Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
Hepatology ; 65(2): 616-630, 2017 02.
Article in English | MEDLINE | ID: mdl-27809334

ABSTRACT

The regenerative capacity of the liver is essential for recovery from surgical resection or injuries induced by trauma or toxins. During liver regeneration, the concentration of nicotinamide adenine dinucleotide (NAD) falls, at least in part due to metabolic competition for precursors. To test whether NAD availability restricts the rate of liver regeneration, we supplied nicotinamide riboside (NR), an NAD precursor, in the drinking water of mice subjected to partial hepatectomy. NR increased DNA synthesis, mitotic index, and mass restoration in the regenerating livers. Intriguingly, NR also ameliorated the steatosis that normally accompanies liver regeneration. To distinguish the role of hepatocyte NAD levels from any systemic effects of NR, we generated mice overexpressing nicotinamide phosphoribosyltransferase, a rate-limiting enzyme for NAD synthesis, specifically in the liver. Nicotinamide phosphoribosyltransferase overexpressing mice were mildly hyperglycemic at baseline and, similar to mice treated with NR, exhibited enhanced liver regeneration and reduced steatosis following partial hepatectomy. Conversely, mice lacking nicotinamide phosphoribosyltransferase in hepatocytes exhibited impaired regenerative capacity that was completely rescued by administering NR. CONCLUSION: NAD availability is limiting during liver regeneration, and supplementation with precursors such as NR may be therapeutic in settings of acute liver injury. (Hepatology 2017;65:616-630).


Subject(s)
Liver Regeneration/drug effects , Liver Regeneration/physiology , Liver/pathology , NAD/biosynthesis , Niacinamide/analogs & derivatives , Animals , Disease Models, Animal , Fluorescent Antibody Technique , Hepatectomy/methods , Immunoblotting , Immunohistochemistry , Liver/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , NAD/metabolism , Niacinamide/pharmacology , Pyridinium Compounds , Random Allocation , Sensitivity and Specificity
2.
FASEB J ; 31(9): 3934-3949, 2017 09.
Article in English | MEDLINE | ID: mdl-28490482

ABSTRACT

Rapamycin is a clinically important drug that is used in transplantation and cancer therapy but which causes a number of side effects, including male infertility. Its canonical target, mammalian target of rapamycin complex 1 (mTORC1), plays a key role in metabolism and binds chromatin; however, its precise role in the male germline has not been elucidated. Here, we inactivate the core component, Raptor, to show that mTORC1 function is critical for male meiosis and the inactivation of sex chromosomes. Disruption of the Raptor gene impairs chromosomal synapsis and prevents the efficient spreading of silencing factors into the XY chromatin. Accordingly, mRNA for XY-linked genes remains inappropriately expressed in Raptor-deficient mice. Molecularly, the failure to suppress gene expression corresponded with deficiencies in 2 repressive chromatin markers, H3K9 dimethylation and H3K9 trimethylation, in the XY body. Together, these results demonstrate that mTORC1 has an essential role in the meiotic progression and silencing of sex chromosomes in the male germline, which may explain the infertility that has been associated with such inhibitors as rapamycin.-Xiong, M., Zhu, Z., Tian, S., Zhu, R., Bai, S., Fu, K., Davis, J. G., Sun, Z., Baur, J. A., Zheng, K., Ye, L. Conditional ablation of Raptor in the male germline causes infertility due to meiotic arrest and impaired inactivation of sex chromosomes.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Infertility, Male/genetics , Meiosis/physiology , Sex Chromosomes/physiology , Adaptor Proteins, Signal Transducing/genetics , Animals , Male , Mechanistic Target of Rapamycin Complex 1 , Mice , Mice, Knockout , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Regulatory-Associated Protein of mTOR , Sex Chromosomes/genetics , Spermatogenesis/genetics , Spermatogenesis/physiology , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism
3.
J Biol Chem ; 290(3): 1546-58, 2015 Jan 16.
Article in English | MEDLINE | ID: mdl-25411251

ABSTRACT

The NAD biosynthetic precursors nicotinamide mononucleotide and nicotinamide riboside are reported to confer resistance to metabolic defects induced by high fat feeding in part by promoting oxidative metabolism in skeletal muscle. Similar effects are obtained by germ line deletion of major NAD-consuming enzymes, suggesting that the bioavailability of NAD is limiting for maximal oxidative capacity. However, because of their systemic nature, the degree to which these interventions exert cell- or tissue-autonomous effects is unclear. Here, we report a tissue-specific approach to increase NAD biosynthesis only in muscle by overexpressing nicotinamide phosphoribosyltransferase, the rate-limiting enzyme in the salvage pathway that converts nicotinamide to NAD (mNAMPT mice). These mice display a ∼50% increase in skeletal muscle NAD levels, comparable with the effects of dietary NAD precursors, exercise regimens, or loss of poly(ADP-ribose) polymerases yet surprisingly do not exhibit changes in muscle mitochondrial biogenesis or mitochondrial function and are equally susceptible to the metabolic consequences of high fat feeding. We further report that chronic elevation of muscle NAD in vivo does not perturb the NAD/NADH redox ratio. These studies reveal for the first time the metabolic effects of tissue-specific increases in NAD synthesis and suggest that critical sites of action for supplemental NAD precursors reside outside of the heart and skeletal muscle.


Subject(s)
Cytokines/metabolism , Muscle, Skeletal/metabolism , NAD/biosynthesis , Nicotinamide Phosphoribosyltransferase/metabolism , Oxygen/metabolism , Animals , Binding Sites , Calorimetry , Chromatography, High Pressure Liquid , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mitochondria, Muscle/metabolism , NAD/metabolism , Nicotinamide Mononucleotide/metabolism , Oxidation-Reduction , Poly(ADP-ribose) Polymerases/metabolism
4.
J Neurosci ; 34(12): 4418-31, 2014 Mar 19.
Article in English | MEDLINE | ID: mdl-24647961

ABSTRACT

Modern society enables a shortening of sleep times, yet long-term consequences of extended wakefulness on the brain are largely unknown. Essential for optimal alertness, locus ceruleus neurons (LCns) are metabolically active neurons that fire at increased rates across sustained wakefulness. We hypothesized that wakefulness is a metabolic stressor to LCns and that, with extended wakefulness, adaptive mitochondrial metabolic responses fail and injury ensues. The nicotinamide adenine dinucleotide-dependent deacetylase sirtuin type 3 (SirT3) coordinates mitochondrial energy production and redox homeostasis. We find that brief wakefulness upregulates SirT3 and antioxidants in LCns, protecting metabolic homeostasis. Strikingly, mice lacking SirT3 lose the adaptive antioxidant response and incur oxidative injury in LCns across brief wakefulness. When wakefulness is extended for longer durations in wild-type mice, SirT3 protein declines in LCns, while oxidative stress and acetylation of mitochondrial proteins, including electron transport chain complex I proteins, increase. In parallel with metabolic dyshomeostasis, apoptosis is activated and LCns are lost. This work identifies mitochondrial stress in LCns upon wakefulness, highlights an essential role for SirT3 activation in maintaining metabolic homeostasis in LCns across wakefulness, and demonstrates that extended wakefulness results in reduced SirT3 activity and, ultimately, degeneration of LCns.


Subject(s)
Locus Coeruleus/metabolism , Nerve Degeneration/metabolism , Neurons/metabolism , Sleep Deprivation/metabolism , Sleep/physiology , Wakefulness/physiology , Animals , Corticosterone/blood , Locus Coeruleus/pathology , Male , Mice , Mice, Knockout , Mitochondria/genetics , Mitochondria/metabolism , Nerve Degeneration/pathology , Neurons/pathology , Oxidative Stress/physiology , Sirtuin 3/genetics , Sirtuin 3/metabolism , Sleep Deprivation/pathology , Up-Regulation
5.
Nat Cardiovasc Res ; 3(10): 1236-1248, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39294272

ABSTRACT

Nicotinamide adenine dinucleotide (NAD+) is an essential co-factor in metabolic reactions and co-substrate for signaling enzymes. Failing human hearts display decreased expression of the major NAD+ biosynthetic enzyme nicotinamide phosphoribosyltransferase (Nampt) and lower NAD+ levels, and supplementation with NAD+ precursors is protective in preclinical models. Here we show that Nampt loss in adult cardiomyocytes caused depletion of NAD+ along with marked metabolic derangements, hypertrophic remodeling and sudden cardiac deaths, despite unchanged ejection fraction, endurance and mitochondrial respiratory capacity. These effects were directly attributable to NAD+ loss as all were ameliorated by restoring cardiac NAD+ levels with the NAD+ precursor nicotinamide riboside (NR). Electrocardiograms revealed that loss of myocardial Nampt caused a shortening of QT intervals with spontaneous lethal arrhythmias causing sudden cardiac death. Thus, changes in NAD+ concentration can have a profound influence on cardiac physiology even at levels sufficient to maintain energetics.


Subject(s)
Arrhythmias, Cardiac , Cardiomyopathy, Hypertrophic , Energy Metabolism , Myocytes, Cardiac , NAD , Nicotinamide Phosphoribosyltransferase , Nicotinamide Phosphoribosyltransferase/metabolism , Nicotinamide Phosphoribosyltransferase/genetics , NAD/metabolism , Animals , Cardiomyopathy, Hypertrophic/metabolism , Cardiomyopathy, Hypertrophic/genetics , Cardiomyopathy, Hypertrophic/pathology , Arrhythmias, Cardiac/metabolism , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Disease Models, Animal , Cytokines/metabolism , Mice, Knockout , Mice, Inbred C57BL , Pyridinium Compounds , Male , Death, Sudden, Cardiac/etiology , Death, Sudden, Cardiac/pathology , Mice , Niacinamide/analogs & derivatives , Niacinamide/pharmacology , Niacinamide/therapeutic use , Niacinamide/metabolism , Electrocardiography
6.
JCI Insight ; 9(16)2024 Jul 18.
Article in English | MEDLINE | ID: mdl-39171530

ABSTRACT

Friedreich's ataxia (FRDA) is a progressive disorder caused by insufficient expression of frataxin, which plays a critical role in assembly of iron-sulfur centers in mitochondria. Individuals are cognitively normal but display a loss of motor coordination and cardiac abnormalities. Many ultimately develop heart failure. Administration of nicotinamide adenine dinucleotide-positive (NAD+) precursors has shown promise in human mitochondrial myopathy and rodent models of heart failure, including mice lacking frataxin in cardiomyocytes. We studied mice with systemic knockdown of frataxin (shFxn), which display motor deficits and early mortality with cardiac hypertrophy. Hearts in these mice do not "fail" per se but become hyperdynamic with small chamber sizes. Data from an ongoing natural history study indicate that hyperdynamic hearts are observed in young individuals with FRDA, suggesting that the mouse model could reflect early pathology. Administering nicotinamide mononucleotide or riboside to shFxn mice increases survival, modestly improves cardiac hypertrophy, and limits increases in ejection fraction. Mechanistically, most of the transcriptional and metabolic changes induced by frataxin knockdown are insensitive to NAD+ precursor administration, but glutathione levels are increased, suggesting improved antioxidant capacity. Overall, our findings indicate that NAD+ precursors are modestly cardioprotective in this model of FRDA and warrant further investigation.


Subject(s)
Disease Models, Animal , Frataxin , Friedreich Ataxia , Iron-Binding Proteins , NAD , Animals , Friedreich Ataxia/metabolism , Friedreich Ataxia/pathology , Friedreich Ataxia/genetics , Iron-Binding Proteins/genetics , Iron-Binding Proteins/metabolism , Mice , Humans , NAD/metabolism , Phenotype , Male , Cardiomegaly/metabolism , Cardiomegaly/pathology , Nicotinamide Mononucleotide/pharmacology , Niacinamide/analogs & derivatives , Niacinamide/pharmacology , Female , Gene Knockdown Techniques , Pyridinium Compounds , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology
7.
Nat Metab ; 5(3): 414-430, 2023 03.
Article in English | MEDLINE | ID: mdl-36914909

ABSTRACT

Our understanding of how global changes in cellular metabolism contribute to human kidney disease remains incompletely understood. Here we show that nicotinamide adenine dinucleotide (NAD+) deficiency drives mitochondrial dysfunction causing inflammation and kidney disease development. Using unbiased global metabolomics in healthy and diseased human kidneys, we identify NAD+ deficiency as a disease signature. Furthermore using models of cisplatin- or ischaemia-reperfusion induced kidney injury in male mice we observed NAD+ depletion Supplemental nicotinamide riboside or nicotinamide mononucleotide restores NAD+ levels and improved kidney function. We find that cisplatin exposure causes cytosolic leakage of mitochondrial RNA (mtRNA) and activation of the cytosolic pattern recognition receptor retinoic acid-inducible gene I (RIG-I), both of which can be ameliorated by restoring NAD+. Male mice with RIG-I knock-out (KO) are protected from cisplatin-induced kidney disease. In summary, we demonstrate that the cytosolic release of mtRNA and RIG-I activation is an NAD+-sensitive mechanism contributing to kidney disease.


Subject(s)
Cisplatin , NAD , Animals , Humans , Male , Mice , Cisplatin/toxicity , Dietary Supplements , Inflammation , Kidney/metabolism , NAD/metabolism , RNA, Mitochondrial
8.
Cell Rep ; 42(3): 112155, 2023 03 28.
Article in English | MEDLINE | ID: mdl-36857182

ABSTRACT

The most abundant cellular divalent cations, Mg2+ (mM) and Ca2+ (nM-µM), antagonistically regulate divergent metabolic pathways with several orders of magnitude affinity preference, but the physiological significance of this competition remains elusive. In mice consuming a Western diet, genetic ablation of the mitochondrial Mg2+ channel Mrs2 prevents weight gain, enhances mitochondrial activity, decreases fat accumulation in the liver, and causes prominent browning of white adipose. Mrs2 deficiency restrains citrate efflux from the mitochondria, making it unavailable to support de novo lipogenesis. As citrate is an endogenous Mg2+ chelator, this may represent an adaptive response to a perceived deficit of the cation. Transcriptional profiling of liver and white adipose reveals higher expression of genes involved in glycolysis, ß-oxidation, thermogenesis, and HIF-1α-targets, in Mrs2-/- mice that are further enhanced under Western-diet-associated metabolic stress. Thus, lowering mMg2+ promotes metabolism and dampens diet-induced obesity and metabolic syndrome.


Subject(s)
Adipose Tissue, Brown , Energy Metabolism , Animals , Mice , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/metabolism , Cation Transport Proteins , Diet , Diet, High-Fat , Energy Metabolism/genetics , Mitochondria/metabolism , Mitochondrial Proteins , Obesity/metabolism , Thermogenesis/genetics
9.
Aging (Albany NY) ; 12(18): 17786-17799, 2020 Sep 22.
Article in English | MEDLINE | ID: mdl-32960787

ABSTRACT

Rapamycin delays multiple age-related conditions and extends lifespan in organisms ranging from yeast to mice. However, the mechanisms by which rapamycin influences longevity are incompletely understood. The objective of this study was to investigate the effect of rapamycin on NAD+/NADH redox balance. We report that the NAD+/NADH ratio of C2C12 myoblasts or differentiated myotubes significantly decreases over time in culture, and that rapamycin prevents this effect. Despite lowering the NADH available to support ATP generation, rapamycin increases ATP availability, consistent with lowering energetic demand. Although rapamycin did not change the NAD+/NADH ratio or steady-state ATP concentration in the livers, kidneys, or muscles of young mice, optical redox imaging revealed that rapamycin caused a substantial decline in the NADH content and an increase in the optical redox ratio (a surrogate of NAD+/NADH redox ratio) in muscles from aged mice. Collectively, these data suggest that rapamycin favors a more oxidized NAD+/NADH ratio in aged muscle, which may influence metabolism and the activity of NAD+-dependent enzymes. This study provides new insight into the mechanisms by which rapamycin might influence the aging process to improve health and longevity among the aging population.

10.
Mol Imaging Biol ; 21(3): 417-425, 2019 06.
Article in English | MEDLINE | ID: mdl-30977079

ABSTRACT

PURPOSE: Optical redox imaging (ORI) technique images cellular autofluorescence of nicotinamide adenine dinucleotide (NADH) and oxidized flavoproteins (Fp containing FAD, i.e., flavin adenine dinucleotide). ORI has found wide applications in the study of cellular energetics and metabolism and may potentially assist in disease diagnosis and prognosis. Fixed tissues have been reported to exhibit autofluorescence with similar spectral characteristics to those of NADH and Fp. However, few studies report on quantitative ORI of formalin-fixed paraffin-embedded (FFPE) unstained tissue slides for disease biomarkers. We investigate whether ORI of FFPE unstained skeletal muscle slides may provide relevant quantitative biological information. PROCEDURES: Living mouse muscle fibers and frozen and FFPE mouse muscle slides were subjected to ORI. Living mouse muscle fibers were imaged ex vivo before and after paraformaldehyde fixation. FFPE muscle slides of three mouse groups (young, mid-age, and muscle-specific overexpression of nicotinamide phosphoribosyltransferase (Nampt) transgenic mid-age) were imaged and compared to detect age-related redox differences. RESULTS: We observed that living muscle fiber and frozen and FFPE slides all had strong autofluorescence signals in the NADH and Fp channels. Paraformaldehyde fixation resulted in a significant increase in the redox ratio Fp/(NADH + Fp) of muscle fibers. Quantitative image analysis on FFPE unstained slides showed that mid-age gastrocnemius muscles had stronger NADH and Fp signals than young muscles. Gastrocnemius muscles from mid-age Nampt mice had lower NADH compared to age-matched controls, but had higher Fp than young controls. Soleus muscles had the same trend of change and appeared to be more oxidative than gastrocnemius muscles. Differential NADH and Fp signals were found between gastrocnemius and soleus muscles within both mid-aged control and Nampt groups. CONCLUSION: Aging effect on redox status quantified by ORI of FFPE unstained muscle slides was reported for the first time. Quantitative information from ORI of FFPE unstained slides may be useful for biomedical applications.


Subject(s)
Muscles/diagnostic imaging , Muscles/metabolism , Optical Imaging , Tissue Fixation , Animals , Flavoproteins/metabolism , Formaldehyde , Male , Mice, Inbred C57BL , Muscle Fibers, Skeletal/metabolism , NAD/metabolism , Oxidation-Reduction , Paraffin Embedding , Polymers , Staining and Labeling
11.
Radiat Res ; 169(6): 615-25, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18494549

ABSTRACT

Malignancy is considered to be a particular risk associated with exposure to the types of ionizing radiation encountered during extended space flight. In the present study, two dietary preparations were evaluated for their ability to prevent carcinogenesis in CBA mice exposed to different forms of space radiation: protons and highly energetic heavy particles (HZE particles). One preparation contained a mixture of antioxidant agents. The other contained the soybean-derived Bowman-Birk protease inhibitor (BBI), used in the form of BBI Concentrate (BBIC). The major finding was that there was a reduced risk of developing malignant lymphoma in animals exposed to space radiation and maintained on diets containing the antioxidant formulation or BBIC compared to the irradiated animals maintained on the control diet. In addition, the two different dietary countermeasures also reduced the yields of a variety of different rare tumor types observed in the animals exposed to space radiation. These results suggest that dietary supplements could be useful in the prevention of malignancies and other neoplastic lesions developing from exposure to space radiation.


Subject(s)
Antioxidants/metabolism , Ions , Iron/chemistry , Lymphoma/drug therapy , Lymphoma/metabolism , Protons , Animals , Antioxidants/pharmacology , Diet , Dietary Supplements , Hematopoietic Stem Cells/cytology , Male , Mice , Mice, Inbred CBA , Neoplasms, Radiation-Induced , Pilot Projects , Time Factors
12.
Diabetes ; 65(4): 927-41, 2016 04.
Article in English | MEDLINE | ID: mdl-26858361

ABSTRACT

Rapamycin extends life span in mice, yet paradoxically causes lipid dysregulation and glucose intolerance through mechanisms that remain incompletely understood. Whole-body energy balance can be influenced by beige/brite adipocytes, which are inducible by cold and other stimuli via ß-adrenergic signaling in white adipose depots. Induction of beige adipocytes is considered a promising strategy to combat obesity because of their ability to metabolize glucose and lipids, dissipating the resulting energy as heat through uncoupling protein 1. Here, we report that rapamycin blocks the ability of ß-adrenergic signaling to induce beige adipocytes and expression of thermogenic genes in white adipose depots. Rapamycin enhanced transcriptional negative feedback on the ß3-adrenergic receptor. However, thermogenic gene expression remained impaired even when the receptor was bypassed with a cell-permeable cAMP analog, revealing the existence of a second inhibitory mechanism. Accordingly, rapamycin-treated mice are cold intolerant, failing to maintain body temperature and weight when shifted to 4°C. Adipocyte-specific deletion of the mTORC1 subunit Raptor recapitulated the block in ß-adrenergic signaling. Our findings demonstrate a positive role for mTORC1 in the recruitment of beige adipocytes and suggest that inhibition of ß-adrenergic signaling by rapamycin may contribute to its physiological effects.


Subject(s)
Adipose Tissue, White/drug effects , Sirolimus/pharmacology , Thermogenesis/drug effects , Adipocytes/drug effects , Adipocytes/physiology , Adipose Tissue, White/metabolism , Animals , Dioxoles/pharmacology , Energy Metabolism/drug effects , Energy Metabolism/genetics , Female , Gene Expression Regulation/drug effects , Male , Mice , Mice, Inbred C57BL , Thermogenesis/genetics
13.
Cell Metab ; 24(2): 269-82, 2016 08 09.
Article in English | MEDLINE | ID: mdl-27508874

ABSTRACT

NAD is an obligate co-factor for the catabolism of metabolic fuels in all cell types. However, the availability of NAD in several tissues can become limited during genotoxic stress and the course of natural aging. The point at which NAD restriction imposes functional limitations on tissue physiology remains unknown. We examined this question in murine skeletal muscle by specifically depleting Nampt, an essential enzyme in the NAD salvage pathway. Knockout mice exhibited a dramatic 85% decline in intramuscular NAD content, accompanied by fiber degeneration and progressive loss of both muscle strength and treadmill endurance. Administration of the NAD precursor nicotinamide riboside rapidly ameliorated functional deficits and restored muscle mass despite having only a modest effect on the intramuscular NAD pool. Additionally, lifelong overexpression of Nampt preserved muscle NAD levels and exercise capacity in aged mice, supporting a critical role for tissue-autonomous NAD homeostasis in maintaining muscle mass and function.


Subject(s)
Homeostasis , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , NAD/metabolism , Administration, Oral , Aging/physiology , Animals , Biological Availability , Energy Metabolism , Glucose/metabolism , Inflammation/pathology , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/metabolism , Muscle Strength , Muscle, Skeletal/enzymology , Muscle, Skeletal/physiopathology , Necrosis , Niacinamide/administration & dosage , Niacinamide/analogs & derivatives , Niacinamide/metabolism , Niacinamide/pharmacology , Nicotinamide Phosphoribosyltransferase/deficiency , Nicotinamide Phosphoribosyltransferase/metabolism , Organ Size , Physical Conditioning, Animal , Pyridinium Compounds , Transcription, Genetic
14.
Neurosci Lett ; 380(1-2): 127-32, 2005.
Article in English | MEDLINE | ID: mdl-15854764

ABSTRACT

We recently reported findings of modest loss of cortical acetylcholinesterase (AChE) activity in patients with overall mild Alzheimer's disease (AD) using N-[11C]methyl-pi-peridin-4-yl propionate ([11C]PMP) AChE positron emission tomography (PET). To determine cognitive correlates of in vivo cortical AChE activity in patients with mild to moderate AD (n=15), and in normal controls (NC, n=12) using [11C]PMP AChE PET imaging. Mean cortical AChE activity in the AD subjects was mildly reduced (-11.1%) compared to the control subjects (P<0.05). Analysis of the cognitive data showed that mean cortical AChE activity was significantly associated with performance on a test of attention and working memory (WAIS-III Digit Span, R=0.46, P=0.01) but not with tests of delayed short or long-term memory functions. Similar findings were present when the analysis was limited to the temporal cortex. Cortical AChE activity is more robustly associated with functions of attention and working memory compared to performance on primary memory tests in AD.


Subject(s)
Acetylcholinesterase/metabolism , Alzheimer Disease/enzymology , Cerebral Cortex/metabolism , Cognition/physiology , Aged , Aged, 80 and over , Alzheimer Disease/physiopathology , Attention/physiology , Carbon Radioisotopes/pharmacokinetics , Case-Control Studies , Cerebral Cortex/drug effects , Cerebral Cortex/physiopathology , Female , Humans , Male , Memory, Short-Term/physiology , Mental Status Schedule , Neuropsychological Tests , Positron-Emission Tomography/methods , Pyrrolidines/pharmacokinetics , Statistics as Topic , Verbal Learning/physiology
16.
Arch Neurol ; 60(12): 1745-8, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14676050

ABSTRACT

BACKGROUND: Pathology reports have shown that cholinergic forebrain neuronal losses in parkinsonian dementia (PDem) are equal to or greater than those in Alzheimer disease (AD). We hypothesized that patients with PDem would have cholinergic deficits that were similar to or greater than those of patients with AD. OBJECTIVE: To determine in vivo cortical acetylcholinesterase (AChE) activity in healthy control subjects and in patients with mild AD, PDem, and Parkinson disease without dementia using AChE positron emission tomography. SETTING: University and Veterans' Administration medical center. Design and Patients Group comparison design of patients with AD (n = 12), PDem (n = 14), and Parkinson disease without dementia (n = 11), and controls (n = 10) who underwent AChE imaging between July 1, 2000, and January 31, 2003. Patients with AD and PDem had approximately equal dementia severity. MAIN OUTCOME MEASURES: Cerebral AChE activity. RESULTS: Compared with controls, mean cortical AChE activity was lowest in patients with PDem (-20.0%), followed by patients with Parkinson disease without dementia (-12.9%; P<.001). Mean cortical AChE activity was relatively preserved in patients with AD (-9.1%), except for regionally selective involvement of the lateral temporal cortex (-15%; P<.001). CONCLUSION: Reduced cortical AChE activity is more characteristic of patients with PDem than of patients with mild AD.


Subject(s)
Acetylcholinesterase/metabolism , Alzheimer Disease/enzymology , Cerebral Cortex/enzymology , Dementia/etiology , Parkinson Disease/complications , Parkinson Disease/enzymology , Aged , Alzheimer Disease/diagnostic imaging , Case-Control Studies , Cerebral Cortex/diagnostic imaging , Female , Humans , Male , Parkinson Disease/diagnostic imaging , Tomography, Emission-Computed
17.
DNA Cell Biol ; 21(7): 505-17, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12162805

ABSTRACT

As part of an effort to identify gene products that are differentially regulated during oligodendrocyte development, we isolated a mouse cDNA that encodes tGolgin-1, a homolog of the human protein known as golgin-245, trans-golgi p230, or 256 kD golgin. Human tGolgin-1 is the target of autoantibodies in patients with Sjögren's syndrome, and is thought to be involved in vesicular transport processes at the trans-Golgi network. Sequencing of cDNAs and EST clones comprising the full-length tGolgin-1 transcript predict marked homology with the amino- and carboxy-terminal regions of the human protein, but more limited homology within the central predicted coiled-coil region. Epitope tagged, truncated forms of mouse tGolgin-1, like those of its human homolog, were localized at steady state to the Golgi/trans-Golgi network in transfected cells. The tGolgin-1 message was expressed in all tissues examined, but was highly upregulated in oligodendrocyte precursors at a stage just prior to myelination. This expression pattern suggests that tGolgin-1 may play a role in specialized transport processes associated with maturation and/or differentiation of oligodendrocyte precursors.


Subject(s)
Autoantigens/genetics , Golgi Apparatus/metabolism , Membrane Proteins , Oligodendroglia/cytology , Oligodendroglia/metabolism , Amino Acid Sequence , Animals , Autoantigens/metabolism , Base Sequence , Cell Differentiation/genetics , DNA, Complementary/genetics , DNA, Complementary/isolation & purification , Gene Expression Regulation, Developmental , Glycolipids/metabolism , Golgi Matrix Proteins , HeLa Cells , Humans , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Proteolipids/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sequence Homology, Amino Acid , Species Specificity , Up-Regulation
18.
Aging Cell ; 13(1): 131-41, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24102714

ABSTRACT

Sleep disruption has detrimental effects on glucose metabolism through pathways that remain poorly defined. Although numerous studies have examined the consequences of sleep deprivation (SD) in the brain, few have directly tested its effects on peripheral organs. We examined several tissues in mice for induction of the unfolded protein response (UPR) following acute SD. In young animals, we found a robust induction of BiP in the pancreas, indicating an active UPR. At baseline, pancreata from aged animals exhibited a marked increase in a pro-apoptotic transcription factor, CHOP, that was amplified by SD, whereas BiP induction was not observed, suggesting a maladaptive response to cellular stress with age. Acute SD increased plasma glucose levels in both young and old animals. However, this change was not overtly related to stress in the pancreatic beta cells, as plasma insulin levels were not lower following acute SD. Accordingly, animals subjected to acute SD remained tolerant to a glucose challenge. In a chronic SD experiment, young mice were found to be sensitized to insulin and have improved glycemic control, whereas aged animals became hyperglycemic and failed to maintain appropriate plasma insulin concentrations. Our results show that both age and SD cooperate to induce the UPR in pancreatic tissue. While changes in insulin secretion are unlikely to play a major role in the acute effects of SD, CHOP induction in pancreatic tissues suggests that chronic SD may contribute to the loss or dysfunction of endocrine cells and that these effects may be exacerbated by normal aging.


Subject(s)
Aging/metabolism , Aging/pathology , Pancreas/metabolism , Pancreas/pathology , Sleep Deprivation/metabolism , Sleep Deprivation/pathology , Unfolded Protein Response , Aging/blood , Animals , Blood Glucose/metabolism , Central Nervous System/pathology , Corticosterone/blood , Food , Glucose Tolerance Test , Homeostasis , Insulin/blood , Male , Mice , Mice, Inbred C57BL , Sleep Deprivation/blood
19.
Aging (Albany NY) ; 5(7): 539-50, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23929887

ABSTRACT

Rapamycin extends lifespan in mice, but can have a number of undesirable effects that may ultimately limit its utility in humans. The canonical target of rapamycin, and the one thought to account for its effects on lifespan, is the mammalian/mechanistic target of rapamycin, complex 1 (mTORC1). We have previously shown that at least some of the detrimental side effects of rapamycin are due to "off target" disruption of mTORC2, suggesting they could be avoided by more specific targeting of mTORC1. However, mTORC1 inhibitionper se can reduce the mRNA expression of mitochondrial genes and compromise the function of mitochondria in cultured muscle cells, implying that defects in bioenergetics might be an unavoidable consequence of targeting mTORC1 in vivo. Therefore, we tested whether rapamycin, at the same doses used to extend lifespan, affects mitochondrial function in skeletal muscle. While mitochondrial transcripts were decreased, particularly in the highly oxidative soleus muscle, we found no consistent change in mitochondrial DNA or protein levels. In agreement with the lack of change in mitochondrial components, rapamycin-treated mice had endurance equivalent to that of untreated controls, and isolated, permeabilized muscle fibers displayed similar rates of oxygen consumption. We conclude that the doses of rapamycin required to extend life do not cause overt mitochondrial dysfunction in skeletal muscle.


Subject(s)
Immunosuppressive Agents/pharmacology , Mitochondria, Muscle/drug effects , Muscle, Skeletal/drug effects , Physical Endurance/drug effects , Sirolimus/pharmacology , Animals , Gene Expression Regulation/drug effects , Male , Mice , Mice, Inbred C57BL , Mitochondria, Muscle/physiology , Motor Activity/drug effects , Muscle, Skeletal/physiology , RNA, Messenger/genetics , RNA, Messenger/metabolism
20.
Science ; 335(6076): 1638-43, 2012 Mar 30.
Article in English | MEDLINE | ID: mdl-22461615

ABSTRACT

Rapamycin, an inhibitor of mechanistic target of rapamycin complex 1 (mTORC1), extends the life spans of yeast, flies, and mice. Calorie restriction, which increases life span and insulin sensitivity, is proposed to function by inhibition of mTORC1, yet paradoxically, chronic administration of rapamycin substantially impairs glucose tolerance and insulin action. We demonstrate that rapamycin disrupted a second mTOR complex, mTORC2, in vivo and that mTORC2 was required for the insulin-mediated suppression of hepatic gluconeogenesis. Further, decreased mTORC1 signaling was sufficient to extend life span independently from changes in glucose homeostasis, as female mice heterozygous for both mTOR and mLST8 exhibited decreased mTORC1 activity and extended life span but had normal glucose tolerance and insulin sensitivity. Thus, mTORC2 disruption is an important mediator of the effects of rapamycin in vivo.


Subject(s)
Insulin Resistance , Longevity , Sirolimus/pharmacology , Adipose Tissue, White/metabolism , Animals , Carrier Proteins/genetics , Carrier Proteins/metabolism , Female , Gluconeogenesis , Glucose/metabolism , Glucose Clamp Technique , Homeostasis , Insulin/administration & dosage , Insulin/blood , Liver/metabolism , Male , Mechanistic Target of Rapamycin Complex 1 , Mice , Mice, Inbred C57BL , Multiprotein Complexes , Muscle, Skeletal/metabolism , Phosphorylation , Proteins/antagonists & inhibitors , Proteins/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rapamycin-Insensitive Companion of mTOR Protein , Signal Transduction , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL