Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Aging Cell ; 16(3): 480-487, 2017 06.
Article in English | MEDLINE | ID: mdl-28229533

ABSTRACT

With aging, there is progressive loss of tissue homeostasis and functional reserve, leading to an impaired response to stress and an increased risk of morbidity and mortality. A key mediator of the cellular response to damage and stress is the transcription factor NF-κB. We demonstrated previously that NF-κB transcriptional activity is upregulated in tissues from both natural aged mice and in a mouse model of a human progeroid syndrome caused by defective repair of DNA damage (ERCC1-deficient mice). We also demonstrated that genetic reduction in the level of the NF-κB subunit p65(RelA) in the Ercc1-/∆ progeroid mouse model of accelerated aging delayed the onset of age-related pathology including muscle wasting, osteoporosis, and intervertebral disk degeneration. Here, we report that the largest fraction of NF-κB -expressing cells in the bone marrow (BM) of aged (>2 year old) mice (C57BL/6-NF-κBEGFP reporter mice) are Gr-1+ CD11b+ myeloid-derived suppressor cells (MDSCs). There was a significant increase in the overall percentage of MDSC present in the BM of aged animals compared with young, a trend also observed in the spleen. However, the function of these cells appears not to be compromised in aged mice. A similar increase of MDSC was observed in BM of progeroid Ercc1-/∆ and BubR1H/H mice. The increase in MDSC in Ercc1-/∆ mice was abrogated by heterozygosity in the p65/RelA subunit of NF-κB. These results suggest that NF-κB activation with aging, at least in part, drives an increase in the percentage of MDSCs, a cell type able to suppress immune cell responses.


Subject(s)
Aging/metabolism , Bone Marrow Cells/metabolism , Cell Proliferation/genetics , Myeloid-Derived Suppressor Cells/metabolism , Transcription Factor RelA/genetics , Aging/genetics , Animals , Bone Marrow Cells/cytology , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Endonucleases/deficiency , Endonucleases/genetics , Gene Expression Regulation , Genes, Reporter , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myeloid-Derived Suppressor Cells/cytology , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Spleen/cytology , Spleen/metabolism , Transcription Factor RelA/metabolism , Transcription, Genetic
2.
J Orthop Res ; 31(7): 1150-7, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23389888

ABSTRACT

Oxidative damage is a well-established driver of aging. Evidence of oxidative stress exists in aged and degenerated discs, but it is unclear how it affects disc metabolism. In this study, we first determined whether oxidative stress negatively impacts disc matrix metabolism using disc organotypic and cell cultures. Mouse disc organotypic culture grown at atmospheric oxygen (20% O(2)) exhibited perturbed disc matrix homeostasis, including reduced proteoglycan synthesis and enhanced expression of matrix metalloproteinases, compared to discs grown at low oxygen levels (5% O(2)). Human disc cells grown at 20% O(2) showed increased levels of mitochondrial-derived superoxide anions and perturbed matrix homeostasis. Treatment of disc cells with the mitochondria-targeted reactive oxygen species (ROS) scavenger XJB-5-131 blunted the adverse effects caused by 20% O(2). Importantly, we demonstrated that treatment of accelerated aging Ercc1(-/Δ) mice, previously established to be a useful in vivo model to study age-related intervertebral disc degeneration (IDD), also resulted in improved disc total glycosaminoglycan content and proteoglycan synthesis. This demonstrates that mitochondrial-derived ROS contributes to age-associated IDD in Ercc1(-/Δ) mice. Collectively, these data provide strong experimental evidence that mitochondrial-derived ROS play a causal role in driving changes linked to aging-related IDD and a potentially important role for radical scavengers in preventing IDD.


