Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
FASEB J ; 35(5): e21590, 2021 05.
Article in English | MEDLINE | ID: mdl-33871093

ABSTRACT

Light is the key regulator of circadian clock, the time-keeping system synchronizing organism physiology and behavior with environmental day and night conditions. In its natural habitat, the strictly subterranean naked mole-rat (Heterocephalus glaber) has lived in a light-free environment for millennia. We questioned if this species retains a circadian clock and if the patterns of this clock and concomitant rhythms differed in liver tissue from mice and naked mole-rats. As expected, in mice, the various circadian clock genes peaked at different times of the day; the Period gene (Per) group peaked in the evening, whereas Brain and Muscle ARNT-like1 (Bmal1) gene peaked in the morning; this phase shift is considered to be fundamental for circadian clock function. In sharp contrast, in the naked mole-rat both Per1 and Per2, as well as Bmal1, peaked at the same time in the morning-around ZT2-suggesting the organization of the molecular circadian oscillator was different. Moreover, gene expression rhythms associated with glucose metabolism and mTOR signaling also differed between the species. Although the activity of mTORC1 was lower, while that of mTORC2 was higher in naked mole-rat livers compared to mice, unlike that of mice where the expression profiles of glucose metabolism genes were not synchronized, these were highly synchronized in naked mole-rats and likely linked to their use of feeding times at zeitgebers.


Subject(s)
CLOCK Proteins/metabolism , Circadian Clocks , Circadian Rhythm , Gene Expression Regulation , Glucose/metabolism , Liver/metabolism , TOR Serine-Threonine Kinases/metabolism , Animals , CLOCK Proteins/genetics , Female , Gene Expression Profiling , Mice , Mice, Inbred C57BL , Mole Rats , TOR Serine-Threonine Kinases/genetics
2.
Aging (Albany NY) ; 12(12): 11185-11199, 2020 06 23.
Article in English | MEDLINE | ID: mdl-32575074

ABSTRACT

Humanin is a member of a new family of peptides that are encoded by short open reading frames within the mitochondrial genome. It is conserved in animals and is both neuroprotective and cytoprotective. Here we report that in C. elegans the overexpression of humanin is sufficient to increase lifespan, dependent on daf-16/Foxo. Humanin transgenic mice have many phenotypes that overlap with the worm phenotypes and, similar to exogenous humanin treatment, have increased protection against toxic insults. Treating middle-aged mice twice weekly with the potent humanin analogue HNG, humanin improves metabolic healthspan parameters and reduces inflammatory markers. In multiple species, humanin levels generally decline with age, but here we show that levels are surprisingly stable in the naked mole-rat, a model of negligible senescence. Furthermore, in children of centenarians, who are more likely to become centenarians themselves, circulating humanin levels are much greater than age-matched control subjects. Further linking humanin to healthspan, we observe that humanin levels are decreased in human diseases such as Alzheimer's disease and MELAS (Mitochondrial Encephalopathy, Lactic Acidosis, and Stroke-like episodes). Together, these studies are the first to demonstrate that humanin is linked to improved healthspan and increased lifespan.


Subject(s)
Alzheimer Disease/blood , Intracellular Signaling Peptides and Proteins/metabolism , Longevity/physiology , MELAS Syndrome/blood , Mitochondria/metabolism , Adult , Aged, 80 and over , Alzheimer Disease/metabolism , Animals , Animals, Genetically Modified , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/metabolism , Case-Control Studies , Child , Cohort Studies , DNA, Mitochondrial/genetics , Female , Forkhead Transcription Factors/metabolism , Gene Dosage , Humans , Infant, Newborn , Intracellular Signaling Peptides and Proteins/blood , Intracellular Signaling Peptides and Proteins/genetics , MELAS Syndrome/metabolism , Macaca mulatta , Mice , Middle Aged , Models, Animal , Mole Rats , Pregnancy , Young Adult
3.
Geroscience ; 42(4): 1147-1155, 2020 08.
Article in English | MEDLINE | ID: mdl-32394346

ABSTRACT

The goal of the current study was to determine the role of maternal diet in the perinatal period on the health and survival of the offspring. AKR/J mice, a model described to be susceptible to leukemia development, was used where females were maintained on either standard diet (SD), high sucrose diet, Western diet, or calorie restriction (CR) as they were mated with SD-fed males. Body weights, pregnancy rates, litter size, and litter survival were used as markers of successful pregnancy and pup health. Data indicated that maternal diet had significant effects on litter size, early pup survival, and early pup body weights. As pups matured, the makeup of their respective maternal diet was a predictor of adult metabolic health and survival. Overall, these results suggest that perinatal maternal diet is an important determinant of the health and survival of the offspring and that these effects continue well into adulthood, strongly correlating with lifespan.


Subject(s)
Diet , Leukemia , Animals , Body Weight , Female , Male , Mice , Pregnancy , Reproduction
4.
Sci Rep ; 10(1): 6966, 2020 04 24.
Article in English | MEDLINE | ID: mdl-32332849

ABSTRACT

The naked mole-rat is a subterranean rodent, approximately the size of a mouse, renowned for its exceptional longevity (>30 years) and remarkable resistance to cancer. To explore putative mechanisms underlying the cancer resistance of the naked mole-rat, we investigated the regulation and function of the most commonly mutated tumor suppressor, TP53, in the naked mole-rat. We found that the p53 protein in naked mole-rat embryonic fibroblasts (NEFs) exhibits a half-life more than ten times in excess of the protein's characterized half-life in mouse and human embryonic fibroblasts. We determined that the long half-life of the naked mole-rat p53 protein reflects protein-extrinsic regulation. Relative to mouse and human p53, a larger proportion of naked mole-rat p53 protein is constitutively localized in the nucleus prior to DNA damage. Nevertheless, DNA damage is sufficient to induce activation of canonical p53 target genes in NEFs. Despite the uniquely long half-life and unprecedented basal nuclear localization of p53 in NEFs, naked mole-rat p53 retains its canonical tumor suppressive activity. Together, these findings suggest that the unique stabilization and regulation of the p53 protein may contribute to the naked mole-rat's remarkable resistance to cancer.


Subject(s)
Cell Nucleus/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Cell Cycle/physiology , Cell Line , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , DNA Damage/physiology , Female , Fluorescent Antibody Technique , Humans , Male , Mice , Mole Rats , Protein Stability
5.
Physiology (Bethesda) ; 35(2): 96-111, 2020 03 01.
Article in English | MEDLINE | ID: mdl-32024425

ABSTRACT

Pedomorphy, maintenance of juvenile traits throughout life, is most pronounced in extraordinarily long-lived naked mole-rats. Many of these traits (e.g., slow growth rates, low hormone levels, and delayed sexual maturity) are shared with spontaneously mutated, long-lived dwarf mice. Although some youthful traits likely evolved as adaptations to subterranean habitats (e.g., thermolability), the nature of these intrinsic pedomorphic features may also contribute to their prolonged youthfulness, longevity, and healthspan.


Subject(s)
Adaptation, Physiological , Aging , Dwarfism/physiopathology , Longevity , Oxidative Stress , Animals , Humans , Mice , Mole Rats , Species Specificity
6.
Geroscience ; 40(3): 357-358, 2018 06.
Article in English | MEDLINE | ID: mdl-29855760

ABSTRACT

The original version of this article unfortunately contained an error.

7.
Geroscience ; 40(2): 105-121, 2018 04.
Article in English | MEDLINE | ID: mdl-29679203

ABSTRACT

Mouse-sized naked mole-rats (Heterocephalus glaber), unlike other mammals, do not conform to Gompertzian laws of age-related mortality; adults show no age-related change in mortality risk. Moreover, we observe negligible hallmarks of aging with well-maintained physiological and molecular functions, commonly altered with age in other species. We questioned whether naked mole-rats, living an order of magnitude longer than laboratory mice, exhibit different plasma metabolite profiles, which could then highlight novel mechanisms or targets involved in disease and longevity. Using a comprehensive, unbiased metabolomics screen, we observe striking inter-species differences in amino acid, peptide, and lipid metabolites. Low circulating levels of specific amino acids, particularly those linked to the methionine pathway, resemble those observed during the fasting period at late torpor in hibernating ground squirrels and those seen in longer-lived methionine-restricted rats. These data also concur with metabolome reports on long-lived mutant mice, including the Ames dwarf mice and calorically restricted mice, as well as fruit flies, and even show similarities to circulating metabolite differences observed in young human adults when compared to older humans. During evolution, some of these beneficial nutrient/stress response pathways may have been positively selected in the naked mole-rat. These observations suggest that interventions that modify the aging metabolomic profile to a more youthful one may enable people to lead healthier and longer lives.


Subject(s)
Aging/metabolism , Longevity/physiology , Metabolomics/methods , Oxidative Stress/physiology , Animals , Male , Mammals , Mice , Mice, Inbred C57BL , Models, Animal , Mole Rats , Rats , Species Specificity
8.
Mamm Genome ; 27(7-8): 259-78, 2016 08.
Article in English | MEDLINE | ID: mdl-27364349

ABSTRACT

Animals have evolved to survive, and even thrive, in different environments. Genetic adaptations may have indirectly created phenotypes that also resulted in a longer lifespan. One example of this phenomenon is the preternaturally long-lived naked mole-rat. This strictly subterranean rodent tolerates hypoxia, hypercapnia, and soil-based toxins. Naked mole-rats also exhibit pronounced resistance to cancer and an attenuated decline of many physiological characteristics that often decline as mammals age. Elucidating mechanisms that give rise to their unique phenotypes will lead to better understanding of subterranean ecophysiology and biology of aging. Comparative genomics could be a useful tool in this regard. Since the publication of a naked mole-rat genome assembly in 2011, analyses of genomic and transcriptomic data have enabled a clearer understanding of mole-rat evolutionary history and suggested molecular pathways (e.g., NRF2-signaling activation and DNA damage repair mechanisms) that may explain the extraordinarily longevity and unique health traits of this species. However, careful scrutiny and re-analysis suggest that some identified features result from incorrect or imprecise annotation and assembly of the naked mole-rat genome: in addition, some of these conclusions (e.g., genes involved in cancer resistance and hairlessness) are rejected when the analysis includes additional, more closely related species. We describe how the combination of better study design, improved genomic sequencing techniques, and new bioinformatic and data analytical tools will improve comparative genomics and ultimately bridge the gap between traditional model and nonmodel organisms.


Subject(s)
Aging/genetics , Genome , Genomics , Longevity/genetics , Animals , Mammals/genetics , Mole Rats , Molecular Sequence Annotation , Rats , Species Specificity , Transcriptome/genetics
9.
Ann N Y Acad Sci ; 1363: 155-70, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26995762

ABSTRACT

Methionine restriction is a widely reported intervention for increasing life span in several model organisms. Low circulating levels of methionine are evident in the long-lived naked mole-rat, suggesting that it naturally presents with a life-extending phenotype akin to that observed in methionine-restricted animals. Similarly, long-lived dwarf mice also appear to have altered methionine metabolism. The mechanisms underlying methionine-restriction effects on life-span extension, however, remain unknown, as do their potential connections with caloric restriction, another well-established intervention for prolonging life span. Paradoxically, methionine is enriched in proteins expressed in mitochondria and may itself serve an important role in the detoxification of reactive oxygen species and may thereby contribute to delayed aging. Collectively, we highlight the evidence that modulation of the methionine metabolic network can extend life span-from yeast to humans-and explore the evidence that sulfur amino acids and the concomitant transsulfuration pathway play a privileged role in this regard. However, systematic studies in single organisms (particularly those that exhibit extreme longevity) are still required to distinguish the fundamental principles concerning the role of methionine and other amino acids in regulating life span.


Subject(s)
Caloric Restriction , Life Expectancy , Longevity , Methionine/metabolism , Yeasts , Aging , Animals , Cysteine/metabolism , Eukaryotic Cells/physiology , Humans , Invertebrates , Metabolic Networks and Pathways , Models, Animal , Rodentia , Yeasts/physiology
10.
Stat Appl Genet Mol Biol ; 14(6): 507-16, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26595407

ABSTRACT

There is an increasing demand for exploration of the transcriptomes of multiple species with extraordinary traits such as the naked-mole rat (NMR). The NMR is remarkable because of its longevity and resistance to developing cancer. It is of scientific interest to understand the molecular mechanisms that impart these traits, and RNA-sequencing experiments with comparator species can correlate transcriptome dynamics with these phenotypes. Comparing transcriptome differences requires a homology mapping of each transcript in one species to transcript(s) within the other. Such mappings are necessary, especially if one species does not have well-annotated genome available. Current approaches for this type of analysis typically identify the best match for each transcript, but the best match analysis ignores the inherent risks of mismatch when there are multiple candidate transcripts with similar homology scores. We present a method that treats the set of homologs from a novel species as a cluster corresponding to a single gene in the reference species, and we compare the cluster-based approach to a conventional best-match analysis in both simulated data and a case study with NMR and mouse tissues. We demonstrate that the cluster-based approach has superior power to detect differential expression.


Subject(s)
Gene Expression Profiling , RNA, Messenger/genetics , Animals , Cluster Analysis , Computer Simulation , Mice , Models, Genetic , Mole Rats , Phenotype , RNA, Messenger/metabolism , Sequence Analysis, RNA , Sequence Homology, Nucleic Acid , Species Specificity , Transcriptome
11.
Cell Metab ; 22(2): 332-43, 2015 Aug 04.
Article in English | MEDLINE | ID: mdl-26244935

ABSTRACT

Biological diversity among mammals is remarkable. Mammalian body weights range seven orders of magnitude and lifespans differ more than 100-fold among species. While genetic, dietary, and pharmacological interventions can be used to modulate these traits in model organisms, it is unknown how they are determined by natural selection. By profiling metabolites in brain, heart, kidney, and liver tissues of 26 mammalian species representing ten taxonomical orders, we report metabolite patterns characteristic of organs, lineages, and species longevity. Our data suggest different rates of metabolite divergence across organs and reveal patterns representing organ-specific functions and lineage-specific physiologies. We identified metabolites that correlated with species lifespan, some of which were previously implicated in longevity control. We also compared the results with metabolite changes in five long-lived mouse models and observed some similar patterns. Overall, this study describes adjustments of the mammalian metabolome according to lifespan, phylogeny, and organ and lineage specialization.


Subject(s)
Brain/metabolism , Kidney/metabolism , Liver/metabolism , Longevity/physiology , Mammals/metabolism , Metabolome/physiology , Myocardium/metabolism , Animals , Mice , Organ Specificity , Species Specificity
12.
Proc Natl Acad Sci U S A ; 112(12): 3722-7, 2015 Mar 24.
Article in English | MEDLINE | ID: mdl-25775529

ABSTRACT

The preternaturally long-lived naked mole-rat, like other long-lived species and experimental models of extended longevity, is resistant to both endogenous (e.g., reactive oxygen species) and environmental stressors and also resists age-related diseases such as cancer, cardiovascular disease, and neurodegeneration. The mechanisms behind the universal resilience of longer-lived organisms to stress, however, remain elusive. We hypothesize that this resilience is linked to the activity of a highly conserved transcription factor, nuclear factor erythroid 2-related factor (Nrf2). Nrf2 regulates the transcription of several hundred cytoprotective molecules, including antioxidants, detoxicants, and molecular chaperones (heat shock proteins). Nrf2 itself is tightly regulated by mechanisms that either promote its activity or increase its degradation. We used a comparative approach and examined Nrf2-signaling activity in naked mole-rats and nine other rodent species with varying maximum lifespan potential (MLSP). We found that constitutive Nrf2-signaling activity was positively correlated (P = 0.0285) with MLSP and that this activity was also manifested in high levels of downstream gene expression and activity. Surprisingly, we found that species longevity was not linked to the protein levels of Nrf2 itself, but rather showed a significant (P < 0.01) negative relationship with the regulators Kelch-like ECH-Associated Protein 1 (Keap1) and ß-transducin repeat-containing protein (ßTrCP), which target Nrf2 for degradation. These findings highlight the use of a comparative biology approach for the identification of evolved mechanisms that contribute to health span, aging, and longevity.


Subject(s)
Gene Expression Regulation , Longevity , NF-E2-Related Factor 2/physiology , Signal Transduction , Animals , Cricetinae , Female , Gerbillinae , Guinea Pigs , Intracellular Signaling Peptides and Proteins/physiology , Kelch-Like ECH-Associated Protein 1 , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Rats , Reactive Oxygen Species/metabolism , Species Specificity , Xenobiotics , beta-Transducin Repeat-Containing Proteins/physiology
13.
Cell Metab ; 18(4): 533-45, 2013 Oct 01.
Article in English | MEDLINE | ID: mdl-24093677

ABSTRACT

Obesity is associated with a chronic, low-grade, systemic inflammation that may contribute to the development of insulin resistance and type 2 diabetes. Resveratrol, a natural compound with anti-inflammatory properties, is shown to improve glucose tolerance and insulin sensitivity in obese mice and humans. Here, we tested the effect of a 2-year resveratrol administration on proinflammatory profile and insulin resistance caused by a high-fat, high-sugar (HFS) diet in white adipose tissue (WAT) from rhesus monkeys. Resveratrol supplementation (80 and 480 mg/day for the first and second year, respectively) decreased adipocyte size, increased sirtuin 1 expression, decreased NF-κB activation, and improved insulin sensitivity in visceral, but not subcutaneous, WAT from HFS-fed animals. These effects were reproduced in 3T3-L1 adipocytes cultured in media supplemented with serum from monkeys fed HFS ± resveratrol diets. In conclusion, chronic administration of resveratrol exerts beneficial metabolic and inflammatory adaptations in visceral WAT from diet-induced obese monkeys.


Subject(s)
Adipose Tissue, White/drug effects , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Diet, High-Fat , Signal Transduction/drug effects , Stilbenes/pharmacology , Adipocytes/cytology , Adipocytes/drug effects , Adipocytes/metabolism , Adipose Tissue, White/metabolism , Animals , Carbohydrates , Cell Line , Inflammation/metabolism , Insulin/blood , Insulin/metabolism , Macaca mulatta/metabolism , Male , Mice , NF-kappa B/metabolism , Obesity/etiology , Obesity/metabolism , Resveratrol , Sirtuin 1/metabolism , Transcriptome , Viscera/metabolism , Viscera/pathology
14.
Antioxid Redox Signal ; 19(12): 1388-99, 2013 Oct 20.
Article in English | MEDLINE | ID: mdl-23025341

ABSTRACT

SIGNIFICANCE: The oxidative stress theory of aging has been the most widely accepted theory of aging providing insights into why we age and die for over 50 years, despite mounting evidence from a multitude of species indicating that there is no direct relationship between reactive oxygen species (ROS) and longevity. Here we explore how different species, including the longest lived rodent, the naked mole-rat, have defied the most predominant aging theory. RECENT ADVANCES: In the case of extremely long-lived naked mole-rat, levels of ROS production are found to be similar to mice, antioxidant defenses unexceptional, and even under constitutive conditions, naked mole-rats combine a pro-oxidant intracellular milieu with high, steady state levels of oxidative damage. Clearly, naked mole-rats can tolerate this level of oxidative stress and must have mechanisms in place to prevent its translation into potentially lethal diseases. CRITICAL ISSUES: In addition to the naked mole-rat, other species from across the phylogenetic spectrum and even certain mouse strains do not support this theory. Moreover, overexpressing or knocking down antioxidant levels alters levels of oxidative damage and even cancer incidence, but does not modulate lifespan. FUTURE DIRECTIONS: Perhaps, it is not oxidative stress that modulates healthspan and longevity, but other cytoprotective mechanisms that allow animals to deal with high levels of oxidative damage and stress, and nevertheless live long, relatively healthy lifespans. Studying these mechanisms in uniquely long-lived species, like the naked mole-rat, may help us tease out the key contributors to aging and longevity.


Subject(s)
Aging , Mole Rats/physiology , Oxidative Stress , Animals , Glutathione/metabolism , Humans , Longevity , Oxidation-Reduction , Oxidoreductases/physiology , Species Specificity
15.
Am J Physiol Endocrinol Metab ; 303(8): E1061-8, 2012 Oct 15.
Article in English | MEDLINE | ID: mdl-22932781

ABSTRACT

Emerging research has shown that subtle factors during pregnancy and gestation can influence long-term health in offspring. In an attempt to be proactive, we set out to explore whether a nonpharmacological intervention, perinatal exercise, might improve offspring health. Female mice were separated into sedentary or exercise cohorts, with the exercise cohort having voluntary access to a running wheel prior to mating and during pregnancy and nursing. Offspring were weaned, and analyses were performed on the mature offspring that did not have access to running wheels during any portion of their lives. Perinatal exercise caused improved glucose disposal following an oral glucose challenge in both female and male adult offspring (P < 0.05 for both). Blood glucose concentrations were reduced to lower values in response to an intraperitoneal insulin tolerance test for both female and male adult offspring of parents with access to running wheels (P < 0.05 and P < 0.01, respectively). Male offspring from exercised dams showed increased percent lean mass and decreased fat mass percent compared with male offspring from sedentary dams (P < 0.01 for both), but these parameters were unchanged in female offspring. These data suggest that short-term maternal voluntary exercise prior to and during healthy pregnancy and nursing can enhance long-term glucose homeostasis in offspring.


Subject(s)
Glucose/metabolism , Homeostasis/physiology , Physical Conditioning, Animal/physiology , Adipose Tissue/metabolism , Animals , Birth Weight/physiology , Blood Glucose/metabolism , Body Composition/physiology , Body Weight/physiology , Deoxyglucose/metabolism , Eating/physiology , Female , Glucose Tolerance Test , Insulin/metabolism , Lactation/physiology , Litter Size/physiology , Mice , Mice, Inbred ICR , Muscle, Skeletal/metabolism , Pregnancy , Running/physiology
16.
Gerontology ; 58(5): 453-62, 2012.
Article in English | MEDLINE | ID: mdl-22572398

ABSTRACT

BACKGROUND: Studies comparing similar-sized species with disparate longevity may elucidate novel mechanisms that abrogate aging and prolong good health. We focus on the longest living rodent, the naked mole-rat. This mouse-sized mammal lives ~8 times longer than do mice and, despite high levels of oxidative damage evident at a young age, it is not only very resistant to spontaneous neoplasia but also shows minimal decline in age-associated physiological traits. OBJECTIVES: We assess the current status of stress resistance and longevity, focusing in particular on the molecular and cellular responses to cytotoxins and other stressors between the short-lived laboratory mouse and the naked mole-rat. RESULTS: Like other experimental animal models of lifespan extension, naked mole-rat fibroblasts are extremely tolerant of a broad spectrum of cytotoxins including heat, heavy metals, DNA-damaging agents and xenobiotics, showing LD(50) values between 2- and 20-fold greater than those of fibroblasts of shorter-lived mice. Our new data reveal that naked mole-rat fibroblasts stop proliferating even at low doses of toxin whereas those mouse fibroblasts that survive treatment rapidly re-enter the cell cycle and may proliferate with DNA damage. Naked mole-rat fibroblasts also show significantly higher constitutive levels of both p53 and Nrf2 protein levels and activity, and this increases even further in response to toxins. CONCLUSION: Enhanced cell signaling via p53 and Nrf2 protects cells against proliferating with damage, augments clearance of damaged proteins and organelles and facilitates the maintenance of both genomic and protein integrity. These pathways collectively regulate a myriad of mechanisms which may contribute to the attenuated aging profile and sustained healthspan of the naked mole-rat. Understanding how these are regulated may be also integral to sustaining positive human healthspan well into old age and may elucidate novel therapeutics for delaying the onset and progression of physiological declines that characterize the aging process.


Subject(s)
Aging/physiology , Mole Rats/physiology , Animals , Cell Survival/drug effects , Female , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Humans , Longevity/physiology , Metals, Heavy/toxicity , Mice , Models, Animal , NF-E2-Related Factor 2/metabolism , Oxidative Stress , Species Specificity , Stress, Physiological , Tumor Suppressor Protein p53/metabolism
17.
Curr Pharm Des ; 17(22): 2290-307, 2011.
Article in English | MEDLINE | ID: mdl-21736541

ABSTRACT

Reactive oxygen species (ROS), by-products of aerobic metabolism, cause oxidative damage to cells and tissue and not surprisingly many theories have arisen to link ROS-induced oxidative stress to aging and health. While studies clearly link ROS to a plethora of divergent diseases, their role in aging is still debatable. Genetic knock-down manipulations of antioxidants alter the levels of accrued oxidative damage, however, the resultant effect of increased oxidative stress on lifespan are equivocal. Similarly the impact of elevating antioxidant levels through transgenic manipulations yield inconsistent effects on longevity. Furthermore, comparative data from a wide range of endotherms with disparate longevity remain inconclusive. Many long-living species such as birds, bats and mole-rats exhibit high-levels of oxidative damage, evident already at young ages. Clearly, neither the amount of ROS per se nor the sensitivity in neutralizing ROS are as important as whether or not the accrued oxidative stress leads to oxidative-damage-linked age-associated diseases. In this review we examine the literature on ROS, its relation to disease and the lessons gleaned from a comparative approach based upon species with widely divergent responses. We specifically focus on the longest lived rodent, the naked mole-rat, which maintains good health and provides novel insights into the paradox of maintaining both an extended healthspan and lifespan despite high oxidative stress from a young age.


Subject(s)
Longevity/physiology , Oxidative Stress/physiology , Reactive Oxygen Species/metabolism , Animals , Antioxidants/metabolism , Autophagy/genetics , Autophagy/physiology , Enzymes/genetics , Humans , Longevity/genetics , Mitochondria/genetics , Mitochondria/physiology , Models, Genetic , Mole Rats , Oxidative Stress/genetics , Oxygen Consumption/genetics , Oxygen Consumption/physiology , Polymorphism, Genetic , Rats , Species Specificity
18.
Integr Comp Biol ; 50(5): 829-43, 2010 Nov.
Article in English | MEDLINE | ID: mdl-21031035

ABSTRACT

Although aging is a ubiquitous process that prevails in all organisms, the mechanisms governing both the rate of decline in functionality and the age of onset remain elusive. A profound constitutively upregulated cytoprotective response is commonly observed in naturally long-lived species and experimental models of extensions to lifespan (e.g., genetically-altered and/or experimentally manipulated organisms), as indicated by enhanced resistance to stress and upregulated downstream components of the cytoprotective nuclear factor erythroid 2-related factor 2 (Nrf2)-signaling pathway. The transcription factor Nrf2 is constitutively expressed in all tissues, although levels may vary among organs, with the key detoxification organs (kidney and liver) exhibiting highest levels. Nrf2 may be further induced by cellular stressors including endogenous reactive-oxygen species or exogenous electrophiles. The Nrf2-signaling pathway mediates multiple avenues of cytoprotection by activating the transcription of more than 200 genes that are crucial in the metabolism of drugs and toxins, protection against oxidative stress and inflammation, as well as playing an integral role in stability of proteins and in the removal of damaged proteins via proteasomal degradation or autophagy. Nrf2 interacts with other important cell regulators such as tumor suppressor protein 53 (p53) and nuclear factor-kappa beta (NF-κB) and through their combined interactions is the guardian of healthspan, protecting against many age-related diseases including cancer and neurodegeneration. We hypothesize that this signaling pathway plays a critical role in the determination of species longevity and that this pathway may indeed be the master regulator of the aging process.


Subject(s)
Longevity/physiology , NF-E2-Related Factor 2/physiology , Signal Transduction/physiology , Aging/physiology , Animals , Cell Survival/physiology , Cytoprotection/physiology
19.
J Neurosci ; 30(29): 9695-707, 2010 Jul 21.
Article in English | MEDLINE | ID: mdl-20660252

ABSTRACT

Conservation of normal cognitive functions relies on the proper performance of the nervous system at the cellular and molecular level. The mammalian nicotinamide-adenine dinucleotide-dependent deacetylase SIRT1 impacts different processes potentially involved in the maintenance of brain integrity, such as chromatin remodeling, DNA repair, cell survival, and neurogenesis. Here we show that SIRT1 is expressed in neurons of the hippocampus, a key structure in learning and memory. Using a combination of behavioral and electrophysiological paradigms, we analyzed the effects of SIRT1 deficiency and overexpression on mouse learning and memory as well as on synaptic plasticity. We demonstrated that the absence of SIRT1 impaired cognitive abilities, including immediate memory, classical conditioning, and spatial learning. In addition, we found that the cognitive deficits in SIRT1 knock-out (KO) mice were associated with defects in synaptic plasticity without alterations in basal synaptic transmission or NMDA receptor function. Brains of SIRT1-KO mice exhibited normal morphology and dendritic spine structure but displayed a decrease in dendritic branching, branch length, and complexity of neuronal dendritic arbors. Also, a decrease in extracellular signal-regulated kinase 1/2 phosphorylation and altered expression of hippocampal genes involved in synaptic function, lipid metabolism, and myelination were detected in SIRT1-KO mice. In contrast, mice with high levels of SIRT1 expression in brain exhibited regular synaptic plasticity and memory. We conclude that SIRT1 is indispensable for normal learning, memory, and synaptic plasticity in mice.


Subject(s)
Cognition/physiology , Hippocampus/physiology , Learning/physiology , Long-Term Potentiation/genetics , Memory/physiology , Neurons/metabolism , Sirtuin 1/genetics , Animals , Dendritic Spines/ultrastructure , Gene Expression Regulation , Hippocampus/cytology , Mice , Mice, Knockout , Neurons/chemistry , Patch-Clamp Techniques , Sirtuin 1/analysis , Tissue Distribution
20.
Cell Metab ; 8(2): 157-68, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18599363

ABSTRACT

A small molecule that safely mimics the ability of dietary restriction (DR) to delay age-related diseases in laboratory animals is greatly sought after. We and others have shown that resveratrol mimics effects of DR in lower organisms. In mice, we find that resveratrol induces gene expression patterns in multiple tissues that parallel those induced by DR and every-other-day feeding. Moreover, resveratrol-fed elderly mice show a marked reduction in signs of aging, including reduced albuminuria, decreased inflammation, and apoptosis in the vascular endothelium, increased aortic elasticity, greater motor coordination, reduced cataract formation, and preserved bone mineral density. However, mice fed a standard diet did not live longer when treated with resveratrol beginning at 12 months of age. Our findings indicate that resveratrol treatment has a range of beneficial effects in mice but does not increase the longevity of ad libitum-fed animals when started midlife.


Subject(s)
Aging/drug effects , Caloric Restriction , Energy Intake/genetics , Longevity/drug effects , Stilbenes/pharmacology , Transcription, Genetic/drug effects , Age Factors , Aging/genetics , Aging/metabolism , Animals , Antioxidants/pharmacology , Antioxidants/therapeutic use , Apoptosis/drug effects , Apoptosis/physiology , Cardiovascular System/drug effects , Cardiovascular System/physiopathology , Food Deprivation/physiology , Food, Formulated , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Inflammation/drug therapy , Inflammation/prevention & control , Longevity/genetics , Male , Mice , Mice, Inbred C57BL , Osteoporosis/drug therapy , Osteoporosis/prevention & control , Resveratrol , Stilbenes/therapeutic use , Transcription, Genetic/genetics , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...