HTATIP2 Antibody

HIV-1 Tat Interactive Protein 2, Mouse Anti Human
Shipped with Ice Packs
In Stock

Description

Introduction to HTATIP2 Antibody

HTATIP2 antibodies are specialized immunoglobulins designed to detect the HIV-1 Tat Interactive Protein 2 (HTATIP2), a tumor suppressor protein implicated in DNA repair, angiogenesis, and cellular apoptosis. These antibodies enable researchers to study HTATIP2's subcellular localization, expression levels, and functional interactions in diseases such as cancer, autoimmune disorders, and vascular diseases.

Applications in Research

HTATIP2 antibodies are critical tools for studying HTATIP2's role in:

  • Cancer Biology: Detecting HTATIP2 expression in glioblastoma, lung adenocarcinoma, and gastric cancer to assess prognosis or therapeutic responses .

  • Angiogenesis and Arteriogenesis: Analyzing HTATIP2's regulation of monocyte/macrophage (Mo/MΦ) function in vascular remodeling .

  • DNA Repair Mechanisms: Tracking HTATIP2-mediated cytoplasmic retention of MPG (N-methylpurine DNA glycosylase) in alkylating agent resistance .

  • T Cell Regulation: Investigating HTATIP2's impact on T cell activation, proliferation, and apoptosis in autoimmune diseases like type 1 diabetes .

Table 1: Comparative Analysis of HTATIP2 Antibodies

SourceTypeApplicationsReactivityMW (kDa)Immunogen
SAB Biotech Rabbit PolyclonalWBHuman27C-terminal fusion protein
Antibodies Online Rabbit MonoclonalWB, ELISA, IHC, IF, IPHuman, Mouse27–30Full-length recombinant protein
Cell Signaling Rabbit PolyclonalWBHuman28, 32Not specified
Sigma-Aldrich Rabbit PolyclonalIB, IF, IHCHuman27AEGFVRVDRDYVLKSAELAKAGGCKHFNLLSSKGADKSSNFLYLQVKGEVEAKVEELKFDRYSVFRPGVLLCDRQESRPGEWLVRKFFGSLPDSWARGHSVPVVTVVRAMLNNVVRPRDKQMELLENKAIHDLGK
Sigma-Aldrich Rabbit PolyclonalIB, IF, IHCHuman27ALSKLREDFRMQNKSVFILGASGETGRVLLKEILEQGLFSKVTLIGRRKLTFDEEAYKNVNQEVVDFEKLDDYASAFQGHDVGFC

Functional Insights from HTATIP2 Antibody Studies

  • Epigenetic Silencing in Glioblastoma: HTATIP2 antibodies revealed promoter methylation-driven downregulation, enhancing nuclear localization of MPG and treatment resistance .

  • Arteriogenic Monocyte Regulation: Silencing HTATIP2 via siRNA increased angiogenic/arteriogenic capacity in monocytes, validated by antibody-based quantification of protein expression .

  • Tumor Hypoxia Adaptation: Absent HTATIP2 expression in lung adenocarcinoma correlated with metabolic plasticity and HIF2α-driven β-catenin signaling, detected via immunoblotting .

Product Specs

Introduction
HTATIP2, a member of the short-chain dehydrogenases/reductases (SDR) family, functions as a tumor suppressor. It achieves this by suppressing metabolism, inhibiting angiogenesis, and inducing the expression of apoptosis-related genes Bad and Siva. HTATIP2 interacts with the activation domain of HIV-1 TAT, enhancing its transcription through phosphorylation of RNA polymerase II (Pol II). Defects in HTATIP2 are linked to hepatocellular carcinomas and apoptotic-resistant tumor cells, suggesting its potential in antitumor therapy.
Physical Appearance
A clear, colorless solution that has been sterilized by filtration.
Formulation
The solution contains 1mg/ml of HTATIP2 antibody in a buffer consisting of PBS at pH 7.4, 10% glycerol, and 0.02% sodium azide.
Storage Procedures
For short-term storage (up to 1 month), keep at 4°C. For longer storage periods, store at -20°C. Avoid repeated freeze-thaw cycles.
Stability / Shelf Life
The product is stable for 12 months when stored at -20°C and for 1 month at 4°C.
Applications
This HTATIP2 antibody has been validated for use in ELISA, Western blot analysis, Flow cytometry, and ICC/IF to ensure its specificity and reactivity. However, optimal working dilutions should be determined empirically for each application.
Synonyms
TIP30, CC3, SDR44U1, HTATIP2, EC=1.1.1.-, HIV-1 TAT-interactive protein 2, FLJ26963.
Purification Method
HTATIP2 antibody was purified from mouse ascitic fluids by protein-A affinity chromatography.
Type
Mouse Anti Human Monoclonal.
Clone
PAT5D6AT.
Immunogen
Anti-human HTATIP2 mAb, is derived from hybridization of mouse F0 myeloma cells with spleen cells from BALB/c mice immunized with recombinant human HTATIP2 amino acids 1-242 purified from E. coli.
Ig Subclass
Mouse IgG2a heavy chain and ? light chain.

Q&A

What is HTATIP2 and why is it significant in research?

HTATIP2, also known as CC3, SDR44U1, or TIP30, is a protein that functions as an oxidoreductase and plays important roles in various cellular processes including apoptosis, cell migration, and angiogenesis. Research significance stems from its involvement in multiple diseases, particularly cancer and vascular disorders. In gastric cancer, HTATIP2 expression inversely correlates with tumor progression and metastasis, suggesting its role as a tumor suppressor gene . Recent evidence also links HTATIP2 to T-cell regulation in type 1 diabetes, indicating its broader involvement in immune regulation and autoimmune conditions . The study of HTATIP2 is therefore crucial for understanding disease mechanisms and identifying potential therapeutic targets across multiple pathological conditions.

What are the primary applications of HTATIP2 antibodies in research?

HTATIP2 antibodies are utilized across various research applications with immunohistochemistry and Western blotting being the most common. In immunohistochemistry, these antibodies enable the visualization of HTATIP2 expression patterns in tissue microarrays, helping researchers categorize samples based on expression levels (weak, moderate, or strong staining) . For Western blotting, HTATIP2 antibodies allow protein quantification to correlate expression levels with disease phenotypes . Additionally, these antibodies support functional studies investigating HTATIP2's role in cellular processes like cell migration, invasion, and epithelial-mesenchymal transition . They are also valuable in validating genetic studies where HTATIP2 expression is manipulated through overexpression or knockdown experiments to understand its mechanistic roles in disease pathways .

How do researchers distinguish between different epitopes of HTATIP2 for antibody selection?

Researchers select HTATIP2 antibodies based on the specific epitopes they target, which is critical for experimental success and reproducibility. When investigating full-length HTATIP2 (Q9BUP3, 242 amino acids), antibodies targeting conserved regions are preferred for cross-species applications. For studies focused on specific HTATIP2 domains, researchers must select antibodies that recognize the relevant functional regions, such as the NAD(P)-binding Rossmann-fold domain or protein interaction interfaces. Antibody validation should include Western blotting with positive controls (tissues/cells known to express HTATIP2) and negative controls (HTATIP2-knockout samples). Additionally, researchers should verify antibody specificity through immunoprecipitation followed by mass spectrometry or through parallel detection with multiple antibodies targeting different HTATIP2 epitopes. This comprehensive validation ensures that experimental results accurately reflect HTATIP2 biology rather than non-specific antibody interactions.

What are the known isoforms of HTATIP2 and how do antibodies differentiate between them?

HTATIP2 exists in multiple isoforms resulting from alternative splicing, with the canonical form (Q9BUP3) comprising 242 amino acids. When selecting antibodies for specific isoform detection, researchers must carefully evaluate the epitope specificity. Polyclonal antibodies typically recognize multiple epitopes and may detect several isoforms, while monoclonal antibodies target specific epitopes that might be isoform-specific. For definitive isoform differentiation, researchers should use antibodies raised against unique peptide sequences present only in specific isoforms. Validation of isoform specificity requires Western blotting with recombinant proteins representing each isoform or tissues with known differential isoform expression patterns. Additionally, parallel RNA analysis using isoform-specific primers helps confirm antibody selectivity. This careful approach is essential when studying isoform-specific functions of HTATIP2 in different tissues or disease states.

What is the optimal protocol for HTATIP2 immunohistochemistry in tissue samples?

The optimal protocol for HTATIP2 immunohistochemistry requires careful attention to fixation, antigen retrieval, and antibody incubation conditions. Based on validated protocols, researchers should follow these methodological steps:

  • Tissue fixation: Fix specimens with 4% paraformaldehyde to preserve tissue morphology while maintaining HTATIP2 antigenicity .

  • Tissue processing: Embed fixed tissues in paraffin and section to 5 μm thickness for optimal antibody penetration.

  • Deparaffinization and rehydration: Use standard xylene and graded alcohol series.

  • Antigen retrieval: Perform heat-induced epitope retrieval using citrate buffer (pH 6.0) at 95-98°C for 20 minutes.

  • Endogenous peroxidase blocking: Incubate sections with 3% hydrogen peroxide for 10 minutes.

  • Primary antibody incubation: Apply HTATIP2 polyclonal antibody (such as PA5-82247) at 1:100-1:200 dilution and incubate overnight at 4°C .

  • Secondary antibody incubation: Use HRP-conjugated secondary antibody (like Dako Real Envision/HRP, K5007) for 15 minutes at room temperature .

  • Visualization: Develop with DAB chromogen and counterstain with hematoxylin.

  • Scoring system: Classify staining intensity into weak, moderate, and strong categories, with HTATIP2 overexpression defined as ≥50% moderate-to-strong staining or ≥25% strong staining .

This protocol allows for consistent evaluation of HTATIP2 expression across different tissue samples, facilitating reliable comparison between experimental groups.

What are the critical controls necessary when using HTATIP2 antibodies in Western blotting?

When performing Western blotting with HTATIP2 antibodies, implementing proper controls is crucial for result validity and reproducibility. The following controls are essential:

  • Positive tissue control: Include lysates from tissues or cell lines with confirmed high HTATIP2 expression (e.g., liver tissue or specific cancer cell lines like MKN74 for gastric cancer studies) .

  • Negative tissue control: Use tissues or cell lines with minimal HTATIP2 expression, or ideally HTATIP2-knockout samples.

  • Loading control: Probe for housekeeping proteins (β-actin, GAPDH, or α-tubulin) to normalize for protein loading variations.

  • HTATIP2 overexpression control: Include lysates from cells transfected with HTATIP2 expression vectors (e.g., pCMV6-HTATIP2-FLAG-DDK) to confirm antibody specificity .

  • HTATIP2 knockdown control: Include lysates from cells treated with HTATIP2-specific siRNA to demonstrate specificity.

  • Molecular weight verification: Confirm that the detected band corresponds to the expected molecular weight of HTATIP2 (approximately 27 kDa).

  • Peptide competition: Pre-incubate the antibody with the immunizing peptide to demonstrate specific binding.

  • Secondary antibody-only control: Omit primary antibody to identify non-specific binding of the secondary antibody.

These controls collectively ensure the specificity, sensitivity, and reliability of HTATIP2 detection in Western blotting experiments.

How should researchers design siRNA experiments to study HTATIP2 function?

Designing effective siRNA experiments to study HTATIP2 function requires careful consideration of multiple factors to ensure specific knockdown and meaningful functional analysis:

  • siRNA design: Target conserved regions of HTATIP2 mRNA sequence with minimal homology to other genes. Design 3-4 different siRNA sequences to identify the most effective option. Maintain GC content between 30-60% and avoid sequences with internal repeats or palindromes.

  • Control selection: Include a non-targeting siRNA control with similar chemical modifications as the experimental siRNA. For studies requiring high specificity, include a rescue control with HTATIP2 cDNA containing silent mutations in the siRNA target sequence.

  • Transfection optimization: Determine optimal transfection conditions (reagent concentration, cell density, incubation time) for each cell type using a fluorescently labeled control siRNA. For immune cells like monocytes, which may be difficult to transfect, electroporation might yield better results .

  • Knockdown verification: Confirm HTATIP2 knockdown at both mRNA level (qRT-PCR, with 70-90% reduction considered successful) and protein level (Western blotting) . Verify knockdown duration, which typically lasts 3-5 days.

  • Functional assays: Based on research from HTATIP2 knockdown studies, appropriate functional assays include:

    • Cell migration and invasion assays using Transwell chambers

    • Angiogenesis assays using HUVEC/Matrigel tubule formation

    • EMT marker analysis (e.g., Snail, Slug expression)

    • Cell viability assessments using assays like CellTiter-Glo

  • Off-target effect monitoring: Perform microarray or RNA-seq analysis to identify potential off-target effects, especially on genes involved in pathways of interest.

This comprehensive approach ensures that observed phenotypes can be confidently attributed to HTATIP2 knockdown rather than experimental artifacts.

What are the best practices for generating and validating HTATIP2 overexpression models?

Creating reliable HTATIP2 overexpression models requires systematic planning and rigorous validation. Based on published methodologies, researchers should follow these best practices:

  • Expression vector selection: Choose appropriate vectors based on experimental needs:

    • For transient expression: pCMV6-based vectors (e.g., pCMV6-HTATIP2-FLAG-DDK)

    • For stable expression: lentiviral or retroviral vectors with selection markers

    • For inducible expression: Tet-On/Off systems when temporal control is needed

  • Tagging considerations: C-terminal tags (FLAG, DDK) are preferable as N-terminal tags may interfere with HTATIP2's functional domains . Verify that the tag doesn't alter protein localization or function through comparative functional assays.

  • Transfection/transduction optimization: Determine optimal conditions for each cell line. For difficult-to-transfect cells like KATO III gastric cancer cells, Lipofectamine 2000 at 2 μg plasmid DNA per standard well has proven effective .

  • Expression validation: Confirm overexpression using:

    • Western blotting (expect 1.5-10 fold increase over endogenous levels)

    • qRT-PCR for mRNA quantification

    • Immunofluorescence to verify subcellular localization

    • Flow cytometry for population-level expression analysis

  • Functional validation: Assess the biological impact through:

    • Cell migration and invasion assays using Transwell chambers with or without Matrigel

    • EMT marker expression (Snail, Slug)

    • Cell proliferation assays

    • Cell-type specific functional assays (e.g., angiogenic activity for vascular studies)

  • Controls: Always include:

    • Empty vector control (e.g., pCMV6)

    • Wild-type cells without transfection

    • If possible, rescue experiments in HTATIP2-knockdown cells

Following these practices ensures the development of reliable overexpression models that accurately reflect HTATIP2's biological functions and potential therapeutic applications.

How does HTATIP2 expression correlate with gastric cancer prognosis and staging?

HTATIP2 expression demonstrates significant correlation with gastric cancer (GC) progression and patient outcomes. According to immunohistochemical analysis of gastric cancer tissues, HTATIP2 overexpression (OE) is associated with several favorable clinicopathological features:

Clinical ParameterHTATIP2 OE GroupHTATIP2 UE GroupP-value
Early GC (T1 stage)Higher percentageLower percentage0.024
Lymph node metastasisLess frequentMore frequent0.008
TNM stageLower stageHigher stage0.017

These clinical correlations are supported by mechanistic studies showing that HTATIP2 overexpression in gastric cancer cell lines (particularly KATO III) significantly inhibits cell migration and invasion capacities . At the molecular level, HTATIP2 overexpression leads to decreased expression of epithelial-mesenchymal transition (EMT) markers Snail and Slug, suggesting that HTATIP2 may suppress metastasis by inhibiting the EMT process .

What role does HTATIP2 play in vascular remodeling and angiogenesis?

HTATIP2 serves as a critical regulator of vascular remodeling and angiogenesis, particularly in the context of ischemic conditions. Research has revealed that HTATIP2 expression is abnormally elevated in monocytes/macrophages (Mo/MΦs) isolated from patients with chronic limb threatening ischemia (CLTI) . This elevated expression appears to significantly impair the pro-arteriogenic capacity of these cells, as demonstrated by both in vitro and in vivo neovascularization assays.

