STMN4 Antibody

Shipped with Ice Packs
In Stock

Product Specs

Buffer
Phosphate Buffered Saline (PBS) containing 0.1% Sodium Azide, 50% Glycerol, adjusted to pH 7.3. Store at -20°C. Avoid repeated freeze-thaw cycles.
Lead Time
We typically dispatch orders within 1-3 business days of receipt. Delivery times may vary depending on the purchasing method and location. Please consult your local distributor for specific delivery timelines.
Synonyms
STMN4 antibody; Stathmin-4 antibody; Stathmin-like protein B3 antibody; RB3 antibody
Target Names
STMN4
Uniprot No.

Target Background

Function
This antibody exhibits microtubule-destabilizing activity.
Database Links

HGNC: 16078

KEGG: hsa:81551

UniGene: Hs.201058

Protein Families
Stathmin family
Subcellular Location
Golgi apparatus. Cell projection, growth cone. Cell projection, axon.

Q&A

Advanced Research Questions

  • How can I differentiate between phosphorylated and non-phosphorylated forms of STMN4 in immunofluorescence experiments?

    Distinguishing phosphorylated from non-phosphorylated STMN4 requires:

    Antibody selection:

    • Use phospho-specific antibodies targeting known STMN4 phosphorylation sites (serine residues S59, S71, and S81 in the SLD domain)

    • Pair with total STMN4 antibodies in dual labeling experiments

    • Validate phospho-antibodies using lambda phosphatase-treated controls

    Sample preparation:

    • Rapid fixation to preserve phosphorylation status

    • Use phosphatase inhibitors throughout sample preparation

    • Consider phospho-protein enrichment for low abundance phospho-forms

    Controls and validation:

    • Include samples treated with phosphatase inhibitors and activators

    • Use tissues/cells with known phosphorylation states (e.g., mitotic vs. G1 phase cells)

    • Perform parallel Western blot analysis with phospho-specific antibodies

    Imaging and quantification:

    • Use spectral unmixing if emission spectra overlap

    • Quantify co-localization between phospho-specific and total STMN4 signals

    • Employ ratio imaging to analyze the proportion of phosphorylated to total STMN4

    This methodology is particularly relevant for studying STMN4's dynamic regulation during neural development, as phosphorylation modulates its microtubule-destabilizing activity .

  • What experimental approaches can resolve contradictory findings about STMN4's role in neuronal differentiation between morpholino knockdown and CRISPR/Cas9 mutant models?

    The discrepancy between morpholino knockdown and CRISPR/Cas9 knockout findings requires a systematic resolution strategy:

    1. Genetic compensation assessment:

      • Perform RNA-seq on both morphants and mutants to identify differentially expressed genes

      • Quantify expression of all stathmin family members (source notes increased stmn1b in stmn4 mutants)

      • Assess transcriptional adaptation mechanisms

    2. Temporal analysis of gene function:

      • Use inducible knockdown/knockout systems to manipulate STMN4 at specific timepoints

      • The shRNA approach with heat shock promoter described in provides a validated methodology

    3. Dosage-dependent effects evaluation:

      • Generate hypomorphic alleles that reduce rather than eliminate STMN4 function

      • Analyze heterozygous mutants alongside homozygotes

      • Compare different morpholino concentrations to identify threshold effects

    4. Epistasis experiments:

      • Manipulate potential compensatory genes (e.g., stmn1b) in stmn4 mutants

      • Test whether double knockouts recapitulate morphant phenotypes

      • Conduct rescue experiments with various stathmin family members

    5. Domain-specific function analysis:

      • Create targeted mutations affecting specific functional domains

      • The serine mutations (S59A, S71A, S81A) described in provide a starting point

  • What methodological considerations are important when studying STMN4's interaction with microtubule dynamics in real-time imaging experiments?

    Real-time imaging of STMN4-microtubule interactions requires:

    Experimental system selection:

    • Primary neural progenitors or neuroblastoma cells with endogenous STMN4 expression

    • Systems with fluorescently-tagged tubulin (e.g., EB3-GFP for plus-end tracking)

    • Microfluidic chambers for controlled manipulation of cellular environment

    Probe design:

    • Fluorescently-tagged STMN4 constructs (ensuring tags don't interfere with microtubule binding)

    • Validation through rescue experiments in STMN4-deficient cells

    • Consider photoactivatable fluorescent proteins for pulse-chase experiments

    Imaging parameters:

    • High numerical aperture objectives (1.3-1.4) for optimal resolution

    • Fast acquisition rates (1-2 seconds/frame) to capture dynamic instability events

    • Extended imaging periods (30-60 minutes) to capture complete catastrophe/rescue cycles

    Analysis methods:

    • Automated tracking of microtubule plus-end movements

    • Quantification of growth rate, shrinkage rate, catastrophe frequency, rescue frequency

    • Time spent in growth/shrinkage/pause phases

    Control experiments:

    • Parallel experiments with other stathmin family members

    • Dose-response studies with varying STMN4 expression levels

    • Comparison of wild-type versus phosphorylation site mutants (S59A, S71A, S81A)

    • Cell cycle synchronization to control for variation across cell cycle phases

  • How can I design experiments to investigate the relationship between STMN4 expression and cell cycle progression in neural progenitors?

    Experimental design strategy for investigating STMN4's role in cell cycle progression:

    1. Cell cycle synchronization and analysis:

      • Synchronize neural progenitors using established methods

      • Analyze STMN4 protein levels and phosphorylation status by Western blot

      • Correlate with cell cycle markers (Cyclin B1, CDK1, phospho-Histone H3)

      • Flow cytometry to quantify cell cycle distribution

    2. Live cell cycle reporters:

      • Use FUCCI (Fluorescent Ubiquitination-based Cell Cycle Indicator) system

      • Co-express fluorescently-tagged STMN4 to correlate with cell cycle phases

      • Time-lapse imaging to track individual cells through complete cycles

      • Quantify STMN4 levels/localization changes relative to cell cycle progression

    3. STMN4 manipulation with cell cycle analysis:

      • Implement inducible STMN4 knockdown/overexpression systems

      • Trigger manipulation at specific cell cycle phases

      • Measure effects on G2/M transition markers (phospho-Cdc25, Cyclin B1)

      • As observed in , measure G2 phase duration using time-lapse microscopy

    4. Rescue experiments:

      • Test whether Cdc25a overexpression rescues cell cycle defects in STMN4-deficient cells

      • Examine other components (CDK1/Cyclin B1 complex, Wnt signaling)

      • Create phosphomimetic STMN4 mutants to determine how phosphorylation affects regulation

    5. In vivo neural progenitor analysis:

      • Use zebrafish or mouse embryonic brain tissue

      • Employ EdU pulse-chase experiments to measure cell cycle parameters

      • Combine with phospho-Histone H3 staining to identify M-phase cells

  • What controls should be implemented when investigating STMN4's role in apoptotic pathways in retinal cells?

    Based on findings that stmn4 deficiency leads to retinal cell apoptosis , implement these controls:

    1. Genetic model validation:

      • Confirm STMN4 knockout/knockdown efficiency using multiple methods

      • Use multiple targeting strategies (morpholinos, CRISPR/Cas9, shRNA)

      • Include scrambled/non-targeting controls for all knockdown approaches

      • Generate rescue lines expressing wild-type STMN4

    2. Apoptosis detection controls:

      • Use multiple independent detection methods:

        • TUNEL assay

        • Cleaved Caspase-3 immunostaining

        • Annexin V/PI flow cytometry

        • Mitochondrial membrane potential assays

      • Include positive controls for each assay

    3. Pathway specificity controls:

      • p53 knockdown experiments (as described in )

      • Bcl-2 family modulation (overexpression of anti-apoptotic members)

      • Caspase inhibitor treatments

      • Examine both intrinsic and extrinsic apoptotic pathways

    4. Temporal controls:

      • Time-course experiments to determine sequence of events

      • Determine whether apoptosis follows cell cycle arrest or occurs independently

      • Use inducible systems to manipulate STMN4 at defined developmental stages

    5. Cell-type specificity controls:

      • Cell sorting of specific retinal populations before analysis

      • Co-labeling with cell-type specific markers

      • Similar to approaches in , which showed overlapping Sox2+ and TUNEL+ signals

  • How can dual immunolabeling be optimized to study the relationship between STMN4 expression and progenitor cell markers?

    Optimizing dual immunolabeling requires:

    Antibody selection and validation:

    • Choose antibodies raised in different host species (e.g., rabbit anti-STMN4 and mouse anti-Sox2)

    • Validate each antibody individually before attempting co-labeling

    • Test for cross-reactivity with controls using each primary antibody alone

    • Consider monoclonal antibodies for greater specificity

    Tissue/cell preparation:

    • Test multiple fixation protocols (PFA concentrations 2-4%, methanol, or combination)

    • Optimize antigen retrieval (both TE buffer pH 9.0 and citrate buffer pH 6.0 can be effective)

    • Test different permeabilization conditions (Triton X-100 concentrations 0.1-0.3%)

    Sequential immunostaining protocol:

    1. First primary antibody incubation (typically the less abundant marker)

    2. First secondary antibody incubation

    3. Optional mild fixation step (0.5% PFA for 10 minutes)

    4. Second primary antibody incubation

    5. Second secondary antibody incubation

    Signal optimization:

    • Tyramide signal amplification for low abundance markers

    • Use Fab fragments to prevent steric hindrance

    • Prolonged incubation times (36-48 hours at 4°C) for thick tissue sections

    Imaging considerations:

    • Acquire fluorescence in separate channels sequentially

    • Include single-labeled controls for spillover compensation

    • Use deconvolution to enhance signal-to-noise ratio

  • What methodological approaches can distinguish between direct and indirect effects of STMN4 on microtubule stability?

    To distinguish direct from indirect effects of STMN4 on microtubules:

    In vitro reconstitution systems:

    • Purified component assays using recombinant STMN4 and tubulin

    • Measure tubulin polymerization kinetics with varying STMN4 concentrations

    • Analyze STMN4-tubulin complex formation using analytical ultracentrifugation

    • Compare wild-type STMN4 versus mutants (particularly SLD domain mutations)

    Structure-function analysis:

    • Generate domain-specific STMN4 mutants:

      • SLD domain mutants (S59A, S71A, S81A)

      • N-terminal membrane binding domain mutants (C20, 22A)

    • Express these in STMN4-deficient cells

    • Quantify microtubule dynamics parameters for each mutant

    Proximity-based interaction studies:

    • BioID or APEX2 proximity labeling with STMN4 as bait

    • FRET or BRET assays between STMN4 and tubulin to detect direct interactions

    • Cross-linking mass spectrometry to map interaction interfaces

    • Immunoprecipitation coupled with Western blot analysis

    Acute manipulation approaches:

    • Optogenetic control of STMN4 activity (e.g., light-inducible degradation)

    • Chemical-genetic approaches for specific inhibition

    • Rapid protein knockdown systems (e.g., dTAG or Trim-Away)

    • Measure immediate changes in microtubule dynamics following manipulation

  • What considerations are important when designing experiments to investigate the Wnt-STMN4 regulatory axis in neural development?

    Source indicates that "inhibition of Wnt could reduce stmn4 transcripts," suggesting a regulatory relationship requiring:

    1. Wnt pathway manipulation:

      • Use multiple methods to modulate Wnt signaling:

        • Small molecules (CHIR99021 for activation, IWR-1 for inhibition)

        • Genetic approaches (β-catenin knockout/knockdown)

        • Ligand manipulation (recombinant Wnt proteins, neutralizing antibodies)

      • Include dose-response and time-course analyses

      • Validate pathway activation/inhibition using established readouts

    2. Developmental timing considerations:

      • Implement stage-specific manipulations using:

        • Heat-shock inducible systems (as described in )

        • Timed drug treatments with Wnt modulators

        • Temporal knockout systems (CreER/loxP with tamoxifen induction)

      • Focus on critical developmental windows based on STMN4 expression patterns

    3. Transcriptional regulation analysis:

      • Perform chromatin immunoprecipitation for β-catenin and TCF/LEF factors on the STMN4 promoter

      • Create STMN4 promoter reporter constructs with wild-type and mutated Wnt responsive elements

      • Employ ATAC-seq to assess chromatin accessibility at the STMN4 locus

      • Implement genome editing of putative Wnt responsive elements

    4. Functional readouts:

      • Cell cycle analysis (particularly G2/M phase transitions)

      • Neuronal differentiation markers (similar to elavl3 expression analysis in )

      • Microtubule dynamics parameters

      • In vivo developmental phenotypes in model organisms

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.