Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Commun Biol ; 4(1): 5, 2021 01 04.
Article in English | MEDLINE | ID: mdl-33398110

ABSTRACT

Previous work has revealed that progerin-lamin A binding inhibitor (JH4) can ameliorate pathological features of Hutchinson-Gilford progeria syndrome (HGPS) such as nuclear deformation, growth suppression in patient's cells, and very short life span in an in vivo mouse model. Despite its favorable effects, JH4 is rapidly eliminated in in vivo pharmacokinetic (PK) analysis. Thus, we improved its property through chemical modification and obtained an optimized drug candidate, Progerinin (SLC-D011). This chemical can extend the life span of LmnaG609G/G609G mouse for about 10 weeks and increase its body weight. Progerinin can also extend the life span of LmnaG609G/+ mouse for about 14 weeks via oral administration, whereas treatment with lonafarnib (farnesyl-transferase inhibitor) can only extend the life span of LmnaG609G/+ mouse for about two weeks. In addition, progerinin can induce histological and physiological improvement in LmnaG609G/+ mouse. These results indicate that progerinin is a strong drug candidate for HGPS.


Subject(s)
Progeria/drug therapy , Adolescent , Animals , Child , Disease Models, Animal , Drug Evaluation, Preclinical , Female , HEK293 Cells , Humans , Lamin Type A/antagonists & inhibitors , Male , Mice , Primary Cell Culture
2.
Acta Physiol (Oxf) ; 228(3): e13374, 2020 03.
Article in English | MEDLINE | ID: mdl-31495066

ABSTRACT

AIM: Apoptosis of vascular smooth muscle cells (VSMCs) influenced by abnormal cyclic stretch is crucial for vascular remodelling during hypertension. Lamin A/C, a nuclear envelope protein, is mechano-responsive, but the role of lamin A/C in VSMC apoptosis is still unclear. METHODS: FX-5000T Strain Unit provided cyclic stretch (CS) in vitro. AnnexinV/PI and cleaved Caspase 3 ELISA detected apoptosis. qPCR was used to investigate the expression of miR-124-3p and a luciferase reporter assay was used to evaluate the ability of miR-124-3p binding to the Lmna 3'UTR. Protein changes of lamin A/C and relevant molecules were detected using western blot. Ingenuity Pathway Analysis and Protein/DNA array detected the potential transcription factors. Renal hypertensive rats verified these changes. RESULTS: High cyclic stretch (15%-CS) induced VSMC apoptosis and repressed lamin A/C expressions compared with normal (5%-CS) control. Downregulation of lamin A/C enhanced VSMC apoptosis. In addition, 15%-CS had no significant effect on mRNA expression of Lmna, and lamin A/C degradation was not induced by autophagy. 15%-CS elevated miR-124-3p bound to the 3'UTR of Lmna and negatively regulated protein expression of lamin A/C. Similar changes occurred in renal hypertensive rats compared with sham controls. Lamin A/C repression affected activity of TP53, CREB1, MYC, STAT1/5/6 and JUN, which may in turn affect apoptosis. CONCLUSION: Our data suggested that the decreased expression of lamin A/C upon abnormal cyclic stretch and hypertension may induce VSMC apoptosis. These mechano-responsive factors play important roles in VSMC apoptosis and might be novel therapeutic targets for vascular remodelling in hypertension.


Subject(s)
Hypertension/pathology , Lamin Type A/antagonists & inhibitors , MicroRNAs/genetics , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/pathology , Animals , Apoptosis/physiology , Cells, Cultured , Disease Models, Animal , Hypertension/genetics , Hypertension/metabolism , Male , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction , Stress, Mechanical
3.
Cell Mol Life Sci ; 76(11): 2199-2216, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30762072

ABSTRACT

The organization of the nuclear periphery is crucial for many nuclear functions. Nuclear lamins form dense network at the nuclear periphery and play a substantial role in chromatin organization, transcription regulation and in organization of nuclear pore complexes (NPCs). Here, we show that TPR, the protein located preferentially within the nuclear baskets of NPCs, associates with lamin B1. The depletion of TPR affects the organization of lamin B1 but not lamin A/C within the nuclear lamina as shown by stimulated emission depletion microscopy. Finally, reduction of TPR affects the distribution of NPCs within the nuclear envelope and the effect can be reversed by simultaneous knock-down of lamin A/C or the overexpression of lamin B1. Our work suggests a novel role for the TPR at the nuclear periphery: the TPR contributes to the organization of the nuclear lamina and in cooperation with lamins guards the interphase assembly of nuclear pore complexes.


Subject(s)
Lamin Type A/genetics , Lamin Type B/genetics , Nuclear Envelope/metabolism , Nuclear Lamina/metabolism , Nuclear Pore Complex Proteins/genetics , Proto-Oncogene Proteins/genetics , Gene Expression Regulation , HeLa Cells , Humans , Lamin Type A/antagonists & inhibitors , Lamin Type A/metabolism , Lamin Type B/metabolism , Molecular Imaging , Nuclear Envelope/ultrastructure , Nuclear Lamina/ultrastructure , Nuclear Pore Complex Proteins/antagonists & inhibitors , Nuclear Pore Complex Proteins/metabolism , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction
4.
Nucleus ; 10(1): 7-20, 2019 12.
Article in English | MEDLINE | ID: mdl-30663495

ABSTRACT

Immunosuppressive drugs such as cyclosporin A (CsA) can elicit hepatotoxicity by affecting gene expression. Here, we address the link between CsA and large-scale chromatin organization in HepG2 hepatocarcinoma cells. We show the existence of lamina-associated domains (LADs) interacting with lamin A, lamin B, or both. These 'A-B', 'A-only' and 'B-only' LADs display distinct fates after CsA treatment: A-B LADs remain constitutive or lose A, A-only LADs mainly lose A or switch to B, and B-only LADs remain B-only or acquire A. LAD rearrangement is overall uncoupled from changes in gene expression. Three-dimensional (3D) genome modeling predicts changes in radial positioning of LADs as LADs switch identities, which are corroborated by fluorescence in situ hybridization. Our results reveal interplay between A- and B-type lamins on radial locus positioning, suggesting complementary contributions to large-scale genome architecture. The data also unveil a hitherto unsuspected impact of cytotoxic drugs on genome conformation.Abbreviations: ChIP-seq: chromatin immunoprecipitation sequencing; CsA: cyclosporin A; FISH; fluorescence in situ hybridization; ICMT: isoprenylcysteine methyltransferase; LAD: lamina-associated domain; TAD: topologically-associated domain.


Subject(s)
Chromatin/metabolism , Lamin Type A/metabolism , Lamin Type B/metabolism , Nuclear Lamina/metabolism , Chromatin/drug effects , Cyclosporine/pharmacology , Hep G2 Cells , Humans , In Situ Hybridization, Fluorescence , Lamin Type A/antagonists & inhibitors , Lamin Type B/antagonists & inhibitors , Models, Genetic , Nuclear Lamina/drug effects , Tumor Cells, Cultured
5.
J Mol Biol ; 431(9): 1780-1791, 2019 04 19.
Article in English | MEDLINE | ID: mdl-30597161

ABSTRACT

RNA is accurately entangled in virtually all pathways that maintain cellular homeostasis. To name but a few, RNA is the "messenger" between DNA encoded information and the resulting proteins. Furthermore, RNAs regulate diverse processes by forming DNA::RNA or RNA::RNA interactions. Finally, RNA itself can be the scaffold for ribonucleoprotein complexes, for example, ribosomes or cellular bodies. Consequently, disruption of any of these processes can lead to disease. This review describes known and emerging RNA-based disease mechanisms like interference with regular splicing, the anomalous appearance of RNA-protein complexes and uncommon RNA species, as well as non-canonical translation. Due to the complexity and entanglement of the above-mentioned pathways, only few drugs are available that target RNA-based disease mechanisms. However, advances in our understanding how RNA is involved in and modulates cellular homeostasis might pave the way to novel treatments.


Subject(s)
DNA/genetics , Molecular Targeted Therapy/methods , Neurodegenerative Diseases/genetics , RNA Splicing , RNA/genetics , Animals , DNA/metabolism , Disease Models, Animal , Dystrophin/antagonists & inhibitors , Dystrophin/genetics , Dystrophin/metabolism , Humans , Lamin Type A/antagonists & inhibitors , Lamin Type A/genetics , Lamin Type A/metabolism , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Neurodegenerative Diseases/therapy , Oligonucleotides, Antisense/administration & dosage , Oligonucleotides, Antisense/genetics , Oligonucleotides, Antisense/metabolism , Protein Biosynthesis , Protein Kinases/genetics , Protein Kinases/metabolism , RNA/classification , RNA/metabolism , RNA Interference , Transcription, Genetic , tau Proteins/antagonists & inhibitors , tau Proteins/genetics , tau Proteins/metabolism
6.
Nucleus ; 9(1): 230-245, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29619860

ABSTRACT

Interphase phosphorylation of lamin-A,C depends dynamically on a cell's microenvironment, including the stiffness of extracellular matrix. However, phosphorylation dynamics is poorly understood for diseased forms such as progerin, a permanently farnesylated mutant of LMNA that accelerates aging of stiff and mechanically stressed tissues. Here, fine-excision alignment mass spectrometry (FEA-MS) is developed to quantify progerin and its phosphorylation levels in patient iPS cells differentiated to mesenchymal stem cells (MSCs). The stoichiometry of total A-type lamins (including progerin) versus B-type lamins measured for Progeria iPS-MSCs prove similar to that of normal MSCs, with total A-type lamins more abundant than B-type lamins. However, progerin behaves more like farnesylated B-type lamins in mechanically-induced segregation from nuclear blebs. Phosphorylation of progerin at multiple sites in iPS-MSCs cultured on rigid plastic is also lower than that of normal lamin-A and C. Reduction of nuclear tension upon i) cell rounding/detachment from plastic, ii) culture on soft gels, and iii) inhibition of actomyosin stress increases phosphorylation and degradation of lamin-C > lamin-A > progerin. Such mechano-sensitivity diminishes, however, with passage as progerin and DNA damage accumulate. Lastly, transcription-regulating retinoids exert equal effects on both diseased and normal A-type lamins, suggesting a differential mechano-responsiveness might best explain the stiff tissue defects in Progeria.


Subject(s)
Induced Pluripotent Stem Cells/metabolism , Lamin Type A/metabolism , Mechanotransduction, Cellular , Mesenchymal Stem Cells/metabolism , Actomyosin/pharmacology , Humans , Induced Pluripotent Stem Cells/drug effects , Lamin Type A/antagonists & inhibitors , Mechanotransduction, Cellular/drug effects , Mesenchymal Stem Cells/drug effects , Phosphorylation/drug effects
7.
PLoS One ; 12(4): e0175953, 2017.
Article in English | MEDLINE | ID: mdl-28422997

ABSTRACT

Nuclear lamins support the nuclear envelope and provide anchorage sites for chromatin. They are involved in DNA synthesis, transcription, and replication. It has previously been reported that the lack of Lamin A/C expression in lymphoma and leukaemia is due to CpG island promoter hypermethylation. Here, we provide evidence that Lamin A/C is silenced via this mechanism in a subset of neuroblastoma cells. Moreover, Lamin A/C expression can be restored with a demethylating agent. Importantly, Lamin A/C reintroduction reduced cell growth kinetics and impaired migration, invasion, and anchorage-independent cell growth. Cytoskeletal restructuring was also induced. In addition, the introduction of lamin Δ50, known as Progerin, caused senescence in these neuroblastoma cells. These cells were stiffer and developed a cytoskeletal structure that differed from that observed upon Lamin A/C introduction. Of relevance, short hairpin RNA Lamin A/C depletion in unmethylated neuroblastoma cells enhanced the aforementioned tumour properties. A cytoskeletal structure similar to that observed in methylated cells was induced. Furthermore, atomic force microscopy revealed that Lamin A/C knockdown decreased cellular stiffness in the lamellar region. Finally, the bioinformatic analysis of a set of methylation arrays of neuroblastoma primary tumours showed that a group of patients (around 3%) gives a methylation signal in some of the CpG sites located within the Lamin A/C promoter region analysed by bisulphite sequencing PCR. These findings highlight the importance of Lamin A/C epigenetic inactivation for a subset of neuroblastomas, leading to enhanced tumour properties and cytoskeletal changes. Additionally, these findings may have treatment implications because tumour cells lacking Lamin A/C exhibit more aggressive behaviour.


Subject(s)
Brain Neoplasms/genetics , DNA Methylation , Gene Expression Regulation, Neoplastic , Gene Silencing , Lamin Type A/genetics , Neuroblastoma/genetics , Actin Cytoskeleton/metabolism , Actin Cytoskeleton/ultrastructure , Base Sequence , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Line , Cell Line, Tumor , Cell Movement , Cell Proliferation , CpG Islands , Humans , Lamin Type A/antagonists & inhibitors , Lamin Type A/metabolism , Neural Stem Cells/metabolism , Neural Stem Cells/pathology , Neuroblastoma/metabolism , Neuroblastoma/pathology , Neuroglia/metabolism , Neuroglia/pathology , Neurons/metabolism , Neurons/pathology , Primary Cell Culture , Promoter Regions, Genetic , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction
8.
Cell Death Dis ; 7: e2105, 2016 Feb 18.
Article in English | MEDLINE | ID: mdl-26890144

ABSTRACT

Hutchinson-Gilford progeria syndrome (HGPS) is a rare genetic disorder characterized by a dramatic appearance of premature aging. HGPS is due to a single-base substitution in exon 11 of the LMNA gene (c.1824C>T) leading to the production of a toxic form of the prelamin A protein called progerin. Because farnesylation process had been shown to control progerin toxicity, in this study we have developed a screening method permitting to identify new pharmacological inhibitors of farnesylation. For this, we have used the unique potential of pluripotent stem cells to have access to an unlimited and relevant biological resource and test 21,608 small molecules. This study identified several compounds, called monoaminopyrimidines, which target two key enzymes of the farnesylation process, farnesyl pyrophosphate synthase and farnesyl transferase, and rescue in vitro phenotypes associated with HGPS. Our results opens up new therapeutic possibilities for the treatment of HGPS by identifying a new family of protein farnesylation inhibitors, and which may also be applicable to cancers and diseases associated with mutations that involve farnesylated proteins.


Subject(s)
Lamin Type A/metabolism , Progeria/pathology , Protein Prenylation/drug effects , Pyrimidines/pharmacology , Binding Sites , Cell Differentiation/drug effects , Farnesyltranstransferase/antagonists & inhibitors , Farnesyltranstransferase/metabolism , Geranyltranstransferase/antagonists & inhibitors , Geranyltranstransferase/metabolism , Humans , Lamin Type A/antagonists & inhibitors , Lamin Type A/genetics , Molecular Docking Simulation , Osteogenesis/drug effects , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/drug effects , Pluripotent Stem Cells/metabolism , Progeria/metabolism , Protein Structure, Tertiary , Pyrimidines/chemistry , Pyrimidines/metabolism , Small Molecule Libraries/chemistry , Small Molecule Libraries/metabolism , Small Molecule Libraries/pharmacology , Structure-Activity Relationship
9.
J Natl Cancer Inst ; 108(1)2016 Jan.
Article in English | MEDLINE | ID: mdl-26563355

ABSTRACT

In metastatic colorectal cancer (CRC), actionable genetic lesions represent potential clinical opportunities. NTRK1, 2, and 3 gene rearrangements encode oncogenic fusions of the tropomyosin-receptor kinase (TRK) family of receptor tyrosine kinases in different tumor types. The TPM3-NTRK1 rearrangement is a recurring event in CRC that renders tumors sensitive to TRKA kinase inhibitors in preclinical models. We identified abnormal expression of the TRKA protein in tumor and liver metastases of a CRC patient refractory to standard therapy. Molecular characterization unveiled a novel LMNA-NTRK1 rearrangement within chromosome 1 with oncogenic potential, and the patient was treated with the pan-TRK inhibitor entrectinib, achieving partial response with decrease in hepatic target lesions from 6.8 and 8.2cm in longest diameter to 4.7 and 4.3cm, respectively. To our knowledge, this is the first clinical evidence of efficacy for therapeutic inhibition of TRKA in a solid tumor, illuminating a genomic-driven strategy to identify CRCs reliant on this oncogene to be clinically targeted with entrectinib.


Subject(s)
Antineoplastic Agents/pharmacology , Colorectal Neoplasms/drug therapy , Gene Fusion , Gene Rearrangement , Lamin Type A/genetics , Liver Neoplasms/drug therapy , Proteins/genetics , Receptor, trkA/genetics , Aged , Anaplastic Lymphoma Kinase , Antineoplastic Agents/administration & dosage , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Drug Administration Schedule , Female , Gene Expression Regulation, Neoplastic , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence , Lamin Type A/antagonists & inhibitors , Liver Neoplasms/secondary , Molecular Targeted Therapy , Protein-Tyrosine Kinases/antagonists & inhibitors , Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/antagonists & inhibitors , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Receptor, trkA/antagonists & inhibitors
10.
Methods ; 96: 46-58, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26341717

ABSTRACT

Hutchinson-Gilford Progeria Syndrome (HGPS) is an early onset lethal premature aging disorder caused by constitutive production of progerin, a mutant form of the nuclear architectural protein lamin A. The presence of progerin causes extensive morphological, epigenetic and DNA damage related nuclear defects that ultimately disrupt tissue and organismal functions. Hypothesis-driven approaches focused on HGPS affected pathways have been used in attempts to identify druggable targets with anti-progeroid effects. Here, we report an unbiased discovery approach to HGPS by implementation of a high-throughput, high-content imaging based screening method that enables systematic identification of small molecules that prevent the formation of multiple progerin-induced aging defects. Screening a library of 2816 FDA approved drugs, we identified retinoids as a novel class of compounds that reverses aging defects in HGPS patient skin fibroblasts. These findings establish a novel approach to anti-progeroid drug discovery.


Subject(s)
Cellular Senescence/drug effects , Fibroblasts/drug effects , High-Throughput Screening Assays , Lamin Type A/antagonists & inhibitors , Molecular Imaging/methods , Retinoids/pharmacology , Cell Line, Transformed , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cell Nucleus/ultrastructure , Cellular Senescence/genetics , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , Epigenesis, Genetic , Fibroblasts/metabolism , Fibroblasts/pathology , Histones/genetics , Histones/metabolism , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Lamin Type A/genetics , Lamin Type A/metabolism , Lamin Type B/genetics , Lamin Type B/metabolism , Plasmids/chemistry , Plasmids/metabolism , Primary Cell Culture , Progeria/genetics , Progeria/metabolism , Progeria/pathology , Small Molecule Libraries/pharmacology , Transfection , Tumor Suppressor p53-Binding Protein 1
11.
Nucleus ; 6(3): 236-46, 2015.
Article in English | MEDLINE | ID: mdl-25996284

ABSTRACT

The cell nucleus is structurally and functionally organized by lamins, intermediate filament proteins that form the nuclear lamina. Point mutations in genes that encode a specific subset of lamins, the A-type lamins, cause a spectrum of diseases termed laminopathies. Recent evidence points to a role for A-type lamins in intracellular redox homeostasis. To determine whether lamin A/C depletion and prelamin A accumulation differentially induce oxidative stress, we have performed a quantitative microscopy-based analysis of reactive oxygen species (ROS) levels and mitochondrial membrane potential (Δψm) in human fibroblasts subjected to sustained siRNA-mediated knockdown of LMNA and ZMPSTE24, respectively. We measured a highly significant increase in basal ROS levels and an even more prominent rise of induced ROS levels in lamin A/C depleted cells, eventually resulting in Δψm hyperpolarization and apoptosis. Depletion of ZMPSTE24 on the other hand, triggered a senescence pathway that was associated with moderately increased ROS levels and a transient Δψm depolarization. Both knockdowns were accompanied by an upregulation of several ROS detoxifying enzymes. Taken together, our data suggest that both persistent prelamin A accumulation and lamin A/C depletion elevate ROS levels, but to a different extent and with different effects on cell fate. This may contribute to the variety of disease phenotypes witnessed in laminopathies.