Subject(s)
Aging/metabolism , Intervertebral Disc Degeneration/metabolism , Mitochondria/metabolism , Oxidative Stress/physiology , Reactive Oxygen Species/metabolism , Adult , Animals , Cells, Cultured , Cyclic N-Oxides/pharmacology , Disease Models, Animal , Free Radical Scavengers/pharmacology , Glycosaminoglycans/metabolism , Homeostasis/drug effects , Homeostasis/physiology , Humans , Intervertebral Disc Degeneration/pathology , Matrix Metalloproteinases/metabolism , Mice , Mice, Inbred C57BL , Middle Aged , Mitochondria/drug effects , Oxidative Stress/drug effects , Oxygen/pharmacology , Proteoglycans/metabolism , Tissue Culture Techniques
3.
J Bone Miner Res ; 28(5): 1214-28, 2013 May.
Article in English | MEDLINE | ID: mdl-23281008

ABSTRACT

Advanced age is one of the most important risk factors for osteoporosis. Accumulation of oxidative DNA damage has been proposed to contribute to age-related deregulation of osteoblastic and osteoclastic cells. Excision repair cross complementary group 1-xeroderma pigmentosum group F (ERCC1-XPF) is an evolutionarily conserved structure-specific endonuclease that is required for multiple DNA repair pathways. Inherited mutations affecting expression of ERCC1-XPF cause a severe progeroid syndrome in humans, including early onset of osteopenia and osteoporosis, or anomalies in skeletal development. Herein, we used progeroid ERCC1-XPF-deficient mice, including Ercc1-null (Ercc1(-/-)) and hypomorphic (Ercc1(-/Δ)) mice, to investigate the mechanism by which DNA damage leads to accelerated bone aging. Compared to their wild-type littermates, both Ercc1(-/-) and Ercc1(-/Δ) mice display severe, progressive osteoporosis caused by reduced bone formation and enhanced osteoclastogenesis. ERCC1 deficiency leads to atrophy of osteoblastic progenitors in the bone marrow stromal cell (BMSC) population. There is increased cellular senescence of BMSCs and osteoblastic cells, as characterized by reduced proliferation, accumulation of DNA damage, and a senescence-associated secretory phenotype (SASP). This leads to enhanced secretion of inflammatory cytokines known to drive osteoclastogenesis, such as interleukin-6 (IL-6), tumor necrosis factor α (TNFα), and receptor activator of NF-κB ligand (RANKL), and thereby induces an inflammatory bone microenvironment favoring osteoclastogenesis. Furthermore, we found that the transcription factor NF-κB is activated in osteoblastic and osteoclastic cells of the Ercc1 mutant mice. Importantly, we demonstrated that haploinsufficiency of the p65 NF-κB subunit partially rescued the osteoporosis phenotype of Ercc1(-/Δ) mice. Finally, pharmacological inhibition of the NF-κB signaling via an I-κB kinase (IKK) inhibitor reversed cellular senescence and SASP in Ercc1(-/Δ) BMSCs. These results demonstrate that DNA damage drives osteoporosis through an NF-κB-dependent mechanism. Therefore, the NF-κB pathway represents a novel therapeutic target to treat aging-related bone disease.


Subject(s)
Bone and Bones/physiology , DNA Damage , NF-kappa B/metabolism , Animals , Cell Differentiation , Cells, Cultured , DNA-Binding Proteins/genetics , Endonucleases/genetics , Mice , Mice, Knockout , Osteoblasts/pathology , Osteoporosis/genetics
4.
Mech Ageing Dev ; 134(1-2): 35-42, 2013.
Article in English | MEDLINE | ID: mdl-23262094

ABSTRACT

Intervertebral disc degeneration (IDD) is the leading cause of debilitating spinal disorders such as chronic lower back pain. Aging is the greatest risk factor for IDD. Previously, we demonstrated IDD in a murine model of a progeroid syndrome caused by reduced expression of a key DNA repair enzyme. This led us to hypothesize that DNA damage promotes IDD. To test our hypothesis, we chronically exposed adult wild-type (Wt) and DNA repair-deficient Ercc1(-/Δ) mice to the cancer therapeutic agent mechlorethamine (MEC) or ionization radiation (IR) to induce DNA damage and measured the impact on disc structure. Proteoglycan, a major structural matrix constituent of the disc, was reduced 3-5× in the discs of MEC- and IR-exposed animals compared to untreated controls. Expression of the protease ADAMTS4 and aggrecan proteolytic fragments was significantly increased. Additionally, new PG synthesis was reduced 2-3× in MEC- and IR-treated discs compared to untreated controls. Both cellular senescence and apoptosis were increased in discs of treated animals. The effects were more severe in the DNA repair-deficient Ercc1(-/Δ) mice than in Wt littermates. Local irradiation of the vertebra in Wt mice elicited a similar reduction in PG. These data demonstrate that genotoxic stress drives degenerative changes associated with IDD.


Subject(s)
Aging/metabolism , DNA Damage , DNA Repair , Intervertebral Disc Degeneration/metabolism , Intervertebral Disc/metabolism , ADAM Proteins/biosynthesis , ADAM Proteins/genetics , ADAMTS4 Protein , Aggrecans/genetics , Aggrecans/metabolism , Aging/genetics , Aging/pathology , Alkylating Agents/pharmacology , Animals , Apoptosis/drug effects , Apoptosis/genetics , Apoptosis/radiation effects , Cellular Senescence/drug effects , Cellular Senescence/genetics , Cellular Senescence/radiation effects , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/genetics , Endonucleases/biosynthesis , Endonucleases/genetics , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Enzymologic/genetics , Gene Expression Regulation, Enzymologic/radiation effects , Intervertebral Disc/pathology , Intervertebral Disc Degeneration/drug therapy , Intervertebral Disc Degeneration/genetics , Intervertebral Disc Degeneration/pathology , Mechlorethamine/pharmacology , Mice , Mice, Knockout , Procollagen N-Endopeptidase/biosynthesis , Procollagen N-Endopeptidase/genetics , Radiation, Ionizing
5.
J Clin Invest ; 122(7): 2601-12, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22706308

ABSTRACT

The accumulation of cellular damage, including DNA damage, is thought to contribute to aging-related degenerative changes, but how damage drives aging is unknown. XFE progeroid syndrome is a disease of accelerated aging caused by a defect in DNA repair. NF-κB, a transcription factor activated by cellular damage and stress, has increased activity with aging and aging-related chronic diseases. To determine whether NF-κB drives aging in response to the accumulation of spontaneous, endogenous DNA damage, we measured the activation of NF-κB in WT and progeroid model mice. As both WT and progeroid mice aged, NF-κB was activated stochastically in a variety of cell types. Genetic depletion of one allele of the p65 subunit of NF-κB or treatment with a pharmacological inhibitor of the NF-κB-activating kinase, IKK, delayed the age-related symptoms and pathologies of progeroid mice. Additionally, inhibition of NF-κB reduced oxidative DNA damage and stress and delayed cellular senescence. These results indicate that the mechanism by which DNA damage drives aging is due in part to NF-κB activation. IKK/NF-κB inhibitors are sufficient to attenuate this damage and could provide clinical benefit for degenerative changes associated with accelerated aging disorders and normal aging.


Subject(s)
Aging/drug effects , Cellular Senescence , DNA Damage , I-kappa B Kinase/antagonists & inhibitors , Transcription Factor RelA/metabolism , Aging/genetics , Animals , Cell Nucleus/metabolism , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p16/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Endonucleases/genetics , Endonucleases/metabolism , Gene Expression Regulation/drug effects , Hepatocytes/drug effects , Hepatocytes/metabolism , Hepatocytes/physiology , I-kappa B Kinase/metabolism , Mice , Mice, Transgenic , Oxidative Stress , Peptides/pharmacology , Phosphorylation , Progeria/drug therapy , Progeria/pathology , Protein Binding , Signal Transduction , Transcription Factor RelA/genetics , Transcriptional Activation
6.
Aging Cell ; 11(4): 714-6, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22530741

ABSTRACT

Accumulation of DNA damage is implicated in aging. This is supported by the fact that inherited defects in DNA repair can cause accelerated aging of tissues. However, clear-cut evidence for DNA damage accumulation in old age is lacking. Numerous studies report measurement of DNA damage in nuclear and mitochondrial DNA from tissues of young and old organisms, with variable outcomes. Variability results from genetic differences between specimens or the instability of some DNA lesions. To control these variables and test the hypothesis that elderly organisms have more oxidative DNA damage than young organisms, we measured 8,5'-cyclopurine-2'-deoxynucleosides (cPu), which are relatively stable, in tissues of young and old wild-type and congenic progeroid mice. We found that cPu accumulate spontaneously in the nuclear DNA of wild-type mice with age and to a greater extent in DNA repair-deficient progeroid mice, with a similar tissue-specific pattern (liver > kidney > brain). These data, generated under conditions where genetic and environmental variables are controlled, provide strong evidence that DNA repair mechanisms are inadequate to clear endogenous lesions over the lifespan of mammals. The similar, although exaggerated, results obtained from progeroid, DNA repair-deficient mice and old normal mice support the conclusion that DNA damage accumulates with, and likely contributes to, aging.


Subject(s)
Aging/metabolism , DNA Damage , Aging/genetics , Aging, Premature/genetics , Aging, Premature/metabolism , Animals , Brain/metabolism , DNA Repair , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Endonucleases/deficiency , Endonucleases/genetics , Female , Kidney/metabolism , Liver/metabolism , Male , Mice , Mice, Congenic , Mice, Knockout , Purines/metabolism
7.
Spine (Phila Pa 1976) ; 37(21): 1819-25, 2012 Oct 01.
Article in English | MEDLINE | ID: mdl-22343279

ABSTRACT

STUDY DESIGN: NF-κB activity was pharmacologically and genetically blocked in an accelerated aging mouse model to mitigate age-related disc degenerative changes. OBJECTIVE: To study the mediatory role of NF-κB-signaling pathway in age-dependent intervertebral disc degeneration. SUMMARY OF BACKGROUND DATA: Aging is a major contributor to intervertebral disc degeneration (IDD), but the molecular mechanism behind this process is poorly understood. NF-κB is a family of transcription factors that play a central role in mediating cellular response to damage, stress, and inflammation. Growing evidence implicates chronic NF-κB activation as a culprit in many aging-related diseases, but its role in aging-related IDD has not been adequately explored. We studied the effects of NF-κB inhibition on IDD, using a DNA repair-deficient mouse model of accelerated aging (Ercc1 mice) previously been reported to exhibit age-related IDD. METHODS: Systemic inhibition of NF-κB activation was achieved either genetically by deletion of 1 allele of the NF-κB subunit p65 (Ercc1p65 mice) or pharmacologically by chronic intraperitoneal administration of the Nemo Binding Domain (8K-NBD) peptide to block the formation of the upstream activator of NF-κB, IκB Inducible Kinase (IKK), in Ercc1 mice. Disc cellularity, total proteoglycan content and proteoglycan synthesis of treated mice, and untreated controls were assessed. RESULTS.: Decreased disc matrix proteoglycan content, a hallmark feature of IDD, and elevated disc NF-κB activity were observed in discs of progeroid Ercc1 mice and naturally aged wild-type mice compared with young wild-type mice. Systemic inhibition of NF-κB by the 8K-NBD peptide in Ercc1 mice increased disc proteoglycan synthesis and ameriolated loss of disc cellularity and matrix proteoglycan. These results were confirmed genetically by using the p65 haploinsufficient Ercc1p65 mice. CONCLUSION: These findings demonstrate that the IKK/NF-κB signaling pathway is a key mediator of age-dependent IDD and represents a therapeutic target for mitigating disc degenerative diseases associated with aging.


Subject(s)
Aging , Intervertebral Disc Degeneration/prevention & control , NF-kappa B/antagonists & inhibitors , Peptides/pharmacology , Amino Acid Sequence , Animals , Awards and Prizes , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Disease Models, Animal , Endonucleases/deficiency , Endonucleases/genetics , Female , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , I-kappa B Kinase/antagonists & inhibitors , I-kappa B Kinase/metabolism , Intervertebral Disc Degeneration/genetics , Intervertebral Disc Degeneration/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Microscopy, Fluorescence , Molecular Sequence Data , NF-kappa B/genetics , NF-kappa B/metabolism , Proteoglycans/metabolism , Transcription Factor RelA/antagonists & inhibitors , Transcription Factor RelA/genetics , Transcription Factor RelA/metabolism
8.
Aging Dis ; 2(6): 449-65, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22396894

