Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 251
Filter
1.
Article in English | MEDLINE | ID: mdl-38831121

ABSTRACT

Once considered a tissue culture-specific phenomenon, cellular senescence has now been linked to various biological processes with both beneficial and detrimental roles in humans, rodents and other species. Much of our understanding of senescent cell biology still originates from tissue culture studies, where each cell in the culture is driven to an irreversible cell cycle arrest. By contrast, in tissues, these cells are relatively rare and difficult to characterize, and it is now established that fully differentiated, postmitotic cells can also acquire a senescence phenotype. The SenNet Biomarkers Working Group was formed to provide recommendations for the use of cellular senescence markers to identify and characterize senescent cells in tissues. Here, we provide recommendations for detecting senescent cells in different tissues based on a comprehensive analysis of existing literature reporting senescence markers in 14 tissues in mice and humans. We discuss some of the recent advances in detecting and characterizing cellular senescence, including molecular senescence signatures and morphological features, and the use of circulating markers. We aim for this work to be a valuable resource for both seasoned investigators in senescence-related studies and newcomers to the field.

2.
Cell ; 179(4): 813-827, 2019 10 31.
Article in English | MEDLINE | ID: mdl-31675495

ABSTRACT

Cellular senescence is a cell state implicated in various physiological processes and a wide spectrum of age-related diseases. Recently, interest in therapeutically targeting senescence to improve healthy aging and age-related disease, otherwise known as senotherapy, has been growing rapidly. Thus, the accurate detection of senescent cells, especially in vivo, is essential. Here, we present a consensus from the International Cell Senescence Association (ICSA), defining and discussing key cellular and molecular features of senescence and offering recommendations on how to use them as biomarkers. We also present a resource tool to facilitate the identification of genes linked with senescence, SeneQuest (available at http://Senequest.net). Lastly, we propose an algorithm to accurately assess and quantify senescence, both in cultured cells and in vivo.


Subject(s)
Aging/genetics , Biomarkers , Cellular Senescence/genetics , Genetic Diseases, Inborn/genetics , Cell Cycle Checkpoints/genetics , Chromatin/genetics , Gene Expression Regulation/genetics , Genetic Diseases, Inborn/therapy , Humans
3.
Cell ; 169(1): 132-147.e16, 2017 03 23.
Article in English | MEDLINE | ID: mdl-28340339

ABSTRACT

The accumulation of irreparable cellular damage restricts healthspan after acute stress or natural aging. Senescent cells are thought to impair tissue function, and their genetic clearance can delay features of aging. Identifying how senescent cells avoid apoptosis allows for the prospective design of anti-senescence compounds to address whether homeostasis can also be restored. Here, we identify FOXO4 as a pivot in senescent cell viability. We designed a FOXO4 peptide that perturbs the FOXO4 interaction with p53. In senescent cells, this selectively causes p53 nuclear exclusion and cell-intrinsic apoptosis. Under conditions where it was well tolerated in vivo, this FOXO4 peptide neutralized doxorubicin-induced chemotoxicity. Moreover, it restored fitness, fur density, and renal function in both fast aging XpdTTD/TTD and naturally aged mice. Thus, therapeutic targeting of senescent cells is feasible under conditions where loss of health has already occurred, and in doing so tissue homeostasis can effectively be restored.


Subject(s)
Aging/pathology , Antibiotics, Antineoplastic/adverse effects , Cell-Penetrating Peptides/pharmacology , Doxorubicin/adverse effects , Aging/drug effects , Animals , Antibiotics, Antineoplastic/administration & dosage , Antibiotics, Antineoplastic/pharmacology , Apoptosis , Cell Cycle Proteins , Cell Line , Cell Survival , Cellular Senescence/drug effects , Doxorubicin/administration & dosage , Doxorubicin/pharmacology , Female , Fibroblasts/cytology , Forkhead Transcription Factors/chemistry , Forkhead Transcription Factors/metabolism , Humans , Inclusion Bodies/drug effects , Inclusion Bodies/metabolism , Inclusion Bodies/pathology , Kidney/drug effects , Kidney/physiology , Liver/drug effects , Liver/physiology , Male , Mice , Trichothiodystrophy Syndromes/drug therapy , Tumor Suppressor Protein p53/metabolism
4.
Cell ; 159(4): 709-13, 2014 Nov 06.
Article in English | MEDLINE | ID: mdl-25417146

ABSTRACT

Mammalian aging can be delayed with genetic, dietary, and pharmacologic approaches. Given that the elderly population is dramatically increasing and that aging is the greatest risk factor for a majority of chronic diseases driving both morbidity and mortality, it is critical to expand geroscience research directed at extending human healthspan.


Subject(s)
Aging/physiology , Chronic Disease , Aging/pathology , Animals , Biomedical Research , Epigenesis, Genetic , Gene-Environment Interaction , Humans
5.
Nature ; 571(7764): 183-192, 2019 07.
Article in English | MEDLINE | ID: mdl-31292558

ABSTRACT

For several decades, understanding ageing and the processes that limit lifespan have challenged biologists. Thirty years ago, the biology of ageing gained unprecedented scientific credibility through the identification of gene variants that extend the lifespan of multicellular model organisms. Here we summarize the milestones that mark this scientific triumph, discuss different ageing pathways and processes, and suggest that ageing research is entering a new era that has unique medical, commercial and societal implications. We argue that this era marks an inflection point, not only in ageing research but also for all biological research that affects the human healthspan.


Subject(s)
Aging/physiology , Biomedical Research , Healthy Aging/physiology , Rejuvenation/physiology , Aging/drug effects , Aging/genetics , Circadian Clocks , Clinical Trials as Topic , Healthy Aging/drug effects , Healthy Aging/genetics , Humans , Inflammation , Longevity/drug effects , Longevity/genetics , Longevity/physiology , Mitochondria/metabolism , Nutritional Status , Oxidative Stress , Signal Transduction
6.
Proc Natl Acad Sci U S A ; 119(49): e2207181119, 2022 12 06.
Article in English | MEDLINE | ID: mdl-36459652

ABSTRACT

Aging is characterized by a progressive loss of brain volume at an estimated rate of 5% per decade after age 40. While these morphometric changes, especially those affecting gray matter and atrophy of the temporal lobe, are predictors of cognitive performance, the strong association with aging obscures the potential parallel, but more specific role, of individual subject physiology. Here, we studied a cohort of 554 human subjects who were monitored using structural MRI scans and blood immune protein concentrations. Using machine learning, we derived a cytokine clock (CyClo), which predicted age with good accuracy (Mean Absolute Error = 6 y) based on the expression of a subset of immune proteins. These proteins included, among others, Placenta Growth Factor (PLGF) and Vascular Endothelial Growth Factor (VEGF), both involved in angiogenesis, the chemoattractant vascular cell adhesion molecule 1 (VCAM-1), the canonical inflammatory proteins interleukin-6 (IL-6) and tumor necrosis factor alpha (TNFα), the chemoattractant IP-10 (CXCL10), and eotaxin-1 (CCL11), previously involved in brain disorders. Age, sex, and the CyClo were independently associated with different functionally defined cortical networks in the brain. While age was mostly correlated with changes in the somatomotor system, sex was associated with variability in the frontoparietal, ventral attention, and visual networks. Significant canonical correlation was observed for the CyClo and the default mode, limbic, and dorsal attention networks, indicating that immune circulating proteins preferentially affect brain processes such as focused attention, emotion, memory, response to social stress, internal evaluation, and access to consciousness. Thus, we identified immune biomarkers of brain aging which could be potential therapeutic targets for the prevention of age-related cognitive decline.