Subject(s)
Fibroblasts/metabolism , Lamin Type A/metabolism , Mitochondria/metabolism , Nuclear Lamina/metabolism , Reactive Oxygen Species/metabolism , Apoptosis , Fibroblasts/cytology , Gene Expression Regulation , Humans , Lamin Type A/antagonists & inhibitors , Lamin Type A/genetics , Membrane Potential, Mitochondrial , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Membrane Proteins/metabolism , Metalloendopeptidases/antagonists & inhibitors , Metalloendopeptidases/genetics , Metalloendopeptidases/metabolism , Mitochondria/pathology , Nuclear Lamina/chemistry , Oxidative Stress , Primary Cell Culture , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Reactive Oxygen Species/agonists , Signal Transduction , Time Factors
12.
PLoS One ; 7(9): e45513, 2012.
Article in English | MEDLINE | ID: mdl-23049808

ABSTRACT

BACKGROUND: Neuroblastoma (NB) is one of the most aggressive tumors that occur in childhood. Although genes, such as MYCN, have been shown to be involved in the aggressiveness of the disease, the identification of new biological markers is still desirable. The induction of differentiation is one of the strategies used in the treatment of neuroblastoma. A-type lamins are components of the nuclear lamina and are involved in differentiation. We studied the role of Lamin A/C in the differentiation and progression of neuroblastoma. METHODOLOGY/PRINCIPAL FINDINGS: Knock-down of Lamin A/C (LMNA-KD) in neuroblastoma cells blocked retinoic acid-induced differentiation, preventing neurites outgrowth and the expression of neural markers. The genome-wide gene-expression profile and the proteomic analysis of LMNA-KD cells confirmed the inhibition of differentiation and demonstrated an increase of aggressiveness-related genes and molecules resulting in augmented migration/invasion, and increasing the drug resistance of the cells. The more aggressive phenotype acquired by LMNA-KD cells was also maintained in vivo after injection into nude mice. A preliminary immunohistochemistry analysis of Lamin A/C expression in nine primary stages human NB indicated that this protein is poorly expressed in most of these cases. CONCLUSIONS/SIGNIFICANCE: We demonstrated for the first time in neuroblastoma cells that Lamin A/C plays a central role in the differentiation, and that the loss of this protein gave rise to a more aggressive tumor phenotype.


Subject(s)
Brain Neoplasms/genetics , Brain Neoplasms/pathology , Lamin Type A/genetics , Neuroblastoma/genetics , Neuroblastoma/pathology , Animals , Antibiotics, Antineoplastic/pharmacology , Brain Neoplasms/drug therapy , Brain Neoplasms/metabolism , Cell Differentiation/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Doxorubicin/pharmacology , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockdown Techniques , Humans , Lamin Type A/antagonists & inhibitors , Lamin Type A/metabolism , Mice , Mice, Nude , Neoplasm Invasiveness , Neoplasm Transplantation , Neurites/drug effects , Neuroblastoma/drug therapy , Neuroblastoma/metabolism , Proteome/genetics , Proteome/metabolism , Tretinoin/pharmacology
13.
Nucleus ; 2(5): 434-43, 2011.
Article in English | MEDLINE | ID: mdl-21983087

ABSTRACT

Up-regulated expression of lamin A has been implicated in increased cell invasiveness and mortality in colorectal cancer. Here we use quantitative proteomics to investigate lamin A regulated changes in the cytoskeleton that might underpin increased cell motility. Using siRNA knockdown of lamin A in a model cell line (SW480/lamA) we confirm that the presence of lamin A promotes cell motility. Using an enhanced technique to prepare cytoskeleton fractions in combination with 2D DiGE we were able to accurately and reproducibly detect changes in the representation of protein species within the cytoskeleton as low as 20%. In total 64 protein spots displayed either increased or decreased representation within the cytoskeleton of SW480/lamA cells compared to controls. Of these the identities of 29 spots were determined by mass spectrometry. A majority were multiple forms of three classes of proteins, including components of the actin and IF cytoskeletons, protein chaperones and translation initiation and elongation factors. In particular our data reveal that the representation of tissue transglutaminase 2, which is known to modify elements of the cytoskeleton and is associated with cancer progression, was highly over-represented in the cytoskeleton fraction of SW480/lamA cells. Overall, our data are consistent with changed protein cross-linking and folding that favours the formation of dynamic actin filaments over stress fibres accounting for the altered cell motility properties in SW480/lamA cells.


Subject(s)
Colorectal Neoplasms/pathology , Cytoskeleton/physiology , Lamin Type A/physiology , Proteomics , Cell Line, Tumor , Colorectal Neoplasms/metabolism , Cytoskeletal Proteins/metabolism , Electrophoresis, Gel, Two-Dimensional , GTP-Binding Proteins/metabolism , Gene Expression Regulation, Neoplastic , Humans , Lamin Type A/antagonists & inhibitors , Lamin Type A/metabolism , Mass Spectrometry , Protein Glutamine gamma Glutamyltransferase 2 , RNA Interference , RNA, Small Interfering/metabolism , Transglutaminases/metabolism
14.
Cell Cycle ; 10(15): 2549-60, 2011 Aug 01.
Article in English | MEDLINE | ID: mdl-21701264

ABSTRACT

A-type lamins are emerging as regulators of nuclear organization and function. Changes in their expression are associated with cancer and mutations are linked to degenerative diseases -laminopathies-. Although a correlation exists between alterations in lamins and genomic instability, the molecular mechanisms remain largely unknown. We previously found that loss of A-type lamins leads to degradation of 53BP1 protein and defective long-range non-homologous end-joining (NHEJ) of dysfunctional telomeres. Here, we determined how loss of A-type lamins affects the repair of short-range DNA double-strand breaks (DSBs) induced by ionizing radiation (IR). We find that lamins deficiency allows activation of the DNA damage response, but compromises the accumulation of 53BP1 at IR-induced foci (IRIF), hindering the fast phase of repair corresponding to classical-NHEJ. Importantly, reconstitution of 53BP1 is sufficient to rescue long-range and short-range NHEJ. Moreover, we demonstrate an unprecedented role for A-type lamins in the maintenance of homologous recombination (HR). Depletion of lamins compromises HR by a mechanism involving transcriptional downregulation of BRCA1 and RAD51 by the repressor complex formed by the Rb family member p130 and E2F4. In line with the DNA repair defects, lamins-deficient cells exhibit increased radiosensitivity. This study demonstrates that A-type lamins promote genomic stability by maintaining the levels of proteins with key roles in DNA DSBs repair by NHEJ and HR. Our results suggest that silencing of A-type lamins by DNA methylation in some cancers could contribute to the genomic instability that drives malignancy. In addition, lamins-deficient tumor cells could represent a good target for radiation therapy.


Subject(s)
DNA Breaks, Double-Stranded , DNA End-Joining Repair , Lamin Type A/metabolism , Animals , BRCA1 Protein/metabolism , Cell Line , Chromosomal Instability , Chromosomal Proteins, Non-Histone/metabolism , DNA-Binding Proteins/metabolism , E2F4 Transcription Factor/metabolism , Homologous Recombination , Humans , Lamin Type A/antagonists & inhibitors , Mice , RNA Interference , RNA, Small Interfering/metabolism , Rad51 Recombinase/metabolism , Radiation, Ionizing , Retinoblastoma-Like Protein p130/metabolism , Tumor Suppressor p53-Binding Protein 1
15.
J Virol ; 84(9): 4524-33, 2010 May.
Article in English | MEDLINE | ID: mdl-20147387

ABSTRACT

The Epstein-Barr virus (EBV)-encoded viral protein kinase, EBV-PK (the BGLF4 gene product), is required for efficient nuclear viral egress in 293 cells. However, since EBV-PK phosphorylates a number of different viral and cellular proteins (including lamin A/C), the relative importance of each target during lytic viral replication remains unclear. We show here that an EBV PK mutant (PKmut; containing stop codons at residues 1 and 5 in EBV-PK) is highly defective for release of infectious virus from 293 cells but not 293T cells. Furthermore, the phenotype of the PKmut in 293 cells is substantially reversed by expression of the simian virus 40 (SV40) large (T) and small (t) T antigens. Efficient rescue requires the presence of both SV40 T/t proteins. We show that 293T cells have a much higher level of constitutive lamin A/C phosphorylation than do 293 cells over residues (S22 and S392) that promote phosphorylation-dependent nuclear disassembly and that both large T and small t contribute to enhanced lamin A/C phosphorylation. Finally, we demonstrate that knockdown of lamin A/C expression using small interfering RNA also rescues the PKmut phenotype in 293 cells. These results suggest that essential roles of EBV-PK during lytic viral replication include the phosphorylation and dispersion of lamin A/C.


Subject(s)
Antigens, Polyomavirus Transforming/biosynthesis , Herpesvirus 4, Human/growth & development , Host-Pathogen Interactions , Lamin Type A/antagonists & inhibitors , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/physiology , Viral Proteins/physiology , Cell Line , Gene Knockdown Techniques , Herpesvirus 4, Human/genetics , Humans , Lamin Type A/metabolism , Phosphorylation , RNA, Small Interfering/genetics , Simian virus 40
16.
Biochim Biophys Acta ; 1792(1): 75-81, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19022376

ABSTRACT

BACKGROUND: Mutations in genes encoding A-type lamins and emerin cause cardiomyopathy and muscular dystrophy. We previously showed activation of the extracellular signal-regulated kinase (ERK) branch of the mitogen-activated protein kinase (MAPK) cascade in hearts of mice with mutations in these genes. Here, we tested the hypothesis that reducing A-type lamins and emerin in cultured cells activate ERK signaling. METHODS: We used siRNA to knockdown A-type lamins and emerin in HeLa and C2C12 cells. Activation of ERK was assessed by immunoblotting and immunofluorescence microscopy with antibodies against phosphorylated protein and by using real-time RT-PCR to measure RNAs encoded by genes for transcription factors stimulated by ERK. RESULTS: Knockdown of A-type lamins and emerin in HeLa and C2C12 stimulated phosphorylation and nuclear translocation of ERK as well as activation of genes encoding downstream transcription factors. A MAPK/ERK kinase (MEK) inhibitor reduced ERK phosphorylation in cells with reduced expression of A-type lamins and emerin. CONCLUSIONS: These results provide proof for the hypothesis that altered expression of emerin and A-type lamins activates ERK signaling, which in turn can cause cardiomyopathy. GENERAL SIGNIFICANCE: ERK is a potential target for the pharmacological treatment of cardiomyopathy caused by mutations in the genes encoding emerin and A-type lamins.


Subject(s)
Lamin Type A/genetics , MAP Kinase Signaling System , Membrane Proteins/genetics , Nuclear Proteins/genetics , Animals , Cardiomyopathies/etiology , Cardiomyopathies/genetics , Cardiomyopathies/metabolism , Cell Line , Gene Expression , HeLa Cells , Humans , Lamin Type A/antagonists & inhibitors , Membrane Proteins/antagonists & inhibitors , Mice , Mitogen-Activated Protein Kinases/metabolism , Mutation , Nuclear Proteins/antagonists & inhibitors , RNA, Small Interfering/genetics
17.
Mol Cell Biochem ; 306(1-2): 123-32, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17657589

ABSTRACT

The technique of RNA interference (RNAi) was trialed in primary human foreskin fibroblasts, both in monolayer culture and in the fibroblast-populated collagen matrix. Knockdown of lamin A/C, p53, and FAK was possible with low-confluency (<50%) monolayer fibroblasts, a transfection vehicle concentration of 1%, and an siRNA concentration of 25-50 nM. Knockdown also was possible in the collagen matrix using similar reagent concentrations and a cellular density of one million fibroblasts per ml of matrix. Optimization of transfection conditions appeared to be important to increase knockdown efficiency. Consistent with prediction, knockdown of FAK induced apoptosis in the fibroblast-populated collagen matrix.


Subject(s)
Collagen/metabolism , Extracellular Matrix/metabolism , Fibroblasts/metabolism , Focal Adhesion Kinase 1/metabolism , Foreskin/cytology , Lamin Type A/metabolism , RNA Interference , Tumor Suppressor Protein p53/metabolism , Cells, Cultured , Fluorescent Antibody Technique , Focal Adhesion Kinase 1/antagonists & inhibitors , Focal Adhesion Kinase 1/genetics , Humans , Kidney/metabolism , Lamin Type A/antagonists & inhibitors , Lamin Type A/genetics , Male , RNA, Small Interfering/pharmacology , Tumor Suppressor Protein p53/antagonists & inhibitors , Tumor Suppressor Protein p53/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...