NPAS1 Antibody

Shipped with Ice Packs
In Stock

Description

Introduction to NPAS1 Antibody

NPAS1 (Neuronal PAS Domain Protein 1) antibodies are specialized immunological tools used to detect and study the NPAS1 transcription factor, which plays critical roles in neurodevelopment and neuropsychiatric disorders. NPAS1 is a basic helix–loop–helix transcription factor expressed in GABAergic neurons, particularly in the basal forebrain (BF), globus pallidus, and cortical interneurons . These antibodies enable researchers to map NPAS1-expressing neuronal populations, investigate their functional roles, and explore their implications in conditions such as insomnia, dementia, and stress-related disorders .

3.1. Neurochemical Phenotyping

NPAS1 antibodies have been instrumental in identifying GABAergic neuronal subpopulations:

  • GABAergic specificity: 75–87% of NPAS1+ neurons in the BF colocalize with GAD67-GFP (a GABAergic marker) . Minimal overlap with glutamatergic (vGlut1/2) or cholinergic (ChAT) markers .

  • Density comparisons: NPAS1+ neurons are 5–6× more abundant than parvalbumin (PV)+ or cholinergic neurons in the BF .

3.2. Functional Insights

  • Sleep-wake regulation: Chemogenetic activation of BF NPAS1+ neurons increases wakefulness and disrupts NREM sleep oscillations, mimicking sleep disturbances in neuropsychiatric disorders .

  • Motivational circuits: NPAS1+ neurons project to the lateral hypothalamus, ventral tegmental area, and nucleus accumbens, linking them to reward and stress responses .

3.3. Developmental Role

NPAS1 antibodies reveal sustained expression in adulthood, suggesting a role in maintaining neuronal identity:

  • 75% of NPAS1-tdTomato+ neurons in adult mice retain NPAS1 protein, particularly in the horizontal diagonal band (HDB) and magnocellular preoptic nucleus (MCPO) .

  • No colocalization with PV (~0% overlap in GPe) , confirming NPAS1+ neurons as a distinct class.

Validation and Technical Considerations

  • Specificity: NPAS1 antibodies show no cross-reactivity in NPAS1 knockout mice .

  • Challenges: Lower antibody penetration in fiber-dense regions like the substantia innominata/ventral pallidum (SI/VP) may underestimate NPAS1+ neurons .

  • Protocols: Staining often requires amplification (e.g., anti-RFP for tdTomato signals) and nuclear localization due to NPAS1’s role as a transcription factor .

Implications for Neuropsychiatric Research

NPAS1 antibodies are critical for studying:

  • Stress-induced insomnia: Linked to hyperactivity of BF NPAS1+ neurons .

  • Dementia: NPAS1+ neuron degeneration correlates with sleep and cognitive deficits .

  • Circuit mapping: Anterograde tracing using NPAS1 antibodies highlights connections to olfactory and limbic systems .

Product Specs

Buffer
PBS with 0.02% Sodium Azide, 50% Glycerol, pH 7.3. Store at -20°C. Avoid freeze-thaw cycles.
Lead Time
Typically, we can ship your order within 1-3 business days of receiving it. Delivery times may vary depending on the method of purchase and location. Please contact your local distributor for specific delivery information.
Synonyms
NPAS1 antibody; BHLHE11 antibody; MOP5 antibody; PASD5 antibody; Neuronal PAS domain-containing protein 1 antibody; Neuronal PAS1 antibody; Basic-helix-loop-helix-PAS protein MOP5 antibody; Class E basic helix-loop-helix protein 11 antibody; bHLHe11 antibody; Member of PAS protein 5 antibody; PAS domain-containing protein 5 antibody
Target Names
NPAS1
Uniprot No.

Target Background

Function
NPAS1 antibody may regulate pathways involved in schizophrenia and psychotic illness. It might play a role in late central nervous system development by modulating EPO expression in response to cellular oxygen levels. NPAS1 forms a heterodimer with ARNT, which binds to the core DNA sequence 5'-TACGTG-3' within the hypoxia response element (HRE). This binding leads to transcriptional repression of its target gene, TH.
Gene References Into Functions
  1. A comprehensive study revealed the full spectrum of genes regulated by NPAS1 and NPAS3, demonstrating their role as master regulators of neuropsychiatric function. This research sheds light on the molecular pathophysiology of NPAS1/3 mutations and highlights the shared, combinatorial nature of molecular pathways involved in distinct neuropsychiatric conditions. PMID: 28499489
  2. Researchers examined the crystal structures of multi-domain NPAS1-ARNT and NPAS3-ARNT-DNA complexes. Their findings revealed four putative ligand-binding pockets within each complex. PMID: 27782878
Database Links

HGNC: 7894

OMIM: 603346

KEGG: hsa:4861

STRING: 9606.ENSP00000405290

UniGene: Hs.79564

Subcellular Location
Nucleus.

Q&A

What is NPAS1 and why is it important in neuroscience research?

NPAS1 (Neuronal PAS domain protein 1) is a basic helix-loop-helix class transcription factor expressed primarily in inhibitory interneurons in the brain . It plays a critical role in regulating the ratio of cortical excitatory and inhibitory neurons during development . NPAS1 is important in neuroscience research because:

  • It functions as a negative regulator of interneuron progenitor proliferation, particularly affecting somatostatin+ (SST) and vasoactive intestinal polypeptide+ (VIP) interneuron populations

  • Genetic studies have linked NPAS1 to neuropsychiatric disorders

  • NPAS1 identifies distinct neuronal subpopulations in multiple brain regions, including the basal forebrain and globus pallidus externa (GPe)

  • NPAS1-deficient mice exhibit behavioral abnormalities including diminished startle response, impaired social recognition, and increased locomotor activity

What are the common applications for NPAS1 antibodies in neuroscience research?

NPAS1 antibodies are utilized across multiple neuroscience research applications:

  • Immunohistochemistry (IHC): For cellular localization studies in brain tissue sections, particularly useful for identifying specific neuronal subpopulations

  • Immunofluorescence (IF): Often used in co-labeling experiments to determine overlap with other neuronal markers like GABA, GAD67, calretinin, and cell-type specific markers

  • Western blot (WB): For quantitative assessment of NPAS1 protein expression in brain tissue samples

  • Immunocytochemistry (ICC): For examining NPAS1 expression in cultured neurons

These applications are essential for understanding NPAS1's role in neuronal development, function, and in pathological conditions related to neuropsychiatric disorders.

How is NPAS1 expression distributed in the mammalian brain?

NPAS1 exhibits a specific expression pattern across different brain regions:

  • Cortex: Expressed in a relatively small number of cells compared to the large excitatory pyramidal neurons, primarily in inhibitory interneurons in layers 1, 4, and 5

  • Hippocampus and dentate gyrus: Expressed in specific populations of inhibitory interneurons

  • Basal forebrain (BF): Expressed in a major subpopulation of GABAergic neurons distinct from parvalbumin+ or cholinergic neurons, with density 5-6 times higher than neighboring cell types

  • Globus pallidus externa (GPe): Forms a distinct class (27% of GPe neurons) separate from parvalbumin-expressing neurons (55% of GPe neurons)

  • Ventral pallidum (VP): Identifies a neuronal population involved in stress responses

  • Subpallium: Expressed in progenitor domains of the mouse basal ganglia, including medial and caudal ganglionic eminences (MGE and CGE)

Nearly all NPAS1-positive cells co-stain with GABA or glutamic acid decarboxylase 67 (GAD-67), confirming their identity as inhibitory neurons .

What are the optimal methods for detecting NPAS1 in brain tissue sections?

For optimal NPAS1 detection in brain tissue sections, consider these methodological approaches:

  • Immunohistochemical staining:

    • Nuclear localization: As NPAS1 is a transcription factor, proper nuclear staining is critical. Use antigen retrieval methods that optimize nuclear protein detection

    • Antibody selection: Use well-validated NPAS1 antibodies that have been tested in knockout tissue as negative controls

    • Signal amplification: Consider using a fluorescent secondary antibody system with signal amplification for weaker signals, particularly in fiber-dense regions like substantia innominata/ventral pallidum (SI/VP)

  • Alternative genetic approaches:

    • β-galactosidase staining: In NPAS1 knockout mice where β-galactosidase gene is placed in frame with truncated NPAS1, this reporter can be used to visualize the expression pattern

    • Transgenic reporter lines: Using Npas1-Cre-2A-tdTomato mice allows visualization of both developmental and current expression

The choice of detection method depends on research questions. For co-localization studies with other markers, double immunofluorescence is preferred. When quantifying cell populations, immunohistochemistry followed by stereological counting provides accurate results .

How should researchers approach co-labeling experiments involving NPAS1?

When designing co-labeling experiments involving NPAS1, consider these methodological approaches:

  • Selection of appropriate markers:

    • GABAergic markers: GAD67-GFP knock-in mice or anti-GABA antibodies for confirming inhibitory phenotype

    • Neuronal subtype markers: Anti-parvalbumin, anti-calretinin, anti-ChAT for distinguishing specific neuronal populations

    • Developmental markers: Lhx6, Nkx2.1 for determining developmental lineage

  • Technical considerations:

    • Primary antibody compatibility: Use NPAS1 antibodies raised in different host species than other target antibodies to prevent cross-reactivity

    • Nuclear vs. cytoplasmic labeling: NPAS1 shows nuclear localization while many other markers are cytoplasmic, requiring careful imaging to establish co-localization

    • Sequential staining protocol: When using multiple antibodies from the same host species, employ sequential staining with intermediate blocking steps

  • Validation approaches:

    • Use transgenic reporter lines for cross-validation (e.g., GAD67-GFP/Npas1-cre-2A-tdTomato crossed mice)

    • Include appropriate controls (primary antibody omission, isotype controls)

    • Validate findings using multiple independent approaches

For quantification of co-labeled cells, analyze at least 3-5 sections per brain region, with balanced sampling across rostral-caudal extent, as regional differences in co-expression have been observed .

What controls should be included when using NPAS1 antibodies?

Robust experimental design requires appropriate controls when using NPAS1 antibodies:

  • Essential negative controls:

    • NPAS1 knockout tissue: The gold standard negative control that confirms antibody specificity

    • Primary antibody omission: Controls for non-specific binding of secondary antibodies

    • Isotype controls: Using non-specific IgG from the same species at matching concentration

  • Positive controls:

    • Brain regions with known high NPAS1 expression (GPe, specific layers of neocortex, dentate gyrus)

    • Heterozygous NPAS1 animals: Show intermediate staining intensity between wild-type and knockout, confirming antibody specificity and dose-dependence

  • Validation strategies:

    • Cross-validation with multiple antibodies targeting different epitopes of NPAS1

    • Comparison with genetic reporters (e.g., Npas1-cre driven reporter expression)

    • RNA expression correlation: Compare protein detection with in situ hybridization patterns for NPAS1 mRNA

The search results demonstrate the importance of comprehensive controls, exemplified by studies that confirmed antibody specificity through diminished staining in NPAS1 heterozygotes and eliminated staining in NPAS1 homozygous knockout tissue .

How can researchers effectively manipulate NPAS1-expressing neurons for functional studies?

For functional manipulation of NPAS1-expressing neurons, several approaches have been successfully implemented:

  • Chemogenetic manipulation:

    • DREADD (Designer Receptors Exclusively Activated by Designer Drugs) technology using Cre-dependent viral vectors in Npas1-Cre mice

    • Successful applications include:

      • AAV5-hSyn-DIO-hM3Dq-mCherry for excitation of Npas1+ neurons

      • AAV5-hSyn-DIO-hM4Di-mCherry for inhibition of Npas1+ neurons

      • AAV5-hSyn-DIO-eYFP as control vector

  • Circuit mapping approaches:

    • Anterograde tracing: Identifying projection targets of NPAS1+ neurons through Cre-dependent viral tracing

    • Optogenetic manipulation: Cre-dependent channelrhodopsin or halorhodopsin expression for precise temporal control

  • Experimental design considerations:

    • Behavioral paradigms: Studies have successfully examined effects on stress response , sleep-wake behavior , and cortical EEG patterns

    • Control populations: Include both NPAS1-negative neurons and sham manipulations

    • Temporal specificity: Consider both acute and chronic manipulation paradigms

Recent studies have demonstrated that chemogenetic activation of NPAS1+ neurons in the ventral pallidum increases susceptibility to social defeat stress, while inhibition enhances resilience . In the basal forebrain, activation increases wakefulness and disrupts NREM sleep oscillations .

What are the molecular mechanisms through which NPAS1 regulates interneuron development?

The molecular mechanisms by which NPAS1 regulates interneuron development involve several complex pathways:

  • Transcriptional regulation:

    • NPAS1 functions as a transcriptional repressor that targets key developmental genes

    • Direct repression of Arx enhancer activity: NPAS1 negatively regulates Arx expression, which is critical for proper interneuron development

    • Complementary relationship with NPAS3: While NPAS1 acts as a negative regulator of proliferation, NPAS3 appears to promote proliferation in ganglionic eminence progenitors

  • Signaling pathway interactions:

    • MAPK/ERK pathway modulation: NPAS1-/- mutants show increased ERK signaling in MGE and CGE progenitors

    • Not through direct FGF receptor regulation, but potentially through downstream effectors

  • Developmental consequences:

    • Selective effects on interneuron subtypes: NPAS1 deficiency leads to overproduction of specific interneuron populations (SST+ and VIP+) while sparing others (PV+)

    • Regional specificity: Effects are most prominent in superficial cortical layers (I and II/III)

    • Long-term consequences: Developmental changes persist into adulthood, affecting cortical inhibitory tone

The research indicates NPAS1 functions within a transcriptional network that precisely regulates the balance of different interneuron subtypes during development, with lasting consequences for adult cortical function and potentially contributing to neurodevelopmental disorder pathophysiology.

How do NPAS1-expressing neurons differ functionally from other neuronal populations in the same brain regions?

NPAS1-expressing neurons exhibit distinct functional properties compared to other neuronal populations in the same brain regions:

  • In the globus pallidus externa (GPe):

    • NPAS1+ neurons are distinct from parvalbumin+ (PV+) neurons in their:

      • Projection patterns: NPAS1+ neurons project primarily to striatum, while PV+ neurons project to subthalamic nucleus

      • Firing characteristics: Different autonomous and driven firing patterns

      • Expression of intrinsic ion conductances

      • Response to dopamine depletion: Different responsiveness to 6-hydroxydopamine lesion

  • In the basal forebrain (BF):

    • NPAS1+ neurons are distinct from cholinergic and PV+ neurons:

      • Size and density: NPAS1+ neurons are 5-6 times more numerous than PV+, ChAT+, or vGlut2+ neurons

      • Projection targets: BF NPAS1+ neurons project to regions involved in sleep-wake control, motivated behavior, and olfaction

      • Functional effects: Activation promotes wakefulness and disrupts NREM sleep and cortical oscillations

  • In the ventral pallidum (VP):

    • NPAS1+ neurons mediate stress susceptibility

    • Project to nucleus accumbens, ventral tegmental area, habenula, and other regions involved in reward processing

These functional differences highlight the importance of using molecular markers like NPAS1 to identify and study specific neuronal subpopulations with distinct roles in circuit function and behavior.

How should researchers address contradictory findings regarding NPAS1 expression patterns?

When confronting contradictory findings regarding NPAS1 expression patterns, researchers should implement a systematic approach:

  • Consider methodological differences:

    • Detection techniques: Different results may arise from using antibody detection versus genetic reporter systems

    • Antibody selection: Variable epitope targeting can yield different detection patterns

    • Tissue preparation: Differences in fixation methods can affect nuclear protein detection

    • Species and age differences: NPAS1 expression patterns may vary across species and developmental stages

  • Specific contradictions highlighted in the literature:

    • Overlap with PV expression: Some studies report minimal overlap between NPAS1 and parvalbumin (1-3%) , while others report higher overlap (up to 12.6%)

    • GABAergic nature of NPAS1+ neurons: While immunohistochemistry consistently shows co-localization with GABAergic markers , transcript analysis using ribosome-associated mRNA in Npas1-cre-Ribotag mice found GABAergic markers were not enriched in VP Npas1+ neurons

  • Resolution strategies:

    • Use multiple independent techniques (e.g., immunohistochemistry, in situ hybridization, reporter lines)

    • Perform careful regional analysis, as expression patterns may differ across brain regions

    • Consider developmental timing, as NPAS1 is a developmental transcription factor with potentially changing expression over time

    • Validate findings using genetic knockout controls and multiple antibodies

For example, researchers resolved discrepancies in PV/NPAS1 overlap through careful quantification across different rostro-caudal levels, finding regional variation in co-expression rates .

What are the key considerations for analyzing NPAS1 knockout phenotypes?

When analyzing NPAS1 knockout phenotypes, researchers should consider several important factors:

  • Developmental vs. acute effects:

    • NPAS1 is a developmental transcription factor, so knockout effects may reflect developmental alterations rather than acute functional requirements

    • Consider using conditional/inducible knockout systems to distinguish between these possibilities

  • Compensation and redundancy:

    • NPAS1 interacts with NPAS3, with partially complementary functions

    • Single knockout phenotypes may be moderated by compensatory mechanisms

    • Double knockout models (NPAS1/NPAS3) may reveal more pronounced phenotypes

  • Cell-type specific effects:

    • NPAS1 regulates specific interneuron subtypes (SST+ and VIP+), while sparing others (PV+)

    • Effects on excitatory/inhibitory balance: NPAS1-/- mice show specific increases in inhibitory tone in superficial cortical layers

    • Regional specificity: Phenotypes may vary across brain regions where NPAS1 is expressed

  • Behavioral interpretation:

    • NPAS1 and NPAS1/NPAS3 knockout mice show complex behavioral phenotypes including:

      • Diminished startle response (prepulse inhibition)

      • Impaired social recognition

      • Stereotypic behavior and increased locomotor activity

    • These behaviors require careful interpretation in relation to specific circuit alterations

  • Molecular correlates:

    • NPAS1/NPAS3-deficient mice show reductions in reelin expression

    • Consider downstream molecular changes that may mediate observed phenotypes

The research suggests NPAS1 knockout effects should be interpreted within a developmental neurobiology framework, recognizing that adult phenotypes likely reflect altered circuit formation rather than just altered gene expression in mature neurons.

What technical challenges are associated with studying NPAS1 in fiber-dense brain regions?

Studying NPAS1 in fiber-dense brain regions presents several technical challenges that researchers must address:

  • Antibody penetration limitations:

    • Fiber-dense regions like substantia innominata/ventral pallidum (SI/VP) show lower apparent co-localization of NPAS1 protein with genetic reporters (67.51 ± 5.13%) compared to less fiber-dense regions like horizontal limb of the diagonal band (HDB) (87.07 ± 3.12%)

    • This discrepancy is likely due to poor antibody penetration rather than biological differences

  • Signal detection optimization:

    • Nuclear localization: As a transcription factor, NPAS1 is localized to the nucleus, requiring nuclear-optimized staining protocols

    • Signal amplification: Consider using tyramide signal amplification or other enhancement techniques for fiber-dense regions

    • Thin section preparation: Use optimal section thickness (30-40μm) to balance antibody penetration with structural preservation

  • Data interpretation considerations:

    • Quantification challenges: Standard stereological methods may be compromised in fiber-dense regions

    • Internal controls: Include less fiber-dense regions in the same section as controls

    • Cross-validation: Supplement immunohistochemistry with in situ hybridization or genetic reporter approaches

  • Solutions from the literature:

    • RFP antibody amplification of tdTomato signal in genetic reporter lines

    • Multiple antibody combinations targeting different epitopes

    • Recognition that quantification in fiber-dense regions likely underestimates true expression levels

Researchers should acknowledge these technical limitations in their interpretations and consider that reported co-localization rates in fiber-dense regions may represent lower bounds rather than absolute values.

How can NPAS1 research inform our understanding of neuropsychiatric disorders?

NPAS1 research provides valuable insights into neuropsychiatric disorder mechanisms:

  • Direct genetic associations:

    • NPAS1 and NPAS3 mutations have been linked to psychiatric disorders in human genetic studies

    • NPAS1/NPAS3-deficient mice exhibit behaviors relevant to psychiatric symptomatology, including impaired prepulse inhibition and social recognition deficits

  • Excitatory/inhibitory balance mechanisms:

    • NPAS1 regulates cortical interneuron development, particularly SST+ and VIP+ populations

    • NPAS1-/- mice show increased inhibitory tone in superficial cortical layers

    • Disrupted E/I balance is implicated in multiple neuropsychiatric conditions including schizophrenia, autism spectrum disorders, and epilepsy

  • Molecular pathway connections:

    • NPAS1/NPAS3-deficient mice show reduced reelin expression, a protein consistently found to be attenuated in postmortem brain tissue of schizophrenia patients

    • NPAS1 regulates Arx expression, mutations in which cause intellectual disability and epilepsy

  • Circuit-specific implications:

    • Ventral pallidum NPAS1+ neurons mediate stress responses, relevant to mood disorders

    • Basal forebrain NPAS1+ neurons regulate sleep-wake behavior and cortical oscillations

    • Activation of BF NPAS1+ neurons reduces NREM slow-wave and sigma power and disrupts sleep spindles, "reminiscent of findings in several neuropsychiatric disorders"

  • Translational research approaches:

    • NPAS1 serves as a developmental marker that allows cross-species identification of conserved neuronal populations

    • This facilitates translational studies of these neurons and their role in "insomnia, dementia, neurodevelopmental disorders, and other conditions involving BF"

By linking molecular mechanisms to circuit function and behavior, NPAS1 research provides a framework for understanding how developmental transcription factors influence adult brain function and potentially contribute to neuropsychiatric disorder pathophysiology.

What emerging technologies could advance NPAS1 research?

Several emerging technologies hold promise for advancing NPAS1 research:

  • Single-cell transcriptomics and multi-omics approaches:

    • Single-cell RNA sequencing to reveal heterogeneity within NPAS1+ populations across brain regions

    • Integration with spatial transcriptomics to maintain anatomical context

    • Multi-omic profiling (transcriptome, epigenome, proteome) of NPAS1+ neurons to understand molecular signatures

  • Advanced genetic manipulation techniques:

    • CRISPR-Cas9 for precise modification of NPAS1 gene or its regulatory elements

    • Split-Cre or intersectional genetic approaches to target specific subsets of NPAS1+ neurons

    • Temporal control systems to distinguish developmental from acute functions

  • Advanced circuit mapping and functional analysis:

    • Whole-brain imaging of NPAS1+ neuron projections using tissue clearing methods (CLARITY, iDISCO)

    • Fiber photometry or miniscope calcium imaging to record NPAS1+ neuron activity during behavior

    • Expansion microscopy for super-resolution imaging of synaptic connections

  • Translational approaches:

    • Development of human stem cell-derived brain organoid models to study NPAS1 function in human neurons

    • Cross-species comparative studies examining evolutionary conservation of NPAS1+ neuron populations

    • Integration with human neuroimaging and genetic findings from psychiatric disorders

These technologies would address current limitations in understanding NPAS1+ neuron heterogeneity, developmental trajectories, and precise contributions to circuit function and behavior.

What are the most significant unanswered questions regarding NPAS1 function?

Despite significant progress, several critical questions about NPAS1 function remain unanswered:

  • Molecular mechanisms:

    • What is the complete set of genes directly regulated by NPAS1 in different cell types?

    • How does NPAS1 interact with other transcription factors in regulatory networks?

    • What upstream signaling pathways regulate NPAS1 expression and activity in different contexts?

  • Developmental biology:

    • What determines the regional specificity of NPAS1's effects on interneuron development?

    • Why does NPAS1 deficiency affect SST+ and VIP+ interneurons but not PV+ interneurons?

    • What is the evolutionary significance of NPAS1's role in forebrain development?

  • Adult function:

    • Does NPAS1 continue to function as a transcription factor in adult neurons, or primarily serve as a lineage marker?

    • What role does NPAS1 play in adult neuronal plasticity and maintenance?

    • How do NPAS1+ neurons respond to various environmental challenges and experiences?

  • Disease relevance:

    • What specific human genetic variants in NPAS1 contribute to neuropsychiatric risk?

    • Do medications used to treat psychiatric disorders affect NPAS1 expression or function?

    • Could NPAS1+ neurons serve as therapeutic targets for specific conditions?

  • Circuit-specific questions:

    • How do NPAS1+ neurons in different brain regions (basal forebrain, ventral pallidum, globus pallidus) communicate?

    • What are the precise mechanisms by which NPAS1+ neurons regulate critical behaviors like sleep-wake transitions and stress responses?

Addressing these questions will require integrated approaches spanning molecular, cellular, circuit, and behavioral levels of analysis.

How might NPAS1 research contribute to potential therapeutic interventions?

NPAS1 research opens several avenues for therapeutic development:

  • Novel drug targets:

    • Molecular pathways regulated by NPAS1 could provide new targets for psychiatric disorder treatment

    • Given its role in interneuron development, NPAS1-regulated pathways might inform interventions for neurodevelopmental disorders

    • The selective effects on specific interneuron populations suggest potential for targeted modulation of E/I balance

  • Circuit-based interventions:

    • BF NPAS1+ neurons influence sleep and cortical oscillations, suggesting potential targets for sleep disorders

    • VP NPAS1+ neurons mediate stress susceptibility, indicating possible targets for stress-related disorders

    • Neural circuit therapies (deep brain stimulation, transcranial magnetic stimulation) could potentially target circuits involving NPAS1+ neurons

  • Biomarker development:

    • NPAS1 expression patterns or downstream targets could serve as biomarkers for specific neuropsychiatric conditions

    • Changes in NPAS1+ neuron activity patterns might provide electrophysiological signatures relevant to diagnosis or treatment monitoring

  • Developmental interventions:

    • Understanding NPAS1's role in interneuron development could inform early interventions for neurodevelopmental disorders

    • Potentially guide timing of therapeutic interventions to critical developmental windows

  • Precision medicine approaches:

    • NPAS1 genetic variants might help stratify patients into subgroups likely to respond to specific treatments

    • Understanding the downstream consequences of NPAS1 dysfunction could inform personalized treatment approaches

While direct targeting of NPAS1 itself may be challenging given its role as a transcription factor, the neural circuits and molecular pathways it regulates offer promising therapeutic avenues for conditions involving disrupted E/I balance, stress responses, or sleep-wake regulation.

What criteria should guide the selection of NPAS1 antibodies for specific applications?

When selecting NPAS1 antibodies for research, consider these critical criteria:

  • Application-specific considerations:

    • Western blot: Select antibodies validated for denatured protein detection, with demonstrated specificity at the expected molecular weight (62.7 kDa)

    • Immunohistochemistry: Choose antibodies optimized for tissue fixation methods, with demonstrated nuclear localization

    • Immunofluorescence: Consider signal strength and compatibility with other antibodies for co-labeling experiments

  • Validation evidence:

    • Testing in knockout tissue: Gold-standard validation showing absence of signal in NPAS1-/- tissue

    • Cross-reactivity testing: Particularly important for multi-species studies (human, mouse, rat)

    • Citation record: Preference for antibodies with published validation in peer-reviewed studies

  • Technical specifications:

    • Epitope location: Consider whether the antibody targets N-terminal, middle region, or C-terminal epitopes

    • Clonality: Monoclonal antibodies may offer higher specificity, while polyclonals may provide stronger signals

    • Host species: Important for avoiding cross-reactivity in multi-labeling experiments

  • Species reactivity:

    • Match to experimental model: Ensure reactivity with species under study (human, mouse, rat)

    • Cross-species conservation: For comparative studies, consider antibodies targeting conserved epitopes

The commercial landscape includes over 154 NPAS1 antibodies from 18 suppliers with varying applications, reactivity profiles, and validation levels . Select those with rigorous validation data specifically for your application and experimental system.

How can researchers optimize immunohistochemical protocols for NPAS1 detection?

Optimizing immunohistochemical protocols for NPAS1 detection requires attention to several key factors:

  • Tissue preparation considerations:

    • Fixation: Optimize paraformaldehyde concentration (typically 4%) and duration (4-24 hours) to preserve nuclear antigens

    • Sectioning: 30-40μm sections balance structural integrity with antibody penetration

    • Antigen retrieval: Include heat-mediated antigen retrieval steps (e.g., sodium citrate buffer, pH 6.0) to expose nuclear epitopes

  • Staining optimization:

    • Blocking: Use comprehensive blocking (normal serum + BSA + mild detergent) to reduce background

    • Antibody concentration: Titrate primary antibody concentrations (typically 1:500-1:2000 dilutions)

    • Incubation conditions: Consider extended incubation times (24-72 hours at 4°C) for optimal penetration

    • Permeabilization: Include adequate Triton X-100 (0.1-0.3%) to facilitate nuclear penetration

  • Signal detection considerations:

    • For fluorescence: Consider signal amplification methods (tyramide signal amplification, ABC amplification)

    • For chromogenic detection: DAB optimization with nickel enhancement can improve sensitivity

    • For fiber-dense regions: Extended antibody incubation times and increased detergent concentration may improve penetration

  • Controls and validation:

    • Include positive control regions with known NPAS1 expression (dentate gyrus, specific cortical layers)

    • Negative controls should include primary antibody omission and ideally NPAS1 knockout tissue

  • Co-labeling optimization:

    • Sequential staining protocols for challenging combinations

    • Careful selection of fluorophores to avoid bleed-through

    • Nuclear counterstains (DAPI, Hoechst) to confirm nuclear localization of NPAS1 signal

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 2025 TheBiotek. All Rights Reserved.