Subject(s)
Brain , Vascular Endothelial Growth Factor A , Humans , Adult , Atrophy , Brain/diagnostic imaging , Aging , Research Personnel , Cytokines
7.
Chromosome Res ; 31(4): 32, 2023 11 01.
Article in English | MEDLINE | ID: mdl-37910282

ABSTRACT

This review investigates the role of aneuploidy and chromosome instability (CIN) in the aging brain. Aneuploidy refers to an abnormal chromosomal count, deviating from the normal diploid set. It can manifest as either a deficiency or excess of chromosomes. CIN encompasses a broader range of chromosomal alterations, including aneuploidy as well as structural modifications in DNA. We provide an overview of the state-of-the-art methodologies utilized for studying aneuploidy and CIN in non-tumor somatic tissues devoid of clonally expanded populations of aneuploid cells.CIN and aneuploidy, well-established hallmarks of cancer cells, are also associated with the aging process. In non-transformed cells, aneuploidy can contribute to functional impairment and developmental disorders. Despite the importance of understanding the prevalence and specific consequences of aneuploidy and CIN in the aging brain, these aspects remain incompletely understood, emphasizing the need for further scientific investigations.This comprehensive review consolidates the present understanding, addresses discrepancies in the literature, and provides valuable insights for future research efforts.


Subject(s)
Aneuploidy , Neoplasms , Animals , Humans , Chromosomal Instability , Chromosome Aberrations , Brain , Chromosomes , Neoplasms/genetics , Mammals/genetics
8.
Mol Cell ; 61(4): 535-546, 2016 Feb 18.
Article in English | MEDLINE | ID: mdl-26833090

ABSTRACT

XPG is a structure-specific endonuclease required for nucleotide excision repair, and incision-defective XPG mutations cause the skin cancer-prone syndrome xeroderma pigmentosum. Truncating mutations instead cause the neurodevelopmental progeroid disorder Cockayne syndrome, but little is known about how XPG loss results in this devastating disease. We identify XPG as a partner of BRCA1 and BRCA2 in maintaining genomic stability through homologous recombination (HRR). XPG depletion causes DNA double-strand breaks, chromosomal abnormalities, cell-cycle delays, defective HRR, inability to overcome replication fork stalling, and replication stress. XPG directly interacts with BRCA2, RAD51, and PALB2, and XPG depletion reduces their chromatin binding and subsequent RAD51 foci formation. Upstream in HRR, XPG interacts directly with BRCA1. Its depletion causes BRCA1 hyper-phosphorylation and persistent chromatin binding. These unexpected findings establish XPG as an HRR protein with important roles in genome stability and suggest how XPG defects produce severe clinical consequences including cancer and accelerated aging.


Subject(s)
BRCA1 Protein/metabolism , BRCA2 Protein/metabolism , Cockayne Syndrome/genetics , DNA-Binding Proteins/genetics , Endonucleases/genetics , Genomic Instability , Homologous Recombination , Nuclear Proteins/genetics , Transcription Factors/genetics , Animals , Cell Line, Tumor , Cockayne Syndrome/metabolism , DNA Repair , DNA-Binding Proteins/metabolism , Endonucleases/metabolism , Fanconi Anemia Complementation Group N Protein , Genome, Human , HeLa Cells , Humans , Mice , Nuclear Proteins/metabolism , Phosphorylation , Rad51 Recombinase/metabolism , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism
9.
PLoS Biol ; 18(1): e3000599, 2020 01.
Article in English | MEDLINE | ID: mdl-31945054

ABSTRACT

The senescence-associated secretory phenotype (SASP) has recently emerged as a driver of and promising therapeutic target for multiple age-related conditions, ranging from neurodegeneration to cancer. The complexity of the SASP, typically assessed by a few dozen secreted proteins, has been greatly underestimated, and a small set of factors cannot explain the diverse phenotypes it produces in vivo. Here, we present the "SASP Atlas," a comprehensive proteomic database of soluble proteins and exosomal cargo SASP factors originating from multiple senescence inducers and cell types. Each profile consists of hundreds of largely distinct proteins but also includes a subset of proteins elevated in all SASPs. Our analyses identify several candidate biomarkers of cellular senescence that overlap with aging markers in human plasma, including Growth/differentiation factor 15 (GDF15), stanniocalcin 1 (STC1), and serine protease inhibitors (SERPINs), which significantly correlated with age in plasma from a human cohort, the Baltimore Longitudinal Study of Aging (BLSA). Our findings will facilitate the identification of proteins characteristic of senescence-associated phenotypes and catalog potential senescence biomarkers to assess the burden, originating stimulus, and tissue of origin of senescent cells in vivo.


Subject(s)
Aging/metabolism , Biomarkers/metabolism , Cellular Senescence/physiology , Proteome/analysis , Secretory Pathway/physiology , Biomarkers/analysis , Cells, Cultured , Databases, Protein , Exosomes/chemistry , Exosomes/metabolism , Female , Humans , Phenotype , Proteome/metabolism , Proteomics
10.
Exp Eye Res ; 225: 109254, 2022 12.
Article in English | MEDLINE | ID: mdl-36150544

ABSTRACT

Advanced age is the most established risk factor for developing age-related macular degeneration (AMD), one of the leading causes of visual impairment in the elderly, in Western and developed countries. Similarly, after middle age, there is an exponential increase in pathologic molecular and cellular events that can induce senescence, traditionally defined as an irreversible loss of the cells' ability to divide and most recently reported to also occur in select post-mitotic and terminally differentiated cells, such as neurons. Together these facts raise the question as to whether or not cellular senescence, may play a role in the development of AMD. A number of studies have reported the effect of ocular-relevant inducers of senescence using primarily in vitro models of poorly polarized, actively dividing retinal pigment epithelial (RPE) cell lines. However, in interpretating the data, the fidelity of these culture models to the RPE in vivo, must be considered. Fewer studies have explored the presence and/or impact of senescent cells in in vivo models that present with phenotypic features of AMD, leaving this an open field for further investigation. The goal of this review is to discuss current thoughts on the potential role of senescence in AMD development and progression, with consideration of the model systems used and their relevance to human disease.


Subject(s)
Macular Degeneration , Retinal Pigment Epithelium , Middle Aged , Humans , Aged , Retinal Pigment Epithelium/metabolism , Macular Degeneration/metabolism , Cellular Senescence
11.
EMBO Rep ; 21(10): e50718, 2020 10 05.
Article in English | MEDLINE | ID: mdl-32785991

ABSTRACT

Senescent cells display senescence-associated (SA) phenotypic programs such as stable proliferation arrest (SAPA) and a secretory phenotype (SASP). Senescence-inducing persistent DNA double-strand breaks (pDSBs) cause an immediate DNA damage response (DDR) and SAPA, but the SASP requires days to develop. Here, we show that following the immediate canonical DDR, a delayed chromatin accumulation of the ATM and MRN complexes coincides with the expression of SASP factors. Importantly, histone deacetylase inhibitors (HDACi) trigger SAPA and SASP in the absence of DNA damage. However, HDACi-induced SASP also requires ATM/MRN activities and causes their accumulation on chromatin, revealing a DNA damage-independent, non-canonical DDR activity that underlies SASP maturation. This non-canonical DDR is required for the recruitment of the transcription factor NF-κB on chromatin but not for its nuclear translocation. Non-canonical DDR further does not require ATM kinase activity, suggesting structural ATM functions. We propose that delayed chromatin recruitment of SASP modulators is the result of non-canonical DDR signaling that ensures SASP activation only in the context of senescence and not in response to transient DNA damage-induced proliferation arrest.


Subject(s)
Cellular Senescence , NF-kappa B , Cellular Senescence/genetics , Chromatin/genetics , DNA Damage , NF-kappa B/metabolism , Signal Transduction
12.
Am J Physiol Heart Circ Physiol ; 321(1): H185-H196, 2021 07 01.
Article in English | MEDLINE | ID: mdl-34114892

ABSTRACT

We assessed the efficacy of oral supplementation with the flavanoid apigenin on arterial function during aging and identified critical mechanisms of action. Young (6 mo) and old (27 mo) C57BL/6N mice (model of arterial aging) consumed drinking water containing vehicle (0.2% carboxymethylcellulose; 10 young and 7 old) or apigenin (0.5 mg/mL in vehicle; 10 young and 9 old) for 6 wk. In vehicle-treated animals, isolated carotid artery endothelium-dependent dilation (EDD), bioassay of endothelial function, was impaired in old versus young (70% ± 9% vs. 92% ± 1%, P < 0.0001) due to reduced nitric oxide (NO) bioavailability. Old mice had greater arterial reactive oxygen species (ROS) production and oxidative stress (higher nitrotyrosine) associated with greater nicotinamide adenine dinucleotide phosphate oxidase (oxidant enzyme) and lower superoxide dismutase 1 and 2 (antioxidant enzymes); ex vivo administration of Tempol (antioxidant) restored EDD to young levels, indicating ROS-mediated suppression of EDD. Old animals also had greater aortic stiffness as indicated by higher aortic pulse wave velocity (PWV, 434 ± 9 vs. 346 ± 5 cm/s, P < 0.0001) due to greater intrinsic aortic wall stiffness associated with lower elastin levels and higher collagen, advanced glycation end products (AGEs), and proinflammatory cytokine abundance. In old mice, apigenin restored EDD (96% ± 2%) by increasing NO bioavailability, normalized arterial ROS, oxidative stress, and antioxidant expression, and abolished ROS inhibition of EDD. Moreover, apigenin prevented foam cell formation in vitro (initiating step in atherosclerosis) and mitigated age-associated aortic stiffening (PWV 373 ± 5 cm/s) by normalizing aortic intrinsic wall stiffness, collagen, elastin, AGEs, and inflammation. Thus, apigenin is a promising therapeutic for arterial aging.NEW & NOTEWORTHY Our study provides novel evidence that oral apigenin supplementation can reverse two clinically important indicators of arterial dysfunction with age, namely, vascular endothelial dysfunction and large elastic artery stiffening, and prevents foam cell formation in an established cell culture model of early atherosclerosis. Importantly, our results provide extensive insight into the biological mechanisms of apigenin action, including increased nitric oxide bioavailability, normalization of age-related increases in arterial ROS production and oxidative stress, reversal of age-associated aortic intrinsic mechanical wall stiffening and adverse remodeling of the extracellular matrix, and suppression of vascular inflammation. Given that apigenin is commercially available as a dietary supplement in humans, these preclinical findings provide the experimental basis for future translational studies assessing the potential of apigenin to treat arterial dysfunction and reduce cardiovascular disease risk with aging.


Subject(s)
Aging/metabolism , Endothelium, Vascular/drug effects , Inflammation/metabolism , Oxidative Stress/drug effects , Spirostans/pharmacology , Vascular Stiffness/drug effects , Animals , Aorta/drug effects , Aorta/metabolism , Blood Pressure/drug effects , Blood Pressure/physiology , Carotid Arteries/drug effects , Carotid Arteries/metabolism , Endothelium, Vascular/metabolism , Mice , Mice, Inbred C57BL , Reactive Oxygen Species/metabolism
13.
Blood ; 133(5): 446-456, 2019 01 31.
Article in English | MEDLINE | ID: mdl-30401703

ABSTRACT

Acute myeloid leukemia (AML) is an age-related disease that is highly dependent on the bone marrow (BM) microenvironment. With increasing age, tissues accumulate senescent cells, characterized by an irreversible arrest of cell proliferation and the secretion of a set of proinflammatory cytokines, chemokines, and growth factors, collectively known as the senescence-associated secretory phenotype (SASP). Here, we report that AML blasts induce a senescent phenotype in the stromal cells within the BM microenvironment and that the BM stromal cell senescence is driven by p16INK4a expression. The p16INK4a-expressing senescent stromal cells then feed back to promote AML blast survival and proliferation via the SASP. Importantly, selective elimination of p16INK4a+ senescent BM stromal cells in vivo improved the survival of mice with leukemia. Next, we find that the leukemia-driven senescent tumor microenvironment is caused by AML-induced NOX2-derived superoxide. Finally, using the p16-3MR mouse model, we show that by targeting NOX2 we reduced BM stromal cell senescence and consequently reduced AML proliferation. Together, these data identify leukemia-generated NOX2-derived superoxide as a driver of protumoral p16INK4a-dependent senescence in BM stromal cells. Our findings reveal the importance of a senescent microenvironment for the pathophysiology of leukemia. These data now open the door to investigate drugs that specifically target the "benign" senescent cells that surround and support AML.


Subject(s)
Bone Marrow/pathology , Cellular Senescence , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Leukemia, Myeloid, Acute/pathology , Tumor Microenvironment , Animals , Bone Marrow/metabolism , Cell Proliferation , Coculture Techniques , Female , Humans , Leukemia, Myeloid, Acute/metabolism , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/pathology , Mice, Inbred C57BL , NADPH Oxidase 2/metabolism , Superoxides/metabolism , Tumor Cells, Cultured
14.
Expert Rev Proteomics ; 17(4): 297-308, 2020 04.
Article in English | MEDLINE | ID: mdl-32425074

ABSTRACT

INTRODUCTION: Cellular senescence is a rapidly growing field with potential relevance for the treatment of multiple human diseases. In the last decade, cellular senescence and the senescence-associated secretory phenotype (SASP) have emerged as central drivers of aging and many chronic diseases, including cancer, neurodegeneration, heart disease and osteoarthritis. Major efforts are underway to develop drugs that selectively eliminate senescent cells (senolytics) or alter the SASP (senomorphics) to treat age-related diseases in humans. The translation of senescence-targeting therapies into humans is still in early stages. Nonetheless, it is clear that proteomic approaches will facilitate the discovery of important SASP proteins, development of senescence- and SASP-derived biomarkers, and identification of therapeutic targets for senolytic and senomorphic drugs. AREAS COVERED: We review recent proteomic studies of cellular senescence and their translational relevance and, particularly, characterization of the secretory phenotype and preclinical development of biomarkers (from 2008-2020, PubMed). We focus on emerging areas, such as the heterogeneity of senescent cells and the SASP, extracellular vesicles released by senescent cells, and validating biomarkers of aging in vivo. EXPERT OPINION: Proteomic and multi-omic approaches will be important for the development of senescence-based biomarkers to facilitate and monitor future therapeutic interventions that target senescent cells.


Subject(s)
Cellular Senescence , Exosomes/metabolism , Proteome/metabolism , Proteomics/methods , Animals , Exosomes/chemistry , Humans , Phenotype , Proteome/chemistry
15.
Immun Ageing ; 17: 16, 2020.
Article in English | MEDLINE | ID: mdl-32518575

ABSTRACT

Cellular senescence is an essentially irreversible arrest of cell proliferation coupled to a complex senescence-associated secretory phenotype (SASP). The senescence arrest prevents the development of cancer, and the SASP can promote tissue repair. Recent data suggest that the prolonged presence of senescent cells, and especially the SASP, could be deleterious, and their beneficial effects early in life can become maladaptive such that they drive aging phenotypes and pathologies late in life. It is therefore important to develop strategies to eliminate senescent cells. There are currently under development or approved several immune cell-based therapies for cancer, which could be redesigned to target senescent cells. This review focuses on this possible use of immune cells and discusses how current cell-based therapies could be used for senescent cell removal.

16.
Nat Rev Mol Cell Biol ; 8(9): 729-40, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17667954

ABSTRACT

Cells continually experience stress and damage from exogenous and endogenous sources, and their responses range from complete recovery to cell death. Proliferating cells can initiate an additional response by adopting a state of permanent cell-cycle arrest that is termed cellular senescence. Understanding the causes and consequences of cellular senescence has provided novel insights into how cells react to stress, especially genotoxic stress, and how this cellular response can affect complex organismal processes such as the development of cancer and ageing.


Subject(s)
Aging , Cellular Senescence/physiology , Neoplasms/etiology , Animals , Cell Cycle , DNA Damage , Humans
17.
Proc Natl Acad Sci U S A ; 112(33): 10407-12, 2015 Aug 18.
Article in English | MEDLINE | ID: mdl-26240345

ABSTRACT

Tissue homeostasis declines with age partly because stem/progenitor cells fail to self-renew or differentiate. Because mitochondrial damage can accelerate aging, we tested the hypothesis that mitochondrial dysfunction impairs stem cell renewal or function. We developed a mouse model, Tg(KRT14-cre/Esr1) (20Efu/J) × Sod2 (tm1Smel) , that generates mitochondrial oxidative stress in keratin 14-expressing epidermal stem/progenitor cells in a temporally controlled manner owing to deletion of Sod2, a nuclear gene that encodes the mitochondrial antioxidant enzyme superoxide dismutase 2 (Sod2). Epidermal Sod2 loss induced cellular senescence, which irreversibly arrested proliferation in a fraction of keratinocytes. Surprisingly, in young mice, Sod2 deficiency accelerated wound closure, increasing epidermal differentiation and reepithelialization, despite the reduced proliferation. In contrast, at older ages, Sod2 deficiency delayed wound closure and reduced epidermal thickness, accompanied by epidermal stem cell exhaustion. In young mice, Sod2 deficiency accelerated epidermal thinning in response to the tumor promoter 12-O-tetradecanoylphorbol-13-acetate, phenocopying the reduced regeneration of older Sod2-deficient skin. Our results show a surprising beneficial effect of mitochondrial dysfunction at young ages, provide a potential mechanism for the decline in epidermal regeneration at older ages, and identify a previously unidentified age-dependent role for mitochondria in skin quality and wound closure.


Subject(s)
Epidermis/pathology , Mitochondria/metabolism , Oxidative Stress , Skin Aging , Stem Cells/cytology , Alleles , Animals , Cell Differentiation , Cell Proliferation , Cellular Senescence , Collagen/chemistry , DNA Primers , Gene Deletion , Gene Expression Profiling , Genetic Pleiotropy , Genotype , Homeostasis , Humans , Keratinocytes/cytology , Mice , Mice, Transgenic , Mitochondria/pathology , Phenotype , Superoxide Dismutase/metabolism , Time Factors , Wound Healing
18.
Eur J Immunol ; 46(10): 2286-2301, 2016 10.
Article in English | MEDLINE | ID: mdl-27595500

ABSTRACT

Major advances in preventing, delaying, or curing individual pathologies are responsible for an increasingly long life span in the developed parts of our planet, and indeed reaching eight to nine decades of life is nowadays extremely frequent. However, medical and sanitary advances have not prevented or delayed the underlying cause of the disparate pathologies occurring in the elderly: aging itself. The identification of the basis of the aging processes that drives the multiple pathologies and loss of function typical of older individuals is a major challenge in current aging research. Among the possible causes, an impairment of the immune system plays a major role, and indeed numerous studies have described immunological changes which occur with age. Far from the intention of being exhaustive, this review will focus on recent advances and views on the role that modifications of cell signalling and remodelling of the immune response play during human aging and longevity, paying particular attention to phenomena which are linked to the so called inflammaging process, such as dysregulation of innate immunity, altered T-cell or B-cell maturation and differentiation, as well as to the implications of immune aging for vaccination strategies in the elderly.


Subject(s)
Aging , B-Lymphocytes/immunology , Immune System , Inflammation , T-Lymphocytes/immunology , Aged , Animals , Humans , Immunity, Cellular , Immunity, Innate , Longevity , Vaccination
19.
Annu Rev Physiol ; 75: 685-705, 2013.
Article in English | MEDLINE | ID: mdl-23140366

ABSTRACT

For most species, aging promotes a host of degenerative pathologies that are characterized by debilitating losses of tissue or cellular function. However, especially among vertebrates, aging also promotes hyperplastic pathologies, the most deadly of which is cancer. In contrast to the loss of function that characterizes degenerating cells and tissues, malignant (cancerous) cells must acquire new (albeit aberrant) functions that allow them to develop into a lethal tumor. This review discusses the idea that, despite seemingly opposite characteristics, the degenerative and hyperplastic pathologies of aging are at least partly linked by a common biological phenomenon: a cellular stress response known as cellular senescence. The senescence response is widely recognized as a potent tumor suppressive mechanism. However, recent evidence strengthens the idea that it also drives both degenerative and hyperplastic pathologies, most likely by promoting chronic inflammation. Thus, the senescence response may be the result of antagonistically pleiotropic gene action.


Subject(s)
Aging/physiology , Cellular Senescence/physiology , Neoplasms/physiopathology , Animals , DNA Damage/physiology , Disease Models, Animal , Humans , Stress, Physiological/physiology
SELECTION OF CITATIONS
SEARCH DETAIL