Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
1.
Cell Death Dis ; 12(11): 1075, 2021 11 11.
Article in English | MEDLINE | ID: mdl-34764236

ABSTRACT

An early event in lung oncogenesis is loss of the tumour suppressor gene LIMD1 (LIM domains containing 1); this encodes a scaffold protein, which suppresses tumorigenesis via a number of different mechanisms. Approximately 45% of non-small cell lung cancers (NSCLC) are deficient in LIMD1, yet this subtype of NSCLC has been overlooked in preclinical and clinical investigations. Defining therapeutic targets in these LIMD1 loss-of-function patients is difficult due to a lack of 'druggable' targets, thus alternative approaches are required. To this end, we performed the first drug repurposing screen to identify compounds that confer synthetic lethality with LIMD1 loss in NSCLC cells. PF-477736 was shown to selectively target LIMD1-deficient cells in vitro through inhibition of multiple kinases, inducing cell death via apoptosis. Furthermore, PF-477736 was effective in treating LIMD1-/- tumours in subcutaneous xenograft models, with no significant effect in LIMD1+/+ cells. We have identified a novel drug tool with significant preclinical characterisation that serves as an excellent candidate to explore and define LIMD1-deficient cancers as a new therapeutic subgroup of critical unmet need.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Intracellular Signaling Peptides and Proteins/deficiency , LIM Domain Proteins/deficiency , Lung Neoplasms/genetics , Animals , Carcinoma, Non-Small-Cell Lung/pathology , Female , Humans , Lung Neoplasms/pathology , Mice , Mice, Inbred NOD , Proof of Concept Study , Transfection
2.
ERJ Open Res ; 7(1)2021 Jan.
Article in English | MEDLINE | ID: mdl-33532474

ABSTRACT

Genetic studies have identified several epithelial-derived genes associated with airway diseases. However, techniques used to study gene function frequently exceed the proliferative potential of primary human bronchial epithelial cells (HBECs) isolated from patients. Increased expression of the polycomb group protein BMI-1 extends the lifespan of HBECs while maintaining cell context plasticity. Herein we aimed to assess how BMI-1 expression impacted cellular functions and global mRNA expression. HBECs from six donors were transduced with lentivirus containing BMI-1 and cells were characterised, including by RNA sequencing and impedance measurement. BMI-1-expressing HBECs (B-HBECs) have a proliferative advantage and show comparable in vitro properties to low passage primary HBECs, including cell attachment/spreading and barrier formation. The B-HBEC mRNA signature was modestly different to HBECs, with only 293 genes differentially expressed (5% false discovery rate). Genes linked to epithelial mesenchymal transition and cell cycle were enriched in B-HBECs. We investigated the expression of genes implicated in asthma from genetic and expression studies and found that 97.6% of genes remained unaltered. We have shown that increased BMI-1 expression in HBECs delays lung epithelial cell senescence by promoting cell cycle progression and highlighted the flexible utility for B-HBECs as an important platform for studying airway epithelial mechanisms.

3.
EMBO Rep ; 20(4)2019 04.
Article in English | MEDLINE | ID: mdl-30833344

ABSTRACT

Determining the mechanisms that distinguish protective immunity from pathological chronic inflammation remains a fundamental challenge. miR-132 has been shown to play largely immunoregulatory roles in immunity; however, its role in CD4+ T cell function is poorly understood. Here, we show that CD4+ T cells express high levels of miR-132 and that T cell activation leads to miR-132 up-regulation. The transcriptomic hallmark of splenic CD4+ T cells lacking the miR-132/212 cluster during chronic infection is an increase in mRNA levels of ribosomal protein (RP) genes. BTAF1, a co-factor of B-TFIID and novel miR-132/212-3p target, and p300 contribute towards miR-132/212-mediated regulation of RP transcription. Following infection with Leishmania donovani, miR-132-/- CD4+ T cells display enhanced expression of IL-10 and decreased IFNγ. This is associated with reduced hepatosplenomegaly and enhanced pathogen load. The enhanced IL-10 expression in miR-132-/- Th1 cells is recapitulated in vitro following treatment with phenylephrine, a drug reported to promote ribosome synthesis. Our results uncover that miR-132/212-mediated regulation of RP expression is critical for optimal CD4+ T cell activation and protective immunity against pathogens.


Subject(s)
Gene Expression Regulation , MicroRNAs/genetics , RNA Interference , Ribosomal Proteins/genetics , Ribosomal Proteins/metabolism , Th1 Cells/immunology , Th1 Cells/metabolism , Animals , Binding Sites , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Cytokines/biosynthesis , Female , Gene Expression Profiling , Gene Regulatory Networks , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Mice , Mice, Transgenic , Protein Binding , Spleen/immunology , Spleen/metabolism , Spleen/microbiology , Transcription Factor TFIID/metabolism , p300-CBP Transcription Factors/metabolism
4.
Endocr Relat Cancer ; 26(1): 165-180, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30345732

ABSTRACT

Primary cilia are sensory organelles involved in regulation of cellular signaling. Cilia loss is frequently observed in tumors; yet, the responsible mechanisms and consequences for tumorigenesis remain unclear. We demonstrate that cilia structure and function is disrupted in human pheochromocytomas - endocrine tumors of the adrenal medulla. This is concomitant with transcriptional changes within cilia-mediated signaling pathways that are associated with tumorigenesis generally and pheochromocytomas specifically. Importantly, cilia loss was most dramatic in patients with germline mutations in the pseudohypoxia-linked genes SDHx and VHL. Using a pheochromocytoma cell line derived from rat, we show that hypoxia and oncometabolite-induced pseudohypoxia are key drivers of cilia loss and identify that this is dependent on activation of an Aurora-A/HDAC6 cilia resorption pathway. We also show cilia loss drives dramatic transcriptional changes associated with proliferation and tumorigenesis. Our data provide evidence for primary cilia dysfunction contributing to pathogenesis of pheochromocytoma by a hypoxic/pseudohypoxic mechanism and implicates oncometabolites as ciliary regulators. This is important as pheochromocytomas can cause mortality by mechanisms including catecholamine production and malignant transformation, while hypoxia is a general feature of solid tumors. Moreover, pseudohypoxia-induced cilia resorption can be pharmacologically inhibited, suggesting potential for therapeutic intervention.


Subject(s)
Adrenal Gland Neoplasms , Cilia , Pheochromocytoma , Adolescent , Adult , Aged , Animals , Child , Female , Humans , Male , Middle Aged , PC12 Cells , Rats , Young Adult
5.
EMBO Mol Med ; 10(8)2018 08.
Article in English | MEDLINE | ID: mdl-29930174

ABSTRACT

The adaptive cellular response to low oxygen tensions is mediated by the hypoxia-inducible factors (HIFs), a family of heterodimeric transcription factors composed of HIF-α and HIF-ß subunits. Prolonged HIF expression is a key contributor to cellular transformation, tumorigenesis and metastasis. As such, HIF degradation under hypoxic conditions is an essential homeostatic and tumour-suppressive mechanism. LIMD1 complexes with PHD2 and VHL in physiological oxygen levels (normoxia) to facilitate proteasomal degradation of the HIF-α subunit. Here, we identify LIMD1 as a HIF-1 target gene, which mediates a previously uncharacterised, negative regulatory feedback mechanism for hypoxic HIF-α degradation by modulating PHD2-LIMD1-VHL complex formation. Hypoxic induction of LIMD1 expression results in increased HIF-α protein degradation, inhibiting HIF-1 target gene expression, tumour growth and vascularisation. Furthermore, we report that copy number variation at the LIMD1 locus occurs in 47.1% of lung adenocarcinoma patients, correlates with enhanced expression of a HIF target gene signature and is a negative prognostic indicator. Taken together, our data open a new field of research into the aetiology, diagnosis and prognosis of LIMD1-negative lung cancers.


Subject(s)
Adenocarcinoma/genetics , Gene Expression Regulation, Neoplastic , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Intracellular Signaling Peptides and Proteins/metabolism , LIM Domain Proteins/metabolism , Lung Neoplasms/genetics , Adenocarcinoma/diagnosis , Adenocarcinoma/metabolism , Adenocarcinoma/mortality , Adult , Aged , Aged, 80 and over , Animals , Carcinogenesis/genetics , Carcinogenesis/metabolism , Cell Hypoxia/genetics , Cell Hypoxia/physiology , Cell Line, Tumor , Feedback, Physiological , Female , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Intracellular Signaling Peptides and Proteins/genetics , LIM Domain Proteins/genetics , Lung Neoplasms/diagnosis , Lung Neoplasms/metabolism , Lung Neoplasms/mortality , Male , Mice , Middle Aged , Prognosis , Survival Analysis , Vascular Endothelial Growth Factor A/genetics
6.
Biochem J ; 475(10): 1793-1806, 2018 05 31.
Article in English | MEDLINE | ID: mdl-29654110

ABSTRACT

To understand the mechanism of cellular stress in basal-parabasal layers of normal cervical epithelium and during different stages of cervical carcinoma, we analyzed the alterations (expression/methylation/copy number variation/mutation) of HIF-1α and its associated genes LIMD1, VHL and VEGF in disease-free normal cervix (n = 9), adjacent normal cervix of tumors (n = 70), cervical intraepithelial neoplasia (CIN; n = 32), cancer of uterine cervix (CACX; n = 174) samples and two CACX cell lines. In basal-parabasal layers of normal cervical epithelium, LIMD1 showed high protein expression, while low protein expression of VHL was concordant with high expression of HIF-1α and VEGF irrespective of HPV-16 (human papillomavirus 16) infection. This was in concordance with the low promoter methylation of LIMD1 and high in VHL in the basal-parabasal layers of normal cervix. LIMD1 expression was significantly reduced while VHL expression was unchanged during different stages of cervical carcinoma. This was in concordance with their frequent methylation during different stages of this tumor. In different stages of cervical carcinoma, the expression pattern of HIF-1α and VEGF was high as seen in basal-parabasal layers and inversely correlated with the expression of LIMD1 and VHL. This was validated by demethylation experiments using 5-aza-2'-deoxycytidine in CACX cell lines. Additional deletion of LIMD1 and VHL in CIN/CACX provided an additional growth advantage during cervical carcinogenesis through reduced expression of genes and associated with poor prognosis of patients. Our data showed that overexpression of HIF-1α and its target gene VEGF in the basal-parabasal layers of normal cervix was due to frequent inactivation of VHL by its promoter methylation. This profile was maintained during different stages of cervical carcinoma with additional methylation/deletion of VHL and LIMD1.


Subject(s)
Gene Expression Regulation, Neoplastic , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , LIM Domain Proteins/metabolism , Uterine Cervical Neoplasms/pathology , Vascular Endothelial Growth Factor A/metabolism , Von Hippel-Lindau Tumor Suppressor Protein/metabolism , DNA Copy Number Variations , DNA Methylation , Female , HeLa Cells , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Intracellular Signaling Peptides and Proteins/genetics , LIM Domain Proteins/genetics , Mutation , Neoplasm Staging , Promoter Regions, Genetic , Survival Rate , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/metabolism , Vascular Endothelial Growth Factor A/genetics , Von Hippel-Lindau Tumor Suppressor Protein/genetics
7.
Oncotarget ; 9(5): 6282-6297, 2018 Jan 19.
Article in English | MEDLINE | ID: mdl-29464072

ABSTRACT

LIMD1 (LIM domain-containing protein 1) is considered as a tumor suppressor, being deregulated in many cancers to include hematological malignancies; however, very little is known about the underlying mechanisms of its deregulation and its roles in carcinogenesis. Epstein-Barr Virus (EBV) is associated with a panel of malignancies of lymphocytic and epithelial origin. Using high throughput expression profiling, we have previously identified LIMD1 as a common marker associated with the oncogenic transcription factor IRF4 in EBV-related lymphomas and other hematological malignancies. In this study, we have identified potential conserved IRF4- and NFκB-binding motifs in the LIMD1 gene promoter, and both are demonstrated functional by promoter-reporter assays. We further show that LIMD1 is partially upregulated by EBV latent membrane protein 1 (LMP1) via IRF4 and NFκB in EBV latency. As to its role in the setting of EBV latent infection, we show that LIMD1 interacts with TRAF6, a crucial mediator of LMP1 signal transduction. Importantly, LIMD1 depletion impairs LMP1 signaling and functions, potentiates ionomycin-induced DNA damage and apoptosis, and inhibits p62-mediated selective autophagy. Taken together, these results show that LIMD1 is upregulated in EBV latency and plays an oncogenic role rather than that of a tumor suppressor. Our findings have identified LIMD1 as a novel player in EBV latency and oncogenesis, and open a novel research avenue, in which LIMD1 and p62 play crucial roles in linking DNA damage response (DDR), apoptosis, and autophagy and their potential interplay during viral oncogenesis.

8.
Stem Cell Res ; 25: 233-244, 2017 12.
Article in English | MEDLINE | ID: mdl-29172153

ABSTRACT

The class Ia anti-arrhythmic drug ajmaline is used clinically to unmask latent type I ECG in Brugada syndrome (BrS) patients, although its mode of action is poorly characterised. Our aims were to identify ajmaline's mode of action in human induced pluripotent stem cell (hiPSC)-derived cardiomyocytes (CMs), and establish a simple BrS hiPSC platform to test whether differences in ajmaline response could be determined between BrS patients and controls. Control hiPSCs were differentiated into spontaneously contracting cardiac clusters. It was found using multi electrode array (MEA) that ajmaline treatment significantly lengthened cluster activation-recovery interval. Patch clamping of single CMs isolated from clusters revealed that ajmaline can block both INa and IKr. Following generation of hiPSC lines from BrS patients (absent of pathogenic SCN5A sodium channel mutations), analysis of hiPSC-CMs from patients and controls revealed that differentiation and action potential parameters were similar. Comparison of cardiac clusters by MEA showed that ajmaline lengthened activation-recovery interval consistently across all lines. We conclude that ajmaline can block both depolarisation and repolarisation of hiPSC-CMs at the cellular level, but that a more refined integrated tissue model may be necessary to elicit differences in its effect between BrS patients and controls.


Subject(s)
Ajmaline/administration & dosage , Anti-Arrhythmia Agents/administration & dosage , Brugada Syndrome/drug therapy , Heart/drug effects , Myocytes, Cardiac/drug effects , Pluripotent Stem Cells/drug effects , Adult , Brugada Syndrome/genetics , Brugada Syndrome/metabolism , Brugada Syndrome/physiopathology , Cell Differentiation/drug effects , Heart/physiopathology , Humans , Male , Middle Aged , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , NAV1.5 Voltage-Gated Sodium Channel/genetics , NAV1.5 Voltage-Gated Sodium Channel/metabolism , Patch-Clamp Techniques , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism
9.
Pathol Res Pract ; 213(12): 1477-1481, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29033184

ABSTRACT

It is evident that the association of LimD1 and VHL could regulate stabilization of HIF1α protein. In this study, the expression of LimD1, VHL and HIF1α was analyzed in primary renal cell carcinoma (RCC) samples to understand their association with development of the disease. For this purpose, immunohistochemical expression analysis of LimD1, VHL and HIF1α was performed along with proliferation marker PCNA in 32 primary RCC samples in different subtypes at different clinical stages of Indian patients (year: 2014-2016). Significant decrease (P<0.05) in LimD1 and VHL expression was observed in different subtypes of RCC and in early invasive lesions. High nuclear expression of HIF1α was seen in different subtypes as well as in early invasive lesions. The LimD1 and VHL expression strongly correlated with each other, while inversely correlated with HIF1α. On the other hand, increased expression of PCNA was seen in different sub types and in early invasive lesions. The PCNA expression was negatively correlated with LimD1 and VHL expression and positively correlated with HIF1α. Thus, our data indicates that the reduced expression of LimD1 and VHL might have synergistic effect on induction of HIF1α resulting increased cellular proliferation and progression of the disease.


Subject(s)
Carcinoma, Renal Cell/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , LIM Domain Proteins/metabolism , Von Hippel-Lindau Tumor Suppressor Protein/metabolism , Aged , Carcinoma, Renal Cell/pathology , Cell Line, Tumor , Female , Humans , Kidney Neoplasms/pathology , Male , Middle Aged , Tumor Suppressor Proteins/metabolism
10.
J Biol Chem ; 292(50): 20683-20693, 2017 12 15.
Article in English | MEDLINE | ID: mdl-29066622

ABSTRACT

Programmed death ligand-1 (PD-L1) is a critical regulator of T cell function contributing to peripheral immune tolerance. Although it has been shown that posttranscriptional regulatory mechanisms control PD-L1 expression in cancer, it remains unknown whether such regulatory loops operate also in non-transformed cells. Here we studied PD-L1 expression in human dermal lymphatic endothelial cells (HDLECs), which play key roles in immunity and cancer. Treatment of HDLECs with the pro-inflammatory cytokines IFN-γ and TNF-α synergistically up-regulated PD-L1 expression. IFN-γ and TNF-α also affected expression of several microRNAs (miRNAs) that have the potential to suppress PD-L1 expression. The most highly up-regulated miRNA following IFN-γ and TNF-α treatment in HDLECs was miR-155, which has a central role in the immune system and cancer. Induction of miR-155 was driven by TNF-α, the effect of which was significantly enhanced by IFN-γ. The PD-L1 3'-UTR contains two functional miR-155-binding sites. Endogenous miR-155 controlled the kinetics and maximal levels of PD-L1 induction upon IFN-γ and TNF-α treatments. We obtained similar findings in dermal fibroblasts, demonstrating that the IFN-γ/TNF-α/miR-155/PD-L1 pathway is not restricted to HDLECs. These results reveal miR-155 as a critical component of an inflammation-induced regulatory loop controlling PD-L1 expression in primary cells.