ABSTRACT

Stochastic damage to cellular macromolecules and organelles is thought to be a driving force behind aging and associated degenerative changes. However, stress response pathways activated by this damage may also contribute to aging. The IKK/NF-κB signaling pathway has been proposed to be one of the key mediators of aging. It is activated by genotoxic, oxidative, and inflammatory stresses and regulates expression of cytokines, growth factors, and genes that regulate apoptosis, cell cycle progression, cell senescence, and inflammation. Transcriptional activity of NF-κB is increased in a variety of tissues with aging and is associated with numerous age-related degenerative diseases including Alzheimer's, diabetes and osteoporosis. In mouse models, inhibition of NF-κB leads to delayed onset of age-related symptoms and pathologies. In addition, NF-κB activation is linked with many of the known lifespan regulators including insulin/IGF-1, FOXO, SIRT, mTOR, and DNA damage. Thus NF-κB represents a possible therapeutic target for extending mammalian healthspan.

9.
DNA Repair (Amst) ; 9(7): 842-7, 2010 Jul 01.
Article in English | MEDLINE | ID: mdl-20462807

ABSTRACT

The DNA of all organisms is constantly damaged by exogenous and endogenous agents. Base excision repair (BER) is important for the removal of several non-bulky lesions from the DNA, however not much is known about the contributions of other DNA repair pathways to the processing of non-bulky lesions. Here we utilized a luciferase reporter system to assess the contributions of transcription-coupled repair (TCR), BER and nucleotide excision repair (NER) to the repair of two non-bulky lesions, 8-oxoguanine (8OG) and uracil (U), in vivo under non-growth conditions. We demonstrate that both TCR and NER are utilized by Escherichia coli to repair 8OG and U. Additionally, the relative level of recognition of these lesions by BER and NER suggests that TCR can utilize components of either pathway for lesion removal, depending upon their availability. These findings indicate a dynamic flexibility of DNA repair pathways in the removal of non-bulky DNA lesions in prokaryotes, and reveal their respective contributions to the repair of 8OG and U in vivo.


Subject(s)
DNA Repair , Escherichia coli/growth & development , Escherichia coli/genetics , Guanine/analogs & derivatives , Guanine/metabolism , Models, Genetic , Transcription, Genetic , Uracil/metabolism
10.
Proc Natl Acad Sci U S A ; 107(8): 3657-62, 2010 Feb 23.
Article in English | MEDLINE | ID: mdl-20142484

ABSTRACT

DNA damage occurs continuously, and faithful replication and transcription are essential for maintaining cell viability. Cells in nature are not dividing and replicating DNA often; therefore it is important to consider the outcome of RNA polymerase (RNAP) encounters with DNA damage. Base damage in the DNA can affect transcriptional fidelity, leading to production of mutant mRNA and protein in a process termed transcriptional mutagenesis (TM). Abasic (AP) sites and strand breaks are frequently occurring, spontaneous damages that are also base excision repair (BER) intermediates. In vitro studies have demonstrated that these lesions can be bypassed by RNAP; however this has never been assessed in vivo. This study demonstrates that RNAP is capable of bypassing AP sites and strand breaks in Escherichia coli and results in TM through adenine incorporation in nascent mRNA. Elimination of the enzymes that process these lesions further increases TM; however, such mutants can still complete repair by other downstream pathways. These results show that AP sites and strand breaks can result in mutagenic RNAP bypass and have important implications for the biologic endpoints of DNA damage.


Subject(s)
DNA Breaks, Double-Stranded , DNA Breaks, Single-Stranded , DNA Replication , Escherichia coli/genetics , Mutagenesis/genetics , Transcription, Genetic , DNA Repair , DNA, Bacterial/genetics , DNA-Directed RNA Polymerases/genetics , DNA-Directed RNA Polymerases/metabolism , Endonucleases/genetics , Endonucleases/metabolism , Mutation/genetics , RNA, Messenger/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...