Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 63
Filtrar
1.
Br J Anaesth ; 131(4): 745-763, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37567808

RESUMO

BACKGROUND: Neuropathic pain impairs quality of life, is widely prevalent, and incurs significant costs. Current pharmacological therapies have poor/no efficacy and significant adverse effects; safe and effective alternatives are needed. Hyperpolarisation-activated cyclic nucleotide-regulated (HCN) channels are causally implicated in some forms of peripherally mediated neuropathic pain. Whilst 2,6-substituted phenols, such as 2,6-di-tert-butylphenol (26DTB-P), selectively inhibit HCN1 gating and are antihyperalgesic, the development of therapeutically tolerable, HCN-selective antihyperalgesics based on their inverse agonist activity requires that such drugs spare the cardiac isoforms and do not cross the blood-brain barrier. METHODS: In silico molecular dynamics simulation, in vitro electrophysiology, and in vivo rat spared nerve injury methods were used to test whether 'hindered' variants of 26DTB-P (wherein a hydrophilic 'anchor' is attached in the para-position of 26DTB-P via an acyl chain 'tether') had the desired properties. RESULTS: Molecular dynamics simulation showed that membrane penetration of hindered 26DTB-Ps is controlled by a tethered diol anchor without elimination of head group rotational freedom. In vitro and in vivo analysis showed that BP4L-18:1:1, a variant wherein a diol anchor is attached to 26DTB-P via an 18-carbon tether, is an HCN1 inverse agonist and an orally available antihyperalgesic. With a CNS multiparameter optimisation score of 2.25, a >100-fold lower drug load in the brain vs blood, and an absence of adverse cardiovascular or CNS effects, BP4L-18:1:1 was shown to be poorly CNS penetrant and cardiac sparing. CONCLUSIONS: These findings provide a proof-of-concept demonstration that anchor-tethered drugs are a new chemotype for treatment of disorders involving membrane targets.


Assuntos
Agonismo Inverso de Drogas , Neuralgia , Ratos , Animais , Qualidade de Vida , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização/uso terapêutico , Neuralgia/tratamento farmacológico , Fenômenos Eletrofisiológicos
2.
Int J Mol Sci ; 24(13)2023 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-37446292

RESUMO

Numerous efforts in basic and clinical studies have explored the potential anti-aging and health-promoting effects of NAD+-boosting compounds such as nicotinamide riboside (NR) and nicotinamide mononucleotide (NMN). Despite these extensive efforts, our understanding and characterization of their whole-body pharmacodynamics, impact on NAD+ tissue distribution, and mechanism of action in various tissues remain incomplete. In this study, we administered NMN via intraperitoneal injection or oral gavage and conducted a rigorous evaluation of NMN's pharmacodynamic effects on whole-body NAD+ homeostasis in mice. To provide more confident insights into NMN metabolism and NAD+ biosynthesis across different tissues and organs, we employed a novel approach using triple-isotopically labeled [18O-phosphoryl-18O-carbonyl-13C-1-ribosyl] NMN. Our results provide a more comprehensive characterization of the NMN impact on NAD+ concentrations and absolute amounts in various tissues and the whole body. We also demonstrate that mice primarily rely on the nicotinamide and NR salvage pathways to generate NAD+ from NMN, while the uptake of intact NMN plays a minimal role. Overall, the tissue-specific pharmacodynamic effects of NMN administration through different routes offer novel insights into whole-body NAD+ homeostasis, laying a crucial foundation for the development of NMN as a therapeutic supplement in humans.


Assuntos
NAD , Mononucleotídeo de Nicotinamida , Humanos , Camundongos , Animais , Mononucleotídeo de Nicotinamida/farmacologia , Mononucleotídeo de Nicotinamida/metabolismo , NAD/metabolismo , Envelhecimento , Transporte Biológico
3.
mSystems ; 7(1): e0122321, 2022 02 22.
Artigo em Inglês | MEDLINE | ID: mdl-35076274

RESUMO

The effort to use nutrients as interventions to treat human disease has been important to medicine. A current example in this vein pertains to NAD+ boosters, such as nicotinamide riboside (NR) and nicotinamide mononucleotide (NMN), which are in many clinical trials in a variety of disease conditions. Independent laboratories have shown that ingested NR (or NMN) has mitigating effects on metabolic syndrome in mice. V. V. Lozada-Fernández, O. deLeon, S. L. Kellogg, F. L. Saravia, et al. (mSystems 7:e00230-21, 2022, https://doi.org/10.1128/mSystems.00230-21) show that NR shifts gut microbiome contents and that the transplantation of an NR-conditioned microbiome by fecal transfer reproduces some effects of NR in mice on a high-fat diet. The involvement of the gut microbiome as a factor in NR effects is linked to changes to the gut microbiome and its activity to transform NR and downstream catabolites. This commentary draws attention to these findings and focuses on some puzzling aspects of NAD+ boosters, exploring the still murky interactions between NAD+ metabolism, energy homeostasis, and the gut microbiome.


Assuntos
Microbioma Gastrointestinal , Doenças Metabólicas , Microbiota , Humanos , Animais , Camundongos , NAD/metabolismo , Dieta Hiperlipídica , Mononucleotídeo de Nicotinamida/metabolismo , Aumento de Peso
4.
Methods Mol Biol ; 2310: 271-285, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34096008

RESUMO

NAD+ is a redox cofactor essential to the proper functioning of a variety of important metabolic pathways, including key steps in mitochondrial energy metabolism. In addition, it serves as a signaling substrate for enzymes such as sirtuins and the poly-ADP ribosyl-polymerase family of enzymes. Sirtuins, which are NAD+-dependent protein deacylases, harness changes in cellular NAD+ concentrations to produce changes in protein acylation status, thereby affecting downstream functions including energy metabolism, stress resistance, and cell survival. Thus, the availability of NAD+ in cells, or in specific organelles such as the mitochondrion, regulates downstream signaling and key biological processes. This concept has driven a need for researchers to easily and precisely measure NAD+ concentrations in biological samples. We herein describe several protocols for the measurement of NAD+ and NADH concentrations in tissues, cells, or subcellular compartments such as mitochondria. These protocols include a cycling assay that can quickly measure NAD+ or NADH levels using a plate reader equipped with fluorescence measurement capabilities. This plate assay relies only upon commercially available materials in addition to the biological samples of interest. In addition, we describe a protocol employing stable isotope-labeled NAD+ as an internal standard to determine biological NAD+ content by isotope-dilution methods. This method requires mass spectrometry to ratio endogenous NAD+ with exogenous isotope-labeled NAD+ to obtain quantification using HPLC and mass spectrometry.


Assuntos
Cromatografia Líquida de Alta Pressão , Metabolismo Energético , Técnicas de Diluição do Indicador , Espectrometria de Massas , Mitocôndrias/metabolismo , NAD/metabolismo , Fracionamento Celular , Células Cultivadas , Espectrometria de Fluorescência
5.
Nat Metab ; 2(4): 364-379, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32694608

RESUMO

Dihydronicotinamide riboside (NRH) has been suggested to act as a precursor for the synthesis of NAD+, but the biochemical pathway converting it has been unknown. Here, we show that NRH can be converted into NAD+ via a salvage pathway in which adenosine kinase (ADK, also known as AK) acts as an NRH kinase. Using isotope-labelling approaches, we demonstrate that NRH is fully incorporated into NAD+, with NMNH acting as an intermediate. We further show that AK is enriched in fractions from cell lysates with NRH kinase activity, and that AK can convert NRH into NAD+. In cultured cells and mouse liver, pharmacological or genetic inhibition of AK blocks formation of reduced nicotinamide mononucleotide (NMNH) and inhibits NRH-stimulated NAD+ biosynthesis. Finally, we confirm the presence of endogenous NRH in the liver with metabolomics. Our findings establish NRH as a natural precursor of NAD+ and reveal a new route for NAD+ biosynthesis via an NRH salvage pathway involving AK.


Assuntos
Adenosina Quinase/metabolismo , NAD/metabolismo , Niacinamida/análogos & derivados , Animais , Células Cultivadas , Camundongos , Niacinamida/metabolismo , Fosforilação
6.
J Biol Chem ; 294(23): 9295-9307, 2019 06 07.
Artigo em Inglês | MEDLINE | ID: mdl-30948509

RESUMO

Interest in pharmacological agents capable of increasing cellular NAD+ concentrations has stimulated investigations of nicotinamide riboside (NR) and nicotinamide mononucleotide (NMN). NR and NMN require large dosages for effect. Herein, we describe synthesis of dihydronicotinamide riboside (NRH) and the discovery that NRH is a potent NAD+ concentration-enhancing agent, which acts within as little as 1 h after administration to mammalian cells to increase NAD+ concentrations by 2.5-10-fold over control values. Comparisons with NR and NMN show that in every instance, NRH provides greater NAD+ increases at equivalent concentrations. NRH also provides substantial NAD+ increases in tissues when administered by intraperitoneal injection to C57BL/6J mice. NRH substantially increases NAD+/NADH ratio in cultured cells and in liver and no induction of apoptotic markers or significant increases in lactate levels in cells. Cells treated with NRH are resistant to cell death caused by NAD+-depleting genotoxins such as hydrogen peroxide and methylmethane sulfonate. Studies to identify its biochemical mechanism of action showed that it does not inhibit NAD+ consumption, suggesting that it acts as a biochemical precursor to NAD+ Cell lysates possess an ATP-dependent kinase activity that efficiently converts NRH to the compound NMNH, but independent of Nrk1 or Nrk2. These studies identify a putative new metabolic pathway to NAD+ and a potent pharmacologic agent for NAD+ concentration enhancement in cells and tissues.


Assuntos
Apoptose/efeitos dos fármacos , NAD/metabolismo , Niacinamida/análogos & derivados , Animais , Linhagem Celular , Cromatografia Líquida de Alta Pressão , Humanos , Peróxido de Hidrogênio/farmacologia , Injeções Intraperitoneais , Ácido Láctico/metabolismo , Fígado/metabolismo , Masculino , Metanossulfonato de Metila/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , NAD/análise , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Niacinamida/administração & dosagem , Niacinamida/síntese química , Niacinamida/farmacologia
7.
Cell Metab ; 27(3): 667-676.e4, 2018 03 06.
Artigo em Inglês | MEDLINE | ID: mdl-29514072

RESUMO

The role in longevity and healthspan of nicotinamide (NAM), the physiological precursor of NAD+, is elusive. Here, we report that chronic NAM supplementation improves healthspan measures in mice without extending lifespan. Untargeted metabolite profiling of the liver and metabolic flux analysis of liver-derived cells revealed NAM-mediated improvement in glucose homeostasis in mice on a high-fat diet (HFD) that was associated with reduced hepatic steatosis and inflammation concomitant with increased glycogen deposition and flux through the pentose phosphate and glycolytic pathways. Targeted NAD metabolome analysis in liver revealed depressed expression of NAM salvage in NAM-treated mice, an effect counteracted by higher expression of de novo NAD biosynthetic enzymes. Although neither hepatic NAD+ nor NADP+ was boosted by NAM, acetylation of some SIRT1 targets was enhanced by NAM supplementation in a diet- and NAM dose-dependent manner. Collectively, our results show health improvement in NAM-supplemented HFD-fed mice in the absence of survival effects.


Assuntos
Suplementos Nutricionais , Envelhecimento Saudável/metabolismo , Fígado , NAD/metabolismo , Niacinamida/farmacologia , Animais , Dieta Hiperlipídica , Modelos Animais de Doenças , Fígado Gorduroso/tratamento farmacológico , Inflamação/tratamento farmacológico , Fígado/efeitos dos fármacos , Fígado/metabolismo , Longevidade , Camundongos Endogâmicos C57BL , Niacinamida/administração & dosagem , Estresse Oxidativo/efeitos dos fármacos , Sirtuína 1/metabolismo
8.
Prog Mol Biol Transl Sci ; 154: 71-104, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29413178

RESUMO

NAD+ acts as a crucial regulator of cell physiology and as an integral participant in cellular metabolism. By virtue of a variety of signaling activities this central metabolite can exert profound effects on organism health status. Thus, while it serves as a well-known metabolic cofactor functioning as a redox-active substrate, it can also function as a substrate for signaling enzymes, such as sirtuins, poly (ADP-ribosyl) polymerases, mono (ADP-ribosyl) transferases, and CD38. Sirtuins function as NAD+-dependent protein deacetylases (deacylases) and catalyze the reaction of NAD+ with acyllysine groups to remove the acyl modification from substrate proteins. This deacetylation provides a regulatory function and integrates cellular NAD+ metabolism into a large spectrum of cellular processes and outcomes, such as cell metabolism, cell survival, cell cycle, apoptosis, DNA repair, mitochondrial homeostasis and mitochondrial biogenesis, and even lifespan. Increased attention to how regulated and pharmacologic changes in NAD+ concentrations can impact sirtuin activities has motivated openings of new areas of research, including investigations of how NAD+ levels are regulated at the subcellular level, and searches for more potent NAD+ precursors typified by nicotinamide riboside (NR) and nicotinamide mononucleotide (NMN). This review describes current results and thinking of how NAD+ metabolic pathways regulate sirtuin activities and how regulated NAD+ levels can impact cell physiology. In addition, NAD+ precursors are discussed, with attention to how these might be harnessed to generate novel therapeutic options to treat the diseases of aging.


Assuntos
Redes e Vias Metabólicas , Mitocôndrias/metabolismo , NAD/metabolismo , Sirtuínas/metabolismo , Animais , Humanos , Transdução de Sinais
9.
Curr Protoc Nucleic Acid Chem ; 71: 14.14.1-14.14.9, 2017 12 24.
Artigo em Inglês | MEDLINE | ID: mdl-29275540

RESUMO

A two-step chemical method for the synthesis of ß-nicotinamide riboside (NR) is described. NR has achieved wide use as an NAD+ precursor (vitamin B3) and can significantly increase central metabolite NAD+ concentrations in mammalian cells. ß-NR can be prepared with an efficient two-step procedure. The synthesis is initiated via coupling of commercially available 1,2,3,5-tetra-O-acetyl-ß-D-ribofuranose with ethyl nicotinate in the presence of trimethylsilyl trifluoromethanesulfonate (TMSOTf). 1 H NMR showed that the product was formed with complete stereoselectivity to produce only the ß-isomer in high yield (>90% versus starting sugar). The clean stereochemical result suggests that the coupling proceeds via a cationic cis-1,2-acyloxonium-sugar intermediate, which controls addition by nucleophiles to generate predominantly ß-stereochemistry. The subsequent deprotection of esters in methanolic ammonia generates the desired product in 85% overall yield versus sugar. © 2017 by John Wiley & Sons, Inc.


Assuntos
Niacinamida/análogos & derivados , Animais , Cromatografia Líquida de Alta Pressão , NAD/química , Niacinamida/síntese química , Niacinamida/química , Ácidos Nicotínicos/química , Espectroscopia de Prótons por Ressonância Magnética , Compostos de Piridínio , Estereoisomerismo
10.
Sci Transl Med ; 8(361): 361ra139, 2016 10 19.
Artigo em Inglês | MEDLINE | ID: mdl-27798264

RESUMO

Neuromuscular diseases are often caused by inherited mutations that lead to progressive skeletal muscle weakness and degeneration. In diverse populations of normal healthy mice, we observed correlations between the abundance of mRNA transcripts related to mitochondrial biogenesis, the dystrophin-sarcoglycan complex, and nicotinamide adenine dinucleotide (NAD+) synthesis, consistent with a potential role for the essential cofactor NAD+ in protecting muscle from metabolic and structural degeneration. Furthermore, the skeletal muscle transcriptomes of patients with Duchene's muscular dystrophy (DMD) and other muscle diseases were enriched for various poly[adenosine 5'-diphosphate (ADP)-ribose] polymerases (PARPs) and for nicotinamide N-methyltransferase (NNMT), enzymes that are major consumers of NAD+ and are involved in pleiotropic events, including inflammation. In the mdx mouse model of DMD, we observed significant reductions in muscle NAD+ levels, concurrent increases in PARP activity, and reduced expression of nicotinamide phosphoribosyltransferase (NAMPT), the rate-limiting enzyme for NAD+ biosynthesis. Replenishing NAD+ stores with dietary nicotinamide riboside supplementation improved muscle function and heart pathology in mdx and mdx/Utr-/- mice and reversed pathology in Caenorhabditis elegans models of DMD. The effects of NAD+ repletion in mdx mice relied on the improvement in mitochondrial function and structural protein expression (α-dystrobrevin and δ-sarcoglycan) and on the reductions in general poly(ADP)-ribosylation, inflammation, and fibrosis. In combination, these studies suggest that the replenishment of NAD+ may benefit patients with muscular dystrophies or other neuromuscular degenerative conditions characterized by the PARP/NNMT gene expression signatures.


Assuntos
Músculo Esquelético/fisiopatologia , Distrofias Musculares/patologia , NAD/química , Poli ADP Ribosilação , Difosfato de Adenosina/química , Animais , Caenorhabditis elegans , Linhagem Celular , Citocinas/química , Fibrose/patologia , Perfilação da Expressão Gênica , Inflamação , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Doenças Musculares/patologia , Nicotinamida Fosforribosiltransferase/química , Nitrosaminas/química , RNA Mensageiro/metabolismo , Tiramina/análogos & derivados , Tiramina/química
11.
Biochim Biophys Acta ; 1864(12): 1787-1800, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27374990

RESUMO

We survey the historical development of scientific knowledge surrounding Vitamin B3, and describe the active metabolite forms of Vitamin B3, the pyridine dinucleotides NAD+ and NADP+ which are essential to cellular processes of energy metabolism, cell protection and biosynthesis. The study of NAD+ has become reinvigorated by new understandings that dynamics within NAD+ metabolism trigger major signaling processes coupled to effectors (sirtuins, PARPs, and CD38) that reprogram cellular metabolism using NAD+ as an effector substrate. Cellular adaptations include stimulation of mitochondrial biogenesis, a process fundamental to adjusting cellular and tissue physiology to reduced nutrient availability and/or increased energy demand. Several mammalian metabolic pathways converge to NAD+, including tryptophan-derived de novo pathways, nicotinamide salvage pathways, nicotinic acid salvage and nucleoside salvage pathways incorporating nicotinamide riboside and nicotinic acid riboside. Key discoveries highlight a therapeutic potential for targeting NAD+ biosynthetic pathways for treatment of human diseases. A recent emergence of understanding that NAD+ homeostasis is vulnerable to aging and disease processes has stimulated testing to determine if replenishment or augmentation of cellular or tissue NAD+ can have ameliorative effects on aging or disease phenotypes. This experimental approach has provided several proofs of concept successes demonstrating that replenishment or augmentation of NAD+ concentrations can provide ameliorative or curative benefits. Thus NAD+ metabolic pathways can provide key biomarkers and parameters for assessing and modulating organism health.


Assuntos
NAD/metabolismo , Animais , Metabolismo Energético , Humanos , Doenças Metabólicas/metabolismo , Doenças Metabólicas/terapia , Redes e Vias Metabólicas , Modelos Biológicos , NADP/metabolismo , Niacina/metabolismo , Niacinamida/metabolismo , Nucleosídeos/metabolismo , Transdução de Sinais , Sirtuínas/metabolismo , Triptofano/metabolismo
12.
Hepatology ; 63(4): 1190-204, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26404765

RESUMO

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.


Assuntos
Fígado Gorduroso/tratamento farmacológico , Fígado Gorduroso/patologia , NAD/metabolismo , Niacinamida/análogos & derivados , Resposta a Proteínas não Dobradas/efeitos dos fármacos , Análise de Variância , Animais , Área Sob a Curva , Biópsia por Agulha , Dieta Hiperlipídica/métodos , Modelos Animais de Doenças , Fígado Gorduroso/metabolismo , Imuno-Histoquímica , Metabolismo dos Lipídeos/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , NAD/efeitos dos fármacos , Niacinamida/farmacologia , Compostos de Piridínio , Distribuição Aleatória , Sensibilidade e Especificidade , Resultado do Tratamento
13.
J Clin Invest ; 125(12): 4592-600, 2015 Nov 03.
Artigo em Inglês | MEDLINE | ID: mdl-26529255

RESUMO

BACKGROUND: Activation of the NLRP3 inflammasome is associated with metabolic dysfunction, and intermittent fasting has been shown to improve clinical presentation of NLRP3 inflammasome-linked diseases. As mitochondrial perturbations, which function as a damage-associated molecular pattern, exacerbate NLRP3 inflammasome activation, we investigated whether fasting blunts inflammasome activation via sirtuin-mediated augmentation of mitochondrial integrity. METHODS: We performed a clinical study of 19 healthy volunteers. Each subject underwent a 24-hour fast and then was fed a fixed-calorie meal. Blood was drawn during the fasted and fed states and analyzed for NRLP3 inflammasome activation. We enrolled an additional group of 8 healthy volunteers to assess the effects of the sirtuin activator, nicotinamide riboside, on NLRP3 inflammasome activation. RESULTS: In the fasting/refeeding study, individuals showed less NLRP3 inflammasome activation in the fasted state compared with that in refed conditions. In a human macrophage line, depletion of the mitochondrial-enriched sirtuin deacetylase SIRT3 increased NLRP3 inflammasome activation in association with excessive mitochondrial ROS production. Furthermore, genetic and pharmacologic SIRT3 activation blunted NLRP3 activity in parallel with enhanced mitochondrial function in cultured cells and in leukocytes extracted from healthy volunteers and from refed individuals but not in those collected during fasting. CONCLUSIONS: Together, our data indicate that nutrient levels regulate the NLRP3 inflammasome, in part through SIRT3-mediated mitochondrial homeostatic control. Moreover, these results suggest that deacetylase-dependent inflammasome attenuation may be amenable to targeting in human disease. TRIAL REGISTRATION: ClinicalTrials.gov NCT02122575 and NCT00442195. FUNDING: Division of Intramural Research, NHLBI of the NIH.


Assuntos
Proteínas de Transporte/sangue , Ingestão de Alimentos , Jejum/sangue , Inflamassomos/sangue , Mitocôndrias/metabolismo , Adulto , Feminino , Humanos , Masculino , Proteína 3 que Contém Domínio de Pirina da Família NLR , Niacinamida/administração & dosagem , Niacinamida/análogos & derivados , Compostos de Piridínio
14.
Cell Rep ; 13(3): 533-545, 2015 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-26456827

RESUMO

Both iron overload and iron deficiency have been associated with cardiomyopathy and heart failure, but cardiac iron utilization is incompletely understood. We hypothesized that the transferrin receptor (Tfr1) might play a role in cardiac iron uptake and used gene targeting to examine the role of Tfr1 in vivo. Surprisingly, we found that decreased iron, due to inactivation of Tfr1, was associated with severe cardiac consequences. Mice lacking Tfr1 in the heart died in the second week of life and had cardiomegaly, poor cardiac function, failure of mitochondrial respiration, and ineffective mitophagy. The phenotype could only be rescued by aggressive iron therapy, but it was ameliorated by administration of nicotinamide riboside, an NAD precursor. Our findings underscore the importance of both Tfr1 and iron in the heart, and may inform therapy for patients with heart failure.


Assuntos
Cardiomiopatias/genética , Miocárdio/metabolismo , Receptores da Transferrina/genética , Animais , Cardiomiopatias/tratamento farmacológico , Cardiomiopatias/patologia , Respiração Celular , Ferro/metabolismo , Camundongos , Mitofagia , Miocárdio/patologia , Niacinamida/análogos & derivados , Niacinamida/uso terapêutico , Compostos de Piridínio , Receptores da Transferrina/metabolismo
15.
Methods Mol Biol ; 1241: 39-48, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25308486

RESUMO

Nicotinamide adenine dinucleotide (NAD(+)) is a central metabolic coenzyme/cosubstrate involved in cellular energy metabolism and energy production. It can readily be reduced by two electron equivalents and forms the NADH form, which is the minority species to NAD(+) under most physiologic conditions. NAD(+) plays an important role in not only oxidation-reduction reactions in cells but also as a signaling molecule. For example, NAD(+) plays a key role in mitochondrial function via participation in pyruvate dehydrogenase, tricarboxylic acid cycle, and oxidative phosphorylation chemistries. It also serves as a substrate for deacylases SIRT3, SIRT4, and SIRT5, which modify protein posttranslational modifications on lysine within the mitochondrial compartment. Recent work has highlighted the biological significance of dynamic changes to mitochondrial NAD(+). This has increased the need for standardized and effective methods to measure NAD(+) contents in this organelle. To determine NAD(+) concentrations in cells, and specifically in mitochondria, we describe two assays for NAD(+) determinations: An Enzymatic Cycling Assay and Isotope Dilution. The cycling assay contains sample NAD(+), lactate, lactate dehydrogenase, diaphorase, and resazurin. The isotope dilution assay uses synthetic (18)O-NAD(+) as an internal standard, and treated samples are fractionated by HPLC and then NAD(+) concentration determined by the (16)O- and (18)O-NAD(+) peak (664/666) ratio in positive mode MS.


Assuntos
Cromatografia Líquida de Alta Pressão/métodos , Espectrometria de Massas/métodos , Mitocôndrias/metabolismo , NAD/metabolismo , Cromatografia Líquida de Alta Pressão/normas , Espectrometria de Massas/normas , Padrões de Referência
16.
Oncotarget ; 5(17): 7303-15, 2014 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-25229978

RESUMO

In our prior publications we characterized a conserved acetylation motif (K(R)xxKK) of evolutionarily related nuclear receptors. Recent reports showed that peroxisome proliferator activated receptor gamma (PPARγ) deacetylation by SIRT1 is involved in delaying cellular senescence and maintaining the brown remodeling of white adipose tissue. However, it still remains unknown whether lysyl residues 154 and 155 (K154/155) of the conserved acetylation motif (RIHKK) in Pparγ1 are acetylated. Herein, we demonstrate that Pparγ1 is acetylated and regulated by both endogenous TSA-sensitive and NAD-dependent deacetylases. Acetylation of lysine 154 was identified by mass spectrometry (MS) while deacetylation of lysine 155 by SIRT1 was confirmed by in vitro deacetylation assay. An in vivo labeling assay revealed K154/K155 as bona fide acetylation sites. The conserved acetylation sites of Pparγ1 and the catalytic domain of SIRT1 are both required for the interaction between Pparγ1 and SIRT1. Sirt1 and Pparγ1 converge to govern lipid metabolism in vivo. Acetylation-defective mutants of Pparγ1 were associated with reduced lipid synthesis in ErbB2 overexpressing breast cancer cells. Together, these results suggest that the conserved lysyl residues K154/K155 of Pparγ1 are acetylated and play an important role in lipid synthesis in ErbB2-positive breast cancer cells.


Assuntos
Neoplasias da Mama/metabolismo , Senescência Celular/fisiologia , Metabolismo dos Lipídeos/fisiologia , PPAR gama/metabolismo , Acetilação , Motivos de Aminoácidos/fisiologia , Animais , Western Blotting , Linhagem Celular Tumoral , Sequência Conservada , Humanos , Imunoprecipitação , Espectrometria de Massas , Camundongos , Mutação , Análise de Sequência com Séries de Oligonucleotídeos , Estrutura Terciária de Proteína/fisiologia , Sirtuína 1/metabolismo , Transfecção
17.
Biochim Biophys Acta ; 1844(12): 2182-92, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25091197

RESUMO

The ability to derivatize antibodies is currently limited by the chemical structure of antibodies as polypeptides. Modern methods of bioorthogonal and biocompatible chemical modifications could make antibody functionalization more predictable and easier, without compromising the functions of the antibody. To explore this concept, we modified the well-known anti-epidermal growth factor receptor (EGFR) drug, cetuximab (Erbitux®), with 5-azido-2-nitro-benzoyl (ANB) modifications by optimization of an acylation protocol. We then show that the resulting ANB-cetuximab can be reliably modified with dyes (TAMRA and carboxyrhodamine) or a novel synthesized cyclooctyne modified biotin. The resulting dye- and biotin-modified cetuximabs were then tested across several assay platforms with several cell lines including U87, LN229, F98EGFR, F98WT and HEK293 cells. The assay platforms included fluorescence microscopy, FACS and biotin-avidin based immunoprecipitation methods. The modified antibody performs consistently in all of these assay platforms, reliably determining relative abundances of EGFR expression on EGFR expressing cells (LN229 and F98EGFR) and failing to cross react with weak to negative EGFR expressing cells (U87, F98WT and HEK293). The ease of achieving diverse and assay relevant functionalizations as well as the consequent rapid construction of highly correlated antigen expression data sets highlights the power of bioorthogonal and biocompatible methods to conjugate macromolecules. These data provide a proof of concept for a multifunctionalization strategy that leverages the biochemical versatility and antigen specificity of antibodies.

18.
Cell Metab ; 19(6): 1034-41, 2014 Jun 03.
Artigo em Inglês | MEDLINE | ID: mdl-24814482

RESUMO

We previously demonstrated that the deletion of the poly(ADP-ribose)polymerase (Parp)-1 gene in mice enhances oxidative metabolism, thereby protecting against diet-induced obesity. However, the therapeutic use of PARP inhibitors to enhance mitochondrial function remains to be explored. Here, we show tight negative correlation between Parp-1 expression and energy expenditure in heterogeneous mouse populations, indicating that variations in PARP-1 activity have an impact on metabolic homeostasis. Notably, these genetic correlations can be translated into pharmacological applications. Long-term treatment with PARP inhibitors enhances fitness in mice by increasing the abundance of mitochondrial respiratory complexes and boosting mitochondrial respiratory capacity. Furthermore, PARP inhibitors reverse mitochondrial defects in primary myotubes of obese humans and attenuate genetic defects of mitochondrial metabolism in human fibroblasts and C. elegans. Overall, our work validates in worm, mouse, and human models that PARP inhibition may be used to treat both genetic and acquired muscle dysfunction linked to defective mitochondrial function.


Assuntos
Metabolismo Energético/fisiologia , Mitocôndrias/metabolismo , Fibras Musculares Esqueléticas/metabolismo , Inibidores de Poli(ADP-Ribose) Polimerases , Poli(ADP-Ribose) Polimerases/metabolismo , Animais , Benzamidas/farmacologia , Benzimidazóis/farmacologia , Caenorhabditis elegans , Células Cultivadas , Inibidores Enzimáticos/farmacologia , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Obesidade/metabolismo , Ftalazinas/farmacologia , Piperazinas/farmacologia , Poli(ADP-Ribose) Polimerases/biossíntese , Sirtuína 1/genética , Sirtuína 1/metabolismo
19.
Cell Metab ; 19(6): 1042-9, 2014 Jun 03.
Artigo em Inglês | MEDLINE | ID: mdl-24814483

RESUMO

Mitochondrial disorders are highly heterogeneous conditions characterized by defects of the mitochondrial respiratory chain. Pharmacological activation of mitochondrial biogenesis has been proposed as an effective means to correct the biochemical defects and ameliorate the clinical phenotype in these severely disabling, often fatal, disorders. Pathways related to mitochondrial biogenesis are targets of Sirtuin1, a NAD(+)-dependent protein deacetylase. As NAD(+) boosts the activity of Sirtuin1 and other sirtuins, intracellular levels of NAD(+) play a key role in the homeostatic control of mitochondrial function by the metabolic status of the cell. We show here that supplementation with nicotinamide riboside, a natural NAD(+) precursor, or reduction of NAD(+) consumption by inhibiting the poly(ADP-ribose) polymerases, leads to marked improvement of the respiratory chain defect and exercise intolerance of the Sco2 knockout/knockin mouse, a mitochondrial disease model characterized by impaired cytochrome c oxidase biogenesis. This strategy is potentially translatable into therapy of mitochondrial disorders in humans.


Assuntos
Metabolismo Energético/fisiologia , NAD/metabolismo , Niacinamida/análogos & derivados , Inibidores de Poli(ADP-Ribose) Polimerases , Sirtuína 1/metabolismo , Animais , Suplementos Nutricionais , Modelos Animais de Doenças , Complexo IV da Cadeia de Transporte de Elétrons/biossíntese , Complexo IV da Cadeia de Transporte de Elétrons/genética , Ativação Enzimática , Expressão Gênica , Camundongos , Camundongos Knockout , Mitocôndrias/patologia , Doenças Mitocondriais/tratamento farmacológico , Chaperonas Moleculares , Niacinamida/farmacologia , Fosforilação Oxidativa , Fenantrenos/farmacologia , Fenótipo , Poli(ADP-Ribose) Polimerase-1 , Poli(ADP-Ribose) Polimerases/genética , Compostos de Piridínio
20.
Diabetes ; 63(11): 3626-36, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24848064

RESUMO

Excessive dietary fat intake causes systemic metabolic toxicity, manifested in weight gain, hyperglycemia, and insulin resistance. In addition, carbohydrate utilization as a fuel is substantially inhibited. Correction or reversal of these effects during high-fat diet (HFD) intake is of exceptional interest in light of widespread occurrence of diet-associated metabolic disorders in global human populations. Here we report that mangiferin (MGF), a natural compound (the predominant constituent of Mangifera indica extract from the plant that produces mango), protected against HFD-induced weight gain, increased aerobic mitochondrial capacity and thermogenesis, and improved glucose and insulin profiles. To obtain mechanistic insight into the basis for these effects, we determined that mice exposed to an HFD combined with MGF exhibited a substantial shift in respiratory quotient from fatty acid toward carbohydrate utilization. MGF treatment significantly increased glucose oxidation in muscle of HFD-fed mice without changing fatty acid oxidation. These results indicate that MGF redirects fuel utilization toward carbohydrates. In cultured C2C12 myotubes, MGF increased glucose and pyruvate oxidation and ATP production without affecting fatty acid oxidation, confirming in vivo and ex vivo effects. Furthermore, MGF inhibited anaerobic metabolism of pyruvate to lactate but enhanced pyruvate oxidation. A key target of MGF appears to be pyruvate dehydrogenase, determined to be activated by MGF in a variety of assays. These findings underscore the therapeutic potential of activation of carbohydrate utilization in correction of metabolic syndrome and highlight the potential of MGF to serve as a model compound that can elicit fuel-switching effects.


Assuntos
Metabolismo dos Carboidratos/efeitos dos fármacos , Xantonas/farmacologia , Animais , Dieta Hiperlipídica , Metabolismo Energético/efeitos dos fármacos , Cetona Oxirredutases/metabolismo , Metabolismo dos Lipídeos/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Oxirredução/efeitos dos fármacos , Ácido Pirúvico/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...