Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
Cell ; 177(7): 1797-1813.e18, 2019 06 13.
Article in English | MEDLINE | ID: mdl-31104839

ABSTRACT

Accurate regulation of mRNA termination is required for correct gene expression. Here, we describe a role for SCAF4 and SCAF8 as anti-terminators, suppressing the use of early, alternative polyadenylation (polyA) sites. The SCAF4/8 proteins bind the hyper-phosphorylated RNAPII C-terminal repeat domain (CTD) phosphorylated on both Ser2 and Ser5 and are detected at early, alternative polyA sites. Concomitant knockout of human SCAF4 and SCAF8 results in altered polyA selection and subsequent early termination, leading to expression of truncated mRNAs and proteins lacking functional domains and is cell lethal. While SCAF4 and SCAF8 work redundantly to suppress early mRNA termination, they also have independent, non-essential functions. SCAF8 is an RNAPII elongation factor, whereas SCAF4 is required for correct termination at canonical, distal transcription termination sites in the presence of SCAF8. Together, SCAF4 and SCAF8 coordinate the transition between elongation and termination, ensuring correct polyA site selection and RNAPII transcriptional termination in human cells.


Subject(s)
RNA Polymerase II/metabolism , RNA, Messenger/biosynthesis , RNA-Binding Proteins/metabolism , Serine-Arginine Splicing Factors/metabolism , Transcription Elongation, Genetic , Transcription Termination, Genetic , HEK293 Cells , Humans , Poly A/genetics , Poly A/metabolism , Protein Domains , RNA Polymerase II/genetics , RNA, Messenger/genetics , RNA-Binding Proteins/genetics , Serine-Arginine Splicing Factors/genetics
2.
EMBO J ; 41(21): e110727, 2022 11 02.
Article in English | MEDLINE | ID: mdl-36124427

ABSTRACT

Better understanding on interactions between SARS-CoV-2 and host cells should help to identify host factors that may be targetable to combat infection and COVID-19 pathology. To this end, we have conducted a genome-wide CRISPR/Cas9-based loss-of-function screen in human lung cancer cells infected with SARS-CoV-2-pseudotyped lentiviruses. Our results recapitulate many findings from previous screens that used full SARS-CoV-2 viruses, but also unveil two novel critical host factors: the lysosomal efflux transporter SPNS1 and the plasma and lysosomal membrane protein PLAC8. Functional experiments with full SARS-CoV-2 viruses confirm that loss-of-function of these genes impairs viral entry. We find that PLAC8 is a key limiting host factor, whose overexpression boosts viral infection in eight different human lung cancer cell lines. Using single-cell RNA-Seq data analyses, we demonstrate that PLAC8 is highly expressed in ciliated and secretory cells of the respiratory tract, as well as in gut enterocytes, cell types that are highly susceptible to SARS-CoV-2 infection. Proteomics and cell biology studies suggest that PLAC8 and SPNS1 regulate the autophagolysosomal compartment and affect the intracellular fate of endocytosed virions.


Subject(s)
COVID-19 , Lung Neoplasms , Humans , SARS-CoV-2 , Angiotensin-Converting Enzyme 2 , Lysosomal Membrane Proteins , Autophagy , Proteins
3.
Genes Dev ; 28(7): 765-82, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-24636986

ABSTRACT

The mammary gland is a very dynamic organ that undergoes continuous remodeling. The critical regulators of this process are not fully understood. Here we identify the microRNA cluster miR-424(322)/503 as an important regulator of epithelial involution after pregnancy. Through the generation of a knockout mouse model, we found that regression of the secretory acini of the mammary gland was compromised in the absence of miR-424(322)/503. Mechanistically, we show that miR-424(322)/503 orchestrates cell life and death decisions by targeting BCL-2 and IGF1R (insulin growth factor-1 receptor). Furthermore, we demonstrate that the expression of this microRNA cluster is regulated by TGF-ß, a well-characterized regulator of mammary involution. Overall, our data suggest a model in which activation of the TGF-ß pathway after weaning induces the transcription of miR-424(322)/503, which in turn down-regulates the expression of key genes. Here, we unveil a previously unknown, multilayered regulation of epithelial tissue remodeling coordinated by the microRNA cluster miR-424(322)/503.


Subject(s)
Epithelium/metabolism , Gene Expression Regulation, Developmental , Mammary Glands, Animal/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Animals , Cell Death/genetics , Cell Line , Female , Gene Knockout Techniques , Humans , Mammary Glands, Animal/cytology , Mice, Knockout , Promoter Regions, Genetic/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Receptor, IGF Type 1/metabolism , Signal Transduction , Transforming Growth Factor beta1/metabolism , Weaning
4.
PLoS Biol ; 16(10): e2006247, 2018 10.
Article in English | MEDLINE | ID: mdl-30346946

ABSTRACT

Different microRNAs (miRNAs), including miR-29 family, may play a role in the development of heart failure (HF), but the underlying molecular mechanisms in HF pathogenesis remain unclear. We aimed at characterizing mice deficient in miR-29 in order to address the functional relevance of this family of miRNAs in the cardiovascular system and its contribution to heart disease. In this work, we show that mice deficient in miR-29a/b1 develop vascular remodeling and systemic hypertension, as well as HF with preserved ejection fraction (HFpEF) characterized by myocardial fibrosis, diastolic dysfunction, and pulmonary congestion, and die prematurely. We also found evidence that the absence of miR-29 triggers the up-regulation of its target, the master metabolic regulator PGC1α, which in turn generates profound alterations in mitochondrial biogenesis, leading to a pathological accumulation of small mitochondria in mutant animals that contribute to cardiac disease. Notably, we demonstrate that systemic hypertension and HFpEF caused by miR-29 deficiency can be rescued by PGC1α haploinsufficiency, which reduces cardiac mitochondrial accumulation and extends longevity of miR-29-mutant mice. In addition, PGC1α is overexpressed in hearts from patients with HF. Collectively, our findings demonstrate the in vivo role of miR-29 in cardiovascular homeostasis and unveil a novel miR-29/PGC1α regulatory circuitry of functional relevance for cell metabolism under normal and pathological conditions.


Subject(s)
Heart Failure/genetics , MicroRNAs/genetics , MicroRNAs/physiology , Animals , Fibrosis , Heart/physiology , Humans , Hypertension/genetics , Mice , Mice, Inbred C57BL , Mitochondria , Myocardium/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/physiology , Up-Regulation , Vascular Remodeling/genetics
5.
EMBO Rep ; 18(4): 549-557, 2017 04.
Article in English | MEDLINE | ID: mdl-28274951

ABSTRACT

Cancer cells modulate their metabolic networks to support cell proliferation and a higher demand of building blocks. These changes may restrict the availability of certain amino acids for protein synthesis, which can be utilized for cancer therapy. However, little is known about the amino acid demand changes occurring during aggressive and invasive stages of cancer. Recently, we developed diricore, an approach based on ribosome profiling that can uncover amino acid limitations. Here, we applied diricore to a cellular model in which epithelial breast cells respond rapidly to TGFß1, a cytokine essential for cancer progression and metastasis, and uncovered shortage of leucine. Further analyses indicated that TGFß1 treatment of human breast epithelial cells reduces the expression of SLC3A2, a subunit of the leucine transporter, which diminishes leucine uptake and inhibits cell proliferation. Thus, we identified a specific amino acid limitation associated with the TGFß1 response, a vulnerability that might be associated with aggressiveness in cancer.


Subject(s)
Codon , Leucine/genetics , Leucine/metabolism , Protein Biosynthesis , Ribosomes/metabolism , Transforming Growth Factor beta1/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Line, Tumor , Female , Fusion Regulatory Protein 1, Heavy Chain/genetics , Fusion Regulatory Protein 1, Heavy Chain/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Humans , Protein Biosynthesis/drug effects , Signal Transduction , Transforming Growth Factor beta1/pharmacology
6.
Nat Rev Genet ; 14(7): 496-506, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23774734

ABSTRACT

The 3' end of most protein-coding genes and long non-coding RNAs is cleaved and polyadenylated. Recent discoveries have revealed that a large proportion of these genes contains more than one polyadenylation site. Therefore, alternative polyadenylation (APA) is a widespread phenomenon, generating mRNAs with alternative 3' ends. APA contributes to the complexity of the transcriptome by generating isoforms that differ either in their coding sequence or in their 3' untranslated regions (UTRs), thereby potentially regulating the function, stability, localization and translation efficiency of target RNAs. Here, we review our current understanding of the polyadenylation process and the latest progress in the identification of APA events, mechanisms that regulate poly(A) site selection, and biological processes and diseases resulting from APA.


Subject(s)
Gene Expression Regulation , Polyadenylation , 3' Untranslated Regions , Animals , Binding Sites , Cell Differentiation , Chromatin/metabolism , Chromosome Mapping , Expressed Sequence Tags , Genetic Diseases, Inborn/genetics , Humans , Models, Genetic , Oligonucleotide Array Sequence Analysis , Poly A , Protein Isoforms , RNA/genetics , RNA Splicing
7.
PLoS Genet ; 12(2): e1005879, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26908102

ABSTRACT

Most mammalian genes often feature alternative polyadenylation (APA) sites and hence diverse 3'UTR lengths. Proliferating cells were reported to favor APA sites that result in shorter 3'UTRs. One consequence of such shortening is escape of mRNAs from targeting by microRNAs (miRNAs) whose binding sites are eliminated. Such a mechanism might provide proliferation-related genes with an expression gain during normal or cancerous proliferation. Notably, miRNA sites tend to be more active when located near both ends of the 3'UTR compared to those located more centrally. Accordingly, miRNA sites located near the center of the full 3'UTR might become more active upon 3'UTR shortening. To address this conjecture we performed 3' sequencing to determine the 3' ends of all human UTRs in several cell lines. Remarkably, we found that conserved miRNA binding sites are preferentially enriched immediately upstream to APA sites, and this enrichment is more prominent in pro-differentiation/anti-proliferative genes. Binding sites of the miR17-92 cluster, upregulated in rapidly proliferating cells, are particularly enriched just upstream to APA sites, presumably conferring stronger inhibitory activity upon shortening. Thus 3'UTR shortening appears not only to enable escape from inhibition of growth promoting genes but also to potentiate repression of anti-proliferative genes.


Subject(s)
3' Untranslated Regions , Cell Proliferation/genetics , MicroRNAs/genetics , Binding Sites , Cell Line , Gene Expression Regulation, Developmental , Humans , MicroRNAs/metabolism , Polyadenylation , RNA, Long Noncoding
8.
Circ Res ; 118(3): 433-8, 2016 Feb 05.
Article in English | MEDLINE | ID: mdl-26671978

ABSTRACT

RATIONALE: Alternative cleavage and polyadenylation (APA) of mRNA represents a layer of gene regulation that to date has remained unexplored in the heart. This phenomenon may be relevant, as the positioning of the poly(A) tail in mRNAs influences the length of the 3'-untranslated region (UTR), a critical determinant of gene expression. OBJECTIVE: To investigate whether the 3'UTR length is regulated by APA in the human heart and whether this changes in the failing heart. METHODS AND RESULTS: We used 3'end RNA sequencing (e3'-Seq) to directly measure global patterns of APA in healthy and failing human heart specimens. By monitoring polyadenylation profiles in these hearts, we identified disease-specific APA signatures in numerous genes. Interestingly, many of the genes with shortened 3'UTRs in heart failure were enriched for functional groups such as RNA binding, whereas genes with longer 3'UTRs were enriched for cytoskeletal organization and actin binding. RNA sequencing in a larger series of human hearts revealed that these APA candidates are often differentially expressed in failing hearts, with an inverse correlation between 3'UTR length and the level of gene expression. Protein levels of the APA regulator, poly(A)-binding protein nuclear-1 were substantially downregulated in failing hearts. CONCLUSIONS: We provide genome-wide, high-resolution polyadenylation maps of the human heart and show that the 3'end formation of mRNA is dynamic in heart failure, suggesting that APA-mediated 3'UTR length modulation represents an additional layer of gene regulation in failing hearts.


