NMNAT1 Human, Active

Nicotinamide Nucleotide Adenylyltransferase 1 Human Recombinant , Active
Shipped with Ice Packs
In Stock

Description

Introduction to NMNAT1 Human, Active

NMNAT1 (Nicotinamide Mononucleotide Adenylyltransferase 1) is a nuclear enzyme critical for NAD⁺ biosynthesis, playing pivotal roles in cellular metabolism, DNA repair, and gene regulation. Recombinant human NMNAT1 is a highly purified, catalytically active protein derived from E. coli, widely used in biochemical assays and therapeutic research. Its activity is quantified by its ability to synthesize NAD⁺ from nicotinamide mononucleotide (NMN) and ATP, with a specific activity exceeding 5 U/mg protein .

Catalytic Activity

NMNAT1 catalyzes the adenylylation of NMN and nicotinic acid mononucleotide (NaMN) to form NAD⁺ and NaAD, respectively. It also exhibits pyrophosphorolytic activity, cleaving NAD⁺ into NMN and pyrophosphate . The enzyme’s catalytic domain requires intact binding sites for NMN (W170) and ATP (H24), as mutations in these residues abolish activity .

Subcellular Localization

NMNAT1 is primarily nuclear, where it interacts with PARP1 to generate nuclear ATP for chromatin remodeling and DNA repair . It is ubiquitously expressed but indispensable in tissues with high metabolic demands, such as the retina and brain .

Research Applications and Findings

ApplicationKey FindingsSources
Retinal DevelopmentNMNAT1 deletion causes photoreceptor degeneration via disrupted NAD⁺ metabolism and impaired gene regulation (e.g., downregulation of photoreceptor-specific genes) .
Cancer TherapyNMNAT1 depletion sensitizes AML to venetoclax by reducing nuclear NAD⁺, activating p53, and impairing DNA repair .
NeurodegenerationOverexpression reduces amyloid plaques in Alzheimer’s models by promoting autophagic clearance of APP-CTF .
Axonal ProtectionBlocks SARM1-mediated axon degeneration by preventing NAD⁺ depletion post-injury .

Disease-Targeted Mechanisms

DiseaseMechanismOutcomeSources
Leber Congenital Amaurosis (LCA9)Mutations in NMNAT1 disrupt NAD⁺ synthesis, causing photoreceptor degeneration .Severe retinal dystrophy and blindness.
Acute Myeloid Leukemia (AML)Inhibition reduces nuclear NAD⁺, enhancing chemosensitivity to venetoclax .Improved survival in preclinical models.
Alzheimer’s DiseasePromotes autophagy, reducing Aβ plaques by enhancing Atg8a lipidation .Potential therapeutic target for proteinopathies.

Challenges and Future Directions

  1. Compartmentalized NAD⁺ Regulation: NMNAT1’s nuclear-specific role complicates systemic targeting. Research focuses on isoform-specific inhibitors or activators .

  2. Metabolic vs. Genetic Roles: NMNAT1 influences both energy metabolism and gene expression (e.g., SIRT1-mediated deacetylation in autophagy) .

  3. Cancer Dependency: NMNAT1’s dispensability in normal hematopoiesis but essentiality in AML suggests a therapeutic window for targeting .

Product Specs

Introduction
NMNAT1, an essential enzyme for NAD biosynthesis, catalyzes the condensation of nicotinamide mononucleotide (NMN) or nicotinic acid mononucleotide (NaMN) with ATP's AMP moiety to form NAD or NaAD. This enzyme is widely distributed in the body, with high concentrations found in skeletal muscle, heart, liver, and kidney. NMNAT1 has been shown to have protective effects against axonal degeneration caused by mechanical or toxic damage.
Description
Recombinant Human NMNAT1, produced in E. coli, is a single, non-glycosylated polypeptide chain containing 315 amino acids (specifically amino acids 1-279) and has a molecular weight of 36 kDa. This protein is fused to a 36 amino acid His-Tag at the N-terminus and purified using proprietary chromatographic methods.
Physical Appearance
A clear, colorless solution that has been sterilized by filtration.
Formulation
The NMNAT1 solution is provided at a concentration of 1mg/ml and contains 20mM Tris-HCl buffer (pH 8.0), 20% glycerol, 0.1M NaCl, 1mM DTT, and 1mM EDTA.
Stability
For short-term storage (up to 2-4 weeks), the product can be stored at 4°C. For long-term storage, it is recommended to store the product frozen at -20°C. Adding a carrier protein (0.1% HSA or BSA) is advised for long-term storage. Repeated freezing and thawing of the product should be avoided.
Purity
The purity of the NMNAT1 protein is greater than 95.0% as determined by SDS-PAGE analysis.
Biological Activity
The specific activity of the enzyme is greater than 7,000 pmol/min/ug. This value was determined by measuring the amount of beta-NAD produced from nicotinamide mononucleotide and ATP per minute at a pH of 8.0 and a temperature of 37°C.
Synonyms
NMNAT, NMNAT1, PNAT1, Nicotinamide mononucleotide adenylyltransferase 1, NMN adenylyltransferase 1, Nicotinate-nucleotide adenylyltransferase 1, NaMN adenylyltransferase 1, EC=2.7.7.1, EC=2.7.7.18.
Source
Escherichia Coli.
Amino Acid Sequence
MRGSHHHHHH GMASMTGGQQ MGRDLYDDDD KDRWGSMENS EKTEVVLLAC GSFNPITNMH LRLFELAKDY MNGTGRYTVV KGIISPVGDA YKKKGLIPAY HRVIMAELAT KNSKWVEVDT WESLQKEWKE TLKVLRHHQE KLEASDCDHQ QNSPTLERPG RKRKWTETQD SSQKKSLEPK TKAVPKVKLL CGADLLESFA VPNLWKSEDI TQIVANYGLI CVTRAGNDAQ KFIYESDVLW KHRSNIHVVN EWIANDISST KIRRALRRGQSIRYLVPDLV QEYIEKHNLY SSESEDRNAG VILAPLQRNT AEAKT.

Q&A

What is human NMNAT1 and what is its primary cellular localization?

NMNAT1 (Nicotinamide Mononucleotide Adenylyltransferase 1) is a critical enzyme in the NAD+ biosynthesis pathway. It is primarily localized in the nucleus of human cells, where it catalyzes the conversion of nicotinamide mononucleotide (NMN) to nicotinamide adenine dinucleotide (NAD+). This nuclear localization is essential for its function in maintaining nuclear NAD+ pools. Experimental verification of NMNAT1's nuclear localization can be performed using subcellular fractionation followed by western blotting or immunofluorescence microscopy with specific anti-NMNAT1 antibodies. Consistent with this localization pattern, studies in MOLM13 cells have confirmed that NMNAT1 protein is predominantly found in the nucleus, and its deletion significantly reduces nuclear NAD+ levels .

What is the catalytic function of NMNAT1 and why is it essential?

NMNAT1 functions as an adenylyltransferase that catalyzes the condensation of nicotinamide mononucleotide (NMN) with adenosine triphosphate (ATP) to form NAD+ and pyrophosphate. This reaction is a critical step in both the de novo and salvage pathways of NAD+ biosynthesis. The catalytic activity of NMNAT1 requires specific amino acid residues in its active site, particularly tryptophan-170 (W170) and histidine-24 (H24), which are essential for binding to NMN and ATP, respectively .

The essentiality of NMNAT1's catalytic function has been demonstrated through mutation studies. Research shows that mutations in the catalytic domain (W170A or H24A) that impair binding to NMN or ATP render the enzyme non-functional. In experimental settings, while wild-type murine Nmnat1 can rescue the phenotype of NMNAT1-ablated cells, mutant forms with W170A or H24A substitutions fail to do so, confirming that the catalytic activity is indispensable for cellular function .

How does NMNAT1 contribute to cellular NAD+ homeostasis?

NMNAT1 serves as a gatekeeper of nuclear NAD+ biosynthesis, maintaining adequate levels of this crucial coenzyme for nuclear processes. Cellular studies demonstrate that NMNAT1 deficiency results in reduced nuclear NAD+ levels, which cannot be fully compensated by other NMNAT isoforms or pathways. In fibroblasts from patients with NMNAT1 mutations (such as p.Val9Met), NAD+ content was decreased by approximately 16% compared to wild-type controls .

Importantly, NMNAT1 appears to be critical for the cellular response to NAD+ precursors. While nicotinic acid treatment can increase total cellular NAD+ content by 53% in control cells, it shows no significant effect on NAD+ levels in cells with NMNAT1 mutations. This observation underscores NMNAT1's role as an essential enzyme in the conversion pathway of NAD+ precursors . The maintenance of nuclear NAD+ pools by NMNAT1 is particularly important for nuclear processes like DNA repair, gene expression, and the activity of NAD+-dependent enzymes such as sirtuins.

What are the key structural domains of human NMNAT1 and their significance?

Human NMNAT1 contains several functional domains that are critical for its enzymatic activity and cellular function. The most significant domains include:

  • Acetyltransferase domain: This catalytic domain is essential for the enzyme's function. Experiments using sgRNAs targeting different regions of NMNAT1 have shown that disruption of this domain significantly reduces the survival of acute myeloid leukemia (AML) cells, whereas targeting the C-terminal tail has minimal effects .

  • NMN-binding site: Contains the conserved tryptophan-170 (W170) residue which is crucial for substrate binding. Mutation of this residue (W170A) abolishes enzyme activity and cellular function.

  • ATP-binding site: Contains the histidine-24 (H24) residue essential for ATP binding. Similar to W170, mutation of H24 (H24A) renders the enzyme non-functional.

These structural elements are highly conserved, reflecting their critical role in enzyme function. Researchers can study these domains through site-directed mutagenesis, protein crystallography, and enzyme kinetics to understand structure-function relationships. The importance of these domains has been validated through rescue experiments where mutant forms of NMNAT1 with alterations in catalytic residues fail to restore function in NMNAT1-depleted cells .

What experimental models are available to study NMNAT1 function?

Several experimental models have been developed to study NMNAT1 function in both normal and pathological contexts:

  • Cell line models: CRISPR-Cas9 genome editing has been used to create NMNAT1-knockout cell lines, particularly in AML cell lines such as MOLM13. These models allow for studying the immediate effects of NMNAT1 deletion on cellular NAD+ levels, gene expression, and cellular phenotypes .

  • Patient-derived fibroblasts: Fibroblasts from patients with NMNAT1 mutations (such as those with Leber congenital amaurosis) provide valuable models to study the consequences of reduced NMNAT1 activity on cellular metabolism and NAD+ homeostasis .

  • Conditional knockout mouse models: Mx1-Cre;Nmnat1 conditional knockout mice have been generated to study the role of NMNAT1 in normal hematopoiesis and leukemogenesis. These models allow for temporal control of Nmnat1 deletion using polyinosinic:polycytidylic acid [poly(I:C)] induction of Cre recombinase .

  • Patient-derived xenograft (PDX) models: Human AML samples with specific genetic alterations (such as t(9;11) translocation or FLT3-ITD mutation) can be transplanted into immunocompromised NSG-SGM3 mice after CRISPR-Cas9-mediated editing of NMNAT1. These models are particularly valuable for studying the role of NMNAT1 in human leukemia maintenance and drug response in vivo .

  • Retroviral transduction models: Wild-type or mutant forms of NMNAT1 can be expressed in NMNAT1-depleted cells to perform rescue experiments, allowing for the assessment of specific structural domains or catalytic residues .

Each of these models offers unique advantages for addressing specific research questions related to NMNAT1 function in different physiological and pathological contexts.

How does NMNAT1 dysfunction contribute to Leber congenital amaurosis?

Leber congenital amaurosis (LCA) is an infantile-onset form of inherited retinal degeneration characterized by severe vision loss. Mutations in NMNAT1 have been identified as causative for a subset of LCA cases, representing the first disease association with any NMNAT isoform .

The pathogenic mechanism appears to involve impaired enzymatic function of NMNAT1, leading to disrupted NAD+ homeostasis in the retina. Research on fibroblasts from LCA patients with NMNAT1 mutations (such as p.Val9Met) has revealed:

  • Reduced cellular NAD+ levels (approximately 16% decrease compared to controls), suggesting that even partial impairment of NMNAT1 activity can affect cellular NAD+ content.

  • Inability to respond to nicotinic acid supplementation. While treatment with 10 mM nicotinic acid for 24 hours significantly increased NAD+ content by 53% in control cells, it had no effect on NAD+ levels in cells with NMNAT1 mutations .

These findings suggest that NMNAT1 mutations in LCA patients result in significant deficiency in cellular NMNAT enzymatic activity, leading to disrupted NAD+ metabolism. The retina may be particularly vulnerable to NMNAT1 dysfunction due to its high metabolic demands and unique requirements for redox balance and energy metabolism.

To study this mechanism, researchers can use patient-derived cells, animal models with NMNAT1 mutations, or retinal organoids derived from induced pluripotent stem cells. Biochemical assays to measure NAD+ levels and NMNAT enzymatic activity, combined with assessments of retinal structure and function, can provide insights into how NMNAT1 mutations lead to retinal degeneration.

What is the role of NMNAT1 in acute myeloid leukemia (AML) progression?

NMNAT1 plays a critical role in the progression and maintenance of acute myeloid leukemia (AML) through regulation of nuclear NAD+ homeostasis. Several experimental approaches have revealed its importance:

  • CRISPR-Cas9 genetic screens: Whole-genome CRISPR screening and pan-cancer genetic dependency mapping have identified NMNAT1 as an AML dependency governing NAD+ biosynthesis .

  • In vitro studies: Deletion of NMNAT1 in AML cell lines results in:

    • Reduced nuclear NAD+ levels

    • Cell cycle arrest with increased proportion of G0/G1 phase and reduced S phase

    • Increased apoptosis

    • Activation of p53 signaling, evidenced by increased phospho-p53 levels and induction of p53 target genes (CDKN1A/p21, NOXA, BAX, PUMA)

  • In vivo studies: NMNAT1-ablated MOLM13 cells rarely develop AML when transplanted into immunocompromised mice, in contrast to control cells which consistently develop leukemia. This defect in leukemogenesis can be rescued by expression of wild-type murine Nmnat1 but not catalytically inactive mutants (W170A or H24A) .

  • Murine leukemia models: Deletion of Nmnat1 in MLL-AF9-driven murine AML significantly delays leukemogenesis, ameliorates leukocytosis, and reduces AML cell frequency in the blood. Even when deleting Nmnat1 in established AML, there is significant reduction in AML cells and extended survival of mice .

  • Human PDX models: In patient-derived xenograft models with t(9;11) translocation or FLT3-ITD mutation, NMNAT1 editing results in reduced AML expansion in the bone marrow, decreased leukemia stem cells, and extended survival of recipient mice .

These findings establish NMNAT1 as a critical factor in AML development and maintenance, making it a potential therapeutic target.

What is the molecular mechanism linking NMNAT1 to p53 activation in cancer cells?

NMNAT1 regulates p53 activity through control of nuclear NAD+ levels and subsequent modulation of sirtuin activity. The molecular mechanism linking NMNAT1 and p53 involves several steps:

  • NAD+ depletion affects sirtuin activity: NMNAT1 deletion reduces nuclear NAD+ levels, which impairs the activity of NAD+-dependent sirtuin family proteins, particularly SIRT6 and SIRT7 .

  • Impaired deacetylation of p53: SIRT6 and SIRT7 normally deacetylate p53, keeping its activity under control. With reduced NAD+ availability, these sirtuins cannot effectively deacetylate p53, leading to its hyperacetylation .

  • Acetylated p53 is activated: Hyperacetylated p53 has increased stability and transcriptional activity, leading to induction of p53 target genes involved in cell cycle arrest (p21/CDKN1A) and apoptosis (NOXA, BAX, PUMA) .

  • DNA damage response activation: NMNAT1 deletion also leads to increased levels of γH2AX, indicating DNA damage response activation, which further reinforces p53 activation .

  • p53-dependent apoptosis: The activated p53 pathway ultimately leads to apoptosis, as evidenced by increased proportion of apoptotic cells in NMNAT1-deleted AML .

The critical role of p53 in this mechanism is confirmed by the observation that co-deletion of TP53 and NMNAT1 significantly reduces the frequency of apoptotic cells and improves survival of NMNAT1-ablated AML cells . This molecular pathway explains why NMNAT1 is particularly important for cancer cells, which often rely on tight control of p53 activity for their survival.

Why is NMNAT1 essential for leukemia stem cells but dispensable for normal hematopoietic stem cells?

One of the most intriguing aspects of NMNAT1 biology is its differential requirement in leukemia stem cells (LSCs) versus normal hematopoietic stem cells (HSCs). This therapeutic window makes NMNAT1 an attractive target for AML therapy. Several experimental approaches have revealed this distinction:

  • Conditional knockout studies: Deletion of Nmnat1 in Mx1-Cre;Nmnat1 conditional knockout mice does not significantly affect normal hematopoiesis. These mice maintain normal:

    • Complete blood counts

    • Bone marrow cellularity

    • Frequencies of HSCs and progenitor populations

    • Colony-forming ability of HSPCs

    • Competitive repopulation capacity of HSCs

  • Leukemia stem cell studies: In contrast, NMNAT1 is essential for leukemia stem cell function in both mouse models and human patient-derived xenografts:

    • Deletion of Nmnat1 in MLL-AF9-driven AML significantly reduces leukemia development

    • NMNAT1 editing in human PDX models reduces CD34+CD38- LSCs and extends survival

  • Metabolic differences: This differential requirement likely stems from fundamental metabolic differences between normal HSCs and LSCs:

    • LSCs have higher energy demands and potentially greater reliance on NAD+-dependent processes

    • LSCs may be more vulnerable to p53 activation resulting from NAD+ depletion

    • Normal HSCs typically reside in a quiescent state with lower metabolic demands

This intrinsic difference in NMNAT1 dependency creates a potential therapeutic window for targeting NMNAT1 in AML without significantly affecting normal hematopoiesis. Researchers can exploit this difference by developing NMNAT1 inhibitors that might selectively target leukemia cells while sparing normal hematopoietic cells.

How does NMNAT1 inhibition sensitize AML cells to venetoclax treatment?

The relationship between NMNAT1 inhibition and venetoclax sensitivity represents an important potential therapeutic strategy for AML. Experimental evidence supports a mechanistic connection:

  • Correlation between NAD+ levels and venetoclax resistance: Analysis of cancer cell line databases has revealed a correlation between NAD+ biosynthesis pathways and resistance to venetoclax, suggesting that higher NAD+ levels may protect against venetoclax-induced apoptosis .

  • In vitro sensitivity studies: NMNAT1 deletion sensitizes AML cells to venetoclax treatment in vitro, with NMNAT1-deleted cells showing significantly increased sensitivity to the drug .

  • Mechanistic link through p53 activation: NMNAT1 deletion activates p53 through reduced NAD+ availability and impaired sirtuin-mediated deacetylation. Prior studies have shown that p53 activation with MDM2 inhibitors sensitizes AML cells to venetoclax, while p53 deletion renders cells resistant to venetoclax .

  • In vivo confirmation: In murine MLL-AF9-driven AML models, reducing NMNAT1 function (using heterozygous Nmnat1 deletion) significantly enhances the efficacy of venetoclax treatment:

    • Venetoclax extends median survival of mice with Nmnat1 WT AML from 25 to 32 days

    • The same treatment extends survival of mice with Nmnat1 heterozygous AML from 38.5 to 60 days

    • This enhanced effect correlates with reduced numbers of AML cells in the blood

These findings suggest that targeting NMNAT1 could be a valuable strategy to overcome venetoclax resistance in AML, particularly through activation of p53-dependent apoptotic pathways. For researchers, this presents opportunities to develop combination therapies targeting both NMNAT1 and BCL-2 (the target of venetoclax) for more effective AML treatment.

What methodologies can be used to measure NMNAT1 activity and nuclear NAD+ levels?

Accurate measurement of NMNAT1 enzymatic activity and nuclear NAD+ levels is essential for research on NAD+ metabolism. Several methodologies are available:

  • NMNAT1 enzymatic activity assays:

    • Spectrophotometric assays: Measuring the formation of NAD+ by monitoring absorbance at 340 nm

    • Coupled enzyme assays: Using auxiliary enzymes like alcohol dehydrogenase to amplify signals

    • Radioactive assays: Using radiolabeled substrates (e.g., 14C-NMN) to track product formation

    • HPLC or LC-MS methods: For direct quantification of NAD+ production

  • Nuclear NAD+ measurement:

    • Subcellular fractionation: Careful isolation of nuclear fractions while preventing NAD+ leakage

    • Enzymatic cycling assays: Amplification of NAD+ signal through enzymatic cycling reactions

    • HPLC-based methods: Direct quantification of NAD+ from nuclear extracts

    • Fluorescent NAD+ biosensors: Genetically encoded sensors that can report on NAD+ levels within specific cellular compartments

  • Validation approaches:

    • Nicotinic acid stimulation: Treatment with NAD+ precursors (10 mM nicotinic acid for 24 hours) increases NAD+ in cells with normal NMNAT1 function but not in cells with NMNAT1 deficiency

    • Genetic complementation: Expression of wild-type NMNAT1 should restore NAD+ levels in NMNAT1-deficient cells, while catalytically inactive mutants (W170A or H24A) should not

When measuring nuclear NAD+ specifically, it's critical to verify that nuclear isolation does not result in leakage of NAD+ from the nucleus. Additionally, rapid processing of samples is essential due to the dynamic nature and rapid turnover of NAD+ in cells. Quantification should be normalized to cell number, protein content, or DNA content depending on the experimental context.

What experimental approaches can be used to study NMNAT1 in patient-derived xenograft models?

Patient-derived xenograft (PDX) models provide valuable insights into the role of NMNAT1 in human leukemia. The following experimental approaches can be used:

  • Generation of NMNAT1-edited PDX models:

    • Electroporation of Cas9 protein together with sgRNA targeting NMNAT1 into patient-derived AML cells

    • Validation of editing efficiency by genomic DNA analysis (e.g., T7 endonuclease assay, Sanger sequencing)

    • Confirmation of protein depletion by western blotting (approximately 90% depletion can be achieved)

  • Transplantation and monitoring:

    • Injection of NMNAT1-edited or control-edited AML cells (typically 1 × 10^5 cells per mouse) into immunocompromised NSG-SGM3 mice via tail vein

    • Monitoring of AML development by regular blood sampling and flow cytometry

    • Analysis of animal survival and disease progression

  • Assessment of leukemia stem cells:

    • Flow cytometric analysis of bone marrow samples for CD34+CD38- leukemia stem cells

    • Evaluation of stem cell frequency and phenotype

    • Secondary transplantation assays to assess self-renewal capacity

  • Therapeutic studies:

    • Combination of NMNAT1 editing with drug treatments (e.g., venetoclax)

    • Evaluation of drug sensitivity in NMNAT1-edited versus control cells

    • Assessment of synergistic effects and mechanisms

  • Molecular and metabolic analyses:

    • Measurement of nuclear NAD+ levels in isolated cells

    • Assessment of p53 activation and target gene expression

    • Evaluation of cell cycle status and apoptosis

    • Gene expression profiling to identify downstream pathways

These approaches have been successfully used to demonstrate that NMNAT1 is required for human AML development in vivo, as evidenced by reduced AML expansion in the bone marrow, decreased CD34+CD38- LSCs, and extended survival in mice receiving NMNAT1-edited cells from two different PDX models (one with t(9;11) translocation/MLL-AF9 fusion gene and another with FLT3-ITD mutation) .

Product Science Overview

Introduction

Nicotinamide Nucleotide Adenylyltransferase 1 (NMNAT1) is a crucial enzyme involved in the biosynthesis of nicotinamide adenine dinucleotide (NAD), a coenzyme essential for various metabolic processes. This enzyme catalyzes the formation of NAD from nicotinamide mononucleotide (NMN) and ATP, playing a pivotal role in cellular metabolism and energy production .

Structure and Function

NMNAT1 is a protein-coding gene that produces an enzyme localized specifically to the cell nucleus. The enzyme’s activity leads to the activation of a nuclear deacetylase, which functions in the protection of damaged neurons . The enzyme is composed of 279 amino acids and has a calculated molecular mass of approximately 31.9 kDa . It contains a conserved N-terminal adenylyltransferase motif, an N-terminal N-glycosylation site, and several potential transmembrane regions .

Expression and Localization

NMNAT1 is widely expressed in various tissues, with high levels observed in skeletal muscle, heart, liver, and kidney . Northern blot analysis has detected two transcripts of NMNAT1, with the 3.1-kb transcript being more abundant . Immunofluorescence microscopy has localized NMNAT1 to the nucleus in human fibroblasts and hepatoma cell lines .

Biological Significance

The enzyme’s activity is crucial for NAD biosynthesis, which is involved in hundreds of metabolic redox reactions. NAD and its derivatives are utilized in protein ADP-ribosylation, histone deacetylation, and some calcium signaling pathways . NMNAT1’s role in NAD biosynthesis makes it essential for cellular metabolism and energy production.

Clinical Relevance

Mutations in the NMNAT1 gene have been associated with several disorders, including Leber congenital amaurosis 9, a severe retinal dystrophy that leads to blindness . Additionally, NMNAT1 is linked to spondyloepiphyseal dysplasia, sensorineural hearing loss, impaired intellectual development, and other conditions .

Recombinant NMNAT1

Recombinant NMNAT1 is produced using recombinant DNA technology, which allows for the expression of the human enzyme in various host cells. This recombinant form retains the enzyme’s activity and is used in research to study its function and potential therapeutic applications .

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2024 Thebiotek. All Rights Reserved.