Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
JCI Insight ; 8(15)2023 08 08.
Article in English | MEDLINE | ID: mdl-37551712

ABSTRACT

Age-associated sarcopenia, characterized by a progressive loss in muscle mass and strength, is the largest cause of frailty and disability in the elderly worldwide. Current treatments involve nonpharmacological guidelines that few subjects can abide by, highlighting the need for effective drugs. Preclinical models were employed to test the benefits of RJx-01, a combination drug composed of metformin and galantamine, on sarcopenia. In worms, RJx-01 treatment improved lifespan, locomotion, pharyngeal pumping, and muscle fiber organization. The synergistic effects of RJx-01 were recapitulated in a transgenic mouse model that displays an exacerbated aging phenotype (Opa1-/-). In these mice, RJx-01 ameliorated physical performance, muscle mass and force, neuromuscular junction stability, and systemic inflammation. RJx-01 also improved physical performance and muscle strength in 22-month-old WT mice and also improved skeletal muscle ultrastructure, mitochondrial morphology, autophagy, lysosomal function, and satellite cell content. Denervation and myofiber damage were decreased in RJx-01-treated animals compared with controls. RJx-01 improved muscle quality rather than quantity, indicating that the improvement in quality underlies the beneficial effects of the combination drug. The studies herein indicate synergistic beneficial effects of RJx-01 in the treatment of sarcopenia and support the pursuit of RJx-01 in a human clinical trial as a therapeutic intervention for sarcopenia.


Subject(s)
Metformin , Sarcopenia , Humans , Mice , Animals , Aged , Infant , Sarcopenia/drug therapy , Galantamine/pharmacology , Metformin/pharmacology , Aging/physiology , Muscle, Skeletal/pathology , Mice, Transgenic
3.
Free Radic Biol Med ; 152: 650-658, 2020 05 20.
Article in English | MEDLINE | ID: mdl-31953150

ABSTRACT

Caloric restriction (CR) is the leading non-pharmaceutical dietary intervention to improve health- and lifespan in most model organisms. A wide array of cellular pathways is induced in response to CR and CR-mimetics, including the transcriptional activator Nuclear factor erythroid-2-related factor 2 (Nrf2), which is essential in the upregulation of multiple stress-responsive and mitochondrial enzymes. Nrf2 is necessary in tumor protection but is not essential for the lifespan extending properties of CR in outbred mice. Here, we sought to study Nrf2-knockout (KO) mice and littermate controls in male C57BL6/J, an inbred mouse strain. Deletion of Nrf2 resulted in shortened lifespan compared to littermate controls only under ad libitum conditions. CR-mediated lifespan extension and physical performance improvements did not require Nrf2. Metabolic and protein homeostasis and activation of tissue-specific cytoprotective proteins were dependent on Nrf2 expression. These results highlight an important contribution of Nrf2 for normal lifespan and stress response.


Subject(s)
Caloric Restriction , Longevity , Animals , Longevity/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-E2-Related Factor 2/genetics
4.
J Gerontol A Biol Sci Med Sci ; 74(2): 155-162, 2019 01 16.
Article in English | MEDLINE | ID: mdl-29733330

ABSTRACT

Caloric restriction (CR) is the most potent nonpharmacological intervention known to both protect against carcinogenesis and delay aging in laboratory animals. There is a growing number of anticarcinogens and CR mimetics that activate NAD(P)H:quinone oxidoreductase 1 (NQO1). We have previously shown that NQO1, an antioxidant enzyme that acts as an energy sensor through modulation of intracellular redox and metabolic state, is upregulated by CR. Here, we used NQO1-knockout (KO) mice to investigate the role of NQO1 in both the aging process and tumor susceptibility, specifically in the context of CR. We found that NQO1 is not essential for the beneficial effects of CR on glucose homeostasis, physical performance, metabolic flexibility, life-span extension, and (unlike our previously observation with Nrf2) chemical-induced tumorigenesis.


Subject(s)
Body Composition , Caloric Restriction/adverse effects , Longevity , NAD(P)H Dehydrogenase (Quinone)/metabolism , Neoplasms, Experimental/prevention & control , Oxidative Stress , Animals , Biomarkers, Tumor/metabolism , Carcinogenesis , Immunoblotting , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoplasms, Experimental/etiology , Neoplasms, Experimental/metabolism
5.
J Clin Invest ; 128(3): 1087-1105, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29431735

ABSTRACT

The skeleton has emerged as an important regulator of systemic glucose homeostasis, with osteocalcin and insulin representing prime mediators of the interplay between bone and energy metabolism. However, genetic evidence indicates that osteoblasts can influence global energy metabolism through additional, as yet unknown, mechanisms. Here, we report that constitutive or postnatally induced deletion of the hypoxia signaling pathway component von Hippel-Lindau (VHL) in skeletal osteolineage cells of mice led to high bone mass as well as hypoglycemia and increased glucose tolerance, not accounted for by osteocalcin or insulin. In vitro and in vivo data indicated that Vhl-deficient osteoblasts displayed massively increased glucose uptake and glycolysis associated with upregulated HIF-target gene expression, resembling the Warburg effect that typifies cancer cells. Overall, the glucose consumption by the skeleton was increased in the mutant mice, as revealed by 18F-FDG radioactive tracer experiments. Moreover, the glycemia levels correlated inversely with the level of skeletal glucose uptake, and pharmacological treatment with the glycolysis inhibitor dichloroacetate (DCA), which restored glucose metabolism in Vhl-deficient osteogenic cells in vitro, prevented the development of the systemic metabolic phenotype in the mutant mice. Altogether, these findings reveal a novel link between cellular glucose metabolism in osteoblasts and whole-body glucose homeostasis, controlled by local hypoxia signaling in the skeleton.


Subject(s)
Glucose/metabolism , Osteoblasts/metabolism , Von Hippel-Lindau Tumor Suppressor Protein/genetics , Adenocarcinoma/pathology , Animals , Bone Marrow/metabolism , Bone Neoplasms/secondary , Cell Lineage , Female , Glycolysis , Humans , Hypoxia , Lung Neoplasms/pathology , Male , Mice , Mice, Knockout , Mutation , Neoplasm Metastasis/pathology , Osteocalcin/metabolism , Signal Transduction , X-Ray Microtomography
6.
NPJ Aging Mech Dis ; 3: 8, 2017.
Article in English | MEDLINE | ID: mdl-28649426

ABSTRACT

Aging is a complex phenomenon involving functional decline in multiple physiological systems. We undertook a comparative analysis of skeletal muscle from four different species, i.e. mice, rats, rhesus monkeys, and humans, at three different representative stages during their lifespan (young, middle, and old) to identify pathways that modulate function and healthspan. Gene expression profiling and computational analysis revealed that pathway complexity increases from mice to humans, and as mammals age, there is predominantly an upregulation of pathways in all species. Two downregulated pathways, the electron transport chain and oxidative phosphorylation, were common among all four species in response to aging. Quantitative PCR, biochemical analysis, mitochondrial DNA measurements, and electron microscopy revealed a conserved age-dependent decrease in mitochondrial content, and a reduction in oxidative phosphorylation complexes in monkeys and humans. Western blot analysis of key proteins in mitochondrial biogenesis discovered that (i) an imbalance toward mitochondrial fusion occurs in aged skeletal muscle and (ii) mitophagy is not overtly affected, presumably leading to the observed accumulation of abnormally large, damaged mitochondria with age. Select transcript expression analysis uncovered that the skeletal inflammatory profile differentially increases with age, but is most pronounced in humans, while increased oxidative stress (as assessed by protein carbonyl adducts and 4-hydroxynonenal) is common among all species. Expression studies also found that there is unique dysregulation of the nutrient sensing pathways among the different species with age. The identification of conserved pathways indicates common molecular mechanisms intrinsic to health and lifespan, whereas the recognition of species-specific pathways emphasizes the importance of human studies for devising optimal therapeutic modalities to slow the aging process.

7.
Am J Respir Cell Mol Biol ; 56(3): 393-401, 2017 03.
Article in English | MEDLINE | ID: mdl-27875656

ABSTRACT

Lung tissue remodeling in chronic obstructive pulmonary disease (COPD) is characterized by airway wall thickening and/or emphysema. Although the bronchial and alveolar compartments are functionally independent entities, we recently showed comparable alterations in matrix composition comprised of decreased elastin content and increased collagen and hyaluronan contents of alveolar and small airway walls. Out of several animal models tested, surfactant protein C (SPC)-TNF-α mice showed remodeling in alveolar and airway walls similar to what we observed in patients with COPD. Epithelial cells are able to undergo a phenotypic shift, gaining mesenchymal properties, a process in which c-Jun N-terminal kinase (JNK) signaling is involved. Therefore, we hypothesized that TNF-α induces JNK-dependent epithelial plasticity, which contributes to lung matrix remodeling. To this end, the ability of TNF-α to induce a phenotypic shift was assessed in A549, BEAS2B, and primary bronchial epithelial cells, and phenotypic markers were studied in SPC-TNF-α mice. Phenotypic markers of mesenchymal cells were elevated both in vitro and in vivo, as shown by the expression of vimentin, plasminogen activator inhibitor-1, collagen, and matrix metalloproteinases. Concurrently, the expression of the epithelial markers, E-cadherin and keratin 7 and 18, was attenuated. A pharmacological inhibitor of JNK attenuated this phenotypic shift in vitro, demonstrating involvement of JNK signaling in this process. Interestingly, activation of JNK signaling was also clearly present in lungs of SPC-TNF-α mice and patients with COPD. Together, these data show a role for TNF-α in the induction of a phenotypic shift in vitro, resulting in increased collagen production and the expression of elastin-degrading matrix metalloproteinases, and provide evidence for involvement of the TNF-α-JNK axis in extracellular matrix remodeling.


Subject(s)
Extracellular Matrix/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Animals , Biomarkers/metabolism , Cell Hypoxia/drug effects , Cell Line , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Extracellular Matrix/drug effects , Humans , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , Lung/drug effects , Lung/metabolism , Mesoderm/metabolism , Mice , Phenotype , Phosphorylation/drug effects , Pulmonary Disease, Chronic Obstructive/metabolism , Pulmonary Disease, Chronic Obstructive/pathology , Pulmonary Surfactant-Associated Protein C/metabolism , Signal Transduction/drug effects
8.
Am J Physiol Lung Cell Mol Physiol ; 307(7): L557-65, 2014 Oct 01.
Article in English | MEDLINE | ID: mdl-25106431

ABSTRACT

Remodeling in chronic obstructive pulmonary disease (COPD) has at least two dimensions: small airway wall thickening and destruction of alveolar walls. Recently we showed comparable alterations of the extracellular matrix (ECM) compounds collagen, hyaluoran, and elastin in alveolar and small airway walls of COPD patients. The aim of this study was to characterize and assess similarities in alveolar and small airway wall matrix remodeling in chronic COPD models. From this comparative characterization of matrix remodeling we derived and elaborated underlying mechanisms to the matrix changes reported in COPD. Lung tissue sections of chronic models for COPD, either induced by exposure to cigarette smoke, chronic intratracheal lipopolysaccharide instillation, or local tumor necrosis factor (TNF) expression [surfactant protein C (SPC)-TNFα mice], were stained for elastin, collagen, and hyaluronan. Furthermore TNF-α matrix metalloproteinase (MMP)-2, -9, and -12 mRNA expression was analyzed using qPCR and localized using immunohistochemistry. Both collagen and hyaluronan were increased in alveolar and small airway walls of all three models. Interestingly, elastin contents were differentially affected, with a decrease in both alveolar and airway walls in SPC-TNFα mice. Furthermore TNF-α and MMP-2 and -9 mRNA and protein levels were found to be increased in alveolar walls and around airway walls only in SPC-TNFα mice. We show that only SPC-TNFα mice show changes in elastin remodeling that are comparable to what has been observed in COPD patients. This reveals that the SPC-TNFα model is a suitable model to study processes underlying matrix remodeling and in particular elastin breakdown as seen in COPD. Furthermore we indicate a possible role for MMP-2 and MMP-9 in the breakdown of elastin in airways and alveoli of SPC-TNFα mice.


Subject(s)
Extracellular Matrix/metabolism , Pulmonary Disease, Chronic Obstructive/metabolism , Tumor Necrosis Factor-alpha/physiology , Airway Remodeling/immunology , Animals , Disease Models, Animal , Elastin/metabolism , Extracellular Matrix/pathology , Fibrillar Collagens/metabolism , Gene Expression , Hyaluronic Acid/metabolism , Lipopolysaccharides/pharmacology , Male , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Inbred C57BL , Pulmonary Alveoli/immunology , Pulmonary Alveoli/metabolism , Pulmonary Alveoli/pathology , Pulmonary Disease, Chronic Obstructive/etiology , Pulmonary Disease, Chronic Obstructive/immunology , Smoking/adverse effects
9.
Mol Cell Biol ; 34(16): 3106-19, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24891619

ABSTRACT

The mammalian RNA-binding protein AUF1 (AU-binding factor 1, also known as heterogeneous nuclear ribonucleoprotein D [hnRNP D]) binds to numerous mRNAs and influences their posttranscriptional fate. Given that many AUF1 target mRNAs encode muscle-specific factors, we investigated the function of AUF1 in skeletal muscle differentiation. In mouse C2C12 myocytes, where AUF1 levels rise at the onset of myogenesis and remain elevated throughout myocyte differentiation into myotubes, RNP immunoprecipitation (RIP) analysis indicated that AUF1 binds prominently to Mef2c (myocyte enhancer factor 2c) mRNA, which encodes the key myogenic transcription factor MEF2C. By performing mRNA half-life measurements and polysome distribution analysis, we found that AUF1 associated with the 3' untranslated region (UTR) of Mef2c mRNA and promoted MEF2C translation without affecting Mef2c mRNA stability. In addition, AUF1 promoted Mef2c gene transcription via a lesser-known role of AUF1 in transcriptional regulation. Importantly, lowering AUF1 delayed myogenesis, while ectopically restoring MEF2C expression levels partially rescued the impairment of myogenesis seen after reducing AUF1 levels. We propose that MEF2C is a key effector of the myogenesis program promoted by AUF1.


Subject(s)
Heterogeneous-Nuclear Ribonucleoprotein D/physiology , Muscle Development/genetics , Muscle, Skeletal/embryology , RNA-Binding Proteins/physiology , 3' Untranslated Regions/genetics , Animals , Cell Differentiation/genetics , Cell Line , Gene Expression Regulation, Developmental , Heterogeneous Nuclear Ribonucleoprotein D0 , Heterogeneous-Nuclear Ribonucleoprotein D/genetics , MEF2 Transcription Factors/biosynthesis , MEF2 Transcription Factors/genetics , Mice , Mice, Inbred C57BL , Muscle Fibers, Skeletal/metabolism , Protein Binding/genetics , Protein Biosynthesis/genetics , RNA Interference , RNA, Messenger/genetics , RNA, Small Interfering , RNA-Binding Proteins/genetics , Regeneration/genetics , Transcription, Genetic/genetics , Transcriptional Activation/genetics
10.
Aging Cell ; 13(5): 787-96, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24931715

ABSTRACT

Increased expression of SIRT1 extends the lifespan of lower organisms and delays the onset of age-related diseases in mammals. Here, we show that SRT2104, a synthetic small molecule activator of SIRT1, extends both mean and maximal lifespan of mice fed a standard diet. This is accompanied by improvements in health, including enhanced motor coordination, performance, bone mineral density, and insulin sensitivity associated with higher mitochondrial content and decreased inflammation. Short-term SRT2104 treatment preserves bone and muscle mass in an experimental model of atrophy. These results demonstrate it is possible to design a small molecule that can slow aging and delay multiple age-related diseases in mammals, supporting the therapeutic potential of SIRT1 activators in humans.


Subject(s)
Bone and Bones/drug effects , Heterocyclic Compounds, 2-Ring/pharmacology , Aging , Animals , Body Composition , Body Mass Index , Bone and Bones/metabolism , Diet , Humans , Kaplan-Meier Estimate , Male , Mice , Mice, Inbred C57BL , Survival Analysis
11.
Cell Rep ; 6(5): 836-43, 2014 Mar 13.
Article in English | MEDLINE | ID: mdl-24582957

ABSTRACT

The prevention or delay of the onset of age-related diseases prolongs survival and improves quality of life while reducing the burden on the health care system. Activation of sirtuin 1 (SIRT1), an NAD(+)-dependent deacetylase, improves metabolism and confers protection against physiological and cognitive disturbances in old age. SRT1720 is a specific SIRT1 activator that has health and lifespan benefits in adult mice fed a high-fat diet. We found extension in lifespan, delayed onset of age-related metabolic diseases, and improved general health in mice fed a standard diet after SRT1720 supplementation. Inhibition of proinflammatory gene expression in both liver and muscle of SRT1720-treated animals was noted. SRT1720 lowered the phosphorylation of NF-κB pathway regulators in vitro only when SIRT1 was functionally present. Combined with our previous work, the current study further supports the beneficial effects of SRT1720 on health across the lifespan in mice.


Subject(s)
Heterocyclic Compounds, 4 or More Rings/metabolism , Sirtuin 1/metabolism , Animals , Diet , Longevity , Male , Mice , Mice, Inbred C57BL , Sirtuin 1/genetics , Survival Analysis , Transcriptome
12.
Aging Cell ; 13(1): 193-6, 2014 Feb.
Article in English | MEDLINE | ID: mdl-23941528

ABSTRACT

The SIRT1 deacetylase is one of the best-studied putative mediators of some of the anti-aging effects of calorie restriction (CR), but its role in CR-dependent lifespan extension has not been demonstrated. We previously found that mice lacking both copies of SIRT1 displayed a shorter median lifespan than wild-type mice on an ad libitum diet. Here, we report that median lifespan extension in CR heterozygote SIRT1(+/-) mice was identical (51%) to that observed in wild-type mice, but SIRT1(+/-) mice displayed a higher frequency of certain pathologies. Although larger studies in additional genetic backgrounds are needed, these results provide strong initial evidence for the requirement of SIRT1 for the lifespan extension effects of CR, but suggest that its high expression is not required for CR-induced lifespan extension.


Subject(s)
Caloric Restriction , Gene Expression Regulation , Longevity , Sirtuin 1/genetics , Sirtuin 1/metabolism , Animals , Longevity/genetics , Mice , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sirtuin 1/biosynthesis
13.
Cell ; 155(7): 1624-38, 2013 Dec 19.
Article in English | MEDLINE | ID: mdl-24360282

ABSTRACT

Ever since eukaryotes subsumed the bacterial ancestor of mitochondria, the nuclear and mitochondrial genomes have had to closely coordinate their activities, as each encode different subunits of the oxidative phosphorylation (OXPHOS) system. Mitochondrial dysfunction is a hallmark of aging, but its causes are debated. We show that, during aging, there is a specific loss of mitochondrial, but not nuclear, encoded OXPHOS subunits. We trace the cause to an alternate PGC-1α/ß-independent pathway of nuclear-mitochondrial communication that is induced by a decline in nuclear NAD(+) and the accumulation of HIF-1α under normoxic conditions, with parallels to Warburg reprogramming. Deleting SIRT1 accelerates this process, whereas raising NAD(+) levels in old mice restores mitochondrial function to that of a young mouse in a SIRT1-dependent manner. Thus, a pseudohypoxic state that disrupts PGC-1α/ß-independent nuclear-mitochondrial communication contributes to the decline in mitochondrial function with age, a process that is apparently reversible.


Subject(s)
Aging/pathology , Cell Nucleus/metabolism , Mitochondria/metabolism , NAD/metabolism , Oxidative Phosphorylation , AMP-Activated Protein Kinases/metabolism , Animals , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mice , Muscle, Skeletal/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Reactive Oxygen Species/metabolism , Sirtuin 1/metabolism , Transcription Factors/metabolism
14.
Aging (Albany NY) ; 5(9): 692-703, 2013 Sep.
Article in English | MEDLINE | ID: mdl-24036467

ABSTRACT

The levels of microRNAs (miRNAs) are altered under different conditions such as cancer, senescence, and aging. Here, we have identified differentially expressed miRNAs in skeletal muscle from young and old rhesus monkeys using RNA sequencing. In old muscle, several miRNAs were upregulated, including miR-451, miR-144, miR-18a and miR-15a, while a few miRNAs were downregulated, including miR-181a and miR-181b. A number of novel miRNAs were also identified, particularly in old muscle. We also examined the impact of caloric restriction (CR) on miRNA abundance by reverse transcription (RT) followed by real-time, quantitative (q)PCR analysis and found that CR rescued the levels of miR-181b and chr1:205580546, and also dampened the age-induced increase in miR-451 and miR-144 levels. Our results reveal that there are changes in expression of known and novel miRNAs with skeletal muscle aging and that CR may reverse some of these changes to a younger phenotype.