Subject(s)
3' Untranslated Regions , Heart Failure/genetics , Polyadenylation , RNA, Messenger/genetics , Adult , Aged , Base Sequence , Case-Control Studies , Female , Gene Expression Profiling/methods , Gene Expression Regulation , Genome-Wide Association Study , Heart Failure/diagnosis , Heart Failure/metabolism , Humans , Male , Middle Aged , Molecular Sequence Data , Poly(A)-Binding Protein I/metabolism , RNA, Messenger/metabolism
11.
EMBO J ; 30(11): 2219-32, 2011 Jun 01.
Article in English | MEDLINE | ID: mdl-21522133

ABSTRACT

Aging is a multifactorial process that affects most of the biological functions of the organism and increases susceptibility to disease and death. Recent studies with animal models of accelerated aging have unveiled some mechanisms that also operate in physiological aging. However, little is known about the role of microRNAs (miRNAs) in this process. To address this question, we have analysed miRNA levels in Zmpste24-deficient mice, a model of Hutchinson-Gilford progeria syndrome. We have found that expression of the miR-29 family of miRNAs is markedly upregulated in Zmpste24(-/-) progeroid mice as well as during normal aging in mouse. Functional analysis revealed that this transcriptional activation of miR-29 is triggered in response to DNA damage and occurs in a p53-dependent manner since p53(-/-) murine fibroblasts do not increase miR-29 expression upon doxorubicin treatment. We have also found that miR-29 represses Ppm1d phosphatase, which in turn enhances p53 activity. Based on these results, we propose the existence of a novel regulatory circuitry involving miR-29, Ppm1d and p53, which is activated in aging and in response to DNA damage.


Subject(s)
Aging , DNA Damage , Gene Expression Regulation , MicroRNAs/biosynthesis , Phosphoprotein Phosphatases/biosynthesis , Tumor Suppressor Protein p53/biosynthesis , Animals , Cells, Cultured , Disease Models, Animal , Fibroblasts/physiology , Membrane Proteins/deficiency , Metalloendopeptidases/deficiency , Mice , Mice, Knockout , Molecular Sequence Data , Protein Phosphatase 2C , Sequence Analysis, DNA
12.
Article in English | MEDLINE | ID: mdl-38394352

ABSTRACT

Aging is a multifactorial process characterized by an age-related decline in organismal fitness. This deterioration is the major risk factor for chronic diseases such as cardiovascular pathologies, neurodegeneration, or cancer, and it represents one of the main challenges of modern society. Therefore, understanding why and how we age would be a fundamental pillar to design strategies to promote a healthy aging. In the last decades, the study of the molecular bases of disease has been revolutionized by the discovery of different types of noncoding RNAs (ncRNAs) with regulatory potential. In this work, we will review the implication of ncRNAs in aging, with the aim to provide a first approach to the different aging-associated ncRNAs, their mechanism of action, and their potential relevance as therapeutic targets and disease biomarkers.


Subject(s)
Longevity , MicroRNAs , Longevity/genetics , RNA, Untranslated/genetics , MicroRNAs/genetics
13.
Proc Natl Acad Sci U S A ; 107(37): 16268-73, 2010 Sep 14.
Article in English | MEDLINE | ID: mdl-20805469

ABSTRACT

Zmpste24 (also called FACE-1) is a metalloproteinase involved in the maturation of lamin A, an essential component of the nuclear envelope. Zmpste24-deficient mice exhibit multiple defects that phenocopy human accelerated aging processes such as Hutchinson-Gilford progeria syndrome. In this work, we report that progeroid Zmpste24(-/-) mice present profound transcriptional alterations in genes that regulate the somatotroph axis, together with extremely high circulating levels of growth hormone (GH) and a drastic reduction in plasma insulin-like growth factor 1 (IGF-1). We also show that recombinant IGF-1 treatment restores the proper balance between IGF-1 and GH in Zmpste24(-/-) mice, delays the onset of many progeroid features, and significantly extends the lifespan of these progeroid animals. Our findings highlight the importance of IGF/GH balance in longevity and may be of therapeutic interest for devastating human progeroid syndromes associated with nuclear envelope abnormalities.


