value to distinguish bladder cancer patients from healthy persons. The area under the ROC curve (AUC) was 0.948. The specificity and sensitivity were 92.5% and 83.8%, respectively. Conclusions GOLPH3 was highly expressed in bladder cancer patients and could be used as a diagnostic tool.
Collapse
Affiliation(s)
- Jianlei Lu
- Department of Surgical Urology, First People's Hospital, Jining, Shandong, China (mainland)
| | - Feng Zhong
- Department of Surgical Urology, Affiliated Hospital of the Academy of Medical Sciences, Jinan, Shandong, China (mainland)
| | - Beibei Sun
- Department of Operating Rooms, Second People's Hospital, Jining, Shandong, China (mainland)
| | - Chao Wang
- Department of Surgical Urology, First People's Hospital, Jining, Shandong, China (mainland)
| |
Collapse
|
9
|
The Great Escape: how phosphatidylinositol 4-kinases and PI4P promote vesicle exit from the Golgi (and drive cancer). Biochem J 2019; 476:2321-2346. [DOI: 10.1042/bcj20180622] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 08/06/2019] [Accepted: 08/12/2019] [Indexed: 12/13/2022]
Abstract
Abstract
Phosphatidylinositol 4-phosphate (PI4P) is a membrane glycerophospholipid and a major regulator of the characteristic appearance of the Golgi complex as well as its vesicular trafficking, signalling and metabolic functions. Phosphatidylinositol 4-kinases, and in particular the PI4KIIIβ isoform, act in concert with PI4P to recruit macromolecular complexes to initiate the biogenesis of trafficking vesicles for several Golgi exit routes. Dysregulation of Golgi PI4P metabolism and the PI4P protein interactome features in many cancers and is often associated with tumour progression and a poor prognosis. Increased expression of PI4P-binding proteins, such as GOLPH3 or PITPNC1, induces a malignant secretory phenotype and the release of proteins that can remodel the extracellular matrix, promote angiogenesis and enhance cell motility. Aberrant Golgi PI4P metabolism can also result in the impaired post-translational modification of proteins required for focal adhesion formation and cell–matrix interactions, thereby potentiating the development of aggressive metastatic and invasive tumours. Altered expression of the Golgi-targeted PI 4-kinases, PI4KIIIβ, PI4KIIα and PI4KIIβ, or the PI4P phosphate Sac1, can also modulate oncogenic signalling through effects on TGN-endosomal trafficking. A Golgi trafficking role for a PIP 5-kinase has been recently described, which indicates that PI4P is not the only functionally important phosphoinositide at this subcellular location. This review charts new developments in our understanding of phosphatidylinositol 4-kinase function at the Golgi and how PI4P-dependent trafficking can be deregulated in malignant disease.
Collapse
|
10
|
Chen MJ, Deng J, Chen C, Hu W, Yuan YC, Xia ZK. LncRNA H19 promotes epithelial mesenchymal transition and metastasis of esophageal cancer via STAT3/EZH2 axis. Int J Biochem Cell Biol 2019; 113:27-36. [PMID: 31102664 DOI: 10.1016/j.biocel.2019.05.011] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Revised: 05/05/2019] [Accepted: 05/14/2019] [Indexed: 01/18/2023]
Abstract
BACKGROUND Long non-coding RNA H19 (lncRNA H19) has been widely reported in esophageal cancer (EC), and previous study had found that lncRNAH19 was up-regulated in EC and promoted cell proliferation and metastasis. However, the mechanism still needs further studied. METHODS Levels of lncRNA H19 were analyzed by qRT-PCR in matched samples from 30 patients. Expression levels of lncRNA H19, let-7, STAT3 and EZH2 were additionally identified by qRT-PCR and western blotting in five EC cell lines. The effects of lncRNA H19 on cell proliferation, migration, invasion and apoptosis in cell lines were performed by MTT assay, colony formation assay, Transwell assay and flow cytometry in vitro, and tumor formation was detected by xenograft nude mice model in vivo. The expression level of STAT3, EZH2, β-catenin, and EMT and metastasis related molecules such as E-cadherin, N-cadherin, Snail-1 and MMP-9 was assessed by qRT-PCR and western blotting. Finally, luciferase reporter assay and RIP assay were used to verify the interaction between lncRNA H19 and let-7c, and their subsequent regulation of STAT3. RESULTS Knockdown of lncRNA H19 repressed cell proliferation, migration and invasion as well as EMT and metastasis via STAT3-EZH2-β-catenin pathway, while lncRNA H19 regulated STAT3 negatively regulated let-7c in EC cell lines. CONCLUSIONS lncRNA H19 facilitates EMT and metastasis of EC through let-7c/STAT3/EZH2/β-catenin axis.
Collapse
Affiliation(s)
- Ming-Jiu Chen
- Department of Thoracic Surgery, The Second Xiangya Hospital of Central South University, Changsha 410011, PR China
| | - Jie Deng
- Department of Respiratory Medicine, The First Hospital of Changsha, Changsha 410005, PR China
| | - Chen Chen
- Department of Thoracic Surgery, The Second Xiangya Hospital of Central South University, Changsha 410011, PR China
| | - Wen Hu
- Department of Thoracic Surgery, The Second Xiangya Hospital of Central South University, Changsha 410011, PR China
| | - Yun-Chang Yuan
- Department of Thoracic Surgery, The Second Xiangya Hospital of Central South University, Changsha 410011, PR China
| | - Zhen-Kun Xia
- Department of Thoracic Surgery, The Second Xiangya Hospital of Central South University, Changsha 410011, PR China.
| |
Collapse
|
11
|
Chen XJ, Shen XN, Chen L. Remifentanil regulates proliferation and apoptosis of gastric cancer cells by regulating miR-206/GOLPH3. Shijie Huaren Xiaohua Zazhi 2019; 27:228-237. [DOI: 10.11569/wcjd.v27.i4.228] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Remifentanil is a commonly used anesthetic in clinical practice. In recent years, its new efficacy has been continuously discovered, especially its anti-cancer function. However, the role and mechanism of remifentanil in gastric cancer (GC) are still not clear.
AIM To investigate the effect of remifentanil on the expression of miR-206, GOLPH3, cell proliferation, and apoptosis in human GC cell lines AGS and MKN-45.
METHODS The expression of miR-206 and GOLPH3 and the viability and apoptosis of AGS and MKN-45 cells after treatment with 40 nmol/L remifentanil were detected by qRT-PCR, Western blot, MTT assay, and flow cytometry, respectively. Cell viability and apoptosis of AGS and MKN-45 cells with overexpression of miR-206 or knockdown of GOLPH3 were detected by MTT assay and flow cytometry, respectively. The targeting relationship between miR-206 and GOLPH3 was verified by Targetscan online prediction, dual-luciferase assay, and Western blot. After transfection with miR-206 inhibitor or pcDNA-GOLPH3, AGS and MKN-45 cells were treated with 40 nmol/L remifentanil and then detected for cell viability and apoptosis.
RESULTS After treatment with remifentanil, the expression of miR-206 and apoptosis rate were increased while the expression of GOLPH3 and cell viability were decreased in AGS and MKN-45 cells. Cell viability was decreased and apoptotic rate was increased in AGS and MKN-45 cells after overexpression of miR-206 or knockdown of GOLPH3. The results of Targetscan online prediction, dual-luciferase assay, and Western blot indicted that miR-206 could regulate the expression of GOLPH3 protein. Down-regulation of miR-206 or overexpression of GOLPH3 could reverse the inhibition of proliferation and apoptosis of AGS and MKN-45 cells by remifentanil.
CONCLUSION Remifentanil could inhibit the proliferation and induce apoptosis of AGS and MKN-45 cells by regulating the expression of miR-206 and GOLPH3.
Collapse
Affiliation(s)
- Xiao-Jun Chen
- Department of Anesthesiology, Yiwu Central Hospital, Yiwu 322000, Zhejiang Province, China
| | - Xin-Ning Shen
- Department of Anesthesiology, Yiwu Central Hospital, Yiwu 322000, Zhejiang Province, China
| | - Liang Chen
- Department of Oncology, Yiwu Central Hospital, Yiwu 322000, Zhejiang Province, China
| |
Collapse
|
12
|
The knocking down of the oncoprotein Golgi phosphoprotein 3 in T98G cells of glioblastoma multiforme disrupts cell migration by affecting focal adhesion dynamics in a focal adhesion kinase-dependent manner. PLoS One 2019; 14:e0212321. [PMID: 30779783 PMCID: PMC6380552 DOI: 10.1371/journal.pone.0212321] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Accepted: 01/31/2019] [Indexed: 01/29/2023] Open
Abstract
Golgi phosphoprotein 3 (GOLPH3) is a conserved protein of the Golgi apparatus that in humans has been implicated in tumorigenesis. However, the precise function of GOLPH3 in malignant transformation is still unknown. Nevertheless, clinicopathological data shows that in more than a dozen kinds of cancer, including gliomas, GOLPH3 could be found overexpressed, which correlates with poor prognosis. Experimental data shows that overexpression of GOLPH3 leads to transformation of primary cells and to tumor growth enhancement. Conversely, the knocking down of GOLPH3 in GOLPH3-overexpressing tumor cells reduces tumorigenic features, such as cell proliferation and cell migration and invasion. The cumulative evidence indicate that GOLPH3 is an oncoprotein that promotes tumorigenicity by a mechanism that impact at different levels in different types of cells, including the sorting of Golgi glycosyltransferases, signaling pathways, and the actin cytoskeleton. How GOLPH3 connects mechanistically these processes has not been determined yet. Further studies are important to have a more complete understanding of the role of GOLPH3 as oncoprotein. Given the genetic diversity in cancer, a still outstanding aspect is how in this inherent heterogeneity GOLPH3 could possibly exert its oncogenic function. We have aimed to evaluate the contribution of GOLPH3 overexpression in the malignant phenotype of different types of tumor cells. Here, we analyzed the effect on cell migration that resulted from stable, RNAi-mediated knocking down of GOLPH3 in T98G cells of glioblastoma multiforme, a human glioma cell line with unique features. We found that the reduction of GOLPH3 levels produced dramatic changes in cell morphology, involving rearrangements of the actin cytoskeleton and reduction in the number and dynamics of focal adhesions. These effects correlated with decreased cell migration and invasion due to affected persistence and directionality of cell motility. Moreover, the knocking down of GOLPH3 also caused a reduction in autoactivation of focal adhesion kinase (FAK), a cytoplasmic tyrosine kinase that regulates focal adhesions. Our data support a model in which GOLPH3 in T98G cells promotes cell migration by stimulating the activity of FAK.
