NAIP Recombinant Monoclonal Antibody

Shipped with Ice Packs
In Stock

Description

Table 1: Key Features of NAIP Recombinant Monoclonal Antibodies

FeatureDetails
Target EpitopeNAIP amino acids 1–100 (human), Asn923–Val1148 (isoform-specific)
Host SpeciesRabbit, Mouse
ApplicationsWestern Blot (WB), Immunohistochemistry (IHC), ELISA, Immunofluorescence (IF)
ReactivityHuman, Mouse (cross-reactivity tested)
StoragePBS with 0.05% BSA, 50% glycerol; -20°C

Apoptosis and Neurological Studies

  • NAIP recombinant antibodies detect endogenous NAIP in human brain tissues (e.g., cerebellar Purkinje neurons) .

  • Validated in spinal muscular atrophy models, where NAIP mutations correlate with motor neuron apoptosis .

Inflammasome Activation

  • Antibodies identify NAIP’s role in NLRC4 inflammasome assembly during bacterial infections (e.g., Legionella pneumophila) .

Technical Performance

  • Western Blot: Detects NAIP at ~160 kDa in HEK293 lysates and human brain tissues . Observed bands at 110–150 kDa suggest splice variants or degradation .

  • IHC: Optimized protocols use heat-induced epitope retrieval and HRP-DAB staining .

Challenges and Considerations

  • Cross-Reactivity: Some antibodies show minor bands in mouse lysates .

  • Storage: Sodium azide-free formulations (e.g., Thermo Fisher’s MA5-50166) enhance compatibility with live-cell assays .

Future Directions

  • Multiplex Labeling: Subclass-switched IgG variants enable simultaneous detection of NAIP with other neuronal markers .

  • Therapeutic Potential: Engineered high-affinity NAIP antibodies may modulate inflammasome activity in autoimmune diseases .

Product Specs

Buffer
Rabbit IgG in phosphate buffered saline, pH 7.4, 150mM NaCl, 0.02% sodium azide and 50% glycerol.
Description

The NAIP recombinant monoclonal antibody is produced through in vitro expression systems, utilizing cloned DNA sequences encoding NAIP antibodies from immunoreactive rabbits. The immunogen employed in this process is a synthetic peptide derived from the human NAIP protein. Subsequently, the NAIP antibody genes are inserted into plasmid vectors, which are then transfected into host cells for antibody expression. The NAIP recombinant monoclonal antibody is purified via affinity chromatography and rigorously tested for functionality in ELISA and IHC applications, demonstrating reactivity with the human NAIP protein in these assays.

NAIP is a critical protein involved in the detection of intracellular bacterial pathogens and the initiation of an immune response through activation of the NLRC4 inflammasome. This protein plays a crucial role in the host's defense against infections caused by bacteria that utilize type III secretion systems to manipulate host cells.

Form
Liquid
Lead Time
We typically ship products within 1-3 business days after receiving your order. Delivery times may vary depending on the purchasing method or location. Please consult your local distributors for specific delivery time estimates.
Synonyms
Baculoviral IAP repeat-containing protein 1 (Neuronal apoptosis inhibitory protein), NAIP, BIRC1
Target Names
Uniprot No.

Target Background

Function

NAIP is an anti-apoptotic protein that functions by inhibiting the activities of CASP3, CASP7, and CASP9. It can inhibit the autocleavage of pro-CASP9 and cleavage of pro-CASP3 by CASP9. NAIP is capable of inhibiting CASP9 autoproteolysis at 'Asp-315' and decreasing the rate of autoproteolysis at 'Asp-330'. This protein acts as a mediator of neuronal survival in pathological conditions and prevents motor-neuron apoptosis induced by various signals. NAIP may play a role in preventing spinal muscular atrophy, which appears to be caused by inappropriate persistence of motor-neuron apoptosis. Mutated or deleted forms of NAIP have been found in individuals with severe spinal muscular atrophy.

NAIP also acts as a sensor component of the NLRC4 inflammasome, specifically recognizing and binding needle protein CprI from the pathogenic bacterium _C.violaceum_. The association of pathogenic bacterial proteins triggers the assembly and activation of the NLRC4 inflammasome, promoting caspase-1 activation, cytokine production, and macrophage pyroptosis. The NLRC4 inflammasome is activated as part of the innate immune response to a range of intracellular bacteria, such as _C.violaceum_ and _L.pneumophila_.

Gene References Into Functions
  1. Our current research suggests that NAIP may be broadly relevant to ALS symptoms as a risk factor and a promising prognostic biomarker. PMID: 29311650
  2. Our data document a previously unknown localization of NAIP throughout the entire cytokinetic process, exhibiting unique dynamic behavior. PMID: 28059125
  3. NAIP expression is most abundant in M2 macrophages, while cIAP1 and cIAP2 show an inverse pattern of expression in polarized cells. cIAP2 is preferentially expressed in M1-macrophages, and cIAP1 in M2-macrophages. Treatment with IAP antagonists of resting M0 macrophages prior to polarization stimulation induced upregulation of NAIP in M2 and downregulation of cIAP1 in both M1 and M2, but an induction of cIAP2 in M1 macrophages. PMID: 29518103
  4. Deletions in the NAIP gene are associated with spinal muscular atrophy. PMID: 27754957
  5. NAIP and survivin expressions were significantly reduced following varicocele induction compared to sham animals, whereas PDRN-treated rats showed an increase in NAIP and survivin levels. PMID: 26347229
  6. The copy numbers and gene structures of NAIP genes differed in Chinese spinal muscular atrophy patients and healthy controls. PMID: 25888055
  7. Results indicated that SMN2 and NAIP copy numbers significantly influenced the age at onset, risk of death, and life expectancy in spinal muscular atrophy patients, with SMN2 having a more pronounced effect. PMID: 25330799
  8. Human Naip functions to activate the inflammasome in response to flagellin, similar to murine Naip5/6. PMID: 26109648
  9. Modulation of chemotherapeutic drug resistance in neuroblastoma SK-N-AS cells by the neural apoptosis inhibitory protein and miR-520f. PMID: 25137037
  10. Copy number variations of SMN2 and NAIP genes in patients are related to spinal muscular atrophy clinical types (P < 0.05). PMID: 24711022
  11. /NAIP1 and NAIP2/5 formed a large oligomeric complex with NLRC4 in the presence of corresponding bacterial ligands, and could support reconstitution of the NLRC4 inflammasome in a ligand-specific manner. PMID: 23940371
  12. We identified an intronic region of the NAIP gene responding to TEAD1/YAP activity, suggesting that regulation of NAIP by TEAD1/YAP occurs at the transcriptional level. PMID: 23994529
  13. The NAIP5-NLRC4 inflammasome is induced by direct interactions with conserved N- and C-terminal regions of flagellin. PMID: 23012363
  14. NAIPFull gene duplication might have been evolutionarily maintained, or even selected for, because it may confer an advantage to the host against flagellated bacteria. PMID: 22067212
  15. There is a close relationship between SMN2, NAIP, and H4F5 gene copy number and spinal muscular atrophy disease severity. PMID: 21821450
  16. The NOD domain is essential for effective inhibition of procaspase-9 and procaspase-3 cleavage by the NAIP protein in apoptosis. PMID: 21371431
  17. NAIP is an inhibitor of procaspase-9, preventing apoptosis at the initiation stage. PMID: 20171302
  18. Expression of NAIP may be associated with enhanced survival of prostate cancer in response to castration. PMID: 20044205
  19. Our results provide the first structures of BIR domains from human NAIP and cIAP2. PMID: 19923725
  20. NAIP gene deletion was higher in type I spinal muscular atrophy than in type U or V. In type I patients lacking the NAIP gene, deterioration in their respiratory function is more rapid than in those type I patients retaining the NAIP gene. PMID: 11912351
  21. NAIP-deltaEx10-11: a novel splice variant of the apoptosis inhibitor NAIP is differentially expressed in drug-sensitive and multidrug-resistant HL60 leukemia cells. NAIP transcripts might be involved in tumor resistance to chemotherapeutic agents. PMID: 12127562
  22. NAIP:Structural requirements for binding hippocalcin and effects on survival of sympathetic neurons. PMID: 12445469
  23. NAIP does not interact with Smac and requires ATP to bind caspase-9. PMID: 15280366
  24. Alterations in C/CAAT enhancer binding protein alpha and neuronal apoptosis inhibitory protein expression occurred in human adipose stromal-vascular cells after weight loss. PMID: 15340105
  25. Multiple, domesticated long terminal repeats (LTRs) of endogenous retroviral elements provide NAIP promoter function in human, mouse, and rat. PMID: 17222062
  26. A role for NAIP in increasing the survival of cells undergoing terminal differentiation, as well as the possibility that the protein serves as an intestinal pathogen recognition protein, was suggested. PMID: 17510375
  27. 80% neuronal apoptosis inhibitory protein gene deletion in 5q-spinal muscular atrophy patients (91% spinal muscular atrophy-I, 50% spinal muscular atrophy-II and -III), and in 5% (two of forty) of spinal muscular atrophy parents, was found. PMID: 17903057
  28. While there was no evidence of NAIP expression in the normal breast tissue, NAIP was expressed in all breast cancer samples. PMID: 17923748
  29. NAIP may be a modifying factor for disease severity of spinal muscular atrophy. PMID: 17932457
  30. The present study is the first one giving detailed information on SMN and NAIP deletion rates in Iranian SMA patients. PMID: 18071605
  31. Data show elevated expression of NAIP in peripheral mononuclear cells from children with Fabry disease. PMID: 18339188
  32. hNAIP and hIpaf mediate innate intracellular defense against flagellated Legionella in human cells. PMID: 18453601
  33. The presence of one NAIP copy, that is, heterozygous NAIP deletion, was common in Vietnamese SMA, regardless of clinical phenotype. PMID: 18533950
  34. HIAP-1 and HIAP-2 mRNA levels were elevated in resting T cells while NAIP mRNA was increased in whole blood in multiple sclerosis. PMID: 18566024
  35. In glioma & glioblastoma multiforme, selective upregulation of miRNA-221 & down-regulation of a miRNA-221 mRNA target encoding BIRC1 were observed; expression of BIRC5 & caspase-3 were found to be significantly up-regulated, particularly in stage IV GBM. PMID: 18759060
  36. Data show that NAIP deletion predicts disease severity in spinal muscular atrophy. PMID: 18842367
  37. Among the SMA Type I patients, 43% showed deletions of SMN1 and NAIP. PMID: 18974562
  38. Findings of homozygous deletions of exon 7 and/or exon 8 of the SMN1 gene confirmed the diagnosis of SMA, and suggested that the deletion of SMN1 exon 7 is a major cause of SMA in southern Chinese children. PMID: 19198020
  39. A higher number of SMN2 copies makes the clinical symptoms more benign, and the NAIP gene deletion is associated with a more severe phenotype. PMID: 19287802
  40. A novel NAIP isoform derives from intragenic Alu SINE promoters. PMID: 19488400

Show More

Hide All

Database Links

HGNC: 7634

OMIM: 600355

KEGG: hsa:4671

STRING: 9606.ENSP00000428657

UniGene: Hs.646951

Tissue Specificity
Expressed in motor neurons, but not in sensory neurons. Found in liver and placenta, and to a lesser extent in spinal cord.

Q&A

What is NAIP and what are its primary biological functions?

NAIP (also known as BIRC1 or Baculoviral IAP repeat-containing protein 1) is an anti-apoptotic protein that inhibits the activities of caspases 3, 7, and 9. It prevents motor-neuron apoptosis induced by various signals and acts as a mediator of neuronal survival in pathological conditions. NAIP also functions as a sensor component of the NLRC4 inflammasome, specifically recognizing and binding needle protein CprI from pathogenic bacteria such as C. violaceum. This interaction drives assembly and activation of the NLRC4 inflammasome, promoting caspase-1 activation, cytokine production, and macrophage pyroptosis as part of the innate immune response to intracellular bacteria like C. violaceum and L. pneumophila.

How do recombinant monoclonal antibodies differ from traditional monoclonal antibodies?

Recombinant monoclonal antibodies are generated using recombinant DNA technology through in vitro cloning processes, which eliminates the need for animal immunization and hybridoma creation. Unlike traditional monoclonal antibodies that may suffer from genetic drift and instability issues, recombinant antibodies are produced from entirely defined genetic sequences with consistent performance. They offer superior batch-to-batch consistency, scalability, easy engineering/modification capabilities, and ethical advantages through animal-free production methods. These antibodies are created by inserting genes for antibody light and heavy chains into expression vectors (plasmids), which are then introduced into host cells for expression, ensuring consistent high-quality antibody production.

What applications are NAIP antibodies most commonly used for in research?

NAIP antibodies are widely utilized in various research applications including Western blot (WB), immunohistochemistry (IHC-P), immunocytochemistry/immunofluorescence (ICC/IF), and enzyme-linked immunosorbent assay (ELISA). These antibodies are particularly valuable for studying neuronal survival mechanisms, investigating spinal muscular atrophy pathology, examining inflammasome activation in immune responses, and analyzing apoptotic pathways. In specific examples, Human NAIP antibodies have been used to detect NAIP at approximately 160 kDa in transfected HEK293 cells via Western blot and to visualize NAIP in Purkinje neurons of human cerebellum through immunohistochemistry.

How should I determine the optimal antibody concentration for my Western blot experiments with NAIP antibodies?

When optimizing NAIP antibody concentration for Western blot, begin with a titration experiment using concentrations ranging from 0.5-2.0 μg/mL (based on reported optimal concentration of 1 μg/mL for Human NAIP Monoclonal Antibody in published protocols). Use positive control samples such as human brain (cerebellum) tissue or HEK293 cells transfected with human NAIP, alongside negative controls. Perform your Western blot under reducing conditions using appropriate immunoblot buffers (e.g., Immunoblot Buffer Group 2). After probing with primary antibody, use a compatible HRP-conjugated secondary antibody and visualization system. Compare signal-to-noise ratios across different concentrations to determine optimal antibody dilution that produces clear detection of the expected ~160 kDa NAIP band with minimal background. Document all parameters carefully for reproducibility in future experiments.

What tissue preparation and antigen retrieval methods are recommended for immunohistochemical detection of NAIP?

For optimal immunohistochemical detection of NAIP in tissues, begin with proper fixation using 10% neutral buffered formalin for 24-48 hours followed by paraffin embedding. Section tissues at 5-7 μm thickness and mount on positively charged slides. Heat-induced epitope retrieval is critical - use a basic antigen retrieval solution (pH 9.0) and heat at 95-100°C for 20 minutes, followed by cooling to room temperature. For NAIP detection in neural tissues such as cerebellum, an overnight incubation at 4°C with primary antibody (recommended concentration 10-15 μg/mL) has been shown to be effective. Use an appropriate detection system such as HRP-DAB for visualization with hematoxylin counterstaining. This protocol has successfully demonstrated specific NAIP staining in Purkinje neurons in human cerebellum samples.

How can I validate the specificity of my NAIP antibody before proceeding with complex experiments?

A comprehensive validation strategy for NAIP antibodies should include multiple complementary approaches. First, perform Western blot analysis comparing NAIP-expressing samples (e.g., brain tissue or transfected cells) with appropriate negative controls, looking for a specific band at ~160 kDa. Second, conduct knockdown/knockout validation by testing the antibody on samples with NAIP expression reduced through siRNA or CRISPR-Cas9 technologies. Third, implement peptide competition assays by pre-incubating the antibody with the immunizing peptide prior to application. Fourth, verify results across multiple detection methods (e.g., if results are observed in Western blot, confirm with immunohistochemistry). Finally, compare reactivity patterns with alternative antibodies targeting different epitopes of NAIP. Document all validation steps with quantitative measures of specificity and include these controls in your experimental reports.

How can I optimize co-immunoprecipitation protocols to study NAIP interactions with inflammasome components?

For investigating NAIP interactions with NLRC4 inflammasome components, implement a carefully optimized co-immunoprecipitation protocol. Begin with appropriate cell models (e.g., macrophages stimulated with bacterial ligands or HEK293 cells co-transfected with NAIP and potential interacting partners). Lyse cells in a gentle, non-denaturing buffer (25 mM Tris-HCl pH 7.4, 150 mM NaCl, 1% NP-40, 1 mM EDTA, protease inhibitors) at 4°C. Pre-clear lysates with protein A/G beads before immunoprecipitation with anti-NAIP antibody (recommend 5 μg antibody per 500 μg total protein) overnight at 4°C. For optimal specificity, use recombinant monoclonal NAIP antibodies that have been validated for immunoprecipitation. After washing, elute complexes and analyze by Western blot using antibodies against potential interacting partners (NLRC4, caspase-1, ASC). Include IgG control immunoprecipitations and input samples to demonstrate specificity. Consider crosslinking approaches for transient interactions and native PAGE for intact inflammasome complexes.

What are the critical considerations when designing experiments to study NAIP's role in neuronal survival using recombinant antibodies?

When investigating NAIP's role in neuronal survival, several critical experimental design factors must be considered. First, select appropriate neuronal models that express NAIP endogenously (primary motor neurons or relevant neuronal cell lines) and establish reliable stress induction protocols (excitotoxicity, oxidative stress, or growth factor deprivation). For loss-of-function studies, implement siRNA knockdown or CRISPR-Cas9 knockout of NAIP, validated by both qRT-PCR and Western blot using well-characterized NAIP recombinant antibodies. For gain-of-function studies, use lentiviral or AAV vectors for stable NAIP overexpression. Assess neuronal survival through multiple complementary assays (e.g., MTT/XTT viability assays, TUNEL staining, caspase activity measurements, and assessment of mitochondrial membrane potential). To connect NAIP to downstream pathways, monitor caspase-3, -7, and -9 activation states using specific antibodies or activity assays. Finally, include temporal analyses to distinguish immediate versus long-term effects of NAIP modulation on neuronal survival pathways.

How should I approach comparative analysis of NAIP expression in spinal muscular atrophy patient samples versus controls?

When analyzing NAIP expression in spinal muscular atrophy (SMA) patient samples compared to controls, implement a systematic approach that accounts for the unique challenges of this research. First, carefully match patient and control samples for age, sex, postmortem interval, and tissue preservation methods. Use multiple detection methods: quantitative immunoblotting with recombinant NAIP antibodies for protein quantification, RT-qPCR for transcript analysis, and immunohistochemistry for spatial distribution assessment. For immunohistochemical analysis of spinal cord sections, employ heat-induced epitope retrieval with basic pH buffer and overnight primary antibody incubation at 4°C (10-15 μg/mL), followed by appropriate detection systems. Quantify NAIP expression in motor neurons specifically through co-staining with motor neuron markers (e.g., ChAT). Calculate the percentage of NAIP-positive motor neurons and measure intensity values using digital image analysis software. Correlate NAIP expression with clinical parameters and SMN1/SMN2 gene status to establish meaningful associations. Include comprehensive controls and blinded quantification to strengthen findings.

What strategies can resolve inconsistent Western blot results when detecting NAIP protein?

Inconsistent Western blot results for NAIP detection typically stem from several technical challenges. First, optimize protein extraction by using specialized lysis buffers containing protease inhibitors that effectively preserve the ~160 kDa NAIP protein. RIPA buffer with complete protease inhibitor cocktail works well for most applications. Second, adjust sample preparation by avoiding excessive heating (limit to 70°C for 5 minutes) and using fresh DTT or β-mercaptoethanol as reducing agents. Third, for gel electrophoresis, use lower percentage gels (6-8%) to better resolve the high molecular weight NAIP protein and extend transfer time (overnight at low voltage) when transferring to PVDF membranes. Fourth, implement stringent blocking (5% BSA rather than milk) to reduce background. Fifth, if sensitivity is an issue, consider signal amplification systems or higher primary antibody concentration (1-2 μg/mL). Finally, compare results using different validated NAIP antibodies targeting distinct epitopes, as certain epitopes may be masked by protein interactions or post-translational modifications in your specific samples.

How can I differentiate between specific and non-specific staining in NAIP immunohistochemistry?

Differentiating specific from non-specific staining in NAIP immunohistochemistry requires a comprehensive validation approach. First, implement essential controls: (1) negative controls omitting primary antibody, (2) isotype controls using non-specific antibodies of the same isotype, and (3) peptide competition assays where pre-incubation of the antibody with the immunizing peptide should abolish specific staining. Second, compare staining patterns with known NAIP expression data - for example, Purkinje neurons in cerebellum should show positive staining based on published results. Third, use NAIP-overexpressing and knockdown/knockout tissues or cell blocks as positive and negative controls. Fourth, perform dual-labeling experiments with established cell-type specific markers (e.g., neuronal markers when examining brain tissue) to confirm that NAIP localization aligns with expected cellular distribution. Fifth, evaluate staining using multiple NAIP antibodies targeting different epitopes - consistent localization patterns across different antibodies strongly supports specificity. Finally, quantify and compare signal-to-background ratios across all controls to establish objective criteria for distinguishing specific from non-specific signals.

What are the potential causes and solutions for detecting multiple bands when using NAIP antibodies in Western blot?

The detection of multiple bands when using NAIP antibodies in Western blot can result from several factors with specific solutions for each. First, NAIP splice variants - human NAIP gene produces multiple isoforms; characterize these using RT-PCR and sequence verification, then compare migration patterns with predicted molecular weights of known variants. Second, post-translational modifications - examine phosphorylation, ubiquitination, or other modifications using phosphatase treatments or specific modification detection reagents. Third, protein degradation - implement more rigorous sample handling (maintain samples at 4°C, add additional protease inhibitors, avoid freeze-thaw cycles) and compare fresh versus stored samples. Fourth, cross-reactivity with related proteins - perform peptide competition assays and test antibody specificity using NAIP knockout/knockdown samples. Fifth, incomplete denaturation - optimize sample preparation conditions including SDS concentration, reducing agent strength, and heat treatment duration. Create a detailed table documenting the molecular weights of all observed bands across different sample types, and systematically test each potential cause through controlled experiments. When reporting results, clearly indicate which band represents full-length NAIP (~160 kDa) and provide evidence supporting the identity of any additional bands.

How do recombinant monoclonal and polyclonal NAIP antibodies compare in detecting low abundance NAIP in different experimental contexts?

For optimal detection of low abundance NAIP, implement signal amplification methods appropriate to each antibody type. With recombinant monoclonals, use high-sensitivity detection systems (e.g., SuperSignal West Femto) and longer exposure times. For polyclonals, optimize blocking conditions and use more stringent washing to minimize background. When developing new assays, systematically compare both antibody types on identical samples using standardized protocols, documenting specificity (signal-to-noise ratio), sensitivity (detection limit), and reproducibility metrics for each application context.

What are the optimal methodological approaches for studying NAIP's interaction with the NLRC4 inflammasome using recombinant antibodies?

Studying NAIP's interaction with NLRC4 inflammasome components requires a multi-faceted methodological approach. Begin with co-immunoprecipitation using recombinant monoclonal NAIP antibodies in macrophages stimulated with relevant bacterial ligands (e.g., flagellin or needle proteins from C. violaceum). Use gentle lysis conditions (1% NP-40 or digitonin-based buffers) to preserve protein-protein interactions and confirm results through reciprocal immunoprecipitation with NLRC4 antibodies.

For spatial analysis, implement proximity ligation assays (PLA) or FRET microscopy using fluorescently-tagged recombinant antibodies against NAIP and NLRC4, allowing visualization of interactions in situ. Complement these approaches with functional assays measuring inflammasome assembly kinetics and downstream effects (caspase-1 activation, IL-1β secretion, pyroptosis) in response to specific bacterial ligands, comparing wild-type cells to those with NAIP knockdown/knockout.

For biochemical characterization, use recombinant purified domains of NAIP and NLRC4 in conjunction with surface plasmon resonance (SPR) or microscale thermophoresis (MST) to determine binding affinities and kinetics. Finally, validate key findings using reconstitution experiments in HEK293 cells expressing fluorescently tagged NAIP and NLRC4 variants to visualize complex formation dynamics in real-time through live-cell imaging.

What experimental design is recommended for studying NAIP expression changes in neurodegenerative disease models using recombinant antibodies?

For investigating NAIP expression changes in neurodegenerative disease models, implement a comprehensive experimental design combining multiple analytical approaches. First, select appropriate disease models (transgenic animals, induced pluripotent stem cell-derived neurons from patients, or relevant cell lines under disease-mimicking stressors) with proper controls. Establish a temporal analysis framework to track NAIP expression changes throughout disease progression by collecting samples at multiple time points.

Utilize quantitative Western blot with recombinant NAIP monoclonal antibodies (1 μg/mL concentration) for protein quantification, normalizing to appropriate housekeeping proteins and implementing technical replicates. Complement this with RT-qPCR to assess transcript levels and determine whether changes occur at transcriptional or post-transcriptional levels. For spatial analysis, perform immunohistochemistry on brain sections using optimized antigen retrieval (heat-induced at basic pH) and antibody concentration (10-15 μg/mL), followed by detailed image analysis quantifying both the number of NAIP-positive cells and staining intensity within specific neuronal populations.

Correlate NAIP expression with functional outcomes (neuronal survival, electrophysiological properties) and disease-specific pathological markers (protein aggregates, inflammatory markers) through co-staining experiments. Finally, implement intervention studies using NAIP overexpression or knockdown to establish causality between NAIP expression changes and disease phenotypes, documenting effects on neuronal survival pathways and caspase activation.

Protocol Optimization Table for NAIP Antibody Applications

ApplicationRecommended Antibody TypeOptimal ConcentrationCritical Buffer ComponentsValidation ControlsTroubleshooting Tips
Western BlotRecombinant Monoclonal1-2 μg/mLReducing conditions, Immunoblot Buffer Group 2HEK293 cells transfected with human NAIP, Human brain tissueUse 6-8% gels for better resolution of 160 kDa band; extended transfer time for large proteins
ImmunohistochemistryRecombinant Monoclonal10-15 μg/mLHeat-induced epitope retrieval with basic pH bufferHuman cerebellum (Purkinje neurons), Peptide competitionOvernight incubation at 4°C; specific staining in Purkinje neurons
ImmunofluorescenceRecombinant Monoclonal or Polyclonal5-10 μg/mL0.1% Triton X-100 permeabilizationNAIP transfected vs. non-transfected cellsUse confocal microscopy for subcellular localization studies
ImmunoprecipitationRecombinant Monoclonal5 μg per 500 μg proteinNon-denaturing lysis buffer (1% NP-40)IgG control IP, Input samplesPre-clear lysates thoroughly; use protein A/G mix beads
Flow CytometryRecombinant Monoclonal1-5 μg per 10^6 cells0.5% BSA in PBSIsotype control, NAIP knockdown cellsFix with 2% paraformaldehyde; avoid methanol fixation
ELISARecombinant Monoclonal1-2 μg/mL for captureCarbonate buffer (pH 9.6) for coatingStandard curve with recombinant NAIPOptimize blocking (5% BSA) to reduce background

How might NAIP antibodies be utilized in developing novel biomarkers for neurodegenerative diseases?

The development of NAIP-based biomarkers for neurodegenerative diseases represents a promising research direction. NAIP's role in preventing motor neuron apoptosis and its altered expression in conditions such as spinal muscular atrophy positions it as a candidate biomarker for neurodegeneration. Future research should focus on developing ultrasensitive detection methods using recombinant monoclonal antibodies to quantify NAIP in accessible biospecimens like cerebrospinal fluid or blood exosomes.

A methodological approach would include developing sandwich ELISA or digital ELISA (Simoa) assays using recombinant monoclonal antibodies targeting different NAIP epitopes, with optimization for detection in biological fluids. These assays should be validated against gold standard diagnostic methods and evaluated in longitudinal cohorts to determine sensitivity and specificity for disease prediction, progression monitoring, and treatment response assessment.

Additional research should investigate NAIP post-translational modifications as potential disease-specific markers and develop antibodies specifically recognizing these modified forms. Integration of NAIP measurement with other biomarkers through machine learning approaches may enhance diagnostic accuracy and disease stratification potential.

What methodological considerations are important when developing recombinant antibodies against different NAIP epitopes for inflammasome research?

Developing recombinant antibodies against different NAIP epitopes for inflammasome research requires strategic epitope selection and rigorous validation approaches. First, perform comprehensive sequence analysis to identify conserved domains versus variable regions, targeting epitopes within NACHT domain (for inflammasome assembly studies), BIR domains (for caspase interaction studies), or LRR domain (for ligand recognition studies). For bacterial ligand binding studies, prioritize epitopes outside the ligand-binding region to avoid competition with bacterial components.

The recombinant antibody generation process should utilize phage display or similar in vitro selection methods with stringent screening against both recombinant NAIP domains and native NAIP in cellular contexts. Functional validation is critical - assess whether antibodies interfere with or enhance NAIP interactions with bacterial ligands, NLRC4, or downstream inflammasome components. For research applications, develop panels of non-competing antibodies recognizing distinct epitopes to enable simultaneous detection of multiple NAIP interactions.

Optimize antibody formats based on application needs - fragment antibodies (Fab, scFv) may better access epitopes in complex assemblies, while full IgG formats typically provide stronger signals in standard assays. Finally, characterize each antibody's performance in various buffer conditions relevant to inflammasome assembly (varying pH, ion concentrations, detergents) to ensure reliable performance across experimental conditions.

What experimental approaches would best utilize NAIP recombinant antibodies to investigate the relationship between neuronal survival and inflammasome activity?

Investigating the relationship between NAIP's dual roles in neuronal survival and inflammasome regulation requires sophisticated experimental approaches leveraging recombinant antibodies. Design a multi-compartment neuronal culture system (microfluidic devices) separating neuronal cell bodies from microglial cells, allowing selective manipulation and analysis of each compartment. Utilize cell-specific promoters to express fluorescently-tagged NAIP variants in either neurons or microglia, allowing real-time visualization with live-cell imaging.

Implement proximity labeling techniques (BioID or APEX2) fused to NAIP in different cellular contexts to identify cell-type-specific interaction partners, followed by validation using co-immunoprecipitation with recombinant NAIP antibodies. Develop FRET-based biosensors using recombinant antibody fragments to monitor NAIP conformational changes upon interaction with different binding partners (caspases versus bacterial ligands) in living neurons and immune cells.

Establish conditional and cell-type-specific NAIP knockout models using CRISPR-Cas9, followed by comprehensive phenotyping using recombinant antibodies to assess impacts on both neuronal survival (caspase activation, apoptotic markers) and inflammasome function (ASC speck formation, IL-1β production). This approach will help determine whether NAIP's functions in neuronal protection and inflammasome regulation represent distinct mechanisms or interconnected pathways that could be therapeutically targeted in neuroinflammatory and neurodegenerative conditions.

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.