Subject(s)
Aging, Premature/drug therapy , Insulin-Like Growth Factor I/therapeutic use , Longevity/drug effects , Somatotrophs/drug effects , Aging, Premature/blood , Animals , Base Sequence , Disease Models, Animal , Gene Expression Profiling , Gene Expression Regulation , Growth Hormone/blood , Insulin-Like Growth Factor I/genetics , Insulin-Like Growth Factor I/metabolism , Liver/metabolism , Membrane Proteins/deficiency , Membrane Proteins/metabolism , Metalloendopeptidases/deficiency , Metalloendopeptidases/metabolism , Mice , Mice, Knockout , MicroRNAs/genetics
14.
Biochem Soc Trans ; 39(6): 1710-4, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22103512

ABSTRACT

Progeroid laminopathies are accelerated aging syndromes caused by defects in nuclear envelope proteins. Accordingly, mutations in the LMNA gene and functionally related genes have been described to cause HGPS (Hutchinson-Gilford progeria syndrome), MAD (mandibuloacral dysplasia) or RD (restrictive dermopathy). Functional studies with animal and cellular models of these syndromes have facilitated the identification of the molecular alterations and regulatory pathways involved in progeria development. We have recently described a novel regulatory pathway involving miR-29 and p53 tumour suppressor which has provided valuable information on the molecular components orchestrating the response to nuclear damage stress. Furthermore, by using progeroid mice deficient in ZMPSTE24 (zinc metalloprotease STE24 homologue) involved in lamin A maturation, we have demonstrated that, besides these abnormal cellular responses to stress, dysregulation of the somatotropic axis is responsible for some of the alterations associated with progeria. Consistent with these observations, pharmacological restoration of the somatotroph axis in these mice delays the onset of their progeroid features, significantly extending their lifespan and supporting the importance of systemic alterations in progeria progression. Finally, we have very recently identified a novel progeroid syndrome with distinctive features from HGPS and MAD, which we have designated NGPS (Néstor-Guillermo progeria syndrome) (OMIM #614008). This disorder is caused by a mutation in BANF1, a gene encoding a protein with essential functions in the assembly of the nuclear envelope, further illustrating the importance of the nuclear lamina integrity for human health and providing additional support to the study of progeroid syndromes as a valuable source of information on human aging.


Subject(s)
Progeria/metabolism , Progeria/pathology , Animals , DNA Damage , Humans , Membrane Proteins/metabolism , MicroRNAs/metabolism , Progeria/genetics , Somatotrophs/metabolism , Somatotrophs/pathology , Tumor Suppressor Protein p53/metabolism
15.
Genome Biol ; 22(1): 54, 2021 01 29.
Article in English | MEDLINE | ID: mdl-33514403

ABSTRACT

BACKGROUND: Frequent activation of the co-transcriptional factor YAP is observed in a large number of solid tumors. Activated YAP associates with enhancer loci via TEAD4-DNA-binding protein and stimulates cancer aggressiveness. Although thousands of YAP/TEAD4 binding-sites are annotated, their functional importance is unknown. Here, we aim at further identification of enhancer elements that are required for YAP functions. RESULTS: We first apply genome-wide ChIP profiling of YAP to systematically identify enhancers that are bound by YAP/TEAD4. Next, we implement a genetic approach to uncover functions of YAP/TEAD4-associated enhancers, demonstrate its robustness, and use it to reveal a network of enhancers required for YAP-mediated proliferation. We focus on EnhancerTRAM2, as its target gene TRAM2 shows the strongest expression-correlation with YAP activity in nearly all tumor types. Interestingly, TRAM2 phenocopies the YAP-induced cell proliferation, migration, and invasion phenotypes and correlates with poor patient survival. Mechanistically, we identify FSTL-1 as a major direct client of TRAM2 that is involved in these phenotypes. Thus, TRAM2 is a key novel mediator of YAP-induced oncogenic proliferation and cellular invasiveness. CONCLUSIONS: YAP is a transcription co-factor that binds to thousands of enhancer loci and stimulates tumor aggressiveness. Using unbiased functional approaches, we dissect YAP enhancer network and characterize TRAM2 as a novel mediator of cellular proliferation, migration, and invasion. Our findings elucidate how YAP induces cancer aggressiveness and may assist diagnosis of cancer metastasis.


Subject(s)
Carcinogenesis/genetics , Enhancer Elements, Genetic , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Animals , Binding Sites , Cell Line, Tumor , Cell Movement , Cell Proliferation , DNA-Binding Proteins/metabolism , Female , Gene Expression Regulation, Neoplastic , Humans , Membrane Glycoproteins/chemistry , Mice , Mice, Inbred NOD , Mice, SCID , TEA Domain Transcription Factors/genetics , TEA Domain Transcription Factors/metabolism , Transcription Factors/metabolism , Transcriptome
16.
Hum Mol Genet ; 17(14): 2196-211, 2008 Jul 15.
Article in English | MEDLINE | ID: mdl-18443001

ABSTRACT

Autophagy is a highly regulated intracellular process involved in the turnover of most cellular constituents and in the maintenance of cellular homeostasis. It is well-established that the basal autophagic activity of living cells decreases with age, thus contributing to the accumulation of damaged macromolecules during aging. Conversely, the activity of this catabolic pathway is required for lifespan extension in animal models such as Caenorhabditis elegans and Drosophila melanogaster. In this work, we describe the unexpected finding that Zmpste24-null mice, which show accelerated aging and are a reliable model of human Hutchinson-Gilford progeria, exhibit an extensive basal activation of autophagy instead of the characteristic decline in this process occurring during normal aging. We also show that this autophagic increase is associated with a series of changes in lipid and glucose metabolic pathways, which resemble those occurring in diverse situations reported to prolong lifespan. These Zmpste24(-/-) mice metabolic alterations are also linked to substantial changes in circulating blood parameters, such as leptin, glucose, insulin or adiponectin which in turn lead to peripheral LKB1-AMPK activation and mTOR inhibition. On the basis of these results, we propose that nuclear abnormalities causing premature aging in Zmpste24(-/-) mice trigger a metabolic response involving the activation of autophagy. However, the chronic activation of this catabolic pathway may turn an originally intended pro-survival strategy into a pro-aging mechanism and could contribute to the systemic degeneration and weakening observed in these progeroid mice.


Subject(s)
Aging, Premature/physiopathology , Autophagy , Membrane Proteins/metabolism , Metalloendopeptidases/metabolism , Progeria/physiopathology , AMP-Activated Protein Kinase Kinases , AMP-Activated Protein Kinases , Aging, Premature/genetics , Animals , Disease Models, Animal , Glucose/metabolism , Hormones/blood , Humans , Lipid Metabolism , Membrane Proteins/genetics , Metalloendopeptidases/genetics , Mice , Mice, Transgenic , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Nuclear Receptor Subfamily 1, Group F, Member 3 , Progeria/genetics , Protein Kinases/genetics , Protein Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Receptors, Retinoic Acid/genetics , Receptors, Retinoic Acid/metabolism , Receptors, Thyroid Hormone/genetics , Receptors, Thyroid Hormone/metabolism , Signal Transduction
17.
Nat Biotechnol ; 34(2): 192-8, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26751173

ABSTRACT

Systematic identification of noncoding regulatory elements has, to date, mainly relied on large-scale reporter assays that do not reproduce endogenous conditions. We present two distinct CRISPR-Cas9 genetic screens to identify and characterize functional enhancers in their native context. Our strategy is to target Cas9 to transcription factor binding sites in enhancer regions. We identified several functional enhancer elements and characterized the role of two of them in mediating p53 (TP53) and ERα (ESR1) gene regulation. Moreover, we show that a genomic CRISPR-Cas9 tiling screen can precisely map functional domains within enhancer elements. Our approach expands the utility of CRISPR-Cas9 to elucidate the functions of the noncoding genome.


Subject(s)
CRISPR-Cas Systems/genetics , Enhancer Elements, Genetic/genetics , Genetic Engineering/methods , Genome, Human/genetics , Genomics/methods , Animals , Cell Line , Gene Knockout Techniques , Humans , MCF-7 Cells , Mice
18.
Nat Commun ; 4: 2268, 2013.
Article in English | MEDLINE | ID: mdl-23917225

ABSTRACT

Defining the relationship between ageing and cancer is a crucial but challenging task. Mice deficient in Zmpste24, a metalloproteinase mutated in human progeria and involved in nuclear prelamin A maturation, recapitulate multiple features of ageing. However, their short lifespan and serious cell-intrinsic and cell-extrinsic alterations restrict the application and interpretation of carcinogenesis protocols. Here we present Zmpste24 mosaic mice that lack these limitations. Zmpste24 mosaic mice develop normally and keep similar proportions of Zmpste24-deficient (prelamin A-accumulating) and Zmpste24-proficient (mature lamin A-containing) cells throughout life, revealing that cell-extrinsic mechanisms are preeminent for progeria development. Moreover, prelamin A accumulation does not impair tumour initiation and growth, but it decreases the incidence of infiltrating oral carcinomas. Accordingly, silencing of ZMPSTE24 reduces human cancer cell invasiveness. Our results support the potential of cell-based and systemic therapies for progeria and highlight ZMPSTE24 as a new anticancer target.


Subject(s)
Neoplasms/pathology , Nuclear Proteins/metabolism , Progeria/metabolism , Progeria/pathology , Protein Precursors/metabolism , Aging/pathology , Animals , Biomarkers/metabolism , Carcinogenesis/pathology , Female , Humans , Lamin Type A , Male , Membrane Proteins/deficiency , Membrane Proteins/metabolism , Metalloendopeptidases/deficiency , Metalloendopeptidases/metabolism , Mice , Mosaicism , Neoplasm Invasiveness , Neoplasms/metabolism , Phenotype
19.
Nucleus ; 2(6): 549-55, 2011.
Article in English | MEDLINE | ID: mdl-22064465

ABSTRACT

Over the last years, the discovery of microRNAs (miRNAs) has revolutionized the classic concepts of gene expression regulation and has introduced a new group of molecules that may contribute to the complex changes observed during aging. Although several Caenorhabditis elegans miRNAs have been proved to influence the nematode life span, the current knowledge about miRNA-mediated regulation of mammalian aging is still limited. Recently, we have analyzed the functional relevance of miRNAs in accelerate aging by using Zmpste24-/- mice, a murine model that phenocopies Hutchinson-Gilford progeria syndrome. These studies have revealed that the nuclear abnormalities present in these mice affect the expression levels of several miRNAs, including a marked upregulation of miR-1 and miR-29. Furthermore, we have found that the altered expression of these miRNAs may contribute to the progeroid phenotype of mutant mice by modulating the levels of key components of the somatroph axis and DNA damage response pathways. Here, we discuss these recent discoveries and summarize the present evidences regarding the involvement of aging-associated miRNAs or geromiRs in senescence and longevity regulation.


Subject(s)
Longevity/physiology , MicroRNAs/metabolism , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , DNA Damage/genetics , Disease Models, Animal , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , Metalloendopeptidases/genetics , Metalloendopeptidases/metabolism , Mice , MicroRNAs/genetics , Progeria/genetics , Progeria/metabolism , RNA, Helminth/genetics , RNA, Helminth/metabolism
20.
Aging (Albany NY) ; 2(12): 1017-22, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21212467

ABSTRACT

We have recently reported that progeroid Zmpste24-/- mice, which exhibit multiple defects that phenocopy Hutchinson-Gilford progeria syndrome, show a profound dysregulation of somatotropic axis, mainly characterized by the occurrence of very high circulating levels of growth hormone (GH) and a drastic reduction in insulin-like growth factor-1 (IGF-1). We have also shown that restoration of the proper GH/IGF-1 balance in Zmpste24-/- mice by treatment with recombinant IGF-1 delays the onset of many progeroid features in these animals and significantly extends their lifespan. Here, we summarize these observations and discuss the importance of GH/IGF-1 balance in longevity as well as its modulation as a putative therapeutic strategy for the treatment of human progeroid syndromes.


Subject(s)
Aging, Premature/metabolism , Growth Hormone/metabolism , Insulin-Like Growth Factor I/metabolism , Progeria/metabolism , Rejuvenation , Signal Transduction , Aging, Premature/drug therapy , Aging, Premature/genetics , Animals , Genotype , Humans , Insulin-Like Growth Factor I/therapeutic use , Longevity , Membrane Proteins/deficiency , Membrane Proteins/genetics , Metalloendopeptidases/deficiency , Metalloendopeptidases/genetics , Mice , Mice, Knockout , Phenotype , Progeria/drug therapy , Progeria/genetics , Recombinant Proteins/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL