Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 54
1.
Article En | MEDLINE | ID: mdl-38394352

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.


Longevity , MicroRNAs , Longevity/genetics , RNA, Untranslated/genetics , MicroRNAs/genetics
2.
Methods Cell Biol ; 181: 73-85, 2024.
Article En | MEDLINE | ID: mdl-38302245

The gut microbiota is a complex community of different microbial species that influence many aspects of health. Consequently, shifts in the composition of gut microbiome have been proposed to exert negative effects on the host physiology, leading to the pathogenesis of various age-related disorders, including cardiovascular and neurological diseases, type 2 diabetes, obesity, non-alcoholic liver disease, and other pathological conditions. Thus, understanding how the gut microbiota influences the aging-related decline is particularly topical. Advances in next-generation sequencing techniques, together with mechanistic experiments in animal models, have provided substantial improvements in microbiome analysis. However, standardization and best practices are needed to limit experimental variation between different studies. Here, we detail a simple method for microbiota composition analysis in mouse fecal samples using 16S rRNA next-generation sequencing.


Diabetes Mellitus, Type 2 , Gastrointestinal Microbiome , Microbiota , Mice , Animals , RNA, Ribosomal, 16S/genetics , Microbiota/genetics , Feces , Gastrointestinal Microbiome/genetics
3.
Front Aging ; 5: 1334261, 2024.
Article En | MEDLINE | ID: mdl-38292053

The inexorability of the aging process has sparked the curiosity of human beings since ancient times. However, despite this interest and the extraordinary scientific advances in the field, the complexity of the process has hampered its comprehension. In this context, The Hallmarks of Aging were defined in 2013 with the aim of establishing an organized, systematic and integrative view of this topic, which would serve as a conceptual framework for aging research. Ten years later and promoted by the progress in the area, an updated version included three new hallmarks while maintaining the original scope. The aim of this review is to determine to what extent The Hallmarks of Aging achieved the purpose that gave rise to them. For this aim, we have reviewed the literature citing any of the two versions of The Hallmarks of Aging and conclude that they have served as a conceptual framework not only for aging research but also for related areas of knowledge. Finally, this review discusses the new candidates to become part of the Hallmarks list, analyzing the evidence that supports whether they should or should not be incorporated.

4.
Cell Death Dis ; 14(1): 60, 2023 01 26.
Article En | MEDLINE | ID: mdl-36702832

The spindle assembly checkpoint (SAC) is an essential mechanism that ensures the accurate chromosome segregation during mitosis, thus preventing genomic instability. Deubiquitinases have emerged as key regulators of the SAC, mainly by determining the fate of proteins during cell cycle progression. Here, we identify USP49 deubiquitinase as a novel regulator of the spindle checkpoint. We show that loss of USP49 in different cancer cell lines impairs proliferation and increases aneuploidy. In addition, USP49-depleted cells overcome the arrest induced by the SAC in the presence of nocodazole. Finally, we report new binding partners of USP49, including ribophorin 1, USP44, and different centrins.


M Phase Cell Cycle Checkpoints , Spindle Apparatus , Humans , Spindle Apparatus/metabolism , Aneuploidy , Mitosis , Deubiquitinating Enzymes/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Ubiquitin Thiolesterase/genetics , Ubiquitin Thiolesterase/metabolism
5.
Nat Med ; 25(8): 1234-1242, 2019 08.
Article En | MEDLINE | ID: mdl-31332389

The gut microbiome is emerging as a key regulator of several metabolic, immune and neuroendocrine pathways1,2. Gut microbiome deregulation has been implicated in major conditions such as obesity, type 2 diabetes, cardiovascular disease, non-alcoholic fatty acid liver disease and cancer3-6, but its precise role in aging remains to be elucidated. Here, we find that two different mouse models of progeria are characterized by intestinal dysbiosis with alterations that include an increase in the abundance of Proteobacteria and Cyanobacteria, and a decrease in the abundance of Verrucomicrobia. Consistent with these findings, we found that human progeria patients also display intestinal dysbiosis and that long-lived humans (that is, centenarians) exhibit a substantial increase in Verrucomicrobia and a reduction in Proteobacteria. Fecal microbiota transplantation from wild-type mice enhanced healthspan and lifespan in both progeroid mouse models, and transplantation with the verrucomicrobia Akkermansia muciniphila was sufficient to exert beneficial effects. Moreover, metabolomic analysis of ileal content points to the restoration of secondary bile acids as a possible mechanism for the beneficial effects of reestablishing a healthy microbiome. Our results demonstrate that correction of the accelerated aging-associated intestinal dysbiosis is beneficial, suggesting the existence of a link between aging and the gut microbiota that provides a rationale for microbiome-based interventions against age-related diseases.


Fecal Microbiota Transplantation , Longevity , Progeria/therapy , Animals , Disease Models, Animal , Dysbiosis , Female , Gastrointestinal Microbiome , Humans , Male , Metabolomics , Mice , Mice, Inbred C57BL
6.
Nat Med ; 25(3): 423-426, 2019 03.
Article En | MEDLINE | ID: mdl-30778239

CRISPR/Cas9-based therapies hold considerable promise for the treatment of genetic diseases. Among these, Hutchinson-Gilford progeria syndrome, caused by a point mutation in the LMNA gene, stands out as a potential candidate. Here, we explore the efficacy of a CRISPR/Cas9-based approach that reverts several alterations in Hutchinson-Gilford progeria syndrome cells and mice by introducing frameshift mutations in the LMNA gene.


CRISPR-Cas Systems , Genetic Therapy/methods , Lamin Type A/genetics , Progeria/therapy , Animals , HEK293 Cells , Humans , Lamin Type A/metabolism , Mice , Point Mutation , Progeria/genetics
7.
Nat Cell Biol ; 21(3): 410, 2019 Mar.
Article En | MEDLINE | ID: mdl-30559458

We, the authors, are retracting this Article due to issues that have come to our attention regarding data availability, data description and figure assembly. Specifically, original numerical data are not available for the majority of the graphs presented in the paper. Although original data were available for most EMSA and immunoblot experiments, those corresponding to the published EMSA data of Supplementary Fig. 8a, the independent replicate immunoblots of Fig. 8b and Supplementary Fig. 1e, and the independent replicate EMSA data of Supplementary Figs 6e, 8b, 8c and 8d, are unavailable. Mistakes were detected in the presentation of Figs 3c, 4i and Supplementary Figs 6a, 8a, 8d, 9, and in some cases the ß-actin immunoblots were erroneously described in the figure legends as loading controls, rather than as sample processing controls that were run on separate gels. Although we, the authors, believe that the key findings of the paper are still valid, given the issues with data availability we have concluded that the most appropriate course of action is to retract the Article. We deeply regret these errors and apologize to the scientific community for any confusion this publication may have caused. All authors agree with the retraction.

8.
PLoS Biol ; 16(10): e2006247, 2018 10.
Article En | MEDLINE | ID: mdl-30346946

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.


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
9.
Cell Rep ; 24(9): 2392-2403, 2018 08 28.
Article En | MEDLINE | ID: mdl-30157432

Dietary intervention constitutes a feasible approach for modulating metabolism and improving the health span and lifespan. Methionine restriction (MR) delays the appearance of age-related diseases and increases longevity in normal mice. However, the effect of MR on premature aging remains to be elucidated. Here, we describe that MR extends lifespan in two different mouse models of Hutchinson-Gilford progeria syndrome (HGPS) by reversing the transcriptome alterations in inflammation and DNA-damage response genes present in this condition. Further, MR improves the lipid profile and changes bile acid levels and conjugation, both in wild-type and in progeroid mice. Notably, treatment with cholic acid improves the health span and lifespan in vivo. These results suggest the existence of a metabolic pathway involved in the longevity extension achieved by MR and support the possibility of dietary interventions for treating progeria.


Bile Acids and Salts/metabolism , Lipid Metabolism/physiology , Methionine/metabolism , Progeria/genetics , Animals , Disease Models, Animal , Humans , Mice
11.
Methods Mol Biol ; 1731: 261-269, 2018.
Article En | MEDLINE | ID: mdl-29318560

Proteases play key roles in the execution and regulation of most if not all biological functions, and alterations in their activity, expression, or location are associated with multiple pathological conditions, including cancer and aging. In this regard, the use of RNA interference-based approaches to specifically target the expression of individual proteases constitutes an invaluable tool to identify enzymes involved in central aspects of these processes and to explore their potential as targets of therapeutic interventions. Here we describe simple protocols to optimize and monitor the specific silencing of cancer- and aging-related proteases.


Aging/metabolism , Membrane Proteins/genetics , Metalloendopeptidases/genetics , Neoplasms/pathology , RNA Interference , Ubiquitin-Specific Proteases/genetics , Aging/genetics , Apoptosis/genetics , Cell Line, Tumor , Gene Knockdown Techniques/instrumentation , Gene Knockdown Techniques/methods , Genetic Vectors/genetics , Humans , Lentivirus/genetics , Membrane Proteins/metabolism , Metalloendopeptidases/metabolism , Neoplasms/genetics , RNA, Small Interfering/metabolism , Real-Time Polymerase Chain Reaction , Ubiquitin-Specific Proteases/metabolism
12.
J Biol Chem ; 293(6): 2183-2194, 2018 02 09.
Article En | MEDLINE | ID: mdl-29273634

Deubiquitinases are proteases with a wide functional diversity that profoundly impact multiple biological processes. Among them, the ubiquitin-specific protease 36 (USP36) has been implicated in the regulation of nucleolar activity. However, its functional relevance in vivo has not yet been fully described. Here, we report the generation of an Usp36-deficient mouse model to examine the function of this enzyme. We show that Usp36 depletion is lethal in preimplantation mouse embryos, where it blocks the transition from morula to blastocyst during embryonic development. USP36 reduces the ubiquitination levels and increases the stability of the DEAH-box RNA helicase DHX33, which is critically involved in ribosomal RNA synthesis and mRNA translation. In agreement with this finding, O-propargyl-puromycin incorporation experiments, Northern blot, and electron microscopy analyses demonstrated the role of USP36 in ribosomal RNA and protein synthesis. Finally, we show that USP36 down-regulation alters cell proliferation in human cancer cells by inducing both apoptosis and cell cycle arrest, and that reducing DHX33 levels through short hairpin RNA interference has the same effect. Collectively, these results support that Usp36 is essential for cell and organism viability because of its role in ribosomal RNA processing and protein synthesis, which is mediated, at least in part, by regulating DHX33 stability.


Blastocyst , DEAD-box RNA Helicases/chemistry , Deubiquitinating Enzymes/physiology , RNA Helicases/chemistry , Ubiquitin Thiolesterase/physiology , Animals , Apoptosis , Cell Line, Tumor , Cell Proliferation , Deubiquitinating Enzymes/genetics , Embryo Loss , Humans , Mice , Mice, Knockout , Protein Biosynthesis , Protein Stability , RNA, Ribosomal , Ubiquitin Thiolesterase/genetics
13.
J Biol Chem ; 292(10): 4164-4175, 2017 03 10.
Article En | MEDLINE | ID: mdl-28154181

KRAS is the most frequently mutated oncogene in human cancer, but its therapeutic targeting remains challenging. Here, we report a synthetic lethal screen with a library of deubiquitinases and identify USP39, which encodes an essential splicing factor, as a critical gene for the viability of KRAS-dependent cells. We show that splicing fidelity inhibitors decrease preferentially the proliferation rate of KRAS-active cells. Moreover, depletion of DHX38, encoding an USP39-interacting splicing factor, also reduces the viability of these cells. In agreement with these results, USP39 depletion caused a significant reduction in pre-mRNA splicing efficiency, as demonstrated through RNA-seq experiments. Furthermore, we show that USP39 is up-regulated in lung and colon carcinomas and its expression correlates with KRAS levels and poor clinical outcome. Accordingly, our work provides critical information for the development of splicing-directed antitumor treatments and supports the potential of USP39-targeting strategies as the basis of new anticancer therapies.


Colonic Neoplasms/pathology , Lung Neoplasms/pathology , Mutation/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Ubiquitin-Specific Proteases/metabolism , Animals , Apoptosis , Blotting, Western , Cell Proliferation , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Mice , Mice, Nude , Prognosis , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Survival Rate , Tumor Cells, Cultured , Ubiquitin-Specific Proteases/genetics , Xenograft Model Antitumor Assays
15.
Oncotarget ; 7(46): 74427-74434, 2016 Nov 15.
Article En | MEDLINE | ID: mdl-27769071

Ubiquitin-Specific Proteases (USPs) are deubiquitinating enzymes frequently deregulated in human malignancies. Here, we show that USP54 is overexpressed in intestinal stem cells and demonstrate that its downregulation in colorectal carcinoma cells impedes tumorigenesis. We have generated mutant mice deficient for this deubiquitinase, which are viable and fertile, and protected against chemically-induced colorectal carcinoma. Furthermore, we show that USP54 is upregulated in human colon cancer and associates with poor prognosis. In agreement with these results, Usp54 downregulation in mouse melanoma cells inhibits lung metastasis formation. Collectively, this work has uncovered the pro-tumorigenic properties of USP54, highlighting the importance of deubiquitinating enzymes as promising targets for the development of specific anti-cancer therapies.


Colorectal Neoplasms/genetics , Gene Expression , Neoplastic Stem Cells/metabolism , Ubiquitin-Specific Proteases/genetics , Animals , Biomarkers, Tumor , Cell Line, Tumor , Cell Proliferation , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Disease Progression , Embryonic Development/genetics , Gene Expression Regulation, Neoplastic , Humans , Mice , Mice, Knockout , Neoplasm Invasiveness , Neoplasm Metastasis , Ubiquitin-Specific Proteases/metabolism
16.
J Clin Invest ; 126(10): 3879-3893, 2016 10 03.
Article En | MEDLINE | ID: mdl-27617860

Hutchinson-Gilford progeria syndrome (HGPS) is a rare autosomal dominant genetic disease that is caused by a silent mutation of the LMNA gene encoding lamins A and C (lamin A/C). The G608G mutation generates a more accessible splicing donor site than does WT and produces an alternatively spliced product of LMNA called progerin, which is also expressed in normal aged cells. In this study, we determined that progerin binds directly to lamin A/C and induces profound nuclear aberrations. Given this observation, we performed a random screening of a chemical library and identified 3 compounds (JH1, JH4, and JH13) that efficiently block progerin-lamin A/C binding. These 3 chemicals, particularly JH4, alleviated nuclear deformation and reversed senescence markers characteristic of HGPS cells, including growth arrest and senescence-associated ß-gal (SA-ß-gal) activity. We then used microarray-based analysis to demonstrate that JH4 is able to rescue defects of cell-cycle progression in both HGPS and aged cells. Furthermore, administration of JH4 to LmnaG609G/G609G-mutant mice, which phenocopy human HGPS, resulted in a marked improvement of several progeria phenotypes and an extended lifespan. Together, these findings indicate that specific inhibitors with the ability to block pathological progerin-lamin A/C binding may represent a promising strategy for improving lifespan and health in both HGPS and normal aging.


Acrylates/pharmacology , Coumarins/pharmacology , Lamin Type A/metabolism , Progeria/drug therapy , Acrylates/chemistry , Animals , Cellular Senescence , Coumarins/chemistry , Drug Evaluation, Preclinical , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/metabolism , Female , Gene Expression/drug effects , HEK293 Cells , Humans , Male , Mice, Inbred C57BL , Mice, Transgenic , Phenotype , Progeria/metabolism , Protein Binding , Protein Isoforms/metabolism , Protein Transport/drug effects
17.
Cell ; 166(4): 802-821, 2016 Aug 11.
Article En | MEDLINE | ID: mdl-27518560

Several metabolic alterations accumulate over time along with a reduction in biological fitness, suggesting the existence of a "metabolic clock" that controls aging. Multiple inborn defects in metabolic circuitries accelerate aging, whereas genetic loci linked to exceptional longevity influence metabolism. Each of the nine hallmarks of aging is connected to undesirable metabolic alterations. The main features of the "westernized" lifestyle, including hypercaloric nutrition and sedentariness, can accelerate aging as they have detrimental metabolic consequences. Conversely, lifespan-extending maneuvers including caloric restriction impose beneficial pleiotropic effects on metabolism. The introduction of strategies that promote metabolic fitness may extend healthspan in humans.


Aging/metabolism , Longevity , Aging/blood , Animals , Caloric Restriction , Cellular Senescence , Diet , Diet, Western , Exercise , Humans , Life Style , Metformin/administration & dosage , Mitochondria/metabolism , Stress, Physiological
19.
Nat Med ; 22(1): 91-6, 2016 Jan.
Article En | MEDLINE | ID: mdl-26692333

AIRAPL (arsenite-inducible RNA-associated protein-like) is an evolutionarily conserved regulator of cellular proteostasis linked to longevity in nematodes, but its biological function in mammals is unknown. We show herein that AIRAPL-deficient mice develop a fully-penetrant myeloproliferative neoplastic process. Proteomic analysis of AIRAPL-deficient mice revealed that this protein exerts its antineoplastic function through the regulation of the insulin/insulin-like growth factor 1 (IGF-1) signaling pathway. We demonstrate that AIRAPL interacts with newly synthesized insulin-related growth factor-1 receptor (IGF1R) polypeptides, promoting their ubiquitination and proteasome-mediated degradation. Accordingly, genetic and pharmacological IGF1R inhibitory strategies prevent the hematological disease found in AIRAPL-deficient mice as well as that in mice carrying the Jak2(V617F) mutation, thereby demonstrating the causal involvement of this pathway in the pathogenesis of myeloproliferative neoplasms. Consistent with its proposed role as a tumor suppressor of myeloid transformation, AIRAPL expression is widely abrogated in human myeloproliferative disorders. Collectively, these findings support the oncogenic relevance of proteostasis deregulation in hematopoietic cells, and they unveil novel therapeutic targets for these frequent hematological neoplasias.


Carrier Proteins/genetics , Insulin-Like Growth Factor I/metabolism , Myeloproliferative Disorders/genetics , RNA-Binding Proteins/genetics , Receptor, IGF Type 1/metabolism , Adaptor Proteins, Signal Transducing , Animals , Blotting, Western , CRISPR-Cas Systems , Caenorhabditis elegans , Caenorhabditis elegans Proteins/genetics , Carrier Proteins/metabolism , Flow Cytometry , Fluorescent Antibody Technique , Humans , Immunoprecipitation , Mice , Mice, Knockout , Microfilament Proteins/genetics , Myeloproliferative Disorders/metabolism , Proteasome Endopeptidase Complex/metabolism , Proteomics , Proteostasis Deficiencies , RNA-Binding Proteins/metabolism , Real-Time Polymerase Chain Reaction , Receptor, Insulin/genetics , Signal Transduction , Ubiquitination , Zinc Fingers/genetics
20.
Nat Cell Biol ; 17(8): 1004-13, 2015 Aug.
Article En | MEDLINE | ID: mdl-26214134

Ageing constitutes a critical impediment to somatic cell reprogramming. We have explored the regulatory mechanisms that constitute age-associated barriers, through derivation of induced pluripotent stem cells (iPSCs) from individuals with premature or physiological ageing. We demonstrate that NF-κB activation blocks the generation of iPSCs in ageing. We also show that NF-κB repression occurs during cell reprogramming towards a pluripotent state. Conversely, ageing-associated NF-κB hyperactivation impairs the generation of iPSCs by eliciting the reprogramming repressor DOT1L, which reinforces senescence signals and downregulates pluripotency genes. Genetic and pharmacological NF-κB inhibitory strategies significantly increase the reprogramming efficiency of fibroblasts from Néstor-Guillermo progeria syndrome and Hutchinson-Gilford progeria syndrome patients, as well as from normal aged donors. Finally, we demonstrate that DOT1L inhibition in vivo extends lifespan and ameliorates the accelerated ageing phenotype of progeroid mice, supporting the interest of studying age-associated molecular impairments to identify targets of rejuvenation strategies.


Aging/metabolism , Cell Proliferation , Cellular Reprogramming , Cellular Senescence , Fibroblasts/metabolism , Induced Pluripotent Stem Cells/metabolism , NF-kappa B/metabolism , Progeria/metabolism , Age Factors , Aged, 80 and over , Aging/genetics , Aging/pathology , Animals , Case-Control Studies , Cell Differentiation , Cell Line , Cellular Reprogramming/drug effects , Disease Models, Animal , Female , Fibroblasts/drug effects , Fibroblasts/pathology , Gene Expression Regulation, Developmental , Genetic Predisposition to Disease , Histone-Lysine N-Methyltransferase , Humans , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/pathology , Male , Membrane Proteins/deficiency , Membrane Proteins/genetics , Metalloendopeptidases/deficiency , Metalloendopeptidases/genetics , Methyltransferases/genetics , Methyltransferases/metabolism , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/antagonists & inhibitors , NF-kappa B/genetics , Phenotype , Progeria/genetics , Progeria/pathology , RNA Interference , Signal Transduction , Time Factors , Transfection
...