Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
2.
Article in English | MEDLINE | ID: mdl-37804247

ABSTRACT

The geroscience hypothesis suggests that addressing the fundamental mechanisms driving aging biology will prevent or mitigate the onset of multiple chronic diseases, for which the largest risk factor is advanced age. Research that investigates the root causes of aging is therefore of critical importance given the rising healthcare burden attributable to age-related diseases. The third annual Midwest Aging Consortium symposium was convened as a showcase of such research performed by investigators from institutions across the Midwestern United States. This report summarizes the work presented during a virtual conference across topics in aging biology, including immune function in the lung-particularly timely given the Corona Virus Immune Disease-2019 pandemic-along with the role of metabolism and nutrient-regulated pathways in cellular function with age, the influence of senescence on stem cell function and inflammation, and our evolving understanding of the mechanisms underlying observation of sex dimorphism in aging-related outcomes. The symposium focused on early-stage and emerging investigators, while including keynote presentations from leaders in the biology of aging field, highlighting the diversity and strength of aging research in the Midwest.


Subject(s)
Aging , Multiple Chronic Conditions , Humans , Aging/physiology , Inflammation , Lung , Geroscience
3.
Biogerontology ; 25(2): 329-339, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38150087

ABSTRACT

Over half a century has passed since Alexey Olovnikov's groundbreaking proposal of the end-replication problem in 1971, laying the foundation for our understanding of telomeres and their pivotal role in cellular senescence. This review paper delves into the intricate and multifaceted relationship between cellular senescence, the influence of telomeres in this process, and the far-reaching consequences of telomeres in the context of aging and age-related diseases. Additionally, the paper investigates the various factors that can influence telomere shortening beyond the confines of the end-replication problem and how telomeres can exert their impact on aging, even in the absence of significant shortening. Ultimately, this paper stands as a tribute to the pioneering work of Olovnikov, whose seminal contributions established the solid foundation upon which our ongoing explorations of telomeres and the aging process are based.


Subject(s)
Cellular Senescence , Telomere Shortening , Telomere
4.
bioRxiv ; 2023 Nov 21.
Article in English | MEDLINE | ID: mdl-38045344

ABSTRACT

Genomic instability and inflammation are distinct hallmarks of aging, but the connection between them is poorly understood. Understanding their interrelationship will help unravel new mechanisms and therapeutic targets of aging and age-associated diseases. Here we report a novel mechanism directly linking genomic instability and inflammation in senescent cells, through a mitochondria-regulated molecular circuit that connects the p53 tumor suppressor and cytoplasmic chromatin fragments (CCF), a driver of inflammation through the cGAS-STING pathway. Activation or inactivation of p53 by genetic and pharmacologic approaches showed that p53 suppresses CCF accumulation and the downstream inflammatory senescence-associated secretory phenotype (SASP), independent of its effects on cell cycle arrest. p53 activation suppressed CCF formation by promoting DNA repair, reflected in maintenance of genomic integrity, particularly in subtelomeric regions, as shown by single cell genome resequencing. Activation of p53 by pharmacological inhibition of MDM2 in old mice decreased features of SASP in liver, indicating a senomorphic role in vivo . Remarkably, mitochondria in senescent cells suppressed p53 activity by promoting CCF formation and thereby restricting ATM-dependent nuclear DNA damage signaling. These data provide evidence for a mitochondria-regulated p53-CCF circuit in senescent cells that controls DNA repair, genome integrity and inflammatory SASP, and is a potential target for senomorphic healthy aging interventions.

5.
Nat Cell Biol ; 25(12): 1724-1725, 2023 Dec.
Article in English | MEDLINE | ID: mdl-38012404
6.
Nature ; 622(7983): 627-636, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37821702

ABSTRACT

Senescent cells drive age-related tissue dysfunction partially through the induction of a chronic senescence-associated secretory phenotype (SASP)1. Mitochondria are major regulators of the SASP; however, the underlying mechanisms have not been elucidated2. Mitochondria are often essential for apoptosis, a cell fate distinct from cellular senescence. During apoptosis, widespread mitochondrial outer membrane permeabilization (MOMP) commits a cell to die3. Here we find that MOMP occurring in a subset of mitochondria is a feature of cellular senescence. This process, called minority MOMP (miMOMP), requires BAX and BAK macropores enabling the release of mitochondrial DNA (mtDNA) into the cytosol. Cytosolic mtDNA in turn activates the cGAS-STING pathway, a major regulator of the SASP. We find that inhibition of MOMP in vivo decreases inflammatory markers and improves healthspan in aged mice. Our results reveal that apoptosis and senescence are regulated by similar mitochondria-dependent mechanisms and that sublethal mitochondrial apoptotic stress is a major driver of the SASP. We provide proof-of-concept that inhibition of miMOMP-induced inflammation may be a therapeutic route to improve healthspan.


Subject(s)
Apoptosis , Cellular Senescence , Cytosol , DNA, Mitochondrial , Mitochondria , Animals , Mice , Cytosol/metabolism , DNA, Mitochondrial/metabolism , Mitochondria/genetics , Mitochondria/metabolism , Mitochondrial Transmembrane Permeability-Driven Necrosis , Proof of Concept Study , Inflammation/metabolism , Phenotype , Longevity , Healthy Aging
7.
Regen Biomater ; 10: rbad060, 2023.
Article in English | MEDLINE | ID: mdl-37501679

ABSTRACT

Human lifespan continues to extend as an unprecedented number of people reach their seventh and eighth decades of life, unveiling chronic conditions that affect the older adult. Age-related skin conditions include senile purpura, seborrheic keratoses, pemphigus vulgaris, bullous pemphigoid, diabetic foot wounds and skin cancer. Current methods of drug testing prior to clinical trials require the use of pre-clinical animal models, which are often unable to adequately replicate human skin response. Therefore, a reliable model for aged human skin is needed. The current challenges in developing an aged human skin model include the intrinsic variability in skin architecture from person to person. An ideal skin model would incorporate innate functionality such as sensation, vascularization and regeneration. The advent of 3D bioprinting allows us to create human skin equivalent for use as clinical-grade surgical graft, for drug testing and other needs. In this review, we describe the process of human skin aging and outline the steps to create an aged skin model with 3D bioprinting using skin cells (i.e. keratinocytes, fibroblasts and melanocytes). We also provide an overview of current bioprinted skin models, associated limitations and direction for future research.

8.
Nat Aging ; 3(7): 776-790, 2023 07.
Article in English | MEDLINE | ID: mdl-37400722

ABSTRACT

Cellular senescence is a well-established driver of aging and age-related diseases. There are many challenges to mapping senescent cells in tissues such as the absence of specific markers and their relatively low abundance and vast heterogeneity. Single-cell technologies have allowed unprecedented characterization of senescence; however, many methodologies fail to provide spatial insights. The spatial component is essential, as senescent cells communicate with neighboring cells, impacting their function and the composition of extracellular space. The Cellular Senescence Network (SenNet), a National Institutes of Health (NIH) Common Fund initiative, aims to map senescent cells across the lifespan of humans and mice. Here, we provide a comprehensive review of the existing and emerging methodologies for spatial imaging and their application toward mapping senescent cells. Moreover, we discuss the limitations and challenges inherent to each technology. We argue that the development of spatially resolved methods is essential toward the goal of attaining an atlas of senescent cells.


Subject(s)
Aging , Cellular Senescence , United States , Humans , Animals , Mice , Longevity
9.
Am J Pathol ; 193(1): 11-26, 2023 01.
Article in English | MEDLINE | ID: mdl-36243043

ABSTRACT

Patients with cholestatic liver disease, including those with primary biliary cholangitis, can experience symptoms of impaired cognition or brain fog. This phenomenon remains unexplained and is currently untreatable. Bile duct ligation (BDL) is an established rodent model of cholestasis. In addition to liver changes, BDL animals develop cognitive symptoms early in the disease process (before development of cirrhosis and/or liver failure). The cellular mechanisms underpinning these cognitive symptoms are poorly understood. Herein, the study explored the neurocognitive symptom manifestations, and tested potential therapies, in BDL mice, and used human neuronal cell cultures to explore translatability to humans. BDL animals exhibited short-term memory loss and showed reduced astrocyte coverage of the blood-brain barrier, destabilized hippocampal network activity, and neuronal senescence. Ursodeoxycholic acid (first-line therapy for most human cholestatic diseases) did not reverse symptomatic or mechanistic aspects. In contrast, obeticholic acid (OCA), a farnesoid X receptor agonist and second-line anti-cholestatic agent, normalized memory function, suppressed blood-brain barrier changes, prevented hippocampal network deficits, and reversed neuronal senescence. Co-culture of human neuronal cells with either BDL or human cholestatic patient serum induced cellular senescence and increased mitochondrial respiration, changes that were limited again by OCA. These findings provide new insights into the mechanism of cognitive symptoms in BDL animals, suggesting that OCA therapy or farnesoid X receptor agonism could be used to limit cholestasis-induced neuronal senescence.


Subject(s)
Cholestasis , Memory, Short-Term , Humans , Mice , Animals , Cholestasis/drug therapy , Chenodeoxycholic Acid/pharmacology , Bile Ducts/surgery , Liver , Ligation
10.
Cell Rep ; 41(6): 111596, 2022 11 08.
Article in English | MEDLINE | ID: mdl-36351380

ABSTRACT

Targeting early-stage lung cancer is vital to improve survival. However, the mechanisms and components of the early tumor suppressor response in lung cancer are not well understood. In this report, we study the role of Toll-like receptor 2 (TLR2), a regulator of oncogene-induced senescence, which is a key tumor suppressor response in premalignancy. Using human lung cancer samples and genetically engineered mouse models, we show that TLR2 is active early in lung tumorigenesis, where it correlates with improved survival and clinical regression. Mechanistically, TLR2 impairs early lung cancer progression via activation of cell intrinsic cell cycle arrest pathways and the proinflammatory senescence-associated secretory phenotype (SASP). The SASP regulates non-cell autonomous anti-tumor responses, such as immune surveillance of premalignant cells, and we observe impaired myeloid cell recruitment to lung tumors after Tlr2 loss. Last, we show that administration of a TLR2 agonist reduces lung tumor growth, highlighting TLR2 as a possible therapeutic target.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Mice , Animals , Humans , Toll-Like Receptor 2/genetics , Toll-Like Receptor 2/metabolism , Carcinoma, Non-Small-Cell Lung/genetics , Lung Neoplasms/genetics , Genes, Tumor Suppressor , Lung/metabolism , Cellular Senescence/genetics
11.
Cell ; 184(22): 5506-5526, 2021 10 28.
Article in English | MEDLINE | ID: mdl-34715021

ABSTRACT

Endogenous cytoplasmic DNA (cytoDNA) species are emerging as key mediators of inflammation in diverse physiological and pathological contexts. Although the role of endogenous cytoDNA in innate immune activation is well established, the cytoDNA species themselves are often poorly characterized and difficult to distinguish, and their mechanisms of formation, scope of function and contribution to disease are incompletely understood. Here, we summarize current knowledge in this rapidly progressing field with emphases on similarities and differences between distinct cytoDNAs, their underlying molecular mechanisms of formation and function, interactions between cytoDNA pathways, and therapeutic opportunities in the treatment of age-associated diseases.


Subject(s)
Aging/metabolism , Cytoplasm/metabolism , DNA/metabolism , Disease , Animals , Humans , Micronucleus, Germline/metabolism , Retroelements/genetics
12.
EMBO J ; 40(9): e106048, 2021 05 03.
Article in English | MEDLINE | ID: mdl-33764576

ABSTRACT

Cellular senescence is characterized by an irreversible cell cycle arrest as well as a pro-inflammatory phenotype, thought to contribute to aging and age-related diseases. Neutrophils have essential roles in inflammatory responses; however, in certain contexts their abundance is associated with a number of age-related diseases, including liver disease. The relationship between neutrophils and cellular senescence is not well understood. Here, we show that telomeres in non-immune cells are highly susceptible to oxidative damage caused by neighboring neutrophils. Neutrophils cause telomere dysfunction both in vitro and ex vivo in a ROS-dependent manner. In a mouse model of acute liver injury, depletion of neutrophils reduces telomere dysfunction and senescence. Finally, we show that senescent cells mediate the recruitment of neutrophils to the aged liver and propose that this may be a mechanism by which senescence spreads to surrounding cells. Our results suggest that interventions that counteract neutrophil-induced senescence may be beneficial during aging and age-related disease.


Subject(s)
Acute Lung Injury/immunology , Carbon Tetrachloride/adverse effects , Neutrophils/cytology , Reactive Oxygen Species/metabolism , Telomere Shortening , Acute Lung Injury/chemically induced , Acute Lung Injury/metabolism , Animals , Cell Line , Cellular Senescence , Coculture Techniques , Disease Models, Animal , Female , Fibroblasts/cytology , Fibroblasts/metabolism , Humans , Male , Mice , Neutrophils/metabolism , Oxidative Stress , Paracrine Communication
13.
Aging Cell ; 20(2): e13296, 2021 02.
Article in English | MEDLINE | ID: mdl-33470505

ABSTRACT

Cellular senescence is characterized by an irreversible cell cycle arrest and a pro-inflammatory senescence-associated secretory phenotype (SASP), which is a major contributor to aging and age-related diseases. Clearance of senescent cells has been shown to improve brain function in mouse models of neurodegenerative diseases. However, it is still unknown whether senescent cell clearance alleviates cognitive dysfunction during the aging process. To investigate this, we first conducted single-nuclei and single-cell RNA-seq in the hippocampus from young and aged mice. We observed an age-dependent increase in p16Ink4a senescent cells, which was more pronounced in microglia and oligodendrocyte progenitor cells and characterized by a SASP. We then aged INK-ATTAC mice, in which p16Ink4a -positive senescent cells can be genetically eliminated upon treatment with the drug AP20187 and treated them either with AP20187 or with the senolytic cocktail Dasatinib and Quercetin. We observed that both strategies resulted in a decrease in p16Ink4a exclusively in the microglial population, resulting in reduced microglial activation and reduced expression of SASP factors. Importantly, both approaches significantly improved cognitive function in aged mice. Our data provide proof-of-concept for senolytic interventions' being a potential therapeutic avenue for alleviating age-associated cognitive impairment.


Subject(s)
Cognitive Dysfunction/pathology , Encephalitis/pathology , Age Factors , Animals , Cellular Senescence , Cognitive Dysfunction/metabolism , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Encephalitis/metabolism , Mice , Mice, Transgenic
14.
Cell Cycle ; 19(5): 532-540, 2020 03.
Article in English | MEDLINE | ID: mdl-32065062

ABSTRACT

Cellular senescence is an irreversible cell cycle arrest, which can be triggered by a number of stressors, including telomere damage. Among many other phenotypic changes, senescence is accompanied by increased secretion of pro-inflammatory molecules, also known as the senescence-associated secretory phenotype (SASP). It is thought that accumulation of senescent cells contributes to age-associated tissue dysfunction partly by inducing senescence in neighboring cells through mechanisms involving SASP factors. Here, we will review evidence suggesting that telomeres can become dysfunctional irrespectively of shortening, and that this may be a mechanism-driving senescence in post-mitotic or slow dividing cells. Furthermore, we review recent evidence that supports that senescent melanocytes induce paracrine telomere damage during skin aging, which may be the mechanism responsible for propagation of senescent cells. We propose that telomeres are sensors of imbalances in the cellular milieu and act as beacons of stress, contributing to autocrine and paracrine senescence.


