Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Cells ; 9(2)2020 02 08.
Article in English | MEDLINE | ID: mdl-32046343

ABSTRACT

Hutchinson-Gilford Progeria Syndrome (HGPS) is a segmental premature aging disease causing patient death by early teenage years from cardiovascular dysfunction. Although HGPS does not totally recapitulate normal aging, it does harbor many similarities to the normal aging process, with patients also developing cardiovascular disease, alopecia, bone and joint abnormalities, and adipose changes. It is unsurprising, then, that as physicians and scientists have searched for treatments for HGPS, they have targeted many pathways known to be involved in normal aging, including inflammation, DNA damage, epigenetic changes, and stem cell exhaustion. Although less studied at a mechanistic level, severe metabolic problems are observed in HGPS patients. Interestingly, new research in animal models of HGPS has demonstrated impressive lifespan improvements secondary to metabolic interventions. As such, further understanding metabolism, its contribution to HGPS, and its therapeutic potential has far-reaching ramifications for this disease still lacking a robust treatment strategy.


Subject(s)
Progeria/metabolism , Progeria/physiopathology , Animals , Disease Models, Animal , Energy Metabolism , Humans , Progeria/therapy
2.
Subcell Biochem ; 91: 435-451, 2019.
Article in English | MEDLINE | ID: mdl-30888661

ABSTRACT

The structural nuclear proteins known as "lamins" (A-type and B-type) provide a scaffold for the compartmentalization of genome function that is important to maintain genome stability. Mutations in the LMNA gene -encoding for A-type lamins- are associated with over a dozen of degenerative disorders termed laminopathies, which include muscular dystrophies, lipodystrophies, neuropathies, and premature ageing diseases such as Hutchinson Gilford Progeria Syndrome (HGPS). This devastating disease is caused by the expression of a truncated lamin A protein named "progerin". To date, there is no effective treatment for HGPS patients, who die in their teens from cardiovascular disease. At a cellular level, progerin expression impacts nuclear architecture, chromatin organization, response to mechanical stress, and DNA transactions such as transcription, replication and repair. However, the current view is that key mechanisms behind progerin toxicity still remain to be discovered. Here, we discuss new findings about pathological mechanisms in HGPS, especially the contribution of replication stress to cellular decline, and therapeutic strategies to ameliorate progerin toxicity. In particular, we present evidence for retinoids and calcitriol (hormonal vitamin D metabolite) being among the most potent compounds to ameliorate HGPS cellular phenotypes in vitro, providing the rationale for testing these compounds in preclinical models of the disease in the near term, and in patients in the future.


Subject(s)
Progeria/pathology , Progeria/therapy , Aging, Premature/pathology , Aging, Premature/therapy , Calcitriol/metabolism , Cell Nucleus/metabolism , Cell Nucleus/pathology , Humans , Lamin Type A/metabolism , Retinoids/metabolism
3.
Aging Cell ; 18(1): e12852, 2019 02.
Article in English | MEDLINE | ID: mdl-30548460

ABSTRACT

Hutchinson-Gilford Progeria Syndrome (HGPS) is a devastating premature aging disease. Mouse models have been instrumental for understanding HGPS mechanisms and for testing therapies, which to date have had only marginal benefits in mice and patients. Barriers to developing effective therapies include the unknown etiology of progeria mice early death, seemingly unrelated to the reported atherosclerosis contributing to HGPS patient mortality, and mice not recapitulating the severity of human disease. Here, we show that progeria mice die from starvation and cachexia. Switching progeria mice approaching death from regular diet to high-fat diet (HFD) rescues early lethality and ameliorates morbidity. Critically, feeding the mice only HFD delays aging and nearly doubles lifespan, which is the greatest lifespan extension recorded in progeria mice. The extended lifespan allows for progeria mice to develop degenerative aging pathologies of a severity that emulates the human disease. We propose that starvation and cachexia greatly influence progeria phenotypes and that nutritional/nutraceutical strategies might help modulate disease progression. Importantly, progeria mice on HFD provide a more clinically relevant animal model to study mechanisms of HGPS pathology and to test therapies.


Subject(s)
Feeding Behavior , Longevity , Progeria/pathology , Animals , Diet, High-Fat , Humans , Lamin Type A/genetics , Lamin Type A/metabolism , Mice, Inbred C57BL , Mutation/genetics , Phenotype , Progeria/metabolism
4.
Nucleus ; 9(1): 258-275, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29637811

ABSTRACT

Mammalian nuclei are equipped with a framework of intermediate filaments that function as a karyoskeleton. This nuclear scaffold, formed primarily by lamins (A-type and B-type), maintains the spatial and functional organization of the genome and of sub-nuclear compartments. Over the past decade, a body of evidence has highlighted the significance of these structural nuclear proteins in the maintenance of nuclear architecture and mechanical stability, as well as genome function and integrity. The importance of these structures is now unquestioned given the wide range of degenerative diseases that stem from LMNA gene mutations, including muscular dystrophy disorders, peripheral neuropathies, lipodystrophies, and premature aging syndromes. Here, we review our knowledge about how alterations in nuclear lamins, either by mutation or reduced expression, impact cellular mechanisms that maintain genome integrity. Despite the fact that DNA replication is the major source of DNA damage and genomic instability in dividing cells, how alterations in lamins function impact replication remains minimally explored. We summarize recent studies showing that lamins play a role in DNA replication, and that the DNA damage that accumulates upon lamins dysfunction is elicited in part by deprotection of replication forks. We also discuss the emerging model that DNA damage and replication stress are "sensed" at the cytoplasm by proteins that normally survey this space in search of foreign nucleic acids. In turn, these cytosolic sensors activate innate immune responses, which are materializing as important players in aging and cancer, as well as in the response to cancer immunotherapy.


Subject(s)
DNA/genetics , Genomic Instability , Interferons/genetics , Lamins/genetics , Animals , DNA Damage , DNA Replication , Humans , Interferons/metabolism , Lamins/metabolism , Nuclear Lamina/chemistry , Nuclear Lamina/metabolism
5.
Cell Rep ; 22(8): 2006-2015, 2018 02 20.
Article in English | MEDLINE | ID: mdl-29466729

ABSTRACT

Hutchinson-Gilford progeria syndrome (HGPS) is a premature aging disease caused by a truncated lamin A protein (progerin) that drives cellular and organismal decline. HGPS patient-derived fibroblasts accumulate genomic instability, but its underlying mechanisms and contribution to disease remain poorly understood. Here, we show that progerin-induced replication stress (RS) drives genomic instability by eliciting replication fork (RF) stalling and nuclease-mediated degradation. Rampant RS is accompanied by upregulation of the cGAS/STING cytosolic DNA sensing pathway and activation of a robust STAT1-regulated interferon (IFN)-like response. Reducing RS and the IFN-like response, especially with calcitriol, improves the fitness of progeria cells and increases the efficiency of cellular reprogramming. Importantly, other compounds that improve HGPS phenotypes reduce RS and the IFN-like response. Our study reveals mechanisms underlying progerin toxicity, including RS-induced genomic instability and activation of IFN-like responses, and their relevance for cellular decline in HGPS.


Subject(s)
DNA Replication , Interferons/metabolism , Lamin Type A/metabolism , Stress, Physiological , Animals , Calcitriol/pharmacology , Cytosol/metabolism , DNA/metabolism , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/pathology , Humans , Mice , Phenotype , Progeria/metabolism , Receptors, Pattern Recognition/metabolism , STAT1 Transcription Factor/metabolism
6.
Ageing Res Rev ; 33: 18-29, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27374873

ABSTRACT

Products of the LMNA gene, primarily lamin A and C, are key components of the nuclear lamina, a proteinaceous meshwork that underlies the inner nuclear membrane and is essential for proper nuclear architecture. Alterations in lamin A and C that disrupt the integrity of the nuclear lamina affect a whole repertoire of nuclear functions, causing cellular decline. In humans, hundreds of mutations in the LMNA gene have been identified and correlated with over a dozen degenerative disorders, referred to as laminopathies. These diseases include neuropathies, muscular dystrophies, lipodystrophies, and premature aging diseases. This review focuses on one of the most severe laminopathies, Hutchinson-Gilford Progeria Syndrome (HGPS), which is caused by aberrant splicing of the LMNA gene and expression of a mutant product called progerin. Here, we discuss current views about the molecular mechanisms that contribute to the pathophysiology of this devastating disease, as well as the strategies being tested in vitro and in vivo to counteract progerin toxicity. In particular, progerin accumulation elicits nuclear morphological abnormalities, misregulated gene expression, defects in DNA repair, telomere shortening, and genomic instability, all of which limit cellular proliferative capacity. In patients harboring this mutation, a severe premature aging disease develops during childhood. Interestingly, progerin is also produced in senescent cells and cells from old individuals, suggesting that progerin accumulation might be a factor in physiological aging. Deciphering the molecular mechanisms whereby progerin expression leads to HGPS is an emergent area of research, which could bring us closer to understanding the pathology of aging.


Subject(s)
Aging, Premature/genetics , Aging/physiology , Lamin Type A/genetics , Progeria , Cellular Senescence/physiology , DNA Repair , Disease Management , Genomic Instability , Humans , Mutation , Progeria/genetics , Progeria/physiopathology
7.
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
8.
Methods Mol Biol ; 1411: 419-37, 2016.
Article in English | MEDLINE | ID: mdl-27147057

ABSTRACT

The organization of the genome within the nuclear space is viewed as an additional level of regulation of genome function, as well as a means to ensure genome integrity. Structural proteins associated with the nuclear envelope, in particular lamins (A- and B-type) and lamin-associated proteins, play an important role in genome organization. Interestingly, there is a whole body of evidence that links disruptions of the nuclear lamina with DNA repair defects and genomic instability. Here, we describe a few standard techniques that have been successfully utilized to identify mechanisms behind DNA repair defects and genomic instability in cells with an altered nuclear lamina. In particular, we describe protocols to monitor changes in the expression of DNA repair factors (Western blot) and their recruitment to sites of DNA damage (immunofluorescence); kinetics of DNA double-strand break repair after ionizing radiation (neutral comet assays); frequency of chromosomal aberrations (FISH, fluorescence in situ hybridization); and alterations in telomere homeostasis (Quantitative-FISH). These techniques have allowed us to shed some light onto molecular mechanisms by which alterations in A-type lamins induce genomic instability, which could contribute to the pathophysiology of aging and aging-related diseases.


Subject(s)
DNA Repair , Genomic Instability , Genomics , Nuclear Lamina/metabolism , Blotting, Western , Comet Assay , DNA Damage , Genomics/methods , In Situ Hybridization, Fluorescence , Microscopy, Fluorescence
9.
Curr Opin Cell Biol ; 34: 75-83, 2015 Jun.
Article in English | MEDLINE | ID: mdl-26079711

ABSTRACT

The integrity of the nuclear lamina has emerged as an important factor in the maintenance of genome stability. In particular, mutations in the LMNA gene, encoding A-type lamins (lamin A/C), alter nuclear morphology and function, and cause genomic instability. LMNA gene mutations are associated with a variety of degenerative diseases and devastating premature aging syndromes such as Hutchinson-Gilford Progeria Syndrome (HGPS) and Restrictive Dermopathy (RD). HGPS is a severe laminopathy, with patients dying in their teens from myocardial infarction or stroke. HGPS patient-derived cells exhibit nuclear shape abnormalities, changes in epigenetic regulation and gene expression, telomere shortening, genome instability, and premature senescence. This review highlights recent advances in identifying molecular mechanisms that contribute to the pathophysiology of HGPS, with a special emphasis on DNA repair defects and genome instability.


Subject(s)
DNA Repair , Genomic Instability , Progeria/genetics , Animals , Epigenesis, Genetic , Genome, Human , Humans , Oxidative Stress
10.
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
11.
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
SELECTION OF CITATIONS
SEARCH DETAIL