Subject(s)
B7-H1 Antigen/antagonists & inhibitors , Dermis/metabolism , Endothelium, Lymphatic/metabolism , Gene Expression Regulation , Interferon-gamma/metabolism , MicroRNAs/agonists , Tumor Necrosis Factor-alpha/metabolism , 3' Untranslated Regions , B7-H1 Antigen/genetics , B7-H1 Antigen/metabolism , Base Sequence , Binding Sites , Cells, Cultured , Dermis/cytology , Dermis/immunology , Endothelium, Lymphatic/cytology , Endothelium, Lymphatic/immunology , Gene Expression Profiling , Genes, Reporter , Humans , Interferon-gamma/genetics , Kinetics , MicroRNAs/chemistry , MicroRNAs/metabolism , Microscopy, Fluorescence , RNA Interference , RNA, Small Interfering/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Response Elements
11.
Cell Rep ; 20(1): 173-187, 2017 07 05.
Article in English | MEDLINE | ID: mdl-28683311

ABSTRACT

As core components of the microRNA-induced silencing complex (miRISC), Argonaute (AGO) proteins interact with TNRC6 proteins, recruiting other effectors of translational repression/mRNA destabilization. Here, we show that LIMD1 coordinates the assembly of an AGO-TNRC6 containing miRISC complex by binding both proteins simultaneously at distinct interfaces. Phosphorylation of AGO2 at Ser 387 by Akt3 induces LIMD1 binding, which in turn enables AGO2 to interact with TNRC6A and downstream effector DDX6. Conservation of this serine in AGO1 and 4 indicates this mechanism may be a fundamental requirement for AGO function and miRISC assembly. Upon CRISPR-Cas9-mediated knockout of LIMD1, AGO2 miRNA-silencing function is lost and miRNA silencing becomes dependent on a complex formed by AGO3 and the LIMD1 family member WTIP. The switch to AGO3 utilization occurs due to the presence of a glutamic acid residue (E390) on the interaction interface, which allows AGO3 to bind to LIMD1, AJUBA, and WTIP irrespective of Akt signaling.


Subject(s)
Argonaute Proteins/metabolism , Gene Silencing , Intracellular Signaling Peptides and Proteins/metabolism , LIM Domain Proteins/metabolism , MicroRNAs/genetics , Argonaute Proteins/genetics , Autoantigens/metabolism , DEAD-box RNA Helicases/metabolism , HEK293 Cells , HeLa Cells , Humans , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/genetics , LIM Domain Proteins/chemistry , LIM Domain Proteins/genetics , MicroRNAs/metabolism , Phosphorylation , Protein Binding , Protein Processing, Post-Translational , Proto-Oncogene Proteins/metabolism , RNA-Binding Proteins/metabolism
12.
Am J Physiol Lung Cell Mol Physiol ; 312(2): L258-L267, 2017 02 01.
Article in English | MEDLINE | ID: mdl-27979861

ABSTRACT

Air-liquid interface (ALI) culture of primary airway epithelial cells enables mucociliary differentiation providing an in vitro model of the human airway, but their proliferative potential is limited. To extend proliferation, these cells were previously transduced with viral oncogenes or mouse Bmi-1 + hTERT, but the resultant cell lines did not undergo mucociliary differentiation. We hypothesized that use of human BMI-1 alone would increase the proliferative potential of bronchial epithelial cells while retaining their mucociliary differentiation potential. Cystic fibrosis (CF) and non-CF bronchial epithelial cells were transduced by lentivirus with BMI-1 and then their morphology, replication kinetics, and karyotype were assessed. When differentiated at ALI, mucin production, ciliary function, and transepithelial electrophysiology were measured. Finally, shRNA knockdown of DNAH5 in BMI-1 cells was used to model primary ciliary dyskinesia (PCD). BMI-1-transduced basal cells showed normal cell morphology, karyotype, and doubling times despite extensive passaging. The cell lines underwent mucociliary differentiation when cultured at ALI with abundant ciliation and production of the gel-forming mucins MUC5AC and MUC5B evident. Cilia displayed a normal beat frequency and 9+2 ultrastructure. Electrophysiological characteristics of BMI-1-transduced cells were similar to those of untransduced cells. shRNA knockdown of DNAH5 in BMI-1 cells produced immotile cilia and absence of DNAH5 in the ciliary axoneme as seen in cells from patients with PCD. BMI-1 delayed senescence in bronchial epithelial cells, increasing their proliferative potential but maintaining mucociliary differentiation at ALI. We have shown these cells are amenable to genetic manipulation and can be used to produce novel disease models for research and dissemination.


Subject(s)
Bronchi/cytology , Cell Differentiation , Cilia/metabolism , Epithelial Cells/cytology , Epithelial Cells/metabolism , Mucus/metabolism , Polycomb Repressive Complex 1/metabolism , Animals , Axonemal Dyneins/metabolism , Cell Proliferation , Cell Shape , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Dyneins/metabolism , Electric Impedance , Electrophysiological Phenomena , Gene Knockdown Techniques , HEK293 Cells , Humans , Kartagener Syndrome/metabolism , Kartagener Syndrome/pathology , Kartagener Syndrome/physiopathology , Karyotyping , Mice , Microtubules/metabolism , Models, Biological , Phenotype , Transduction, Genetic
13.
Physiol Rep ; 4(16)2016 08.
Article in English | MEDLINE | ID: mdl-27558999

ABSTRACT

The airway epithelium is altered in respiratory disease and is thought to contribute to disease etiology. A caveat to disease research is that the technique of isolation of bronchial epithelial cells from patients is invasive and cells have a limited lifespan. The aim of this study was to extensively characterize the plasticity of primary human bronchial epithelial cells that have been engineered to delay cell senescence including the ability of these cells to differentiate. Cells were engineered to express BMI-1 or hTERT using viral vector systems. Cells were characterized at passage (p) early (p5), mid (p10), and late (p15) stage for: BMI-1, p16, and CK14 protein expression, viability and the ability to differentiate at air-liquid interface (ALI), using a range of techniques including immunohistochemistry (IHC), immunofluorescence (IF), transepithelial electrical resistance (TEER), scanning electron microscopy (SEM), MUC5AC and beta tubulin (BTUB) staining. BMI-1-expressing cells maintained elevated levels of the BMI-1 protein and the epithelial marker CK14 and showed a suppression of p16. BMI-1-expressing cells had a viability advantage, differentiated at ALI, and had a normal karyotype. In contrast, hTERT-expressing cells had a reduced viability, showed limited differentiation, and had an abnormal karyotype. We therefore provide extensive characterization of the plasticity of BMI-1 expressing cells in the context of the ALI model. These cells retain properties of wild-type cells and may be useful to characterize respiratory disease mechanisms in vitro over sustained periods.


Subject(s)
Bronchi/cytology , Cell Plasticity/genetics , Epithelial Cells/metabolism , Polycomb Repressive Complex 1/metabolism , Adult , Cell Differentiation/genetics , Cell Differentiation/physiology , Cellular Senescence/genetics , Genes, p16 , Genetic Engineering/methods , Humans , In Vitro Techniques , Karyotype , Lentivirus/genetics , Male , Middle Aged , Peptide Fragments/metabolism , Retroviridae/genetics , Telomerase/metabolism
14.
Nucleic Acids Res ; 44(20): 9942-9955, 2016 Nov 16.
Article in English | MEDLINE | ID: mdl-27407113

ABSTRACT

MicroRNAs (miRNAs) are short non-coding RNAs that silence mRNAs. They are generated following transcription and cleavage by the DROSHA/DGCR8 and DICER/TRBP/PACT complexes. Although it is known that components of the miRNA biogenesis machinery can be phosphorylated, it remains poorly understood how these events become engaged during physiological cellular activation. We demonstrate that S6 kinases can phosphorylate the extended C-terminal domain of TRBP and interact with TRBP in situ in primary cells. TRBP serines 283/286 are essential for S6K-mediated TRBP phosphorylation, optimal expression of TRBP, and the S6K-TRBP interaction in human primary cells. We demonstrate the functional relevance of this interaction in primary human dermal lymphatic endothelial cells (HDLECs). Angiopoietin-1 (ANG1) can augment miRNA biogenesis in HDLECs through enhancing TRBP phosphorylation and expression in an S6K2-dependent manner. We propose that the S6K2/TRBP node controls miRNA biogenesis in HDLECs and provides a molecular link between the mTOR pathway and the miRNA biogenesis machinery.


Subject(s)
Endothelial Cells/metabolism , Gene Expression Regulation , MicroRNAs/genetics , RNA-Binding Proteins/metabolism , Ribosomal Protein S6 Kinases/metabolism , Angiopoietin-1/pharmacology , Cell Line , Cells, Cultured , Endothelial Cells/drug effects , Gene Expression Regulation/drug effects , Humans , Phosphorylation , Protein Interaction Domains and Motifs , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/genetics
15.
Cancer Res ; 76(16): 4720-4727, 2016 08 15.
Article in English | MEDLINE | ID: mdl-27325650

ABSTRACT

Other than an association with HPV infection, little is known about the genetic alterations determining the development of penile cancer. Although penile cancer is rare in the developed world, it presents a significant burden in developing countries. Here, we report the findings of whole-exome sequencing (WES) to determine the somatic mutational landscape of penile cancer. WES was performed on penile cancer and matched germline DNA from 27 patients undergoing surgical resection. Targeted resequencing of candidate genes was performed in an independent 70 patient cohort. Mutation data were also integrated with DNA methylation and copy-number information from the same patients. We identified an HPV-associated APOBEC mutation signature and an NpCpG signature in HPV-negative disease. We also identified recurrent mutations in the novel penile cancer tumor suppressor genes CSN1(GPS1) and FAT1 Expression of CSN1 mutants in cells resulted in colocalization with AGO2 in cytoplasmic P-bodies, ultimately leading to the loss of miRNA-mediated gene silencing, which may contribute to disease etiology. Our findings represent the first comprehensive analysis of somatic alterations in penile cancer, highlighting the complex landscape of alterations in this malignancy. Cancer Res; 76(16); 4720-7. ©2016 AACR.


Subject(s)
Cadherins/genetics , Carcinoma, Squamous Cell/genetics , Intracellular Signaling Peptides and Proteins/genetics , Penile Neoplasms/genetics , COP9 Signalosome Complex , DNA Copy Number Variations , DNA Methylation , DNA Mutational Analysis , Fluorescent Antibody Technique , Humans , Male , Mutation , Polymerase Chain Reaction
16.
Int J Biochem Cell Biol ; 69: 75-84, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26475020

ABSTRACT

The abundance of miR-132 ranges from constitutively high in the brain where it is necessary for neuronal development and function, to inducible expression in haematopoietic and endothelial cells where it controls angiogenesis and immune activation. We show that expression of AGO2, a protein central to miRNA-mediated gene silencing and miRNA biogenesis, is negatively regulated by miR-132. Using HeLa cells, we demonstrate that miR-132 interacts with the AGO2 mRNA 3'UTR and suppresses AGO2 expression and AGO2-dependent small RNA-mediated silencing. Similarly, miR-132 over-expression leads to AGO2 suppression in primary human dermal lymphatic endothelial cells (HDLECs). During phorbol myristate acetate (PMA)-activation of HDLECs, miR-132 is induced in a CREB-dependent manner and inhibition of miR-132 results in increased AGO2 expression. In agreement with the role of AGO2 in maintenance of miRNA expression, AGO2 suppression by miR-132 affects the steady state levels of miR-221 and miR-146a, two miRNAs involved in angiogenesis and inflammation, respectively. Our data demonstrate that the miRNA-silencing machinery is subject to autoregulation during primary cell activation through direct suppression of AGO2 by miR-132.


Subject(s)
Argonaute Proteins/genetics , Endothelial Cells/metabolism , MicroRNAs/physiology , 3' Untranslated Regions , Argonaute Proteins/metabolism , Base Sequence , Binding Sites , Conserved Sequence , Gene Expression , HeLa Cells , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Primary Cell Culture , RNA Interference
17.
Future Oncol ; 10(16): 2659-74, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25531051

ABSTRACT

Since the application of molecular biology in cancer biology, lung cancer research has classically focused on molecular drivers of disease. One such pathway, the hypoxic response pathway, is activated by reduced local oxygen concentrations at the tumor site. Hypoxia-driven gene and protein changes enhance epithelial-to-mesenchymal transition, remodel the extracellular matrix, drive drug resistance, support cancer stem cells and aid evasion from immune cells. However, it is not the tumor cells alone which drive this response to hypoxia, but rather their interaction with a complex milieu of supporting cells. This review will focus on recent advances in our understanding of how these cells contribute to the tumor response to hypoxia in non-small-cell lung cancer.


Subject(s)
Carcinogenesis , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Small Cell/genetics , Lung Neoplasms/genetics , Carcinoma, Non-Small-Cell Lung/classification , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Small Cell/classification , Carcinoma, Small Cell/pathology , Cell Hypoxia/genetics , Epithelial-Mesenchymal Transition/genetics , Extracellular Matrix/genetics , Extracellular Matrix/pathology , Humans , Lung Neoplasms/classification , Lung Neoplasms/pathology , Neoplastic Stem Cells/pathology , Oncogenes/genetics , Tumor Microenvironment/genetics , Tumor Suppressor Proteins/genetics
18.
Neuropsychopharmacology ; 39(2): 464-76, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24037344

ABSTRACT

Glutamate is the principle excitatory neurotransmitter in the mammalian brain, and dysregulation of glutamatergic neurotransmission is implicated in the pathophysiology of several psychiatric and neurological diseases. This study utilized novel lentiviral short hairpin RNA (shRNA) vectors to target expression of the vesicular glutamate transporter 1 (VGLUT1) following injection into the dorsal hippocampus of adult mice, as partial reductions in VGLUT1 expression should attenuate glutamatergic signaling and similar reductions have been reported in schizophrenia. The VGLUT1-targeting vector attenuated tonic glutamate release in the dorsal hippocampus without affecting GABA, and selectively impaired novel object discrimination (NOD) and retention (but not acquisition) in the Morris water maze, without influencing contextual fear-motivated learning or causing any adverse locomotor or central immune effects. This pattern of cognitive impairment is consistent with the accumulating evidence for functional differentiation along the dorsoventral axis of the hippocampus, and supports the involvement of dorsal hippocampal glutamatergic neurotransmission in both spatial and nonspatial memory. Future use of this nonpharmacological VGLUT1 knockdown mouse model could improve our understanding of glutamatergic neurobiology and aid assessment of novel therapies for cognitive deficits such as those seen in schizophrenia.


Subject(s)
Cognition Disorders/chemically induced , Cognition Disorders/genetics , Gene Transfer Techniques , Genetic Vectors/administration & dosage , Hippocampus/pathology , RNA, Small Interfering/administration & dosage , Vesicular Glutamate Transport Protein 1/antagonists & inhibitors , Vesicular Glutamate Transport Protein 1/genetics , Animals , Cell Line , Cognition Disorders/metabolism , Genetic Vectors/genetics , Lentivirus/genetics , Male , Mice , Mice, Inbred C57BL , RNA, Small Interfering/genetics , Vesicular Glutamate Transport Protein 1/metabolism
19.
PLoS One ; 8(11): e80470, 2013.
Article in English | MEDLINE | ID: mdl-24260396

ABSTRACT

TNF receptor-associated factors (TRAFs) are multifunctional adaptor proteins involved in temporal and spatial coordination of signals necessary for normal immune function. Here, we report that TRAF3, a TRAF family member with a key role in Toll-like and TNF family receptor signaling and suppressor of lymphomagenesis, is post-translationally modified by the small ubiquitin-related modifier (SUMO). Through yeast two-hybrid and co-immunoprecipitation assays we have identified Ubc9, the SUMO conjugating enzyme, as a novel TRAF3-interacting protein. We show that Ubc9-dependent SUMOylation of TRAF3 modulates optimal association with the CD40 receptor, thereby influencing TRAF3 degradation and non-canonical NF-κB activation upon CD40 triggering. Collectively, our findings describe a novel post-translational modification of a TRAF family member and reveal a link between SUMOylation and TRAF-mediated signal transduction.


Subject(s)
Signal Transduction , TNF Receptor-Associated Factor 3/metabolism , CD40 Antigens/metabolism , Cell Transformation, Neoplastic/metabolism , Humans , Lymphoma/metabolism , NF-kappa B p52 Subunit/metabolism , Protein Binding , Protein Interaction Domains and Motifs , Protein Interaction Mapping/methods , Proteolysis , Sumoylation , Two-Hybrid System Techniques , Ubiquitin-Conjugating Enzymes/metabolism
20.
Regen Med ; 8(6): 771-82, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24147532

ABSTRACT

Regenerative medicine relies on harnessing the capacity of stem cells to grow, divide and differentiate safely and predictably. This may be in the context of expanding stem cells in vitro or encouraging their expansion, mobilization and capacity to regenerate tissues either locally or remotely in vivo. In either case, understanding the stem cell niche is fundamental to recapitulating or manipulating conditions to enable therapy. It has become obvious that hypoxia plays a fundamental role in the maintenance of the stem cell niche. Low O2 benefits the self-renewal of human embryonic, hematopoietic, mesenchymal and neural stem cells, as well as improving the efficiency of genetic reprogramming to induced pluripotency. There is emerging evidence that harnessing or manipulating the hypoxic response can result in safer, more efficacious methodologies for regenerative medicine.


Subject(s)
Cell Differentiation , Stem Cells/cytology , Adult Stem Cells/cytology , Animals , Cell Hypoxia , Humans , Pluripotent Stem Cells/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...