Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 54
Filter
Add more filters










Publication year range
1.
Nutrients ; 13(5)2021 May 14.
Article in English | MEDLINE | ID: mdl-34068917

ABSTRACT

Nicotinamide adenine dinucleotide (NAD) is an essential redox cofactor, but it also acts as a substrate for NAD-consuming enzymes, regulating cellular events such as DNA repair and gene expression. Since such processes are fundamental to support cancer cell survival and proliferation, sustained NAD production is a hallmark of many types of neoplasms. Depleting intratumor NAD levels, mainly through interference with the NAD-biosynthetic machinery, has emerged as a promising anti-cancer strategy. NAD can be generated from tryptophan or nicotinic acid. In addition, the "salvage pathway" of NAD production, which uses nicotinamide, a byproduct of NAD degradation, as a substrate, is also widely active in mammalian cells and appears to be highly exploited by a subset of human cancers. In fact, research has mainly focused on inhibiting the key enzyme of the latter NAD production route, nicotinamide phosphoribosyltransferase (NAMPT), leading to the identification of numerous inhibitors, including FK866 and CHS-828. Unfortunately, the clinical activity of these agents proved limited, suggesting that the approaches for targeting NAD production in tumors need to be refined. In this contribution, we highlight the recent advancements in this field, including an overview of the NAD-lowering compounds that have been reported so far and the related in vitro and in vivo studies. We also describe the key NAD-producing pathways and their regulation in cancer cells. Finally, we summarize the approaches that have been explored to optimize the therapeutic response to NAMPT inhibitors in cancer.


Subject(s)
Antineoplastic Agents/pharmacology , NAD/biosynthesis , NAD/drug effects , Neoplasms/drug therapy , Animals , Biosynthetic Pathways , Cell Death , Cell Line, Tumor , Cell Survival , Cytokines , DNA Damage , DNA Repair , Humans , Niacin/pharmacology , Niacinamide/pharmacology , Nicotinamide Phosphoribosyltransferase , Oxidative Stress
2.
Aging (Albany NY) ; 13(9): 12308-12333, 2021 04 26.
Article in English | MEDLINE | ID: mdl-33901008

ABSTRACT

UV radiation is one of the main contributors to skin photoaging by promoting the accumulation of cellular senescence, which in turn induces a proinflammatory and tissue-degrading state that favors skin aging. The members of the sirtuin family of NAD+-dependent enzymes play an anti-senescence role and their activation suggests a promising approach for preventing UV-induced senescence in the treatment of skin aging. A two-step screening designed to identify compounds able to protect cells from UV-induced senescence through sirtuin activation identified shikimic acid (SA), a metabolic intermediate in many organisms, as a bona-fide candidate. The protective effects of SA against senescence were dependent on specific activation of SIRT1 as the effect was abrogated by the SIRT1 inhibitor EX-527. Upon UV irradiation SA induced S-phase accumulation and a decrease in p16INK4A expression but did not protect against DNA damage or increased polyploidies. In contrast, SA reverted misfolded protein accumulation upon senescence, an effect that was abrogated by EX-527. Consistently, SA induced an increase in the levels of the chaperone BiP, resulting in a downregulation of unfolded protein response (UPR) signaling and UPR-dependent autophagy, avoiding their abnormal hyperactivation during senescence. SA did not directly activate SIRT1 in vitro, suggesting that SIRT1 is a downstream effector of SA signaling specifically in the response to cellular senescence. Our study not only uncovers a shikimic acid/SIRT1 signaling pathway that prevents cellular senescence, but also reinforces the role of sirtuins as key regulators of cell proteostasis.


Subject(s)
NAD/drug effects , Shikimic Acid/pharmacology , Sirtuin 1/drug effects , Skin Aging/drug effects , Cell Proliferation/drug effects , Cellular Senescence/physiology , Humans , NAD/metabolism , Oxidative Stress/drug effects , Protective Agents/pharmacology , Signal Transduction/drug effects , Sirtuin 1/metabolism , Skin/drug effects , Skin/metabolism , Ultraviolet Rays/adverse effects
3.
Aging (Albany NY) ; 12(24): 24504-24521, 2020 12 29.
Article in English | MEDLINE | ID: mdl-33373320

ABSTRACT

Currently, no pharmacotherapy has been proven effective in treating photoreceptor degeneration in patients. Discovering readily available and safe neuroprotectants is therefore highly sought after. Here, we investigated nicotinamide mononucleotide (NMN), a precursor of nicotinamide adenine dinucleotide (NAD+), in a retinal detachment (RD) induced photoreceptor degeneration. NMN administration after RD resulted in a significant reduction of TUNEL+ photoreceptors, CD11b+ macrophages, and GFAP labeled glial activation; a normalization of protein carbonyl content (PCC), and a preservation of the outer nuclear layer (ONL) thickness. NMN administration significantly increased NAD+ levels, SIRT1 protein expression, and heme oxygenase-1 (HO-1) expression. Delayed NMN administration still exerted protective effects after RD. Mechanistic in vitro studies using 661W cells revealed a SIRT1/HO-1 signaling as a downstream effector of NMN-mediated protection under oxidative stress and LPS stimulation. In conclusion, NMN administration exerts neuroprotective effects on photoreceptors after RD and oxidative injury, suggesting a therapeutic avenue to treating photoreceptor degeneration.


Subject(s)
Neuroprotective Agents/pharmacology , Nicotinamide Mononucleotide/pharmacology , Oxidative Stress/drug effects , Photoreceptor Cells, Vertebrate/drug effects , Retinal Degeneration/metabolism , Animals , Apoptosis/drug effects , CD11b Antigen/metabolism , Cell Line , Glial Fibrillary Acidic Protein/drug effects , Glial Fibrillary Acidic Protein/metabolism , Heme Oxygenase-1/drug effects , Heme Oxygenase-1/metabolism , In Situ Nick-End Labeling , Lipopolysaccharides/pharmacology , Macrophages/drug effects , Membrane Proteins/drug effects , Membrane Proteins/metabolism , Mice , NAD/drug effects , NAD/metabolism , Photoreceptor Cells, Vertebrate/metabolism , Photoreceptor Cells, Vertebrate/pathology , Protein Carbonylation/drug effects , Retinal Degeneration/etiology , Retinal Degeneration/pathology , Retinal Detachment/complications , Sirtuin 1/drug effects , Sirtuin 1/metabolism
4.
Cancer Res ; 80(22): 5024-5034, 2020 11 15.
Article in English | MEDLINE | ID: mdl-32998997

ABSTRACT

The aggressive primary brain tumor glioblastoma (GBM) is characterized by aberrant metabolism that fuels its malignant phenotype. Diverse genetic subtypes of malignant glioma are sensitive to selective inhibition of the NAD+ salvage pathway enzyme nicotinamide phosphoribosyltransferase (NAMPT). However, the potential impact of NAD+ depletion on the brain tumor microenvironment has not been elaborated. In addition, systemic toxicity of NAMPT inhibition remains a significant concern. Here we show that microparticle-mediated intratumoral delivery of NAMPT inhibitor GMX1778 induces specific immunologic changes in the tumor microenvironment of murine GBM, characterized by upregulation of immune checkpoint PD-L1, recruitment of CD3+, CD4+, and CD8+ T cells, and reduction of M2-polarized immunosuppressive macrophages. NAD+ depletion and autophagy induced by NAMPT inhibitors mediated the upregulation of PD-L1 transcripts and cell surface protein levels in GBM cells. NAMPT inhibitor modulation of the tumor immune microenvironment was therefore combined with PD-1 checkpoint blockade in vivo, significantly increasing the survival of GBM-bearing animals. Thus, the therapeutic impacts of NAMPT inhibition extended beyond neoplastic cells, shaping surrounding immune effectors. Microparticle delivery and release of NAMPT inhibitor at the tumor site offers a safe and robust means to alter an immune tumor microenvironment that could potentiate checkpoint immunotherapy for glioblastoma. SIGNIFICANCE: Microparticle-mediated local inhibition of NAMPT modulates the tumor immune microenvironment and acts cooperatively with anti-PD-1 checkpoint blockade, offering a combination immunotherapy strategy for the treatment of GBM.


Subject(s)
B7-H1 Antigen/metabolism , Brain Neoplasms/therapy , Cyanides/administration & dosage , Cytokines/antagonists & inhibitors , Glioblastoma/therapy , Guanidines/administration & dosage , NAD/drug effects , Nicotinamide Phosphoribosyltransferase/antagonists & inhibitors , Tumor Microenvironment/drug effects , Acrylamides/administration & dosage , Animals , Autophagy , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/genetics , Brain Neoplasms/immunology , Brain Neoplasms/metabolism , Brain Neoplasms/mortality , CD4-Positive T-Lymphocytes , CD8-Positive T-Lymphocytes , Cell Movement , Cyanides/adverse effects , Delayed-Action Preparations , Drug Carriers/chemical synthesis , Glioblastoma/immunology , Glioblastoma/metabolism , Glioblastoma/mortality , Guanidines/adverse effects , Humans , Injections, Intralesional , Macrophages/drug effects , Membrane Proteins/metabolism , Mice , NAD/analysis , NAD/deficiency , Piperidines/administration & dosage , Polymers/chemical synthesis , RNA, Messenger/metabolism , Signal Transduction , Tumor Microenvironment/immunology , Up-Regulation/drug effects
5.
Am J Physiol Cell Physiol ; 318(4): C796-C805, 2020 04 01.
Article in English | MEDLINE | ID: mdl-32049549

ABSTRACT

Excessive exposure of the eye to ultraviolet B light (UVB) leads to corneal edema and opacification because of the apoptosis of the corneal endothelium. Our previous study found that nicotinamide (NIC), the precursor of nicotinamide adenine dinucleotide (NAD), could inhibit the endothelial-mesenchymal transition and accelerate healing the wound to the corneal endothelium in the rabbit. Here we hypothesize that NIC may possess the capacity to protect the cornea from UVB-induced endothelial apoptosis. Therefore, a mouse model and a cultured cell model were used to examine the effect of NAD+ precursors, including NIC, nicotinamide mononucleotide (NMN), and NAD, on the UVB-induced apoptosis of corneal endothelial cells (CECs). The results showed that UVB irradiation caused apparent corneal edema and cell apoptosis in mice, accompanied by reduced levels of NAD+ and its key biosynthesis enzyme, nicotinamide phosphoribosyltransferase (NAMPT), in the corneal endothelium. However, the subconjunctival injection of NIC, NMN, or NAD+ effectively prevented UVB-induced tissue damage and endothelial cell apoptosis in the mouse cornea. Moreover, pretreatment using NIC, NMN, and NAD+ increased the survival rate and inhibited the apoptosis of cultured human CECs irradiated by UVB. Mechanistically, pretreatment using nicotinamide (NIC) recovered the AKT activation level and decreased the BAX/BCL-2 ratio. In addition, the capacity of NIC to protect CECs was fully reversed in the presence of the AKT inhibitor LY294002. Therefore, we conclude that NAD+ precursors can effectively prevent the apoptosis of the corneal endothelium through reactivating AKT signaling; this represents a potential therapeutic approach for preventing UVB-induced corneal damage.


Subject(s)
Apoptosis/drug effects , Endothelial Cells/drug effects , NAD/drug effects , Nicotinamide Mononucleotide/pharmacology , Animals , Cell Death/drug effects , Cells, Cultured , Endothelial Cells/metabolism , Endothelium, Corneal/drug effects , Endothelium, Corneal/metabolism , Humans , Mice , NAD/metabolism , Protective Agents/pharmacology , Rabbits
6.
Curr Opin HIV AIDS ; 15(2): 101-109, 2020 03.
Article in English | MEDLINE | ID: mdl-31789674

ABSTRACT

PURPOSE OF REVIEW: Some older people living with HIV (PLWH) exhibit features of unsuccessful ageing, such as frailty. Mitochondrial dysfunction is one of the best characterized ageing mechanisms. There has been recent interest in whether some people ageing with HIV may have an excess of mitochondrial dysfunction. This review aims to address this question through: analogy with ageing and chronic disease; discussion of the key unknowns; suggested ways that measures of mitochondrial dysfunction might be incorporated into HIV research studies. RECENT FINDINGS: Recent data suggest that mitochondrial dysfunction in PLWH may not be wholly a legacy effect of historical nucleoside analog reverse transcriptase inhibitor exposures. Research in the non-HIV setting has altered our understanding of the important mediators of mitochondrial dysfunction in ageing. SUMMARY: Mitochondrial dysfunction is a very plausible driver of adverse ageing phenotypes in some older PLWH. As such it may be a target for therapeutic interventions. Currently, however, there remain considerable uncertainties around the extent of this phenomenon, and its relative importance. Current studies are likely to clarify these questions over the next few years.


Subject(s)
Aging , DNA, Mitochondrial , HIV Infections , Mitochondria , Reverse Transcriptase Inhibitors/pharmacology , Aged , Aged, 80 and over , Aging/pathology , Anti-HIV Agents/pharmacology , Biomarkers , Chronic Disease , DNA, Mitochondrial/drug effects , DNA, Mitochondrial/metabolism , Flow Cytometry/methods , Frailty/pathology , HIV Infections/drug therapy , HIV Infections/pathology , Humans , Magnetic Resonance Spectroscopy/methods , Mitochondria/drug effects , Mitochondria/pathology , NAD/drug effects , NAD/metabolism , Oxidative Phosphorylation/drug effects , Reactive Oxygen Species/metabolism , Spectroscopy, Near-Infrared/methods
7.
J Lipid Res ; 60(4): 741-746, 2019 04.
Article in English | MEDLINE | ID: mdl-30782960

ABSTRACT

Niacin, the first antidyslipidemic drug, has been at the center stage of lipid research for many decades before the discovery of statins. However, to date, despite its remarkable effects on lipid profiles, the clinical outcomes of niacin treatment on cardiac events is still debated. In addition to its historically well-defined interactions with central players of lipid metabolism, niacin can be processed by eukaryotic cells to synthesize a crucial cofactor, NAD+ NAD+ acts as a cofactor in key cellular processes, including oxidative phosphorylation, glycolysis, and DNA repair. More recently, evidence has emerged that NAD+ also is an essential cosubstrate for the sirtuin family of protein deacylases and thereby has an impact on a wide range of cellular processes, most notably mitochondrial homeostasis, energy homeostasis, and lipid metabolism. NAD+ achieves these remarkable effects through sirtuin-mediated deacetylation of key transcriptional regulators, such as peroxisome proliferator-activated receptor gamma coactivator 1-α, LXR, and SREBPs, that control these cellular processes. Here, we present an alternative point of view to explain niacin's mechanism of action, with a strong focus on the importance of how this old drug acts as a control switch of NAD+/sirtuin-mediated control of metabolism.


Subject(s)
Hypolipidemic Agents/pharmacology , NAD/drug effects , Niacin/pharmacology , Animals , Humans , Hypolipidemic Agents/chemistry , Lipid Metabolism/drug effects , Molecular Structure , NAD/metabolism , Niacin/chemistry
8.
Neurochem Res ; 44(10): 2280-2287, 2019 Oct.
Article in English | MEDLINE | ID: mdl-30661231

ABSTRACT

Dysfunctions in NAD+ metabolism are associated with neurodegenerative diseases, acute brain injury, diabetes, and aging. Loss of NAD+ levels results in impairment of mitochondria function, which leads to failure of essential metabolic processes. Strategies to replenish depleted NAD+ pools can offer significant improvements of pathologic states. NAD+ levels are maintained by two opposing enzymatic reactions, one is the consumption of NAD+ while the other is the re-synthesis of NAD+. Inhibition of NAD+ degrading enzymes, poly-ADP-ribose polymerase 1 (PARP1) and ectoenzyme CD38, following brain ischemic insult can provide neuroprotection. Preservation of NAD+ pools by administration of NAD+ precursors, such as nicotinamide (Nam) or nicotinamide mononucleotide (NMN), also offers neuroprotection. However, NMN treatment demonstrates to be a promising candidate as a therapeutic approach due to its multi-targeted effect acting as PARP1 and CD38 inhibitor, sirtuins activator, mitochondrial fission inhibitor, and NAD+ supplement. Many neurodegenerative diseases or acute brain injury activate several cellular death pathways requiring a treatment strategy that will target these mechanisms. Since NMN demonstrated the ability to exert its effect on several cellular metabolic pathways involved in brain pathophysiology it seems to be one of the most promising candidates to be used for successful neuroprotection.


Subject(s)
Brain/drug effects , Hydrolases/drug effects , Mitochondria/drug effects , Nicotinamide Mononucleotide/pharmacology , Animals , Brain/metabolism , Humans , Hydrolases/metabolism , Mitochondria/metabolism , NAD/drug effects , NAD/metabolism , Neurodegenerative Diseases/drug therapy , Neurodegenerative Diseases/metabolism , Niacinamide/metabolism , Niacinamide/pharmacology , Nicotinamide Mononucleotide/metabolism
9.
Cytometry A ; 95(1): 110-121, 2019 01.
Article in English | MEDLINE | ID: mdl-30604477

ABSTRACT

Redox changes in live HeLa cervical cancer cells after doxorubicin treatment can either be analyzed by a novel fluorescence lifetime microscopy (FLIM)-based redox ratio NAD(P)H-a2%/FAD-a1%, called fluorescence lifetime redox ratio or one of its components (NAD(P)H-a2%), which is actually driving that ratio and offering a simpler and alternative metric and are both compared. Auto-fluorescent NAD(P)H, FAD lifetime is acquired by 2- photon excitation and Tryptophan by 3-photon, at 4 time points after treatment up to 60 min demonstrating early drug response to doxorubicin. Identical Fields-of-view (FoV) at each interval allows single-cell analysis, showing heterogeneous responses to treatment, largely based on their initial control redox state. Based on a discrete ROI selection method, mitochondrial OXPHOS and cytosolic glycolysis are discriminated. Furthermore, putative FRET interaction and energy transfer between tryptophan residue carrying enzymes and NAD(P)H correlate with NAD(P)H-a2%, as does the NADPH/NADH ratio, highlighting a multi-parametric assay to track metabolic changes in live specimens. © 2019 International Society for Advancement of Cytometry.


Subject(s)
Mitochondria/metabolism , NADP/analysis , NAD/analysis , Tryptophan/chemistry , Cytosol/drug effects , Cytosol/metabolism , Doxorubicin/pharmacology , Energy Metabolism/drug effects , Flavin-Adenine Dinucleotide/analysis , Fluorescence , Fluorescence Resonance Energy Transfer/methods , HeLa Cells , Humans , Microscopy, Fluorescence, Multiphoton/methods , Mitochondria/drug effects , NAD/drug effects , NADP/drug effects , Optical Imaging , Oxidation-Reduction , Oxidative Phosphorylation/drug effects , Reactive Oxygen Species/metabolism , Single-Cell Analysis/methods
10.
Microb Cell Fact ; 17(1): 10, 2018 Jan 22.
Article in English | MEDLINE | ID: mdl-29357933

ABSTRACT

BACKGROUND: Azotobacter vinelandii is a bacterium that produces alginate and polyhydroxybutyrate (P3HB); however, the role of NAD(P)H/NAD(P)+ ratios on the metabolic fluxes through biosynthesis pathways of these biopolymers remains unknown. The aim of this study was to evaluate the NAD(P)H/NAD(P) + ratios and the metabolic fluxes involved in alginate and P3HB biosynthesis, under oxygen-limiting and non-limiting oxygen conditions. RESULTS: The results reveal that changes in the oxygen availability have an important effect on the metabolic fluxes and intracellular NADPH/NADP+ ratio, showing that at the lowest OTR (2.4 mmol L-1 h-1), the flux through the tricarboxylic acid (TCA) cycle decreased 27.6-fold, but the flux through the P3HB biosynthesis increased 6.6-fold in contrast to the cultures without oxygen limitation (OTR = 14.6 mmol L-1 h-1). This was consistent with the increase in the level of transcription of phbB and the P3HB biosynthesis. In addition, under conditions without oxygen limitation, there was an increase in the carbon uptake rate (twofold), as well as in the flux through the pentose phosphate (PP) pathway (4.8-fold), compared to the condition of 2.4 mmol L-1 h-1. At the highest OTR condition, a decrease in the NADPH/NADP+ ratio of threefold was observed, probably as a response to the high respiration rate induced by the respiratory protection of the nitrogenase under diazotrophic conditions, correlating with a high expression of the uncoupled respiratory chain genes (ndhII and cydA) and induction of the expression of the genes encoding the nitrogenase complex (nifH). CONCLUSIONS: We have demonstrated that changes in oxygen availability affect the internal redox state of the cell and carbon metabolic fluxes. This also has a strong impact on the TCA cycle and PP pathway as well as on alginate and P3HB biosynthetic fluxes.


Subject(s)
Azotobacter vinelandii/metabolism , Metabolic Flux Analysis , NADP/analysis , NAD/analysis , Oxygen/metabolism , Alginates/metabolism , Biomass , Biosynthetic Pathways/drug effects , Carbon/metabolism , Citric Acid Cycle/drug effects , Culture Media/chemistry , NAD/drug effects , NAD/metabolism , NADP/drug effects , NADP/metabolism , Oxidation-Reduction , Oxygen/pharmacology , Pentose Phosphate Pathway/drug effects
11.
Neurobiol Dis ; 95: 102-10, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27425894

ABSTRACT

Nicotinamide adenine dinucleotide (NAD(+)) is an essential cofactor for multiple cellular metabolic reactions and has a central role in energy production. Brain ischemia depletes NAD(+) pools leading to bioenergetics failure and cell death. Nicotinamide mononucleotide (NMN) is utilized by the NAD(+) salvage pathway enzyme, nicotinamide adenylyltransferase (Nmnat) to generate NAD(+). Therefore, we examined whether NMN could protect against ischemic brain damage. Mice were subjected to transient forebrain ischemia and treated with NMN or vehicle at the start of reperfusion or 30min after the ischemic insult. At 2, 4, and 24h of recovery, the proteins poly-ADP-ribosylation (PAR), hippocampal NAD(+) levels, and expression levels of NAD(+) salvage pathway enzymes were determined. Furthermore, animal's neurologic outcome and hippocampal CA1 neuronal death was assessed after six days of reperfusion. NMN (62.5mg/kg) dramatically ameliorated the hippocampal CA1 injury and significantly improved the neurological outcome. Additionally, the post-ischemic NMN treatment prevented the increase in PAR formation and NAD(+) catabolism. Since the NMN administration did not affect animal's temperature, blood gases or regional cerebral blood flow during recovery, the protective effect was not a result of altered reperfusion conditions. These data suggest that administration of NMN at a proper dosage has a strong protective effect against ischemic brain injury.


Subject(s)
Brain Injuries/drug therapy , Brain Ischemia/drug therapy , NAD/drug effects , Nicotinamide Mononucleotide/pharmacology , Animals , Brain Injuries/complications , Brain Injuries/metabolism , Brain Ischemia/etiology , Brain Ischemia/metabolism , Cell Death/drug effects , Disease Models, Animal , Hippocampus/drug effects , Hippocampus/metabolism , Male , Mice, Inbred C57BL , NAD/metabolism , Niacinamide/metabolism
12.
Nutrition ; 32(6): 702-8, 2016 Jun.
Article in English | MEDLINE | ID: mdl-26899162

ABSTRACT

OBJECTIVE: Previous studies suggest that low vitamin D status is associated with obesity characterized by excess lipid storage in adipocytes. The aim of the present study was to determine the effects of the most hormonally active form of vitamin D 1,25-dihydroxyvitamin D [1,25(OH)2D] on adipocyte fat storage and lipid metabolism in mature 3T3-L1 cells. METHODS: Differentiated 3T3-L1 cells were treated with various concentrations of 1,25(OH)2D. 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide cell proliferation, intracellular lipid content, and basal and isoproterenol-stimulated lipolysis were measured to investigate the regulatory role of 1,25(OH)2D in adipocyte lipid metabolism. Reverse transcription polymerase chain reaction was performed to determine the effects of 1,25(OH)2D on adipogenesis-related markers, fatty acid oxidation-associated genes, and lipolytic enzymes. Sirtulin 1 (SIRT1) activity, nicotinamide adenine dinucleotide (NAD) and NADH were measured. RESULTS: 1,25(OH)2D treatment (24 h, 100 nmol/L) induced a decrease in intracellular fat accumulation and an increase of basal and isoproterenol-stimulated lipolysis without cell toxicity in adipocytes. Adipogenic gene levels were decreased. In contrast, mRNA levels of ß-oxidation-related genes, lipolytic enzymes, and vitamin D responsive gene were elevated by 1,25(OH)2D. Additionally, significant incremental changes in NAD levels, the ratio of NAD to NADH, and SIRT1 expression and activity were noted in 1,25(OH)2D-treated 3T3-L1 adipocytes. CONCLUSIONS: The observed potent inhibitory effect of 1,25(OH)2D on adipocyte fat storage in mature 3T3-L1 cells suggests that vitamin D might improve adipocyte metabolic function and protect against obesity. Increased NAD concentrations and SIRT1 ​activity may play a role in the mechanism of 1,25(OH)2D action.


Subject(s)
3T3-L1 Cells/drug effects , Adipocytes/drug effects , Lipid Metabolism/drug effects , Lipolysis/drug effects , Sirtuin 1/drug effects , Vitamin D/analogs & derivatives , 3T3-L1 Cells/metabolism , Adipocytes/metabolism , Adipogenesis/drug effects , Animals , Cell Culture Techniques , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Gene Expression/genetics , Humans , Mice , NAD/drug effects , NAD/genetics , NAD/metabolism , Oxidation-Reduction/drug effects , Polymerase Chain Reaction , Sirtuin 1/genetics , Sirtuin 1/metabolism , Vitamin D/pharmacology
13.
Hepatology ; 63(4): 1190-204, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26404765

ABSTRACT

UNLABELLED: With no approved pharmacological treatment, nonalcoholic fatty liver disease (NAFLD) is now the most common cause of chronic liver disease in Western countries and its worldwide prevalence continues to increase along with the growing obesity epidemic. Here, we show that a high-fat high-sucrose (HFHS) diet, eliciting chronic hepatosteatosis resembling human fatty liver, lowers hepatic nicotinamide adenine dinucleotide (NAD(+) ) levels driving reductions in hepatic mitochondrial content, function, and adenosine triphosphate (ATP) levels, in conjunction with robust increases in hepatic weight, lipid content, and peroxidation in C57BL/6J mice. To assess the effect of NAD(+) repletion on the development of steatosis in mice, nicotinamide riboside, a precursor of NAD(+) biosynthesis, was added to the HFHS diet, either as a preventive strategy or as a therapeutic intervention. We demonstrate that NR prevents and reverts NAFLD by inducing a sirtuin (SIRT)1- and SIRT3-dependent mitochondrial unfolded protein response, triggering an adaptive mitohormetic pathway to increase hepatic ß-oxidation and mitochondrial complex content and activity. The cell-autonomous beneficial component of NR treatment was revealed in liver-specific Sirt1 knockout mice (Sirt1(hep-/-) ), whereas apolipoprotein E-deficient mice (Apoe(-/-) ) challenged with a high-fat high-cholesterol diet affirmed the use of NR in other independent models of NAFLD. CONCLUSION: Our data warrant the future evaluation of NAD(+) boosting strategies to manage the development or progression of NAFLD.


Subject(s)
Fatty Liver/drug therapy , Fatty Liver/pathology , NAD/metabolism , Niacinamide/analogs & derivatives , Unfolded Protein Response/drug effects , Analysis of Variance , Animals , Area Under Curve , Biopsy, Needle , Diet, High-Fat/methods , Disease Models, Animal , Fatty Liver/metabolism , Immunohistochemistry , Lipid Metabolism/drug effects , Male , Mice , Mice, Inbred C57BL , Mitochondria/drug effects , Mitochondria/metabolism , NAD/drug effects , Niacinamide/pharmacology , Pyridinium Compounds , Random Allocation , Sensitivity and Specificity , Treatment Outcome
14.
Free Radic Biol Med ; 89: 750-7, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26454079

ABSTRACT

3-Nitrotyrosine (3NT) in liver proteins of mice treated with hepatotoxic doses of acetaminophen (APAP) has been postulated to be causative in toxicity. Nitration is by a reactive nitrogen species formed from nitric oxide (NO). The source of the NO is unclear. iNOS knockout mice were previously found to be equally susceptible to APAP toxicity as wildtype mice and iNOS inhibitors did not decrease toxicity in mice or in hepatocytes. In this work we examined the potential role of nNOS in APAP toxicity in hepatocytes using the specific nNOS inhibitor NANT (10 µM)(N-[(4S)-4-amino-5-[(2-aminoethyl)amino]pentyl]-N'-nitroguanidinetris (trifluoroacetate)). Primary hepatocytes (1 million/ml) from male B6C3F1 mice were incubated with APAP (1mM). Cells were removed and assayed spectrofluorometrically for reactive nitrogen and oxygen species using diaminofluorescein (DAF) and Mitosox red, respectively. Cytotoxicity was determined by LDH release into media. Glutathione (GSH, GSSG), 3NT, GSNO, acetaminophen-cysteine adducts, NAD, and NADH were measured by HPLC. APAP significantly increased cytotoxicity at 1.5-3.0 h. The increase was blocked by NANT. NANT did not alter APAP mediated GSH depletion or acetaminophen-cysteine adducts in proteins which indicated that NANT did not inhibit metabolism. APAP significantly increased spectroflurometric evidence of reactive nitrogen and oxygen formation at 0.5 and 1.0 h, respectively, and increased 3NT and GSNO at 1.5-3.0 h. These increases were blocked by NANT. APAP dramatically increased NADH from 0.5-3.0 h and this increase was blocked by NANT. Also, APAP decreased the Oxygen Consumption Rate (OCR), decreased ATP production, and caused a loss of mitochondrial membrane potential, which were all blocked by NANT.


Subject(s)
Acetaminophen/toxicity , Analgesics, Non-Narcotic/toxicity , Chemical and Drug Induced Liver Injury/metabolism , Enzyme Inhibitors/pharmacology , Guanidines/pharmacology , Hepatocytes/drug effects , Animals , Chromatography, High Pressure Liquid , Male , Membrane Potential, Mitochondrial/drug effects , Mice , Mitochondria, Liver/drug effects , Mitochondria, Liver/metabolism , NAD/drug effects , NAD/metabolism , Nitric Oxide Synthase Type I/antagonists & inhibitors , Oxidative Stress/drug effects , Oxygen Consumption/drug effects , Reactive Nitrogen Species/metabolism , Reactive Oxygen Species/metabolism
15.
Am J Physiol Heart Circ Physiol ; 309(9): H1543-53, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26342067

ABSTRACT

Ventricular fibrillation (VF) is an important cause of sudden cardiac arrest following myocardial infarction. Following resuscitation from VF, decreased cardiac contractile function is a common problem. During and following myocardial ischemia, decreased glucose oxidation, increased anaerobic glycolysis for cardiac energy production are harmful and energetically expensive. The objective of the present study is to determine the effects of dichloroacetate (DCA), a glucose oxidation stimulator, on cardiac contractile dysfunction following ischemia-induced VF. Male Sprague-Dawley rat hearts were Langendorff perfused in Tyrode's buffer. Once stabilized, hearts were subjected to 15 min of global ischemia and 5 min of aerobic reperfusion in the presence or absence of DCA. At the 6th min of reperfusion, VF was induced electrically, and terminated. Left ventricular (LV) pressure was measured using a balloon. Pretreatment with DCA significantly improved post-VF left ventricular developed pressure (LVDP) and dp/dtmax. In DCA-pretreated hearts, post-VF lactate production and pyruvate dehydrogenase (PDH) phosphorylation were significantly reduced, indicative of stimulated glucose oxidation, and inhibited anaerobic glycolysis by activation of PDH. Epicardial NADH fluorescence was increased during global ischemia above preischemic levels, but decreased below preischemia levels following VF, with no differences between nontreated controls and DCA-pretreated hearts, whereas DCA pretreatment increased NADH production in nonischemic hearts. With exogenous fatty acids (FA) added to the perfusion solution, DCA pretreatment also resulted in improvements in post-VF LVDP and dp/dtmax, indicating that the presence of exogenous FA did not affect the beneficial actions of DCA. In conclusion, enhancement of PDH activation by DCA mitigates cardiac contractile dysfunction following ischemia-induced VF.


Subject(s)
Dichloroacetic Acid/pharmacology , Heart/drug effects , Myocardial Contraction/drug effects , Myocardial Ischemia/physiopathology , Myocardium/metabolism , Pressure , Ventricular Dysfunction, Left/physiopathology , Ventricular Fibrillation/physiopathology , Ventricular Function, Left/drug effects , Animals , Lactic Acid/metabolism , Male , Myocardial Ischemia/complications , Myocardial Ischemia/metabolism , NAD/drug effects , NAD/metabolism , Phosphorylation/drug effects , Pyruvate Dehydrogenase Complex/metabolism , Rats , Rats, Sprague-Dawley , Ventricular Dysfunction, Left/etiology , Ventricular Dysfunction, Left/metabolism , Ventricular Fibrillation/complications , Ventricular Fibrillation/metabolism
16.
Nat Commun ; 6: 6582, 2015 Mar 20.
Article in English | MEDLINE | ID: mdl-25791197

ABSTRACT

Organometallic complexes are effective hydrogenation catalysts for organic reactions. For example, Noyori-type ruthenium complexes catalyse reduction of ketones by transfer of hydride from formate. Here we show that such catalytic reactions can be achieved in cancer cells, offering a new strategy for the design of safe metal-based anticancer drugs. The activity of ruthenium(II) sulfonamido ethyleneamine complexes towards human ovarian cancer cells is enhanced by up to 50 × in the presence of low non-toxic doses of formate. The extent of conversion of coenzyme NAD(+) to NADH in cells is dependent on formate concentration. This novel reductive stress mechanism of cell death does not involve apoptosis or perturbation of mitochondrial membrane potentials. In contrast, iridium cyclopentadienyl catalysts cause cancer cell death by oxidative stress. Organometallic complexes therefore have an extraordinary ability to modulate the redox status of cancer cells.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Carcinoma/metabolism , Cell Proliferation/drug effects , Fibroblasts/drug effects , Organometallic Compounds/pharmacology , Ovarian Neoplasms/metabolism , Ruthenium Compounds/pharmacology , Catalysis , Cell Line , Cell Line, Tumor , Drug Design , Drug Screening Assays, Antitumor , Female , Formates/pharmacology , Humans , Hydrogenation , NAD/drug effects , NAD/metabolism , Necrosis
17.
Chemistry ; 18(36): 11362-70, 2012 Sep 03.
Article in English | MEDLINE | ID: mdl-22890974

ABSTRACT

Eliamid is a secondary metabolite isolated from two bacterial strains. This molecule features a linear polyketide backbone terminated by a tetramic acid amide moiety. Among other biological activities, eliamid shows a high and specific cytostatic action on human lymphoma and cervix carcinoma cell lines. The 2,4-anti relative configuration of the C-2,C-4-dimethyl substituted amide fragment was assigned by means of Breit's rule. The absolute configuration of all stereocenters was determined by a combination of degradation methods, structural similarity analysis and total synthesis. The stereogenic centers were introduced by vinylogous Mukaiyama aldol reaction and two consecutive Myers alkylations. The use of pentafluorophenyl ester as acylation agent allowed the efficient formation of tetramic acid amide. The longest linear sequence in the synthesis consist of 13 steps and proceeds with 12% overall yield. Differential spectroscopy experiments with beef heart submitochondrial particles established that eliamid is a potent inhibitor of the NADH-ubiquinone oxidoreductase complex. Additionally, biosynthesis of eliamid was investigated by feeding experiments with (13)C-labeled precursors.


Subject(s)
Antifungal Agents/pharmacology , Cytostatic Agents/pharmacology , Electron Transport Complex I/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Myxococcales/chemistry , Pyrrolidinones/pharmacology , Animals , Antifungal Agents/chemistry , Antifungal Agents/isolation & purification , Cattle , Cell Line , Cell Proliferation/drug effects , Cytostatic Agents/chemistry , Cytostatic Agents/isolation & purification , Dose-Response Relationship, Drug , Electron Transport Complex I/metabolism , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/isolation & purification , Fungi/drug effects , Heart/drug effects , Humans , Mice , Microbial Sensitivity Tests , NAD/drug effects , NAD/metabolism , Oxidation-Reduction , Pyrrolidinones/chemistry , Pyrrolidinones/isolation & purification , Rats , Structure-Activity Relationship
18.
Meat Sci ; 90(3): 564-71, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22030110

ABSTRACT

The effects of 4-hydroxy-2-nonenal (HNE) on mitochondria isolated from bovine hearts (n=5) were assessed using ultrastructure, oxygen consumption, membrane permeability, HNE binding, and metmyoglobin reduction in vitro. Pre-incubation (pH 5.6 and 7.4 at 25°C) of mitochondria with HNE decreased oxygen consumption compared with samples without HNE (P<0.05). Electron microscopy revealed that HNE-treated mitochondria were swollen and had increased membrane permeability at pH 7.4, compared with ethanol controls. Conversely, mitochondria incubated with HNE at pH 5.6 had decreased volume and permeability. Fluorescence studies indicate that HNE binds to the membrane of mitochondria isolated from bovine cardiac muscle (at pH 5.6 and 7.4). HNE-treated mitochondria at both pH 5.6 and 7.4 had lower metmyoglobin reduction and NADH dependent metmyoglobin reductase activity compared with control mitochondria without HNE (P<0.05). In addition to covalent binding with myoglobin, HNE may influence beef color stability by interacting with mitochondria.


Subject(s)
Meat , Metmyoglobin/drug effects , Mitochondria, Heart/drug effects , Mitochondria, Heart/ultrastructure , Oxygen Consumption/drug effects , Aldehydes/pharmacology , Animals , Cattle , Color , Heart/drug effects , Hydrogen-Ion Concentration , Metmyoglobin/metabolism , Microscopy, Electron, Transmission , Mitochondria, Heart/metabolism , NAD/drug effects , Spectrometry, Mass, Electrospray Ionization
19.
Cell Calcium ; 50(6): 548-58, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21978418

ABSTRACT

Neuronal Ca(2+) dyshomeostasis associated with cognitive impairment and mediated by changes in several Ca(2+) sources has been seen in animal models of both aging and diabetes. In the periphery, dysregulation of intracellular Ca(2+) signals may contribute to the development of insulin resistance. In the brain, while it is well-established that type 2 diabetes mellitus is a risk factor for the development of dementia in the elderly, it is not clear whether Ca(2+) dysregulation might also affect insulin sensitivity and glucose utilization. Here we present a combination of imaging techniques testing the disappearance of the fluorescent glucose analog 2-(N-(7-nitrobenz-2-oxa-1,3-diazol-4-yl)amino)-2-deoxyglucose (2-NBDG) as an indication of glycolytic activity in neurons and astrocytes. Our work shows that glucose utilization at rest is greater in neurons compared to astrocytes, and ceases upon activation in neurons with little change in astrocytes. Pretreatment of hippocampal cultures with pioglitazone, a drug used in the treatment of type 2 diabetes, significantly reduced glycolytic activity in neurons and enhanced it in astrocytes. This series of experiments, including Fura-2 and NADH imaging, provides results that are consistent with the idea that Ca(2+) levels may rapidly alter glycolytic activity, and that downstream events beyond Ca(2+) dysregulation with aging, may alter cellular metabolism in the brain.


Subject(s)
4-Chloro-7-nitrobenzofurazan/analogs & derivatives , Astrocytes/metabolism , Calcium/metabolism , Deoxyglucose/analogs & derivatives , Fluorescent Dyes/analysis , NAD/metabolism , Neurons/metabolism , Thiazolidinediones/pharmacology , 4-Chloro-7-nitrobenzofurazan/analysis , Animals , Astrocytes/cytology , Astrocytes/drug effects , Calcium Signaling/drug effects , Cells, Cultured , Deoxyglucose/analysis , Fura-2 , Glucose/metabolism , Glycolysis/drug effects , Hippocampus/cytology , NAD/drug effects , Neurons/cytology , Neurons/drug effects , Pioglitazone , Rats , Rats, Sprague-Dawley
20.
Bull Exp Biol Med ; 150(5): 610-3, 2011 Mar.
Article in English | MEDLINE | ID: mdl-22235397

ABSTRACT

The therapeutic action of adenocine during cardiac insufficiency (heart failure) caused by ischemic (stenosis) or reperfusion (removal of ligature) injury to the myocardium prevents depletion of ATP, the major energy source for myocytes in the right and left ventricles, and a drop in NAD/NADH ratio. The development of energy shortage during heart failure cannot be eliminated by ß-acetyldigoxin, levosimendan, or milrinone: the content of ATP in the right and left ventricular myocardium remained below the normal level by 28 and 29%, 37 and 33%, 32 and 28%, respectively; the NAD/NADH ratio of the energy supply system in cardiomyocytes did not return to normal. Adenocine increased the content of NAD to the normal level in both the right and left ventricles, while it remained below the normal level after administration of ß-acetyldigoxin (by 24 and 19.5%, respectively), levosimendan (by 27 and 29%), and milrinone (by 26 and 24%). In contrast to ß-acetyldigoxin, levosimendan, and milrinone, adenocine inhibited activity of poly(ADP-ribose) polymerase in both ventricles. It is concluded that adenocine directly inhibits the key enzyme triggering apoptosis; we also hypothesized that this drug activates the regulatory and signal mechanisms arresting apoptotic alterations in the myocardium during heart failure.


Subject(s)
Adenosine/pharmacology , Cardiotonic Agents/pharmacology , Heart Failure/drug therapy , Ventricular Function/drug effects , Acetyldigoxins/pharmacology , Adenosine Triphosphate/metabolism , Animals , Apoptosis/drug effects , Cardiotonic Agents/therapeutic use , Constriction, Pathologic , Female , Heart/drug effects , Heart Ventricles/metabolism , Heart Ventricles/pathology , Hydrazones/pharmacology , Male , Milrinone/pharmacology , Myocardium/pathology , Myocytes, Cardiac/drug effects , NAD/analysis , NAD/drug effects , Poly(ADP-ribose) Polymerase Inhibitors , Poly(ADP-ribose) Polymerases/metabolism , Pyridazines/pharmacology , Rabbits , Reperfusion , Simendan
SELECTION OF CITATIONS
SEARCH DETAIL
...