Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
1.
Biochem Biophys Res Commun ; 513(2): 486-493, 2019 05 28.
Article in English | MEDLINE | ID: mdl-30975470

ABSTRACT

Tissue nicotinamide adenine dinucleotide (NAD+) decline has been implicated in aging. We have recently identified CD38 as a central regulator involved in tissue NAD+ decline during the aging process. CD38 is an ecto-enzyme highly expressed in endothelial and inflammatory cells. To date, the mechanisms that regulate CD38 expression in aging tissues characterized by the presence of senescent cells is not completely understood. Cellular senescence has been described as a hallmark of the aging process and these cells are known to secrete several factors including cytokines and chemokines through their senescent associated secretory phenotype (SASP). Here we investigated if the cellular senescence phenotype is involved in the regulation of CD38 expression and its NADase activity. We observed that senescent cells do not have high expression of CD38. However, the SASP factors secreted by senescent cells induced CD38 mRNA and protein expression and increased CD38-NADase activity in non-senescent cells such as endothelial cells or bone marrow derived macrophages. Our data suggest a link between cellular senescence and NAD+ decline in which SASP-mediated upregulation of CD38 can disrupt cellular NAD+ homeostasis.


Subject(s)
ADP-ribosyl Cyclase 1/metabolism , Cellular Senescence , NAD/metabolism , ADP-ribosyl Cyclase 1/analysis , Aging , Animals , Cells, Cultured , Endothelial Cells/cytology , Endothelial Cells/metabolism , Female , Human Umbilical Vein Endothelial Cells , Humans , Macrophages/cytology , Macrophages/metabolism , Mice, Inbred C57BL , Middle Aged
2.
Drug Metab Dispos ; 44(11): 1742-1751, 2016 11.
Article in English | MEDLINE | ID: mdl-27531952

ABSTRACT

Overdose of isoniazid (INH), an antituberculosis drug, can be life-threatening because of neurotoxicity. In clinical practice for management of INH overdose and acute toxicity, the potential of INH-induced hepatotoxicity is also considered. However, the biochemical basis of acute INH toxicity in the liver remains elusive. In the current study, we used an untargeted metabolomic approach to explore the acute effects of INH on endobiotic homeostasis in mouse liver. We found that overdose of INH resulted in accumulation of oleoyl-l-carnitine and linoleoyl-l-carnitine in the liver, indicating mitochondrial dysfunction. We also revealed the interactions between INH and fatty acyl-CoAs by identifying INH-fatty acid amides. In addition, we found that overdose of INH led to the accumulation of heme and oxidized NAD in the liver. We also identified an INH and NAD adduct in the liver. In this adduct, the nicotinamide moiety in NAD was replaced by INH. Furthermore, we illustrated that overdose of INH depleted vitamin B6 in the liver and blocked vitamin B6-dependent cystathionine degradation. These data suggest that INH interacts with multiple biochemical pathways in the liver during acute poisoning caused by INH overdose.


Subject(s)
Antitubercular Agents/adverse effects , Antitubercular Agents/metabolism , Homeostasis/drug effects , Isoniazid/adverse effects , Isoniazid/metabolism , Liver/drug effects , Liver/metabolism , Animals , Carnitine/metabolism , Homeostasis/physiology , Metabolomics/methods , Mice , Oxidation-Reduction/drug effects , Vitamin B 6/metabolism
3.
Aging Cell ; 21(4): e13589, 2022 04.
Article in English | MEDLINE | ID: mdl-35263032

ABSTRACT

Nicotinamide adenine dinucleotide (NAD) levels decline during aging, contributing to physical and metabolic dysfunction. The NADase CD38 plays a key role in age-related NAD decline. Whether the inhibition of CD38 increases lifespan is not known. Here, we show that the CD38 inhibitor 78c increases lifespan and healthspan of naturally aged mice. In addition to a 10% increase in median survival, 78c improved exercise performance, endurance, and metabolic function in mice. The effects of 78c were different between sexes. Our study is the first to investigate the effect of CD38 inhibition in naturally aged animals.


Subject(s)
Longevity , NAD , ADP-ribosyl Cyclase 1/metabolism , Aging/metabolism , Animals , Mice , NAD/metabolism , NAD+ Nucleosidase/metabolism
4.
EMBO Mol Med ; 14(5): e12860, 2022 05 09.
Article in English | MEDLINE | ID: mdl-35298089

ABSTRACT

Duchenne muscular dystrophy (DMD) is characterized by progressive muscle degeneration. Two important deleterious features are a Ca2+ dysregulation linked to Ca2+ influxes associated with ryanodine receptor hyperactivation, and a muscular nicotinamide adenine dinucleotide (NAD+ ) deficit. Here, we identified that deletion in mdx mice of CD38, a NAD+ glycohydrolase-producing modulators of Ca2+ signaling, led to a fully restored heart function and structure, with skeletal muscle performance improvements, associated with a reduction in inflammation and senescence markers. Muscle NAD+ levels were also fully restored, while the levels of the two main products of CD38, nicotinamide and ADP-ribose, were reduced, in heart, diaphragm, and limb. In cardiomyocytes from mdx/CD38-/- mice, the pathological spontaneous Ca2+ activity was reduced, as well as in myotubes from DMD patients treated with isatuximab (SARCLISA® ) a monoclonal anti-CD38 antibody. Finally, treatment of mdx and utrophin-dystrophin-deficient (mdx/utr-/- ) mice with CD38 inhibitors resulted in improved skeletal muscle performances. Thus, we demonstrate that CD38 actively contributes to DMD physiopathology. We propose that a selective anti-CD38 therapeutic intervention could be highly relevant to develop for DMD patients.


Subject(s)
Muscular Dystrophy, Duchenne , ADP-ribosyl Cyclase 1 , Animals , Humans , Mice , Mice, Inbred mdx , Muscle, Skeletal , Muscular Dystrophy, Duchenne/genetics , Myocytes, Cardiac/pathology , NAD/genetics , NAD/therapeutic use , NAD+ Nucleosidase/genetics , Phenotype
5.
Cell Rep ; 36(5): 109481, 2021 08 03.
Article in English | MEDLINE | ID: mdl-34348145

ABSTRACT

Preadipocytes dynamically produce sensory cilia. However, the role of primary cilia in preadipocyte differentiation and adipose homeostasis remains poorly understood. We previously identified transition fiber component FBF1 as an essential player in controlling selective cilia import. Here, we establish Fbf1tm1a/tm1a mice and discover that Fbf1tm1a/tm1a mice develop severe obesity, but surprisingly, are not predisposed to adverse metabolic complications. Obese Fbf1tm1a/tm1a mice possess unexpectedly healthy white fat tissue characterized by spontaneous upregulated beiging, hyperplasia but not hypertrophy, and low inflammation along the lifetime. Mechanistically, FBF1 governs preadipocyte differentiation by constraining the beiging program through an AKAP9-dependent, cilia-regulated PKA signaling, while recruiting the BBS chaperonin to transition fibers to suppress the hedgehog signaling-dependent adipogenic program. Remarkably, obese Fbf1tm1a/tm1a mice further fed a high-fat diet are protected from diabetes and premature death. We reveal a central role for primary cilia in the fate determination of preadipocytes and the generation of metabolically healthy fat tissue.


Subject(s)
Adaptor Proteins, Signal Transducing/deficiency , Adipose Tissue, Beige/metabolism , Adipose Tissue, White/metabolism , 3T3-L1 Cells , Adaptor Proteins, Signal Transducing/metabolism , Adipocytes/metabolism , Adipogenesis , Animals , Cell Respiration , Cilia/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Female , Fibroblasts/metabolism , Hedgehog Proteins/metabolism , Homozygote , Humans , Hyperphagia/complications , Hyperphagia/pathology , Hyperplasia , Inflammation/pathology , Male , Metabolic Syndrome/complications , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microtubule-Associated Proteins/metabolism , Obesity/complications , Signal Transduction , Transcription Factors/metabolism
6.
Nat Metab ; 2(11): 1284-1304, 2020 11.
Article in English | MEDLINE | ID: mdl-33199925

ABSTRACT

Decreased NAD+ levels have been shown to contribute to metabolic dysfunction during aging. NAD+ decline can be partially prevented by knockout of the enzyme CD38. However, it is not known how CD38 is regulated during aging, and how its ecto-enzymatic activity impacts NAD+ homeostasis. Here we show that an increase in CD38 in white adipose tissue (WAT) and the liver during aging is mediated by accumulation of CD38+ immune cells. Inflammation increases CD38 and decreases NAD+. In addition, senescent cells and their secreted signals promote accumulation of CD38+ cells in WAT, and ablation of senescent cells or their secretory phenotype decreases CD38, partially reversing NAD+ decline. Finally, blocking the ecto-enzymatic activity of CD38 can increase NAD+ through a nicotinamide mononucleotide (NMN)-dependent process. Our findings demonstrate that senescence-induced inflammation promotes accumulation of CD38 in immune cells that, through its ecto-enzymatic activity, decreases levels of NMN and NAD+.


Subject(s)
ADP-ribosyl Cyclase 1/metabolism , Aging/metabolism , Membrane Glycoproteins/metabolism , NAD/biosynthesis , ADP-ribosyl Cyclase 1/genetics , ADP-ribosyl Cyclase 1/immunology , Adipocytes, White/metabolism , Adipose Tissue, White/metabolism , Aging/immunology , Animals , Bone Marrow Transplantation , Cellular Senescence , HEK293 Cells , Humans , Inflammation/immunology , Liver/growth & development , Liver/metabolism , Membrane Glycoproteins/genetics , Membrane Glycoproteins/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Nicotinamide Mononucleotide/metabolism , Phenotype
7.
Cell Metab ; 27(5): 1081-1095.e10, 2018 May 01.
Article in English | MEDLINE | ID: mdl-29719225

ABSTRACT

Aging is characterized by the development of metabolic dysfunction and frailty. Recent studies show that a reduction in nicotinamide adenine dinucleotide (NAD+) is a key factor for the development of age-associated metabolic decline. We recently demonstrated that the NADase CD38 has a central role in age-related NAD+ decline. Here we show that a highly potent and specific thiazoloquin(az)olin(on)e CD38 inhibitor, 78c, reverses age-related NAD+ decline and improves several physiological and metabolic parameters of aging, including glucose tolerance, muscle function, exercise capacity, and cardiac function in mouse models of natural and accelerated aging. The physiological effects of 78c depend on tissue NAD+ levels and were reversed by inhibition of NAD+ synthesis. 78c increased NAD+ levels, resulting in activation of pro-longevity and health span-related factors, including sirtuins, AMPK, and PARPs. Furthermore, in animals treated with 78c we observed inhibition of pathways that negatively affect health span, such as mTOR-S6K and ERK, and attenuation of telomere-associated DNA damage, a marker of cellular aging. Together, our results detail a novel pharmacological strategy for prevention and/or reversal of age-related NAD+ decline and subsequent metabolic dysfunction.


Subject(s)
ADP-ribosyl Cyclase 1/antagonists & inhibitors , Aging/drug effects , Cellular Senescence/drug effects , Enzyme Inhibitors/pharmacology , NAD/metabolism , Quinolines/pharmacology , Triazoles/pharmacology , AMP-Activated Protein Kinase Kinases , Aging/metabolism , Animals , DNA Damage/drug effects , Enzyme Inhibitors/chemistry , Glucose Intolerance/blood , Glucose Intolerance/drug therapy , Humans , MAP Kinase Signaling System/drug effects , Mice , Physical Functional Performance , Poly(ADP-ribose) Polymerases/metabolism , Protein Kinases/metabolism , Quinolines/chemistry , Sirtuins/metabolism , TOR Serine-Threonine Kinases/metabolism , Triazoles/chemistry
8.
Mol Cell Endocrinol ; 455: 62-74, 2017 Nov 05.
Article in English | MEDLINE | ID: mdl-27825999

ABSTRACT

Life as we know it cannot exist without the nucleotide nicotinamide adenine dinucleotide (NAD). From the simplest organism, such as bacteria, to the most complex multicellular organisms, NAD is a key cellular component. NAD is extremely abundant in most living cells and has traditionally been described to be a cofactor in electron transfer during oxidation-reduction reactions. In addition to participating in these reactions, NAD has also been shown to play a key role in cell signaling, regulating several pathways from intracellular calcium transients to the epigenetic status of chromatin. Thus, NAD is a molecule that provides an important link between signaling and metabolism, and serves as a key molecule in cellular metabolic sensoring pathways. Importantly, it has now been clearly demonstrated that cellular NAD levels decline during chronological aging. This decline appears to play a crucial role in the development of metabolic dysfunction and age-related diseases. In this review we will discuss the molecular mechanisms responsible for the decrease in NAD levels during aging. Since other reviews on this subject have been recently published, we will concentrate on presenting a critical appraisal of the current status of the literature and will highlight some controversial topics in the field. In particular, we will discuss the potential role of the NADase CD38 as a driver of age-related NAD decline.


Subject(s)
Aging/metabolism , Caloric Restriction , Mitochondria/metabolism , NAD/metabolism , Protein Processing, Post-Translational , ADP-ribosyl Cyclase 1/genetics , ADP-ribosyl Cyclase 1/metabolism , Aging/genetics , Animals , Armadillo Domain Proteins/genetics , Armadillo Domain Proteins/metabolism , Cyclic ADP-Ribose/metabolism , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Humans , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mitochondria/genetics , NADP/analogs & derivatives , NADP/metabolism , Oxidation-Reduction , Poly(ADP-ribose) Polymerases/genetics , Poly(ADP-ribose) Polymerases/metabolism , Signal Transduction , Sirtuins/genetics , Sirtuins/metabolism
9.
Cell Metab ; 23(6): 1127-1139, 2016 06 14.
Article in English | MEDLINE | ID: mdl-27304511

ABSTRACT

Nicotinamide adenine dinucleotide (NAD) levels decrease during aging and are involved in age-related metabolic decline. To date, the mechanism responsible for the age-related reduction in NAD has not been elucidated. Here we demonstrate that expression and activity of the NADase CD38 increase with aging and that CD38 is required for the age-related NAD decline and mitochondrial dysfunction via a pathway mediated at least in part by regulation of SIRT3 activity. We also identified CD38 as the main enzyme involved in the degradation of the NAD precursor nicotinamide mononucleotide (NMN) in vivo, indicating that CD38 has a key role in the modulation of NAD-replacement therapy for aging and metabolic diseases.


Subject(s)
ADP-ribosyl Cyclase 1/metabolism , Aging/metabolism , Mitochondria/metabolism , NAD/metabolism , Sirtuin 3/metabolism , Animals , Diet, High-Fat , Mammals/metabolism , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/ultrastructure , NAD+ Nucleosidase/genetics , NAD+ Nucleosidase/metabolism , Niacinamide/analogs & derivatives , Niacinamide/metabolism , Organ Specificity , Pyridinium Compounds , RNA, Messenger/genetics , RNA, Messenger/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL