75 results on '"X X, Chen"'
Search Results
2. Correction to: KIF26B, a novel oncogene, promotes proliferation and metastasis by activating the VEGF pathway in gastric cancer.
- Author
-
Zhang H, Ma RR, Wang XJ, Su ZX, Chen X, Shi DB, Guo XY, Liu HT, and Gao P
- Published
- 2022
- Full Text
- View/download PDF
3. Correction: eEF-2 kinase is a critical regulator of Warbrug effect through controlling PP2A-A synthesis.
- Author
-
Cheng Y, Ren X, Yuan Y, Shan Y, Li L, Chen X, Zhang L, Takahashi Y, Yang JW, Han B, Liao J, Li Y, Harvey H, Ryazanov A, Robertson GP, Wan G, Liu D, Chen AF, Tao Y, and Yang JM
- Abstract
In this article, the authors recently noticed that the tubulin blots in Figs. 2a and 6a were inadvertently misplaced during the preparation of these figures due to their similarity. The amended versions of the figures are now shown below. The conclusions of this paper are not affected. The authors sincerely apologize for these errors.
- Published
- 2018
- Full Text
- View/download PDF
4. Upregulation of the ALDOA/DNA-PK/p53 pathway by dietary restriction suppresses tumor growth.
- Author
-
Ma D, Chen X, Zhang PY, Zhang H, Wei LJ, Hu S, Tang JZ, Zhou MT, Xie C, Ou R, Xu Y, and Tang KF
- Subjects
- Animals, Apoptosis, Biomarkers, Tumor, Caloric Restriction, Carcinoma, Hepatocellular etiology, Carcinoma, Hepatocellular metabolism, Carcinoma, Hepatocellular pathology, Cell Movement, Cell Proliferation, DNA-Activated Protein Kinase genetics, Female, Fructose-Bisphosphate Aldolase genetics, Humans, Liver Neoplasms etiology, Liver Neoplasms metabolism, Liver Neoplasms pathology, Mice, Mice, Inbred BALB C, Mice, Nude, Neoplasm Invasiveness, Nuclear Proteins genetics, Tumor Cells, Cultured, Tumor Suppressor Protein p53 genetics, Xenograft Model Antitumor Assays, Carcinoma, Hepatocellular prevention & control, DNA-Activated Protein Kinase metabolism, Diet adverse effects, Fructose-Bisphosphate Aldolase metabolism, Gene Expression Regulation, Neoplastic, Liver Neoplasms prevention & control, Nuclear Proteins metabolism, Tumor Suppressor Protein p53 metabolism
- Abstract
Dietary restriction (DR) delays the incidence and decreases the growth of various types of tumors; however, the mechanisms responsible for DR-mediated antitumor effects have not been unequivocally identified. Here, we report that DR suppresses xenograft tumor growth by upregulating a novel signaling pathway. DR led to upregulated aldolase A (ALDOA) expression in xenograft tumors. ALDOA physically interacted with the catalytic subunit of DNA-dependent protein kinase (DNA-PK) and promoted DNA-PK activation. Activated DNA-PK phosphorylated p53 and increased its activity. Although ALDOA can function as an oncogene in cultured cells, it can also activate the tumor suppressor p53. Thus, ALDOA overexpression in the presence of p53 suppressed xenograft tumor growth; however, when p53 was suppressed, ALDOA overexpression promoted xenograft tumor growth. Moreover, we demonstrated that p53 suppression inhibited the antitumor effects of DR. Our results indicate that upregulation of the ALDOA/DNA-PK/p53 pathway is a mechanism accounting for the antitumor effects of DR.
- Published
- 2018
- Full Text
- View/download PDF
5. Intracellular mature IL-37 suppresses tumor metastasis via inhibiting Rac1 activation.
- Author
-
Li Y, Zhao M, Guo C, Chu H, Li W, Chen X, Wang X, Li Y, Jia Y, Koussatidjoa S, Zhu F, Wang J, Wang X, Wang Q, Zhao W, Shi Y, Chen W, and Zhang L
- Subjects
- Adenocarcinoma genetics, Adenocarcinoma metabolism, Animals, Apoptosis, Biomarkers, Tumor genetics, Biomarkers, Tumor metabolism, Cell Cycle, Female, Follow-Up Studies, Humans, Interleukin-1 genetics, Lung Neoplasms genetics, Lung Neoplasms metabolism, Lymphatic Metastasis, Male, Mice, Mice, Inbred BALB C, Mice, Nude, Middle Aged, Neoplasm Invasiveness, Neovascularization, Pathologic, Prognosis, Survival Rate, Tumor Cells, Cultured, Xenograft Model Antitumor Assays, rac1 GTP-Binding Protein genetics, Adenocarcinoma secondary, Cell Movement, Cell Proliferation, Gene Expression Regulation, Neoplastic, Interleukin-1 metabolism, Lung Neoplasms pathology, rac1 GTP-Binding Protein metabolism
- Abstract
IL-37, a newly found anti-inflammatory cytokine of the IL-1 family, has both extracellular and intracellular functions. Accumulating evidences indicate that it is also involved in tumor progression. However, the mechanism and its intracellular target are unclear. In this study, clinical data from 84 patients showed that loss or reduced expression of IL-37 in lung adenocarcinoma tissues was significantly associated with tumor metastasis. We further provided evidence that IL-37 inhibited effectively tumor metastasis in vitro and in vivo. Moreover, we uncovered a novel mechanism by which IL-37 suppressed tumor cell migration via its intracellular mature form (amino acids 46-218). Intracellular mature form of IL-37, but not its extracellular form, markedly inhibited migration of multiple kinds of tumor cells through inhibiting Rac1 activation. Mechanistically, intracellular mature IL-37 directly bound to the CAAX motif in the C-terminal hypervariable region of Rac1, and then inhibited Rac1 membrane translocation and subsequent downstream signaling. Our research identifies intracellular mature IL-37 as a novel endogenous inhibitor of Rac1. Given the crucial roles of Rac1 in tumor angiogenesis and metastasis, intracellular mature IL-37 might serve as a potential strategy for the control of Rac1 activity and tumor progression.
- Published
- 2018
- Full Text
- View/download PDF
6. IL6 derived from cancer-associated fibroblasts promotes chemoresistance via CXCR7 in esophageal squamous cell carcinoma.
- Author
-
Qiao Y, Zhang C, Li A, Wang D, Luo Z, Ping Y, Zhou B, Liu S, Li H, Yue D, Zhang Z, Chen X, Shen Z, Lian J, Li Y, Wang S, Li F, Huang L, Wang L, Zhang B, Yu J, Qin Z, and Zhang Y
- Subjects
- Animals, Antineoplastic Agents pharmacology, Apoptosis, Biomarkers, Tumor, Cancer-Associated Fibroblasts drug effects, Cancer-Associated Fibroblasts metabolism, Carcinoma, Squamous Cell drug therapy, Carcinoma, Squamous Cell metabolism, Cell Proliferation, Epithelial-Mesenchymal Transition, Esophageal Neoplasms drug therapy, Esophageal Neoplasms metabolism, Female, Humans, Interleukin-6 genetics, Lymphatic Metastasis, Male, Mice, Mice, Inbred BALB C, Mice, Nude, Middle Aged, Neoplasm Invasiveness, Neoplasm Recurrence, Local, Prognosis, Receptors, CXCR genetics, Signal Transduction, Survival Rate, Tumor Cells, Cultured, Tumor Microenvironment, Xenograft Model Antitumor Assays, Cancer-Associated Fibroblasts pathology, Carcinoma, Squamous Cell secondary, Drug Resistance, Neoplasm, Esophageal Neoplasms pathology, Gene Expression Regulation, Neoplastic, Interleukin-6 metabolism, Receptors, CXCR metabolism
- Abstract
Various factors and cellular components in the tumor microenvironment are key drivers associated with drug resistance in many cancers. Here, we analyzed the factors and molecular mechanisms involved in chemoresistance in patients with esophageal squamous cell carcinoma (ESCC). We found that interleukin 6 (IL6) derived mainly from cancer-associated fibroblasts played the most important role in chemoresistance by upregulating C-X-C motif chemokine receptor 7 (CXCR7) expression through signal transducer and activator of transcription 3/nuclear factor-κB pathway. CXCR7 knockdown resulted in the inhibition of IL6-induced proliferation and chemoresistance. In addition, CXCR7 silencing significantly decreased gene expression associated with stemness, chemoresistance and epithelial-mesenchymal transition and suppressed the proliferation ability of ESCC cells in three-dimensional culture systems and angiogenesis assay. In clinical samples, ESCC patients with high expression of CXCR7 and IL6 presented a significantly worse overall survival and progression-free survival upon receiving cisplatin after operation. These results suggest that the IL6-CXCR7 axis may provide a promising target for the treatment of ESCC.
- Published
- 2018
- Full Text
- View/download PDF
7. Induction of anti-VEGF therapy resistance by upregulated expression of microseminoprotein (MSMP).
- Author
-
Mitamura T, Pradeep S, McGuire M, Wu SY, Ma S, Hatakeyama H, Lyons YA, Hisamatsu T, Noh K, Villar-Prados A, Chen X, Ivan C, Rodriguez-Aguayo C, Hu W, Lopez-Berestein G, Coleman RL, and Sood AK
- Subjects
- Angiogenesis Inhibitors pharmacology, Animals, Apoptosis, Biomarkers, Tumor, Carcinoma, Ovarian Epithelial drug therapy, Carcinoma, Ovarian Epithelial metabolism, Cell Hypoxia, Cell Proliferation, Fallopian Tube Neoplasms drug therapy, Fallopian Tube Neoplasms metabolism, Female, Follow-Up Studies, Humans, Male, Mice, Mice, Nude, Neoplasm Proteins genetics, Neovascularization, Pathologic, Peritoneal Neoplasms drug therapy, Peritoneal Neoplasms metabolism, Prognosis, Tumor Cells, Cultured, Xenograft Model Antitumor Assays, Bevacizumab pharmacology, Carcinoma, Ovarian Epithelial pathology, Drug Resistance, Neoplasm, Fallopian Tube Neoplasms pathology, Neoplasm Proteins metabolism, Peritoneal Neoplasms pathology, Vascular Endothelial Growth Factor A antagonists & inhibitors
- Abstract
Anti-vascular endothelial growth factor (VEGF) therapy has demonstrated efficacy in treating human metastatic cancers, but therapeutic resistance is a practical limitation and most tumors eventually become unresponsive. To identify microenvironmental factors underlying the resistance of cancer to antiangiogenesis therapy, we conducted genomic analyses of intraperitoneal ovarian tumors in which adaptive resistance to anti-VEGF therapy (B20 antibody) developed. We found that expression of the microseminoprotein, prostate-associated (MSMP) gene was substantially upregulated in resistant compared with control tumors. MSMP secretion from cancer cells was induced by hypoxia, triggering MAPK signaling in endothelial cells to promote tube formation in vitro. Recruitment of the transcriptional repressor CCCTC-binding factor (CTCF) to the MSMP enhancer region was decreased by histone acetylation under hypoxic conditions in cancer cells. MSMP siRNA, delivered in vivo using the DOPC nanoliposomes, restored tumor sensitivity to anti-VEGF therapy. In ovarian cancer patients treated with bevacizumab, serum MSMP concentration increased significantly only in non-responders. These findings imply that MSMP inhibition combined with the use of antiangiogenesis drugs may be a new strategy to overcome resistance to antiangiogenesis therapy.
- Published
- 2018
- Full Text
- View/download PDF
8. Berberine binds RXRα to suppress β-catenin signaling in colon cancer cells.
- Author
-
Ruan H, Zhan YY, Hou J, Xu B, Chen B, Tian Y, Wu D, Zhao Y, Zhang Y, Chen X, Mi P, Zhang L, Zhang S, Wang X, Cao H, Zhang W, Wang H, Li H, Su Y, Zhang XK, and Hu T
- Subjects
- Animals, Berberine metabolism, Berberine therapeutic use, Colonic Neoplasms drug therapy, Humans, Mice, Mice, Inbred BALB C, Xenograft Model Antitumor Assays, Berberine pharmacology, Colonic Neoplasms pathology, Retinoid X Receptor alpha metabolism, Signal Transduction physiology, beta Catenin physiology
- Abstract
Berberine, an isoquinoline alkaloid, is a traditional oriental medicine used to treat diarrhea and gastroenteritis. Recently, we reported that it could inhibit the growth of intestinal polyp in animals and in patients with the familial adenomatous polyposis by downregulating β-catenin signaling. However, the intracellular target mediating the effects of berberine remains elusive. Here, we provide evidence that berberine inhibits β-catenin function via directly binding to a unique region comprising residues Gln275, Arg316 and Arg371 in nuclear receptor retinoid X receptor alpha (RXRα), where berberine concomitantly binding to and synergistically activating RXRα with 9-cis-retinoic acid (9-cis-RA), a natural ligand binding to the classical ligand-binding pocket of RXRα. Berberine binding promotes RXRα interaction with nuclear β-catenin, leading to c-Cbl mediated degradation of β-catenin, and consequently inhibits the proliferation of colon cancer cells. Furthermore, berberine suppresses the growth of human colon carcinoma xenograft in nude mice in an RXRα-dependent manner. Together, our study not only identifies RXRα as a direct protein target for berberine but also dissects their binding mode and validates that berberine indeed suppresses β-catenin signaling and cell growth in colon cancer via binding RXRα, which provide new strategies for the design of new RXRα-based antitumor agents and drug combinations.
- Published
- 2017
- Full Text
- View/download PDF
9. Loss of claudin-3 expression induces IL6/gp130/Stat3 signaling to promote colon cancer malignancy by hyperactivating Wnt/β-catenin signaling.
- Author
-
Ahmad R, Kumar B, Chen Z, Chen X, Müller D, Lele SM, Washington MK, Batra SK, Dhawan P, and Singh AB
- Subjects
- Adenocarcinoma genetics, Adenocarcinoma mortality, Animals, Carcinogenesis metabolism, Cell Transformation, Neoplastic, Claudin-3 genetics, Colon metabolism, Colorectal Neoplasms genetics, Colorectal Neoplasms mortality, Cytokine Receptor gp130 metabolism, Epigenesis, Genetic, Epithelial-Mesenchymal Transition, Gene Expression Regulation, Neoplastic, Humans, Intestinal Mucosa metabolism, Mice, Mice, Knockout, Permeability, STAT3 Transcription Factor metabolism, Up-Regulation, Adenocarcinoma pathology, Claudin-3 metabolism, Colonic Neoplasms metabolism, Colorectal Neoplasms pathology, Wnt Signaling Pathway, beta Catenin metabolism
- Abstract
The hyperactivated Wnt/β-catenin signaling acts as a switch to induce epithelial to mesenchymal transition and promote colorectal cancer. However, due to its essential role in gut homeostasis, therapeutic targeting of this pathway has proven challenging. Additionally, IL-6/Stat-3 signaling, activated by microbial translocation through the dysregulated mucosal barrier in colon adenomas, facilitates the adenoma to adenocarcinomas transition. However, inter-dependence between these signaling pathways and key mucosal barrier components in regulating colon tumorigenesis and cancer progression remains unclear. In current study, we have discovered, using a comprehensive investigative regimen, a novel and tissue-specific role of claudin-3, a tight junction integral protein, in inhibiting colon cancer progression by serving as the common rheostat of Stat-3 and Wnt-signaling activation. Loss of claudin-3 also predicted poor patient survival. These findings however contrasted an upregulated claudin-3 expression in other cancer types and implicated role of the epigenetic regulation. Claudin-3-/- mice revealed dedifferentiated and leaky colonic epithelium, and developed invasive adenocarcinoma when subjected to colon cancer. Wnt-signaling hyperactivation, albeit in GSK-3β independent manner, differentiated colon cancer in claudin-3-/- mice versus WT-mice. Claudin-3 loss also upregulated the gp130/IL6/Stat3 signaling in colonic epithelium potentially assisted by infiltrating immune components. Genetic and pharmacological studies confirmed that claudin-3 loss induces Wnt/β-catenin activation, which is further exacerbated by Stat-3-activation and help promote colon cancer. Overall, these novel findings identify claudin-3 as a therapeutic target for inhibiting overactivation of Wnt-signaling to prevent CRC malignancy.
- Published
- 2017
- Full Text
- View/download PDF
10. KIF26B, a novel oncogene, promotes proliferation and metastasis by activating the VEGF pathway in gastric cancer.
- Author
-
Zhang H, Ma RR, Wang XJ, Su ZX, Chen X, Shi DB, Guo XY, Liu HT, and Gao P
- Subjects
- Animals, Carcinogenesis genetics, Cell Line, Tumor, Cell Movement, Cell Proliferation genetics, Gene Expression Regulation, Neoplastic, Humans, Male, Mice, MicroRNAs metabolism, Neoplasm Invasiveness genetics, Prognosis, Signal Transduction, Up-Regulation, Neoplasm Metastasis genetics, Oncogenes, Stomach Neoplasms genetics, Vascular Endothelial Growth Factor A metabolism
- Abstract
Tumor metastasis is the main reason of cancer-related death for gastric cancer (GC) patients and gene expression microarray data indicate that kinesin family member 26B (KIF26B) is one of the most upregulated genes in metastatic GC samples. Specifically, KIF26B expression was upregulated in a stepwise manner from non-tumorous gastric mucosa, primary GC tissues without metastasis, via primary GC tissues with metastasis, to secondary lymph node metastatic (LNM) foci. Increased expression of KIF26B was correlated with tumor size, positive LNM or distant metastases and poor prognosis. KIF26B, negatively regulated by miR-372, promoted GC cell proliferation and metastasis in vitro and in vivo. Mechanistic investigations confirmed that the main target of KIF26B was the vascular endothelial growth factor (VEGF) signaling pathway, particularly by inhibition or overexpression of VEGFA, PXN, FAK, PIK3CA, BCL2 and CREB1. Thus, KIF26B, a novel oncogene regulated by miR-372, promotes proliferation and metastasis through the VEGF pathway in GC.
- Published
- 2017
- Full Text
- View/download PDF
11. Critical role of Myc activation in mouse hepatocarcinogenesis induced by the activation of AKT and RAS pathways.
- Author
-
Xin B, Yamamoto M, Fujii K, Ooshio T, Chen X, Okada Y, Watanabe K, Miyokawa N, Furukawa H, and Nishikawa Y
- Subjects
- Animals, Apoptosis, Carcinoma, Hepatocellular genetics, Carcinoma, Hepatocellular metabolism, Cell Proliferation, Cell Transformation, Neoplastic genetics, Cell Transformation, Neoplastic metabolism, Humans, Liver Neoplasms genetics, Liver Neoplasms metabolism, Male, Mice, Mice, Inbred C57BL, Proto-Oncogene Proteins c-akt genetics, Proto-Oncogene Proteins c-myc genetics, Proto-Oncogene Proteins p21(ras) genetics, Retrospective Studies, Signal Transduction, Tumor Cells, Cultured, Carcinoma, Hepatocellular pathology, Cell Transformation, Neoplastic pathology, Liver Neoplasms pathology, Proto-Oncogene Proteins c-akt metabolism, Proto-Oncogene Proteins c-myc metabolism, Proto-Oncogene Proteins p21(ras) metabolism
- Abstract
MYC activation at modest levels has been frequently found in hepatocellular carcinoma. However, its significance in hepatocarcinogenesis has remained obscure. Here we examined the role of Myc activation in mouse liver tumours induced by hepatocytic expression of myristoylated AKT (AKT) and/or mutant HRAS
V12 (HRAS) via transposon-mediated gene integration. AKT or HRAS alone required 5 months to induce liver tumours, whereas their combination generated hepatocellular carcinoma within 8 weeks. Co-introduction of AKT and HRAS induced lipid-laden preneoplastic cells that grew into nodules composed of tumour cells with or without intracytoplasmic lipid, with the latter being more proliferative and associated with spontaneous Myc expression. AKT/HRAS-induced tumorigenesis was almost completely abolished when MadMyc, a competitive Myc inhibitor, was expressed simultaneously. The Tet-On induction of MadMyc in preneoplastic cells significantly inhibited the progression of AKT/HRAS-induced tumours; its induction in transformed cells suppressed their proliferative activity with alterations in lipid metabolism and protein translation. Transposon-mediated Myc overexpression facilitated tumorigenesis by AKT or HRAS, and when it was co-introduced with AKT and HRAS, diffusely infiltrating tumours without lipid accumulation developed as early as 2 weeks. Examination of the dose-responses of Myc in the enhancement of AKT/HRAS-induced tumorigenesis revealed that a reduction to one-third retained enhancing effect but three-times greater introduction damped the process with increased apoptosis. Myc overexpression suppressed the mRNA expression of proteins involved in the synthesis of fatty acids, and when combined with HRAS introduction, it also suppressed the mRNA expression of proteins involved in their degradation. Finally, the MYC-positive human hepatocellular carcinoma was characterized by the cytoplasm devoid of lipid accumulation, prominent nucleoli and a higher proliferative activity. Our results demonstrate that in hepatocarcinogenesis induced by both activated AKT and HRAS, activation of endogenous Myc is an enhancing factor and adequate levels of Myc deregulation further facilitate the process with alterations in cellular metabolism.- Published
- 2017
- Full Text
- View/download PDF
12. A novel role for non-ubiquitinated FANCD2 in response to hydroxyurea-induced DNA damage.
- Author
-
Chen X, Bosques L, Sung P, and Kupfer GM
- Abstract
This corrects the article DOI: 10.1038/onc.2015.68.
- Published
- 2017
- Full Text
- View/download PDF
13. Phosphorylation of NFAT3 by CDK3 induces cell transformation and promotes tumor growth in skin cancer.
- Author
-
Xiao T, Zhu JJ, Huang S, Peng C, He S, Du J, Hong R, Chen X, Bode AM, Jiang W, Dong Z, and Zheng D
- Subjects
- Animals, Cell Line, Tumor, Cell Proliferation, Cell Transformation, Neoplastic genetics, Cyclin-Dependent Kinase 3 chemistry, Gene Expression, Gene Expression Regulation, Neoplastic, Humans, Mice, NFATC Transcription Factors genetics, Oncogene Proteins genetics, Oncogene Proteins metabolism, Phosphorylation, Promoter Regions, Genetic, Protein Binding, Skin Neoplasms genetics, Substrate Specificity, Transcriptional Activation, Cell Transformation, Neoplastic metabolism, Cyclin-Dependent Kinase 3 metabolism, NFATC Transcription Factors metabolism, Skin Neoplasms metabolism, Skin Neoplasms pathology
- Abstract
The nuclear factor of activated T cells (NFAT) family proteins are transcription factors that regulate the expression of pro-inflammatory cytokines and other genes during the immune response. Although the NFAT proteins have been extensively investigated in the immune system, their role in cancer progression remains controversial. Here, we report that NFAT3 is highly expressed in various skin cancer cell lines and tumor tissues. Knockdown of endogenous NFAT3 expression by short hairpin RNA (shRNA) significantly inhibited tumor cell proliferation, colony formation and anchorage-independent cell growth. Furthermore, results of the mammalian two-hybrid assay showed that cyclin-dependent kinase 3 (CDK3) directly interacted with NFAT3 and phosphorylated NFAT3 at serine 259 (Ser259), which enhanced the transactivation and transcriptional activity of NFAT3. The phosphorylation site of NFAT3 was critical for epidermal growth factor (EGF)-stimulated cell transformation of the HaCaT immortalized skin cell line and mutation of NFAT3 at Ser259 led to a reduction of colony formation in soft agar. We also found that overexpressing wildtype NFAT3, but not mutant NFAT3-S259A, promoted A431 xenograft tumor growth. Importantly, we showed that CDK3, NFAT3 and phosphorylated NFAT3-Ser259 were highly expressed in skin cancer compared with normal skin tissues. These results provided evidence supporting the oncogenic potential of NFAT3 and suggested that CDK3-mediated phosphorylation of NFAT3 has an important role in skin tumorigenesis.
- Published
- 2017
- Full Text
- View/download PDF
14. Transducin (β)-like 1 X-linked receptor 1 promotes gastric cancer progression via the ERK1/2 pathway.
- Author
-
Zhou Q, Wang X, Yu Z, Wu X, Chen X, Li J, Zhu Z, Liu B, and Su L
- Subjects
- Aged, Animals, Cell Line, Tumor, Cell Movement genetics, Cell Proliferation genetics, Disease Models, Animal, Disease Progression, Epithelial-Mesenchymal Transition, ErbB Receptors metabolism, Female, Gene Expression, Heterografts, Humans, Immunohistochemistry, Male, Matrix Metalloproteinase 7 metabolism, Mice, Middle Aged, Neoplasm Grading, Neoplasm Metastasis, Neoplasm Staging, Nuclear Proteins metabolism, Prognosis, Receptors, Cytoplasmic and Nuclear metabolism, Repressor Proteins metabolism, Signal Transduction, Stomach Neoplasms mortality, Stomach Neoplasms pathology, Tumor Burden, beta Catenin metabolism, MAP Kinase Signaling System, Nuclear Proteins genetics, Receptors, Cytoplasmic and Nuclear genetics, Repressor Proteins genetics, Stomach Neoplasms genetics, Stomach Neoplasms metabolism
- Abstract
Gastric cancer (GC) is one of the most common types of cancer worldwide, and it involves extensive local tumour invasion, metastasis and poor prognosis. Understanding the mechanisms regulating the progression of GC is necessary for the development of effective therapeutic strategies. Transducin (β)-like 1 X-linked receptor 1 (TBL1XR1) is an important regulator controlling gene activation and repression, which has been thought to be involved in tumorigenesis. However, the role of TBL1XR1 in human GC remains largely unknown. Here, we find that TBL1XR1 is aberrantly expressed in human GC tissues, and TBL1XR1 levels are highly correlated with local tumour invasion, late tumor, lymph node, metastasis (TNM) stage and poor prognosis. Knockdown of TBL1XR1 by shRNA inhibits GC cell proliferation, migration, invasion, epithelial-mesenchymal transition (EMT) in vitro, as well as tumorigenesis and peritoneal metastasis in vivo, whereas overexpression of TBL1XR1 produces the opposite effects. These effects are mediated by activation of the ERK1/2 signalling pathway, and inhibition of this pathway with a specific ERK1/2 inhibitor (U0126) significantly impairs the tumour-promoting effects induced by TBL1XR1. Moreover, TBL1XR1 mediated ERK1/2 activation is dependent on the β-catenin/MMP7/EGFR signalling pathway. In conclusion, TBL1XR1 contributes to GC tumorigenesis and progression through the activation of the β-catenin/MMP7/EGFR/ERK signalling pathway and may act as a new therapeutic target for GC.
- Published
- 2017
- Full Text
- View/download PDF
15. eEF-2 kinase is a critical regulator of Warburg effect through controlling PP2A-A synthesis.
- Author
-
Cheng Y, Ren X, Yuan Y, Shan Y, Li L, Chen X, Zhang L, Takahashi Y, Yang JW, Han B, Liao J, Li Y, Harvey H, Ryazanov A, Robertson GP, Wan G, Liu D, Chen AF, Tao Y, and Yang JM
- Subjects
- Animals, Breast Neoplasms enzymology, Breast Neoplasms genetics, Cell Line, Tumor, Elongation Factor 2 Kinase genetics, Female, Heterografts, Humans, MCF-7 Cells, Mice, Mice, Nude, Protein Phosphatase 2 genetics, Transfection, Elongation Factor 2 Kinase metabolism, Protein Phosphatase 2 biosynthesis
- Abstract
Cancer cells predominantly metabolize glucose by glycolysis to produce energy in order to meet their metabolic requirement, a phenomenon known as Warburg effect. Although Warburg effect is considered a peculiarity critical for survival and proliferation of cancer cells, the regulatory mechanisms behind this phenomenon remain incompletely understood. We report here that eukaryotic elongation factor-2 kinase (eEF-2K), a negative regulator of protein synthesis, has a critical role in promoting glycolysis in cancer cells. We showed that deficiency in eEF-2K significantly reduced the uptake of glucose and decreased the productions of lactate and adenosine triphosphate in tumor cells and in the Ras-transformed mouse embryonic fibroblasts. We further demonstrated that the promotive effect of eEF-2K on glycolysis resulted from the kinase-mediated restriction of synthesis of the protein phosphatase 2A-A (PP2A-A), a key factor that facilitates the ubiquitin-proteasomal degradation of c-Myc protein, as knockdown of eEF-2K expression led to a significant increase in PP2A-A protein synthesis and remarkable downregulation of c-Myc and pyruvate kinase M2 isoform, the key glycolytic enzyme transcriptionally activated by c-Myc. In addition, depletion of eEF-2K reduced the ability of the transformed cells to proliferate and enhanced the sensitivity of tumor cells to chemotherapy both in vitro and in vivo. These results, which uncover a role of the eEF-2K-mediated control of PP2A-A in tumor cell glycolysis, provide new insights into the regulation of the Warburg effect.
- Published
- 2016
- Full Text
- View/download PDF
16. Myeloid translocation genes differentially regulate colorectal cancer programs.
- Author
-
Parang B, Bradley AM, Mittal MK, Short SP, Thompson JJ, Barrett CW, Naik RD, Bilotta AJ, Washington MK, Revetta FL, Smith JJ, Chen X, Wilson KT, Hiebert SW, and Williams CS
- Subjects
- Animals, Colorectal Neoplasms metabolism, Colorectal Neoplasms pathology, Female, Humans, Male, Mice, Mice, Inbred C57BL, Nuclear Proteins metabolism, Repressor Proteins metabolism, Signal Transduction, Transcription Factors metabolism, Translocation, Genetic, Tumor Suppressor Proteins metabolism, Colorectal Neoplasms genetics, Nuclear Proteins genetics, Repressor Proteins genetics, Transcription Factors genetics, Tumor Suppressor Proteins genetics
- Abstract
Myeloid translocation genes (MTGs), originally identified as chromosomal translocations in acute myelogenous leukemia, are transcriptional corepressors that regulate hematopoietic stem cell programs. Analysis of The Cancer Genome Atlas (TCGA) database revealed that MTGs were mutated in epithelial malignancy and suggested that loss of function might promote tumorigenesis. Genetic deletion of MTGR1 and MTG16 in the mouse has revealed unexpected and unique roles within the intestinal epithelium. Mtgr1
-/- mice have progressive depletion of all intestinal secretory cells, and Mtg16-/- mice have a decrease in goblet cells. Furthermore, both Mtgr1-/- and Mtg16-/- mice have increased intestinal epithelial cell proliferation. We thus hypothesized that loss of MTGR1 or MTG16 would modify Apc1638/+ -dependent intestinal tumorigenesis. Mtgr1-/- mice, but not Mtg16-/- mice, had a 10-fold increase in tumor multiplicity. This was associated with more advanced dysplasia, including progression to invasive adenocarcinoma, and augmented intratumoral proliferation. Analysis of chromatin immunoprecipitation sequencing data sets for MTGR1 and MTG16 targets indicated that MTGR1 can regulate Wnt and Notch signaling. In support of this, immunohistochemistry and gene expression analysis revealed that both Wnt and Notch signaling pathways were hyperactive in Mtgr1-/- tumors. Furthermore, in human colorectal cancer (CRC) samples MTGR1 was downregulated at both the transcript and protein level. Overall our data indicates that MTGR1 has a context-dependent effect on intestinal tumorigenesis., Competing Interests: There are no conflicts of interest to disclose- Published
- 2016
- Full Text
- View/download PDF
17. Overexpression of maelstrom promotes bladder urothelial carcinoma cell aggressiveness by epigenetically downregulating MTSS1 through DNMT3B.
- Author
-
Li XD, Zhang JX, Jiang LJ, Wang FW, Liu LL, Liao YJ, Jin XH, Chen WH, Chen X, Guo SJ, Zhou FJ, Zeng YX, Guan XY, Liu ZW, and Xie D
- Subjects
- Animals, Carrier Proteins biosynthesis, Cell Line, Tumor, Cell Proliferation genetics, DNA-Binding Proteins, Down-Regulation, Epigenesis, Genetic, Heterografts, Humans, Male, Mice, Mice, SCID, MicroRNAs genetics, MicroRNAs metabolism, Microfilament Proteins metabolism, Middle Aged, Neoplasm Proteins metabolism, Transcription Factors, Urinary Bladder Neoplasms metabolism, Urinary Bladder Neoplasms pathology, DNA Methyltransferase 3B, Carrier Proteins genetics, DNA (Cytosine-5-)-Methyltransferases genetics, Microfilament Proteins genetics, Neoplasm Proteins genetics, Urinary Bladder Neoplasms genetics
- Abstract
We have recently identified and characterized a novel oncogene, maelstrom (MAEL) from 1q24, in the pathogenesis of hepatocellular carcinoma. In this study, MAEL was investigated for its oncogenic role in urothelial carcinoma of the bladder (UCB) tumorigenesis/aggressiveness and underlying molecular mechanisms. Here, we report that overexpression of MAEL in UCB is important in the acquisition of an aggressive and/or poor prognostic phenotype. In UCB cell lines, knockdown of MAEL by short hairpin RNA is sufficient to inhibit cell growth, invasiveness/metastasis and suppressed epithelial-mesenchymal transition (EMT), whereas ectopic overexpression of MAEL promoted cell growth, invasive and/or metastatic capacity and enhanced EMT both in vitro and in vivo. We further demonstrate that MAEL could induce UCB cell EMT by downregulating a critical downstream target, the metastasis suppressor 1 (MTSS1) gene, ultimately leading to an increased invasiveness of cancer cells. Notably, overexpression of MAEL in UCB cells substantially enhanced the enrichment of DNA methyltrans-ferase (DNMT)3B and histone deacetylase (HDAC)1/2 on the promoter of the MTSS1, and thereby epigenetically suppressing the MTSS1 transcription. Downregulation of MTSS1 by MAEL in UCB cells is partially dependent on DNMT3B. Furthermore, we identify that beside the gene amplification of MAEL, miR-186 is a key negative regulator of MAEL and downregulation of miR-186 is another important mechanism for MAEL overexpression in UCBs. These data suggest that overexpression of MAEL, caused by gene amplification and/or decreased miR-186, has a critical oncogenic role in UCB pathogenesis by downregulation of MTSS1, and MAEL could be used as a novel prognostic marker and/or effective therapeutic target for human UCB.
- Published
- 2016
- Full Text
- View/download PDF
18. A novel nickel complex works as a proteasomal deubiquitinase inhibitor for cancer therapy.
- Author
-
Zhao C, Chen X, Zang D, Lan X, Liao S, Yang C, Zhang P, Wu J, Li X, Liu N, Liao Y, Huang H, Shi X, Jiang L, Liu X, He Z, Dou QP, Wang X, and Liu J
- Subjects
- Animals, Apoptosis drug effects, Cell Line, Tumor, Cell Proliferation drug effects, Cell Survival drug effects, DNA Damage drug effects, Disease Models, Animal, Humans, Membrane Potential, Mitochondrial drug effects, Mice, Proteasome Endopeptidase Complex metabolism, Tumor Burden drug effects, Ubiquitin metabolism, Xenograft Model Antitumor Assays, Antineoplastic Agents pharmacology, Nickel pharmacology, Proteasome Inhibitors pharmacology
- Abstract
Based on the central role of the ubiquitin-proteasome system (UPS) in the degradation of cellular proteins, proteasome inhibition has been considered an attractive approach for anticancer therapy. Deubiquitinases (DUBs) remove ubiquitin conjugates from diverse substrates; therefore, they are essential regulators of the UPS. DUB inhibitors, especially the inhibitors of proteasomal DUBs are becoming a research hotspot in targeted cancer therapy. Previous studies have shown that metal complexes, such as copper and zinc complexes, can induce cancer cell apoptosis through inhibiting UPS function. Moreover, we have found that copper pyrithione inhibits both 19S proteasome-associated DUBs and 20S proteasome activity with a mechanism distinct from that of the classical 20S proteasome inhibitor bortezomib. In the present study, we reveal that (i) nickel pyrithione complex (NiPT) potently inhibits the UPS via targeting the 19S proteasome-associated DUBs (UCHL5 and USP14), without effecting on the 20S proteasome; (ii) NiPT selectively induces proteasome inhibition and apoptosis in cultured tumor cells and cancer cells from acute myeloid leukemia human patients; and (iii) NiPT inhibits proteasome function and tumor growth in nude mice. This study, for the first time, uncovers a nickel complex as an effective inhibitor of the 19S proteasomal DUBs and suggests a potentially new strategy for cancer treatment.
- Published
- 2016
- Full Text
- View/download PDF
19. Overexpression of PIK3CA in murine head and neck epithelium drives tumor invasion and metastasis through PDK1 and enhanced TGFβ signaling.
- Author
-
Du L, Chen X, Cao Y, Lu L, Zhang F, Bornstein S, Li Y, Owens P, Malkoski S, Said S, Jin F, Kulesz-Martin M, Gross N, Wang XJ, and Lu SL
- Subjects
- Animals, Animals, Genetically Modified, Cell Proliferation genetics, Cell Transformation, Neoplastic genetics, Class I Phosphatidylinositol 3-Kinases, Disease Models, Animal, Epithelial-Mesenchymal Transition genetics, Epithelium pathology, Female, Gene Expression Regulation, Neoplastic, Head and Neck Neoplasms pathology, Humans, Lymphatic Metastasis, Male, Mice, Mutation, Neoplasm Invasiveness pathology, Neoplastic Stem Cells pathology, Phosphatidylinositol 3-Kinases genetics, Pyruvate Dehydrogenase Acetyl-Transferring Kinase, Signal Transduction, Head and Neck Neoplasms genetics, Neoplasm Invasiveness genetics, Phosphatidylinositol 3-Kinases biosynthesis, Protein Serine-Threonine Kinases genetics, Transforming Growth Factor beta genetics
- Abstract
Head and neck squamous cell carcinoma (HNSCC) patients have a poor prognosis, with invasion and metastasis as major causes of mortality. The phosphatidylinositol 3-kinase (PI3K) pathway regulates a wide range of cellular processes crucial for tumorigenesis, and PIK3CA amplification and mutation are among the most common genetic alterations in human HNSCC. Compared with the well-documented roles of the PI3K pathway in cell growth and survival, the roles of the PI3K pathway in tumor invasion and metastasis have not been well delineated. We generated a PIK3CA genetically engineered mouse model (PIK3CA-GEMM) in which wild-type PIK3CA is overexpressed in head and neck epithelium. Although PIK3CA overexpression alone was not sufficient to initiate HNSCC formation, it significantly increased tumor susceptibility in an oral carcinogenesis mouse model. PIK3CA overexpression in mouse oral epithelium increased tumor invasiveness and metastasis by increasing epithelial-mesenchymal transition and by enriching a cancer stem cell phenotype in tumor epithelial cells. In addition to these epithelial alterations, we also observed marked inflammation in tumor stroma. AKT is a central signaling mediator of the PI3K pathway. However, molecular analysis suggested that progression of PIK3CA-driven HNSCC is facilitated by 3-phosphoinositide-dependent protein kinase (PDK1) and enhanced transforming growth factor β (TGFβ) signaling rather than by AKT. Examination of human HNSCC clinical samples revealed that both PIK3CA and PDK1 protein levels correlated with tumor progression, highlighting the significance of this pathway. In summary, our results offer significant insight into how PIK3CA overexpression drives HNSCC invasion and metastasis, providing a rationale for targeting PI3K/PDK1 and TGFβ signaling in advanced HNSCC patients with PIK3CA amplification., Competing Interests: The authors declare no conflict of interest.
- Published
- 2016
- Full Text
- View/download PDF
20. Oxidative stress enables Epstein-Barr virus-induced B-cell transformation by posttranscriptional regulation of viral and cellular growth-promoting factors.
- Author
-
Chen X, Kamranvar SA, and Masucci MG
- Subjects
- B-Lymphocytes metabolism, B-Lymphocytes pathology, Cell Proliferation genetics, Cell Transformation, Viral genetics, Cells, Cultured, DNA Damage, Epstein-Barr Virus Nuclear Antigens genetics, Epstein-Barr Virus Nuclear Antigens metabolism, Gene Expression Regulation, Herpesvirus 4, Human genetics, Host-Pathogen Interactions, Humans, Immunoblotting, Microscopy, Fluorescence, Phosphorylation, Reactive Oxygen Species metabolism, Reverse Transcriptase Polymerase Chain Reaction, STAT3 Transcription Factor genetics, STAT3 Transcription Factor metabolism, Viral Matrix Proteins genetics, Viral Matrix Proteins metabolism, Viral Proteins genetics, Viral Proteins metabolism, B-Lymphocytes virology, Cell Transformation, Viral physiology, Herpesvirus 4, Human physiology, Oxidative Stress
- Abstract
Infection of human B lymphocytes by Epstein-Barr virus (EBV) leads to the establishment of immortalized lymphoblastoid cell lines (LCLs) that are widely used as a model of viral oncogenesis. An early consequence of infection is the induction of DNA damage and activation of the DNA damage response, which limits the efficiency of growth transformation. The cause of the DNA damage remains poorly understood. We have addressed this question by comparing the response of B lymphocytes infected with EBV or stimulated with a potent B-cell mitogen. We found that although the two stimuli induce comparable proliferation during the first 10 days of culture, the EBV-infected blasts showed significantly higher levels of DNA damage, which correlated with stronger and sustained accumulation of reactive oxygen species (ROS). Treatment with ROS scavengers decreased DNA damage in both mitogen-stimulated and EBV-infected cells. However, while mitogen-induced proliferation was slightly improved, the proliferation of EBV-infected cells and the establishment of LCLs were severely impaired. Quenching of ROS did not affect the kinetics and magnitude of viral gene expression but was associated with selective downregulation of the viral LMP1 and phosphorylated cellular transcription factor STAT3 that have key roles in transformation. Analysis of the mechanism by which high levels of ROS support LMP1 expression revealed selective inhibition of viral microRNAs that target the LMP1 transcript. Our study provides novel insights into the role of EBV-induced oxidative stress in promoting B-cell immortalization and malignant transformation.
- Published
- 2016
- Full Text
- View/download PDF
21. Gankyrin facilitates follicle-stimulating hormone-driven ovarian cancer cell proliferation through the PI3K/AKT/HIF-1α/cyclin D1 pathway.
- Author
-
Chen J, Bai M, Ning C, Xie B, Zhang J, Liao H, Xiong J, Tao X, Yan D, Xi X, Chen X, Yu Y, Bast RC, Zhang Z, Feng Y, and Zheng W
- Subjects
- Carcinoma, Ovarian Epithelial, Cell Line, Tumor, Cell Proliferation drug effects, Female, Humans, Signal Transduction drug effects, Ubiquitination drug effects, Up-Regulation drug effects, Cyclin D1 metabolism, Follicle Stimulating Hormone pharmacology, Hypoxia-Inducible Factor 1, alpha Subunit metabolism, Neoplasms, Glandular and Epithelial pathology, Ovarian Neoplasms pathology, Phosphatidylinositol 3-Kinases metabolism, Proteasome Endopeptidase Complex metabolism, Proto-Oncogene Proteins metabolism, Proto-Oncogene Proteins c-akt metabolism
- Abstract
Gankyrin is a regulatory subunit of the 26kD proteasome complex. As a novel oncoprotein, gankyrin is expressed aberrantly in cancers from several different sites and has been shown to contribute to oncogenesis in endometrial and cervical carcinomas. Neither gankyrin's contribution to the development of epithelial ovarian cancer nor its interaction with follicle-stimulating hormone (FSH)-driven proliferation in ovarian cancer has been studied. Here we have found that gankyrin is overexpressed in ovarian cancers compared with benign ovarian cystadenomas and that gankyrin regulates FSH upregulation of cyclin D1. Importantly, gankyrin regulates PI3K/AKT signaling by downregulating PTEN. Prolonged AKT activation by FSH stimulation of the FSH receptor (FSHR) promotes gankyrin expression, which, in turn, enhances AKT activation by inhibiting PTEN. Overexpression of gankyrin decreases hypoxia inducible factor-1α (HIF-1α) protein levels, but has little effect on HIF-1α mRNA levels, which could be attributed to gankyrin mediating HIF-1α protein stability via the ubiquitin-proteasome pathway. Reduction in HIF-1α protein stability led to attenuation of the binding with cyclin D1 promoter, resulted in abolishment of the negative regulation of cyclin D1 by HIF-1α, which promotes proliferation of ovarian cancer cells. Our results document that gankyrin regulates HIF-1α protein stability and cyclin D1 expression, ultimately mediating FSH-driven ovarian cancer cell proliferation.
- Published
- 2016
- Full Text
- View/download PDF
22. Structural basis of how stress-induced MDMX phosphorylation activates p53.
- Author
-
Chen X, Gohain N, Zhan C, Lu WY, Pazgier M, and Lu W
- Subjects
- Binding Sites, Cell Cycle Proteins, Humans, Hydrogen Bonding, Ligands, Models, Molecular, Neoplasm Proteins chemistry, Neoplasm Proteins metabolism, Nuclear Proteins chemical synthesis, Nuclear Proteins metabolism, Oligopeptides metabolism, Peptide Fragments chemical synthesis, Peptide Fragments chemistry, Peptide Fragments metabolism, Phosphorylation, Phosphotyrosine chemistry, Protein Binding, Protein Conformation, Protein Interaction Mapping, Protein Processing, Post-Translational, Proto-Oncogene Proteins chemical synthesis, Proto-Oncogene Proteins metabolism, Structure-Activity Relationship, Surface Plasmon Resonance, Tumor Suppressor Protein p53 metabolism, DNA Damage, Nuclear Proteins chemistry, Proto-Oncogene Proteins chemistry, Tumor Suppressor Protein p53 chemistry
- Abstract
The tumor-suppressor protein p53 is tightly controlled in normal cells by its two negative regulators--the E3 ubiquitin ligase MDM2 and its homolog MDMX. Under stressed conditions such as DNA damage, p53 escapes MDM2- and MDMX-mediated functional inhibition and degradation, acting to prevent damaged cells from proliferating through induction of cell cycle arrest, DNA repair, senescence or apoptosis. Ample evidence suggests that stress signals induce phosphorylation of MDM2 and MDMX, leading to p53 activation. However, the structural basis of stress-induced p53 activation remains poorly understood because of the paucity of technical means to produce site-specifically phosphorylated MDM2 and MDMX proteins for biochemical and biophysical studies. Herein, we report total chemical synthesis, via native chemical ligation, and functional characterization of (24-108)MDMX and its Tyr99-phosphorylated analog with respect to their ability to interact with a panel of p53-derived peptide ligands and PMI, a p53-mimicking but more potent peptide antagonist of MDMX, using FP and surface plasmon resonance techniques. Phosphorylation of MDMX at Tyr99 weakens peptide binding by approximately two orders of magnitude. Comparative X-ray crystallographic analyses of MDMX and of pTyr99 MDMX in complex with PMI as well as modeling studies reveal that the phosphate group of pTyr99 imposes extensive steric clashes with the C-terminus of PMI or p53 peptide and induces a significant lateral shift of the peptide ligand, contributing to the dramatic decrease in the binding affinity of MDMX for p53. Because DNA damage activates c-Abl tyrosine kinase that phosphorylates MDMX at Tyr99, our findings afford a rare glimpse at the structural level of how stress-induced MDMX phosphorylation dislodges p53 from the inhibitory complex and activates it in response to DNA damage.
- Published
- 2016
- Full Text
- View/download PDF
23. Role of p14ARF-HDM2-p53 axis in SOX6-mediated tumor suppression.
- Author
-
Wang J, Ding S, Duan Z, Xie Q, Zhang T, Zhang X, Wang Y, Chen X, Zhuang H, and Lu F
- Subjects
- Animals, Cell Proliferation genetics, Female, Genes, Tumor Suppressor physiology, HeLa Cells, Humans, Mice, Mice, Inbred C57BL, Mice, Nude, Nucleophosmin, Protein Structure, Tertiary genetics, SOXD Transcription Factors chemistry, SOXD Transcription Factors genetics, Signal Transduction physiology, Tumor Cells, Cultured, Cell Transformation, Neoplastic genetics, Proto-Oncogene Proteins c-mdm2 physiology, SOXD Transcription Factors physiology, Tumor Suppressor Protein p14ARF physiology, Tumor Suppressor Protein p53 pharmacokinetics, Tumor Suppressor Protein p53 physiology
- Abstract
Sex-determining region Y box 6 (SOX6) has been described as a tumor-suppressor gene in several cancers. Our previous work has suggested that SOX6 upregulated p21(Waf1/Cip1)(p21) expression in a p53-dependent manner; however, the underlying mechanism has remained elusive. In this study, we confirmed that SOX6 can suppress cell proliferation in vitro and in vivo by stabilizing p53 protein and subsequently upregulating p21. Co-immunoprecipitation and immunocytofluorescence assays demonstrated that SOX6 can promote formation of the p14ARF-HDM2-p53 ternary complex by promoting translocation of p14ARF (p14 alternate reading frame tumor suppressor) to the nucleoplasm, thereby inhibiting HDM2-mediated p53 nuclear export and degradation. Chromatin immunoprecipitation combined with PCR assay proved that SOX6 can bind to a potential binding site in the regulatory region of the c-Myc gene. Furthermore, we confirmed that SOX6 can downregulate the expression of c-Myc, as well as its direct target gene nucleophosmin 1 (NPM1), and that the SOX6-induced downregulation of NPM1 is linked to translocation of p14ARF to the nucleoplasm. Finally, we showed that the highly conserved high-mobility group (HMG) domain of SOX6 is required for SOX6-mediated p53 stabilization and tumor inhibitory activity. Collectively, these results reveal a new mechanism of SOX6-mediated tumor suppression involving p21 upregulation via the p14ARF-HDM2-p53 axis in an HMG domain-dependent manner.
- Published
- 2016
- Full Text
- View/download PDF
24. Cyclin-dependent kinase 2 (CDK2) is a key mediator for EGF-induced cell transformation mediated through the ELK4/c-Fos signaling pathway.
- Author
-
Peng C, Zeng W, Su J, Kuang Y, He Y, Zhao S, Zhang J, Ma W, Bode AM, Dong Z, and Chen X
- Subjects
- Animals, Cell Cycle genetics, Cell Line, Cell Proliferation, Cell Transformation, Neoplastic genetics, Cyclin-Dependent Kinase 2 biosynthesis, Epidermal Growth Factor genetics, Epidermal Growth Factor metabolism, Humans, Melanoma pathology, Mice, Phosphorylation, Transcriptional Activation genetics, ets-Domain Protein Elk-4 genetics, Cyclin-Dependent Kinase 2 genetics, Melanoma genetics, Proto-Oncogene Proteins c-fos biosynthesis, ets-Domain Protein Elk-4 biosynthesis
- Abstract
Cyclin-dependent kinase 2 (CDK2) is a known regulator in the cell cycle control of the G1/S and S/G2 transitions. However, the role of CDK2 in tumorigenesis is controversial. Evidence from knockout mice as well as colon cancer cell lines indicated that CDK2 is dispensable for cell proliferation. In this study, we found that ectopic CDK2 enhances Ras (G12V)-induced foci formation and knocking down CDK2 expression markedly decreases epidermal growth factor (EGF)-induced cell transformation mediated through the downregulation of c-fos expression. Interestingly, CDK2 directly phosphorylates ELK4 at Thr194 and Ser387 and regulates the ELK4 transcriptional activity, which serves as a mechanism to regulate c-fos expression. In addition, ELK4 is overexpressed in melanoma and knocking down the ELK4 or CDK2 expression significantly attenuated the malignant phenotype of melanoma cells. Taken together, our study reveals a novel function of CDK2 in EGF-induced cell transformation and the associated signal transduction pathways. This indicates that CDK2 is a useful molecular target for the chemoprevention and therapy against skin cancer.
- Published
- 2016
- Full Text
- View/download PDF
25. Sirtuin 3 inhibits hepatocellular carcinoma growth through the glycogen synthase kinase-3β/BCL2-associated X protein-dependent apoptotic pathway.
- Author
-
Song CL, Tang H, Ran LK, Ko BC, Zhang ZZ, Chen X, Ren JH, Tao NN, Li WY, Huang AL, and Chen J
- Subjects
- Apoptosis physiology, Carcinoma, Hepatocellular metabolism, Cell Line, Tumor, Female, Glycogen Synthase Kinase 3 beta, Hep G2 Cells, Humans, Liver Neoplasms metabolism, Male, Middle Aged, Transfection, Carcinoma, Hepatocellular pathology, Glycogen Synthase Kinase 3 metabolism, Liver Neoplasms pathology, Sirtuin 3 metabolism, bcl-2-Associated X Protein metabolism
- Abstract
SIRT3 is a class III histone deacetylase that has been implicated in a variety of cancers. The role of SIRT3 in hepatocellular carcinoma (HCC) remains elusive. In this study, we found that SIRT3 expression was frequently repressed in HCC and its downregulation was closely associated with tumor grade and size. Ectopic expression of SIRT3 inhibited cell growth and induced apoptosis in HCC cells, whereas depletion of SIRT3 in immortalized hepatocyte promoted cell growth and decreased epirubicin-induced apoptosis. Mechanistic studies revealed that SIRT3 deacetylated and activated glycogen synthase kinase-3β (GSK-3β), which subsequently induced expression and mitochondrial translocation of the pro-apoptotic protein BCL2-associated X protein (Bax) to promote apoptosis. GSK-3β inhibitor or gene silencing of BAX reversed SIRT3-induced growth inhibition and apoptosis. Furthermore, SIRT3 overexpression also suppressed tumor growth in vivo. Together, this study reveals a role of SIRT3/GSK-3β/Bax signaling pathway in the suppression of HCC growth, and also suggests that targeting this pathway may represent a potential therapeutic approach for HCC treatment.
- Published
- 2016
- Full Text
- View/download PDF
26. A novel role for non-ubiquitinated FANCD2 in response to hydroxyurea-induced DNA damage.
- Author
-
Chen X, Bosques L, Sung P, and Kupfer GM
- Subjects
- DNA-Binding Proteins metabolism, DNA-Directed DNA Polymerase metabolism, Fanconi Anemia metabolism, Fanconi Anemia Complementation Group D2 Protein deficiency, Gene Knockdown Techniques, HEK293 Cells, Humans, Hydroxyurea adverse effects, Proliferating Cell Nuclear Antigen metabolism, Rad51 Recombinase metabolism, Ubiquitin-Protein Ligases, Ubiquitination, DNA Damage, Fanconi Anemia drug therapy, Fanconi Anemia genetics, Fanconi Anemia Complementation Group D2 Protein genetics, Fanconi Anemia Complementation Group D2 Protein metabolism, Hydroxyurea pharmacology
- Abstract
Fanconi anemia (FA) is a genetic disease of bone marrow failure, cancer susceptibility, and sensitivity to DNA crosslinking agents. FANCD2, the central protein of the FA pathway, is monoubiquitinated upon DNA damage, such as crosslinkers and replication blockers such as hydroxyurea (HU). Even though FA cells demonstrate unequivocal sensitivity to crosslinkers, such as mitomycin C (MMC), we find that they are largely resistant to HU, except for cells absent for expression of FANCD2. FANCD2, RAD51 and RAD18 form a complex, which is enhanced upon HU exposure. Surprisingly, although FANCD2 is required for this enhanced interaction, its monoubiquitination is not. Similarly, non-ubiquitinated FANCD2 can still support proliferation cell nuclear antigen (PCNA) monoubiquitination. RAD51, but not BRCA2, is also required for PCNA monoubiquitination in response to HU, suggesting that this function is independent of homologous recombination (HR). We further show that translesion (TLS) polymerase PolH chromatin localization is decreased in FANCD2 deficient cells, FANCD2 siRNA knockdown cells and RAD51 siRNA knockdown cells, and PolH knockdown results in HU sensitivity only. Our data suggest that FANCD2 and RAD51 have an important role in PCNA monoubiquitination and TLS in a FANCD2 monoubiquitination and HR-independent manner in response to HU. This effect is not observed with MMC treatment, suggesting a non-canonical function for the FA pathway in response to different types of DNA damage.
- Published
- 2016
- Full Text
- View/download PDF
27. Repression of Hox genes by LMP1 in nasopharyngeal carcinoma and modulation of glycolytic pathway genes by HoxC8.
- Author
-
Jiang Y, Yan B, Lai W, Shi Y, Xiao D, Jia J, Liu S, Li H, Lu J, Li Z, Chen L, Chen X, Sun L, Muegge K, Cao Y, and Tao Y
- Subjects
- Cells, Cultured, Citric Acid Cycle, DNA Methylation, Dioxygenases physiology, Glycolysis, Humans, Nasopharyngeal Neoplasms metabolism, RNA Polymerase II metabolism, Genes, Homeobox physiology, Homeodomain Proteins physiology, Nasopharyngeal Neoplasms genetics, Viral Matrix Proteins physiology
- Abstract
Epstein-Barr virus (EBV) causes human lymphoid malignancies, and the EBV product latent membrane protein 1 (LMP1) has been identified as an oncogene in epithelial carcinomas such as nasopharyngeal carcinoma (NPC). EBV can epigenetically reprogram lymphocyte-specific processes and induce cell immortalization. However, the interplay between LMP1 and the NPC host cell remains largely unknown. Here, we report that LMP1 is important to establish the Hox gene expression signature in NPC cell lines and tumor biopsies. LMP1 induces repression of several Hox genes in part via stalling of RNA polymerase II (RNA Pol II). Pol II stalling can be overcome by irradiation involving the epigenetic regulator TET3. Furthermore, we report that HoxC8, one of the genes silenced by LMP1, has a role in tumor growth. Ectopic expression of HoxC8 inhibits NPC cell growth in vitro and in vivo, modulates glycolysis and regulates the expression of tricarboxylic acid (TCA) cycle-related genes. We propose that viral latency products may repress via stalling key mediators that in turn modulate glycolysis.
- Published
- 2015
- Full Text
- View/download PDF
28. PPM1D phosphatase, a target of p53 and RBM38 RNA-binding protein, inhibits p53 mRNA translation via dephosphorylation of RBM38.
- Author
-
Zhang M, Xu E, Zhang J, and Chen X
- Subjects
- 3' Untranslated Regions genetics, Blotting, Western, Breast Neoplasms genetics, Breast Neoplasms pathology, Colonic Neoplasms genetics, Colonic Neoplasms pathology, Female, Flow Cytometry, Humans, Immunoenzyme Techniques, Immunoprecipitation, Phosphoprotein Phosphatases antagonists & inhibitors, Phosphoprotein Phosphatases genetics, Phosphorylation, Protein Phosphatase 2C, RNA Processing, Post-Transcriptional, RNA, Messenger genetics, RNA, Small Interfering genetics, RNA-Binding Proteins antagonists & inhibitors, RNA-Binding Proteins genetics, Real-Time Polymerase Chain Reaction, Reverse Transcriptase Polymerase Chain Reaction, Tumor Cells, Cultured, Tumor Suppressor Protein p53 antagonists & inhibitors, Tumor Suppressor Protein p53 genetics, Breast Neoplasms metabolism, Colonic Neoplasms metabolism, Gene Expression Regulation, Neoplastic, Phosphoprotein Phosphatases metabolism, RNA, Messenger metabolism, RNA-Binding Proteins metabolism, Tumor Suppressor Protein p53 metabolism
- Abstract
PPM1D phosphatase, also called wild-type p53-induced phosphatase 1, promotes tumor development by inactivating the p53 tumor suppressor pathway. RBM38 RNA-binding protein, also called RNPC1 and a target of p53, inhibits p53 messenger RNA (mRNA) translation, which can be reversed by GSK3 protein kinase via phosphorylation of RBM38 at serine 195. Here we showed that ectopic expression of RBM38 increases, whereas knockdown of RBM38 inhibits, PPM1D mRNA translation. Consistent with this, we found that RBM38 directly binds to PPM1D 3'-untranslated region (3'-UTR) and promotes expression of a heterologous reporter gene that carries PPM1D 3'-UTR in a dose-dependent manner. Interestingly, we showed that PPM1D directly interacts with and dephosphorylates RBM38 at serine 195. Furthermore, we showed that PPM1D modulates p53 mRNA translation and p53-dependent growth suppression through dephosphorylation of RBM38. These findings provide evidence that the crosstalk between PPM1D and RBM38, both of which are targets and modulators of p53, has a critical role in p53 expression and activity.
- Published
- 2015
- Full Text
- View/download PDF
29. Pediatric solid tumor genomics and developmental pliancy.
- Author
-
Chen X, Pappo A, and Dyer MA
- Subjects
- Child, Epigenesis, Genetic, Gene Expression Regulation, Neoplastic, Genes, Neoplasm, Genomics, Humans, Mutation, Neoplasms pathology, Neoplasms genetics
- Abstract
Pediatric solid tumors are remarkably diverse in their cellular origins, developmental timing and clinical features. Over the last 5 years, there have been significant advances in our understanding of the genetic lesions that contribute to the initiation and progression of pediatric solid tumors. To date, over 1000 pediatric solid tumors have been analyzed by Next-Generation Sequencing. These genomic data provide the foundation to launch new research efforts to address one of the fundamental questions in cancer biology-why are some cells more susceptible to malignant transformation by particular genetic lesions at discrete developmental stages than others? Because of their developmental, molecular, cellular and genetic diversity, pediatric solid tumors provide an ideal platform to begin to answer this question. In this review, we highlight the diversity of pediatric solid tumors and provide a new framework for studying the cellular and developmental origins of pediatric cancer. We also introduce a new unifying concept called cellular pliancy as a possible explanation for susceptibility to cancer and the developmental origins of pediatric solid tumors.
- Published
- 2015
- Full Text
- View/download PDF
30. Claudin-7 expression induces mesenchymal to epithelial transformation (MET) to inhibit colon tumorigenesis.
- Author
-
Bhat AA, Pope JL, Smith JJ, Ahmad R, Chen X, Washington MK, Beauchamp RD, Singh AB, and Dhawan P
- Subjects
- Adenocarcinoma metabolism, Adenocarcinoma mortality, Adenoma metabolism, Adenoma mortality, Animals, Colonic Neoplasms metabolism, Colonic Neoplasms mortality, Epithelial Cells metabolism, HT29 Cells, Humans, MAP Kinase Signaling System, Mice, Nude, Neoplasm Transplantation, Transcriptome, Tumor Burden, rab GTP-Binding Proteins metabolism, Adenocarcinoma pathology, Adenoma pathology, Carcinogenesis metabolism, Claudins physiology, Colonic Neoplasms pathology, Epithelial-Mesenchymal Transition
- Abstract
In normal colon, claudin-7 is one of the highly expressed claudin proteins and its knockdown in mice results in altered epithelial cell homeostasis and neonatal death. Notably, dysregulation of the epithelial homeostasis potentiates oncogenic transformation and growth. However, the role of claudin-7 in the regulation of colon tumorigenesis remains poorly understood. Using a large colorectal cancer (CRC) patient database and mouse models of colon cancer, we found claudin-7 expression to be significantly downregulated in cancer samples. Most notably, forced claudin-7 expression in poorly differentiated and highly metastatic SW620 colon cancer cells induced epithelial characteristics and inhibited their growth in soft agar and tumor growth in vivo. By contrast, knockdown of claudin-7 in HT-29 or DLD-1 cells induced epithelial-to-mesenchymal transition (EMT), colony formation, xenograft-tumor growth in athymic mice and invasion. Importantly, a claudin-7 signature gene profile generated by overlapping the DEGs (differentially expressed genes in a high-throughput transcriptome analysis using claudin-7-manipulated cells) with human claudin-7 signature genes identified high-risk CRC patients. Furthermore, Rab25, a colon cancer suppressor and regulator of the polarized cell trafficking constituted one of the highly upregulated DEGs in claudin-7 overexpressing cells. Notably, silencing of Rab25 expression counteracted the effects of claudin-7 expression and not only increased proliferation and cell invasion but also increased the expression of p-Src and mitogen-activated protein kinase-extracellular signal-regulated kinase 1/2 that were suppressed upon claudin-7 overexpression. Of interest, CRC cell lines, which exhibited decreased claudin-7 expression, also exhibited promoter DNA hypermethylation, a modification associated with transcriptional silencing. Taken together, our data demonstrate a previously undescribed role of claudin-7 as a colon cancer suppressor and suggest that loss of claudin-7 potentiates EMT to promote colon cancer, in a manner dependent on Rab25.
- Published
- 2015
- Full Text
- View/download PDF
31. A combinatorial microRNA therapeutics approach to suppressing non-small cell lung cancer.
- Author
-
Kasinski AL, Kelnar K, Stahlhut C, Orellana E, Zhao J, Shimer E, Dysart S, Chen X, Bader AG, and Slack FJ
- Subjects
- Animals, Carcinoma, Non-Small-Cell Lung genetics, Carcinoma, Non-Small-Cell Lung pathology, Drug Delivery Systems, Genetic Therapy methods, Humans, Lung Neoplasms genetics, Lung Neoplasms pathology, Mice, Mice, Transgenic, Nanostructures, Tumor Burden drug effects, Tumor Cells, Cultured, Xenograft Model Antitumor Assays, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Carcinoma, Non-Small-Cell Lung drug therapy, Genes, Tumor Suppressor, Lung Neoplasms drug therapy, MicroRNAs administration & dosage
- Abstract
Targeted cancer therapies, although often effective, have limited utility owing to preexisting primary or acquired secondary resistance. Consequently, agents are sometimes used in combination to simultaneously affect multiple targets. MicroRNA mimics are excellent therapeutic candidates because of their ability to repress multiple oncogenic pathways at once. Here we treated the aggressive Kras;p53 non-small cell lung cancer mouse model and demonstrated efficacy with a combination of two tumor-suppressive microRNAs (miRNAs). Systemic nanodelivery of miR-34 and let-7 suppressed tumor growth leading to survival advantage. This combinatorial miRNA therapeutic approach engages numerous components of tumor cell-addictive pathways and highlights the ability to deliver multiple miRNAs in a safe and effective manner to target lung tissue.
- Published
- 2015
- Full Text
- View/download PDF
32. Autocrine CSF1R signaling mediates switching between invasion and proliferation downstream of TGFβ in claudin-low breast tumor cells.
- Author
-
Patsialou A, Wang Y, Pignatelli J, Chen X, Entenberg D, Oktay M, and Condeelis JS
- Subjects
- Animals, Breast Neoplasms pathology, Cell Line, Tumor, Cell Movement physiology, Disease Progression, Female, Gene Expression Regulation, Neoplastic physiology, Humans, MCF-7 Cells, Macrophage Colony-Stimulating Factor metabolism, Mice, Autocrine Communication physiology, Breast Neoplasms metabolism, Cell Proliferation physiology, Claudins metabolism, Neoplasm Invasiveness pathology, Receptor, Macrophage Colony-Stimulating Factor metabolism, Transforming Growth Factor beta metabolism
- Abstract
Patient data suggest that colony-stimulating factor-1 (CSF1) and its receptor (CSF1R) have critical roles during breast cancer progression. We have previously shown that in human breast tumors expressing both CSF1 and CSF1R, invasion in vivo is dependent both on a paracrine interaction with tumor-associated macrophages and an autocrine regulation of CSF1R in the tumor cells themselves. Although the role of the paracrine interaction between tumor cells and macrophages has been extensively studied, very little is known about the mechanism by which the autocrine CSF1R signaling contributes to tumor progression. We show here that breast cancer patients of the claudin-low subtype have significantly increased expression of CSF1R. Using a panel of breast cancer cell lines, we confirm that CSF1R expression is elevated and regulated by TGFβ specifically in claudin-low cell lines. Abrogation of autocrine CSF1R signaling in MDA-MB-231 xenografts (a claudin-low cell line) leads to increased tumor size by enhanced proliferation, but significantly reduced invasion, dissemination and metastasis. Indeed, we show that proliferation and invasion are oppositely regulated by CSF1R downstream of TGFβ only in claudin-low cell lines. Intravital multiphoton imaging revealed that inhibition of CSF1R in the tumor cells leads to decreased in vivo motility and a more cohesive morphology. We show that, both in vitro and in vivo, CSF1R inhibition results in a reversal of claudin-low marker expression by significant upregulation of luminal keratins and tight-junction proteins such as claudins. Finally, we show that artificial overexpression of claudins in MDA-MB-231 cells is sufficient to tip the cells from an invasive state to a proliferative state. Our results suggest that autocrine CSF1R signaling is essential in maintaining low claudin expression and that it mediates a switch between the proliferative and the invasive state in claudin-low tumor cells downstream of TGFβ.
- Published
- 2015
- Full Text
- View/download PDF
33. Targeted resequencing of the microRNAome and 3'UTRome reveals functional germline DNA variants with altered prevalence in epithelial ovarian cancer.
- Author
-
Chen X, Paranjape T, Stahlhut C, McVeigh T, Keane F, Nallur S, Miller N, Kerin M, Deng Y, Yao X, Zhao H, Weidhaas JB, and Slack FJ
- Subjects
- Base Sequence, Breast Neoplasms genetics, Carcinoma, Ovarian Epithelial, Case-Control Studies, DNA genetics, E2F2 Transcription Factor genetics, Female, Genetic Markers genetics, Genetic Predisposition to Disease, Humans, MicroRNAs genetics, Polymorphism, Single Nucleotide, Receptors, Vascular Endothelial Growth Factor genetics, Sequence Analysis, DNA, Vascular Endothelial Growth Factor Receptor-1 genetics, 3' Untranslated Regions genetics, Autoantigens genetics, Biomarkers, Tumor genetics, Cell Cycle Proteins genetics, Neoplasms, Glandular and Epithelial genetics, Ovarian Neoplasms genetics
- Abstract
Ovarian cancer is a major cause of cancer deaths, yet there have been few known genetic risk factors identified, the best known of which are disruptions in protein coding sequences (BRCA1 and 2). Recent findings indicate that there are powerful genetic markers of cancer risk outside of these regions, in the noncoding mRNA control regions. To identify additional cancer-associated, functional non-protein-coding sequence germline variants associated with ovarian cancer risk, we captured DNA regions corresponding to all validated human microRNAs and the 3' untranslated regions (UTRs) of ~6000 cancer-associated genes from 31 ovarian cancer patients. Multiple single-nucleotide polymorphisms in the 3'UTR of the vascular endothelial growth factor receptor/FLT1, E2F2 and PCM1 oncogenes were highly enriched in ovarian cancer patients compared with the 1000 Genome Project. Sequenom validation in a case-control study (267 cases and 89 controls) confirmed a novel variant in the PCM1 3'UTR is significantly associated with ovarian cancer (P=0.0086). This work identifies a potential new ovarian cancer locus and further confirms that cancer resequencing efforts should not ignore the study of noncoding regions of cancer patients.
- Published
- 2015
- Full Text
- View/download PDF
34. Phosphodiesterase 10A: a novel target for selective inhibition of colon tumor cell growth and β-catenin-dependent TCF transcriptional activity.
- Author
-
Li N, Lee K, Xi Y, Zhu B, Gary BD, Ramírez-Alcántara V, Gurpinar E, Canzoneri JC, Fajardo A, Sigler S, Piazza JT, Chen X, Andrews J, Thomas M, Lu W, Li Y, Laan DJ, Moyer MP, Russo S, Eberhardt BT, Yet L, Keeton AB, Grizzle WE, and Piazza GA
- Subjects
- Cell Line, Tumor, Cell Proliferation drug effects, Colonic Neoplasms pathology, Gene Expression Regulation, Neoplastic, Gene Knockdown Techniques, HCT116 Cells, HEK293 Cells, HT29 Cells, Humans, RNA, Small Interfering pharmacology, Signal Transduction drug effects, TCF Transcription Factors metabolism, Transcription, Genetic, beta Catenin metabolism, Colonic Neoplasms genetics, Colonic Neoplasms metabolism, Phosphodiesterase Inhibitors pharmacology, Phosphoric Diester Hydrolases genetics, Phosphoric Diester Hydrolases metabolism, TCF Transcription Factors genetics, beta Catenin genetics
- Abstract
The cyclic nucleotide phosphodiesterase 10A (PDE10) has been mostly studied as a therapeutic target for certain psychiatric and neurological conditions, although a potential role in tumorigenesis has not been reported. Here we show that PDE10 is elevated in human colon tumor cell lines compared with normal colonocytes, as well as in colon tumors from human clinical specimens and intestinal tumors from Apc(Min/+) mice compared with normal intestinal mucosa, respectively. An isozyme and tumor-selective role of PDE10 were evident by the ability of small-molecule inhibitors and small interfering RNA knockdown to suppress colon tumor cell growth with reduced sensitivity of normal colonocytes. Stable knockdown of PDE10 by short hairpin RNA also inhibits colony formation and increases doubling time of colon tumor cells. PDE10 inhibition selectively activates cGMP/cGMP-dependent protein kinase signaling to suppress β-catenin levels and T-cell factor (TCF) transcriptional activity in colon tumor cells. Conversely, ectopic expression of PDE10 in normal and precancerous colonocytes increases proliferation and activates TCF transcriptional activity. These observations suggest a novel role of PDE10 in colon tumorigenesis and that inhibitors may be useful for the treatment or prevention of colorectal cancer.
- Published
- 2015
- Full Text
- View/download PDF
35. Oncogenic Y641 mutations in EZH2 prevent Jak2/β-TrCP-mediated degradation.
- Author
-
Sahasrabuddhe AA, Chen X, Chung F, Velusamy T, Lim MS, and Elenitoba-Johnson KS
- Subjects
- Enhancer of Zeste Homolog 2 Protein, HEK293 Cells, Histones metabolism, Humans, Lymphoma etiology, Methylation, Phosphorylation, Polycomb Repressive Complex 2 metabolism, Proteasome Endopeptidase Complex physiology, Janus Kinase 2 physiology, Mutation, Polycomb Repressive Complex 2 genetics, beta-Transducin Repeat-Containing Proteins physiology
- Abstract
EZH2 (enhancer of zeste homolog 2) is a critical enzymatic subunit of the polycomb repressive complex 2 (PRC2), which trimethylates histone H3 (H3K27) to mediate gene repression. Somatic mutations, overexpression and hyperactivation of EZH2 have been implicated in the pathogenesis of several forms of cancer. In particular, recurrent gain-of-function mutations targeting EZH2 Y641 occur most frequently in follicular lymphoma and aggressive diffuse large B-cell lymphoma and are associated with H3K27me3 hyperactivation, which contributes to lymphoma pathogenesis. However, the post-translational mechanisms of EZH2 regulation are not completely understood. Here we show that EZH2 is a novel interactor and substrate of the SCF E3 ubiquitin ligase β-TrCP (FBXW1). β-TrCP ubiquitinates EZH2 and Jak2-mediated phosphorylation on Y641 directs β-TrCP-mediated EZH2 degradation. RNA interference-mediated silencing of β-TrCP or inhibition of Jak2 results in EZH2 stabilization with attendant increase in H3K27 trimethylation activity. Importantly, the EZH2(Y641) mutants recurrently implicated in lymphoma pathogenesis are unable to bind β-TrCP. Further, endogenous EZH2(Y641) mutants in lymphoma cells exhibit increased EZH2 stability and H3K27me3 hyperactivity. Our studies demonstrate that β-TrCP has an important role in controlling H3K27 trimethylation activity and lymphoma pathogenesis by targeting EZH2 for degradation.
- Published
- 2015
- Full Text
- View/download PDF
36. Transformation by Hras(G12V) is consistently associated with mutant allele copy gains and is reversed by farnesyl transferase inhibition.
- Author
-
Chen X, Makarewicz JM, Knauf JA, Johnson LK, and Fagin JA
- Subjects
- 9,10-Dimethyl-1,2-benzanthracene toxicity, Animals, Cell Line, Tumor, Farnesyltranstransferase metabolism, Gene Dosage, Gene Knock-In Techniques, Humans, Mice, Mutant Strains, Mutation, Papilloma chemically induced, Papilloma drug therapy, Piperidines pharmacology, Pyridines pharmacology, Skin Neoplasms chemically induced, Skin Neoplasms drug therapy, Tetradecanoylphorbol Acetate toxicity, Enzyme Inhibitors pharmacology, Farnesyltranstransferase antagonists & inhibitors, Genes, ras, Papilloma genetics, Skin Neoplasms genetics
- Abstract
RAS-driven malignancies remain a major therapeutic challenge. The two-stage 7,12-dimethylbenz(a)anthracene (DMBA)/12-o-tetradecanoylphorbol-13-acetate (TPA) model of mouse skin carcinogenesis has been used to study mechanisms of epithelial tumor development by oncogenic Hras. We used mice with an Hras(G12V) knock-in allele to elucidate the early events after Hras activation, and to evaluate the therapeutic effectiveness of farnesyltransferase inhibition (FTI). Treatment of Caggs-Cre/FR-Hras(G12V) mice with TPA alone was sufficient to trigger papilloma development with a shorter latency and an ∼10-fold greater tumor burden than DMBA/TPA-treated WT-controls. Hras(G12V) allele copy number was increased in all papillomas induced by TPA. DMBA/TPA treatment of Hras(G12V) knock-in mice induced an even greater incidence of papillomas, which either harbored Hras(G12V) amplification or developed an Hras(Q61L) mutation in the second allele. Laser-capture microdissection of normal skin, hyperplastic skin and papillomas showed that amplification occurred only at the papilloma stage. HRAS-mutant allelic imbalance was also observed in human cancer cell lines, consistent with a requirement for augmented oncogenic HRAS signaling for tumor development. The FTI SCH66336 blocks HRAS farnesylation and delocalizes it from the plasma membrane. NRAS and KRAS are not affected as they are alternatively prenylated. When tested in lines harboring HRAS, NRAS or KRAS mutations, SCH66336 delocalized, inhibited signaling and preferentially inhibited growth only of HRAS-mutant lines. Treatment with SCH66336 also induced near-complete regression of papillomas of TPA-treated Hras(G12V) knock-in mice. These data suggest that farnesyl transferase inhibitors should be reevaluated as targeted agents for human HRAS-driven cancers, such as those of bladder, thyroid and other epithelial lineages.
- Published
- 2014
- Full Text
- View/download PDF
37. Combined PKC and MEK inhibition in uveal melanoma with GNAQ and GNA11 mutations.
- Author
-
Chen X, Wu Q, Tan L, Porter D, Jager MJ, Emery C, and Bastian BC
- Subjects
- Animals, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Apoptosis, Benzimidazoles administration & dosage, Cell Line, Tumor, Cell Proliferation, Drug Synergism, Female, GTP-Binding Protein alpha Subunits, Gq-G11, Humans, Inhibitory Concentration 50, MAP Kinase Kinase Kinases antagonists & inhibitors, MAP Kinase Kinase Kinases metabolism, Melanoma genetics, Melanoma secondary, Mice, Mice, Inbred C57BL, Mice, Nude, Mutation, Missense, Protein Kinase C antagonists & inhibitors, Protein Kinase C metabolism, Protein Kinase Inhibitors administration & dosage, Pyrroles administration & dosage, Quinazolines administration & dosage, Signal Transduction, Tumor Burden drug effects, Uveal Neoplasms genetics, Uveal Neoplasms pathology, Xenograft Model Antitumor Assays, Antineoplastic Combined Chemotherapy Protocols pharmacology, GTP-Binding Protein alpha Subunits genetics, Melanoma drug therapy, Uveal Neoplasms drug therapy
- Abstract
Uveal melanoma (UM) is a genetically and biologically distinct type of melanoma, and once metastatic there is no effective treatment currently available. Eighty percent of UMs harbor mutations in the Gαq family members GNAQ and GNA11. Understanding the effector pathways downstream of these oncoproteins is important to identify opportunities for targeted therapy. We report consistent activation of the protein kinase C (PKC) and MAPK pathways as a consequence of GNAQ or GNA11 mutation. PKC inhibition with AEB071 or AHT956 suppressed PKC and MAPK signalling and induced G1 arrest selectively in melanoma cell lines carrying GNAQ or GNA11 mutations. In contrast, treatment with two different MEK inhibitors, PD0325901 and MEK162, inhibited the proliferation of melanoma cell lines irrespective of their mutation status, indicating that in the context of GNAQ or GNA11 mutation MAPK activation can be attributed to activated PKC. AEB071 significantly slowed the growth of tumors in an allograft model of GNAQ(Q209L)-transduced melanocytes, but did not induce tumor shrinkage. In vivo and in vitro studies showed that PKC inhibitors alone were unable to induce sustained suppression of MAP-kinase signaling. However, combinations of PKC and MEK inhibition, using either PD0325901or MEK162, led to sustained MAP-kinase pathway inhibition and showed a strong synergistic effect in halting proliferation and in inducing apoptosis in vitro. Furthermore, combining PKC and MEK inhibition was efficacious in vivo, causing marked tumor regression in a UM xenograft model. Our data identify PKC as a rational therapeutic target for melanoma patients with GNAQ or GNA11 mutations and demonstrate that combined MEK and PKC inhibition is synergistic, with superior efficacy compared to treatment with either approach alone.
- Published
- 2014
- Full Text
- View/download PDF
38. Pivotal role of augmented αB-crystallin in tumor development induced by deficient TSC1/2 complex.
- Author
-
Wang F, Chen X, Li C, Sun Q, Chen Y, Wang Y, Peng H, Liu Z, Chen R, Liu K, Yan H, Ye BH, Kwiatkowski DJ, and Zhang H
- Subjects
- Animals, Apoptosis, Cell Line, Tumor, Cell Movement, Cell Proliferation, Female, Humans, Kidney Neoplasms metabolism, Leiomyoma metabolism, Lung Neoplasms metabolism, Lymphangioleiomyomatosis metabolism, Male, Mechanistic Target of Rapamycin Complex 2, Mice, Multiprotein Complexes metabolism, NF-kappa B metabolism, Neoplasm Transplantation, Rats, TOR Serine-Threonine Kinases metabolism, Tuberous Sclerosis metabolism, Tuberous Sclerosis Complex 1 Protein, Tuberous Sclerosis Complex 2 Protein, Uterine Neoplasms metabolism, Carcinogenesis metabolism, Tumor Suppressor Proteins deficiency, alpha-Crystallin B Chain physiology
- Abstract
Tuberous sclerosis complex 1 (TSC1) and TSC2 are suppressors of mechanistic target of rapamycin (mTOR). mTOR is the major component of two protein complexes: mTOR complex 1 (mTORC1) and mTORC2. Inactive mutation of either TSC1 or TSC2 unleashes mTOR signaling and consequently causes TSC, a benign tumor syndrome affecting multiple organs. We report here that expression of αB-crystallin was upregulated in Tsc1-/- or Tsc2-/- mouse embryonic fibroblasts, Eker rat uterine leiomyoma-derived Tsc2-deficient ELT3 cells, mutant Tsc2-associated mouse kidney tumors, and human lung lymphangioleiomyomatosis nodules. αB-crystallin was transcriptionally activated by mTOR complex 2 (mTORC2): nuclear factor-kappa B (NFκB) signaling cascade. The augmented αB-crystallin was critical for the migration, invasion and apoptotic resistance of Tsc2-defective cells. Disruption of αB-crystallin suppressed Tsc2-null cell proliferation and tumorigenesis. Therefore, enhanced αB-crystallin has an essential role in TSC1/2 complex deficiency-mediated tumorigenesis, and inhibition of αB-crystallin may complement the current therapy for TSC.
- Published
- 2014
- Full Text
- View/download PDF
39. p53 regulates mesenchymal stem cell-mediated tumor suppression in a tumor microenvironment through immune modulation.
- Author
-
Huang Y, Yu P, Li W, Ren G, Roberts AI, Cao W, Zhang X, Su J, Chen X, Chen Q, Shou P, Xu C, Du L, Lin L, Xie N, Zhang L, Wang Y, and Shi Y
- Subjects
- Animals, Cytokines biosynthesis, Disease Models, Animal, Inflammation Mediators, Melanoma, Experimental, Mice, Mice, Knockout, Neoplasms genetics, Neoplasms pathology, Nitric Oxide biosynthesis, Nitric Oxide Synthase Type II metabolism, Tumor Microenvironment genetics, Tumor Suppressor Protein p53 deficiency, Tumor Suppressor Protein p53 genetics, Immunomodulation genetics, Mesenchymal Stem Cells metabolism, Neoplasms immunology, Neoplasms metabolism, Tumor Microenvironment immunology, Tumor Suppressor Protein p53 metabolism
- Abstract
p53 is one of the most studied genes in cancer biology, and mutations in this gene may be predictive for the development of many types of cancer in humans and in animals. However, whether p53 mutations in non-tumor stromal cells can affect tumor development has received very little attention. In this study, we show that B16F0 melanoma cells form much larger tumors in p53-deficient mice than in wild-type mice, indicating a potential role of p53 deficiency in non-tumor cells of the microenvironment. As mesenchymal stem cells (MSCs) are attracted to tumors and form a major component of the tumor microenvironment, we examined the potential role of p53 status in MSCs in tumor development. We found that larger tumors resulted when B16F0 melanoma cells were co-injected with bone marrow MSCs derived from p53-deficient mice rather than MSCs from wild-type mice. Interestingly, this tumor-promoting effect by p53-deficient MSCs was not observed in non-obese diabetic/severe combined immunodeficiency mice, indicating the immune response has a critical role. Indeed, in the presence of inflammatory cytokines, p53-deficient MSCs expressed more inducible nitric oxide synthase (iNOS) and exhibited greater immunosuppressive capacity. Importantly, tumor promotion by p53-deficient MSCs was abolished by administration of S-methylisothiourea, an iNOS inhibitor. Therefore, our data demonstrate that p53 status in tumor stromal cells has a key role in tumor development by modulating immune responses.
- Published
- 2014
- Full Text
- View/download PDF
40. IL-6 secreted by cancer-associated fibroblasts induces tamoxifen resistance in luminal breast cancer.
- Author
-
Sun X, Mao Y, Wang J, Zu L, Hao M, Cheng G, Qu Q, Cui D, Keller ET, Chen X, Shen K, and Wang J
- Abstract
Cancer-associated fibroblasts (CAFs) have been implicated in the development of resistance to anticancer drugs; however, the role and mechanism underlying CAFs in luminal breast cancer (BrCA) tamoxifen resistance are unclear. We found that stromal fibroblasts isolated from the central or peripheral area of BrCA have similar CAF phenotype and activity. In vitro and in vivo experiments showed that CAFs derived from clinical-luminal BrCAs induce tamoxifen resistance through decreasing estrogen receptor-α (ER-α) level when cultured with luminal BrCA cell lines MCF7 and T47D. CAFs promoted tamoxifen resistance through interleukin-6 (IL-6) secretion, which activates Janus kinase/signal transducers and activators of transcription (JAK/STAT3) and phosphatidylinositol 3-kinase (PI3K)/AKT pathways in tumor cells, followed by induction of epithelial-mesenchymal transition and upregulation of E3 ubiquitin ligase anaphase-promoting complex 10 activity, which targeted ER-α degradation through the ubiquitin-proteasome pathway. Inhibition of proteasome activity, IL-6 activity or either the JAK/STAT3 or PI3K/AKT pathways markedly reduced CAF-induced tamoxifen resistance. In xenograft experiments of CAFs mixed with MCF7 cells, CAF-specific IL-6 knockdown inhibited tumorigenesis and restored tamoxifen sensitivity. These findings indicate that CAFs mediate tamoxifen resistance through IL-6-induced degradation of ER-α in luminal BrCAs.Oncogene advance online publication, 9 June 2014; doi:10.1038/onc.2014.158.
- Published
- 2014
- Full Text
- View/download PDF
41. P63 regulates tubular formation via epithelial-to-mesenchymal transition.
- Author
-
Zhang Y, Yan W, and Chen X
- Subjects
- Animals, Cell Movement, Cell Proliferation, Dogs, Gene Knockdown Techniques, Madin Darby Canine Kidney Cells, Protein Isoforms deficiency, Protein Isoforms genetics, Protein Isoforms metabolism, Tumor Suppressor Proteins deficiency, Tumor Suppressor Proteins genetics, Up-Regulation, Epithelial Cells cytology, Epithelial-Mesenchymal Transition, Tumor Suppressor Proteins metabolism
- Abstract
P63, a p53 family member, is expressed as TA and ΔN isoforms. Interestingly, both TAp63 and ΔNp63 are transcription factors, and regulate both common and distinct sets of target genes. p63 is required for survival of some epithelial cell lineages, and lack of p63 leads to loss of epidermis and other epithelia in humans and mice. Here, we explored the role of p63 isoforms in cell proliferation, migration and tubulogenesis by using Madin-Darby Canine Kidney (MDCK) tubular epithelial cells in two- or three-dimensional (2-D or 3-D) culture. We found that like downregulation of p53, downregulation of p63 and TAp63 decreases expression of growth-suppressing genes, including p21, PUMA and MIC-1, and consequently promotes cell proliferation and migration in 2-D culture. However, in 3-D culture, downregulation of p63, especially TAp63, but not p53, decapacitates MDCK cells to form a cyst structure through enhanced epithelial-to-mesenchymal transition (EMT). In contrast, downregulation of ΔNp63 inhibits MDCK cell proliferation and migration in 2-D culture, and delays but does not block MDCK cell cyst formation and tubulogenesis in 3-D culture. Consistent with this, downregulation of ΔNp63 markedly upregulates growth-suppressing genes, including p21, PUMA and MIC-1. Taken together, these data suggest that TAp63 is the major isoform required for tubulogenesis by maintaining an appropriate level of EMT, whereas ΔNp63 fine-tunes the rate of cyst formation and tubulogenesis by maintaining an appropriate expression level of genes involved in cell cycle arrest and apoptosis.
- Published
- 2014
- Full Text
- View/download PDF
42. Telomere dysfunction and activation of alternative lengthening of telomeres in B-lymphocytes infected by Epstein-Barr virus.
- Author
-
Kamranvar SA, Chen X, and Masucci MG
- Subjects
- DNA Helicases metabolism, Enzyme Activation, Humans, Nuclear Proteins metabolism, Shelterin Complex, Sister Chromatid Exchange, Telomerase metabolism, Telomere-Binding Proteins metabolism, Telomeric Repeat Binding Protein 1 metabolism, Telomeric Repeat Binding Protein 2 metabolism, X-linked Nuclear Protein, B-Lymphocytes metabolism, B-Lymphocytes virology, Epstein-Barr Virus Infections genetics, Herpesvirus 4, Human, Telomere genetics, Telomere Homeostasis genetics
- Abstract
Malignant cells achieve replicative immortality by two alternative mechanisms, a common one dependent on de novo synthesis of telomeric DNA by telomerase, and a rare one based on telomere recombination known as alternative lengthening of telomeres (ALT). Epstein-Barr virus (EBV) transforms human B-lymphocytes into lymphoblastoid cell lines with unlimited growth potential in vitro and in vivo. Here we show that newly EBV-infected cells exhibit multiple signs of telomere dysfunction, including the occurrence of extra-chromosomal telomeres, telomere fusion and telomere length heterogeneity, and undergo progressive increase in telomere length without a parallel increase in telomerase activity. This phenotype is accompanied by the accumulation of telomere-associated promyelocytic leukemia nuclear bodies and telomeric-sister chromatid exchange, suggesting that EBV infection promotes the activation of ALT. Newly infected cells also display a significant reduction of telomere-associated TRF2 and express low levels of TRF1, TRF2, POT1 and ATRX, pointing to telomere de-protection as an important correlate of ALT activation. Collectively, these findings highlight the involvement of recombination-dependent mechanisms for maintenance of telomere homeostasis in EBV-induced B-cell immortalization.
- Published
- 2013
- Full Text
- View/download PDF
43. Stat3-coordinated Lin-28-let-7-HMGA2 and miR-200-ZEB1 circuits initiate and maintain oncostatin M-driven epithelial-mesenchymal transition.
- Author
-
Guo L, Chen C, Shi M, Wang F, Chen X, Diao D, Hu M, Yu M, Qian L, and Guo N
- Subjects
- Animals, Breast Neoplasms metabolism, Cell Line, Tumor, Down-Regulation, Epithelial-Mesenchymal Transition genetics, Female, Gene Knockdown Techniques, HMGA2 Protein biosynthesis, HMGA2 Protein genetics, Heterografts, Homeodomain Proteins, Humans, Inflammation, Kruppel-Like Transcription Factors, Lung Neoplasms secondary, MCF-7 Cells, Mammary Neoplasms, Animal metabolism, Mice, Mice, Inbred BALB C, MicroRNAs biosynthesis, Neoplasm Transplantation, Oncostatin M biosynthesis, Promoter Regions, Genetic, RNA-Binding Proteins biosynthesis, RNA-Binding Proteins genetics, Up-Regulation, Zinc Finger E-box-Binding Homeobox 1, Epithelial-Mesenchymal Transition physiology, MicroRNAs metabolism, Oncostatin M metabolism, STAT3 Transcription Factor metabolism
- Abstract
Inflammation can act as a crucial mediator of epithelial-to-mesenchymal transition (EMT). In this study, we show that oncostatin M (OSM) is expressed in an autocrine/paracrine fashion in invasive breast carcinoma. OSM stimulation promotes spontaneous lung metastasis of MCF-7 xenografts in nude mice. A conspicuous epigenetic transition was induced by OSM stimulation not only in breast cancer cell lines but also in MCF-7 xenografts in nude mice. The expression of miR-200 and let-7 family members in response to OSM stimulation was downregulated in a signal transducer and activator of transcription factor 3 (Stat3)-dependent manner, resulting in comprehensive alterations of the transcription factors and oncoproteins targeted by these microRNAs. Inhibition of Stat3 activation or the ectopic expression of let-7 and miR-200 effectively reversed the mesenchymal phenotype of breast cancer cells. Stat3 promotes the transcription of Lin-28 by directly binding to the Lin-28 promoter, resulting in the repression of let-7 expression and concomitant upregulation of the let-7 target, high-mobility group A protein 2 (HMGA2). Knock down of HMGA2 significantly impairs OSM-driven EMT. Our data indicate that downregulation of let-7 and miR-200 levels initiates and maintains OSM-induced EMT phenotypes, and HMGA2 acts as a master switch of OSM-induced EMT. These findings highlight the importance of Stat3-coordinated Lin-28B-let-7-HMGA2 and miR-200-ZEB1 circuits in the cytokine-mediated phenotypic reprogramming of breast cancer cells.
- Published
- 2013
- Full Text
- View/download PDF
44. mTORC1 enhancement of STIM1-mediated store-operated Ca2+ entry constrains tuberous sclerosis complex-related tumor development.
- Author
-
Peng H, Liu J, Sun Q, Chen R, Wang Y, Duan J, Li C, Li B, Jing Y, Chen X, Mao Q, Xu KF, Walker CL, Li J, Wang J, and Zhang H
- Subjects
- Adaptor Proteins, Signal Transducing, Animals, Calcium Channels genetics, Calcium Channels physiology, Cell Line, Cell Line, Tumor, Female, Fibroblasts, Humans, Kidney Neoplasms genetics, Kidney Neoplasms pathology, Leiomyoma pathology, Mechanistic Target of Rapamycin Complex 1, Mice, Mice, Inbred BALB C, Mice, Nude, Neovascularization, Pathologic physiopathology, ORAI1 Protein, Phosphorylation, Protein Processing, Post-Translational, RNA Interference, Rats, Recombinant Fusion Proteins physiology, Stromal Interaction Molecule 1, Tuberous Sclerosis genetics, Tuberous Sclerosis metabolism, Tuberous Sclerosis Complex 2 Protein, Tumor Necrosis Factor Receptor-Associated Peptides and Proteins metabolism, Tumor Suppressor Proteins physiology, Uterine Neoplasms pathology, Calcium Signaling physiology, Cell Transformation, Neoplastic, Membrane Proteins physiology, Multiprotein Complexes physiology, Neoplasm Proteins physiology, TOR Serine-Threonine Kinases physiology, Tuberous Sclerosis pathology
- Abstract
The protein complex of tuberous sclerosis complex (TSC)1 and TSC2 tumor suppressors is a key negative regulator of mammalian target of rapamycin (mTOR). Hyperactive mTOR signaling due to the loss-of-function of mutations in either TSC1 or TSC2 gene causes TSC, an autosomal dominant disorder featured with benign tumors in multiple organs. As the ubiquitous second messenger calcium (Ca(2+)) regulates various cellular processes involved in tumorigenesis, we explored the potential role of mTOR in modulation of cellular Ca(2+) homeostasis, and in turn the effect of Ca(2+) signaling in TSC-related tumor development. We found that loss of Tsc2 potentiated store-operated Ca(2+) entry (SOCE) in an mTOR complex 1 (mTORC1)-dependent way. The endoplasmic reticulum Ca(2+) sensor, stromal interaction molecule 1 (STIM1), was upregulated in Tsc2-deficient cells, and was suppressed by mTORC1 inhibitor rapamycin. In addition, SOCE repressed AKT1 phosphorylation. Blocking SOCE either by depleting STIM1 or ectopically expressing dominant-negative Orai1 accelerated TSC-related tumor development, likely because of restored AKT1 activity and enhanced tumor angiogenesis. Our data, therefore, suggest that mTORC1 enhancement of store-operated Ca(2+) signaling hinders TSC-related tumor growth through suppression of AKT1 signaling. The augmented SOCE by hyperactive mTORC1-STIM1 cascade may contribute to the benign nature of TSC-related tumors. Application of SOCE agonists could thus be a contraindication for TSC patients. In contrast, SOCE agonists should attenuate mTOR inhibitors-mediated AKT reactivation and consequently potentiate their efficacy in the treatment of the patients with TSC.
- Published
- 2013
- Full Text
- View/download PDF
45. MDM2 expression is repressed by the RNA-binding protein RNPC1 via mRNA stability.
- Author
-
Xu E, Zhang J, and Chen X
- Subjects
- 3' Untranslated Regions, Animals, Cell Line, Tumor, Gene Expression, Half-Life, Humans, Mice, Protein Binding, Protein Isoforms physiology, Proto-Oncogene Proteins c-mdm2 metabolism, RNA, Messenger genetics, RNA, Messenger metabolism, Tumor Suppressor Protein p53 metabolism, Gene Silencing, Proto-Oncogene Proteins c-mdm2 genetics, RNA Stability, RNA-Binding Proteins physiology
- Abstract
The RNA-binding protein (RBP) RNPC1 is a target of the p53 family and forms a feedback regulatory loop with the p53 family proteins. The murine double minute-2 (MDM2) oncogene, a key negative regulator of p53, has a critical role in a variety of fundamental cellular processes. MDM2 expression is found to be regulated via gene amplification, transcription, protein translation and protein stability. In the current study, we reported a novel regulation of MDM2 by RNPC1 via mRNA stability. Specifically, we found that overexpression of RNPC1 decreases, whereas knockdown or knockout of RNPC1 increases, the level of MDM2 transcript and protein independent of p53. To uncover the underlying mechanism, we found that RNPC1 is able to destabilize the MDM2 transcript via binding to multiple AU-/U-rich elements in MDM2 3'untranslated region (3'UTR). Consistent with this, we showed that RNPC1 inhibits expression of exogenous MDM2 from an expression vector as long as the vector contains an AU-/U-rich element from MDM2 3'UTR. Finally, we showed that the RNA-binding activity of RNPC1 is required for binding to MDM2 transcript and consequently, for inhibiting MDM2 expression. Together, we uncover a novel regulation of MDM2 by the RBP RNPC1 via mRNA stability.
- Published
- 2013
- Full Text
- View/download PDF
46. Lenalidomide promotes p53 degradation by inhibiting MDM2 auto-ubiquitination in myelodysplastic syndrome with chromosome 5q deletion.
- Author
-
Wei S, Chen X, McGraw K, Zhang L, Komrokji R, Clark J, Caceres G, Billingsley D, Sokol L, Lancet J, Fortenbery N, Zhou J, Eksioglu EA, Sallman D, Wang H, Epling-Burnette PK, Djeu J, Sekeres M, Maciejewski JP, and List A
- Subjects
- Animals, Chromosome Deletion, Humans, Lenalidomide, Mice, Thalidomide pharmacology, Ubiquitination, Chromosomes, Human, Pair 5, Myelodysplastic Syndromes genetics, Proto-Oncogene Proteins c-mdm2 metabolism, Thalidomide analogs & derivatives, Tumor Suppressor Protein p53 metabolism
- Abstract
Allelic deletion of the RPS14 gene is a key effector of the hypoplastic anemia in patients with myelodysplastic syndrome (MDS) and chromosome 5q deletion (del(5q)). Disruption of ribosome integrity liberates free ribosomal proteins to bind to and trigger degradation of mouse double minute 2 protein (MDM2), with consequent p53 transactivation. Herein we show that p53 is overexpressed in erythroid precursors of primary bone marrow del(5q) MDS specimens accompanied by reduced cellular MDM2. More importantly, we show that lenalidomide (Len) acts to stabilize MDM2, thereby accelerating p53 degradation. Biochemical and molecular analyses showed that Len inhibits the haplodeficient protein phosphatase 2A catalytic domain alpha (PP2Acα) phosphatase resulting in hyperphosphorylation of inhibitory serine-166 and serine-186 residues on MDM2, and displaces binding of RPS14 to suppress MDM2 autoubiquitination whereas PP2Acα overexpression promotes drug resistance. Bone marrow specimens from del(5q) MDS patients resistant to Len overexpressed PP2Acα accompanied by restored accumulation of p53 in erythroid precursors. Our findings indicate that Len restores MDM2 functionality in the 5q- syndrome to overcome p53 activation in response to nucleolar stress, and therefore may warrant investigation in other disorders of ribosomal biogenesis.
- Published
- 2013
- Full Text
- View/download PDF
47. Histone deacetylase inhibitors suppress mutant p53 transcription via histone deacetylase 8.
- Author
-
Yan W, Liu S, Xu E, Zhang J, Zhang Y, Chen X, and Chen X
- Subjects
- Cell Line, Tumor, Gene Knockdown Techniques, Histone Deacetylases genetics, Homeodomain Proteins metabolism, Humans, Mutation, Repressor Proteins antagonists & inhibitors, Repressor Proteins genetics, Transcription, Genetic drug effects, Vorinostat, Genes, p53, Histone Deacetylase Inhibitors pharmacology, Histone Deacetylases metabolism, Hydroxamic Acids pharmacology, Repressor Proteins metabolism
- Abstract
Mutation of the p53 gene is the most common genetic alteration in human cancer and contributes to malignant process by enhancing transformed properties of cells and resistance to anticancer therapy. Mutant p53 is often highly expressed in tumor cells at least, in part, due to its increased half-life. However, whether mutant p53 expression is regulated by other mechanisms in tumors is unclear. Here we found that histone deacetylase (HDAC) inhibitors suppress both wild-type and mutant p53 transcription in time- and dose-dependent manners. Consistent with this, the levels of wild-type and mutant p53 proteins are decreased upon treatment with HDAC inhibitors. Importantly, we found that upon knockdown of each class I HDAC, only HDAC8 knockdown leads to decreased expression of wild-type and mutant p53 proteins and transcripts. Conversely, we found that ectopic expression of wild-type, but not mutant HDAC8, leads to increased transcription of p53. Furthermore, we found that knockdown of HDAC8 results in reduced expression of HoxA5 and consequently, attenuated ability of HoxA5 to activate p53 transcription, which can be rescued by ectopic expression of HoxA5. Because of the fact that HDAC8 is required for expression of both wild-type and mutant p53, we found that targeted disruption of HDAC8 expression remarkably triggers proliferative defect in cells with a mutant, but not wild-type, p53. Together, our data uncover a regulatory mechanism of mutant p53 transcription via HDAC8 and suggest that HDAC inhibitors and especially HDAC8-targeting agents might be explored as an adjuvant for tumors carrying a mutant p53.
- Published
- 2013
- Full Text
- View/download PDF
48. Myc represses miR-15a/miR-16-1 expression through recruitment of HDAC3 in mantle cell and other non-Hodgkin B-cell lymphomas.
- Author
-
Zhang X, Chen X, Lin J, Lwin T, Wright G, Moscinski LC, Dalton WS, Seto E, Wright K, Sotomayor E, and Tao J
- Subjects
- Acetylation, Cell Line, Tumor, Epigenesis, Genetic, Gene Expression Regulation, Neoplastic, Histone Deacetylase Inhibitors pharmacology, Histones metabolism, Humans, Hydroxamic Acids pharmacology, Lymphoma, Non-Hodgkin metabolism, MicroRNAs genetics, Promoter Regions, Genetic, Proto-Oncogene Proteins c-myc genetics, RNA, Long Noncoding, RNA, Small Interfering metabolism, Transferases, Tumor Suppressor Proteins genetics, Tumor Suppressor Proteins metabolism, Vorinostat, Histone Deacetylases metabolism, Lymphoma, Non-Hodgkin pathology, MicroRNAs biosynthesis, Proto-Oncogene Proteins c-myc biosynthesis
- Abstract
Our recent study demonstrated miR-15a/16-1 downregulation in mantle cell lymphoma (MCL). Here, we investigated mechanisms of miR-15a/16-1 transcriptional repression and its epigenetic regulation by c-Myc and histone deacetylase (HDAC) in MCL. c-Myc expression was detected in MCL cell lines and in the primary MCL samples, and pri-miR-15a/16-1 mRNAs were significantly upregulated in Mino and Jeko-1 cells with c-Myc knockdown by small interfering RNAs (siRNAs). Our co-immunoprecipitation analysis showed that c-Myc interacted with HDAC3. Moreover, using chromatin immunoprecipitation, we demonstrated that both c-Myc and HDAC3 co-localized to the two promoters of the miR-15a/16-1 cluster gene, DLEU2, and inhibition of HDAC3 increased histone acetylation of the DLEU2 promoters. Luciferase reporter assay confirmed the dependence of Myc-mediated DLEU2 transcriptional repression on HDAC3. Treatment with the pan-HDAC inhibitor, suberoylanilide hydroxamic acid and HDAC3 siRNA resulted in increased miR-15a/16-1 expression. The regulatory mechanism of miR-15a/16-1 was further demonstrated in Burkitt lymphoma and Myc overexpressing cell lines. These findings highlight the role of HDAC3 in Myc-induced miR-15a/16-1 changes and reveal novel mechanisms for c-Myc-driven microRNA suppression and malignant transformation in aggressive B-cell malignancies.
- Published
- 2012
- Full Text
- View/download PDF
49. IRX1 influences peritoneal spreading and metastasis via inhibiting BDKRB2-dependent neovascularization on gastric cancer.
- Author
-
Jiang J, Liu W, Guo X, Zhang R, Zhi Q, Ji J, Zhang J, Chen X, Li J, Zhang J, Gu Q, Liu B, Zhu Z, and Yu Y
- Subjects
- Animals, Cell Line, Tumor, Chick Embryo, Endothelial Cells, Homeodomain Proteins physiology, Human Umbilical Vein Endothelial Cells pathology, Humans, Lung Neoplasms prevention & control, Lung Neoplasms secondary, Male, Mice, Mice, Nude, Neoplasm Invasiveness, Neoplasm Metastasis, RNA Interference, RNA, Small Interfering pharmacology, Stomach Neoplasms blood supply, Stomach Neoplasms pathology, Transcription Factors physiology, Transfection, Homeodomain Proteins genetics, Neovascularization, Pathologic genetics, Receptor, Bradykinin B2 genetics, Stomach Neoplasms genetics, Transcription Factors genetics
- Abstract
The overexpression of IRX1 gene correlates with the growth arrest in gastric cancer. Furthermore, overexpression of IRX1 gene suppresses peritoneal spreading and long distance metastasis. To explore the precise mechanisms, we investigated whether restoring IRX1 expression affects the angiogenesis or vasculogenic mimicry (VM). Human umbilical vein endothelial cells (HUVECs) and chick embryo and SGC-7901 gastric cancer cells were used for angiogenesis and VM analysis. Small interfering RNA was used for analyzing the function of BDKRB2, a downstream target gene of IRX1. As results, the remarkable suppression on peritoneal spreading and pulmonary metastasis of SGC-7901 cells by IRX1 transfectant correlates to reduced angiogenesis as well as VM formation. Using the supernatant from SGC-7901/IRX1 cells, we found a strong inhibiting effect on angiogenesis both in vitro and in chick embryo. SGC-7901/IRX1 cells revealed strong inhibiting effect on VM formation too. By gene-specific RNA interference for BDKRB2, or its effector PAK1, we got an effective inhibition on tube formation, cell proliferation, cell migration and invasion in vitro. In conclusion, enforcing IRX1 expression effectively suppresses peritoneal spreading and pulmonary metastasis via anti-angiogenesis and anti-VM mechanisms, in addition to previously found cell growth and invasion. BDKRB2 and its downstream effector might be potential targets for anti-cancer strategy.
- Published
- 2011
- Full Text
- View/download PDF
50. NANOG promotes cancer stem cell characteristics and prostate cancer resistance to androgen deprivation.
- Author
-
Jeter CR, Liu B, Liu X, Chen X, Liu C, Calhoun-Davis T, Repass J, Zaehres H, Shen JJ, and Tang DG
- Subjects
- Animals, Carcinoma, Embryonal pathology, Cell Line, Tumor, Chromatin Immunoprecipitation, Green Fluorescent Proteins metabolism, Humans, Lentivirus genetics, Male, Mice, Mice, Inbred NOD, Mice, SCID, Models, Genetic, Nanog Homeobox Protein, Phenotype, RNA, Messenger metabolism, Androgens metabolism, Homeodomain Proteins physiology, Neoplastic Stem Cells cytology, Prostatic Neoplasms drug therapy, Prostatic Neoplasms pathology
- Abstract
Cancer cell molecular mimicry of stem cells (SC) imbues neoplastic cells with enhanced proliferative and renewal capacities. In support, numerous mediators of SC self-renewal have been evinced to show oncogenic potential. We have recently reported that short-hairpin RNA-mediated knockdown of the embryonic stem cell (ESC) self-renewal gene NANOG significantly reduced the clonogenic and tumorigenic capabilities of various cancer cells. In this study, we sought to test the potential pro-tumorigenic functions of NANOG, particularly, in prostate cancer (PCa). Using qRT-PCR, we first confirmed that PCa cells expressed NANOG mRNA primarily from the NANOGP8 locus on chromosome 15q14. We then constructed a lentiviral promoter reporter in which the -3.8-kb NANOGP8 genomic fragment was used to drive the expression of green fluorescence protein (GFP). We observed that NANOGP8-GFP(+) PCa cells showed cancer stem cell (CSC) characteristics such as enhanced clonal growth and tumor regenerative capacity. To further investigate the functions and mechanisms of NANOG in tumorigenesis, we established tetracycline-inducible NANOG-overexpressing cancer cell lines, including both PCa (Du145 and LNCaP) and breast (MCF-7) cancer cells. NANOG induction promoted drug resistance in MCF-7 cells, tumor regeneration in Du145 cells and, most importantly, castration-resistant tumor development in LNCaP cells. These pro-tumorigenic effects of NANOG were associated with key molecular changes, including an upregulation of molecules such as CXCR4, IGFBP5, CD133 and ALDH1. The present gain-of-function studies, coupled with our recent loss-of-function work, establish the integral role for NANOG in neoplastic processes and shed light on its mechanisms of action.
- Published
- 2011
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.