HOMER1 Antibody

Homer Homolog-1, Mouse Anti Human
Shipped with Ice Packs
In Stock

Description

Table 1: Core Properties of HOMER1 Antibodies

SupplierClonalityHost SpeciesReactivityApplications (Dilution)
VWR PolyclonalRabbitHuman, Mouse, Rat, DogWB (N/A), ICC (N/A)
Bio-Techne MonoclonalMouseHuman, Mouse, RatWB (1:1,000), ICC (1:100)
Proteintech MonoclonalMouseHuman, Mouse, Rat, PigWB (1:5,000–50,000), ICC (1:400–1,600)
R&D Systems MonoclonalMouseHuman, Mouse, RatWB (2 µg/mL), Simple Western (20 µg/mL)
Synaptic Systems MonoclonalRabbitHuman, Mouse, RatWB, ICC, ELISA

Synaptic Plasticity and AMPAR Regulation

  • HOMER1 knockout (KO) studies show disrupted AMPAR subunit GluA2 distribution, reducing synaptic polyamine sensitivity and long-term potentiation (LTP) maintenance .

  • Overexpression of the short isoform Homer1a decreases AMPA/NMDA current ratios and destabilizes LTP, highlighting its role in synaptic AMPAR dynamics .

Protein Interaction Networks

  • HOMER1 anchors mGluR5 and Shank3 at synapses. Depolarization dissociates these complexes, altering cortical synaptic proteomes .

  • In Homer1 KO mice, ankyrin-G levels increase while Shank3 decreases, suggesting compensatory mechanisms in spine stability .

Disease Implications

  • Dysregulated Homer1 interactions are linked to ASD, schizophrenia, and bipolar disorder due to disrupted synaptic protein networks .

Table 2: Validation Data Across Platforms

ApplicationObserved Molecular WeightKey FindingsSource
Western Blot40–52 kDaDetected in brain lysates (human, mouse, rat) .
ImmunofluorescenceN/ALabels excitatory synapses colocalized with GluR2 .
ELISAN/AUsed for quantitative analysis of synaptic extracts .

Critical Notes:

  • Batch Variability: Anti-HOMER1 antibodies show species-specific reactivity; cross-validate using KO controls .

  • Storage: Lyophilized antibodies require reconstitution in PBS with sucrose and storage at -20°C .

Limitations and Future Directions

  • Cross-Reactivity: Some antibodies detect all Homer isoforms due to conserved N-terminal domains .

  • Research Gaps: Functional studies on non-neuronal roles of HOMER1 (e.g., striated muscle) remain underexplored .

Product Specs

Introduction
HOMER1, a member of the Homer family of proteins, plays a crucial role in regulating signal transduction, trafficking of glutamate receptors, and maintaining extracellular glutamate levels in corticolimbic brain regions. It is primarily located at excitatory synapses and exhibits binding affinity for group 1 metabotropic glutamate receptors (mGluRs), influencing the susceptibility to TRAIL-mediated apoptosis.
Formulation
The antibody is supplied in a solution containing 1mg/ml in PBS buffer at pH 7.4 with 0.1% sodium azide.
Storage Procedures
For short-term storage (up to 1 month), maintain the antibody at 4°C. For extended periods, store at -20°C. Avoid repeated freeze-thaw cycles to ensure stability.
Stability / Shelf Life
The antibody remains stable for 12 months when stored at -20°C and for 1 month at 4°C.
Applications
This HOMER1 antibody has undergone rigorous testing using ELISA and Western blot analysis, confirming its specificity and reactivity. While the recommended dilution range for Western blot analysis is 1:1000 to 1:2000, with a starting dilution of 1:1000, it is advised to optimize the dilution for each specific application to achieve optimal results.
Synonyms
HOMER, SYN47, Ves-1, HOMER1A, HOMER1B, HOMER1C, HOMER1, Homer protein homolog 1.
Purification Method
HOMER1 antibody was purified from mouse ascitic fluids by protein-G affinity chromatography.
Type
Mouse Anti Human Monoclonal.
Clone
PAT1F3AT.
Immunogen
Anti-human HOMER1 mAb is derived from hybridization of mouse F0 myeloma cells with spleen cells from BALB/c mice immunized with recombinant human HOMER1 amino acids 1-354 purified from E. coli.
Ig Subclass
Mouse IgG2b heavy chain and κ light chain.

Q&A

What is HOMER1 and why is it important in neuroscience research?

HOMER1 is a postsynaptic density (PSD) scaffold protein that plays critical roles in synaptic plasticity, calcium signaling, and is implicated in several neurological disorders. Its importance stems from its function as a molecular scaffold that binds and cross-links cytoplasmic regions of various proteins including metabotropic glutamate receptors (GRM1, GRM5), inositol trisphosphate receptors (ITPR1), dynamin (DNM3), ryanodine receptors (RYR1, RYR2), and SHANK proteins (SHANK1, SHANK3). By physically linking surface receptors like GRM1 and GRM5 with ER-associated ITPR1 receptors, HOMER1 facilitates the coupling of surface receptor activation to intracellular calcium release, a crucial mechanism in neuronal signaling .

