Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 65
Filter
1.
Cancer Sci ; 112(3): 1075-1083, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33503272

ABSTRACT

Defensins, a class of small cysteine-rich cationic polypeptides across cellular life, are identified as antimicrobial compounds that display direct antimicrobial and immune signaling activities that are involved in the host defense. In addition to their roles in the innate immune system, accumulating studies have reported that some members of defensins are expressed and involved in some cancer cells, such as colon cancer, colorectal cancer, lung cancer and renal cell carcinomas. However, the roles of α-Defensin 5 (DEFA5) in tumorigenesis and development remain unknown. In the present study, bioinformatics analysis and quantitative PCR results showed that the expression level of DEFA5 was dramatically downregulated in human gastric cancer. Overexpression of human DEFA5 in gastric cancer cell lines SGC7901 and BGC823 effectively diminished cell proliferation and reduced the colony forming ability. Moreover, DEFA5 overexpression induced cell cycle arrest by significantly increasing the number of G1-phase cells. Consistently, in vivo tumor formation experiments in nude mice showed the suppression of the tumor growth by DEFA5 overexpression, suggesting an inhibitory effect of DEFA5 in gastric cancer. Mechanistically, DEFA5 directly binds to BMI1, which subsequently decreased its binding at the CDKN2a locus and upregulated the expression of 2 cyclin-dependent kinase inhibitors, p16 and p19. Taken together, we concluded that DEFA5 showed an inhibitory effect in gastric cancer cell growth and may serve as a potential tumor suppressor in gastric cancer.


Subject(s)
Carcinogenesis/genetics , Polycomb Repressive Complex 1/metabolism , Stomach Neoplasms/genetics , alpha-Defensins/metabolism , Adult , Animals , Cell Line, Tumor , Cell Proliferation/genetics , Computational Biology , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p19/genetics , Down-Regulation , Female , G1 Phase Cell Cycle Checkpoints/genetics , Gastrectomy , Gene Expression Regulation, Neoplastic , Gene Knockout Techniques , HEK293 Cells , Humans , Male , Mice , Mice, Nude , Middle Aged , Polycomb Repressive Complex 1/genetics , Stomach/pathology , Stomach/surgery , Stomach Neoplasms/pathology , Stomach Neoplasms/surgery , Up-Regulation , alpha-Defensins/genetics
2.
J Pathol Clin Res ; 7(3): 243-252, 2021 05.
Article in English | MEDLINE | ID: mdl-33428330

ABSTRACT

Adult-type granulosa cell tumors (aGCTs) account for 90% of malignant ovarian sex cord-stromal tumors and 2-5% of all ovarian cancers. These tumors are usually diagnosed at an early stage and are treated with surgery. However, one-third of patients relapse between 4 and 8 years after initial diagnosis, and there are currently no effective treatments other than surgery for these relapsed patients. As the majority of aGCTs (>95%) harbor a somatic mutation in FOXL2 (c.C402G; p.C134W), the aim of this study was to identify genetic mutations besides FOXL2 C402G in aGCTs that could explain the clinical diversity of this disease. Whole-genome sequencing of 10 aGCTs and their matched normal blood was performed to identify somatic mutations. From this analysis, a custom amplicon-based panel was designed to sequence 39 genes of interest in a validation cohort of 83 aGCTs collected internationally. KMT2D inactivating mutations were present in 10 of 93 aGCTs (10.8%), and the frequency of these mutations was similar between primary and recurrent aGCTs. Inactivating mutations, including a splice site mutation in candidate tumor suppressor WNK2 and nonsense mutations in PIK3R1 and NLRC5, were identified at a low frequency in our cohort. Missense mutations were identified in cell cycle-related genes TP53, CDKN2D, and CDK1. From these data, we conclude that aGCTs are comparatively a homogeneous group of tumors that arise from a limited set of genetic events and are characterized by the FOXL2 C402G mutation. Secondary mutations occur in a subset of patients but do not explain the diverse clinical behavior of this disease. As the FOXL2 C402G mutation remains the main driver of this disease, progress in the development of therapeutics for aGCT would likely come from understanding the functional consequences of the FOXL2 C402G mutation.


Subject(s)
Biomarkers, Tumor/genetics , Forkhead Box Protein L2/genetics , Granulosa Cell Tumor/genetics , Mutation , Ovarian Neoplasms/genetics , Adult , Aged , Boston , British Columbia , CDC2 Protein Kinase/genetics , Class Ia Phosphatidylinositol 3-Kinase/genetics , Cyclin-Dependent Kinase Inhibitor p19/genetics , DNA Mutational Analysis , DNA-Binding Proteins/genetics , Europe , Female , Genetic Predisposition to Disease , Granulosa Cell Tumor/pathology , Humans , Intracellular Signaling Peptides and Proteins/genetics , Middle Aged , Neoplasm Proteins/genetics , Ovarian Neoplasms/pathology , Protein Serine-Threonine Kinases/genetics , Tumor Suppressor Protein p53/genetics , Whole Genome Sequencing
3.
Blood Cells Mol Dis ; 85: 102477, 2020 11.
Article in English | MEDLINE | ID: mdl-32711219

ABSTRACT

Chronic myeloid leukemia (CML) is a kind of myeloproliferative disorder caused by a constitutively active BCR-ABL tyrosine kinase. Tyrosine kinase inhibitors (TKIs), imatinib and its derivatives, have achieved great progress in the treatment of CML. However, many CML patients do not respond to TKIs alone. p19INK4d, a cyclin-dependent kinase inhibitor, plays important roles in proliferation, DNA damage repair, apoptosis and cell differentiation, but its role in CML is unknown. Herein, we found that the expression of p19INK4d in CML patients was significantly lower than that in healthy controls. p19INK4d overexpression inhibits cell proliferation through cell cycle arrest, and cooperates with imatinib to inhibit CML more effectively in vitro and in vivo. Mechanistically, p19INK4d decreased the expression of BCR-ABL and its downstream molecules p-Mek1/2, moreover, the expression of Gli-1, c-myc, MUC1, Shh and TC48 also reduced significantly. Collectively, p19INK4d inhibits proliferation and enhances imatinib efficacy in the treatment of CML. These findings maybe have implications for developing potential targets to increase imatinib sensitivity for CML.


Subject(s)
Antineoplastic Agents/therapeutic use , Cyclin-Dependent Kinase Inhibitor p19/genetics , Fusion Proteins, bcr-abl/genetics , Gene Expression Regulation, Leukemic , Imatinib Mesylate/therapeutic use , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Animals , Antineoplastic Agents/pharmacology , Cell Proliferation/drug effects , Cyclin-Dependent Kinase Inhibitor p19/metabolism , Fusion Proteins, bcr-abl/metabolism , Gene Expression Regulation, Leukemic/drug effects , Humans , Imatinib Mesylate/pharmacology , K562 Cells , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Mice, Nude , Signal Transduction/drug effects
4.
J Immunol ; 205(2): 346-358, 2020 07 15.
Article in English | MEDLINE | ID: mdl-32554431

ABSTRACT

IL-23 promotes autoimmune disease, including Th17 CD4 T cell development and autoantibody production. In this study, we show that a deficiency of the p19 component of IL-23 in the autoimmune BXD2 (BXD2-p19-/- ) mouse leads to a shift of the follicular T helper cell program from follicular T helper (Tfh)-IL-17 to Tfh-IFN-γ. Although the germinal center (GC) size and the number of GC B cells remained the same, BXD2-p19-/- mice exhibited a lower class-switch recombination (CSR) in the GC B cells, leading to lower serum levels of IgG2b. Single-cell transcriptomics analysis of GC B cells revealed that whereas Ifngr1, Il21r, and Il4r genes exhibited a synchronized expression pattern with Cxcr5 and plasma cell program genes, Il17ra exhibited a synchronized expression pattern with Cxcr4 and GC program genes. Downregulation of Ighg2b in BXD2-p19-/- GC B cells was associated with decreased expression of CSR-related novel base excision repair genes that were otherwise predominantly expressed by Il17ra + GC B cells in BXD2 mice. Together, these results suggest that although IL-23 is dispensable for GC formation, it is essential to promote a population of Tfh-IL-17 cells. IL-23 acts indirectly on Il17ra + GC B cells to facilitate CSR-related base excision repair genes during the dark zone phase of GC B cell development.


Subject(s)
Autoimmune Diseases/immunology , B-Lymphocytes/immunology , Germinal Center/immunology , Immunoglobulin G/metabolism , Interleukin-23/metabolism , T-Lymphocyte Subsets/immunology , Th17 Cells/immunology , Animals , Cell Differentiation , Cyclin-Dependent Kinase Inhibitor p19/genetics , Immunoglobulin Class Switching , Immunoglobulin G/genetics , Interferon-gamma/metabolism , Interleukin-23/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout
5.
Curr Drug Targets ; 21(1): 96-102, 2020.
Article in English | MEDLINE | ID: mdl-31400265

ABSTRACT

Cyclin-dependent kinase inhibitors (CDKIs) are important cell cycle regulators. The CDKI family is composed of the INK4 family and the CIP/KIP family. p19INK4d belongs to the INK4 gene family and is involved in a series of normal physiological activities and the pathogenesis of diseases. Many factors play regulatory roles in the p19INK4d gene expression at the transcriptional and posttranscriptional levels. p19INK4d not only regulates the cell cycle but also plays regulatory roles in apoptosis, DNA damage repair, cell differentiation of hematopoietic cells, and cellular senescence. In this review, the regulatory network of the p19INK4d gene expression and its biological functions are summarized, which provides a basis for further study of p19INK4d as a drug target for disease treatment.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p19/physiology , Animals , Apoptosis , Cell Cycle , Cell Differentiation , Cellular Senescence , Cyclin-Dependent Kinase Inhibitor p19/biosynthesis , Cyclin-Dependent Kinase Inhibitor p19/genetics , Cyclin-Dependent Kinase Inhibitor p19/metabolism , DNA Repair , Gene Expression Regulation , Humans , Mice
6.
Free Radic Biol Med ; 138: 23-32, 2019 07.
Article in English | MEDLINE | ID: mdl-31029787

ABSTRACT

AIMS: Oxidative stress is known to induce early replicative senescence. Senescence has been proposed to work as a barrier to immortalization and tumor development. Here, we aimed to evaluate the impact of the loss of peroxisome proliferator activated receptor γ co-activator 1α (PGC-1α), a master regulator of oxidative metabolism and mitochondrial reactive oxygen species (ROS) generation, on replicative senescence and immortalization in mouse embryonic fibroblasts (MEFs). RESULTS: We found that primary MEFs lacking PGC-1α showed higher levels of ROS than wild-type MEFs at all cell passages tested. The elevated production of ROS was associated with higher levels of oxidative DNA damage and the increased formation of DNA double-strand breaks. Evaluation of the induction of DNA repair systems in response to γ-radiation indicated that the loss of PGC-1α also resulted in a small but significant reduction in their activity. DNA damage induced the early activation of senescence markers, including an increase in the number of ß-galactosidase-positive cells, the induction of p53 phosphorylation, and the increase in p16 and p19 protein. These changes were, however, not sufficient to reduce proliferation rates of PGC-1α-deficient MEFs at any cell passage tested. Moreover, PGC-1α-deficient cells escaped replicative senescence. INNOVATION & CONCLUSION: PGC-1α plays an important role in the control of cellular senescence and immortalization.


Subject(s)
Cellular Senescence/radiation effects , DNA Repair , DNA/genetics , Fibroblasts/radiation effects , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Animals , Biomarkers/metabolism , Cell Proliferation/genetics , Cell Proliferation/radiation effects , Cellular Senescence/genetics , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Cyclin-Dependent Kinase Inhibitor p19/genetics , Cyclin-Dependent Kinase Inhibitor p19/metabolism , DNA/metabolism , DNA Breaks, Double-Stranded/radiation effects , Embryo, Mammalian , Fibroblasts/cytology , Fibroblasts/metabolism , Gamma Rays , Gene Expression Regulation , Mice , Mice, Knockout , Mitochondria/metabolism , Mitochondria/radiation effects , Oxidative Stress , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/deficiency , Phosphorylation/radiation effects , Reactive Oxygen Species/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , beta-Galactosidase/genetics , beta-Galactosidase/metabolism
7.
J Med Food ; 21(7): 678-688, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29851371

ABSTRACT

Schisantherin A (SCA) was evaluated for possible function in restoring the learning and memory impairment induced by D-galactose in mice. ICR mice were treated with D-galactose subcutaneously (220 mg·kg-1), and followed by SCA in different doses (1.25, 2.50 and 5.00 mg·kg-1, administered orally) for 42 days. Effects of SCA on learning and memory were examined by step-through tests and Morris water maze tests. The activity of superoxide dismutase (SOD), the content of malondialdehyde (MDA) in the peripheral blood and hippocampus of mice were assayed by water-soluble tetrazolium-1 (WST-1) and thiobarbituric acid (TBA) methods. The contents of 8 hydroxy deoxy guanosine (8-OHdG) in the hippocampus of mice were detected by immunosorbent assay methods, respectively. Quantitative real-time PCR and Western Blot were respectively used to detect the expression of p19, p53, p21, cyclin D1, CDK4 and RB genes, and the phosphorylation of RB in the hippocampus of mice. We found that SCA significantly improved the learning and memory impairment induced by D-galactose in mice. After SCA treatment, SOD activity was increased and the content of MDA was decreased in both peripheral blood and hippocampus of mice. 8-OHDG content was also decreased in the hippocampus of mice. Furthermore, the expression of p19, p53 and p21 genes was reduced and the expression of cyclin D1 and CDK4 and the phosphorylation of RB protein were elevated in the hippocampus. SCA may improve the learning and memory impairment induced by D-galactose by enhancing the antioxidant capacity, and regulating the expression of p19/p53/p21/cyclinD1/CDK4 genes, and the phosphorylation of RB protein in the hippocampus of mice.


Subject(s)
Cyclooctanes/administration & dosage , Dioxoles/administration & dosage , Drugs, Chinese Herbal/administration & dosage , Learning Disabilities/drug therapy , Lignans/administration & dosage , Memory Disorders/drug therapy , Schisandra/chemistry , Animals , Cyclin D1/genetics , Cyclin D1/metabolism , Cyclin-Dependent Kinase 4/genetics , Cyclin-Dependent Kinase 4/metabolism , Cyclin-Dependent Kinase Inhibitor p19/genetics , Cyclin-Dependent Kinase Inhibitor p19/metabolism , Galactose/adverse effects , Gene Expression/drug effects , Hippocampus/drug effects , Hippocampus/metabolism , Humans , Learning Disabilities/genetics , Learning Disabilities/metabolism , Learning Disabilities/psychology , Male , Memory/drug effects , Memory Disorders/genetics , Memory Disorders/metabolism , Memory Disorders/psychology , Mice , Mice, Inbred ICR , Retinoblastoma Protein/genetics , Retinoblastoma Protein/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , rho GTP-Binding Proteins/genetics , rho GTP-Binding Proteins/metabolism
8.
J Cell Physiol ; 233(11): 8617-8629, 2018 11.
Article in English | MEDLINE | ID: mdl-29761857

ABSTRACT

Parkinson's disease (PD) is a common neurodegenerative disorder due to the loss of dopaminergic neurons in the substantia nigra. This study focuses on the effect of microRNA-329 (miR-329) on nigral dopaminergic neurons in a rat model of PD via the FoxO3a signaling pathway by binding to CDKN2D. Brain tissues from the substantia nigra were taken from the rats in two groups. TUNEL staining was used to observe tyrosine hydroxylase (TH)-positive neurons. Nigral dopaminergic neurons were randomized into the normal, blank, negative control (NC), miR-329 mimics, miR-329 inhibitors, small interfering (siRNA)-CDKN2D, and miR-329 inhibitors + siRNA-CDKN2D groups. Expressions of miR-329, CDKN2D, FoxO3a, AKT, caspase-3 and Bcl-2 were determined using RT-qPCR and western blotting. Apoptosis rate of nigral dopaminergic neurons in 7 groups was determined by flow cytometry. Compared with the blank and NC groups, the miR-329 mimics group showed increased miR-329 and caspase-3 expressions as well as decreased expressions of CDKN2D, FoxO3a, AKT, and Bcl-2, the siRNA-CDKN2D group indicated enhanced expressions of caspase-3 and declined expressions of CDKN2D, FoxO3a, AKT, and Bcl-2, and the miR-329 inhibitors group revealed decreased miR-329 and caspase-3 expressions and increased expressions of CDKN2D, FoxO3a, AKT, and Bcl-2. The apoptosis rate of nigral dopaminergic neurons was significantly increased in the miR-329 mimics and siRNA-CDKN2D groups, but was decreased in the miR-329 inhibitors group. Our data suggested that downregulated miR-329 could inhibit apoptosis of nigral dopaminergic neurons in a rat model of PD by upregulating the expression of CDKN2D via the activation of the FoxO3a signaling pathway.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p19/genetics , Forkhead Box Protein O3/genetics , MicroRNAs/genetics , Parkinson Disease/genetics , Animals , Apoptosis/genetics , Caspase 3/genetics , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/pathology , Gene Expression Regulation , Humans , MicroRNAs/antagonists & inhibitors , Parkinson Disease/pathology , Proto-Oncogene Proteins c-bcl-2/genetics , RNA, Small Interfering/genetics , Rats , Signal Transduction , Substantia Nigra/metabolism , Substantia Nigra/pathology
9.
Sci Rep ; 7(1): 16577, 2017 11 29.
Article in English | MEDLINE | ID: mdl-29185453

ABSTRACT

YAP is a downstream nuclear transcription factor of Hippo pathway which plays an essential role in development, cell growth, organ size and homeostasis. It was previously identified that elevation of YAP in genomics of genetic engineered mouse (GEM) model of prostate cancer is associated with Pten/Trp53 inactivation and ARF elevation hypothesizing the essential crosstalk of AKT/mTOR/YAP with ARF in prostate cancer. However, the detailed function and trafficking of YAP in cancer cells remains unclear. Using GEM microarray model, we found ARF dysregulates Hippo and Wnt pathways. In particular, ARF knockdown reduced non-nuclear localization of YAP which led to an increase in F-actin. Mechanistically, ARF knockdown suppressed protein turnover of ß-catenin/YAP, and therefore enhanced the activity of AKT and phosphorylation of YAP. Moreover, we found tea-derived carbon dots can interact with ARF in nucleus that may further lead to the non-nuclear localization of YAP. Thus, we reported a novel crosstalk of ARF/ß-catenin dysregulated YAP in Hippo pathway and a new approach to stimulate ARF-mediated signaling to inhibit nuclear YAP using nanomaterials implicating an innovative avenue for treatment of cancer.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Carbon/chemistry , Nanotechnology/methods , Phosphoproteins/metabolism , Tea/chemistry , Animals , Cell Cycle Proteins , Cell Nucleus/metabolism , Cyclin-Dependent Kinase Inhibitor p19/genetics , Cyclin-Dependent Kinase Inhibitor p19/metabolism , Epithelial-Mesenchymal Transition/genetics , Epithelial-Mesenchymal Transition/physiology , Hippo Signaling Pathway , Male , Mice , Microscopy, Confocal , Prostatic Neoplasms/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Wnt Signaling Pathway/genetics , Wnt Signaling Pathway/physiology , YAP-Signaling Proteins
10.
Curr Biol ; 27(21): 3302-3314.e6, 2017 Nov 06.
Article in English | MEDLINE | ID: mdl-29112861

ABSTRACT

Mesothelioma is a fatal tumor of the pleura and is strongly associated with asbestos exposure. The molecular mechanisms underlying the long latency period of mesothelioma and driving carcinogenesis are unknown. Moreover, late diagnosis means that mesothelioma research is commonly focused on end-stage disease. Although disruption of the CDKN2A (INK4A/ARF) locus has been reported in end-stage disease, information is lacking on the status of this key tumor suppressor gene in pleural lesions preceding mesothelioma. Manufactured carbon nanotubes (CNTs) are similar to asbestos in terms of their fibrous shape and biopersistent properties and thus may pose an asbestos-like inhalation hazard. Here we show that instillation of either long CNTs or long asbestos fibers into the pleural cavity of mice induces mesothelioma that exhibits common key pro-oncogenic molecular events throughout the latency period of disease progression. Sustained activation of pro-oncogenic signaling pathways, increased proliferation, and oxidative DNA damage form a common molecular signature of long-CNT- and long-asbestos-fiber-induced pathology. We show that hypermethylation of p16/Ink4a and p19/Arf in CNT- and asbestos-induced inflammatory lesions precedes mesothelioma; this results in silencing of Cdkn2a (Ink4a/Arf) and loss of p16 and p19 protein, consistent with epigenetic alterations playing a gatekeeper role in cancer. In end-stage mesothelioma, silencing of p16/Ink4a is sustained and deletion of p19/Arf is detected, recapitulating human disease. This study addresses the long-standing question of which early molecular changes drive carcinogenesis during the long latency period of mesothelioma development and shows that CNT and asbestos pose a similar health hazard.


Subject(s)
Asbestos/toxicity , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Cyclin-Dependent Kinase Inhibitor p19/metabolism , Lung Neoplasms/chemically induced , Lung Neoplasms/pathology , Mesothelioma/chemically induced , Mesothelioma/pathology , Nanotubes, Carbon/toxicity , Aged , Animals , Carcinogenesis/chemically induced , Carcinogenesis/genetics , Cell Proliferation/drug effects , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p19/genetics , Female , Humans , Lung Neoplasms/genetics , Male , Mesothelioma/genetics , Mesothelioma, Malignant , Methylation/drug effects , Mice , Mice, Inbred C57BL , Middle Aged
11.
Ann Clin Lab Sci ; 47(5): 523-528, 2017 Sep.
Article in English | MEDLINE | ID: mdl-29066476

ABSTRACT

Medullary thyroid carcinoma (MTC), an aggressive form of thyroid cancer, occurs sporadically in approximately 75% of MTCs. RET and RAS mutations play a role in about 40% and 15%, respectively, of sporadic MTCs and are predominant drivers in MTC pathways. These mutations are some of the most comprehensively described and screened for in MTC patients; however, in recent studies, other mutations in the CDKN2C gene (p18) have been implicated in the tumorigenesis of MTC. Comparative genomic hybridization analysis revealed that approximately 40% of sporadic MTC samples have loss of CDKN2C at chromosome 1p32 in addition to frequent losses of CDKN2D (p19) at chromosome 19p13. However, no feasible routine method had been established to detect loss of heterozygosity (LOH) of CDKN2C and CD-KN2D The aim of this study is to assess the feasibility of using Fluorescence in situ Hybridization (FISH) to screen MTC patients for CDKN2C and CDKN2D deletions. We subjected 5 formalin-fixed, paraffin-embedded (FFPE) MTC samples with defined RET/RAS mutations to dual-color FISH assays to detect loss of CDKN2C and/or CDKN2D We prepared spectrum orange probes using the bacterial artificial chromosomes RP11-779F9 for CDKN2C (p18) and RP11-177J4 for CDKN2D (p19) and prepared spectrum green control probes to the 1q25.2 and 19q11 regions (RP11-1146A3 and RP11-942P7, respectively). Nine FFPE normal thyroid tissue samples were used to establish the cutoff values for the FISH signal patterns. Of the five FFPE MTC samples, four and one yielded a positive significant result for CDKNN2C loss and CDKN2D loss, respectively. The results of a Clinical Laboratory Improvement Amendments validation with a CDKN2C/CKS1B probe set for CDKN2C (p18) loss of heterozygosity were 100% concordant with the FISH results obtained in this study. Thus, FISH is a fast and reliable diagnostic or prognostic indicator of gene loss in MTC.


Subject(s)
Carcinoma, Neuroendocrine/genetics , Cyclin-Dependent Kinase Inhibitor p18/genetics , Cytogenetic Analysis/methods , Neoplasm Proteins/genetics , Thyroid Gland/metabolism , Thyroid Neoplasms/genetics , Cancer Care Facilities , Carcinoma, Neuroendocrine/diagnosis , Carcinoma, Neuroendocrine/metabolism , Carcinoma, Neuroendocrine/pathology , Chromosomes, Artificial, Bacterial/metabolism , Cyclin-Dependent Kinase Inhibitor p18/metabolism , Cyclin-Dependent Kinase Inhibitor p19/genetics , Cyclin-Dependent Kinase Inhibitor p19/metabolism , DNA, Complementary/metabolism , DNA, Recombinant/metabolism , Feasibility Studies , Gene Deletion , Humans , In Situ Hybridization, Fluorescence , Loss of Heterozygosity , Neoplasm Proteins/metabolism , Prognosis , Recombinant Proteins/metabolism , Reproducibility of Results , Texas , Thyroid Gland/pathology , Thyroid Neoplasms/diagnosis , Thyroid Neoplasms/metabolism , Thyroid Neoplasms/pathology , Time Factors
12.
Aging Cell ; 16(5): 1035-1042, 2017 10.
Article in English | MEDLINE | ID: mdl-28613007

ABSTRACT

Transcriptional coactivator with PDZ-binding motif (TAZ) directly interacts with transcription factors and regulates their transcriptional activity. Extensive functional studies have shown that TAZ plays critical regulatory roles in stem cell proliferation, differentiation, and survival and also modulates the development of organs such as the lung, kidney, heart, and bone. Despite the importance of TAZ in stem cell maintenance, TAZ function has not yet been evaluated in spermatogenic stem cells of the male reproductive system. Here, we investigated the expression and functions of TAZ in mouse testis. TAZ was expressed in spermatogenic stem cells; however, its deficiency caused significant structural abnormalities, including atrophied tubules, widened interstitial space, and abnormal Leydig cell expansion, thereby resulting in lowered sperm counts and impaired fertility. Furthermore, TAZ deficiency increased the level of apoptosis and senescence in spermatogenic cells and Leydig cells upon aging. The expression of senescence-associated ß-galactosidase (SA-ßgal), secretory phenotypes, and cyclin-dependent kinase inhibitors (p16, p19, and p21) significantly increased in the absence of TAZ. TAZ downregulation in testicular cells further increased SA-ßgal and p21 expression induced by oxidative stress, whereas TAZ overexpression decreased p21 induction and prevented senescence. Mechanistic studies showed that TAZ suppressed DNA-binding activity of p53 through a direct interaction and thus attenuated p53-induced p21 gene transcription. Our results suggested that TAZ may suppress apoptosis and premature senescence in spermatogenic cells by inhibiting the p53-p21 signaling pathway, thus playing important roles in the maintenance and control of reproductive function.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Aging/genetics , Cyclin-Dependent Kinase Inhibitor p21/genetics , Oligospermia/genetics , Spermatogonia/metabolism , Tumor Suppressor Protein p53/genetics , Adaptor Proteins, Signal Transducing/deficiency , Aging/metabolism , Animals , Apoptosis , Cellular Senescence , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Cyclin-Dependent Kinase Inhibitor p19/genetics , Cyclin-Dependent Kinase Inhibitor p19/metabolism , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Gene Expression Regulation, Developmental , Leydig Cells/metabolism , Leydig Cells/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Oligospermia/metabolism , Oligospermia/physiopathology , Signal Transduction , Spermatogenesis/genetics , Spermatogonia/pathology , Stem Cells/metabolism , Stem Cells/pathology , Trans-Activators , Tumor Suppressor Protein p53/metabolism , beta-Galactosidase/genetics , beta-Galactosidase/metabolism
13.
Tumour Biol ; 39(6): 1010428317704821, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28653900

ABSTRACT

Prostate cancer is the most commonly diagnosed malignancy and is the second leading deadly reason among male cancer. WDFY2, which is found to be a cancer-specific fusion gene with CDKN2D in ovarian cancer, is a new gene with unknown function in carcinogenesis. In this study, we investigated the role of WDFY2 in prostate cancer development. We examined WDFY2 expression in human prostate tissue specimens and prostate cancer cell lines BPH-1, LNCaP, PC3, and DU-145. Overexpression of WDFY2 was performed to evaluate the role of WDFY2 in cell proliferation, migration, and colony formation of prostate cancer cells. We analyzed the clinical impact and prognosis of WDFY2 expression on the progress of prostate cancer through data from online datasets. Our results showed that WDFY2 had lower expression level in prostate tumors than in normal tissues. Overexpression of WDFY2 in prostate cancer cells DU145 and PC-3 led to the suppression of cancer cell migration and colony formation. Furthermore, we found that WDFY2 exerted its role by suppressing the activity of Akt pathway other than the epithelial-mesenchymal transition progression. In conclusion, we have uncovered WDFY2 as a tumor suppressor gene and a new potential biomarker for cancer progression. Our results showed that WDFY2 inhibited cancer cell colony formation and migration via suppressing Akt pathway, making it a potential new therapeutic target in prostate cancer.


Subject(s)
Cell Proliferation/genetics , Epithelial-Mesenchymal Transition/genetics , Intracellular Signaling Peptides and Proteins/genetics , Prostatic Neoplasms/genetics , Aged , Carcinogenesis/genetics , Cell Line, Tumor , Cell Movement/genetics , Cyclin-Dependent Kinase Inhibitor p19/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Intracellular Signaling Peptides and Proteins/biosynthesis , Male , Middle Aged , Prognosis , Prostatic Neoplasms/pathology , Signal Transduction
14.
Cell Rep ; 18(4): 977-990, 2017 01 24.
Article in English | MEDLINE | ID: mdl-28122246

ABSTRACT

The identity of the glioblastoma (GBM) cell of origin and its contributions to disease progression and treatment response remain largely unknown. We have analyzed how the phenotypic state of the initially transformed cell affects mouse GBM development and essential GBM cell (GC) properties. We find that GBM induced in neural stem-cell-like glial fibrillary acidic protein (GFAP)-expressing cells in the subventricular zone of adult mice shows accelerated tumor development and produces more malignant GCs (mGC1GFAP) that are less resistant to cancer drugs, compared with those originating from more differentiated nestin- (mGC2NES) or 2,'3'-cyclic nucleotide 3'-phosphodiesterase (mGC3CNP)-expressing cells. Transcriptome analysis of mouse GCs identified a 196 mouse cell origin (MCO) gene signature that was used to partition 61 patient-derived GC lines. Human GC lines that clustered with the mGC1GFAP cells were also significantly more self-renewing, tumorigenic, and sensitive to cancer drugs compared with those that clustered with mouse GCs of more differentiated origin.


Subject(s)
Brain Neoplasms/pathology , Glioblastoma/pathology , 2',3'-Cyclic-Nucleotide Phosphodiesterases/genetics , 2',3'-Cyclic-Nucleotide Phosphodiesterases/metabolism , Adult , Aged , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Brain/cytology , Brain/metabolism , Brain/pathology , Brain Neoplasms/drug therapy , Brain Neoplasms/mortality , Cell Differentiation , Cell Proliferation/drug effects , Cell Self Renewal , Cell Survival/drug effects , Cyclin-Dependent Kinase Inhibitor p19/deficiency , Cyclin-Dependent Kinase Inhibitor p19/genetics , Disease-Free Survival , Female , Glial Fibrillary Acidic Protein/genetics , Glial Fibrillary Acidic Protein/metabolism , Glioblastoma/drug therapy , Glioblastoma/mortality , Humans , Male , Mice , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Middle Aged , Nestin/genetics , Nestin/metabolism , Tumor Cells, Cultured
15.
Sci Signal ; 10(461)2017 01 10.
Article in English | MEDLINE | ID: mdl-28074012

ABSTRACT

The tumor suppressor p16INK4a, one protein encoded by the INK4/ARF locus, is frequently absent in multiple cancers, including non-small cell lung cancer (NSCLC). Whereas increased methylation of the encoding gene (CDKN2A) accounts for its loss in a third of patients, no molecular explanation exists for the remainder. We unraveled an alternative mechanism for the silencing of the INK4/ARF locus involving the E3 ubiquitin ligase and transcriptional cofactor E6AP (also known as UBE3A). We found that the expression of three tumor suppressor genes encoded in the INK4/ARF locus (p15INK4b, p16INK4a, and p19ARF) was decreased in E6AP-/- mouse embryo fibroblasts. E6AP induced the expression of the INK4/ARF locus at the transcriptional level by inhibiting CDC6 transcription, a gene encoding a key repressor of the locus. Luciferase assays revealed that E6AP inhibited CDC6 expression by reducing its E2F1-dependent transcription. Chromatin immunoprecipitation analysis indicated that E6AP reduced the amount of E2F1 at the CDC6 promoter. In a subset of NSCLC samples, an E6AP-low/CDC6-high/p16INK4a-low protein abundance profile correlated with low methylation of the gene encoding p16INK4a (CDKN2A) and poor patient prognosis. These findings define a previously unrecognized tumor-suppressive role for E6AP in NSCLC, reveal an alternative silencing mechanism of the INK4/ARF locus, and reveal E6AP as a potential prognostic marker in NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Cyclin-Dependent Kinase Inhibitor p15/genetics , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p19/genetics , Lung Neoplasms/genetics , Ubiquitin-Protein Ligases/genetics , Animals , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p15/metabolism , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Cyclin-Dependent Kinase Inhibitor p19/metabolism , DNA Methylation , E2F1 Transcription Factor/genetics , E2F1 Transcription Factor/metabolism , Embryo, Mammalian/cytology , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Expression Regulation, Neoplastic , Humans , Kaplan-Meier Estimate , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mice, Knockout , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Promoter Regions, Genetic/genetics , Protein Binding , Ubiquitin-Protein Ligases/metabolism
16.
Semin Cell Dev Biol ; 65: 96-105, 2017 05.
Article in English | MEDLINE | ID: mdl-27864084

ABSTRACT

Vestibular sensation is essential for gaze stabilization, balance, and perception of gravity. The vestibular receptors in mammals, Type I and Type II hair cells, are located in five small organs in the inner ear. Damage to hair cells and their innervating neurons can cause crippling symptoms such as vertigo, visual field oscillation, and imbalance. In adult rodents, some Type II hair cells are regenerated and become re-innervated after damage, presenting opportunities for restoring vestibular function after hair cell damage. This article reviews features of vestibular sensory cells in mammals, including their basic properties, how they develop, and how they are replaced after damage. We discuss molecules that control vestibular hair cell regeneration and highlight areas in which our understanding of development and regeneration needs to be deepened.


Subject(s)
Cell Lineage/genetics , Gravity Sensing/physiology , Hair Cells, Vestibular/cytology , Postural Balance/physiology , Regeneration/genetics , Animals , Cell Differentiation , Cell Proliferation , Cyclin-Dependent Kinase Inhibitor p19/genetics , Cyclin-Dependent Kinase Inhibitor p19/metabolism , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Gene Expression Regulation, Developmental , Hair Cells, Auditory/cytology , Hair Cells, Auditory/metabolism , Hair Cells, Vestibular/classification , Hair Cells, Vestibular/metabolism , Mice , Organogenesis/genetics , Signal Transduction , beta Catenin/genetics , beta Catenin/metabolism
17.
Cell Death Dis ; 7(10): e2430, 2016 10 20.
Article in English | MEDLINE | ID: mdl-27763644

ABSTRACT

A better understanding of the mechanisms involved in megakaryocyte maturation will facilitate the generation of platelets in vitro and their clinical applications. A microRNA, miR-125b, has been suggested to have important roles in the self-renewal of megakaryocyte-erythroid progenitors and in platelet generation. However, miR-125b is also critical for hematopoietic stem cell self-renewal. Thus, the function of miR-125b and the complex signaling pathways regulating megakaryopoiesis remain to be elucidated. In this study, an attentive examination of the endogenous expression of miR-125b during megakaryocyte differentiation was performed. Accordingly, the differentiation of hematopoietic stem cells requires the downregulation of miR-125b, whereas megakaryocyte determination and maturation synchronize with miR-125b accumulation. The overexpression of miR-125b improves megakaryocytic differentiation of K562 and UT-7 cells. Furthermore, stage-specific overexpression of miR-125b in primary cells demonstrates that miR-125b mediates an enhancement of megakaryocytic differentiation after megakaryocyte determination, the stage at which megakaryocytes are negative for the expression of the hematopoietic progenitor marker CD34. The identification of miR-125b targets during megakaryopoiesis was focused on negative regulators of cell cycle because the transition of the G1/S phase has been associated with megakaryocyte polyploidization. Real-time PCR, western blot and luciferase reporter assay reveal that p19INK4D is a direct target of miR-125b. P19INK4D knockdown using small interfering RNA (siRNA) in megakaryocyte-induced K562 cells, UT-7 cells and CD61+ promegakaryocytes results in S-phase progression and increased polyploidy, as well as improved megakaryocyte differentiation, similarly to the effects of miR-125b overexpression. P19INK4D overexpression reverses these effects, as indicated by reduced expression of megakaryocyte markers, G1-phase arrest and polyploidy decrease. P19INK4D knockdown in miR-125b downregulated cells or p19INK4D overexpression in miR-125b upregulated cells rescued the effect of miR-125b. Taken together, these findings suggest that miR-125b expression positively regulates megakaryocyte development since the initial phases of megakaryocyte determination, and p19INK4D is one of the key mediators of miR-125b activity during the onset of megakaryocyte polyploidization.


Subject(s)
Cell Differentiation/genetics , Cyclin-Dependent Kinase Inhibitor p19/genetics , Megakaryocytes/cytology , MicroRNAs/metabolism , Base Sequence , Blood Platelets/drug effects , Blood Platelets/metabolism , Colony-Forming Units Assay , Cyclin-Dependent Kinase Inhibitor p19/metabolism , Down-Regulation/drug effects , Fetal Blood/cytology , G1 Phase/drug effects , Gene Knockdown Techniques , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/metabolism , Humans , K562 Cells , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , Megakaryocytes/drug effects , Megakaryocytes/metabolism , MicroRNAs/genetics , Polyploidy , Tetradecanoylphorbol Acetate/pharmacology , Up-Regulation/drug effects , Up-Regulation/genetics
18.
Exp Hematol ; 44(4): 231-7.e4, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26854485

ABSTRACT

The MOZ-TIF2 translocation, which fuses monocytic leukemia zinc finger protein (MOZ) histone acetyltransferase (HAT) with the nuclear co-activator TIF2, is associated with the development of acute myeloid leukemia. We recently found that in the absence of MOZ HAT activity, p16(INK4a) transcriptional levels are significantly increased, triggering an early entrance into replicative senescence. Because oncogenic fusion proteins must bypass cellular safeguard mechanisms, such as senescence and apoptosis, to induce leukemia, we hypothesized that this repressive activity of MOZ over p16(INK4a) transcription could be preserved, or even reinforced, in MOZ leukemogenic fusion proteins, such as MOZ-TIF2. We describe here that, indeed, MOZ-TIF2 silences expression of the CDKN2A locus (p16(INK4a) and p19(ARF)), inhibits the triggering of senescence and enhances proliferation, providing conditions favorable to the development of leukemia. Furthermore, we describe that abolishing the MOZ HAT activity of the fusion protein leads to a significant increase in expression of the CDKN2A locus and the number of hematopoietic progenitors undergoing senescence. Finally, we report that inhibition of senescence by MOZ-TIF2 is associated with increased apoptosis, suggesting a role for the fusion protein in p53 apoptosis-versus-senescence balance. Our results underscore the importance of the HAT activity of MOZ, preserved in the fusion protein, for repression of the CDKN2A locus transcription and the subsequent block of senescence, a necessary step for the survival of leukemic cells.


Subject(s)
Cellular Senescence/genetics , Gene Expression , Histone Acetyltransferases/genetics , Nuclear Receptor Coactivator 2/genetics , Oncogene Proteins, Fusion/genetics , Animals , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Cyclin-Dependent Kinase Inhibitor p19/genetics , Cyclin-Dependent Kinase Inhibitor p19/metabolism , Flow Cytometry , Genetic Loci , Histone Acetyltransferases/metabolism , Mice , Nuclear Receptor Coactivator 2/metabolism , Oncogene Proteins, Fusion/metabolism , Proto-Oncogene Proteins c-kit/genetics , Proto-Oncogene Proteins c-kit/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transcription, Genetic , Transduction, Genetic
19.
World J Gastroenterol ; 21(19): 5867-76, 2015 May 21.
Article in English | MEDLINE | ID: mdl-26019450

ABSTRACT

AIM: To investigate the underlying molecular mechanisms of miR-451 to inhibit proliferation of esophageal carcinoma cell line EC9706. METHODS: Assays for cell growth, apoptosis and invasion were used to evaluate the effects of miR-451 expression on EC cells. Luciferase reporter and Western blot assays were used to test whether cyclin-dependent kinase inhibitor 2D (CDKN2D) and MAP3K1 act as major targets of miR-451. RESULTS: The results showed that CDKN2D and MAP3K1 are direct targets of miR-451. CDKN2D and MAP3K1 overexpression reversed the effect of miR-451. MiR-451 inhibited the proliferation of EC9706 by targeting CDKN2D and MAP3K1. CONCLUSION: These findings suggest that miR-451 might be a novel prognostic biomarker and a potential target for the treatment of esophageal squamous cell carcinoma in the future.


Subject(s)
Carcinoma, Squamous Cell/enzymology , Carcinoma/enzymology , Cell Proliferation , Cyclin-Dependent Kinase Inhibitor p19/metabolism , Esophageal Neoplasms/enzymology , MAP Kinase Kinase Kinase 1/metabolism , MicroRNAs/metabolism , Carcinoma/genetics , Carcinoma/pathology , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p19/genetics , Esophageal Neoplasms/genetics , Esophageal Neoplasms/pathology , Esophageal Squamous Cell Carcinoma , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Humans , MAP Kinase Kinase Kinase 1/genetics , MicroRNAs/genetics , Signal Transduction , Transfection
20.
Mol Cell ; 57(5): 860-872, 2015 Mar 05.
Article in English | MEDLINE | ID: mdl-25702873

ABSTRACT

During apoptosis, the mitochondrial outer membrane is permeabilized, leading to the release of cytochrome c that activates downstream caspases. Mitochondrial outer membrane permeabilization (MOMP) has historically been thought to occur synchronously and completely throughout a cell, leading to rapid caspase activation and apoptosis. Using a new imaging approach, we demonstrate that MOMP is not an all-or-nothing event. Rather, we find that a minority of mitochondria can undergo MOMP in a stress-regulated manner, a phenomenon we term "minority MOMP." Crucially, minority MOMP leads to limited caspase activation, which is insufficient to trigger cell death. Instead, this caspase activity leads to DNA damage that, in turn, promotes genomic instability, cellular transformation, and tumorigenesis. Our data demonstrate that, in contrast to its well-established tumor suppressor function, apoptosis also has oncogenic potential that is regulated by the extent of MOMP. These findings have important implications for oncogenesis following either physiological or therapeutic engagement of apoptosis.


Subject(s)
Apoptosis/physiology , DNA Damage , Genomic Instability , Mitochondrial Membranes/physiology , Animals , Apoptosis/drug effects , Biphenyl Compounds/pharmacology , Blotting, Western , Caspases/metabolism , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p19/deficiency , Cyclin-Dependent Kinase Inhibitor p19/genetics , Dose-Response Relationship, Drug , Embryo, Mammalian/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , HCT116 Cells , HeLa Cells , Histones/metabolism , Humans , MCF-7 Cells , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Confocal , Nitrophenols/pharmacology , Permeability , Piperazines/pharmacology , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Proto-Oncogene Proteins c-bcl-2/metabolism , Staurosporine/pharmacology , Sulfonamides/pharmacology , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...