Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
1.
Nature ; 613(7942): 169-178, 2023 01.
Article in English | MEDLINE | ID: mdl-36544018

ABSTRACT

Tissue regeneration requires coordination between resident stem cells and local niche cells1,2. Here we identify that senescent cells are integral components of the skeletal muscle regenerative niche that repress regeneration at all stages of life. The technical limitation of senescent-cell scarcity3 was overcome by combining single-cell transcriptomics and a senescent-cell enrichment sorting protocol. We identified and isolated different senescent cell types from damaged muscles of young and old mice. Deeper transcriptome, chromatin and pathway analyses revealed conservation of cell identity traits as well as two universal senescence hallmarks (inflammation and fibrosis) across cell type, regeneration time and ageing. Senescent cells create an aged-like inflamed niche that mirrors inflammation associated with ageing (inflammageing4) and arrests stem cell proliferation and regeneration. Reducing the burden of senescent cells, or reducing their inflammatory secretome through CD36 neutralization, accelerates regeneration in young and old mice. By contrast, transplantation of senescent cells delays regeneration. Our results provide a technique for isolating in vivo senescent cells, define a senescence blueprint for muscle, and uncover unproductive functional interactions between senescent cells and stem cells in regenerative niches that can be overcome. As senescent cells also accumulate in human muscles, our findings open potential paths for improving muscle repair throughout life.


Subject(s)
Aging , Cellular Senescence , Inflammation , Muscle, Skeletal , Regeneration , Stem Cell Niche , Aged , Animals , Humans , Mice , Aging/metabolism , Aging/physiology , Cellular Senescence/physiology , Inflammation/metabolism , Inflammation/physiopathology , Muscle, Skeletal/physiology , Muscle, Skeletal/physiopathology , Stem Cells/physiology , Fibrosis/physiopathology , Stem Cell Niche/physiology , Transcriptome , Chromatin/genetics , Geroscience
3.
Nature ; 529(7584): 37-42, 2016 Jan 07.
Article in English | MEDLINE | ID: mdl-26738589

ABSTRACT

During ageing, muscle stem-cell regenerative function declines. At advanced geriatric age, this decline is maximal owing to transition from a normal quiescence into an irreversible senescence state. How satellite cells maintain quiescence and avoid senescence until advanced age remains unknown. Here we report that basal autophagy is essential to maintain the stem-cell quiescent state in mice. Failure of autophagy in physiologically aged satellite cells or genetic impairment of autophagy in young cells causes entry into senescence by loss of proteostasis, increased mitochondrial dysfunction and oxidative stress, resulting in a decline in the function and number of satellite cells. Re-establishment of autophagy reverses senescence and restores regenerative functions in geriatric satellite cells. As autophagy also declines in human geriatric satellite cells, our findings reveal autophagy to be a decisive stem-cell-fate regulator, with implications for fostering muscle regeneration in sarcopenia.


Subject(s)
Autophagy/physiology , Cellular Senescence , Satellite Cells, Skeletal Muscle/cytology , Aging/pathology , Animals , Cell Count , Cyclin-Dependent Kinase Inhibitor p16/genetics , Epigenesis, Genetic , Homeostasis , Humans , Male , Mice , Mitochondria/metabolism , Mitochondria/pathology , Mitophagy , Muscle, Skeletal/cytology , Muscle, Skeletal/pathology , Organelles/metabolism , Oxidative Stress , Proteins/metabolism , Reactive Oxygen Species/metabolism , Regeneration , Sarcopenia/pathology , Sarcopenia/prevention & control , Satellite Cells, Skeletal Muscle/pathology
4.
Nature ; 506(7488): 316-21, 2014 Feb 20.
Article in English | MEDLINE | ID: mdl-24522534

ABSTRACT

Regeneration of skeletal muscle depends on a population of adult stem cells (satellite cells) that remain quiescent throughout life. Satellite cell regenerative functions decline with ageing. Here we report that geriatric satellite cells are incapable of maintaining their normal quiescent state in muscle homeostatic conditions, and that this irreversibly affects their intrinsic regenerative and self-renewal capacities. In geriatric mice, resting satellite cells lose reversible quiescence by switching to an irreversible pre-senescence state, caused by derepression of p16(INK4a) (also called Cdkn2a). On injury, these cells fail to activate and expand, undergoing accelerated entry into a full senescence state (geroconversion), even in a youthful environment. p16(INK4a) silencing in geriatric satellite cells restores quiescence and muscle regenerative functions. Our results demonstrate that maintenance of quiescence in adult life depends on the active repression of senescence pathways. As p16(INK4a) is dysregulated in human geriatric satellite cells, these findings provide the basis for stem-cell rejuvenation in sarcopenic muscles.


Subject(s)
Aging/metabolism , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Satellite Cells, Skeletal Muscle/cytology , Satellite Cells, Skeletal Muscle/metabolism , Adult , Animals , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p16/deficiency , Cyclin-Dependent Kinase Inhibitor p16/genetics , E2F1 Transcription Factor/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Progeria/metabolism , Progeria/pathology , Regeneration , Rejuvenation , Retinoblastoma Protein/metabolism , Young Adult
5.
J Exp Clin Cancer Res ; 43(1): 127, 2024 Apr 29.
Article in English | MEDLINE | ID: mdl-38685100

ABSTRACT

BACKGROUND: TP53, the most frequently mutated gene in human cancers, orchestrates a complex transcriptional program crucial for cancer prevention. While certain TP53-dependent genes have been extensively studied, others, like the recently identified RNF144B, remained poorly understood. This E3 ubiquitin ligase has shown potent tumor suppressor activity in murine Eµ Myc-driven lymphoma, emphasizing its significance in the TP53 network. However, little is known about its targets and its role in cancer development, requiring further exploration. In this work, we investigate RNF144B's impact on tumor suppression beyond the hematopoietic compartment in human cancers. METHODS: Employing TP53 wild-type cells, we generated models lacking RNF144B in both non-transformed and cancerous cells of human and mouse origin. By using proteomics, transcriptomics, and functional analysis, we assessed RNF144B's impact in cellular proliferation and transformation. Through in vitro and in vivo experiments, we explored proliferation, DNA repair, cell cycle control, mitotic progression, and treatment resistance. Findings were contrasted with clinical datasets and bioinformatics analysis. RESULTS: Our research underscores RNF144B's pivotal role as a tumor suppressor, particularly in lung adenocarcinoma. In both human and mouse oncogene-expressing cells, RNF144B deficiency heightened cellular proliferation and transformation. Proteomic and transcriptomic analysis revealed RNF144B's novel function in mediating protein degradation associated with cell cycle progression, DNA damage response and genomic stability. RNF144B deficiency induced chromosomal instability, mitotic defects, and correlated with elevated aneuploidy and worse prognosis in human tumors. Furthermore, RNF144B-deficient lung adenocarcinoma cells exhibited resistance to cell cycle inhibitors that induce chromosomal instability. CONCLUSIONS: Supported by clinical data, our study suggests that RNF144B plays a pivotal role in maintaining genomic stability during tumor suppression.


Subject(s)
Genomic Instability , Tumor Suppressor Protein p53 , Ubiquitin-Protein Ligases , Animals , Humans , Mice , Cell Line, Tumor , Cell Proliferation , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Lung Neoplasms/metabolism , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Protein p53/genetics , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics
6.
Nat Aging ; 2: 851-866, 2022 09.
Article in English | MEDLINE | ID: mdl-36438588

ABSTRACT

Cellular senescence is a stable type of cell cycle arrest triggered by different stresses. As such, senescence drives age-related diseases and curbs cellular replicative potential. Here, we show that 3-deazaadenosine (3DA), an S-adenosyl homocysteinase (AHCY) inhibitor, alleviates replicative and oncogene-induced senescence. 3DA-treated senescent cells showed reduced global Histone H3 Lysine 36 trimethylation (H3K36me3), an epigenetic modification that marks the bodies of actively transcribed genes. By integrating transcriptome and epigenome data, we demonstrate that 3DA treatment affects key factors of the senescence transcriptional program. Remarkably, 3DA treatment alleviated senescence and increased the proliferative and regenerative potential of muscle stem cells from very old mice in vitro and in vivo. Moreover, ex vivo 3DA treatment was sufficient to enhance the engraftment of human umbilical cord blood (UCB) cells in immunocompromised mice. Together, our results identify 3DA as a promising drug enhancing the efficiency of cellular therapies by restraining senescence.


Subject(s)
Cellular Senescence , Histones , Humans , Mice , Animals , Histones/genetics , Cellular Senescence/genetics , Tubercidin/pharmacology , Epigenesis, Genetic
7.
Nat Commun ; 11(1): 189, 2020 01 13.
Article in English | MEDLINE | ID: mdl-31929511

ABSTRACT

A unique property of skeletal muscle is its ability to adapt its mass to changes in activity. Inactivity, as in disuse or aging, causes atrophy, the loss of muscle mass and strength, leading to physical incapacity and poor quality of life. Here, through a combination of transcriptomics and transgenesis, we identify sestrins, a family of stress-inducible metabolic regulators, as protective factors against muscle wasting. Sestrin expression decreases during inactivity and its genetic deficiency exacerbates muscle wasting; conversely, sestrin overexpression suffices to prevent atrophy. This protection occurs through mTORC1 inhibition, which upregulates autophagy, and AKT activation, which in turn inhibits FoxO-regulated ubiquitin-proteasome-mediated proteolysis. This study reveals sestrin as a central integrator of anabolic and degradative pathways preventing muscle wasting. Since sestrin also protected muscles against aging-induced atrophy, our findings have implications for sarcopenia.


Subject(s)
Heat-Shock Proteins/metabolism , Muscle, Skeletal/pathology , Muscular Atrophy/prevention & control , Nuclear Proteins/metabolism , Signal Transduction , Aging , Animals , Autophagy , Disease Models, Animal , Forkhead Box Protein O1/genetics , Forkhead Box Protein O1/metabolism , Forkhead Box Protein O3/genetics , Forkhead Box Protein O3/metabolism , Gene Expression , Heat-Shock Proteins/genetics , Humans , Male , Mechanistic Target of Rapamycin Complex 1/genetics , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Muscle Proteins/genetics , Muscle Proteins/metabolism , Muscle, Skeletal/metabolism , Muscular Atrophy/genetics , Muscular Atrophy/metabolism , Muscular Atrophy/pathology , Nuclear Proteins/genetics , Sarcopenia/genetics , Sarcopenia/metabolism , Sarcopenia/pathology , Sarcopenia/prevention & control
8.
Nat Cell Biol ; 22(11): 1307-1318, 2020 11.
Article in English | MEDLINE | ID: mdl-33106654

ABSTRACT

Tissue regeneration declines with ageing but little is known about whether this arises from changes in stem-cell heterogeneity. Here, in homeostatic skeletal muscle, we identify two quiescent stem-cell states distinguished by relative CD34 expression: CD34High, with stemness properties (genuine state), and CD34Low, committed to myogenic differentiation (primed state). The genuine-quiescent state is unexpectedly preserved into later life, succumbing only in extreme old age due to the acquisition of primed-state traits. Niche-derived IGF1-dependent Akt activation debilitates the genuine stem-cell state by imposing primed-state features via FoxO inhibition. Interventions to neutralize Akt and promote FoxO activity drive a primed-to-genuine state conversion, whereas FoxO inactivation deteriorates the genuine state at a young age, causing regenerative failure of muscle, as occurs in geriatric mice. These findings reveal transcriptional determinants of stem-cell heterogeneity that resist ageing more than previously anticipated and are only lost in extreme old age, with implications for the repair of geriatric muscle.


Subject(s)
Antigens, CD34/metabolism , Cell Proliferation , Cell Self Renewal , Cellular Senescence , Forkhead Transcription Factors/metabolism , Muscle, Skeletal/metabolism , Regeneration , Satellite Cells, Skeletal Muscle/metabolism , Age Factors , Animals , Cardiotoxins/toxicity , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Proliferation/drug effects , Cell Proliferation/genetics , Cell Self Renewal/drug effects , Cell Self Renewal/genetics , Cells, Cultured , Cellular Senescence/drug effects , Cellular Senescence/genetics , Forkhead Box Protein O1/genetics , Forkhead Box Protein O1/metabolism , Forkhead Box Protein O3/genetics , Forkhead Box Protein O3/metabolism , Forkhead Transcription Factors/genetics , Gene Expression Regulation , Male , Mice, Inbred C57BL , Mice, Knockout , Mice, SCID , Muscle, Skeletal/drug effects , Muscle, Skeletal/pathology , Muscle, Skeletal/transplantation , Phenotype , Proto-Oncogene Proteins c-akt/metabolism , Regeneration/drug effects , Regeneration/genetics , Satellite Cells, Skeletal Muscle/drug effects , Satellite Cells, Skeletal Muscle/pathology , Satellite Cells, Skeletal Muscle/transplantation , Signal Transduction , Stem Cell Niche
9.
Front Biosci ; 12: 4722-30, 2007 May 01.
Article in English | MEDLINE | ID: mdl-17485408

ABSTRACT

B-cell lymphoma-2 (Bcl-2) family members have been demonstrated to play a crucial role in the regulation of apoptosis as mediators in between the apical stimuli sensing steps and the executory mechanisms of apoptosis. Deregulation of their role may subvert the homeostasis of a given tissue and collaborate in the genesis of a myriad of diseases characterised by exacerbated or insufficient apoptosis, including diseases such as neurodegenerative diseases or cancer. Structural studies have defined homology regions shared by the members of the family that are responsible of the network of interactions established amongst the members of the family. These proteins usually form heterodimers between the so called antiapoptotic multidomain members and the proapoptotic BH3-only proteins. As a consequence, mitochondrial apoptogenic proteins are released to the cytoplasm and the apoptotic signal proceeds towards the final, execution phase of the apoptotic process. The high complexity of the family (more than 20 members have been isolated) makes the study of individual proteins difficult. Genetic approaches have revealed a high degree of redundancy in the family. Only a few proteins belonging to the antiapoptotic group have been proven to be essential for correct embryonic development. Genetic inactivation in mice shows a dramatic phenotype characterised by massive cell death in multiple tissues during embryogenesis, which leads from very early up to perinatal death. This genetic evidence proves the importance of the members of the family for the regulation of apoptosis in order to achieve the proper development and homeostasis of tissues and organs.


Subject(s)
Apoptosis , Proto-Oncogene Proteins c-bcl-2/physiology , Animals , Mice , Mice, Knockout , Models, Theoretical , Proto-Oncogene Proteins c-bcl-2/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL