Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 708
Filter
1.
Nat Commun ; 15(1): 6652, 2024 Aug 06.
Article in English | MEDLINE | ID: mdl-39103368

ABSTRACT

Nicotinamide adenine dinucleotide (NAD+) is a redox cofactor and signal central to cell metabolisms. Disrupting NAD homeostasis in plant alters growth and stress resistance, yet the underlying mechanisms remain largely unknown. Here, by combining genetics with multi-omics, we discover that NAD+ deficiency in qs-2 caused by mutation in NAD+ biosynthesis gene-Quinolinate Synthase retards growth but induces biosynthesis of defense compounds, notably aliphatic glucosinolates that confer insect resistance. The elevated defense in qs-2 is resulted from activated jasmonate biosynthesis, critically hydroperoxidation of α-linolenic acid by the 13-lipoxygenase (namely LOX2), which is escalated via the burst of chloroplastic ROS-singlet oxygen (1O2). The NAD+ deficiency-mediated JA induction and defense priming sequence in plants is recapitulated upon insect infestation, suggesting such defense mechanism operates in plant stress response. Hence, NAD homeostasis is a pivotal metabolic checkpoint that may be manipulated to navigate plant growth and defense metabolism for stress acclimation.


Subject(s)
Arabidopsis Proteins , Arabidopsis , Cyclopentanes , NAD , Oxylipins , Cyclopentanes/metabolism , Oxylipins/metabolism , NAD/metabolism , NAD/biosynthesis , Arabidopsis/genetics , Arabidopsis/metabolism , Arabidopsis Proteins/metabolism , Arabidopsis Proteins/genetics , Gene Expression Regulation, Plant , Homeostasis , Animals , Mutation , Lipoxygenase/metabolism , Lipoxygenase/genetics , Glucosinolates/metabolism , Glucosinolates/biosynthesis , Reactive Oxygen Species/metabolism , Stress, Physiological
2.
Nutrients ; 16(14)2024 Jul 20.
Article in English | MEDLINE | ID: mdl-39064797

ABSTRACT

Nicotinamide mononucleotide (NMN), the direct precursor of nicotinamide adenine dinucleotide (NAD+), is involved in the regulation of many physiological and metabolic reactions in the body. NMN can indirectly affect cellular metabolic pathways, DNA repair, and senescence, while also being essential for maintaining tissues and dynamic metabolic equilibria, promoting healthy aging. Therefore, NMN has found many applications in the food, pharmaceutical, and cosmetics industries. At present, NMN synthesis strategies mainly include chemical synthesis and biosynthesis. Despite its potential benefits, the commercial production of NMN by organic chemistry approaches faces environmental and safety problems. With the rapid development of synthetic biology, it has become possible to construct microbial cell factories to produce NMN in a cost-effective way. In this review, we summarize the chemical and biosynthetic strategies of NMN, offering an overview of the recent research progress on host selection, chassis cell optimization, mining of key enzymes, metabolic engineering, and adaptive fermentation strategies. In addition, we also review the advances in the role of NMN in aging, metabolic diseases, and neural function. This review provides comprehensive technical guidance for the efficient biosynthesis of NMN as well as a theoretical basis for its application in the fields of food, medicine, and cosmetics.


Subject(s)
Metabolic Engineering , Nicotinamide Mononucleotide , Nicotinamide Mononucleotide/metabolism , Humans , Metabolic Engineering/methods , Animals , Aging , Metabolic Networks and Pathways , Fermentation , NAD/biosynthesis , NAD/metabolism
3.
FASEB J ; 38(14): e23804, 2024 Jul 31.
Article in English | MEDLINE | ID: mdl-39037422

ABSTRACT

Natural Nicotinamide Adenine Dinucleotide (NAD+) precursors have attracted much attention due to their positive effects in promoting ovarian health. However, their target tissue, synthesis efficiency, advantages, and disadvantages are still unclear. This review summarizes the distribution of NAD+ at the tissue, cellular and subcellular levels, discusses its biosynthetic pathways and the latest findings in ovary, include: (1) NAD+ plays distinct roles both intracellularly and extracellularly, adapting its distribution in response to requirements. (2) Different precursors differs in target tissues, synthetic efficiency, biological utilization, and adverse effects. Importantly: tryptophan is primarily utilized in the liver and kidneys, posing metabolic risks in excess; nicotinamide (NAM) is indispensable for maintaining NAD+ levels; nicotinic acid (NA) constructs a crucial bridge between intestinal microbiota and the host with diverse functions; nicotinamide riboside (NR) and nicotinamide mononucleotide (NMN) increase NAD+ systemically and can be influenced by delivery route, tissue specificity, and transport efficiency. (3) The biosynthetic pathways of NAD+ are intricately intertwined. They provide multiple sources and techniques for NAD+ synthesis, thereby reducing the dependence on a single molecule to maintain cellular NAD+ levels. However, an excess of a specific precursor potentially influencing other pathways. In addition, Protein expression analysis suggest that ovarian tissues may preferentially utilize NAM and NMN. These findings summarize the specific roles and potential of NAD+ precursors in enhancing ovarian health. Future research should delve into the molecular mechanisms and intervention strategies of different precursors, aiming to achieve personalized prevention or treatment of ovarian diseases, and reveal their clinical application value.


Subject(s)
NAD , Niacinamide , Ovary , Humans , NAD/metabolism , NAD/biosynthesis , Ovary/metabolism , Female , Animals , Niacinamide/metabolism , Niacinamide/biosynthesis , Biosynthetic Pathways , Nicotinamide Mononucleotide/metabolism
4.
Toxicol Sci ; 201(1): 118-128, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-38830045

ABSTRACT

High-dose perfluorooctanoic acid (PFOA) impairs oocyte maturation and offspring quality. However, the physiological concentrations of PFOA in follicular fluids of patients with premature ovarian insufficiency (POI) were detected at lower levels, thus the relationship between physiological PFOA and reproductive disorders remains elusive. Here, we investigated whether physiological PFOA exposure affects gonad function in adult zebrafish. Physiological PFOA exposure resulted in POI-like phenotypes in adult females, which exhibited decreased spawning frequency, reduced number of ovulated eggs, abnormal gonadal index, and aberrant embryonic mortality. Meanwhile, oocytes from PFOA-exposed zebrafish showed mitochondrial disintegration and diminished mitochondrial membrane potential. Unlike the high-dose treated oocytes exhibiting high reactive oxygen species (ROS) levels and excessive apoptosis, physiological PFOA reduced the ROS levels and did not trigger apoptosis. Interestingly, physiological PFOA exposure would not affect testis function, indicating specific toxicity in females. Mechanistically, PFOA suppressed the NAD+ biosynthesis and impaired mitochondrial function in oocytes, thus disrupting oocyte maturation and ovarian fertility. Nicotinamide mononucleotide (NMN), a precursor for NAD+ biosynthesis, alleviated the PFOA-induced toxic effects in oocytes and improved the oocyte maturation and fertility upon PFOA exposure. Our findings discover new insights into PFOA-induced reproductive toxicity and provide NMN as a potential drug for POI therapy.


Subject(s)
Caprylates , Fluorocarbons , Mitochondria , NAD , Oocytes , Primary Ovarian Insufficiency , Zebrafish , Animals , Fluorocarbons/toxicity , Caprylates/toxicity , Female , Mitochondria/drug effects , Mitochondria/metabolism , Oocytes/drug effects , Oocytes/metabolism , Primary Ovarian Insufficiency/chemically induced , Primary Ovarian Insufficiency/metabolism , NAD/metabolism , NAD/biosynthesis , Reactive Oxygen Species/metabolism , Male , Apoptosis/drug effects , Ovary/drug effects , Ovary/metabolism , Membrane Potential, Mitochondrial/drug effects , Water Pollutants, Chemical/toxicity
5.
Cell Rep ; 43(7): 114379, 2024 Jul 23.
Article in English | MEDLINE | ID: mdl-38889006

ABSTRACT

The protein phosphatase 2A (PP2A) regulatory subunit PPP2R2A is involved in the regulation of immune response. We report that lupus-prone mice with T cells deficient in PPP2R2A display less autoimmunity and nephritis. PPP2R2A deficiency promotes NAD+ biosynthesis through the nicotinamide riboside (NR)-directed salvage pathway in T cells. NR inhibits murine Th17 and promotes Treg cell differentiation, in vitro, by PΑRylating histone H1.2 and causing its reduced occupancy in the Foxp3 loci and increased occupancy in the Il17a loci, leading to increased Foxp3 and decreased Il17a transcription. NR treatment suppresses disease in MRL.lpr mice and restores NAD+-dependent poly [ADP-ribose] polymerase 1 (PARP1) activity in CD4 T cells from patients with systemic lupus erythematosus (SLE), while reducing interferon (IFN)-γ and interleukin (IL)-17 production. We conclude that PPP2R2A controls the level of NAD+ through the NR-directed salvage pathway and promotes systemic autoimmunity. Translationally, NR suppresses lupus nephritis in mice and limits the production of proinflammatory cytokines by SLE T cells.


Subject(s)
Autoimmunity , Cell Differentiation , Lupus Erythematosus, Systemic , NAD , Protein Phosphatase 2 , Animals , Female , Humans , Mice , Forkhead Transcription Factors/metabolism , Histones/metabolism , Interferon-gamma/metabolism , Interleukin-17/metabolism , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/pathology , Lupus Nephritis/pathology , Lupus Nephritis/immunology , Lupus Nephritis/genetics , Lupus Nephritis/metabolism , Mice, Inbred C57BL , Mice, Inbred MRL lpr , NAD/metabolism , NAD/biosynthesis , Niacinamide/analogs & derivatives , Poly (ADP-Ribose) Polymerase-1/metabolism , Protein Phosphatase 2/metabolism , Pyridinium Compounds , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocytes, Regulatory/immunology , Th17 Cells/immunology , Th17 Cells/metabolism
6.
Antiviral Res ; 228: 105939, 2024 08.
Article in English | MEDLINE | ID: mdl-38909960

ABSTRACT

Viruses have developed sophisticated strategies to control metabolic activity of infected cells in order to supply replication machinery with energy and metabolites. Dengue virus (DENV), a mosquito-borne flavivirus responsible for dengue fever, is no exception. Previous reports have documented DENV interactions with metabolic pathways and shown in particular that glycolysis is increased in DENV-infected cells. However, underlying molecular mechanisms are still poorly characterized and dependence of DENV on this pathway has not been investigated in details yet. Here, we identified an interaction between the non-structural protein 3 (NS3) of DENV and glucokinase regulator protein (GCKR), a host protein that inhibits the liver-specific hexokinase GCK. NS3 expression was found to increase glucose consumption and lactate secretion in hepatic cell line expressing GCK. Interestingly, we observed that GCKR interaction with GCK decreases DENV replication, indicating the dependence of DENV to GCK activity and supporting the role of NS3 as an inhibitor of GCKR function. Accordingly, in the same cells, DENV replication both induces and depends on glycolysis. By targeting NAD(H) biosynthesis with the antimetabolite 6-Amino-Nicotinamide (6-AN), we decreased cellular glycolytic activity and inhibited DENV replication in hepatic cells. Infection of primary organotypic liver cultures (OLiC) from hamsters was also inhibited by 6-AN. Altogether, our results show that DENV has evolved strategies to control glycolysis in the liver, which could account for hepatic dysfunctions associated to infection. Besides, our findings suggest that lowering intracellular availability of NAD(H) could be a valuable therapeutic strategy to control glycolysis and inhibit DENV replication in the liver.


Subject(s)
Dengue Virus , Dengue , Glucokinase , Glycolysis , NAD , Viral Nonstructural Proteins , Virus Replication , Glycolysis/drug effects , Dengue Virus/drug effects , Glucokinase/metabolism , Glucokinase/antagonists & inhibitors , Humans , Virus Replication/drug effects , Viral Nonstructural Proteins/metabolism , Viral Nonstructural Proteins/genetics , Animals , Dengue/drug therapy , Dengue/virology , Dengue/metabolism , NAD/metabolism , NAD/biosynthesis , Cell Line , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , Glucose/metabolism , Liver/virology , Liver/metabolism , Antiviral Agents/pharmacology , Viral Proteases , Serine Endopeptidases , Nucleoside-Triphosphatase , DEAD-box RNA Helicases
7.
Nat Struct Mol Biol ; 31(9): 1413-1425, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38769389

ABSTRACT

NADH/NAD+ redox balance is pivotal for cellular metabolism. Systematic identification of NAD(H) redox regulators, although currently lacking, would help uncover unknown effectors critically implicated in the coordination of growth metabolism. In this study, we performed a genome-scale RNA interference (RNAi) screen to globally survey the genes involved in redox modulation and identified the HES family bHLH transcription factor HES4 as a negative regulator of NADH/NAD+ ratio. Functionally, HES4 is shown to be crucial for maintaining mitochondrial electron transport chain (ETC) activity and pyrimidine synthesis. More specifically, HES4 directly represses transcription of SLC44A2 and SDS, thereby inhibiting mitochondrial choline oxidation and cytosolic serine deamination, respectively, which, in turn, ensures coenzyme Q reduction capacity for DHODH-mediated UMP synthesis and serine-derived dTMP production. Accordingly, inhibition of choline oxidation preserves mitochondrial serine catabolism and ETC-coupled redox balance. Furthermore, HES4 protein stability is enhanced under EGFR activation, and increased HES4 levels facilitate EGFR-driven tumor growth and predict poor prognosis of lung adenocarcinoma. These findings illustrate an unidentified mechanism, underlying pyrimidine biosynthesis in the intersection between serine and choline catabolism, and underscore the physiological importance of HES4 in tumor metabolism.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors , Mitochondria , Oxidation-Reduction , Pyrimidines , RNA Interference , Humans , Pyrimidines/pharmacology , Pyrimidines/metabolism , Mitochondria/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Basic Helix-Loop-Helix Transcription Factors/genetics , Choline/metabolism , NAD/metabolism , NAD/biosynthesis , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Lung Neoplasms/genetics , Mice , Cell Line, Tumor , ErbB Receptors/metabolism , ErbB Receptors/genetics , Serine/metabolism , Adenocarcinoma of Lung/pathology , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/metabolism , Homeodomain Proteins/metabolism , Homeodomain Proteins/genetics , Repressor Proteins/metabolism , Repressor Proteins/genetics
8.
Am J Physiol Cell Physiol ; 326(5): C1423-C1436, 2024 05 01.
Article in English | MEDLINE | ID: mdl-38497113

ABSTRACT

Nicotinamide adenine dinucleotide (NAD+) is a pivotal coenzyme, essential for cellular reactions, metabolism, and mitochondrial function. Depletion of kidney NAD+ levels and reduced de novo NAD+ synthesis through the tryptophan-kynurenine pathway are linked to acute kidney injury (AKI), whereas augmenting NAD+ shows promise in reducing AKI. We investigated de novo NAD+ biosynthesis using in vitro, ex vivo, and in vivo models to understand its role in AKI. Two-dimensional (2-D) cultures of human primary renal proximal tubule epithelial cells (RPTECs) and HK-2 cells showed limited de novo NAD+ synthesis, likely due to low pathway enzyme gene expression. Using three-dimensional (3-D) spheroid culture model improved the expression of tubular-specific markers and enzymes involved in de novo NAD+ synthesis. However, de novo NAD+ synthesis remained elusive in the 3-D spheroid culture, regardless of injury conditions. Further investigation revealed that 3-D cultured cells could not metabolize tryptophan (Trp) beyond kynurenine (KYN). Intriguingly, supplementation of 3-hydroxyanthranilic acid into RPTEC spheroids was readily incorporated into NAD+. In a human precision-cut kidney slice (PCKS) ex vivo model, de novo NAD+ synthesis was limited due to substantially downregulated kynurenine 3-monooxygenase (KMO), which is responsible for KYN to 3-hydroxykynurenine conversion. KMO overexpression in RPTEC 3-D spheroids successfully reinstated de novo NAD+ synthesis from Trp. In addition, in vivo study demonstrated that de novo NAD+ synthesis is intact in the kidney of the healthy adult mice. Our findings highlight disrupted tryptophan-kynurenine NAD+ synthesis in in vitro cellular models and an ex vivo kidney model, primarily attributed to KMO downregulation.NEW & NOTEWORTHY Nicotinamide adenine dinucleotide (NAD+) is essential in regulating mitochondrial function. Reduced NAD+ synthesis through the de novo pathway is associated with acute kidney injury (AKI). Our study reveals a disruption in de novo NAD+ synthesis in proximal tubular models, but not in vivo, attributed to downregulation of enzyme kynurenine 3-monooxygenase (KMO). These findings highlight a crucial role of KMO in governing de novo NAD+ biosynthesis within the kidney, shedding light on potential AKI interventions.


Subject(s)
Epithelial Cells , Kidney Tubules, Proximal , Kynurenine 3-Monooxygenase , NAD , Tryptophan , Animals , Humans , Mice , Acute Kidney Injury/metabolism , Acute Kidney Injury/pathology , Acute Kidney Injury/enzymology , Cell Line , Cells, Cultured , Epithelial Cells/metabolism , Kidney Tubules, Proximal/metabolism , Kynurenine/metabolism , Kynurenine 3-Monooxygenase/metabolism , Kynurenine 3-Monooxygenase/genetics , Mice, Inbred C57BL , NAD/metabolism , NAD/biosynthesis , Tryptophan/metabolism
9.
Biochimie ; 220: 84-98, 2024 May.
Article in English | MEDLINE | ID: mdl-38182101

ABSTRACT

Nicotinamide Adenine Dinucleotide (NAD+), a coenzyme, is ubiquitously distributed and serves crucial functions in diverse biological processes, encompassing redox reactions, energy metabolism, and cellular signalling. This review article explores the intricate realm of NAD + metabolism, with a particular emphasis on the complex relationship between its structure, function, and the pivotal enzyme, Nicotinate Nucleotide Adenylyltransferase (NNAT), also known as nicotinate mononucleotide adenylyltransferase (NaMNAT), in the process of its biosynthesis. Our findings indicate that NAD + biosynthesis in humans and bacteria occurs via the same de novo synthesis route and the pyridine ring salvage pathway. Maintaining NAD homeostasis in bacteria is imperative, as most bacterial species cannot get NAD+ from their surroundings. However, due to lower sequence identity and structurally distant relationship of bacteria, including E. faecium and K. pneumonia, to its human counterpart, inhibiting NNAT, an indispensable enzyme implicated in NAD + biosynthesis, is a viable alternative in curtailing infections orchestrated by E. faecium and K. pneumonia. By merging empirical and computational discoveries and connecting the intricate NAD + metabolism network with NNAT's crucial role, it becomes clear that the synergistic effect of these insights may lead to a more profound understanding of the coenzyme's function and its potential applications in the fields of therapeutics and biotechnology.


Subject(s)
NAD , Nicotinamide-Nucleotide Adenylyltransferase , Nicotinamide-Nucleotide Adenylyltransferase/metabolism , Nicotinamide-Nucleotide Adenylyltransferase/chemistry , NAD/metabolism , NAD/biosynthesis , Humans , Bacterial Proteins/metabolism , Bacterial Proteins/chemistry
10.
Aging (Albany NY) ; 16(1): 106-128, 2023 12 28.
Article in English | MEDLINE | ID: mdl-38157259

ABSTRACT

BACKGROUND: Patients with chronic kidney disease (CKD) lack efficacious treatment. Jian-Pi-Yi-Shen formula (JPYSF) has demonstrated significant clinical efficacy in treating CKD for decades. However, its renoprotective mechanism has not been fully elucidated. This study aimed to determine whether JPYSF could delay renal fibrosis progression in CKD by restoring nicotinamide adenine dinucleotide (NAD+) biosynthesis. METHODS: Adenine-diet feeding was used to model CKD in C57BL/6 mice. JPYSF was orally administered for 4 weeks. Human proximal tubular epithelial cells (HK-2) cells were stimulated with transforming growth factor-ß1 (TGF-ß1) with or without JPYSF treatment. Renal function of mice was assessed by serum creatinine and blood urea nitrogen levels. Renal histopathological changes were assessed using Periodic acid-Schiff and Masson's trichrome staining. Cell viability was assessed using a cell counting kit-8 assay. NAD+ concentrations were detected by a NAD+/NADH assay kit. Western blotting, immunohistochemistry, and immunofluorescence were employed to examine fibrosis-related proteins and key NAD+ biosynthesis enzymes expression in the CKD kidney and TGF-ß1-induced HK-2 cells. RESULTS: JPYSF treatment could not only improve renal function and pathological injury but also inhibit renal fibrosis in CKD mice. Additionally, JPYSF reversed fibrotic response in TGF-ß1-induced HK-2 cells. Moreover, JPYSF rescued the decreased NAD+ content in CKD mice and TGF-ß1-induced HK-2 cells through restoring expression of key enzymes in NAD+ biosynthesis, including quinolinate phosphoribosyltransferase, nicotinamide mononucleotide adenylyltransferase 1, and nicotinamide riboside kinase 1. CONCLUSIONS: JPYSF alleviated renal fibrosis in CKD mice and reversed fibrotic response in TGF-ß1-induced HK-2 cells, which may be related to the restoration of NAD+ biosynthesis.


Subject(s)
NAD , Renal Insufficiency, Chronic , Animals , Humans , Mice , Fibrosis , Kidney/pathology , Mice, Inbred C57BL , NAD/biosynthesis , Renal Insufficiency, Chronic/metabolism , Transforming Growth Factor beta1/metabolism
11.
JCI Insight ; 7(1)2022 01 11.
Article in English | MEDLINE | ID: mdl-34793337

ABSTRACT

The biosynthetic routes leading to de novo nicotinamide adenine dinucleotide (NAD+) production are involved in acute kidney injury (AKI), with a critical role for quinolinate phosphoribosyl transferase (QPRT), a bottleneck enzyme of de novo NAD+ biosynthesis. The molecular mechanisms determining reduced QPRT in AKI, and the role of impaired NAD+ biosynthesis in the progression to chronic kidney disease (CKD), are unknown. We demonstrate that a high urinary quinolinate-to-tryptophan ratio, an indirect indicator of impaired QPRT activity and reduced de novo NAD+ biosynthesis in the kidney, is a clinically applicable early marker of AKI after cardiac surgery and is predictive of progression to CKD in kidney transplant recipients. We also provide evidence that the endoplasmic reticulum (ER) stress response may impair de novo NAD+ biosynthesis by repressing QPRT transcription. In conclusion, NAD+ biosynthesis impairment is an early event in AKI embedded with the ER stress response, and persistent reduction of QPRT expression is associated with AKI to CKD progression. This finding may lead to identification of noninvasive metabolic biomarkers of kidney injury with prognostic and therapeutic implications.


Subject(s)
Acute Kidney Injury/metabolism , Endoplasmic Reticulum Stress/physiology , Kidney/metabolism , NAD/biosynthesis , Animals , Cell Line , Male , Mice , Mice, Inbred C57BL , Pentosyltransferases/metabolism , Quinolinic Acid/urine , Tryptophan/urine
12.
Hum Exp Toxicol ; 40(12_suppl): S666-S675, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34752167

ABSTRACT

Background: Nicotinamide phosphoribosyltransferase (NAMPT) catalyzes the rate-limiting step in the salvage pathway of mammalian nicotinamide adenine dinucleotide (NAD+) biosynthesis. Through its NAD+-biosynthetic activity, NAMPT is able to regulate the development of hepatic steatosis and inflammation induced by diet or alcohol. However, the roles NAMPT plays in the development of liver fibrosis remain obscure. Purpose: To investigate the roles of NAMPT-mediated NAD+ biosynthesis in hepatic stellate cell (HSC) activation and liver fibrosis. Research Design: Realtime RT-PCR and western blot analyses were performed to analyze the expression of profibrogenic genes. Sirius red staining was conducted to examine the fibrosis in liver. Mouse liver fibrosis was induced by intraperitoneal injection of carbon tetrachloride (CCl4) 2 times a week for 6 weeks. Adenovirus-mediated NAMPT overexpression or nicotinamide mononucleotide (NMN) administration was carried out to study the effects of elevation of NAD+ levels on protecting CCl4-induced liver fibrosis in mice. LX2 cells or primary HSCs were used to study the role of NAMPT overexpression or NMN treatment in reducing profibrogenic gene expression in vitro. ResultsCCl4 administration suppresses NAMPT expression in liver and reduces hepatic NAD+ content. Tgfß1 treatment decreases intracellular NAD+ levels and NAMPT expression in LX2 cells. Adenovirus-mediated NAMPT overexpression augments liver NAD+ levels, inhibits HSC activation and alleviates CCl4-induced liver fibrosis in mice. Administration of NMN also suppresses HSC activation and protects against CCl4-induced liver fibrosis in mice. Conclusions: NAMPT-mediated NAD+ biosynthesis inhibits HSC activation and protects against CCl4-induced liver fibrosis.


Subject(s)
Carbon Tetrachloride Poisoning/complications , Hepatic Stellate Cells/metabolism , Liver Cirrhosis/etiology , NAD/biosynthesis , Nicotinamide Phosphoribosyltransferase/metabolism , Animals , Carbon Tetrachloride/toxicity , Carbon Tetrachloride Poisoning/prevention & control , Mice , Mice, Inbred C57BL
13.
Genes (Basel) ; 12(11)2021 11 09.
Article in English | MEDLINE | ID: mdl-34828382

ABSTRACT

Neurodegenerative diseases result in the progressive deterioration of the nervous system, with motor and cognitive impairments being the two most observable problems. Motor dysfunction could be caused by motor neuron diseases (MNDs) characterized by the loss of motor neurons, such as amyotrophic lateral sclerosis and Charcot-Marie-Tooth disease, or other neurodegenerative diseases with the destruction of brain areas that affect movement, such as Parkinson's disease and Huntington's disease. Nicotinamide adenine dinucleotide (NAD+) is one of the most abundant metabolites in the human body and is involved with numerous cellular processes, including energy metabolism, circadian clock, and DNA repair. NAD+ can be reversibly oxidized-reduced or directly consumed by NAD+-dependent proteins. NAD+ is synthesized in cells via three different paths: the de novo, Preiss-Handler, or NAD+ salvage pathways, with the salvage pathway being the primary producer of NAD+ in mammalian cells. NAD+ metabolism is being investigated for a role in the development of neurodegenerative diseases. In this review, we discuss cellular NAD+ homeostasis, looking at NAD+ biosynthesis and consumption, with a focus on the NAD+ salvage pathway. Then, we examine the research, including human clinical trials, focused on the involvement of NAD+ in MNDs and other neurodegenerative diseases with motor dysfunction.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , Charcot-Marie-Tooth Disease/metabolism , NAD/biosynthesis , Amyotrophic Lateral Sclerosis/genetics , Charcot-Marie-Tooth Disease/genetics , Circadian Clocks , Clinical Trials as Topic , DNA Repair , Energy Metabolism , Humans
14.
Sci Rep ; 11(1): 19648, 2021 10 04.
Article in English | MEDLINE | ID: mdl-34608200

ABSTRACT

The gut microbiota has tremendous potential to affect the host's health, in part by synthesizing vitamins and generating nutrients from food that is otherwise indigestible by the host. 1,5-Anhydro-D-fructose (1,5-AF) is a monosaccharide with a wide range of bioactive potentials, including anti-oxidant, anti-inflammatory, and anti-microbial effects. Based on its potential benefits and minimal toxicity, it is anticipated that 1,5-AF will be used as a dietary supplement to support general health. However, the effects of 1,5-AF on the gut microbiota are yet to be clarified. Here, using an unbiased metagenomic approach, we profiled the bacterial taxa and functional genes in the caecal microbiota of mice fed a diet containing either 2% 1,5-AF or a reference sweetener. Supplementation with 1,5-AF altered the composition of the gut microbiota, enriching the proportion of Faecalibacterium prausnitzii. 1,5-AF also altered the metabolomic profile of the gut microbiota, enriching genes associated with nicotinamide adenine dinucleotide biosynthesis. These findings support the potential benefits of 1,5-AF, but further studies are required to clarify the impact of 1,5-AF on health and disease.


Subject(s)
Fructose/analogs & derivatives , Gastrointestinal Microbiome , Animals , Diet , Dietary Supplements , Fructose/metabolism , Fructose/pharmacology , Gastrointestinal Microbiome/drug effects , Metagenome , Metagenomics/methods , Mice , NAD/biosynthesis , Nutrients/biosynthesis , Vitamins/biosynthesis
15.
Int J Mol Sci ; 22(19)2021 Sep 30.
Article in English | MEDLINE | ID: mdl-34638936

ABSTRACT

Nicotinamide adenine dinucleotide (NAD+) and its reduced form (NADH) are coenzymes employed in hundreds of metabolic reactions. NAD+ also serves as a substrate for enzymes such as sirtuins, poly(ADP-ribose) polymerases (PARPs) and ADP-ribosyl cyclases. Given the pivotal role of NAD(H) in health and disease, studying NAD+ metabolism has become essential to monitor genetic- and/or drug-induced perturbations related to metabolic status and diseases (such as ageing, cancer or obesity), and its possible therapies. Here, we present a strategy based on liquid chromatography-tandem mass spectrometry (LC-MS/MS), for the analysis of the NAD+ metabolome in biological samples. In this method, hydrophilic interaction chromatography (HILIC) was used to separate a total of 18 metabolites belonging to pathways leading to NAD+ biosynthesis, including precursors, intermediates and catabolites. As redox cofactors are known for their instability, a sample preparation procedure was developed to handle a variety of biological matrices: cell models, rodent tissues and biofluids, as well as human biofluids (urine, plasma, serum, whole blood). For clinical applications, quantitative LC-MS/MS for a subset of metabolites was demonstrated for the analysis of the human whole blood of nine volunteers. Using this developed workflow, our methodology allows studying NAD+ biology from mechanistic to clinical applications.


Subject(s)
Metabolome , NAD/biosynthesis , Plasma/metabolism , Serum/metabolism , Tandem Mass Spectrometry/methods , Urine/physiology , Animals , Blood Donors , Chromatography, Liquid/methods , Hep G2 Cells , Humans , Hydrophobic and Hydrophilic Interactions , Metabolomics/methods , Mice , Mice, Inbred C57BL , Monitoring, Physiologic/methods , Oxidation-Reduction , Pilot Projects , Plasma/chemistry , Serum/chemistry , Urine/chemistry
16.
Nutrients ; 13(10)2021 Sep 28.
Article in English | MEDLINE | ID: mdl-34684434

ABSTRACT

Despite the progress in the development of new anticancer strategies, cancer is rapidly spreading around the world and remains one of the most common diseases. For more than 40 years, doxorubicin has been widely used in the treatment of solid and hematological tumors. At the same time, the problem of its cardiotoxicity remains unresolved, despite the high efficiency of this drug. Symptomatic therapy is used as a treatment for side-effects of doxorubicin or pathological conditions that have already appeared in their background. To date, there are no treatment methods for doxorubicin cardiomyopathy as such. A drug such as nicotinamide riboside can play an important role in solving this problem. Nicotinamide riboside is a pyridine nucleoside similar to vitamin B3 that acts as a precursor to NAD+. There is no published research on nicotinamide riboside effects on cardiomyopathy, despite the abundance of works devoted to the mechanisms of its effects in various pathologies. The review analyzes information about the effects of nicotinamide riboside on various experimental models of pathologies, its role in the synthesis of NAD+, and also considers the possibility and prospects of its use for the prevention of doxorubicin cardiomyopathy.


Subject(s)
Antibiotics, Antineoplastic/adverse effects , Cardiomyopathies/drug therapy , Cardiomyopathies/etiology , Cardiotonic Agents/therapeutic use , Doxorubicin/adverse effects , Niacinamide/analogs & derivatives , Pyridinium Compounds/therapeutic use , Animals , Biomarkers , Cardiomyopathies/metabolism , Cardiomyopathies/prevention & control , Cardiotonic Agents/pharmacology , Cardiotoxicity/drug therapy , Cardiotoxicity/etiology , Cardiotoxicity/prevention & control , Disease Management , Disease Models, Animal , Disease Susceptibility , Humans , Metabolic Networks and Pathways , NAD/biosynthesis , Niacinamide/pharmacology , Niacinamide/therapeutic use , Oxidative Stress/drug effects , Pyridinium Compounds/pharmacology , Signal Transduction/drug effects , Sirtuins/metabolism
17.
Mech Ageing Dev ; 199: 111567, 2021 10.
Article in English | MEDLINE | ID: mdl-34517020

ABSTRACT

NAD+ is a fundamental molecule in human life and health as it participates in energy metabolism, cell signalling, mitochondrial homeostasis, and in dictating cell survival or death. Emerging evidence from preclinical and human studies indicates an age-dependent reduction of cellular NAD+, possibly due to reduced synthesis and increased consumption. In preclinical models, NAD+ repletion extends healthspan and / or lifespan and mitigates several conditions, such as premature ageing diseases and neurodegenerative diseases. These findings suggest that NAD+ replenishment through NAD+ precursors has great potential as a therapeutic target for ageing and age-predisposed diseases, such as Alzheimer's disease. Here, we provide an updated review on the biological activity, safety, and possible side effects of NAD+ precursors in preclinical and clinical studies. Major NAD+ precursors focused on by this review are nicotinamide riboside (NR), nicotinamide mononucleotide (NMN), and the new discovered dihydronicotinamide riboside (NRH). In summary, NAD+ precursors have an exciting therapeutic potential for ageing, metabolic and neurodegenerative diseases.


Subject(s)
Aging , Alzheimer Disease/drug therapy , NAD , Niacinamide/analogs & derivatives , Nicotinamide Mononucleotide/pharmacology , Pyridinium Compounds/pharmacology , Aging/drug effects , Aging/metabolism , Cell Survival/physiology , Drug Development , Energy Metabolism/physiology , Humans , NAD/biosynthesis , NAD/metabolism , Niacinamide/metabolism , Niacinamide/pharmacology , Nicotinamide Mononucleotide/metabolism , Pyridinium Compounds/metabolism , Signal Transduction/physiology
18.
Mech Ageing Dev ; 199: 111569, 2021 10.
Article in English | MEDLINE | ID: mdl-34509469

ABSTRACT

Nicotinamide adenine dinucleotide (NAD+) is a vital coenzyme in redox reactions. NAD+ is also important in cellular signalling as it is consumed by PARPs, SARM1, sirtuins and CD38. Cellular NAD+ levels regulate several essential processes including DNA repair, immune cell function, senescence, and chromatin remodelling. Maintenance of these cellular processes is important for healthy ageing and lifespan. Interestingly, the levels of NAD+ decline during ageing in several organisms, including humans. Declining NAD+ levels have been linked to several age-related diseases including various metabolic diseases and cognitive decline. Decreasing tissue NAD+ concentrations have been ascribed to an imbalance between biosynthesis and consumption of the dinucleotide, resulting from, for instance, reduced levels of the rate limiting enzyme NAMPT along with an increased activation state of the NAD+-consuming enzymes PARPs and CD38. The progression of some age-related diseases can be halted or reversed by therapeutic augmentation of NAD+ levels. NAD+ metabolism has therefore emerged as a potential target to ameliorate age-related diseases. The present review explores how ageing affects NAD+ metabolism and current approaches to reverse the age-dependent decline of NAD+.


Subject(s)
ADP-ribosyl Cyclase 1/metabolism , Aging , Armadillo Domain Proteins/metabolism , Cytoskeletal Proteins/metabolism , NAD , Poly(ADP-ribose) Polymerases/metabolism , Sirtuins/metabolism , Aging/drug effects , Aging/physiology , Animals , Cognitive Dysfunction/metabolism , Cognitive Dysfunction/therapy , Drug Discovery , Humans , Metabolic Diseases/metabolism , Metabolic Diseases/therapy , NAD/biosynthesis , NAD/metabolism , Oxidation-Reduction , Signal Transduction
19.
Proc Natl Acad Sci U S A ; 118(31)2021 08 03.
Article in English | MEDLINE | ID: mdl-34330829

ABSTRACT

The evolutionary context of why caloric restriction (CR) activates physiological mechanisms that slow the process of aging remains unclear. The main goal of this analysis was to identify, using metabolomics, the common pathways that are modulated across multiple tissues (brown adipose tissue, liver, plasma, and brain) to evaluate two alternative evolutionary models: the "disposable soma" and "clean cupboards" ideas. Across the four tissues, we identified more than 10,000 different metabolic features. CR altered the metabolome in a graded fashion. More restriction led to more changes. Most changes, however, were tissue specific, and in some cases, metabolites changed in opposite directions in different tissues. Only 38 common metabolic features responded to restriction in the same way across all four tissues. Fifty percent of the common altered metabolites were carboxylic acids and derivatives, as well as lipids and lipid-like molecules. The top five modulated canonical pathways were l-carnitine biosynthesis, NAD (nicotinamide adenine dinucleotide) biosynthesis from 2-amino-3-carboxymuconate semialdehyde, S-methyl-5'-thioadenosine degradation II, NAD biosynthesis II (from tryptophan), and transfer RNA (tRNA) charging. Although some pathways were modulated in common across tissues, none of these reflected somatic protection, and each tissue invoked its own idiosyncratic modulation of pathways to cope with the reduction in incoming energy. Consequently, this study provides greater support for the clean cupboards hypothesis than the disposable soma interpretation.


Subject(s)
Caloric Restriction , Carnitine/biosynthesis , Energy Metabolism/physiology , NAD/biosynthesis , RNA, Transfer/metabolism , Animal Feed/analysis , Animals , Diet/veterinary , Gene Expression Regulation/physiology , Male , Mice , Mice, Inbred C57BL , RNA, Transfer/genetics , Random Allocation , Tissue Distribution
20.
Mech Ageing Dev ; 198: 111545, 2021 09.
Article in English | MEDLINE | ID: mdl-34302821

ABSTRACT

Strategies to correct declining nicotinamide adenine dinucleotide (NAD+) levels in neurological disease and biological ageing are promising therapeutic candidates. These strategies include supplementing with NAD+ precursors, small molecule activation of NAD+ biosynthetic enzymes, and treatment with small molecule inhibitors of NAD+ consuming enzymes such as CD38, SARM1 or members of the PARP family. While these strategies have shown efficacy in animal models of neurological disease, each of these has the mechanistic potential for adverse events that could preclude their preclinical use. Here, we discuss the implications of these strategies for treating neurological diseases, including potential off-target effects that may be unique to the brain.


Subject(s)
Aging , Molecular Targeted Therapy , NAD , Nervous System Diseases , Aging/drug effects , Aging/metabolism , Animals , Biosynthetic Pathways/drug effects , Biosynthetic Pathways/physiology , Humans , Molecular Targeted Therapy/adverse effects , Molecular Targeted Therapy/methods , NAD/biosynthesis , NAD/metabolism , Nervous System Diseases/enzymology , Nervous System Diseases/therapy , Risk Assessment
SELECTION OF CITATIONS
SEARCH DETAIL