Subject(s)
Aging/genetics , Aging/metabolism , Caloric Restriction , Macaca mulatta/genetics , Macaca mulatta/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Muscle, Skeletal/metabolism , Animals , Down-Regulation , Humans , Macaca mulatta/growth & development , Male , Muscle, Skeletal/growth & development , Muscular Diseases/genetics , Muscular Diseases/metabolism , Species Specificity , Up-Regulation
15.
Nat Commun ; 4: 2192, 2013.
Article in English | MEDLINE | ID: mdl-23900241

ABSTRACT

Metformin is a drug commonly prescribed to treat patients with type 2 diabetes. Here we show that long-term treatment with metformin (0.1% w/w in diet) starting at middle age extends healthspan and lifespan in male mice, while a higher dose (1% w/w) was toxic. Treatment with metformin mimics some of the benefits of calorie restriction, such as improved physical performance, increased insulin sensitivity, and reduced low-density lipoprotein and cholesterol levels without a decrease in caloric intake. At a molecular level, metformin increases AMP-activated protein kinase activity and increases antioxidant protection, resulting in reductions in both oxidative damage accumulation and chronic inflammation. Our results indicate that these actions may contribute to the beneficial effects of metformin on healthspan and lifespan. These findings are in agreement with current epidemiological data and raise the possibility of metformin-based interventions to promote healthy aging.


Subject(s)
Health , Longevity/drug effects , Metformin/pharmacology , AMP-Activated Protein Kinases , Animals , Antioxidants/pharmacology , Biomarkers/blood , Caloric Restriction , Electron Transport/drug effects , Enzyme Activation/drug effects , Inflammation/blood , Inflammation/drug therapy , Inflammation/pathology , Male , Metformin/therapeutic use , Mice , Mice, Inbred C57BL , Mitochondria/drug effects , Mitochondria/metabolism , Survival Analysis , Transcriptome/drug effects , Transcriptome/genetics
16.
Aging Cell ; 12(4): 645-51, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23601134

ABSTRACT

Caloric restriction (CR) and down-regulation of the insulin/IGF pathway are the most robust interventions known to increase longevity in lower organisms. However, little is known about the molecular adaptations induced by CR in humans. Here, we report that long-term CR in humans inhibits the IGF-1/insulin pathway in skeletal muscle, a key metabolic tissue. We also demonstrate that CR induces dramatic changes of the skeletal muscle transcriptional profile that resemble those of younger individuals. Finally, in both rats and humans, CR evoked similar responses in the transcriptional profiles of skeletal muscle. This common signature consisted of three key pathways typically associated with longevity: IGF-1/insulin signaling, mitochondrial biogenesis, and inflammation. Furthermore, our data identify promising pathways for therapeutic targets to combat age-related diseases and promote health in humans.


Subject(s)
Caloric Restriction , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction , Transcription, Genetic , Transcriptome , Adipose Tissue/cytology , Adipose Tissue/metabolism , Adult , Aging , Animals , Female , Forkhead Box Protein O3 , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Humans , Insulin-Like Growth Factor I/genetics , Insulin-Like Growth Factor I/metabolism , Kaplan-Meier Estimate , Male , Mitochondrial Turnover , Phosphatidylinositol 3-Kinases/genetics , Principal Component Analysis , Quadriceps Muscle/cytology , Quadriceps Muscle/metabolism , Rats
17.
Mol Cell ; 45(6): 826-35, 2012 Mar 30.
Article in English | MEDLINE | ID: mdl-22387028

ABSTRACT

Although expression of the mammalian RNA-binding protein HuD was considered to be restricted to neurons, we report that HuD is present in pancreatic ß cells, where its levels are controlled by the insulin receptor pathway. We found that HuD associated with a 22-nucleotide segment of the 5' untranslated region (UTR) of preproinsulin (Ins2) mRNA. Modulating HuD abundance did not alter Ins2 mRNA levels, but HuD overexpression decreased Ins2 mRNA translation and insulin production, and conversely, HuD silencing enhanced Ins2 mRNA translation and insulin production. Following treatment with glucose, HuD rapidly dissociated from Ins2 mRNA and enabled insulin biosynthesis. Importantly, HuD-knockout mice displayed higher insulin levels in pancreatic islets, while HuD-overexpressing mice exhibited lower insulin levels in islets and in plasma. In sum, our results identify HuD as a pivotal regulator of insulin translation in pancreatic ß cells.


Subject(s)
ELAV Proteins/metabolism , Insulin/genetics , Insulin/metabolism , Protein Biosynthesis , 5' Untranslated Regions , Animals , ELAV Proteins/genetics , ELAV-Like Protein 4 , Glucose/metabolism , Humans , Insulin-Secreting Cells/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Protein Precursors/genetics
18.
Ageing Res Rev ; 11(3): 390-8, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22210414

ABSTRACT

During aging there is an increasing imbalance of energy intake and expenditure resulting in obesity, frailty, and metabolic disorders. For decades, research has shown that caloric restriction (CR) and exercise can postpone detrimental aspects of aging. These two interventions invoke a similar physiological signature involving pathways associated with stress responses and mitochondrial homeostasis. Nonetheless, CR is able to delay aging processes that result in an increase of both mean and maximum lifespan, whereas exercise primarily increases healthspan. Due to the strict dietary regime necessary to achieve the beneficial effects of CR, most studies to date have focused on rodents and non-human primates. As a consequence, there is vast interest in the development of compounds such as resveratrol, metformin and rapamycin that would activate the same metabolic- and stress-response pathways induced by these interventions without actually restricting caloric intake. Therefore the scope of this review is to (i) describe the benefits of CR and exercise in healthy individuals, (ii) discuss the role of these interventions in the diseased state, and (iii) examine some of the promising pharmacological alternatives such as CR- and exercise-mimetics.


Subject(s)
Aging/physiology , Biomimetic Materials/metabolism , Caloric Restriction , Exercise/physiology , Animals , Biomimetic Materials/therapeutic use , Caloric Restriction/methods , Energy Intake/physiology , Humans , Longevity/physiology , Metabolic Diseases/drug therapy , Metabolic Diseases/metabolism , Metabolic Diseases/prevention & control , Mice
19.
J Gerontol A Biol Sci Med Sci ; 66(3): 247-56, 2011 Mar.
Article in English | MEDLINE | ID: mdl-20974733

ABSTRACT

Nuclear factor E2-related factor-2 (Nrf2) transcription factor is one of the main regulators of intracellular redox balance and a sensor of oxidative and electrophilic stress. Low Nrf2 activity is usually associated with carcinogenesis, but Nrf2 is also considered as an oncogene because it increases survival of transformed cells. Because intracellular redox balance alterations are involved in both senescence and tumorigenesis, we investigated the impact of Nrf2 genetic deletion on cellular immortalization and life span of murine embryonic fibroblasts. We report that Nrf2 genetic deletion promotes immortalization due to an early loss of p53-dependent gene expression. However, compared with control cells, immortalized Nrf2-/- murine embryonic fibroblasts exhibited decreased growth, lower cyclin E levels, and impaired expression of NQO1 and cytochrome b5 reductase. Moreover, SirT1 was also significantly reduced in immortalized Nrf2-/- murine embryonic fibroblasts, and these cells exhibited shorter life span. Our results underscore the dual role of Nrf2 in protection against carcinogenesis and in the delay of cellular aging.


Subject(s)
Cellular Senescence/genetics , Fibroblasts/metabolism , Gene Expression Regulation , NF-E2-Related Factor 2/genetics , Animals , Mice , Sequence Deletion
20.
Sci Rep ; 1: 70, 2011.
Article in English | MEDLINE | ID: mdl-22355589

ABSTRACT

Sirt1 is an NAD(+)-dependent deacetylase that extends lifespan in lower organisms and improves metabolism and delays the onset of age-related diseases in mammals. Here we show that SRT1720, a synthetic compound that was identified for its ability to activate Sirt1 in vitro, extends both mean and maximum lifespan of adult mice fed a high-fat diet. This lifespan extension is accompanied by health benefits including reduced liver steatosis, increased insulin sensitivity, enhanced locomotor activity and normalization of gene expression profiles and markers of inflammation and apoptosis, all in the absence of any observable toxicity. Using a conditional SIRT1 knockout mouse and specific gene knockdowns we show SRT1720 affects mitochondrial respiration in a Sirt1- and PGC-1α-dependent manner. These findings indicate that SRT1720 has long-term benefits and demonstrate for the first time the feasibility of designing novel molecules that are safe and effective in promoting longevity and preventing multiple age-related diseases in mammals.


Subject(s)
Heterocyclic Compounds, 4 or More Rings/pharmacology , Longevity/drug effects , Obesity/physiopathology , Animals , Apoptosis/drug effects , Body Composition/drug effects , Dietary Fats/administration & dosage , Gene Expression/drug effects , Glucose/metabolism , Homeostasis/drug effects , Liver/drug effects , Liver/pathology , Male , Mice , Mice, Inbred C57BL , Pancreas/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...