NME1 antibodies are immunoreagents designed to detect the NME1 protein, which is encoded by the NME1 gene located at chromosome 17q21.33. These antibodies are validated for use in techniques such as Western blotting (WB), immunohistochemistry (IHC), immunofluorescence (IF), and enzyme-linked immunosorbent assays (ELISA). Key epitopes recognized include regions critical for NME1’s enzymatic activities, such as the histidine kinase domain and nucleotide-binding sites .
Multiple monoclonal and polyclonal NME1 antibodies have been developed, with rigorous validation in preclinical models and human tissues:
CPTC-NME1-2 exhibits cytoplasmic and nuclear staining in tumor tissues, correlating with clinical outcomes .
Antibody #3345 demonstrates specificity across species, with no cross-reactivity to NME2 .
NME1 antibodies have been instrumental in elucidating the protein’s dual roles in cancer progression:
Breast Cancer: NME1 loss correlates with increased MT1-MMP surface levels and invasiveness in triple-negative tumors .
Melanoma: NME1 suppresses metastatic activity by regulating genes like MMP1 and ANGPT2 .
Lung Adenocarcinoma: Nuclear NME1 localization is linked to upregulated CYP24A1 expression and poor prognosis .
Cytoplasmic NME1 associates with dynamin-2 in clathrin-coated vesicles, modulating endocytosis .
Radiation-induced nuclear translocation enhances tumor cell migration and invasion .
NME1 expression levels serve as prognostic markers in multiple cancers:
Sensitivity: CPTC-NME1-2 detects NME1 at dilutions up to 1:100 in IHC .
Cross-Reactivity: Limited to NME1 in most clones, though CPTC-NME1-2 shows partial reactivity with NME2 mutants .
Post-Translational Modifications: Antibodies detect CoA-modified NME1 under oxidative stress, revealing regulatory interactions .
Dual Roles: NME1 exhibits context-dependent pro- and anti-metastatic activities, complicating therapeutic targeting .
Technical Limitations: Antibody specificity for nuclear vs. cytoplasmic NME1 requires further optimization .
Emerging Applications: Investigating NME1’s histidine kinase activity in neuroblastoma differentiation .
Nucleoside diphosphate kinase (NDK), composed of 'A' (encoded by NME1) and 'B' (encoded by NME2) isoforms and forming a hexamer, plays a role in cell proliferation, differentiation, development, signal transduction, G protein-coupled receptor endocytosis, and gene expression. NME1 is known to suppress tumor metastasis and is crucial for the synthesis of nucleoside triphosphates, excluding ATP. It is essential for neural development, particularly neural patterning and cell fate determination. Notably, NME1 gene expression varies across tumor types, with its levels linked to both reduced and increased metastatic potential. A decrease in NME1 expression is associated with aggressive behavior in melanoma, breast, colon, and gastric carcinomas. Conversely, elevated NME1 expression is observed in advanced thyroid carcinomas. Somatic mutations in the NME1 gene are found in neuroblastoma, where increased NME1 levels are correlated with aggressive tumor characteristics.
The solution contains 1mg/ml of the antibody in a buffer of PBS at pH 7.4, supplemented with 10% glycerol and 0.02% sodium azide.
This NME1 antibody has undergone testing in various applications, including ELISA, Western blot analysis, Flow cytometry, and ICC/IF, to confirm its specificity and reactivity. However, it's important to note that optimal working dilutions may vary depending on the specific application and should be determined through titration.
Nucleoside diphosphate kinase A, NDP kinase A, NDK A, Tumor metastatic process-associated protein, Metastasis inhibition factor nm23, nm23-H1, Granzyme A-activated DNase, GAAD, NME1, NDPKA, NM23, NB, AWD, NBS, NDPK-A.
NME1 antibody was purified from mouse ascitic fluids by protein-A affinity chromatography.
PAT5F4AT.
Anti-human NME1 mAb, is derived from hybridization of mouse F0 myeloma cells with spleen cells from BALB/c mice immunized with recombinant human NME1 amino acids 1-152 purified from E. coli.
Mouse IgG2b heavy chain and k light chain.
NME1 (also known as nm23-H1) serves as a metastasis suppressor protein involved in multiple cellular processes. It has significant importance in cancer research due to its metastasis suppression role. Studies have demonstrated that NME1 is upregulated in ductal carcinoma in situ (DCIS) compared to normal breast epithelial tissues, but its levels decrease in microinvasive and invasive components of breast tumor cells . This expression pattern makes NME1 a crucial protein for understanding cancer progression mechanisms, particularly the transition from in situ to invasive disease. Additionally, NME1 functions as a nucleoside diphosphate kinase and has been identified as a coenzyme A (CoA) binding protein that undergoes modification under oxidative stress conditions .
Researchers have access to several types of NME1 antibodies with validated applications:
Mouse monoclonal antibodies such as CPTC-NME1-2 (IgG2a isotype), which have been extensively characterized for multiple applications
Antibody pairs consisting of rabbit polyclonal capture antibodies combined with mouse monoclonal detection antibodies for quantitative assays
Specialized antibodies validated for specific applications like imaging mass cytometry
Many of these antibodies have been comprehensively validated across multiple tissue types, including both normal tissues (liver, bone marrow, spleen, prostate, colon, pancreas, breast, lung, testis, endometrium, and appendix) and cancer tissues (breast, colon, ovarian, lung, and prostate) .
NME1 antibodies have been validated for numerous research applications:
For IHC applications, the CPTC-NME1-2 antibody shows specific cytoplasmic staining in multiple tissue types, including various cancer tissues. This antibody has also been successfully employed in the NCI60 Cell Line Array with variable expression patterns across different cell lines .
For optimal immunohistochemistry results with NME1 antibodies, follow these methodological guidelines:
Start with a validated dilution range - 1:50 has been successfully used for antibodies like CPTC-NME1-2 on tissue microarrays
Perform antigen retrieval optimization for your specific tissue type (heat-induced epitope retrieval in citrate buffer pH 6.0 is often effective)
Include appropriate positive control tissues (colon, lung cancer, breast tissues have shown reliable staining)
Run a dilution series (1:25, 1:50, 1:100, 1:200) to determine optimal concentration for your specific tissues
For multiplex applications like imaging mass cytometry, a 1:100 dilution of 0.5mg/mL stock has shown positive results across multiple tissue types
The staining pattern should be predominantly cytoplasmic, though some nuclear staining may be observed depending on the cell type and physiological state. Always evaluate staining specificity by comparing with the known biology of NME1 and including appropriate negative controls.
To study NME1-protein interactions, several approaches have proven effective:
Pull-down assays: As demonstrated in studies of NME1-CaMKII interactions, use 6xHis-tagged proteins immobilized on nickel-nitrilotriacetic acid agarose resin and incubate with cell/tissue lysates containing NME1 or with purified NME1 protein . After washing, elute bound proteins and analyze by SDS-PAGE followed by immunoblotting with anti-NME1 antibodies.
Co-immunoprecipitation: Use anti-NME1 antibodies to immunoprecipitate NME1 along with its binding partners. For example:
Prepare cell lysates under non-denaturing conditions
Pre-clear with protein A/G beads
Incubate with anti-NME1 antibody overnight at 4°C
Capture complexes with protein A/G beads
Analyze by western blotting for potential interaction partners
Proximity ligation assays: For in situ detection of protein interactions in fixed cells or tissues, using pairs of antibodies against NME1 and potential interacting proteins.
This approach has successfully identified interactions between NME1 and proteins such as CaMKII, revealing that NME1 directly interacts with CaMKII and modulates its activity in a concentration-dependent manner .
Cross-reactivity between NME1 and NME2 is an important consideration due to their high sequence homology. Research-validated approaches to address this include:
Antibody selection: Review cross-reactivity data for your selected antibody. For example, cross-reactivity data is available for antibodies like CPTC-NME1-2
Validation controls:
Include recombinant NME1 and NME2 proteins as positive and specificity controls
For critical experiments, include NME1 knockout/knockdown samples
Use western blotting to distinguish between NME1 and NME2 based on their slight molecular weight differences
Multiple antibody approach: Validate key findings with multiple antibodies targeting different epitopes
Isoform-specific detection: When possible, target epitopes in regions that differ between NME1 and NME2
Biochemical separation: Use techniques like 2D gel electrophoresis to separate the isoforms before immunodetection
This systematic approach ensures reliable discrimination between these highly similar proteins and prevents misinterpretation of experimental results.
To investigate NME1's biphasic effect on CaMKII activity (enhancement at nanomolar concentrations and inhibition at micromolar concentrations), researchers can employ these methodological approaches:
In vitro kinase assays:
Use purified components (recombinant NME1 and CaMKII)
Test a concentration range of NME1 (20 nM to 10 μM) with constant CaMKII
Monitor activity via:
Autophosphorylation studies:
Cellular studies:
Generate cell lines with titratable NME1 expression
Measure endogenous CaMKII activity using phospho-specific antibodies
Correlate CaMKII activity with NME1 expression levels
These approaches have revealed that NME1 significantly enhances CaMKII activity at 500 nM concentration but strongly inhibits activity at 4 μM concentration. This inhibition appears independent of calmodulin concentration, suggesting NME1 does not act by sequestering calmodulin .
To investigate NME1 CoAlation (covalent modification by Coenzyme A) under oxidative stress conditions, employ the following research-validated approaches:
Recombinant protein studies:
Cellular oxidative stress models:
Transfect cells with His-tagged NME1 constructs (wild-type and C109A mutant)
Treat with oxidizing agents (diamide at 500 μM for 30 min or H₂O₂ at 100 μM to 2.5 mM for 30 min)
For metabolic stress, culture in glucose- and pyruvate-free media for 20 hours
Harvest cells with NEM to prevent post-lysis thiol modifications
Perform His-tag pull-downs using Ni-NTA resin
Mass spectrometry confirmation:
Perform affinity purification of NME1 from treated cells
Digest with trypsin and analyze by LC-MS/MS
Identify the CoAlated peptide containing Cys109
Compare spectral counts between oxidizing conditions and controls
This approach has successfully identified NME1 as a major CoA binding protein in cultured cells and tissues, with specific CoAlation occurring at Cys109 under oxidative stress conditions .
To study NME1's function in regulating the invasive transition in breast cancer, employ these methodological approaches:
Expression analysis in clinical samples:
Mechanistic studies:
Functional analysis in cell models:
In vivo validation:
Research has shown that NME1 levels drop during the progression from DCIS to invasive breast cancer, and this downregulation enhances MT1-MMP surface levels by inhibiting its endocytic clearance, thereby promoting ECM degradation and invasion .
For rigorous NME1 antibody experiments, incorporate these controls:
Positive controls:
Negative controls:
Specificity controls:
Pre-absorption with recombinant NME1 protein
Validation with multiple antibodies targeting different epitopes
Peptide competition assays
Cross-reactivity controls:
Recombinant NME1 and NME2 proteins to assess specificity
Western blot analysis to distinguish based on molecular weight
These controls ensure reliable and reproducible results across different experimental applications and prevent misinterpretation due to non-specific binding or false-positive/negative outcomes.
When encountering staining issues with NME1 antibodies, use these empirically-validated troubleshooting approaches:
For weak staining:
Optimize antibody concentration - validated dilutions for IHC include 1:50 for CPTC-NME1-2
Extend primary antibody incubation (overnight at 4°C instead of 1 hour at room temperature)
Improve antigen retrieval:
Test different buffers (citrate pH 6.0 vs. EDTA pH 9.0)
Extend retrieval time or increase temperature
Use signal amplification systems (tyramide signal amplification, polymer-based detection)
Check tissue fixation protocols (overfixation can mask epitopes)
For non-specific staining:
Optimize blocking:
Increase blocking duration (1 hour minimum)
Try different blocking agents (BSA, normal serum from detection antibody species)
Dilute antibody further
Perform more stringent washing (increased duration, higher salt concentration)
Try alternative antibodies targeting different epitopes
For IHC with avidin-biotin systems, include biotin blocking step
Following these methodological adjustments has proven effective in optimizing NME1 detection across multiple tissue types and experimental conditions .
For critical experiments requiring high confidence in antibody specificity, implement this comprehensive validation strategy:
Biochemical validation:
Perform western blot analysis to confirm detection of a band at the correct molecular weight (~17-20 kDa for NME1)
Run recombinant NME1 as positive control
Include NME2 to assess cross-reactivity
Genetic validation:
Compare samples from NME1 knockout models with wild-type
Analyze siRNA/shRNA knockdown samples alongside non-targeting controls
Use graded knockdown to establish correlation between signal and expression level
Epitope validation:
Perform peptide competition assays
Use multiple antibodies targeting different epitopes and compare staining patterns
For custom antibodies, validate with epitope-mutated constructs
Orthogonal validation:
Compare protein detection with mRNA expression (RNA-seq, qPCR, in situ hybridization)
Verify subcellular localization patterns match known biology
Perform mass spectrometry on immunoprecipitated samples
This multi-layered validation strategy ensures that experimental findings truly reflect NME1 biology rather than artifacts of antibody cross-reactivity or non-specific binding.
Imaging mass cytometry (IMC) enables highly multiplexed protein detection in tissues. For NME1 analysis via IMC, follow this methodological workflow:
Antibody preparation:
Sample preparation:
Prepare FFPE tissue sections (4-5 μm thickness)
Perform deparaffinization and antigen retrieval
Block with BSA/normal serum
Multiplex staining:
Combine metal-labeled NME1 antibody with other markers of interest
Include markers for tissue architecture (e.g., E-cadherin, cytokeratins)
Add immune cell markers if studying tumor microenvironment
Data acquisition and analysis:
Acquire data using a Hyperion Imaging System or equivalent
Process using specialized software (Visiopharm, HALO)
Perform neighborhood analysis to identify cellular interactions
This approach has been successfully applied to multiple tissue types including normal colon, liver, bone marrow, spleen, and various cancer tissues, allowing visualization of NME1 expression in spatial context .
To study the functional relationship between NME1 and CaMKII in cancer, implement this research strategy based on their concentration-dependent interaction :
Expression correlation analysis:
Perform dual immunofluorescence or sequential IHC on cancer tissue microarrays
Quantify NME1 and phospho-CaMKII (activated form) levels
Analyze correlation at different cancer stages and in different tissue compartments
Functional studies in cell models:
Generate cell lines with:
NME1 knockdown/knockout
NME1 overexpression (wild-type and binding mutants)
Inducible/titratable NME1 expression
Measure:
CaMKII activity via phospho-specific antibodies
Cancer-related phenotypes (migration, invasion, proliferation)
Rescue experiments with CaMKII inhibitors
Mechanistic investigation:
Study NME1-CaMKII complex formation at different NME1 concentrations
Analyze downstream CaMKII substrates in relation to NME1 levels
Examine how oxidative stress affects the NME1-CaMKII axis
This approach leverages the finding that NME1 enhances CaMKII activity at nanomolar concentrations but inhibits it at micromolar concentrations , which may be relevant to its concentration-dependent effects in different cancer contexts.
To investigate how CoAlation of NME1 at Cys109 affects its metastasis suppressor function, employ this comprehensive research strategy:
Cellular model development:
Generate stable cell lines expressing:
Wild-type NME1
C109A mutant (CoAlation-deficient)
C109D mutant (phosphomimetic to simulate modification)
Validate expression by western blotting with NME1 antibodies
Functional phenotype assessment:
Invasion assays (Boyden chamber, 3D matrix)
Migration assays (wound healing, single-cell tracking)
Adhesion and colony formation assays
Compare phenotypes under normal and oxidative stress conditions
Biochemical characterization:
Measure nucleoside diphosphate kinase activity of wild-type vs. mutant NME1
Analyze protein-protein interactions under oxidative stress
Assess ability to modulate CaMKII activity
In vivo studies:
Orthotopic xenograft models with wild-type or C109A mutant NME1 cells
Tail vein injection metastasis assays
Analyze primary tumors and metastases by IHC with anti-NME1 antibodies
Translational relevance:
Examine CoAlation status of NME1 in patient samples using specialized antibodies
Correlate with oxidative stress markers and metastatic outcomes
This approach connects the biochemical modification of NME1 by CoA with its established role in suppressing metastasis , potentially revealing new therapeutic strategies targeting this regulatory mechanism.
Non-Metastatic Cells 1 (NME1), also known as NM23-H1, is a gene that encodes a protein involved in various cellular processes, including cell proliferation, differentiation, and apoptosis. The study of NME1 has gained significant attention due to its role in suppressing metastasis in cancer cells. The use of mouse anti-human antibodies targeting NME1 has been instrumental in advancing our understanding of its function and potential therapeutic applications.
NME1 was first identified in the early 1990s as a gene associated with reduced metastatic potential in melanoma cells. Subsequent studies revealed that NME1 is a member of the nucleoside diphosphate kinase (NDPK) family, which plays a crucial role in maintaining cellular energy homeostasis by catalyzing the transfer of phosphate groups from nucleoside triphosphates to nucleoside diphosphates.
NME1 is involved in several critical cellular functions:
NME1 is ubiquitously expressed in various tissues, with higher expression levels observed in tissues with high proliferative capacity, such as the liver, spleen, and thymus. In cancer, NME1 expression is often downregulated in metastatic cells compared to non-metastatic cells, highlighting its role as a metastasis suppressor.
The expression and activity of NME1 are tightly regulated by multiple mechanisms:
The use of mouse anti-human NME1 antibodies has provided valuable insights into the therapeutic potential of targeting NME1 in cancer treatment. These antibodies have been used in various experimental models to study the effects of NME1 modulation on tumor growth and metastasis. The findings suggest that enhancing NME1 activity or expression could be a promising strategy for preventing cancer metastasis and improving patient outcomes.