Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 598
Filter
1.
Cell Death Dis ; 15(10): 744, 2024 Oct 11.
Article in English | MEDLINE | ID: mdl-39394148

ABSTRACT

Extensive studies indicate that mitochondria dysfunction is pivotal for Alzheimer's disease (AD) pathogenesis; while cumulative evidence suggests that increased mitochondrial stress response (MSR) may mitigate neurodegeneration in AD, explorations to develop a MSR-targeted therapeutic strategy against AD are scarce. We combined cell biology, molecular biology, and pharmacological approaches to unravel a novel molecular pathway by which NAD+-boosting agent nicotinamide mononucleotide (NMN) regulates MSR in AD models. Here, we report dyshomeostasis plasma UPRmt-mitophagy-mediated MSR profiles in AD patient samples. NMN restores NAD+ metabolic profiles and improves MSR through the ATF4-dependent UPRmt pathway in AD-related cross-species models. At the organismal level, NAD+ repletion with NMN supplementation ameliorates mitochondrial proteotoxicity, decreases hippocampal synaptic disruption, decreases neuronal loss, and brain atrophy in mice model of AD. Remarkably, omics features of the hippocampus with NMN show that NMN leads to transcriptional changes of genes and proteins involved in MSR characteristics, principally within the astrocyte unit rather than microglia and oligodendrocytes. In brief, our work provides evidence that MSR has an active role in the pathogenesis of AD, as reducing mitochondrial homeostasis via atf4 depletion in AD mice aggravates the hallmarks of the disease; conversely, bolstering mitochondrial proteostasis by NMN decreases protein aggregation, restores memory performance, and delays disease progression, ultimately translating to increased healthspan.


Subject(s)
Activating Transcription Factor 4 , Alzheimer Disease , Mitochondria , NAD , Nicotinamide Mononucleotide , Unfolded Protein Response , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Animals , Mitochondria/metabolism , Mitochondria/drug effects , Activating Transcription Factor 4/metabolism , Humans , Mice , Nicotinamide Mononucleotide/pharmacology , NAD/metabolism , Unfolded Protein Response/drug effects , Disease Models, Animal , Hippocampus/metabolism , Hippocampus/drug effects , Hippocampus/pathology , Male , Mice, Inbred C57BL
2.
Theranostics ; 14(15): 5999-6015, 2024.
Article in English | MEDLINE | ID: mdl-39346542

ABSTRACT

The process of skeletal regeneration initiated by stem cells following injury, especially in fractures, is significantly impaired by aging and adverse factors. Nicotinamide mononucleotide (NMN), a critical endogenous precursor of nicotinamide adenine dinucleotide (NAD), has garnered extensive attention for its multifaceted regulatory functions in living organisms and its wide-ranging therapeutic potential. However, whether NMN contributes to trauma-induced skeletal regeneration remains unclear. Methods: The transverse femoral shaft fracture model was employed to evaluate the potential advantages of NMN administration for overall repair during the initial fracture stages in male mice through micro-CT analysis, histochemistry, and biomechanical testing. The pro-proliferative function of NMN on skeletal stem cells (SSCs) was investigated through flow cytometry, qRT-PCR, NAD content measurement, and cell proliferation assay. Results: In this study, we observed that the administration of NMN during the initial phase of fracture in mice led to a larger callus and corresponding improvement in micro-CT parameters. NMN enhances the cartilaginous component of the callus by elevating the NAD content, consequently accelerating subsequent endochondral ossification and the fracture healing process. Subsequent analyses elucidated that NMN was beneficial in promoting the expansion of diverse stem cells in vivo and in vitro potentially via modulation of the Notch signaling pathway. Moreover, the depletion of macrophages profoundly obstructs the proliferation of SSCs. Conclusion: Our discoveries provide a potential strategy for enhancing fracture healing through stimulation of callus SSC proliferation at an early stage, shedding light on the translational value of NMN as an enhancer for skeletal regeneration and highlighting the pivotal role of macrophage-stem cell interactions in governing the regenerative influence of NMN on stem cells.


Subject(s)
Cell Proliferation , Fracture Healing , Nicotinamide Mononucleotide , Animals , Mice , Cell Proliferation/drug effects , Fracture Healing/drug effects , Nicotinamide Mononucleotide/pharmacology , Male , Stem Cells/drug effects , Stem Cells/metabolism , X-Ray Microtomography , Osteogenesis/drug effects , Disease Models, Animal , Femoral Fractures/drug therapy , Femoral Fractures/pathology , Mice, Inbred C57BL , Signal Transduction/drug effects , Bony Callus/drug effects , Bone Regeneration/drug effects , NAD/metabolism
3.
Biochem Biophys Res Commun ; 733: 150623, 2024 Nov 12.
Article in English | MEDLINE | ID: mdl-39255619

ABSTRACT

After prolonged space operations, astronauts showed maladaptive atrophy within mostly left-ventricular myocardium, resulting in cardiac dysfunction. However, the mechanism of cardiac dysfunction under microgravity conditions is unclear, and the relevant prevention and treatment measures also need to be explored. Through simulating the microgravity environment with a tail suspension (TS) model, we found that long-term exposure to microgravity promotes aging of mouse hearts, which is closely related to cardiac dysfunction. The intravenous administration of adipose-derived mesenchymal stem cells (ADSCs) emerged preventive and therapeutic effect against myocardial senescence and the decline in cardiac function. Plasma metabolomics analysis suggests the loss of NAD+ in TS mice and motivated myocardial NAD + metabolism and utilization in ADSCs-treated mice, likely accounting for ADSCs' function. Oral administration of nicotinamide mononucleotide (NMN, a NAD + precursor) showed similar therapeutic effect to ADSCs treatment. Collectively, these data implicate the effect of ADSCs in microgravity-induced cardiac dysfunction and provide new therapeutic ideas for aging-related maladaptive cardiac remodeling.


Subject(s)
Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Mice, Inbred C57BL , Myocardium , NAD , Weightlessness , Animals , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/cytology , NAD/metabolism , Weightlessness/adverse effects , Myocardium/metabolism , Myocardium/pathology , Mice , Mesenchymal Stem Cell Transplantation/methods , Male , Nicotinamide Mononucleotide/pharmacology , Nicotinamide Mononucleotide/metabolism , Hindlimb Suspension/adverse effects , Aging/metabolism , Cellular Senescence/drug effects , Heart Diseases/metabolism , Heart Diseases/etiology , Heart Diseases/pathology , Heart Diseases/therapy , Heart Diseases/prevention & control
4.
J Zhejiang Univ Sci B ; 25(9): 723-735, 2024 Sep 15.
Article in English, Chinese | MEDLINE | ID: mdl-39308064

ABSTRACT

Aging and age-related ailments have emerged as critical challenges and great burdens within the global contemporary society. Addressing these concerns is an imperative task, with the aims of postponing the aging process and finding effective treatments for age-related degenerative diseases. Recent investigations have highlighted the significant roles of nicotinamide adenine dinucleotide (NAD+) in the realm of anti-aging. It has been empirically evidenced that supplementation with nicotinamide mononucleotide (NMN) can elevate NAD+ levels in the body, thereby ameliorating certain age-related degenerative diseases. The principal anti-aging mechanisms of NMN essentially lie in its impact on cellular energy metabolism, inhibition of cell apoptosis, modulation of immune function, and preservation of genomic stability, which collectively contribute to the deferral of the aging process. This paper critically reviews and evaluates existing research on the anti-aging mechanisms of NMN, elucidates the inherent limitations of current research, and proposes novel avenues for anti-aging investigations.


Subject(s)
Aging , NAD , Nicotinamide Mononucleotide , Nicotinamide Mononucleotide/pharmacology , Humans , Aging/drug effects , NAD/metabolism , Animals , Apoptosis/drug effects , Energy Metabolism/drug effects , Genomic Instability/drug effects
5.
Int J Mol Sci ; 25(17)2024 Sep 02.
Article in English | MEDLINE | ID: mdl-39273473

ABSTRACT

Nicotinamide adenine dinucleotide (NAD+) is an essential metabolite that plays a crucial role in diverse biological processes, including energy metabolism, gene expression, DNA repair, and mitochondrial function. An aberrant NAD+ level mediates the development of cardiovascular dysfunction and diseases. Both in vivo and in vitro studies have demonstrated that nicotinamide mononucleotide (NMN), as a NAD+ precursor, alleviates the development of cardiovascular diseases such as heart failure, atherosclerosis, and myocardial ischemia/reperfusion injury. Importantly, NMN has suggested pharmacological activities mostly through its involvement in NAD+ biosynthesis. Several clinical studies have been conducted to investigate the efficacy and safety of NMN supplementation, indicating its potential role in cardiovascular protection without significant adverse effects. In this review, we systematically summarize the impact of NMN as a nutraceutical and potential therapeutic drug on cardiovascular diseases and emphasize the correlation between NMN supplementation and cardiovascular protection.


Subject(s)
Cardiovascular Diseases , Nicotinamide Mononucleotide , Humans , Nicotinamide Mononucleotide/therapeutic use , Nicotinamide Mononucleotide/metabolism , Nicotinamide Mononucleotide/pharmacology , Cardiovascular Diseases/metabolism , Cardiovascular Diseases/drug therapy , Animals , Dietary Supplements , NAD/metabolism
6.
Anal Chim Acta ; 1325: 343074, 2024 Oct 09.
Article in English | MEDLINE | ID: mdl-39244299

ABSTRACT

BACKGROUND: ß-nicotinamide mononucleotide stands out as an essential breakthrough in "anti-aging" and consistently leads the list of top-selling nutritional supplements in terms of quantity. As the metabolites of ß-nicotinamide mononucleotide, the detection of nicotinamide and N1-methylnicotinamide is of great significance for evaluating the nutritional effect of dietary supplements of ß-nicotinamide mononucleotide. However, due to the extremely low concentration of nicotinamide and N1-methylnicotinamide in vivo and the serious matrix interference in biological samples, there is an increasing demand for materials and methods of pre-treatment. RESULTS: In this study, Fe3O4@hydroxypropyl methyl cellulose@dodecylbenzenesulfonic acid magnetic fluid was synthesized for the first time, and it was used as pretreatment material to detect nicotinamide and N1- methylnicotinamide in urine samples by high performance liquid chromatography. Compared with other adsorption materials, Fe3O4@hydroxypropyl methyl cellulose@dodecylbenzenesulfonic acid nanoparticles are a kind of uniform magnetic fluid. Furthermore, they have more types and quantities of interaction sites (electrostatic interactions, hydrophobic interactions, hydrogen bonding interactions, and π-π interactions), so they own greater adsorption capacity. When the pH of the solution is 4, they can be adsorbed quickly within 10 s. The method successfully detected trace nicotinamide and N1-methylnicotinamide in urine samples in the linear range of 0.1-80 µg mL-1, the low detection limits were 0.30 ng mL-1 and 1.5 ng mL-1 respectively, and the quantification limits were 1.0 ng mL-1 and 5.0 ng mL-1, respectively. At the same time, the standard urine samples of nicotinamide and N1-methylnicotinamide showed satisfactory recovery rate 94.50-109.1 %. The results indicated that the established method can accurately and quantitatively determine trace nicotinamide and N1-methylnicotinamide in urine samples. SIGNIFICANCE: Consequently, low concentration of ß-nicotinamide mononucleotide metabolites can be detected simultaneously, and the interference can be eliminated during the detection process, which provides an efficient and convenient pretreatment method and a rapid and sensitive detection method for the analysis of ß-nicotinamide mononucleotide metabolites. What's more, it has a wide application prospect in the analysis of other similar metabolites in biological samples.


Subject(s)
Niacinamide , Nicotinamide Mononucleotide , Nicotinamide Mononucleotide/chemistry , Nicotinamide Mononucleotide/urine , Nicotinamide Mononucleotide/metabolism , Humans , Niacinamide/urine , Niacinamide/analogs & derivatives , Niacinamide/metabolism , Niacinamide/analysis , Niacinamide/chemistry , Chromatography, High Pressure Liquid , Magnetite Nanoparticles/chemistry , Adsorption , Limit of Detection
7.
Sci Rep ; 14(1): 19392, 2024 08 20.
Article in English | MEDLINE | ID: mdl-39169144

ABSTRACT

Cuproptosis is characterized by lipoylated protein aggregation and loss of iron-sulfur (Fe-S) proteins, which are crucial for a wide range of important cellular functions, including DNA replication and damage repair. Sirt2 and sirt4 are lipoamidases that remove the lipoyl moiety from lipoylated proteins using nicotinamide adenine dinucleotide (NAD+) as a cofactor. However, to date, it is not clear whether nicotinamide mononucleotide (NMN), a precursor of NAD+, affects cellular sensitivity to cuproptosis. Therefore, in the current study, cuproptosis was induced by the copper (Cu) ionophore elesclomol (Es) in HeLa cells. It was also found that Es/Cu treatment increased cellular DNA damage level. On the other hand, NMN treatment partially rescued cuproptosis in a dose-dependent manner, as well as reduced cellular DNA damage level. In addition, NMN upregulated the expression of Fe-S protein POLD1, without affecting the aggregation of lipoylated proteins. Mechanistic study revealed that NMN increased the expression of sirt2 and cellular reduced nicotinamide adenine dinucleotide phosphate (NADPH) level. Overexpression of sirt2 and sirt4 did not change the aggregation of lipoylated proteins, however, sirt2, but not sirt4, increased cellular NADPH levels and partially rescued cuproptosis. Inhibition of NAD+ kinase (NADK), which is responsible for generating NADPH, abolished the rescuing function of NMN and sirt2 for Es/Cu induced cell death. Taken together, our results suggested that DNA damage is a characteristic feature of cuproptosis. NMN can partially rescue cuproptosis by upregulating sirt2, increase intracellular NADPH content and maintain the level of Fe-S proteins, independent of the lipoamidase activity of sirt2.


Subject(s)
DNA Damage , NADP , Nicotinamide Mononucleotide , Sirtuin 2 , Up-Regulation , Humans , Sirtuin 2/metabolism , Sirtuin 2/genetics , HeLa Cells , NADP/metabolism , DNA Damage/drug effects , Up-Regulation/drug effects , Nicotinamide Mononucleotide/pharmacology , Nicotinamide Mononucleotide/metabolism , Copper/pharmacology , Copper/metabolism , Sirtuins/metabolism
8.
JCI Insight ; 9(16)2024 Jul 18.
Article in English | MEDLINE | ID: mdl-39171530

ABSTRACT

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


Subject(s)
Disease Models, Animal , Frataxin , Friedreich Ataxia , Iron-Binding Proteins , NAD , Animals , Friedreich Ataxia/metabolism , Friedreich Ataxia/pathology , Friedreich Ataxia/genetics , Iron-Binding Proteins/genetics , Iron-Binding Proteins/metabolism , Mice , Humans , NAD/metabolism , Phenotype , Male , Cardiomegaly/metabolism , Cardiomegaly/pathology , Nicotinamide Mononucleotide/pharmacology , Niacinamide/analogs & derivatives , Niacinamide/pharmacology , Female , Gene Knockdown Techniques , Pyridinium Compounds , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology
9.
EMBO Mol Med ; 16(10): 2583-2618, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39169162

ABSTRACT

Chemotherapy induced ovarian failure and infertility is an important concern in female cancer patients of reproductive age or younger, and non-invasive, pharmacological approaches to maintain ovarian function are urgently needed. Given the role of reduced nicotinamide adenine dinucleotide phosphate (NADPH) as an essential cofactor for drug detoxification, we sought to test whether boosting the NAD(P)+ metabolome could protect ovarian function. We show that pharmacological or transgenic strategies to replenish the NAD+ metabolome ameliorates chemotherapy induced female infertility in mice, as measured by oocyte yield, follicle health, and functional breeding trials. Importantly, treatment of a triple-negative breast cancer mouse model with the NAD+ precursor nicotinamide mononucleotide (NMN) reduced tumour growth and did not impair the efficacy of chemotherapy drugs in vivo or in diverse cancer cell lines. Overall, these findings raise the possibility that NAD+ precursors could be a non-invasive strategy for maintaining ovarian function in cancer patients, with potential benefits in cancer therapy.


Subject(s)
Antineoplastic Agents , Metabolome , Animals , Female , Mice , Metabolome/drug effects , Humans , Antineoplastic Agents/pharmacology , NADP/metabolism , Nicotinamide Mononucleotide/pharmacology , Nicotinamide Mononucleotide/metabolism , NAD/metabolism , Cell Line, Tumor , Infertility, Female/drug therapy , Infertility, Female/metabolism , Disease Models, Animal , Ovary/drug effects , Ovary/metabolism , Ovary/pathology , Fertility/drug effects
10.
Biotechnol J ; 19(8): e2400311, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39167557

ABSTRACT

In the previous study, the culture medium was treated with nicotinamide adenine dinucleotide (NAD+) under the hypothesis that NAD+ regeneration is a major factor causing excessive lactate accumulation in Chinese hamster ovary (CHO) cells. The NAD+ treatment improved metabolism by not only reducing the Warburg effect but also enhancing oxidative phosphorylation, leading to enhanced antibody production. Building on this, four NAD+ precursors - nicotinamide mononucleotide (NMN), nicotinic acid (NA), nicotinamide riboside (NR), and nicotinamide (NAM) - were tested to elevate intracellular NAD+ levels more economically. First, the ability of CHO cells to utilize both the salvage and Preiss-Handler pathways for NAD+ biosynthesis was verified, and then the effect of NAD+ precursors on CHO cell cultures was evaluated. These precursors increased intracellular NAD+ levels by up to 70.6% compared to the non-treated group. Culture analysis confirmed that all the precursors induced metabolic changes and that NMN, NA, and NR improved productivity akin to NAD+ treatment, with comparable integral viable cell density. Despite the positive effects such as the increase in the specific productivity and changes in cellular glucose metabolism, none of the precursors surpassed direct NAD+ treatment in antibody titer, presumably due to the reduction in nucleoside availability, as evidenced by the decrease in ATP levels in the NAD+ precursor-treated groups. These results underscore the complexity of cellular metabolism as well as the necessity for further investigation to optimize NAD+ precursor treatment strategies, potentially with the supplementation of nucleoside precursors. Our findings suggest a feasible approach for improving CHO cell culture performances by using NAD+ precursors as medium and feed components for the biopharmaceutical production.


Subject(s)
Cricetulus , NAD , Niacinamide , CHO Cells , Animals , NAD/metabolism , Niacinamide/metabolism , Niacinamide/analogs & derivatives , Culture Media/chemistry , Culture Media/metabolism , Nicotinamide Mononucleotide/metabolism , Niacin/metabolism , Pyridinium Compounds/metabolism , Cricetinae , Cell Culture Techniques/methods , Antibodies, Monoclonal/metabolism , Glucose/metabolism
11.
Cell Biochem Funct ; 42(5): e4087, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38953407

ABSTRACT

ß-Nicotinamide mononucleotide (NMN) is a biologically active nucleotide that regulates the physiological metabolism of the body by rapidly increasing nicotinamide adenine dinucleotide (NAD+). To determine the safety and biological activity of NMN resources, we constructed a recombinant strain of P. pastoris that heterologously expresses nicotinamide-phosphate ribosyltransferase (NAMPT), and subsequently catalyzed and purified the expressed product to obtain NMN. Consequently, this study established a high-fat diet (HFD) obese model to investigate the lipid-lowering activity of NMN. The findings showed that NMN supplementation directly increased the NAD+ levels, and reduced HFD-induced liver injury and lipid deposition. NMN treatment significantly decreased total cholesterol (TC) and triglyceride (TG) in serum and liver, as well as alanine aminotransferase (ALT) and insulin levels in serum (p < .05 or p < .01). In conclusion, this study combined synthetic biology with nutritional evaluation to confirm that P. pastoris-generated NMN modulated lipid metabolism in HFD mice, offering a theoretical framework and evidence for the application of microbially created NMN.


Subject(s)
Diet, High-Fat , Lipid Metabolism , Liver , Mice, Inbred C57BL , Nicotinamide Mononucleotide , Animals , Nicotinamide Mononucleotide/metabolism , Nicotinamide Mononucleotide/pharmacology , Lipid Metabolism/drug effects , Mice , Liver/metabolism , Male , Nicotinamide Phosphoribosyltransferase/metabolism
12.
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
13.
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
14.
Nat Rev Mol Cell Biol ; 25(10): 822-840, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39026037

ABSTRACT

Nicotinamide adenine dinucleotide, in its oxidized (NAD+) and reduced (NADH) forms, is a reduction-oxidation (redox) co-factor and substrate for signalling enzymes that have essential roles in metabolism. The recognition that NAD+ levels fall in response to stress and can be readily replenished through supplementation has fostered great interest in the potential benefits of increasing or restoring NAD+ levels in humans to prevent or delay diseases and degenerative processes. However, much about the biology of NAD+ and related molecules remains poorly understood. In this Review, we discuss the current knowledge of NAD+ metabolism, including limitations of, assumptions about and unappreciated factors that might influence the success or contribute to risks of NAD+ supplementation. We highlight several ongoing controversies in the field, and discuss the role of the microbiome in modulating the availability of NAD+ precursors such as nicotinamide riboside (NR) and nicotinamide mononucleotide (NMN), the presence of multiple cellular compartments that have distinct pools of NAD+ and NADH, and non-canonical NAD+ and NADH degradation pathways. We conclude that a substantial investment in understanding the fundamental biology of NAD+, its detection and its metabolites in specific cells and cellular compartments is needed to support current translational efforts to safely boost NAD+ levels in humans.


Subject(s)
NAD , NAD/metabolism , Humans , Animals , Oxidation-Reduction , Niacinamide/metabolism , Niacinamide/analogs & derivatives , Nicotinamide Mononucleotide/metabolism , Pyridinium Compounds
15.
Enzyme Microb Technol ; 180: 110482, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39059289

ABSTRACT

ß-nicotinamide mononucleotide (ß-NMN) is a key precursor of nicotinamide adenine dinucleotide, and becomes attractive in the nutrition and health care fields, but its enzymatic synthesis is expensive. In this study, a six-enzyme cascade catalytic system was constructed to produce ß-NMN. Using D-ribose and nicotinamide as substrates, the ß-NMN yield reached 97.5 % catalyzed by purified enzymes. Then, after knocking out the genes encoding proteins that consume ß-NMN in E. coli BL21(DE3), the similar ß-NMN yield, 97.2 %, using the crude enzymes could be also obtained. After that, ß-NMN synthesis was performed under increased substrate concentration, and 'modular' crude enzymes cascade catalytic reaction system was proposed to reduce the inhibition of polyphosphate on ribose-phosphate diphosphokinase activity, and the ß-NMN yield reached 78.4 % at 10 mM D-ribose, which is 1.82 times of that in 'one-pot' reaction and represents the highest ß-NMN preparation level with phosphoribosylpyrophosphate as the core reported till now.


Subject(s)
Escherichia coli , Nicotinamide Mononucleotide , Escherichia coli/genetics , Escherichia coli/enzymology , Escherichia coli/metabolism , Nicotinamide Mononucleotide/metabolism , Ribose/metabolism , Biocatalysis , Niacinamide/metabolism , Niacinamide/chemistry , Escherichia coli Proteins/metabolism , Escherichia coli Proteins/genetics , Escherichia coli Proteins/chemistry , Catalysis , Substrate Specificity
16.
Neurochem Res ; 49(10): 2888-2896, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39037560

ABSTRACT

With the aging global population, Alzheimer's disease (AD) has become a significant social and economic burden, necessitating the development of novel therapeutic strategies. This study investigates the therapeutic potential of nicotinamide mononucleotide (NMN) synbiotics, a combination of NMN, Lactiplantibacillus plantarum CGMCC 1.16089, and lactulose, in mitigating AD pathology. APP/PS1 mice were supplemented with NMN synbiotics and compared against control groups. The effects on amyloid-ß (Aß) deposition, intestinal histopathology, tight junction proteins, inflammatory cytokines, and reactive oxygen species (ROS) levels were assessed. NMN synbiotics intervention significantly reduced Aß deposition in the cerebral cortex and hippocampus by 67% and 60%, respectively. It also ameliorated histopathological changes in the colon, reducing crypt depth and restoring goblet cell numbers. The expression of tight junction proteins Claudin-1 and ZO-1 was significantly upregulated, enhancing intestinal barrier integrity. Furthermore, NMN synbiotics decreased the expression of proinflammatory cytokines IL-1ß, IL-6, and TNF-α, and reduced ROS levels, indicative of attenuated oxidative stress. The reduction in Aß deposition, enhancement of intestinal barrier function, decrease in neuroinflammation, and alleviation of oxidative stress suggest that NMN synbiotics present a promising therapeutic intervention for AD by modulating multiple pathological pathways. Further research is required to elucidate the precise mechanisms, particularly the role of the NLRP3 inflammasome pathway, which may offer a novel target for AD treatment.


Subject(s)
Alzheimer Disease , Synbiotics , Animals , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Synbiotics/administration & dosage , Nicotinamide Mononucleotide/therapeutic use , Nicotinamide Mononucleotide/pharmacology , Mice, Transgenic , Mice , Amyloid beta-Peptides/metabolism , Reactive Oxygen Species/metabolism , Oxidative Stress/drug effects , Male , Cytokines/metabolism
17.
Biomed Pharmacother ; 178: 117199, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39053426

ABSTRACT

Alzheimer's disease (AD) is a neurodegenerative disorder characterized by the deposition of ß-amyloid (Aß) plaques and neurofibrillary tangles composed of tau protein in the brain. These neuropathological hallmarks contribute to cognitive impairment by inducing neuronal loss in the cerebral cortex and hippocampus. Unfortunately, current therapeutic approaches only target symptomatic relief and do not impede disease progression. Nicotinamide mononucleotide (NMN), a precursor of nicotinamide adenine dinucleotide (NAD+), has emerged as a promising candidate for the treatment of age-related neurodegenerative disorders. NMN supplementation could restore NAD+ levels, thereby alleviating neuronal damage and slowing the progression of AD and other aging-associated diseases. AD is closely associated with autophagic impairment and oxidative stress. Our in vivo experiments demonstrated that NMN could ameliorate pathological and behavioral impairments in AD mice. Specifically, NMN enhanced autophagy and promoted p-tau clearance. Meanwhile, NMN could activate the Nrf2/Keap1/NQO1 pathway, thereby reducing the oxidative stress. Immunofluorescence results demonstrated that NMN could alleviate neuronal damage in AD mice. Furthermore, in vitro results showed that the p-tau clearance and antioxidant stress effects of NMN were suppressed by autophagy inhibitor, chloroquine (CQ) or bafilomycin A1 (BafA1), in Aß-induced PC12 cells. Lastly, when Nrf2 was knocked down, the antioxidant stress, autophagy enhancement, and p-tau clearance effects of NMN were all inhibited. In conclusion, our research indicates that NMN exerts therapeutic effect against AD by activating autophagy and the Nrf2/Keap1/NQO1 pathway through a mutual regulating mechanism of autophagy and antioxidative stress. These findings highlight the promising potential of NMN for the treatment of AD.


Subject(s)
Alzheimer Disease , Autophagy , NF-E2-Related Factor 2 , Nicotinamide Mononucleotide , Oxidative Stress , Animals , Oxidative Stress/drug effects , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Autophagy/drug effects , Nicotinamide Mononucleotide/pharmacology , Nicotinamide Mononucleotide/therapeutic use , Mice , NF-E2-Related Factor 2/metabolism , Antioxidants/pharmacology , Antioxidants/therapeutic use , tau Proteins/metabolism , Kelch-Like ECH-Associated Protein 1/metabolism , Mice, Transgenic , Male , Disease Models, Animal , NAD(P)H Dehydrogenase (Quinone)/metabolism , PC12 Cells , Rats , Amyloid beta-Peptides/metabolism , Mice, Inbred C57BL , Signal Transduction/drug effects
18.
Biochem Biophys Res Commun ; 728: 150346, 2024 Oct 08.
Article in English | MEDLINE | ID: mdl-38972085

ABSTRACT

Tissue-specific deficiency of nicotinamide phosphoribosyl transferase (NAMPT), the rate-limiting enzyme of the nicotinamide adenine dinucleotide (NAD+)-salvage pathway, causes a decrease of NAD+ in the tissue, resulting in functional abnormalities. The NAD+-salvage pathway is drastically activated in the mammary gland during lactation, but the significance of this has not been established. To investigate the impact of NAD+ perturbation in the mammary gland, we generated two new lines of mammary gland epithelial-cell-specific Nampt-knockout mice (MGKO). LC-MS/MS analyses confirmed that the levels of NAD+ and its precursor nicotinamide mononucleotide (NMN) were significantly increased in lactating mammary glands. We found that murine milk contained a remarkably high level of NMN. MGKO exhibited a significant decrease in tissue NAD+ and milk NMN levels in the mammary gland during lactation periods. Despite the decline in NAD+ levels, the mammary glands of MGKO appeared to develop normally. Transcriptome analysis revealed that the gene profiles of MGKO were indistinguishable from those of their wild-type counterparts, except for Nampt. Although the NMN levels in milk from MGKO were decreased, the metabolomic profile of milk was otherwise unaltered. The mammary gland also contains adipocytes, but adipocyte-specific deficiency of Nampt did not affect mammary gland NAD+ metabolism or mammary gland development. These results demonstrate that the NAD+ -salvage pathway is activated in mammary epithelial cells during lactation and suggest that this activation is required for production of milk NMN rather than mammary gland development. Our MGKO mice could be a suitable model for exploring the potential roles of NMN in milk.


Subject(s)
Epithelial Cells , Lactation , Mammary Glands, Animal , Mice, Knockout , Milk , Nicotinamide Mononucleotide , Nicotinamide Phosphoribosyltransferase , Animals , Nicotinamide Phosphoribosyltransferase/metabolism , Nicotinamide Phosphoribosyltransferase/genetics , Nicotinamide Mononucleotide/metabolism , Mammary Glands, Animal/metabolism , Female , Epithelial Cells/metabolism , Milk/metabolism , Mice , Lactation/metabolism , Cytokines/metabolism , NAD/metabolism , Mice, Inbred C57BL
19.
Sci Rep ; 14(1): 16557, 2024 07 17.
Article in English | MEDLINE | ID: mdl-39019993

ABSTRACT

Nicotinamide adenine dinucleotide (NAD +) plays a pivotal role in numerous cellular functions. Reduced NAD + levels are postulated to be associated with cancer. As interest in understanding NAD + dynamics in cancer patients with therapeutic applications in mind grows, there remains a shortage of comprehensive data. This study delves into NAD + dynamics in patients undergoing surgery for different digestive system cancers. This prospective study enrolled 99 patients with eight different cancers. Fasting blood samples were obtained during the perioperative period. The concentrations of NAD + , nicotinamide mononucleotide (NMN), and nicotinamide riboside were analyzed using tandem mass spectrometry. After erythrocyte volume adjustment, NAD + remained relatively stable after surgery. Meanwhile, NMN decreased the day after surgery and displayed a recovery trend. Interestingly, liver and pancreatic cancer patients exhibited poor postoperative NMN recovery, suggesting a potential cancer type-specific influence on NAD + metabolism. This study illuminated the behavior of NAD + in surgically treated cancer patients. We identified which cancer types have particularly low levels and at what point depletion occurs during the perioperative period. These insights suggest the need for personalized NAD + supplementation strategies, calibrated to individual patient needs and treatment timelines. Clinical trial registration jRCT1020210066.


Subject(s)
NAD , Niacinamide , Adult , Aged , Aged, 80 and over , Female , Humans , Male , Middle Aged , NAD/metabolism , Neoplasms/surgery , Neoplasms/metabolism , Niacinamide/therapeutic use , Niacinamide/metabolism , Niacinamide/analogs & derivatives , Nicotinamide Mononucleotide/metabolism , Prospective Studies , Pyridinium Compounds , Tandem Mass Spectrometry
20.
ACS Synth Biol ; 13(8): 2425-2435, 2024 Aug 16.
Article in English | MEDLINE | ID: mdl-39023319

ABSTRACT

Nicotinamide mononucleotide (NMN) serves as a precursor for NAD+ synthesis and has been shown to have positive effects on the human body. Previous research has predominantly focused on the nicotinamide phosphoribosyltransferase-mediated route (NadV-mediated route) for NMN biosynthesis. In this study, we have explored the de novo NMN biosynthesis route as an alternative pathway to enhance NMN production. Initially, we systematically engineered Escherichia coli to enhance its capacity for NMN synthesis and accumulation, resulting in a remarkable over 100-fold increase in NMN yield. Subsequently, we progressively enhanced the de novo NMN biosynthesis route to further augment NMN production. We screened and identified the crucial role of MazG in catalyzing the enzymatic cleavage of NAD+ to NMN. And the de novo NMN biosynthesis route was optimized and integrated with the NadV-mediated NMN biosynthetic pathways, leading to an intracellular concentration of 844.10 ± 17.40 µM NMN. Furthermore, the introduction of two transporters enhanced the uptake of NAM and the excretion of NMN, resulting in NMN production of 1293.73 ± 61.38 µM. Finally, by engineering an E. coli strain with optimized PRPP synthetase, we achieved the highest NMN production, reaching 3067.98 ± 27.25 µM after 24 h of fermentation at the shake flask level. In addition to constructing an efficient E. coli cell factory for NMN production, our findings provide new insights into understanding the NAD+ salvage pathway and its role in energy metabolism within E. coli.


Subject(s)
Escherichia coli , Metabolic Engineering , NAD , Nicotinamide Mononucleotide , Escherichia coli/metabolism , Escherichia coli/genetics , Nicotinamide Mononucleotide/metabolism , Metabolic Engineering/methods , NAD/metabolism , Escherichia coli Proteins/metabolism , Escherichia coli Proteins/genetics , Nicotinamide Phosphoribosyltransferase/metabolism , Nicotinamide Phosphoribosyltransferase/genetics , Biosynthetic Pathways/genetics
SELECTION OF CITATIONS
SEARCH DETAIL