Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Oncotarget ; 7(21): 30018-31, 2016 May 24.
Article in English | MEDLINE | ID: mdl-27145372

ABSTRACT

Hutchinson-Gilford Progeria Syndrome (HGPS) is a devastating incurable premature aging disease caused by accumulation of progerin, a toxic lamin A mutant protein. HGPS patient-derived cells exhibit nuclear morphological abnormalities, altered signaling pathways, genomic instability, and premature senescence. Here we uncover new molecular mechanisms contributing to cellular decline in progeria. We demonstrate that HGPS cells reduce expression of vitamin D receptor (VDR) and DNA repair factors BRCA1 and 53BP1 with progerin accumulation, and that reconstituting VDR signaling via 1α,25-dihydroxyvitamin D3 (1,25D) treatment improves HGPS phenotypes, including nuclear morphological abnormalities, DNA repair defects, and premature senescence. Importantly, we discovered that the 1,25D/VDR axis regulates LMNA gene expression, as well as expression of DNA repair factors. 1,25D dramatically reduces progerin production in HGPS cells, while stabilizing BRCA1 and 53BP1, two key factors for genome integrity. Vitamin D/VDR axis emerges as a new target for treatment of HGPS and potentially other lamin-related diseases exhibiting VDR deficiency and genomic instability. Because progerin expression increases with age, maintaining vitamin D/VDR signaling could keep the levels of progerin in check during physiological aging.


Subject(s)
Aging, Premature/metabolism , Calcitriol/pharmacology , Lamin Type A/metabolism , Progeria/metabolism , Receptors, Calcitriol/metabolism , Signal Transduction , Vitamins/pharmacology , Aging, Premature/genetics , Calcitriol/therapeutic use , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cells, Cultured , Cellular Senescence/drug effects , DNA Repair/drug effects , Down-Regulation , Fibroblasts , Fluorescent Antibody Technique , Gene Expression Regulation , Genomic Instability , Humans , Lamin Type A/genetics , Mutation , Nuclear Lamina/genetics , Nuclear Lamina/metabolism , Phenotype , Primary Cell Culture , Progeria/drug therapy , Progeria/genetics , RNA Interference , RNA, Small Interfering , Tumor Suppressor Proteins/metabolism , Tumor Suppressor p53-Binding Protein 1/metabolism , Ubiquitin Thiolesterase/metabolism , Vitamins/therapeutic use
2.
Nucleus ; 4(5): 410-9, 2013.
Article in English | MEDLINE | ID: mdl-24153156

ABSTRACT

Over 300 mutations in the LMNA gene, encoding A-type lamins, are associated with 15 human degenerative disorders and premature aging syndromes. Although genomic instability seems to contribute to the pathophysiology of some laminopathies, there is limited information about what mutations cause genomic instability and by which molecular mechanisms. Mouse embryonic fibroblasts depleted of A-type lamins or expressing mutants lacking exons 8-11 (Lmna(Δ8-11/Δ8-11)) exhibit alterations in telomere biology and DNA repair caused by cathepsin L-mediated degradation of 53BP1 and reduced expression of BRCA1 and RAD51. Thus, a region encompassing exons 8-11 seems essential for genome integrity. Given that deletion of lamin A exon 9 in the mouse (Lmna(Δ9/Δ9)) results in a progeria phenotype, we tested if this domain is important for genome integrity. Lmna(Δ9/Δ9) MEFs exhibit telomere shortening and heterochromatin alterations but do not activate cathepsin L-mediated degradation of 53BP1 and maintain expression of BRCA1 and RAD51. Accordingly, Lmna(Δ9/Δ9) MEFs do not present genomic instability, and expression of mutant lamin A Δexon9 in lamin-depleted cells restores DNA repair factors levels and partially rescues nuclear abnormalities. These data reveal that the domain encoded by exon 9 is important to maintain telomere homeostasis and heterochromatin structure but does not play a role in DNA repair, thus pointing to other exons in the lamin A tail as responsible for the genomic instability phenotype in Lmna(Δ8-11/Δ8-11) mice. Our study also suggests that the levels of DNA repair factors 53BP1, BRCA1 and RAD51 could potentially serve as biomarkers to identify laminopathies that present with genomic instability.


Subject(s)
Chromatin/genetics , Exons/genetics , Genomic Instability/genetics , Lamin Type A/genetics , Sequence Deletion/genetics , Telomere/genetics , Animals , BRCA1 Protein/metabolism , Cell Line , Chromatin/chemistry , Chromatin/pathology , Chromosomal Proteins, Non-Histone/metabolism , DNA Repair/genetics , DNA-Binding Proteins/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Heterochromatin/chemistry , Heterochromatin/genetics , Heterochromatin/metabolism , Humans , Mice , Rad51 Recombinase/metabolism , Telomere/pathology , Telomere Shortening/genetics , Tumor Suppressor p53-Binding Protein 1
3.
Cell Cycle ; 12(23): 3629-39, 2013 Dec 01.
Article in English | MEDLINE | ID: mdl-24091731

ABSTRACT

BRCA1 and 53BP1 play decisive roles in the choice of DNA double-strand break repair mechanisms. BRCA1 promotes DNA end resection and homologous recombination (HR) during S/G 2 phases of the cell cycle, while 53BP1 inhibits end resection and facilitates non-homologous end-joining (NHEJ), primarily during G 1. This competitive relationship is critical for genome integrity during cell division. However, their relationship in the many cells in our body that are not cycling is unknown. We discovered profound differences in 53BP1 and BRCA1 regulation between cycling and non-cycling cells. Cellular growth arrest results in transcriptional downregulation of BRCA1 and activation of cathepsin-L (CTSL)-mediated degradation of 53BP1. Accordingly, growth-arrested cells do not form BRCA1 or 53BP1 ionizing radiation-induced foci (IRIF). Interestingly, cell cycle re-entry reverts this scenario, with upregulation of BRCA1, downregulation of CTSL, stabilization of 53BP1, and 53BP1 IRIF formation throughout the cycle, indicating that BRCA1 and 53BP1 are important in replicating cells and dispensable in non-cycling cells. We show that CTSL-mediated degradation of 53BP1, previously associated with aggressive breast cancers, is an endogenous mechanism of non-cycling cells to balance NHEJ (53BP1) and HR (BRCA1). Breast cancer cells exploit this mechanism to ensure genome stability and viability, providing an opportunity for targeted therapy.


Subject(s)
BRCA1 Protein/metabolism , Gene Expression Regulation , Intracellular Signaling Peptides and Proteins/metabolism , BRCA1 Protein/genetics , Cathepsin L/antagonists & inhibitors , Cathepsin L/genetics , Cathepsin L/metabolism , Cell Cycle Checkpoints/radiation effects , Cell Line , DNA Breaks, Double-Stranded/radiation effects , DNA End-Joining Repair , DNA Replication/drug effects , HeLa Cells , Humans , Hydroxyurea/pharmacology , Intracellular Signaling Peptides and Proteins/genetics , Leucine/analogs & derivatives , Leucine/pharmacology , MCF-7 Cells , RNA Interference , RNA, Small Interfering/metabolism , Radiation, Ionizing , Tumor Suppressor p53-Binding Protein 1
4.
J Cell Biol ; 200(2): 187-202, 2013 Jan 21.
Article in English | MEDLINE | ID: mdl-23337117

ABSTRACT

Loss of 53BP1 rescues BRCA1 deficiency and is associated with BRCA1-deficient and triple-negative breast cancers (TNBC) and with resistance to genotoxic drugs. The mechanisms responsible for decreased 53BP1 transcript and protein levels in tumors remain unknown. Here, we demonstrate that BRCA1 loss activates cathepsin L (CTSL)-mediated degradation of 53BP1. Activation of this pathway rescued homologous recombination repair and allowed BRCA1-deficient cells to bypass growth arrest. Importantly, depletion or inhibition of CTSL with vitamin D or specific inhibitors stabilized 53BP1 and increased genomic instability in response to radiation and poly(adenosine diphosphate-ribose) polymerase inhibitors, compromising proliferation. Analysis of human breast tumors identified nuclear CTSL as a positive biomarker for TNBC, which correlated inversely with 53BP1. Importantly, nuclear levels of CTSL, vitamin D receptor, and 53BP1 emerged as a novel triple biomarker signature for stratification of patients with BRCA1-mutated tumors and TNBC, with potential predictive value for drug response. We identify here a novel pathway with prospective relevance for diagnosis and customization of breast cancer therapy.


Subject(s)
BRCA1 Protein/genetics , Breast Neoplasms/genetics , Cathepsin L/physiology , Intracellular Signaling Peptides and Proteins/metabolism , Cathepsin L/genetics , Cell Line, Tumor , DNA Repair/genetics , Female , Gene Expression Regulation, Neoplastic , Genomic Instability , Germ-Line Mutation , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/physiology , Tumor Suppressor p53-Binding Protein 1
5.
EMBO J ; 30(16): 3383-96, 2011 Jul 12.
Article in English | MEDLINE | ID: mdl-21750527

ABSTRACT

Genomic instability due to telomere dysfunction and defective repair of DNA double-strand breaks (DSBs) is an underlying cause of ageing-related diseases. 53BP1 is a key factor in DNA DSBs repair and its deficiency is associated with genomic instability and cancer progression. Here, we uncover a novel pathway regulating the stability of 53BP1. We demonstrate an unprecedented role for the cysteine protease Cathepsin L (CTSL) in the degradation of 53BP1. Overexpression of CTSL in wild-type fibroblasts leads to decreased 53BP1 protein levels and changes in its cellular distribution, resulting in defective repair of DNA DSBs. Importantly, we show that the defects in DNA repair associated with 53BP1 deficiency upon loss of A-type lamins are due to upregulation of CTSL. Furthermore, we demonstrate that treatment with vitamin D stabilizes 53BP1 and promotes DNA DSBs repair via inhibition of CTSL, providing an as yet unsuspected link between vitamin D action and DNA repair. Given that CTSL upregulation is a hallmark of cancer and progeria, regulation of this pathway could be of great therapeutic significance for these diseases.


Subject(s)
Cathepsin L/physiology , Chromosomal Proteins, Non-Histone/metabolism , DNA Repair/physiology , DNA-Binding Proteins/metabolism , Lamin Type A/physiology , Vitamin D/physiology , Animals , Calcitriol/pharmacology , Cathepsin L/antagonists & inhibitors , Cathepsin L/biosynthesis , Cathepsin L/genetics , Cell Line , Enzyme Precursors/metabolism , Fibroblasts/drug effects , Fibroblasts/metabolism , Humans , Lamin Type A/deficiency , Lamin Type A/genetics , Leupeptins/pharmacology , Mice , Proteasome Endopeptidase Complex/metabolism , Protein Stability , Recombinant Fusion Proteins/physiology , Species Specificity , Transfection , Tumor Suppressor p53-Binding Protein 1
SELECTION OF CITATIONS
SEARCH DETAIL