Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Nat Commun ; 15(1): 467, 2024 Jan 11.
Article in English | MEDLINE | ID: mdl-38212606

ABSTRACT

Dietary restriction (DR) delays aging, but the mechanism remains unclear. We identified polymorphisms in mtd, the fly homolog of OXR1, which influenced lifespan and mtd expression in response to DR. Knockdown in adulthood inhibited DR-mediated lifespan extension in female flies. We found that mtd/OXR1 expression declines with age and it interacts with the retromer, which regulates trafficking of proteins and lipids. Loss of mtd/OXR1 destabilized the retromer, causing improper protein trafficking and endolysosomal defects. Overexpression of retromer genes or pharmacological restabilization with R55 rescued lifespan and neurodegeneration in mtd-deficient flies and endolysosomal defects in fibroblasts from patients with lethal loss-of-function of OXR1 variants. Multi-omic analyses in flies and humans showed that decreased Mtd/OXR1 is associated with aging and neurological diseases. mtd/OXR1 overexpression rescued age-related visual decline and tauopathy in a fly model. Hence, OXR1 plays a conserved role in preserving retromer function and is critical for neuronal health and longevity.


Subject(s)
Aging , Nervous System Diseases , Humans , Female , Aging/genetics , Longevity/genetics , Neurons/metabolism , Nervous System Diseases/metabolism , Brain/metabolism , Caloric Restriction , Mitochondrial Proteins/metabolism
2.
Elife ; 122023 09 20.
Article in English | MEDLINE | ID: mdl-37728328

ABSTRACT

The Maillard reaction, a chemical reaction between amino acids and sugars, is exploited to produce flavorful food ubiquitously, from the baking industry to our everyday lives. However, the Maillard reaction also occurs in all cells, from prokaryotes to eukaryotes, forming advanced glycation end-products (AGEs). AGEs are a heterogeneous group of compounds resulting from the irreversible reaction between biomolecules and α-dicarbonyls (α-DCs), including methylglyoxal (MGO), an unavoidable byproduct of anaerobic glycolysis and lipid peroxidation. We previously demonstrated that Caenorhabditis elegans mutants lacking the glod-4 glyoxalase enzyme displayed enhanced accumulation of α-DCs, reduced lifespan, increased neuronal damage, and touch hypersensitivity. Here, we demonstrate that glod-4 mutation increased food intake and identify that MGO-derived hydroimidazolone, MG-H1, is a mediator of the observed increase in food intake. RNAseq analysis in glod-4 knockdown worms identified upregulation of several neurotransmitters and feeding genes. Suppressor screening of the overfeeding phenotype identified the tdc-1-tyramine-tyra-2/ser-2 signaling as an essential pathway mediating AGE (MG-H1)-induced feeding in glod-4 mutants. We also identified the elt-3 GATA transcription factor as an essential upstream regulator for increased feeding upon accumulation of AGEs by partially controlling the expression of tdc-1 gene. Furthermore, the lack of either tdc-1 or tyra-2/ser-2 receptors suppresses the reduced lifespan and rescues neuronal damage observed in glod-4 mutants. Thus, in C. elegans, we identified an elt-3 regulated tyramine-dependent pathway mediating the toxic effects of MG-H1 AGE. Understanding this signaling pathway may help understand hedonistic overfeeding behavior observed due to modern AGE-rich diets.


Subject(s)
Caenorhabditis elegans Proteins , Caenorhabditis elegans , Animals , Caenorhabditis elegans/physiology , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Pyruvaldehyde/metabolism , Magnesium Oxide/metabolism , GATA Transcription Factors/genetics , GATA Transcription Factors/metabolism , Signal Transduction , Tyramine/metabolism , Glycation End Products, Advanced/metabolism , Eating
3.
Nat Commun ; 13(1): 3156, 2022 06 07.
Article in English | MEDLINE | ID: mdl-35672419

ABSTRACT

Many vital processes in the eye are under circadian regulation, and circadian dysfunction has emerged as a potential driver of eye aging. Dietary restriction is one of the most robust lifespan-extending therapies and amplifies circadian rhythms with age. Herein, we demonstrate that dietary restriction extends lifespan in Drosophila melanogaster by promoting circadian homeostatic processes that protect the visual system from age- and light-associated damage. Altering the positive limb core molecular clock transcription factor, CLOCK, or CLOCK-output genes, accelerates visual senescence, induces a systemic immune response, and shortens lifespan. Flies subjected to dietary restriction are protected from the lifespan-shortening effects of photoreceptor activation. Inversely, photoreceptor inactivation, achieved via mutating rhodopsin or housing flies in constant darkness, primarily extends the lifespan of flies reared on a high-nutrient diet. Our findings establish the eye as a diet-sensitive modulator of lifespan and indicates that vision is an antagonistically pleiotropic process that contributes to organismal aging.


Subject(s)
Drosophila Proteins , Drosophila melanogaster , Eye , Animals , Circadian Rhythm/genetics , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Gene Expression Regulation , Longevity/genetics , Transcription Factors/genetics
4.
Elife ; 82019 02 21.
Article in English | MEDLINE | ID: mdl-30789342

ABSTRACT

In the present study we show that the master myogenic regulatory factor, MYOD1, is a positive modulator of molecular clock amplitude and functions with the core clock factors for expression of clock-controlled genes in skeletal muscle. We demonstrate that MYOD1 directly regulates the expression and circadian amplitude of the positive core clock factor Bmal1. We identify a non-canonical E-box element in Bmal1 and demonstrate that is required for full MYOD1-responsiveness. Bimolecular fluorescence complementation assays demonstrate that MYOD1 colocalizes with both BMAL1 and CLOCK throughout myonuclei. We demonstrate that MYOD1 and BMAL1:CLOCK work in a synergistic fashion through a tandem E-box to regulate the expression and amplitude of the muscle specific clock-controlled gene, Titin-cap (Tcap). In conclusion, these findings reveal mechanistic roles for the muscle specific transcription factor MYOD1 in the regulation of molecular clock amplitude as well as synergistic regulation of clock-controlled genes in skeletal muscle.


Subject(s)
Circadian Rhythm Signaling Peptides and Proteins/biosynthesis , Gene Expression Regulation , Muscle, Skeletal/enzymology , MyoD Protein/metabolism , ARNTL Transcription Factors/metabolism , Animals , CLOCK Proteins/metabolism , Circadian Clocks , Connectin/metabolism , Mice, Inbred C57BL
5.
Int J Mol Sci ; 19(12)2018 Nov 29.
Article in English | MEDLINE | ID: mdl-30501099

ABSTRACT

Aging is often defined as the accumulation of damage at the molecular and cellular levels which, over time, results in marked physiological impairments throughout the organism. Dietary restriction (DR) has been recognized as one of the strongest lifespan extending therapies observed in a wide array of organisms. Recent studies aimed at elucidating how DR promotes healthy aging have demonstrated a vital role of the digestive tract in mediating the beneficial effects of DR. Here, we review how dietary restriction influences gut metabolic homeostasis and immune function. Our discussion is focused on studies of the Drosophila digestive tract, where we describe in detail the potential mechanisms in which DR enhances maintenance of the intestinal epithelial barrier, up-regulates lipid metabolic processes, and improves the ability of the gut to deal with damage or stress. We also examine evidence of a tissue-tissue crosstalk between gut and neighboring organs including brain and fat body. Taken together, we argue that the Drosophila gut plays a critical role in DR-mediated lifespan extension.


Subject(s)
Caloric Restriction , Longevity , Animals , Drosophila , Drosophila Proteins/metabolism , Gastrointestinal Tract/metabolism , Humans , Intestinal Mucosa/metabolism
6.
Ecol Evol ; 7(7): 2346-2356, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28405298

ABSTRACT

Mobile species will migrate considerable distances to find habitats suitable for meeting life history requirements, and stream-dwelling salmonids are no exception. In April-October 2014, we used radio-telemetry to examine habitat use and movement of 36 Colorado River cutthroat trout Oncorhynchus clarkii pleuriticus (CRCT) in a 14.9-km fragment of Milk Creek, a relatively low-elevation stream in the Rocky Mountains (Colorado). We also used a network of data loggers to track stream temperature across time and space. Our objectives were to (1) characterize distribution and movement of CRCT, (2) evaluate seasonal differences in distribution and movement of CRCT, and (3) explore the relationship between stream temperature and distribution and movement of CRCT. During the course of our study, median range of CRCT was 4.81 km (range = 0.14-10.94) and median total movement was 5.94 km (range = 0.14-26.02). Median location of CRCT was significantly further upstream in summer than in spring, whereas range and movement of CRCT were greater in spring than in summer. Twenty-six of the 27 CRCT tracked through mid-June displayed a potamodromous (freshwater migratory) life history, migrating 1.8-8.0 km upstream during the spring spawning season. Four of the seven CRCT tracked through July migrated >1.4 km in summer. CRCT selected relatively cool reaches during summer months, and early-summer movement was positively correlated with mean stream temperature. Study fish occupied stream segments in spring and fall that were thermally unsuitable, if not lethal, to the species in summer. Although transmitter loss limited the scope of inference, our findings suggest that preferred habitat is a moving target in Milk Creek, and that CRCT move to occupy that target. Because mobile organisms move among complementary habitats and exploit seasonally-unsuitable reaches, we recommend that spatial and temporal variability be accounted for in delineations of distributional boundaries.

7.
Skelet Muscle ; 6: 12, 2016.
Article in English | MEDLINE | ID: mdl-27486508

ABSTRACT

BACKGROUND: Diabetes is the seventh leading cause of death in the USA, and disruption of circadian rhythms is gaining recognition as a contributing factor to disease prevalence. This disease is characterized by hyperglycemia and glucose intolerance and symptoms caused by failure to produce and/or respond to insulin. The skeletal muscle is a key insulin-sensitive metabolic tissue, taking up ~80 % of postprandial glucose. To address the role of the skeletal muscle molecular clock to insulin sensitivity and glucose tolerance, we generated an inducible skeletal muscle-specific Bmal1 (-/-) mouse (iMSBmal1 (-/-)). RESULTS: Progressive changes in body composition (decreases in percent fat) were seen in the iMSBmal1 (-/-) mice from 3 to 12 weeks post-treatment as well as glucose intolerance and non-fasting hyperglycemia. Ex vivo analysis of glucose uptake revealed that the extensor digitorum longus (EDL) muscles did not respond to either insulin or 5-aminoimidazole-4-carboxamide ribonucleotide (AICAR) stimulation. RT-PCR and Western blot analyses demonstrated a significant decrease in mRNA expression and protein content of the muscle glucose transporter (Glut4). We also found that both mRNA expression and activity of two key rate-limiting enzymes of glycolysis, hexokinase 2 (Hk2) and phosphofructokinase 1 (Pfk1), were significantly reduced in the iMSBmal1 (-/-) muscle. Lastly, results from metabolomics analyses provided evidence of decreased glycolytic flux and uncovered decreases in some tricarboxylic acid (TCA) intermediates with increases in amino acid levels in the iMSBmal1 (-/-) muscle. These findings suggest that the muscle is relying predominantly on fat as a fuel with increased protein breakdown to support the TCA cycle. CONCLUSIONS: These data support a fundamental role for Bmal1, the endogenous circadian clock, in glucose metabolism in the skeletal muscle. Our findings have implicated altered molecular clock dictating significant changes in altered substrate metabolism in the absence of feeding or activity changes. The changes in body composition in our model also highlight the important role that changes in skeletal muscle carbohydrate, and fat metabolism can play in systemic metabolism.


Subject(s)
ARNTL Transcription Factors/physiology , Blood Glucose/metabolism , Circadian Rhythm , Insulin/metabolism , Muscle, Skeletal/metabolism , Adipose Tissue/metabolism , Aminoimidazole Carboxamide/administration & dosage , Aminoimidazole Carboxamide/analogs & derivatives , Animals , Blood Glucose/analysis , Body Weight , Female , Glucose Transporter Type 4/metabolism , Hexokinase/metabolism , Homeostasis , Hypoglycemic Agents/administration & dosage , Insulin/administration & dosage , Insulin/blood , Male , Mice , Mice, Knockout , Motor Activity , Phosphofructokinase-1, Muscle Type/metabolism , RNA, Messenger/metabolism , Ribonucleotides/administration & dosage
9.
J Cell Sci ; 129(8): 1671-84, 2016 Apr 15.
Article in English | MEDLINE | ID: mdl-26945058

ABSTRACT

Collagen VI myopathies are genetic disorders caused by mutations in collagen 6 A1, A2 and A3 genes, ranging from the severe Ullrich congenital muscular dystrophy to the milder Bethlem myopathy, which is recapitulated by collagen-VI-null (Col6a1(-/-)) mice. Abnormalities in mitochondria and autophagic pathway have been proposed as pathogenic causes of collagen VI myopathies, but the link between collagen VI defects and these metabolic circuits remains unknown. To unravel the expression profiling perturbation in muscles with collagen VI myopathies, we performed a deep RNA profiling in both Col6a1(-/-)mice and patients with collagen VI pathology. The interactome map identified common pathways suggesting a previously undetected connection between circadian genes and collagen VI pathology. Intriguingly, Bmal1(-/-)(also known as Arntl) mice, a well-characterized model displaying arrhythmic circadian rhythms, showed profound deregulation of the collagen VI pathway and of autophagy-related genes. The involvement of circadian rhythms in collagen VI myopathies is new and links autophagy and mitochondrial abnormalities. It also opens new avenues for therapies of hereditary myopathies to modulate the molecular clock or potential gene-environment interactions that might modify muscle damage pathogenesis.


Subject(s)
ARNTL Transcription Factors/genetics , Circadian Clocks/physiology , Collagen Type VI/genetics , Contracture/genetics , Mitochondria/physiology , Muscular Dystrophies/congenital , Mutation/genetics , Sclerosis/genetics , Animals , Autophagy/genetics , Gene Expression Profiling , Humans , Mice , Mice, Knockout , Microarray Analysis , Muscular Dystrophies/genetics , RNA/analysis
10.
J Physiol ; 593(24): 5387-404, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26486627

ABSTRACT

KEY POINTS: The endogenous molecular clock in skeletal muscle is necessary for maintenance of phenotype and function. Loss of Bmal1 solely from adult skeletal muscle (iMSBmal1(-/-) ) results in reductions in specific tension, increased oxidative fibre type and increased muscle fibrosis with no change in feeding or activity. Disruption of the molecular clock in adult skeletal muscle is sufficient to induce changes in skeletal muscle similar to those seen in the Bmal1 knockout mouse (Bmal1(-/-) ), a model of advanced ageing. iMSBmal1(-/-) mice develop increased bone calcification and decreased joint collagen, which in combination with the functional changes in skeletal muscle results in altered gait. This study uncovers a fundamental role for the skeletal muscle clock in musculoskeletal homeostasis with potential implications for ageing. ABSTRACT: Disruption of circadian rhythms in humans and rodents has implicated a fundamental role for circadian rhythms in ageing and the development of many chronic diseases including diabetes, cardiovascular disease, depression and cancer. The molecular clock mechanism underlies circadian rhythms and is defined by a transcription-translation feedback loop with Bmal1 encoding a core molecular clock transcription factor. Germline Bmal1 knockout (Bmal1 KO) mice have a shortened lifespan, show features of advanced ageing and exhibit significant weakness with decreased maximum specific tension at the whole muscle and single fibre levels. We tested the role of the molecular clock in adult skeletal muscle by generating mice that allow for the inducible skeletal muscle-specific deletion of Bmal1 (iMSBmal1). Here we show that disruption of the molecular clock, specifically in adult skeletal muscle, is associated with a muscle phenotype including reductions in specific tension, increased oxidative fibre type, and increased muscle fibrosis similar to that seen in the Bmal1 KO mouse. Remarkably, the phenotype observed in the iMSBmal1(-/-) mice was not limited to changes in muscle. Similar to the germline Bmal1 KO mice, we observed significant bone and cartilage changes throughout the body suggesting a role for the skeletal muscle molecular clock in both the skeletal muscle niche and the systemic milieu. This emerging area of circadian rhythms and the molecular clock in skeletal muscle holds the potential to provide significant insight into intrinsic mechanisms of the maintenance of muscle quality and function as well as identifying a novel crosstalk between skeletal muscle, cartilage and bone.


Subject(s)
ARNTL Transcription Factors/metabolism , Biological Clocks , Muscle, Skeletal/metabolism , ARNTL Transcription Factors/genetics , Animals , Bone and Bones/pathology , Calcinosis/genetics , Collagen/metabolism , Fibrosis , Gait , Joints/pathology , Mice , Mice, Inbred C57BL , Muscle, Skeletal/growth & development , Muscle, Skeletal/pathology , Phenotype
11.
Skelet Muscle ; 5: 17, 2015.
Article in English | MEDLINE | ID: mdl-26000164

ABSTRACT

BACKGROUND: Skeletal muscle is a major contributor to whole-body metabolism as it serves as a depot for both glucose and amino acids, and is a highly metabolically active tissue. Within skeletal muscle exists an intrinsic molecular clock mechanism that regulates the timing of physiological processes. A key function of the clock is to regulate the timing of metabolic processes to anticipate time of day changes in environmental conditions. The purpose of this study was to identify metabolic genes that are expressed in a circadian manner and determine if these genes are regulated downstream of the intrinsic molecular clock by assaying gene expression in an inducible skeletal muscle-specific Bmal1 knockout mouse model (iMS-Bmal1 (-/-) ). METHODS: We used circadian statistics to analyze a publicly available, high-resolution time-course skeletal muscle expression dataset. Gene ontology analysis was utilized to identify enriched biological processes in the skeletal muscle circadian transcriptome. We generated a tamoxifen-inducible skeletal muscle-specific Bmal1 knockout mouse model and performed a time-course microarray experiment to identify gene expression changes downstream of the molecular clock. Wheel activity monitoring was used to assess circadian behavioral rhythms in iMS-Bmal1 (-/-) and control iMS-Bmal1 (+/+) mice. RESULTS: The skeletal muscle circadian transcriptome was highly enriched for metabolic processes. Acrophase analysis of circadian metabolic genes revealed a temporal separation of genes involved in substrate utilization and storage over a 24-h period. A number of circadian metabolic genes were differentially expressed in the skeletal muscle of the iMS-Bmal1 (-/-) mice. The iMS-Bmal1 (-/-) mice displayed circadian behavioral rhythms indistinguishable from iMS-Bmal1 (+/+) mice. We also observed a gene signature indicative of a fast to slow fiber-type shift and a more oxidative skeletal muscle in the iMS-Bmal1 (-/-) model. CONCLUSIONS: These data provide evidence that the intrinsic molecular clock in skeletal muscle temporally regulates genes involved in the utilization and storage of substrates independent of circadian activity. Disruption of this mechanism caused by phase shifts (that is, social jetlag) or night eating may ultimately diminish skeletal muscle's ability to efficiently maintain metabolic homeostasis over a 24-h period.

12.
J Comp Physiol B ; 181(8): 1045-62, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21630040

ABSTRACT

We measured the ontogeny of salinity tolerance and the preparatory hypo-osmoregulatory physiological changes for seawater entry in green sturgeon (Acipenser medirostris), an anadromous species occurring along the Pacific Coast of North America. Salinity tolerance was measured every 2 weeks starting in 40-day post-hatch (dph) juveniles and was repeated until 100% survival at 34‰ was achieved. Fish were subjected to step increases in salinity (5‰ 12 h(-1)) that culminated in a 72-h exposure to a target salinity, and treatment groups (0, 15, 20, 25, 30, 34‰; and abrupt exposure to 34‰) were adjusted as fish developed. After 100% survival was achieved (134 dph), a second experiment tested two sizes of fish for 28-day seawater (33‰) tolerance, and gill and gastrointestinal tract tissues were sampled. Their salinity tolerance increased and plasma osmolality decreased with increasing size and age, and electron microscopy revealed three types of mitochondria-rich cells: one in fresh water and two in seawater. In addition, fish held on a natural photoperiod in fresh water at 19°C showed peaks in cortisol, thyroid hormones and gill and pyloric ceca Na(+), K(+)-ATPase activities at body sizes associated with seawater tolerance. Therefore, salinity tolerance in green sturgeon increases during ontogeny (e.g., as these juveniles may move down estuaries to the ocean) with increases in body size. Also, physiological and morphological changes associated with seawater readiness increased in freshwater-reared juveniles and peaked at their seawater-tolerant ages and body sizes. Their seawater-ready body size also matched that described for swimming performance decreases, presumably associated with downstream movements. Therefore, juvenile green sturgeon develop structures and physiological changes appropriate for seawater entry while growing in fresh water, indicating that hypo-osmoregulatory changes may proceed by multiple routes in sturgeons.


Subject(s)
Fishes/physiology , Growth and Development/physiology , Salt Tolerance/physiology , Seawater , Age Factors , Animal Structures/enzymology , Animals , Body Size/physiology , Chlorides/blood , Epithelial Cells/ultrastructure , Fresh Water , Gills/cytology , Gills/enzymology , Hematocrit , Hydrocortisone/blood , Intestines/enzymology , Microscopy, Electron, Scanning , Microscopy, Electron, Transmission , Mitochondria/ultrastructure , Osmolar Concentration , Plasma/chemistry , Salinity , Sodium/blood , Sodium-Potassium-Exchanging ATPase/metabolism , Survival Rate , Thyroxine/blood , Triiodothyronine/blood
13.
Empl Benefits J ; 28(4): 53-61, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14712737

ABSTRACT

All employee benefit plans are potential targets of fraudulent schemes. Smaller plans are targeted by unscrupulous brokers and promoters selling fraudulent policies; plans large enough to be self-insured face greater risks of fraud by providers and participants misrepresenting claims. Plan trustees, administrators and consultants should be alert to the many ways fraudulent schemes manifest themselves and to the legal remedies available; establish investigative programs to detect and discourage fraud; and promote education and plan incentives for participants to report fraud.


Subject(s)
Financial Audit/standards , Fraud/prevention & control , Health Benefit Plans, Employee/economics , Criminal Law , Employee Retirement Income Security Act , Fees and Charges , Fraud/legislation & jurisprudence , Health Benefit Plans, Employee/legislation & jurisprudence , Hospitals, Proprietary/economics , Hospitals, Proprietary/legislation & jurisprudence , Insurance Claim Reporting/legislation & jurisprudence , Liability, Legal , Marketing , Salaries and Fringe Benefits , United States
SELECTION OF CITATIONS
SEARCH DETAIL
...