Collapse
|
13
|
Fang C, Jiang B, Shi X, Fan C. Hes3 Enhances the Malignant Phenotype of Lung Cancer through Upregulating Cyclin D1, Cyclin D3 and MMP7 Expression. Int J Med Sci 2019; 16:470-476. [PMID: 30911281 PMCID: PMC6428979 DOI: 10.7150/ijms.28139] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Accepted: 02/08/2019] [Indexed: 12/23/2022] Open
Abstract
Hes3 is a basic helix-loop-helix factor gene, which was found to be involved in neural cell differentiation. Expression and clinicopathological significance of Hes3 in non-small cell lung cancer was not clear. In this study, we used immunohistochemistry to examine Hes3 expression in normal human lung and non-small cell lung cancer tissues. Hes3 expression was detected in cytoplasm and nucleus. Hes3 expression in bronchial epithelial cells and epithelial cells of submucosal glands was relatively weak and the positive rate was of 30.3% (10/33). Hes3 expression in non-small cell lung cancer tissues (51.8% (58/112)) was significantly higher than that in normal lung tissues (p < 0.05). Hes3 expression in cancer tissues was significantly associated with poor differentiation, advanced TNM stages, lymph node metastasis, and a shorter patient survival time (p < 0.05). In vitro study showed that overexpression of Hes3 in A549 cells significantly promoted cancer cell proliferation and invasion, while inhibition of Hes3 expression significantly downregulated cancer cell proliferation and invasion (p < 0.05). Western blotting showed that overexpression of Hes3 significantly upregulated expression of Cyclin D1, Cyclin D3, and MMP7 in A549 cells, while inhibition of Hes3 expression in LK2 cells significantly downregulated the expression of these molecules (p < 0.05). These results indicated that Hes3 may contribute to the malignant phenotype of non-small cell lung cancer, possibly through regulation of Cyclin D1, Cyclin D3, and MMP7, and may be a promising cancer marker.
Collapse
Affiliation(s)
- Changqing Fang
- Department of Pathology, First Affiliated Hospital and College of Basic Medical Sciences of China Medical University, 110001, Shenyang, China
| | - Biying Jiang
- Department of Pathology, First Affiliated Hospital and College of Basic Medical Sciences of China Medical University, 110001, Shenyang, China
| | - Xiuying Shi
- Department of Pathology, First Affiliated Hospital and College of Basic Medical Sciences of China Medical University, 110001, Shenyang, China
| | - Chuifeng Fan
- Department of Pathology, First Affiliated Hospital and College of Basic Medical Sciences of China Medical University, 110001, Shenyang, China
| |
Collapse
|
14
|
Kuna RS, Field SJ. GOLPH3: a Golgi phosphatidylinositol(4)phosphate effector that directs vesicle trafficking and drives cancer. J Lipid Res 2018; 60:269-275. [PMID: 30266835 DOI: 10.1194/jlr.r088328] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 09/25/2018] [Indexed: 12/17/2022] Open
Abstract
GOLPH3 is a peripheral membrane protein localized to the Golgi and its vesicles, but its purpose had been unclear. We found that GOLPH3 binds specifically to the phosphoinositide phosphatidylinositol(4)phosphate [PtdIns(4)P], which functions at the Golgi to promote vesicle exit for trafficking to the plasma membrane. PtdIns(4)P is enriched at the trans-Golgi and so recruits GOLPH3. Here, a GOLPH3 complex is formed when it binds to myosin18A (MYO18A), which binds F-actin. This complex generates a pulling force to extract vesicles from the Golgi; interference with this GOLPH3 complex results in dramatically reduced vesicle trafficking. The GOLPH3 complex has been identified as a driver of cancer in humans, likely through multiple mechanisms that activate secretory trafficking. In this review, we summarize the literature that identifies the nature of the GOLPH3 complex and its role in cancer. We also consider the GOLPH3 complex as a hub with the potential to reveal regulation of the Golgi and suggest the possibility of GOLPH3 complex inhibition as a therapeutic approach in cancer.
Collapse
Affiliation(s)
- Ramya S Kuna
- Division of Endocrinology and Metabolism, Department of Medicine, University of California at San Diego, La Jolla, CA
| | - Seth J Field
- Division of Endocrinology and Metabolism, Department of Medicine, University of California at San Diego, La Jolla, CA
| |
Collapse
|