Subject(s)
Autocrine Communication , Cellular Senescence , DNA Damage , Melanocytes/pathology , Paracrine Communication , Skin Aging/pathology , Telomere/pathology , Animals , Bystander Effect , Cellular Senescence/genetics , DNA Damage/genetics , Humans , Skin Aging/genetics , Stress, Physiological
16.
EMBO J ; 38(23): e101982, 2019 12 02.
Article in English | MEDLINE | ID: mdl-31633821

ABSTRACT

Cellular senescence has been shown to contribute to skin ageing. However, the role of melanocytes in the process is understudied. Our data show that melanocytes are the only epidermal cell type to express the senescence marker p16INK4A during human skin ageing. Aged melanocytes also display additional markers of senescence such as reduced HMGB1 and dysfunctional telomeres, without detectable telomere shortening. Additionally, senescent melanocyte SASP induces telomere dysfunction in paracrine manner and limits proliferation of surrounding cells via activation of CXCR3-dependent mitochondrial ROS. Finally, senescent melanocytes impair basal keratinocyte proliferation and contribute to epidermal atrophy in vitro using 3D human epidermal equivalents. Crucially, clearance of senescent melanocytes using the senolytic drug ABT737 or treatment with mitochondria-targeted antioxidant MitoQ suppressed this effect. In conclusion, our study provides proof-of-concept evidence that senescent melanocytes affect keratinocyte function and act as drivers of human skin ageing.


Subject(s)
Aging/pathology , Atrophy/pathology , Cellular Senescence , Melanocytes/pathology , Skin/pathology , Telomere/pathology , Adult , Aged , Aged, 80 and over , Aging/drug effects , Atrophy/chemically induced , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Epidermis/drug effects , Epidermis/pathology , Female , Humans , Male , Melanocytes/metabolism , Middle Aged , Paracrine Communication , Reactive Oxygen Species/metabolism , Receptors, CXCR4/metabolism , Skin/metabolism , Telomere/metabolism , Young Adult
17.
EBioMedicine ; 47: 446-456, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31542391

ABSTRACT

BACKGROUND: Senescent cells, which can release factors that cause inflammation and dysfunction, the senescence-associated secretory phenotype (SASP), accumulate with ageing and at etiological sites in multiple chronic diseases. Senolytics, including the combination of Dasatinib and Quercetin (D + Q), selectively eliminate senescent cells by transiently disabling pro-survival networks that defend them against their own apoptotic environment. In the first clinical trial of senolytics, D + Q improved physical function in patients with idiopathic pulmonary fibrosis (IPF), a fatal senescence-associated disease, but to date, no peer-reviewed study has directly demonstrated that senolytics decrease senescent cells in humans. METHODS: In an open label Phase 1 pilot study, we administered 3 days of oral D 100 mg and Q 1000 mg to subjects with diabetic kidney disease (N = 9; 68·7 ±â€¯3·1 years old; 2 female; BMI:33·9 ±â€¯2·3 kg/m2; eGFR:27·0 ±â€¯2·1 mL/min/1·73m2). Adipose tissue, skin biopsies, and blood were collected before and 11 days after completing senolytic treatment. Senescent cell and macrophage/Langerhans cell markers and circulating SASP factors were assayed. FINDINGS: D + Q reduced adipose tissue senescent cell burden within 11 days, with decreases in p16INK4A-and p21CIP1-expressing cells, cells with senescence-associated ß-galactosidase activity, and adipocyte progenitors with limited replicative potential. Adipose tissue macrophages, which are attracted, anchored, and activated by senescent cells, and crown-like structures were decreased. Skin epidermal p16INK4A+ and p21CIP1+ cells were reduced, as were circulating SASP factors, including IL-1α, IL-6, and MMPs-9 and -12. INTERPRETATION: "Hit-and-run" treatment with senolytics, which in the case of D + Q have elimination half-lives <11 h, significantly decreases senescent cell burden in humans. FUND: NIH and Foundations. ClinicalTrials.gov Identifier: NCT02848131. Senescence, Frailty, and Mesenchymal Stem Cell Functionality in Chronic Kidney Disease: Effect of Senolytic Agents.


Subject(s)
Cellular Senescence/drug effects , Dasatinib/pharmacology , Diabetic Nephropathies/metabolism , Quercetin/pharmacology , Adipocytes/drug effects , Adipocytes/metabolism , Adipose Tissue/drug effects , Adipose Tissue/metabolism , Aged , Biomarkers , Biopsy , Clinical Trials, Phase I as Topic , Dasatinib/therapeutic use , Diabetic Nephropathies/diagnosis , Diabetic Nephropathies/drug therapy , Drug Therapy, Combination , Female , Humans , Immunohistochemistry , Kidney Function Tests , Macrophages/drug effects , Macrophages/metabolism , Male , Middle Aged , Quercetin/therapeutic use
19.
Aging Cell ; 18(3): e12945, 2019 06.
Article in English | MEDLINE | ID: mdl-30920115

ABSTRACT

Cardiovascular disease is the leading cause of death in individuals over 60 years old. Aging is associated with an increased prevalence of coronary artery disease and a poorer prognosis following acute myocardial infarction (MI). With age, senescent cells accumulate in tissues, including the heart, and contribute to age-related pathologies. However, the role of senescence in recovery following MI has not been investigated. In this study, we demonstrate that treatment of aged mice with the senolytic drug, navitoclax, eliminates senescent cardiomyocytes and attenuates profibrotic protein expression in aged mice. Importantly, clearance of senescent cells improved myocardial remodelling and diastolic function as well as overall survival following MI. These data provide proof-of-concept evidence that senescent cells are major contributors to impaired function and increased mortality following MI and that senolytics are a potential new therapeutic avenue for MI.


Subject(s)
Aging/drug effects , Aniline Compounds/pharmacology , Antineoplastic Agents/pharmacology , Cellular Senescence/drug effects , Myocardial Infarction/drug therapy , Sulfonamides/pharmacology , Acute Disease , Aniline Compounds/administration & dosage , Animals , Antineoplastic Agents/administration & dosage , Cell Survival/drug effects , Male , Mice , Mice, Inbred C57BL , Myocardial Infarction/pathology , Sulfonamides/administration & dosage
20.
EMBO J ; 38(5)2019 03 01.
Article in English | MEDLINE | ID: mdl-30737259

ABSTRACT

Ageing is the biggest risk factor for cardiovascular disease. Cellular senescence, a process driven in part by telomere shortening, has been implicated in age-related tissue dysfunction. Here, we address the question of how senescence is induced in rarely dividing/post-mitotic cardiomyocytes and investigate whether clearance of senescent cells attenuates age-related cardiac dysfunction. During ageing, human and murine cardiomyocytes acquire a senescent-like phenotype characterised by persistent DNA damage at telomere regions that can be driven by mitochondrial dysfunction and crucially can occur independently of cell division and telomere length. Length-independent telomere damage in cardiomyocytes activates the classical senescence-inducing pathways, p21CIP and p16INK4a, and results in a non-canonical senescence-associated secretory phenotype, which is pro-fibrotic and pro-hypertrophic. Pharmacological or genetic clearance of senescent cells in mice alleviates detrimental features of cardiac ageing, including myocardial hypertrophy and fibrosis. Our data describe a mechanism by which senescence can occur and contribute to age-related myocardial dysfunction and in the wider setting to ageing in post-mitotic tissues.


Subject(s)
Cardiomegaly/pathology , Cellular Senescence , DNA Damage , Fibrosis/pathology , Mitosis , Myocytes, Cardiac/pathology , Telomere Shortening , Aging , Animals , Cardiomegaly/etiology , Female , Fibrosis/etiology , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Monoamine Oxidase/physiology , Myocytes, Cardiac/metabolism , Phenotype , RNA/physiology , Rats, Sprague-Dawley , Telomerase/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...