Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
2.
Cancer Res ; 78(15): 4191-4202, 2018 08 01.
Article in English | MEDLINE | ID: mdl-29880481

ABSTRACT

Triple-negative breast cancer (TNBC) has a poor prognosis due to its aggressive characteristics and lack of targeted therapies. Cytotoxic chemotherapy may reduce tumor bulk, but leaves residual disease due to the persistence of chemotherapy-resistant breast cancer stem cells (BCSC), which are critical for tumor recurrence and metastasis. Here, we demonstrate that hypoxia-inducible factor (HIF)-1-dependent regulation of mitogen-activated protein kinase (MAPK) signaling pathways contributes to chemotherapy-induced BCSC enrichment. Chemotherapy increased DUSP9 expression and decreased DUSP16 expression in a HIF1-dependent manner, leading to inhibition of ERK and activation of p38 signaling pathways, respectively. Inhibition of ERK caused transcriptional induction of the pluripotency factor Nanog through decreased inactivating phosphorylation of FoxO3, while activation of p38 stabilized Nanog and Klf4 mRNA through increased inactivating phosphorylation of RNA-binding protein ZFP36L1, both of which promoted specification of the BCSC phenotype. Inhibition of HIF1 or p38 signaling blocked chemotherapy-induced pluripotency factor expression and BCSC enrichment. These surprising results delineate a mechanism by which a transcription factor switches cells from ERK to p38 signaling in response to chemotherapy and suggest that therapeutic targeting of HIF1 or the p38 pathway in combination with chemotherapy will block BCSC enrichment and improve outcome in TNBC.Significance: These findings provide a molecular mechanism that may account for the increased relapse rate of women with TNBC who are treated with cytotoxic chemotherapy and suggest that combining chemotherapy with an inhibitor of HIF1 or p38 activity may increase patient survival. Cancer Res; 78(15); 4191-202. ©2018 AACR.


Subject(s)
Dual-Specificity Phosphatases/metabolism , Hypoxia-Inducible Factor 1/metabolism , MAP Kinase Signaling System/physiology , Mitogen-Activated Protein Kinase Phosphatases/metabolism , Neoplastic Stem Cells/metabolism , Triple Negative Breast Neoplasms/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Female , Forkhead Box Protein O3/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/physiology , Humans , Kruppel-Like Factor 4 , MAP Kinase Signaling System/drug effects , Neoplastic Stem Cells/drug effects , Phosphorylation/drug effects , Phosphorylation/physiology , RNA, Messenger/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology , Transcription, Genetic/drug effects , Transcription, Genetic/physiology , Triple Negative Breast Neoplasms/drug therapy
3.
Medicine (Baltimore) ; 96(22): e7076, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28562575

ABSTRACT

The purpose of this study was to evaluate the predictability and stability of laser-assisted subepithelial keratectomy (LASEK) with mitomycin C (MMC) in correction of high myopia (≤-6.0 diopters [D]) as compared to low-to-moderate myopia (>-6.0 D).This is a retrospective, comparative, cohort study which included 43 eyes of 43 consecutive patients who underwent LASEK with MMC in a private hospital in Hong Kong by a single surgeon. Twenty-five eyes had high myopia (mean spherical equivalent [SE] = -8.53 ±â€Š1.82 D) and 18 eyes had low-to-moderate myopia (mean SE = -3.99 ±â€Š1.37 D) before surgery.In terms of refractive predictability, mean SE was significantly better in eyes with preoperative low-to-moderate myopia than high myopia at 6 months (0.04 ±â€Š0.23 vs 0.31 ±â€Š0.52 D, P = .035). In terms of refractive stability, between 1 and 3 months, both groups had mean absolute change of SE of around 0.25 D. Between 3 and 6 months, preoperative low-to-moderate myopia group had significantly less absolute change of SE compared to high myopia group (0.07 vs 0.23 D, P = .003). More eyes with preoperative high myopia changed SE by more than 0.25 D than those with low-to-moderate myopia between 3 and 6 months (32.0% vs 5.6%, P = .057).In conclusion, LASEK with MMC is more unpredictable and unstable in correction of high myopia than low-to-moderate myopia. The refractive outcome of most low-to-moderate myopia correction stabilizes at 3 months. Stability is not achieved until after 6 months in high myopia correction.


Subject(s)
Keratectomy, Subepithelial, Laser-Assisted , Mitomycin/therapeutic use , Myopia/drug therapy , Myopia/surgery , Nucleic Acid Synthesis Inhibitors/therapeutic use , Adult , Combined Modality Therapy , Female , Hong Kong , Humans , Keratectomy, Subepithelial, Laser-Assisted/adverse effects , Male , Middle Aged , Mitomycin/adverse effects , Nucleic Acid Synthesis Inhibitors/adverse effects , Refraction, Ocular , Retrospective Studies , Severity of Illness Index , Time Factors , Treatment Outcome , Young Adult
4.
Biomed Pharmacother ; 91: 899-905, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28501777

ABSTRACT

Reactive astrocyte proliferation after spinal cord injury (SCI) contributes to glial scar formation that impedes axonal regeneration. The mechanisms underlying astrocyte proliferation upon injury remain partially understood. MicroRNAs (miRNAs) function as a major class of post-transcriptional gene expression regulators that participate in many biological processes. In this study, we focused on the functional role of miR-140 in normal human astrocyte (NHA) cell proliferation. Ectopic miR-140 expression significantly inhibited NHA cell viability and proliferation; miR-140 inhibition exerted the opposite function. Commonly, miRNAs exert functions through targeting downstream genes to inhibit their expression. In the present study, brain-derived neurotrophic factor (BDNF), a regulator of astrocyte proliferation and differentiation, confirmed as a direct target of miR-140 in NHA. Through binding to the 3'UTR of BDNF, miR-140 inhibited BDNF expression. BDNF overexpression significantly promoted NHA cell viability and proliferation; the regulatory effect of miR-140/BDNF on NHA proliferation was mediated by PI3K/AKT pathway. Moreover, we evaluated the functional role of miR-140 in Lipopolysaccharide (LPS)-induced in vitro injury model of astroglial cultures; a significantly up-regulated BDNF, interleukin (IL)-6 and tumor necrosis factor (TNF)-α expression in response to LPS stimulation was observed. After ectopic miR-140 expression, the promotive effect of LPS on BDNF, IL-6 and TGF-α expression was partially restored. Taken together, miR-140/BDNF axis regulates NHA proliferation through PI3K/AKT pathway; miR-140 could inhibit BDNF, IL-6 and TGF-α expression in LPS-induced in vitro injury model. MiR-140/BDNF might serve as a promising target in strategy against reactive astrocyte proliferation after SCI.


Subject(s)
Astrocytes/cytology , Astrocytes/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Interleukin-6/metabolism , Lipopolysaccharides/pharmacology , MicroRNAs/metabolism , Tumor Necrosis Factor-alpha/metabolism , 3' Untranslated Regions/genetics , Base Sequence , Cell Proliferation/drug effects , Humans , Inflammation/pathology , Phosphatidylinositol 3-Kinases/metabolism , Protein Binding/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects
5.
Cell Rep ; 18(8): 1946-1957, 2017 02 21.
Article in English | MEDLINE | ID: mdl-28228260

ABSTRACT

Breast cancer stem cells (BCSCs) play a critical role in tumor recurrence and metastasis. Exposure of breast cancer cells to chemotherapy leads to an enrichment of BCSCs. Here, we find that chemotherapy induces the expression of glutathione S-transferase omega 1 (GSTO1), which is dependent on hypoxia-inducible factor 1 (HIF-1) and HIF-2. Knockdown of GSTO1 expression abrogates carboplatin-induced BCSC enrichment, decreases tumor initiation and metastatic capacity, and delays tumor recurrence after chemotherapy. GSTO1 interacts with the ryanodine receptor RYR1 and promotes calcium release from the endoplasmic reticulum. Increased cytosolic calcium levels activate PYK2 → SRC → STAT3 signaling, leading to increased expression of pluripotency factors and BCSC enrichment. HIF inhibition blocks chemotherapy-induced GSTO1 expression and BCSC enrichment. Combining HIF inhibitors with chemotherapy may improve clinical outcome in breast cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Calcium/metabolism , Neoplastic Stem Cells/metabolism , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Line, Tumor , Cell Transformation, Neoplastic/metabolism , Cytosol/metabolism , Female , Glutathione Transferase/metabolism , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , MCF-7 Cells , Neoplastic Stem Cells/physiology , Ryanodine Receptor Calcium Release Channel/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects
6.
Oncotarget ; 7(40): 64527-64542, 2016 Oct 04.
Article in English | MEDLINE | ID: mdl-27590511

ABSTRACT

Exposure of breast cancer cells to hypoxia increases the percentage of breast cancer stem cells (BCSCs), which are required for tumor initiation and metastasis, and this response is dependent on the activity of hypoxia-inducible factors (HIFs). We previously reported that exposure of breast cancer cells to hypoxia induces the ALKBH5-mediated demethylation of N6-methyladenosine (m6A) in NANOG mRNA leading to increased expression of NANOG, which is a pluripotency factor that promotes BCSC specification. Here we report that exposure of breast cancer cells to hypoxia also induces ZNF217-dependent inhibition of m6A methylation of mRNAs encoding NANOG and KLF4, which is another pluripotency factor that mediates BCSC specification. Although hypoxia induced the BCSC phenotype in all breast-cancer cell lines analyzed, it did so through variable induction of pluripotency factors and ALKBH5 or ZNF217. However, in every breast cancer line, the hypoxic induction of pluripotency factor and ALKBH5 or ZNF217 expression was HIF-dependent. Immunohistochemistry revealed that expression of HIF-1α and ALKBH5 was concordant in all human breast cancer biopsies analyzed. ALKBH5 knockdown in MDA-MB-231 breast cancer cells significantly decreased metastasis from breast to lungs in immunodeficient mice. Thus, HIFs stimulate pluripotency factor expression and BCSC specification by negative regulation of RNA methylation.


Subject(s)
AlkB Homolog 5, RNA Demethylase/metabolism , Breast Neoplasms/metabolism , Neoplastic Stem Cells/physiology , RNA, Messenger/genetics , Trans-Activators/metabolism , Adenosine/analogs & derivatives , Adenosine/metabolism , Animals , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Hypoxia , Female , Gene Expression Regulation, Neoplastic , Humans , Hypoxia , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , MCF-7 Cells , Methylation , Mice , Mice, SCID , Nanog Homeobox Protein/genetics , Nanog Homeobox Protein/metabolism , RNA, Small Interfering/genetics , Xenograft Model Antitumor Assays
7.
Proc Natl Acad Sci U S A ; 113(14): E2047-56, 2016 Apr 05.
Article in English | MEDLINE | ID: mdl-27001847

ABSTRACT

N(6)-methyladenosine (m(6)A) modification of mRNA plays a role in regulating embryonic stem cell pluripotency. However, the physiological signals that determine the balance between methylation and demethylation have not been described, nor have studies addressed the role of m(6)A in cancer stem cells. We report that exposure of breast cancer cells to hypoxia stimulated hypoxia-inducible factor (HIF)-1α- and HIF-2α-dependent expression of AlkB homolog 5 (ALKBH5), an m(6)A demethylase, which demethylated NANOG mRNA, which encodes a pluripotency factor, at an m(6)A residue in the 3'-UTR. Increased NANOG mRNA and protein expression, and the breast cancer stem cell (BCSC) phenotype, were induced by hypoxia in an HIF- and ALKBH5-dependent manner. Insertion of the NANOG 3'-UTR into a luciferase reporter gene led to regulation of luciferase activity by O2, HIFs, and ALKBH5, which was lost upon mutation of the methylated residue. ALKBH5 overexpression decreased NANOG mRNA methylation, increased NANOG levels, and increased the percentage of BCSCs, phenocopying the effect of hypoxia. Knockdown of ALKBH5 expression in MDA-MB-231 human breast cancer cells significantly reduced their capacity for tumor initiation as a result of reduced numbers of BCSCs. Thus, HIF-dependent ALKBH5 expression mediates enrichment of BCSCs in the hypoxic tumor microenvironment.


Subject(s)
AlkB Homolog 5, RNA Demethylase/physiology , Basic Helix-Loop-Helix Transcription Factors/physiology , Breast Neoplasms/pathology , Cell Hypoxia , Nanog Homeobox Protein/genetics , Neoplastic Stem Cells/pathology , RNA, Messenger/metabolism , AlkB Homolog 5, RNA Demethylase/genetics , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Catalysis , Cell Line, Tumor , Female , Gene Knockdown Techniques , Humans , Methylation
8.
Oncotarget ; 7(6): 6379-97, 2016 Feb 09.
Article in English | MEDLINE | ID: mdl-26837221

ABSTRACT

Hypoxia-inducible factors (HIFs) control the transcription of genes that are crucial for the pathogenesis of cancer and other human diseases. The transcriptional activity of HIFs is rapidly increased upon exposure to hypoxia, but expression of some HIF target genes decreases during prolonged hypoxia. However, the underlying mechanism for feedback inhibition is not completely understood. Here, we report that peroxiredoxin 2 (PRDX2) and PRDX4 interact with HIF-1α and HIF-2α in vitro and in hypoxic HeLa cells. Prolonged hypoxia increases the nuclear translocation of PRDX2 and PRDX4. As a result, PRDX2 and PRDX4 impair HIF-1 and HIF-2 binding to the hypoxia response elements of a subset of HIF target genes, thereby inhibiting gene transcription in cells exposed to prolonged hypoxia. PRDX2 and PRDX4 have no effect on the recruitment of p300 and RNA polymerase II to HIF target genes and the enzymatic activity of PRDX2 and PRDX4 is not required for inhibition of HIF-1 and HIF-2. We also demonstrate that PRDX2 is a direct HIF target gene and that PRDX2 expression is induced by prolonged hypoxia. These findings uncover a novel feedback mechanism for inhibition of HIF transcriptional activity under conditions of prolonged hypoxia.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Embryo, Mammalian/metabolism , Fibroblasts/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia/physiopathology , Peroxiredoxins/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Blotting, Western , Cells, Cultured , Chromatin Immunoprecipitation , Embryo, Mammalian/pathology , Fibroblasts/pathology , Gene Expression Regulation , HEK293 Cells , HeLa Cells , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Immunoprecipitation , Luciferases/metabolism , Mice , Peroxiredoxins/genetics , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Transcriptional Activation
9.
Proc Natl Acad Sci U S A ; 112(33): E4600-9, 2015 Aug 18.
Article in English | MEDLINE | ID: mdl-26229077

ABSTRACT

Triple negative breast cancer (TNBC) accounts for 10-15% of all breast cancer but is responsible for a disproportionate share of morbidity and mortality because of its aggressive characteristics and lack of targeted therapies. Chemotherapy induces enrichment of breast cancer stem cells (BCSCs), which are responsible for tumor recurrence and metastasis. Here, we demonstrate that chemotherapy induces the expression of the cystine transporter xCT and the regulatory subunit of glutamate-cysteine ligase (GCLM) in a hypoxia-inducible factor (HIF)-1-dependent manner, leading to increased intracellular glutathione levels, which inhibit mitogen-activated protein kinase kinase (MEK) activity through copper chelation. Loss of MEK-ERK signaling causes FoxO3 nuclear translocation and transcriptional activation of the gene encoding the pluripotency factor Nanog, which is required for enrichment of BCSCs. Inhibition of xCT, GCLM, FoxO3, or Nanog blocks chemotherapy-induced enrichment of BCSCs and impairs tumor initiation. These results suggest that, in combination with chemotherapy, targeting BCSCs by inhibiting HIF-1-regulated glutathione synthesis may improve outcome in TNBC.


Subject(s)
Antineoplastic Agents/chemistry , Copper/chemistry , Glutathione/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Neoplastic Stem Cells/cytology , Triple Negative Breast Neoplasms/metabolism , Animals , Cell Line, Tumor , Chelating Agents/chemistry , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , MAP Kinase Kinase Kinases/metabolism , MAP Kinase Signaling System , Mice , Mice, SCID , Neoplasm Transplantation , Oligonucleotides/genetics , Paclitaxel/chemistry , Phenotype , Phosphorylation , RNA, Messenger/metabolism
10.
J Cataract Refract Surg ; 40(1): 29-36, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24269139

ABSTRACT

PURPOSE: To report the early experience and complications during cataract surgery with a noncontact femtosecond laser system. SETTING: Hong Kong Sanatorium and Hospital, Hong Kong Special Administrative Region, China. DESIGN: Retrospective case series. METHODS: All patients had anterior capsulotomy or combined anterior capsulotomy and lens fragmentation using a noncontact femtosecond laser system (Lensar) before phacoemulsification. Chart and video reviews were performed retrospectively to determine the intraoperative complication rate. Risk factors associated with the complications were also analyzed. RESULTS: One hundred seventy eyes were included. Free-floating capsule buttons were found in 151 eyes (88.8%). No suction break occurred in any case. Radial anterior capsule tears occurred in 9 eyes (5.3%); they did not extend to the equator or posterior capsule. One eye (0.6%) had a posterior capsule tear. No capsular block syndrome developed, and no nuclei were dropped during irrigation/aspiration (I/A). Anterior capsule tags and miosis occurred in 4 eyes (2.4%) and 17 eyes (10.0%), respectively. Different severities of subconjunctival hemorrhages developed in 71 (43.8%) of 162 eyes after the laser procedure. The mean surgical time from the beginning to the end of suction was 6.72 minutes ± 4.57 (SD) (range 2 to 28 minutes). CONCLUSIONS: Cataract surgery with the noncontact femtosecond laser system was safe. No eye lost vision because of complications. Caution should be taken during phacoemulsification and I/A to avoid radial anterior capsule tears and posterior capsule tears.


Subject(s)
Cataract Extraction/methods , Intraoperative Complications , Laser Therapy/methods , Adult , Aged , Aged, 80 and over , Anterior Capsule of the Lens/surgery , Female , Humans , Lens Implantation, Intraocular , Male , Middle Aged , Refraction, Ocular/physiology , Retrospective Studies , Risk Factors , Visual Acuity/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...