What are the key differences between HOMER1 isoforms that researchers should consider when selecting antibodies?

When selecting HOMER1 antibodies, researchers must consider the specific isoform they want to target. HOMER1 exists in both long forms (Homer 1b/c) and short forms (Homer 1a), which have distinct cellular distributions and functions. Immunohistochemistry and biochemical studies show that the long forms are enriched at the PSD, while Homer 1a is dispersed throughout neuronal somal and dendritic cytoplasm with little affiliation with the PSD . These isoforms have different roles - the long forms are constitutively expressed and function in maintaining synaptic structure, while Homer 1a acts as a natural dominant negative that dynamically competes with the long forms to regulate synaptic metabotropic glutamate function and may be involved in structural changes during neuronal plasticity and development . Antibodies raised against specific regions may recognize different isoforms, so researchers should verify the epitope recognition and validate antibody specificity for their target isoform .

What is the expected molecular weight of HOMER1 in Western blot applications?

When performing Western blot analysis, researchers should expect to detect HOMER1 at approximately 48-52 kDa, depending on the specific isoform and post-translational modifications. In published experimental data, HOMER1 has been detected at approximately 52 kDa in human brain (cerebellum) tissue and IMR-32 human neuroblastoma cell line using Western blot analysis . Using Simple Western analysis (an automated Western blot alternative), HOMER1 was detected at approximately 51 kDa in human brain tissue . Some resources report the protein to be approximately 48 kDa in cytoplasmic preparations, while others note it as 40.3 kilodaltons in theoretical mass . These slight variations in molecular weight may be attributed to differences in experimental conditions, post-translational modifications, or the specific isoform being detected .

How should researchers optimize antibody dilutions for different applications involving HOMER1?

Optimization of HOMER1 antibody dilutions should follow a systematic approach, starting with the manufacturer's recommended dilution range as a baseline. For Western blot applications, begin with a concentration around 2 μg/mL as demonstrated in successful experiments using Mouse Anti-HOMER1 Monoclonal Antibody followed by HRP-conjugated secondary antibody . For immunocytochemistry and immunohistochemistry applications, initial dilutions may need to be higher, but should be determined empirically for each tissue type. When optimizing:

  • Perform a dilution series (e.g., 1:100, 1:200, 1:500, 1:1000)

  • Include appropriate positive controls (e.g., brain tissue known to express HOMER1)

  • Include negative controls (tissues or cells without HOMER1 expression)

  • Evaluate signal-to-noise ratio at each dilution

  • Consider fixation method, antigen retrieval protocols, and blocking agents which may affect epitope accessibility

Remember that optimal dilutions can vary significantly between applications (WB, IHC, ICC, IF) and even between different tissue preparations within the same application . Document all optimization parameters systematically to ensure reproducibility.

What are the most appropriate positive and negative controls for validating HOMER1 antibody specificity?

Proper validation of HOMER1 antibody specificity requires carefully selected positive and negative controls:

Positive controls:

  • Human, mouse, or rat brain tissue (particularly cerebellum) where HOMER1 is highly expressed

  • IMR-32 human neuroblastoma cell line, which has been validated to express HOMER1

  • Skeletal muscle and heart tissue, which also express HOMER1

  • Recombinant HOMER1 protein (especially when working with a new antibody)

Negative controls:

  • Tissues or cell lines with HOMER1 knocked down via siRNA or CRISPR-Cas9

  • Pre-absorption of the antibody with the immunizing peptide before application

  • Isotype control antibodies (same isotype, irrelevant specificity)

  • Secondary antibody-only controls to check for non-specific binding

  • For HOMER1 isoform-specific antibodies, tissues from corresponding knockout models

When validating specificity, researchers should observe the expected molecular weight band (approximately 48-52 kDa) in positive controls and absence of this band in negative controls. Cross-reactivity with other Homer family members (Homer2, Homer3) should be assessed, especially when using antibodies targeting conserved regions .

How can researchers effectively distinguish between HOMER1 isoforms in their experiments?

Distinguishing between HOMER1 isoforms requires strategic experimental approaches due to their structural similarities:

  • Antibody selection: Use isoform-specific antibodies that target unique regions. For example, antibodies raised against the C-terminal region will detect long forms (Homer 1b/c) but not the short form (Homer 1a) which lacks this region .

  • Western blot analysis: Long and short HOMER1 isoforms can be distinguished by their molecular weights. Homer 1a (~45 kDa) migrates slightly faster than Homer 1b/c (~47-52 kDa) .

  • Subcellular fractionation: Biochemical fractionation coupled with Western blotting can help distinguish isoforms based on their characteristic localization patterns. Long forms are enriched in PSD fractions, while Homer 1a is predominantly found in cytosolic fractions .

  • Immunostaining patterns: Immunohistochemistry or immunofluorescence with pan-HOMER1 antibodies followed by careful analysis of localization patterns can help distinguish isoforms. Homer 1b/c shows punctate staining at synapses, while Homer 1a exhibits diffuse cytoplasmic staining .

  • Temporal expression analysis: In certain experimental paradigms, Homer 1a is induced as an immediate early gene following neuronal activation, while long forms maintain relatively stable expression. Time-course experiments following stimulation can help distinguish these patterns .

  • RT-PCR or qPCR: Design primers that specifically amplify different isoforms based on their unique exon usage to quantify transcript levels of specific isoforms.

Documentation of isoform-specific molecular weights, subcellular distribution patterns, and validation using knockout controls is essential for reliable isoform discrimination .

How should researchers interpret unexpected molecular weight bands when using HOMER1 antibodies?

When encountering unexpected molecular weight bands in HOMER1 antibody applications, systematic interpretation and troubleshooting are necessary:

  • Bands at ~30-40 kDa: May represent degradation products of HOMER1, especially if samples were not properly handled or protease inhibitors were omitted. Alternative explanation could be cross-reactivity with other Homer family members or detection of splice variants.

  • Bands at ~60-70 kDa: Could indicate post-translational modifications like phosphorylation, ubiquitination, or SUMOylation of HOMER1. Alternatively, might represent incomplete denaturation leading to dimerization, as Homer proteins can form multimers through their coiled-coil domains.

  • Multiple bands between 45-55 kDa: Likely represent different HOMER1 isoforms (Homer 1a, 1b, 1c) or post-translationally modified forms. Western blots have detected HOMER1 at approximately 51-52 kDa in brain tissue samples .

  • High molecular weight bands (>100 kDa): May indicate protein aggregates, particularly if sample preparation involved insufficient reducing conditions or boiling time.

To distinguish between these possibilities:

  • Compare with positive control tissues known to express HOMER1

  • Pre-absorb antibody with immunizing peptide to identify specific vs. non-specific bands

  • Use phosphatase treatment to identify phosphorylated forms

  • Perform subcellular fractionation to correlate band patterns with expected localization

  • Consider cross-reactivity with other proteins containing similar epitopes, particularly other Homer family members

Document all experimental conditions systematically, as HOMER1 detection can be affected by sample preparation methods, reducing agents, gel percentage, and transfer conditions .

What are common sources of variability in HOMER1 antibody experiments and how can they be controlled?

Researchers working with HOMER1 antibodies should be aware of several sources of variability and implement appropriate controls:

  • Tissue/sample preparation variability:

    • Use consistent protocols for tissue extraction and processing

    • Standardize post-mortem intervals for animal tissue collection

    • Include protease and phosphatase inhibitors in all buffers

    • Maintain consistent fixation times for immunohistochemistry samples

  • Expression level differences:

    • HOMER1 expression varies across brain regions and is highly enriched in the postsynaptic density

    • Expression of different isoforms can change dynamically in response to neuronal activity (especially Homer 1a)

    • Standardize loading controls and quantification methods

    • Consider developmental stage and circadian timing in experimental design

  • Antibody variability:

    • Use antibodies from consistent lots when possible

    • Include standard positive controls with every experiment

    • Validate each new antibody lot against previous lots

    • Document epitope information and cross-reactivity profiles

  • Technical variability:

    • Standardize gel percentage, transfer conditions, and blocking reagents

    • Maintain consistent antibody incubation times and temperatures

    • Implement automated systems where possible to reduce handling variation

    • Document all protocol deviations

  • Biological variability:

    • Increase biological replicates to account for individual variation

    • Control for sex, age, and strain differences in animal models

    • Consider circadian effects on HOMER1 expression

    • Document health status and treatment history of sample sources

Implementation of a detailed laboratory protocol with standardized positive controls (such as human brain cerebellum tissue) and quality control checkpoints can substantially reduce variability across experiments .

How can researchers reconcile contradictory findings when using different HOMER1 antibodies?

When faced with contradictory results from different HOMER1 antibodies, researchers should implement a systematic approach to identify the source of discrepancies:

  • Compare antibody characteristics:

    • Epitope mapping: Different antibodies may target distinct domains of HOMER1, affecting detection of specific isoforms or binding partners

    • Clonality: Monoclonal antibodies may provide higher specificity but potentially lower sensitivity compared to polyclonal antibodies

    • Host species: Different host species can affect background in certain applications

  • Validate antibodies using orthogonal methods:

    • Confirm specificity using HOMER1 knockout/knockdown systems

    • Perform epitope competition assays with immunizing peptides

    • Use multiple antibodies targeting different HOMER1 epitopes

    • Complement antibody-based detection with mRNA analysis

  • Contextualize with experimental conditions:

    • Certain fixation methods may mask specific epitopes in immunohistochemistry

    • Reducing conditions in Western blot may affect epitope accessibility

    • Sample preparation methods may cause differential extraction of HOMER1 isoforms

    • Cross-linking in chromatin immunoprecipitation may affect epitope availability

  • Systematic comparison experiment:

    • Design an experiment using identical samples processed in parallel

    • Apply multiple antibodies side-by-side under identical conditions

    • Document all variables including lot numbers, dilutions, and incubation conditions

    • Include appropriate positive controls (e.g., brain tissue) and negative controls

  • Collaboration and external validation:

    • Compare results with published literature

    • Consider sending samples to reference laboratories

    • Contact antibody manufacturers with documented discrepancies

Remember that different HOMER1 antibodies have demonstrated variability in detecting specific isoforms. For instance, antibodies against long forms (Homer 1b/c) show enrichment at PSDs, while antibodies against Homer 1a show dispersed cytoplasmic labeling , which could appear contradictory if not properly interpreted.

How can researchers effectively use HOMER1 antibodies in studies of synaptic plasticity and neuronal signaling?

Researchers studying synaptic plasticity and neuronal signaling can leverage HOMER1 antibodies in several sophisticated experimental paradigms:

  • High-resolution imaging of synaptic architecture:

    • Use HOMER1 antibodies (particularly against long forms) as postsynaptic markers in super-resolution microscopy (STORM, PALM) to visualize PSD organization

    • Combine with presynaptic markers to assess synaptic morphology changes during plasticity

    • Measure HOMER1 cluster size and intensity as indicators of synaptic strength

    • Implement live-cell imaging using tagged antibody fragments to track dynamic changes

  • Activity-dependent HOMER1 expression:

    • Design time-course experiments to track Homer 1a induction following neuronal stimulation

    • Compare HOMER1 isoform ratios before and after long-term potentiation (LTP) or depression (LTD)

    • Correlate HOMER1 expression changes with electrophysiological measurements

    • Use specific antibodies to distinguish activity-induced Homer 1a from constitutive Homer 1b/c

  • Protein interaction studies:

    • Employ HOMER1 antibodies in co-immunoprecipitation experiments to identify binding partners

    • Use proximity ligation assays to visualize HOMER1 interactions with metabotropic glutamate receptors, IP3 receptors, or SHANK proteins in situ

    • Implement FRET-based approaches to study dynamic interactions

    • Consider that HOMER1 binds to cytoplasmic regions of GRM1, GRM5, ITPR1, DNM3, RYR1, RYR2, SHANK1, and SHANK3

  • Calcium signaling dynamics:

    • Combine HOMER1 immunostaining with calcium imaging to correlate scaffold organization with signaling outputs

    • Assess the role of HOMER1 in coupling surface receptors to intracellular calcium release

    • Investigate HOMER1's role in regulating ER-plasma membrane contact sites

    • Examine how different HOMER1 isoforms affect calcium oscillation patterns

  • Disease model applications:

    • Investigate HOMER1 expression and localization in models of neurological disorders associated with HOMER1 dysfunction (autism, bipolar disorder, Phelan-McDermid Syndrome)

    • Assess therapeutic interventions targeting HOMER1 scaffolding function

    • Correlate HOMER1 complex formation with behavioral phenotypes

When designing these experiments, consider that the long form (Homer 1b/c) is enriched at PSDs while the short form (Homer 1a) shows diffuse cytoplasmic distribution, allowing for distinct experimental readouts depending on the target isoform .

What are the most effective strategies for studying HOMER1 binding partners using immunoprecipitation with HOMER1 antibodies?

Studying HOMER1 binding partners through immunoprecipitation requires careful experimental design:

  • Antibody selection and validation:

    • Choose antibodies that recognize epitopes outside known protein interaction domains (EVH1 domain, coiled-coil region) to avoid interference with binding partner interactions

    • Verify that the antibody effectively immunoprecipitates HOMER1 in pilot studies

    • Consider using multiple antibodies targeting different HOMER1 regions to compare binding partner profiles

    • Epitope tags may be useful for clean immunoprecipitation but could disrupt certain interactions

  • Optimized tissue/cell preparation:

    • Use gentle lysis conditions to preserve protein-protein interactions

    • Consider crosslinking approaches for capturing transient interactions

    • Include phosphatase inhibitors to maintain phosphorylation-dependent interactions

    • Optimize salt concentration to balance specificity with yield

    • Use appropriate detergents (mild non-ionic) to maintain membrane protein interactions

  • Control experiments:

    • Include immunoprecipitation with isotype control antibodies

    • Pre-clear lysates to reduce non-specific binding

    • Consider performing reciprocal immunoprecipitations with antibodies against known binding partners

    • Include negative controls from tissues lacking HOMER1 expression

  • Advanced analytical approaches:

    • Couple immunoprecipitation with mass spectrometry for unbiased identification of binding partners

    • Consider SILAC or TMT labeling for quantitative comparison across conditions

    • Implement BioID or proximity labeling approaches as complementary methods

    • Use sequential immunoprecipitation to identify higher-order complexes

  • Context-specific considerations:

    • Different HOMER1 isoforms have distinct binding partner profiles; Homer 1a lacks the coiled-coil domain necessary for multimerization

    • HOMER1 binding partners may vary by subcellular compartment; consider fractionation before immunoprecipitation

    • Activity-dependent interactions may require stimulation protocols before lysis

    • The binding landscape of HOMER1 is complex and involves motifs beyond the canonical PPxxF sequence

Researchers should note that HOMER1 binding partners include GRM1, GRM5, ITPR1, DNM3, RYR1, RYR2, SHANK1, and SHANK3 , and the interaction with these partners occurs through specific domains that should be considered when designing immunoprecipitation experiments.

How can researchers integrate HOMER1 antibody-based approaches with advanced imaging techniques to study neuronal microdomains?

Integrating HOMER1 antibody detection with advanced imaging techniques creates powerful approaches for investigating neuronal microdomains:

  • Super-resolution microscopy applications:

    • Implement STORM or PALM imaging with HOMER1 antibodies to visualize nanoscale organization of postsynaptic densities

    • Use multi-color super-resolution to map HOMER1 relative to binding partners within the PSD

    • Quantify HOMER1 cluster properties (size, density, shape) as readouts of synaptic organization

    • Apply correlative light and electron microscopy (CLEM) to integrate molecular specificity with ultrastructural context

  • Live-cell imaging strategies:

    • Utilize HOMER1 antibody fragments (Fab, nanobodies) conjugated to cell-permeable peptides for live imaging

    • Combine with optogenetic tools to simultaneously manipulate and visualize HOMER1-containing structures

    • Implement fluorescence recovery after photobleaching (FRAP) to study HOMER1 dynamics at synapses

    • Use single-particle tracking to follow HOMER1 movement between synaptic and extrasynaptic regions

  • Spatially-resolved protein interaction analysis:

    • Apply proximity ligation assays (PLA) to visualize HOMER1 interactions with binding partners in situ

    • Implement FRET/FLIM approaches to study protein-protein interactions in living neurons

    • Use split-fluorescent protein complementation to visualize specific HOMER1 complexes

    • Combine with optogenetic tools to manipulate interactions while imaging

  • Volumetric and whole-brain approaches:

    • Apply tissue clearing methods (CLARITY, iDISCO) combined with HOMER1 immunolabeling for whole-brain analysis

    • Implement array tomography for high-resolution 3D reconstruction of HOMER1 distribution

    • Use light-sheet microscopy for rapid volumetric imaging of HOMER1 across brain regions

    • Develop computational approaches to quantify HOMER1-positive synapses throughout neural circuits

  • Integrative multi-modal imaging:

    • Combine HOMER1 immunodetection with functional calcium imaging to correlate structure with activity

    • Implement correlative electrophysiology and HOMER1 imaging to link function with molecular organization

    • Use micro-scaled devices to manipulate local environments while imaging HOMER1 organization

    • Apply machine learning approaches to identify patterns in HOMER1 distribution across neuronal populations

When designing these experiments, consider the distinct localization patterns of different HOMER1 isoforms: Homer 1b/c enriched at the PSD versus the cytoplasmic distribution of Homer 1a . This differential localization can be leveraged as an internal control when validating new imaging approaches.

How should researchers design experiments to study HOMER1 dysfunction in neurological and psychiatric disorders?

Designing experiments to investigate HOMER1 dysfunction in neurological and psychiatric disorders requires careful consideration of multiple factors:

  • Disease model selection:

    • Choose models relevant to HOMER1-associated conditions (Phelan-McDermid Syndrome, bipolar disorder, autism, fragile X syndrome)

    • Consider both genetic models (e.g., HOMER1 knockouts/knockins) and environmental/pharmacological models

    • Validate model relevance by confirming HOMER1 expression/function changes

    • Compare findings across multiple model systems for robustness

  • Molecular analysis approaches:

    • Quantify total HOMER1 protein levels and isoform ratios (Homer 1a vs. Homer 1b/c) in disease-relevant brain regions

    • Assess post-translational modifications that may be altered in disease states

    • Examine HOMER1 subcellular localization using fractionation and immunohistochemistry

    • Analyze HOMER1 binding partner interactions and complex formation in disease contexts

  • Structural and functional readouts:

    • Correlate HOMER1 alterations with synaptic structural changes (spine density, morphology)

    • Implement electrophysiological recordings to assess synaptic function

    • Analyze calcium signaling dynamics, particularly in contexts where HOMER1 couples surface receptors to intracellular calcium release

    • Assess circuit-level alterations using in vivo recording or imaging techniques

  • Translational considerations:

    • Include analyses of human postmortem tissue when available

    • Design interventions targeting HOMER1 function (peptide inhibitors, viral overexpression)

    • Develop biomarkers based on HOMER1 complexes or signaling pathways

    • Consider therapeutic approaches that normalize HOMER1 function or expression

  • Technical approaches:

    • Use multiple validated HOMER1 antibodies targeting different epitopes

    • Implement quantitative approaches (automated image analysis, ELISA)

    • Include appropriate age-matched and sex-balanced controls

    • Consider developmental trajectories and age-dependent effects

Remember that HOMER1 dysfunction may manifest differently across disorders. For example, in autism spectrum disorders, alterations in the balance between long and short HOMER1 isoforms may impact synaptic plasticity and metabotropic glutamate receptor signaling, while in Phelan-McDermid Syndrome, interactions with SHANK3 may be particularly relevant .

What are the key methodological challenges in comparing HOMER1 expression patterns between normal and pathological tissues?

Comparing HOMER1 expression patterns between normal and pathological tissues presents several methodological challenges that researchers must address:

  • Tissue quality and preservation challenges:

    • Postmortem interval effects can significantly impact protein integrity and epitope availability

    • Different fixation protocols between clinical and research samples may affect antibody binding

    • Storage conditions and freezing methods can introduce variability

    • Solution: Implement strict quality control metrics for all samples and document preservation parameters

  • Cell-type specific expression considerations:

    • HOMER1 expression varies across neuronal subtypes and non-neuronal cells

    • Disease states may alter cellular composition of regions being analyzed

    • Solution: Combine HOMER1 immunolabeling with cell-type specific markers; consider single-cell approaches

  • Isoform-specific detection challenges:

    • Disease states may alter the ratio of different HOMER1 isoforms rather than total levels

    • Antibodies may recognize multiple isoforms with different affinities

    • Solution: Use isoform-specific antibodies; complement with mRNA analysis of splice variants

  • Quantification standardization:

    • Variability in immunostaining intensity between batches complicates cross-sample comparison

    • Regional heterogeneity requires systematic sampling approaches

    • Solution: Include standard reference samples in each batch; implement rigorous randomization and blinding

  • Distinguishing primary from secondary changes:

    • HOMER1 alterations may be compensatory rather than causative in disease states

    • Medication effects in clinical samples may confound results

    • Solution: Correlate findings with disease progression metrics; compare medicated vs. unmedicated samples when possible

  • Technical variables in protein extraction:

    • Different extraction protocols may preferentially isolate certain HOMER1 pools

    • Synaptic HOMER1 may be difficult to extract completely using standard protocols

    • Solution: Compare multiple extraction methods; consider subcellular fractionation protocols optimized for synaptic proteins

  • Control selection challenges:

    • Age, sex, and comorbidity matching between pathological and control samples

    • Regional and demographic variability in baseline HOMER1 expression

    • Solution: Implement careful matching protocols; increase control sample numbers to account for variability

These challenges highlight the importance of implementing rigorous experimental design with appropriate controls, standardized protocols, and multiple complementary methodological approaches when comparing HOMER1 expression between normal and pathological states.

How might advances in antibody engineering and development enhance HOMER1 research?

Advances in antibody engineering offer significant potential to enhance HOMER1 research in several dimensions:

  • Isoform-specific antibody development:

    • Creation of highly specific antibodies that can distinguish between closely related HOMER1 isoforms (Homer 1a, 1b, 1c)

    • Development of conformation-specific antibodies that recognize particular structural states of HOMER1

    • Engineering antibodies that selectively recognize post-translationally modified forms of HOMER1

    • Potential for creating antibodies that detect specific HOMER1 protein complexes

  • Intrabody and live-cell applications:

    • Development of cell-permeable single-domain antibodies (nanobodies) against HOMER1 for live-cell imaging

    • Creation of genetically encoded intrabodies that can be expressed within neurons to track HOMER1 dynamics

    • Engineering split-antibody complementation systems to visualize HOMER1 interactions in living cells

    • Development of HOMER1-specific FASTags or other protein-tagging antibody fragments

  • Functional antibody applications:

    • Design of function-blocking antibodies that can interfere with specific HOMER1 interactions

    • Creation of antibodies that stabilize or promote particular HOMER1 complexes

    • Development of antibody-based tools to manipulate HOMER1 localization or function

    • Engineering bispecific antibodies that can bring HOMER1 into proximity with specific binding partners

  • Advanced detection strategies:

    • Integration of HOMER1 antibodies with click chemistry for multiplexed detection

    • Development of proximity-sensing antibody pairs for detecting HOMER1 complexes

    • Creation of antibody-based sensors that respond to changes in HOMER1 conformation or interaction state

    • Engineering of renewable synthetic HOMER1 antibodies with precisely controlled binding properties

  • Therapeutic and diagnostic applications:

    • Development of antibody-based imaging agents for visualization of HOMER1 in neurological disorders

    • Creation of antibody-drug conjugates to target therapies to HOMER1-enriched synapses

    • Engineering of blood-brain barrier-penetrant antibodies for in vivo HOMER1 targeting

    • Development of antibody-based assays for HOMER1 complexes as potential biomarkers

These advances would build upon current understanding of HOMER1 binding properties, particularly its EVH1 domain's interaction with the PPxxF motif and the more complex binding landscape that has been recently elaborated . The creation of more specific tools would help resolve current challenges in distinguishing between HOMER1 isoforms and understanding their dynamic roles in neuronal function.

What novel methodological approaches might help uncover previously unidentified HOMER1 functions or interactions?

Innovative methodological approaches could reveal new dimensions of HOMER1 biology beyond its known functions:

  • Proximity-based labeling technologies:

    • Implement BioID, TurboID, or APEX2 fusions with HOMER1 to identify proximal proteins in living cells

    • Apply spatially-restricted enzymatic tagging to map the HOMER1 interactome in specific subcellular compartments

    • Use split-BioID systems to capture proteins that interact with specific HOMER1 complexes

    • These approaches may identify transient or context-specific interactions missed in traditional co-immunoprecipitation studies

  • High-throughput screening approaches:

    • Apply CRISPR screening to identify genes that modify HOMER1 function or localization

    • Implement drug screens to identify compounds that modulate HOMER1 interactions

    • Use synthetic peptide libraries to identify novel binding motifs beyond the classical PPxxF sequence

    • Apply functional genomics to systematically map HOMER1-dependent cellular processes

  • Single-molecule techniques:

    • Implement single-molecule tracking to study HOMER1 dynamics at synapses

    • Apply optical tweezers or AFM to measure binding forces between HOMER1 and partners

    • Use single-molecule pull-down assays to study composition of individual HOMER1 complexes

    • Implement single-molecule FRET to study conformational changes in HOMER1 during binding events

  • Advanced imaging modalities:

    • Apply expansion microscopy to visualize HOMER1 nanoscale organization

    • Use lattice light-sheet microscopy for long-term imaging of HOMER1 dynamics

    • Implement cryo-electron tomography to visualize HOMER1 complexes in their native context

    • Apply correlative light and electron microscopy to link HOMER1 molecular patterns with ultrastructure

  • Systems biology approaches:

    • Develop computational models of HOMER1 scaffolding dynamics

    • Apply network analysis to position HOMER1 within broader signaling networks

    • Implement multi-omics approaches to correlate HOMER1 function with transcriptome, proteome, and metabolome changes

    • Use machine learning to identify patterns in HOMER1 distribution or function across conditions

  • In vivo molecular tools:

    • Apply genetically encoded sensors that report on HOMER1 binding events

    • Implement optogenetic approaches to spatiotemporally control HOMER1 interactions

    • Use chemogenetic tools to selectively disrupt specific HOMER1 functions

    • Apply in vivo genome editing to study HOMER1 variants in intact circuits

Recent research has revealed that HOMER1 binding preferences are more complex than previously thought, with an N-terminally overlapping motif that is also bound by Ena/VASP proteins . These novel methodological approaches could further elucidate the evolutionary relationships between these paralogous domains and identify additional binding partners and functions.

How might researchers better integrate HOMER1 antibody-based research with other emerging neuroscience technologies?

Integration of HOMER1 antibody-based research with emerging neuroscience technologies can create powerful synergistic approaches:

  • Integration with spatial transcriptomics:

    • Combine HOMER1 immunolabeling with in situ sequencing to correlate protein localization with local transcriptome

    • Apply Slide-seq or Visium spatial genomics with HOMER1 antibody staining on adjacent sections

    • Implement MERFISH with HOMER1 immunolabeling to correlate mRNA expression of binding partners with protein localization

    • These approaches could reveal spatial regulation of HOMER1 isoform expression and correlation with binding partner availability

  • Combination with connectomics:

    • Apply array tomography with HOMER1 immunogold labeling for volume electron microscopy

    • Correlate HOMER1 expression patterns with circuit connectivity maps

    • Implement expansion microscopy with HOMER1 antibodies followed by volume imaging

    • Use HOMER1 as a marker to identify specific synapse types within connectomic datasets

  • Integration with functional recording technologies:

    • Combine HOMER1 antibody labeling with voltage imaging to correlate structure with function

    • Apply post-hoc HOMER1 immunostaining after calcium imaging experiments

    • Correlate electrophysiological recordings with HOMER1 distribution in the same neurons

    • Implement activity-dependent labeling techniques with HOMER1 immunostaining

  • Combination with optogenetic and chemogenetic approaches:

    • Use HOMER1 antibodies to assess structural changes following optogenetic stimulation

    • Apply HOMER1 immunolabeling after chemogenetic manipulation of specific neuronal populations

    • Design activity-dependent HOMER1 reporters that can be monitored during optogenetic stimulation

    • Implement multiplexed imaging of HOMER1 with optogenetic actuators

  • Integration with computational approaches:

    • Apply machine learning to identify patterns in HOMER1 distribution across brain regions

    • Develop computational models that integrate HOMER1 scaffolding properties with circuit function

    • Use deep learning for automated analysis of HOMER1 immunolabeling in large datasets

    • Implement systems biology approaches to position HOMER1 within broader neuronal signaling networks

  • Combination with protein engineering approaches:

    • Use HOMER1 antibodies to validate genetically encoded HOMER1 sensors

    • Apply split-protein complementation with HOMER1 segments and validate with antibodies

    • Implement CRISPR-based tagging of endogenous HOMER1 and confirm with antibody validation

    • Design protein interaction perturbation tools and assess effects using HOMER1 antibodies

These integrative approaches would build upon current understanding of HOMER1's role in postsynaptic organization and its complex binding landscape , providing multi-dimensional insights into how this scaffold protein contributes to neuronal function and dysfunction in health and disease.

What are the most effective computational approaches for analyzing HOMER1 distribution and colocalization in complex microscopy datasets?

Analyzing HOMER1 distribution and colocalization in complex microscopy datasets requires sophisticated computational approaches:

  • Advanced segmentation strategies:

    • Implement deep learning-based segmentation (U-Net, Mask R-CNN) to identify HOMER1-positive puncta

    • Apply instance segmentation to distinguish individual synapses in densely labeled regions

    • Use adaptive thresholding approaches that account for regional intensity variations

    • Implement multi-channel segmentation that leverages correlations between HOMER1 and other synaptic markers

    • These approaches are particularly valuable given HOMER1's punctate distribution at postsynaptic densities

  • Colocalization analysis methods:

    • Move beyond simple Pearson's correlation to more sophisticated measures like Manders' coefficients or object-based colocalization

    • Implement point pattern analysis to assess spatial relationships between HOMER1 and binding partners

    • Apply coordinate-based colocalization for super-resolution datasets

    • Use distance-based analysis to quantify proximity between HOMER1 and other proteins

    • Consider that different HOMER1 isoforms show distinct localization patterns that require appropriate analysis methods

  • 3D and 4D analysis approaches:

    • Implement 3D object-based analysis for volumetric imaging of HOMER1 distribution

    • Apply temporal tracking algorithms for time-lapse imaging of HOMER1 dynamics

    • Use registration algorithms to align HOMER1 imaging with other modalities

    • Implement trajectory analysis for single-particle tracking of HOMER1 molecules

  • Machine learning integration:

    • Train classifiers to identify specific patterns of HOMER1 distribution associated with different physiological states

    • Apply dimensionality reduction techniques to identify major modes of variation in HOMER1 organization

    • Implement anomaly detection algorithms to identify abnormal HOMER1 distribution patterns

    • Use generative models to predict HOMER1 organization under different conditions

  • Quantitative feature extraction:

    • Develop morphometric analysis pipelines for HOMER1-positive structures

    • Apply intensity distribution analysis to quantify HOMER1 concentration gradients

    • Implement graph-based approaches to analyze HOMER1 network topology

    • Use spatial statistics to characterize HOMER1 clustering properties

  • Validation and quality control:

    • Implement automated quality control metrics to identify imaging artifacts

    • Apply bootstrapping or cross-validation to assess reliability of measurements

    • Use synthetic data generation to benchmark analysis algorithms

    • Implement ground truth comparison where possible (e.g., correlative electron microscopy)

Product Science Overview

Introduction

Homer protein homolog 1 (HOMER1) is a neuronal protein encoded by the HOMER1 gene in humans. It is also known by other names such as Vesl and PSD-Zip45 . HOMER1 plays a crucial role in the central nervous system, particularly in the regulation of synaptic transmission and plasticity.

Structure and Function

HOMER1 is an approximately 48 kDa cytoplasmic protein that regulates the cell surface localization of neuronal metabotropic glutamate receptors (mGluR1 and mGluR5) . These receptors are involved in various neural processes, including learning, memory, and synaptic plasticity. HOMER1 interacts with these receptors to modulate their signaling pathways and ensure proper synaptic function.

Antibody Specificity

The mouse anti-human HOMER1 antibody is a monoclonal antibody that specifically detects human HOMER1 in various applications, including Western blot and ELISA . The antibody is produced by immunizing mice with a synthetic peptide corresponding to a specific region of the human HOMER1 protein . This ensures high specificity and affinity for the target protein.

Applications

The mouse anti-human HOMER1 antibody is widely used in research to study the expression and function of HOMER1 in different tissues and cell types. It is particularly useful in neuroscience research to investigate the role of HOMER1 in synaptic transmission and plasticity. The antibody can be used in various experimental techniques, including:

  • Western Blotting: To detect HOMER1 protein levels in tissue and cell lysates.
  • Immunohistochemistry: To visualize the localization of HOMER1 in tissue sections.
  • ELISA: To quantify HOMER1 protein levels in biological samples.

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.