5-Amino-1MQ
5-Amino-1MQ
5-Amino-1MQ: Next-Generation NNMT Inhibitor for Metabolic Research
Disclaimer
Products described here are supplied for research use only and are not intended for diagnostic, therapeutic, or clinical application. All statements regarding biological activity reflect preclinical and in vitro findings exclusively and have not been evaluated by the U.S. Food and Drug Administration. These materials and related content are provided for educational and investigational purposes only and are intended solely for qualified researchers in laboratory and academic settings.
Introduction: A Breakthrough in Metabolic Modulation
5-amino-1-methylquinolinium (5-amino-1MQ) represents a significant advancement in the field of metabolic research, emerging as a potent, selective inhibitor of nicotinamide N-methyltransferase (NNMT). This small molecule compound was developed through sophisticated structure-guided design and binding calculations as part of a systematic effort to create membrane-permeable NNMT inhibitors with enhanced potency and selectivity. Unlike broader metabolic interventions, 5-amino-1MQ targets a specific enzymatic bottleneck in cellular NAD+ metabolism, offering researchers a precise tool for investigating the intricate relationships between methylation cycles, energy metabolism, and cellular homeostasis.
The discovery of 5-amino-1MQ emerged from groundbreaking research demonstrating that NNMT serves as a critical regulatory node in metabolic health. This enzyme catalyzes the N-methylation of nicotinamide using S-adenosyl-L-methionine (SAM) as a methyl donor, effectively creating a "metabolic drain" that depletes both NAD+ precursors and cellular methylation capacity. In laboratory investigations, researchers found that NNMT overexpression correlates with obesity and metabolic dysfunction, while its inhibition produces remarkable metabolic benefits. The development of 5-amino-1MQ as a selective NNMT inhibitor represents a paradigm shift in metabolic research, providing scientists with the ability to modulate this pathway with unprecedented precision and potency.
What distinguishes 5-amino-1MQ in the research landscape is its exceptional selectivity profile and membrane permeability characteristics. Unlike earlier compounds that affected multiple methyltransferases, 5-amino-1MQ demonstrates remarkable specificity for NNMT without inhibiting related SAM-dependent enzymes or components of the NAD+ salvage pathway. This selectivity, combined with its ability to efficiently cross cellular membranes, makes it an invaluable research tool for dissecting the specific contributions of NNMT to metabolic regulation. The compound's development represents years of medicinal chemistry optimization, resulting in a 10-fold improvement in potency over parent compounds while maintaining excellent safety margins in preclinical studies.
Primary Mechanism: Selective NNMT Inhibition and NAD+ Restoration
At the molecular level, 5-amino-1MQ functions as a competitive inhibitor of NNMT, demonstrating remarkable potency with an IC₅₀ of 1.2 ± 0.1 μM under standard assay conditions (50 μM SAM, 100 μM nicotinic acid). This represents a dramatic 10-fold improvement over the parent compound 1-methylquinolinium, achieved through strategic amino group substitution that enhances binding affinity to the NNMT active site. The compound's mechanism centers on preventing the methylation of nicotinamide to 1-methylnicotinamide (1-MNA), thereby preserving nicotinamide for recycling back to NAD+ through the salvage pathway. This intervention effectively blocks what researchers have termed the "NNMT metabolic drain" – a process that simultaneously depletes NAD+ precursors and consumes cellular methylation capacity.
The downstream effects of NNMT inhibition by 5-amino-1MQ create a cascade of beneficial metabolic changes at the cellular level. By preventing nicotinamide methylation, the compound increases intracellular NAD+ availability, which in turn activates sirtuins and other NAD+-dependent enzymes critical for metabolic homeostasis. Simultaneously, the preservation of SAM levels maintains cellular methylation capacity for essential epigenetic modifications and protein functions. Research demonstrates that 5-amino-1MQ administration at 30 μM concentration in adipocytes for 24 hours significantly increases intracellular NAD+ while reducing 1-MNA levels with an EC₅₀ of 2.3 μM. This dual preservation of both NAD+ and methylation resources represents a unique mechanism among metabolic modulators, positioning 5-amino-1MQ as a bridge between energy metabolism and epigenetic regulation.
The selectivity profile of 5-amino-1MQ is particularly noteworthy for research applications, as comprehensive enzyme panel screening reveals no inhibition of related SAM-dependent methyltransferases or enzymes in the NAD+ salvage pathway at concentrations up to 10 μM. This includes nicotinamide riboside kinases, nicotinamide phosphoribosyltransferase, and other methyltransferases that could confound experimental results. The compound's selectivity extends to its membrane permeability characteristics, which allow efficient cellular uptake without affecting membrane integrity or cellular viability up to 100 μM concentrations. This exceptional selectivity profile enables researchers to attribute observed effects specifically to NNMT inhibition, rather than off-target activities that plague less specific compounds.
Metabolic Effects: Adipose Tissue Regulation and Energy Balance
One of the most striking applications of 5-amino-1MQ in metabolic research lies in its profound effects on adipose tissue metabolism and lipid storage regulation. In diet-induced obesity animal models, NNMT inhibition demonstrated metabolic changes including alterations in adipocyte lipid content without affecting food intake patterns, coupled with significant reductions in white adipose tissue mass in treated mice. These changes occurred through direct effects on adipocyte biology rather than appetite suppression, as research models maintained normal feeding behavior while exhibiting significant reductions in individual adipocyte size. Laboratory investigations reveal that 5-amino-1MQ suppresses lipogenesis in adipocytes with an EC₅₀ of 30 μM, mechanistically achieved through restoration of cellular NAD+/SAM ratios that favor lipolysis over lipid storage.
The molecular mechanisms underlying 5-amino-1MQ's anti-adipogenic effects involve complex interactions between NAD+ metabolism, methylation status, and transcriptional regulation. By inhibiting NNMT, the compound prevents the depletion of both NAD+ and SAM in adipocytes, two factors that are critical for maintaining metabolic flexibility. Research shows that NNMT overexpression in adipose tissue creates a metabolic environment that favors lipid accumulation by depleting the very cofactors needed for efficient fatty acid oxidation. 5-amino-1MQ reverses this process, restoring cellular capacity for fatty acid oxidation while simultaneously reducing the availability of methylation resources needed for lipogenic gene expression. This dual mechanism explains why the compound produces such pronounced effects on adipose tissue lipid content without affecting lean tissue or overall energy intake in research models.
Beyond direct adipose effects, 5-amino-1MQ influences whole-body energy metabolism through its actions on multiple tissue types. In experimental models, research models demonstrate altered insulin sensitivity and glucose handling parameters, with plasma total cholesterol levels showing reductions. The compound's effects extend to muscle tissue, where NNMT inhibition modulates cellular energetics and contractile function. Recent breakthrough research in 2024 demonstrated that 5-amino-1MQ combined with exercise training in aged murine models produced enhanced adaptations compared to exercise alone, with grip strength improvements of approximately 60% and alterations in daily locomotor activity patterns with modified recovery kinetics. These findings suggest that NNMT inhibition may modulate the metabolic responses to physical activity by altering cellular energy metabolism across multiple tissue types.
Cellular Mechanisms: NAD+ Homeostasis and Methylation Balance
At the cellular level, 5-amino-1MQ orchestrates a complex rebalancing of fundamental metabolic processes through its effects on the NAD+ salvage pathway and one-carbon metabolism. The compound's primary target, NNMT, sits at a critical metabolic intersection where NAD+ precursor availability meets cellular methylation capacity. Under normal conditions, NNMT activity can consume substantial amounts of both nicotinamide and SAM, particularly in metabolically active tissues or during periods of metabolic stress. 5-amino-1MQ intervention effectively removes this metabolic drain, allowing cells to maintain higher steady-state levels of both NAD+ and SAM. This restoration of metabolic cofactor availability enables enhanced function of NAD+-dependent enzymes including sirtuins, poly(ADP-ribose) polymerases, and various deacetylases that regulate cellular metabolism, stress responses, and aging processes.
The implications of NNMT inhibition extend far beyond simple cofactor availability, influencing fundamental cellular processes including mitochondrial biogenesis, oxidative stress management, and epigenetic regulation. Research demonstrates that 5-amino-1MQ administration enhances mitochondrial respiratory capacity through improved NAD+ availability for complex I function and TCA cycle enzymes. Simultaneously, the preservation of SAM levels maintains cellular capacity for critical methylation reactions including histone modifications, DNA methylation, and protein methylation that regulate gene expression and protein function. This dual preservation creates a unique metabolic environment where cells can simultaneously maintain high energy production capacity and robust epigenetic regulation – a combination that typically requires careful balance between competing metabolic demands.
The temporal dynamics of 5-amino-1MQ's cellular effects reveal sophisticated regulatory mechanisms that extend well beyond immediate enzyme inhibition. Within hours of treatment, research models begin showing increased NAD+ levels and reduced 1-MNA production, but the full spectrum of metabolic changes develops over days to weeks as downstream signaling cascades adjust to the new metabolic environment. Long-term studies in laboratory models suggest that sustained NNMT inhibition may reprogram cellular metabolism toward enhanced oxidative capacity and improved stress resistance. This temporal progression indicates that 5-amino-1MQ may serve as more than a simple enzyme inhibitor, potentially acting as a metabolic reprogramming agent that fundamentally alters cellular energy management strategies over extended periods.
Research Applications: From Basic Metabolism to Aging Biology
The research utility of 5-amino-1MQ extends across multiple disciplines within metabolic biology, offering investigators a powerful tool for dissecting the relationships between NAD+ metabolism, methylation biology, and cellular aging processes. In aging research, the compound provides a unique approach to investigating theories of metabolic decline, as NNMT expression increases with age in many tissues, potentially contributing to the NAD+ decline associated with aging processes. Laboratory studies using 5-amino-1MQ in aged animal models have revealed that NNMT inhibition modulates many age-associated metabolic parameters, including alterations in muscle tissue metabolism and cellular stress response pathways. These findings position the compound as a valuable research tool for investigating whether NNMT modulation influences age-associated metabolic changes in mammalian aging models.
In cancer metabolism research, 5-amino-1MQ offers investigators a precise tool for studying the role of NNMT in tumor cell biology and metabolic reprogramming. Research has demonstrated that the compound shows anti-proliferative activity in specific cancer cell lines, with HeLa cells showing concentration and time-dependent growth inhibition across a 0.1-500 μM range while HEK-293 cells remain unaffected. Molecular analyses reveal that 5-amino-1MQ administration alters expression of key genes involved in epithelial-mesenchymal transition, including increased ZEB1 and SIRT1 expression alongside decreased TWIST and SERPIN1 levels. These findings suggest that NNMT inhibition may influence tumor cell line phenotypes in vitro through both metabolic and epigenetic mechanisms, making 5-amino-1MQ a valuable tool for investigating the metabolic characteristics of different tumor cell lines.
Exercise physiology research represents another expanding application area for 5-amino-1MQ, particularly given recent findings demonstrating altered metabolic responses to contractile activity in laboratory models. The compound's effects on muscle tissue metabolism and cellular energetics during activity-recovery cycles provide a tool for studying the molecular basis of exercise-induced adaptations in mammalian muscle. Researchers are investigating whether NNMT inhibition modulates the cellular metabolic responses to repeated contractile stress, potentially explaining the differential metabolic profiles observed when combining NNMT inhibition with exercise protocols in research models. These investigations provide insights into the metabolic regulation of contractile adaptation and the cellular mechanisms underlying activity-induced metabolic remodeling in muscle tissue across different experimental conditions.
Safety Profile and Research Considerations
The safety profile of 5-amino-1MQ in preclinical studies demonstrates favorable tolerability profiles across multiple research models, providing researchers with a substantial safety margin for experimental applications. Comprehensive toxicology evaluations reveal that the compound does not affect food intake, body temperature regulation, or basic physiological parameters at effective doses in animal models, indicating that its metabolic effects occur through specific pathway modulation rather than general physiological disruption. In vitro studies confirm minimal cytotoxicity with no impact on cell viability up to 100 μM concentrations across multiple cell types, though higher concentrations may produce non-specific effects that could confound experimental results.
For research applications, investigators should consider several important factors when designing studies with 5-amino-1MQ. The compound's high membrane permeability and metabolic stability make it suitable for both acute and chronic study designs, but researchers should be aware that its effects on NAD+ and methylation metabolism may require time to fully manifest. Dose-response relationships appear to be tissue-specific, with adipose tissue showing high sensitivity while other tissues may require higher concentrations for detectable effects. The compound's selectivity profile minimizes concerns about off-target effects, but investigators should consider potential interactions with other NAD+-modulating interventions or compounds that affect methylation metabolism.
Current regulatory status places 5-amino-1MQ as restricted to research use only in laboratory and academic settings. Comprehensive preclinical safety evaluations across multiple animal models demonstrate favorable tolerability profiles with excellent safety margins for investigational research applications. As with all research compounds, investigators should implement appropriate laboratory safety protocols and follow institutional guidelines for handling investigational chemical compounds in controlled research environments.
Future Directions and Research Implications
The emergence of 5-amino-1MQ as a research tool opens numerous avenues for investigating fundamental questions in metabolic biology, aging research, and cellular energetics. Current research priorities include large-scale studies to better understand the tissue-specific effects of NNMT inhibition and the temporal dynamics of metabolic reprogramming following treatment. Investigators are particularly interested in determining the mechanisms by which effects observed in murine models relate to broader mammalian metabolic regulation, and whether different experimental conditions (young vs. aged models, healthy vs. metabolically compromised states) show differential responses to NNMT inhibition. These studies will be critical for understanding the research utility of this pathway and identifying optimal applications for future investigational development.
Mechanistic research continues to reveal new aspects of NNMT biology that may expand the applications of 5-amino-1MQ in experimental settings. Recent discoveries suggest that NNMT may play important roles in tissue-specific metabolic regulation beyond its established functions in liver and adipose tissue, including potential roles in brain metabolism, immune cell function, and tissue repair processes. Advanced analytical techniques are enabling researchers to track the compound's effects on global metabolite profiles, revealing complex interactions between NAD+ metabolism, methylation cycles, and other metabolic pathways that were previously unappreciated. These systems-level insights may identify new research applications and combination strategies that enhance the compound's utility as an experimental tool.
The development of 5-amino-1MQ also represents a broader proof-of-concept for targeting metabolic intersections as a research strategy. The success of this approach in modulating both energy metabolism and epigenetic regulation suggests that other metabolic nodes may offer similar opportunities for dual-pathway intervention. Future medicinal chemistry efforts may yield next-generation compounds with enhanced properties, including improved tissue selectivity, modified pharmacokinetics, or additional mechanistic activities that complement NNMT inhibition. Such developments would provide researchers with an expanded toolkit for investigating the complex relationships between metabolism, aging, and disease, potentially accelerating discoveries across multiple fields of biomedical research.
Conclusion: A Precise Tool for Metabolic Research
5-amino-1MQ represents a significant advancement in metabolic research tools, offering investigators unprecedented precision in modulating the critical intersection between NAD+ metabolism and cellular methylation capacity. The compound's exceptional selectivity for NNMT, combined with its favorable safety profile and membrane permeability characteristics, positions it as an invaluable resource for researchers investigating fundamental questions in metabolism, aging, and cellular energetics. From its origins in structure-guided drug design to its current applications across multiple research disciplines, 5-amino-1MQ exemplifies how targeted enzyme inhibition can provide insights into complex biological systems while maintaining the specificity required for rigorous scientific investigation.
The compound's unique mechanism of action – simultaneously preserving NAD+ availability and methylation capacity through selective NNMT inhibition – offers researchers a powerful approach to investigating metabolic flexibility and cellular adaptation. Whether applied to studies of aging biology, exercise physiology, cancer metabolism, or fundamental cellular energetics, 5-amino-1MQ provides the precision and reliability required for advancing our understanding of how metabolic networks integrate to maintain cellular and organismal health. As research continues to reveal new applications and mechanistic insights, this compound stands poised to contribute to breakthrough discoveries across the expanding landscape of metabolic biology and translational research.
References
- Neelakantan, H., et al. (2018). Selective and membrane-permeable small molecule inhibitors of nicotinamide N-methyltransferase reverse high fat diet-induced obesity in mice. Biochemical Pharmacology 147:141-152. https://doi.org/10.1016/j.bcp.2017.11.007
- Akar, S., et al. (2021). Small molecule inhibitor of nicotinamide N-methyltransferase shows anti-proliferative activity in HeLa cells. Journal of Obstetrics and Gynaecology 41(8):1316-1322. https://doi.org/10.1080/01443615.2020.1854696
- Awosemo, O., et al. (2021). Development & validation of LC-MS/MS assay for 5-amino-1-methyl quinolinium in rat plasma: Application to pharmacokinetic and oral bioavailability studies. Journal of Pharmaceutical and Biomedical Analysis 204:114255. https://doi.org/10.1016/j.jpba.2021.114255
- Dimet-Wiley, A.L., et al. (2021). Reduced calorie diet combined with NNMT inhibition establishes a distinct microbiome in DIO mice. Scientific Reports 12:484. https://doi.org/10.1038/s41598-021-03670-5
- Kraus, D., et al. (2014). Nicotinamide N-methyltransferase knockdown protects against diet-induced obesity. Nature 508(7495):258-262. https://doi.org/10.1038/nature13198
- Ulanovskaya, O.A., et al. (2013). NNMT promotes epigenetic remodeling in cancer by creating a metabolic methylation sink. Nature Chemical Biology 9(5):300-306. https://doi.org/10.1038/nchembio.1204
- Pissios, P. (2017). Nicotinamide N-methyltransferase: More than a vitamin B3 clearance enzyme. Trends in Endocrinology & Metabolism 28(5):340-353. https://doi.org/10.1016/j.tem.2017.02.004
- Hong, S., et al. (2015). Nicotinamide N-methyltransferase regulates hepatic nutrient metabolism through Sirt1 protein stabilization. Nature Medicine 21(8):887-894. https://doi.org/10.1038/nm.3882
- Dimet-Wiley, A.L., et al. (2024). Nicotinamide N-methyltransferase inhibition mimics and boosts exercise-mediated improvements in muscle function in aged mice. Scientific Reports 14:15554. https://doi.org/10.1038/s41598-024-66034-9
- Babula, P., et al. (2024). Nicotinamide N-methyltransferase inhibition mitigates obesity-related metabolic dysfunction. Diabetes, Obesity and Metabolism. https://doi.org/10.1111/dom.15879
- Liu, J.R., et al. (2021). Roles of Nicotinamide N-Methyltransferase in Obesity and Type 2 Diabetes. BioMed Research International 2021:9924314. https://doi.org/10.1155/2021/9924314
- Roberti, A., et al. (2021). Nicotinamide N-methyltransferase: At the crossroads between cellular metabolism and epigenetic regulation. Molecular Metabolism 45:101165. https://doi.org/10.1016/j.molmet.2021.101165
- Liang, Y., et al. (2024). Identification of nicotinamide N-methyltransferase as a promising therapeutic target for sarcopenia. Aging Cell. https://doi.org/10.1111/acel.14236
- Kannt, A., et al. (2018). A small molecule inhibitor of nicotinamide N-methyltransferase for the treatment of metabolic disorders. Scientific Reports 8:3660. https://doi.org/10.1038/s41598-018-22081-7
- Neelakantan, H., et al. (2021). Combined nicotinamide N-methyltransferase inhibition and reduced-calorie diet normalizes body composition and enhances metabolic benefits in obese mice. Scientific Reports 11:6244. https://doi.org/10.1038/s41598-021-85051-6
- PubChem. (2024). 5-Amino-1-methylquinolinium. Compound CID 950107. National Center for Biotechnology Information. https://pubchem.ncbi.nlm.nih.gov/compound/5-Amino-1-methylquinolinium
| CAS Number | 685079-15-6 |
|---|---|
| Molecular Formula | C10H11IN2 |
| Molecular Weight | 286.11 g/Mol |
| Purity | 99.5% |
| Lot Number | 25039 |
| Quantity | 6.47mg |
| Sequence | 1-methylquinolin-1-ium-5-amine |
Cosmic Peptides
Certificate of Analysis
Unable to load certificate. Download COA