Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
J Mol Histol ; 50(3): 239-251, 2019 Jun.
Article in English | MEDLINE | ID: mdl-31049798

ABSTRACT

Reduced expression of endothelial nitric oxide synthase (eNOS) is a hallmark of endothelial dysfunction in diabetes, which predisposes diabetic patients to numerous cardiovascular complications including blunted angiogenesis. The Krüppel-like factor (KLF) five has been implicated as a central regulator of cardiovascular remodeling, but its role in endothelial cells (ECs) remains poorly understood. We show here that expression of endothelial KLF5 was significantly increased in the ECs from mouse diabetes mellitus type 2 (T2DM) model, when compared to non-diabetic or T1DM mouse. KLF5 up-regulation by insulin was dependent on activation of multiple pathways, including mammalian target of rapamycin, oxidative stress and Protein kinase C pathways. Hyperinsulinemia-induced KLF5 inhibited endothelial function and migration, and thereby compromised in vitro and in vivo angiogenesis. Mechanistically, KLF5 acted in concert with the MTA1 coregulator to negatively regulate NOS3 transcription, thereby leading to the diminished eNOS levels in ECs. Conversely, potentiation of cGMP content (the essential downstream effector of eNOS signaling) by pharmacological approaches successfully rescued the endothelial proliferation and in vitro tube formation, in the HUVECs overexpressing the exogenous KLF5. Collectively, the available data suggest that the augmentation of endothelial KLF5 expression by hyperinsulinemia may represent a novel mechanism for negatively regulating eNOS expression, and may thus help to explain for the T2DM-related endothelial dysfunction at the transcriptional level.


Subject(s)
Hyperinsulinism/genetics , Kruppel-Like Transcription Factors/genetics , Neovascularization, Pathologic/genetics , Nitric Oxide Synthase Type III/genetics , Animals , Cell Movement/genetics , Cell Proliferation/genetics , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/pathology , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/pathology , Endothelial Cells/metabolism , Endothelial Cells/pathology , Gene Expression/genetics , Human Umbilical Vein Endothelial Cells , Humans , Hyperinsulinism/pathology , Male , Mice , Oxidative Stress/genetics , Protein Kinase C/genetics , Signal Transduction/genetics
2.
Cancer Chemother Pharmacol ; 84(2): 427-439, 2019 08.
Article in English | MEDLINE | ID: mdl-31087138

ABSTRACT

OBJECTIVE: Although DNA-mismatch-repair-deficient (dMMR) status and aberrant expression of miRNAs are both critically implicated in the pathogenesis of resistance to 5-fluorouracil (5-FU) in colorectal cancer (CRC), whether these two factors regulate tumor response to 5-FU in a coordinated manner remains unknown. This study is designed to elucidate whether changes in miR-552 expression levels correlate to 5-FU-based chemoresistance in CRC, and to further identify the putative targets of miR-552 using multiple approaches. METHODS: miR-552 expression was assessed in 5-FU-resistant CRC tissues and cells using real-time PCR. Effects of miR-552 dysregulation on 5-FU resistance in CRC cells were determined by measuring cell viability, apoptosis and in vivo oncogenic capacity. Finally, we studied the posttranscriptional regulation of SMAD2 by miR-552 using multiple approaches including luciferase reporter assay, site-directed mutagenesis and transient/stable transfection, at molecular and functional levels. RESULTS: Expression of miR-552 was significantly downregulated in 5-FU-resistant CRC tissues and cells, and this downregulation, regulated by dMMR, was associated with poor postchemotherapy prognosis. Functionally, forced expression of miR-552 exhibited a proapoptotic effect and attenuated 5-FU resistance, whereas inhibition of miR-552 expression potentiated 5-FU resistance in CRC cells. Mechanically, miR-552 directly targeted the 3'-UTR of SMAD2, and stable ablation of SMAD2 neutralized the promoting effects of miR-552 deficiency-induced 5-FU resistance. CONCLUSIONS: Overall, our findings have revealed a critical role of miR-552/SMAD2 cascade in modulating cellular response to 5-FU chemotherapy. miR-552 may act as an efficient mechanistic link synchronizing dMMR and 5-FU resistance in CRC.


Subject(s)
Colorectal Neoplasms/drug therapy , DNA Mismatch Repair/genetics , MicroRNAs/metabolism , Smad2 Protein/genetics , Animals , Colorectal Neoplasms/genetics , Disease Models, Animal , Fluorouracil/pharmacology , Humans , Mice , Signal Transduction
3.
Cancer Chemother Pharmacol ; 83(4): 625-637, 2019 04.
Article in English | MEDLINE | ID: mdl-30631898

ABSTRACT

OBJECTIVE: Compelling evidence establishes the etiological role of viral proteins E6 and E7 of high-risk human papillomaviruses (HPV) in cervical carcinogenesis, but their contribution in chemoresistance that leads to advanced metastatic lesions remains poorly defined. Since metastasis-associated protein 1 (MTA1) upregulation and augmentation of APOBEC3B expression are both strongly associated with cervical cancer (CCa) development, and both molecules have been shown to be functionally associated with NF-κB pathway, we therefore sought to investigate the potential mechanistic link between MTA1, APOBEC3B and NF-κB during the pathogenesis of cisplatin (CDDP) resistance in HPV-positive CCa cells. METHODS: MTA1 expression was assessed in HPV-transfected CCa cells using quantitative RT-PCR and immunoblotting. Effects of MTA1 deregulation on CDDP chemosensitivity in CCa cells were determined by measuring cell viability, apoptosis and in vivo oncogenic capacity. Finally, we studied the transcriptional regulation of the antiviral DNA cytosine deaminase APOBEC3B by MTA1 using multiple approaches including DNA deaminase activity assay, luciferase reporter assay, chromatin immunoprecipitation, co-immunoprecipitation and transient/stable transfection, at the molecular and functional levels. RESULTS: Expression levels of MTA1 were significantly induced in HPV-positive CCa cells. Transduction experiments showed that the E6 oncoprotein alone was sufficient to cause MTA1 upregulation. Moreover, MTA1 knockdown potentiated CDDP sensitivity in highly metastatic CCa cells. Mechanistically, MTA1 acted as an indirect upstream modulator of APOBEC3B transcription during the pathogenesis of CDDP chemoresistance. HPV-mediated stimulation of APOBEC3B expression was accompanied by the enhanced recruitment of Iκκ α/ß and p65 to the NF-κB consensus sites in the APOBEC3B promoter, and this recruitment was substantially abrogated by MTA1 siRNA treatment. CONCLUSIONS: These findings reveal an obligatory coregulatory role of MTA1 in the indirect regulation of APOBEC3B expression via classical NF-κB pathway, and also suggest that inhibition of MTA1/NF-κB/APOBEC3B cascade may be repositioned to suppress cancer mutagenesis, dampen tumor evolution, and decrease the probability of adverse outcomes from CDDP resistance in CCa.


Subject(s)
Antineoplastic Agents/pharmacology , Cisplatin/pharmacology , Papillomavirus Infections/complications , Repressor Proteins/genetics , Trans-Activators/genetics , Uterine Cervical Neoplasms/drug therapy , Animals , Cytidine Deaminase/metabolism , DNA-Binding Proteins/metabolism , Female , Humans , Mice, Inbred BALB C , Mice, Nude , Minor Histocompatibility Antigens/metabolism , NF-kappa B/metabolism , Oncogene Proteins, Viral/metabolism , Up-Regulation , Uterine Cervical Neoplasms/virology , Xenograft Model Antitumor Assays
4.
Cancer Chemother Pharmacol ; 82(4): 593-605, 2018 10.
Article in English | MEDLINE | ID: mdl-30032449

ABSTRACT

OBJECTIVE: The mainstay of treatment for advanced bladder cancer (BC) is cisplatin (CDDP)-based systematic chemotherapy. However, acquired chemoresistance induced by as yet unidentified mechanisms is encountered frequently and often results in treatment failure and disease progression. The present study was designed to elucidate the expression and potential role of the gene associated with retinoid-interferon-induced mortality-19 (GRIM19) in the pathogenesis of CDDP resistance in BC. METHODS: RT-qPCR and immunoblotting were employed to evaluate the expression profile of GRIM19 in clinical BC samples and in different BC cells. Using cell viability assay, apoptotic ELISA, xenografts mouse model, and Transwell assay, the effects of GRIM19 inhibition or GRIM19 overexpression on CDDP resistance were determined in different BC cells. Lastly, using co-immunoprecipitation, we provided the molecular evidence for the interaction between GRIM19 and Bcl-xL. RESULTS: Expression levels of GRIM19 were significantly down-regulated in recurrent BC specimens, and in experimentally induced CDDP-resistant BC cells. Functionally, overexpression of the exogenous GRIM19 potentiated CDDP sensitivity and suppressed the survival and invasion of BC cells in the presence of CDDP challenge. Mechanistically, the compromised CDDP chemosensitization induced by GRIM19 loss was at least partially attributed to the attenuation of Bcl-xL polyubiquitination and subsequent degradation, because (1) GRIM19 colocalized with Bcl-xL in the mitochondria of BC cells and (2) GRIM19 overexpression promoted the ubiquitination of Bcl-xL, and this event could be effectively reversed by pretreatment with inhibitors of p38-MAPK and JNK pathways, indicating that GRIM19 overexpression-induced Bcl-xL ubiquitination may achieve in a p38/JNK-dependent manner. Using the UMUC-3 cells stably depleted of endogenous GRIM19, we further show that inhibition of Bcl-xL rectified GRIM19 deficiency-caused CDDP resistance in BC cells. In addition, BCL2L1 mRNA levels were negatively correlated with GRIM19 mRNA levels in CDDP-associated clinical BC tissues. CONCLUSIONS: Disruption of GRIM19/Bcl-xL is a key mechanism of CDDP resistance in advanced BC. Therapeutically, enhancement of GRIM19 expression or employment of p38/JNK inhibitors may serve as resensitizing therapies for subgroups of CDDP-resistant or refractory BC patients.


Subject(s)
Cisplatin/pharmacology , Drug Resistance, Neoplasm , NADH, NADPH Oxidoreductases , Urinary Bladder Neoplasms , bcl-X Protein/metabolism , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Apoptosis Regulatory Proteins/metabolism , Cell Line, Tumor , Down-Regulation/drug effects , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Gene Expression Profiling , Humans , Mice , NADH, NADPH Oxidoreductases/genetics , NADH, NADPH Oxidoreductases/metabolism , Neoplasm Recurrence, Local , Neoplasm Staging , Protein Subunits , Signal Transduction/drug effects , Ubiquitination/drug effects , Urinary Bladder Neoplasms/drug therapy , Urinary Bladder Neoplasms/pathology , Xenograft Model Antitumor Assays
5.
Biochem Biophys Res Commun ; 503(4): 2293-2300, 2018 09 18.
Article in English | MEDLINE | ID: mdl-29964012

ABSTRACT

Cisplatin (CDDP)-based systematic chemotherapy remains the mainstay of treatment for muscle-invasive bladder cancer (MIBC). However, acquired resistance to CDDP, a multifactorial process governed by an array of signals acting at different levels, is the major problem in BC treatment. Here, we report for the first time that, expression of Paired-box gene 5 (PAX5), a B-cell essential transcription factor, was significantly induced in CDDP-resistant BC tissues and in experimentally-induced CDDP-resistant BC cells. Inhibition of PAX5 expression by shRNA treatment effectively improved CDDP sensitivity in BC cells, whereas overexpression of PAX5 potentiated CDDP resistance through supporting BC cell survival. Mechanistically, using luciferase reporter and chromatin immunoprecipitation assays, we identified prostaglandin-endoperoxide synthase 2 (PTGS2, also called COX2), a potent enzyme responsible for prostanoids formation and inflammatory response, as the direct down-stream target of PAX5. PAX5 exerted its oncogenic function during the pathogenesis of CDDP resistance via stimulation of PTGS2 transcription. These observations collectively suggest that dysregulation of PAX5/PTGS2 cascade plays a causal role in the induction of CDDP resistance and gene silencing approaches targeting this pathway may therefore provide a novel therapeutic strategy for overcoming CDDP resistance in BC.


Subject(s)
Cisplatin/pharmacology , Cyclooxygenase 2/genetics , Drug Resistance, Neoplasm/genetics , PAX5 Transcription Factor/genetics , Transcriptional Activation , Urinary Bladder Neoplasms/pathology , Humans , Muscle Neoplasms/secondary , Neoplasm Invasiveness , Signal Transduction , Tumor Cells, Cultured
6.
Biomed Pharmacother ; 99: 859-866, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29710485

ABSTRACT

Type 2 diabetes mellitus (T2DM) promotes a high oxidative stress and hypercoagulable state that drives microvascular injury and multiple-organ abnormality. Elevated thrombin activity underlies T2DM-linked endothelial dysfunction, but the mechanistic links between T2DM/oxidative stress axis and thrombin-associated endothelial pathologies are incompletely understood. In this work, immunohistochemical studies and quantitative analysis using isolated endothelial cells (ECs) identified accumulated Kru¨ppel-like family of transcription factor 14 (KLF14) deposits in ECs from multiple organs as distinct features of T2DM mice. KLF14 upregulation in ECs, which was stimulated by thrombin treatment, was dependent on multiple pathways including calcium mobilization, activation of PKC and AMPK pathways. Functionally, inhibition of endogenous KLF14 expression significantly attenuated thrombin-induced endotheliocyte proliferation, endothelial cell migration and oxidative stress. Molecularly, by directly binding the promoter, KLF14 functions as a transcriptional activator of PLK1, a polo-like kinase whose overexpression induced excessive reactive oxygen species (ROS) production. Transient knockdown of PLK1 was sufficient to suppress KLF14 overexpression-potentiated endothelial dysfunction. Collectively, these data provide proof of concept that deregulation of KLF14/PLK1 cascade plays a key role in thrombin-induced endothelial dysfunction and targeting KLF14 or PLK1 may limit thrombin-associated pathologies in T2DM patients.


Subject(s)
Cell Cycle Proteins/genetics , Diabetes Mellitus, Experimental/physiopathology , Diabetes Mellitus, Type 2/physiopathology , Kruppel-Like Transcription Factors/genetics , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins/genetics , Animals , Cell Movement/genetics , Cell Proliferation/genetics , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Type 2/genetics , Endothelial Cells/pathology , Humans , Male , Mice , Mice, Inbred BALB C , Oxidative Stress , Reactive Oxygen Species/metabolism , Signal Transduction , Thrombin/metabolism , Polo-Like Kinase 1
7.
Biochem Biophys Res Commun ; 493(1): 847-854, 2017 11 04.
Article in English | MEDLINE | ID: mdl-28709873

ABSTRACT

Mechanistic insight into estrogen deficiency by polycystic ovary syndrome (PCOS) remains a longstanding challenge in reproductive medicine. Recent advance suggest that Wingless-type MMTV integration site family members (WNTs), in concert with its Frizzled (FZD) receptors, regulate normal folliculogenesis, luteogenesis and ovarian steroidogenesis. However, no studies have so far investigated any causality between WNT-FZDs interactions and disrupted estrogen synthesis under certain pathological conditions. Here, we show that (i) FZD3 expression was significantly up-regulated in the cumulus cells (CCs) from PCOS patients. This up-regulation, along with the activation of WNT2/ß-Catenin pathway, was tightly associated with insulin resistance and estrogen deficiency, two hallmarks of PCOS. (ii) Overexpression of exogenous FZD3 in human granulosa cell COV434 impaired long-term FSH incubation-induced CYP19A1 transactivation and the recruitment of ß-Catenin onto CYP19A1 promoter, and subsequently compromised FSH-stimulated estrogen production. (iii) Conversely, inhibition of FZD3 expression exhibited a therapeutic effect on estrogen synthesis in PCOS CCs. Thus, excessive FZD3 expression in CCs may act as a brake on steroidogenic activation that is normally overcome by FSH stimulation. Future endeavor in this field should help to elucidate the complicated crosstalk between energy metabolism and endocrine cells through WNT/FZD signaling molecules.


Subject(s)
Cumulus Cells/metabolism , Estrogens/biosynthesis , Frizzled Receptors/metabolism , Polycystic Ovary Syndrome/metabolism , Wnt Signaling Pathway , Wnt2 Protein/metabolism , beta Catenin/metabolism , Cells, Cultured , Cumulus Cells/pathology , Down-Regulation , Female , Humans , Polycystic Ovary Syndrome/pathology
8.
Biochem Biophys Res Commun ; 491(2): 545-551, 2017 09 16.
Article in English | MEDLINE | ID: mdl-28351619

ABSTRACT

Despite numerous epidemiological data linking type 2 diabetes mellitus (T2DM) and breast cancer (BCa), there is limited experimental evidence of this association. The clinically relevant question is at what stage diabetes may exert its tumor-promoting activity. Moreover, identification of major pathophysiological pathways underlying this activity should provide valuable information for treatment. In the present study, the BCa cells isolated from long-term T2DM-treated tumors from diabetic nude mice were found to have increased cell proliferation, invasiveness and docetaxel (DTX) resistance. Importantly, this stimulatory effect was only observable in estrogen receptor (ER)-positive BCa cells. Mechanistically, T2DM-elicited hyperinsulinemia induced HIF-1α expression by reducing its ubiquitination, which was accompanied with upregulated oxidative stress. Furthermore, in vivo inhibition of HIF-1α expression effectively reversed the above-mentioned tumor-promoting activity and partially attenuated T2DM-elicited oxidative stress. Altogether, the results provide novel and compelling experimental evidence that (i) prolonged exposure to T2DM promotes BCa progression; (ii) the hyperinsulinemia/HIF-1α/oxidative stress cascade is the major mediator of this effect.


Subject(s)
Breast Neoplasms/genetics , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Type 2/genetics , Gene Expression Regulation, Neoplastic , Hyperinsulinism/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Animals , Antineoplastic Agents/pharmacology , Breast Neoplasms/complications , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Docetaxel , Drug Resistance, Neoplasm/genetics , Female , Humans , Hyperinsulinism/complications , Hyperinsulinism/metabolism , Hyperinsulinism/pathology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Insulin/metabolism , Insulin Resistance , MCF-7 Cells , Mice , Mice, Inbred BALB C , Mice, Nude , Reactive Oxygen Species/metabolism , Receptors, Estrogen/genetics , Receptors, Estrogen/metabolism , Signal Transduction , Taxoids/pharmacology , Ubiquitination , Xenograft Model Antitumor Assays
9.
FEBS Lett ; 591(6): 914-923, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28117896

ABSTRACT

Members of the Drosophila behavior/human splicing protein family, including splicing factor proline/glutamine rich (SFPQ), non-POU domain-containing octamer-binding protein (NONO), and paraspeckle protein component 1 (PSPC1), are abundantly expressed in testicular Sertoli cells (SCs), but their roles remain obscure. Here, we show that treatment with mono-(2-ethylhexyl) phthalate (MEHP), a well-known SC toxicant, selectively stimulates the expression levels of NONO and PSPC1. Simultaneous inhibition of NONO and PSPC1 expression in SCs enhances MEHP-induced oxidative stress and potentiates SC death. Mechanistically, NONO and PSPC1 transcriptionally activate aldehyde dehydrogenase 1 (Aldh1a1), by synergistically binding to the distinct CCGGAGTC sequence in the Aldh1a1 promoter. Together, the NONO/PSPC1-ALDH1A1 cascade may serve as an indispensable defense mechanism against MEHP insult in SCs.


Subject(s)
Aldehyde Dehydrogenase/metabolism , DNA-Binding Proteins/metabolism , Diethylhexyl Phthalate/analogs & derivatives , Nuclear Proteins/metabolism , RNA-Binding Proteins/metabolism , Sertoli Cells/metabolism , Aldehyde Dehydrogenase/genetics , Animals , Base Sequence , Binding Sites/genetics , Cell Line , DNA-Binding Proteins/genetics , Diethylhexyl Phthalate/pharmacology , Humans , Immunoblotting , Immunohistochemistry , Male , Mice, Inbred C57BL , Promoter Regions, Genetic/genetics , Protein Binding , Reverse Transcriptase Polymerase Chain Reaction , Sertoli Cells/drug effects , Signal Transduction/drug effects , Transcriptional Activation/drug effects , Up-Regulation
10.
Urology ; 105: 208.e11-208.e17, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28088556

ABSTRACT

OBJECTIVE: To identify the potential downstream targets of hsa-miR-125a-3p, a mature form of miR-125a, during the pathogenesis of chemoresistance in prostate cancer (PCa). MATERIALS AND METHODS: The expression levels of hsa-miR-125a-3p were assessed in chemoresistant PCa tissues and experimentally established chemoresistant cells using quantitative reverse transcription-polymerase chain reaction. The effect of hsa-miR-125a-3p knockdown or hsa-miR-125a-3p overexpression on the Dox-induced cell death was evaluated using apoptosis ELISA in chemosensitive PC-3 cells or in chemoresistant PC-3 cells (PC-3R). Finally, using multiple assays, the regulation of metastasis-associated protein 1 (MTA1), an essential component of the Mi-2-nucleosome remodeling deacetylation complex, by hsa-miR-125a-3p was studied at both molecular and functional levels. RESULTS: The expression of hsa-miR-125a-3p was significantly downregulated in chemoresistant PCa tissues and cells. Inhibition of hsa-miR-125a-3p significantly increased docetaxel (Dox) resistance in PC-3 cells, whereas upregulation of hsa-miR-125a-3p effectively reduced Dox resistance in PC-3R, suggesting that this microRNA (miRNA) may act as a tumor suppressor along the pathogenesis of drug resistance. Mechanistically, hsa-miR-125a-3p induced apoptosis and Dox sensitivity in PCa cells through regulating MTA1. CONCLUSION: Our results collectively indicate that miRNA-MTA1 can form a delicate regulatory loop to maintain a bistable state in the Dox chemosensitivity, and future endeavor in this filed should provide important clues to develop miRNA-based therapies that benefit advanced PCa patients through modulating the functional status of MTA1.


Subject(s)
Antineoplastic Agents/pharmacology , Docetaxel/pharmacology , Histone Deacetylases/metabolism , MicroRNAs/metabolism , Prostatic Neoplasms/metabolism , Repressor Proteins/metabolism , Cell Culture Techniques , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Humans , Male , Prostatic Neoplasms/pathology , Trans-Activators
11.
Biochem Biophys Res Commun ; 482(4): 1469-1476, 2017 Jan 22.
Article in English | MEDLINE | ID: mdl-27965096

ABSTRACT

Deregulation of epigenetic modification by microRNAs (miRNAs) contributes to the development of estrogen deficiency, a hallmark of the multigenic endocrine disorder polycystic ovary syndrome (PCOS), but its etiology remains unclear. Previous study has pointed to a tight association between miR-320a expression and oocyte development in human follicular fluid. Given that the bi-directional communication existing between cumulus cells (CCs) and follicular fluid is essential for ovarian steroidogenesis and CCs are the main site where estrogen is finally synthesized, it is intriguing to know whether miR-320a have any regulatory roles in this unique cell. Here we report that miR-320a expression is significantly down-regulated in primary CCs from PCOS patients and this down-regulation promotes estrogen deficiency in CCs. From a mechanistic standpoint, IGF1 regulates miR-320a expression in CCs, and miR-320a could potentiate the steroidogenesis in CCs through modulation of CYP11A1 and CYP19A1 expression, by directly targeting 3'untranslated region (3'UTR) of the osteogenic transcription factor RUNX2. Overall, our results strongly suggest that deregulation of miR-320a/RUNX2/CYP11A1 (CYP19A1) cascade plays an important role in the development of estrogen deficiency in human CCs. Testing patients for miR-320a/RUNX2 expression ratios may provide more accurate diagnostic information and could influence the recommended course of treatment for PCOS.


Subject(s)
Core Binding Factor Alpha 1 Subunit/metabolism , Cumulus Cells/metabolism , Gene Expression Regulation , Granulosa Cells/metabolism , MicroRNAs/metabolism , Polycystic Ovary Syndrome/metabolism , 3' Untranslated Regions , Adult , Aromatase/metabolism , Case-Control Studies , Cholesterol Side-Chain Cleavage Enzyme/metabolism , Down-Regulation , Estradiol/chemistry , Estrogens/deficiency , Female , Fertilization in Vitro , Gene Expression Profiling , Humans , Oocytes/cytology , Progesterone/chemistry , RNA, Small Interfering/metabolism , Sperm Injections, Intracytoplasmic
12.
Biochem Biophys Res Commun ; 480(4): 682-689, 2016 11 25.
Article in English | MEDLINE | ID: mdl-27983982

ABSTRACT

The regulatory transcriptional factor PATZ1 is abnormally up-regulated in diabetic endothelial cells (ECs) where it acts as an anti-angiogenic factor via modulation of fatty acid-binding protein 4 (FABP4) signaling. The aim of the present work was to elucidate the upstream molecular events regulating PATZ1 expression in diabetic angiogenesis. The bioinformatics search for microRNAs (miRNAs) able to potentially target PATZ1 led to the identification of several miRNAs. Among them we focused on the miR-24 since the multiple targets of miR-24, which have so far been identified in beta cells, cardiomyocytes and macrophages, are all involved in diabetic complications. miR-24 expression was significantly impaired in the ECs isolated from diabetic hearts. Functionally, endothelial migration was profoundly inhibited by miR-24 suppression in Ctrl ECs, whereas miR-24 overexpression by mimics treatment effectively restored the migration rate in diabetic ECs. Mechanistically, miR-24 directly targeted the 3'untranslated region (3'UTR) of PATZ1, and miR-24 accumulation potentiated endothelial migration by reducing the mRNA stability of PATZ1. Together, these results suggest a novel mechanism regulating endothelial PATZ1 expression based on the down-regulation of miR-24 expression caused by hyperglycemia. Interfering with PATZ1 expression via miRNAs or miRNA mimics could potentially represent a new way to target endothelial PATZ1-dependent signaling of vascular dysfunction in diabetes.


Subject(s)
BTB-POZ Domain , Diabetic Angiopathies/metabolism , Diabetic Angiopathies/pathology , Endothelial Cells/metabolism , MicroRNAs/metabolism , Neoplasm Proteins/metabolism , Repressor Proteins/metabolism , Animals , Cells, Cultured , Diabetic Angiopathies/prevention & control , Endothelial Cells/pathology , Gene Silencing , Male , Mice , Mice, Inbred C57BL
13.
Biochem Biophys Res Commun ; 479(4): 893-900, 2016 Oct 28.
Article in English | MEDLINE | ID: mdl-27693788

ABSTRACT

Epigenetic gene inactivation by microRNAs (miRNAs) plays a key role in malignant transformation, prevention of apoptosis, drug resistance and metastasis. It has been shown that miR-125a is down-regulated in HER2-amplified and HER2-overexpressing breast cancers (BCa), and this miRNA is believed to serve as an important tumor suppressor. miR-125a has two mature forms: hsa-miR-125a-3p and hsa-miR-125a-5p. However, the functional details of these miRNAs in BCa, particularly during pathogenesis of drug resistance, remain largely unexplored. Herein, we reported that hsa-miR-125a-3p expression was significantly reduced in chemoresistant BCa tissues and in experimentally established chemoresistant BCa cells. hsa-miR-125a-3p knockdown promoted cell proliferation and compromised docetaxel (Dox)-induced cell death, whereas overexpression of hsa-miR-125a-3p attenuated Dox chemoresistance in BCa cells. From a mechanistic standpoint, hsa-miR-125a-3p directly targeted 3'-untranslated regions (3'-UTRs) of breast cancer early onset gene 1 (BRCA1) and inhibits its protein expression via translational repression mechanism. In addition, suppression of BRCA1 expression by siRNA treatment effectively improved hsa-miR-125a-3p deficiency-triggered chemoresistance in BCa cells. Collectively, these findings suggest that hsa-miR-125a-3p may function as a tumor suppressor by regulating the BRCA1 signaling, and reintroduction of hsa-miR-125a-3p analogs could be a potential adjunct therapy for advanced/chemoresistant BCa.


Subject(s)
Antineoplastic Agents/pharmacology , BRCA1 Protein/genetics , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , MicroRNAs/genetics , Taxoids/pharmacology , 3' Untranslated Regions , BRCA1 Protein/antagonists & inhibitors , BRCA1 Protein/metabolism , Base Sequence , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Proliferation , Docetaxel , Drug Resistance, Neoplasm/genetics , Female , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Genes, BRCA1 , Humans , MCF-7 Cells , MicroRNAs/antagonists & inhibitors , MicroRNAs/metabolism , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism , Signal Transduction
14.
Biochem Biophys Res Commun ; 479(1): 40-7, 2016 10 07.
Article in English | MEDLINE | ID: mdl-27603575

ABSTRACT

The Metastasis-associated protein 1 (MTA1) coregulator, an essential component of the nucleosome remodeling and deacetylase (NuRD) complex, potentiates neuroprotective effects against ischemia/reperfusion (I/R) injury. But the underlying mechanism(s) remain largely unknown. Here, we discovered that neuronal MTA1 was a target of oxidative stress, and stimulation of neurons with oxygen glucose deprivation (OGD) treatment significantly inhibited MTA1 expression. Additionally, MTA1 depletion augmented ischemic oxidative stress and thus promoted oxidative stress-induced neuronal cell death by OGD. While studying the impact of MTA1 status on global neuronal gene expression, we unexpectedly discovered that MTA1 may modulate OGD-induced neuronal damage via regulation of distinct nitric oxide synthase (NOS) (namely neuronal NOS, nNOS) signaling. We provided in vitro evidence that NOS1 is a chromatin target of MTA1 in OGD-insulted neurons. Mechanistically, neuronal ischemia-mediated repression of NOS1 expression is accompanied by the enhanced recruitment of MTA1 along with histone deacetylases (HDACs) to the NOS1 promoter, which could be effectively blocked by a pharmacological inhibitor of the HDACs. These findings collectively reveal a previously unrecognized, critical homeostatic role of MTA1, both as a target and as a component of the neuronal oxidative stress, in the regulation of acute neuronal responses against brain I/R damage. Our study also provides a molecular mechanistic explanation for the previously reported neurovascular protection by selective nNOS inhibitors.


Subject(s)
Histone Deacetylases/metabolism , Neurons/metabolism , Nitric Oxide Synthase Type I/metabolism , Oxidative Stress , Repressor Proteins/metabolism , Apoptosis/genetics , Blotting, Western , Cell Hypoxia , Cell Line, Tumor , Chromatin/genetics , Chromatin/metabolism , Gene Expression , Glucose/metabolism , Histone Deacetylases/genetics , Humans , Nitric Oxide/metabolism , Nitric Oxide Synthase Type I/genetics , Oxygen/metabolism , Promoter Regions, Genetic/genetics , RNA Interference , Reactive Oxygen Species/metabolism , Repressor Proteins/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Time Factors , Trans-Activators
15.
Biochem Biophys Res Commun ; 477(4): 548-555, 2016 09 02.
Article in English | MEDLINE | ID: mdl-27297106

ABSTRACT

Vascular endothelial dysfunction, a central hallmark of diabetes, predisposes diabetic patients to numerous cardiovascular complications. The POZ/BTB and AT-hook-containing zinc finger protein 1 (PATZ1), is an important transcriptional regulatory factor and regulates divergent pathways depending on the cellular context, but its role in endothelial cells remains poorly understood. Herein, we report for the first time that endothelial PATZ1 expression was abnormally upregulated in diabetic endothelial cells (ECs) regardless of diabetes classification. This stimulatory effect was further confirmed in the high glucose-treated human umbilical vein endothelial cells (HUVECs). From a functional standpoint, transgenic overexpression of PATZ1 in endothelial colony forming cells (ECFCs) blunted angiogenesis in vivo and rendered endothelial cells unresponsive to established angiogenic factors. Mechanistically, PATZ1 acted as a potent transcriptional corepressor of fatty acid-binding protein 4 (FABP4), an essential convergence point for angiogenic and metabolic signaling pathways in ECs. Taken together, endothelial PATZ1 thus potently inhibits endothelial function and angiogenesis via inhibition of FABP4 expression, and abnormal induction of endothelial PATZ1 may contribute to multiple aspects of vascular dysfunction in diabetes.


Subject(s)
Fatty Acid-Binding Proteins/metabolism , Hyperglycemia/metabolism , Neoplasm Proteins/metabolism , Repressor Proteins/metabolism , Signal Transduction , Animals , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Fatty Acid-Binding Proteins/genetics , Glucose/administration & dosage , Human Umbilical Vein Endothelial Cells , Humans , Male , Mice , Mice, Inbred C57BL
SELECTION OF CITATIONS
SEARCH DETAIL
...