The functional significance of HTATIP2 in angiogenesis was established through knockdown experiments. When HTATIP2 expression was reduced using siRNA technology (achieving approximately 77% reduction in gene expression), there was a marked enhancement of angiogenic activity as measured in HUVEC/Matrigel tubule formation assays . This effect was consistent and maintained for at least 5 days in culture without affecting cell viability, as confirmed by 7-AAD staining and flow cytometric analysis.

Mechanistically, HTATIP2 appears to modulate cellular pathways involved in vascular remodeling, potentially through:

  • Regulation of pro-angiogenic gene expression

  • Modulation of immune cell phenotypes important for arteriogenesis

  • Influence on endothelial cell interaction and activation

These findings suggest that targeting HTATIP2 could represent a novel therapeutic approach for enhancing vascular remodeling in patients with ischemic vascular diseases. Particularly, the ability to manipulate HTATIP2 expression in autologous cells before therapeutic administration could potentially improve the currently modest efficacy of cell therapy in patients with CLTI.

How is HTATIP2 implicated in autoimmune diseases such as type 1 diabetes?

Recent genome-wide association studies have implicated HTATIP2 as a significant gene in type 1 diabetes (T1D) pathogenesis, specifically through its role in T cell regulation . This represents an emerging area of research that links HTATIP2 to autoimmune mechanisms beyond its previously established roles in cancer and vascular biology.

In T1D research, experimental approaches have involved PCR amplification of the HTATIP2 gene from Mammalian Gene Collection cDNA clones, followed by ligation into expression vectors containing T7 RNA polymerase promoters for in vitro transcription/translation . These constructs enable the generation of capped-and-tailed mRNA using specialized kits like the HiScribe T7 ARCA mRNA Kit with tailing. The quality of these transcripts is verified through spectrophotometry and electrophoresis on denaturing formaldehyde/MOPS agarose gels before functional studies.

Functional investigations have employed primary human CD4+ T cells, which were expanded using anti-CD3/CD28 beads and then transfected with HTATIP2 mRNA through electroporation techniques . The impact of HTATIP2 expression on T cell function is then assessed through various assays, including cell viability measurements using CellTiter-Glo 2.0 assay.

While the complete mechanisms remain under investigation, current evidence suggests that HTATIP2 may influence T cell activation, proliferation, or regulatory functions, potentially affecting the autoimmune processes that lead to pancreatic β-cell destruction in T1D. This emerging research direction could offer new insights into autoimmune disease pathways and potentially identify novel therapeutic targets for T1D and other autoimmune conditions.

How do structural changes in antibodies impact HTATIP2 binding and downstream signaling?

The binding of antibodies to HTATIP2 involves complex structural dynamics that can significantly influence downstream signaling pathways. According to structural analysis studies of antibody-antigen interactions, the binding interface between antibodies and their targets can undergo conformational changes that propagate throughout the antibody structure . For anti-HTATIP2 antibodies, these structural changes may be particularly relevant due to the functional domains present in HTATIP2.

When antibodies bind to HTATIP2, conformational changes can occur in both the variable and constant domains. Research indicates that the constant domains (CH1 and CL) often demonstrate more movement than the variable domains (VH and VL) . These movements can affect the relative orientation of VH-VL domains and potentially influence signal propagation from variable to constant domains. The RMSD (Root Mean Square Deviation) analysis of antibody structures before and after antigen binding provides quantitative measures of these structural changes:

These structural movements have functional implications, as they may affect complement activation and immune response triggering. Previous research has demonstrated that antibodies with similar variable domains but different constant domains can bind their targets with significantly different affinities . This suggests that the selection of appropriate antibody formats (full IgG, Fab, scFv) for HTATIP2 detection or targeting should consider these potential structural dynamics and their impact on binding affinity and downstream signaling.

What are the cutting-edge approaches for studying HTATIP2 protein interactions using antibody-based techniques?

Cutting-edge antibody-based techniques are revolutionizing the study of HTATIP2 protein interactions. These advanced methodologies enable researchers to capture the complex interactome of HTATIP2 with unprecedented precision and comprehensiveness.

Proximity-based labeling approaches represent a significant advancement in studying HTATIP2 interactions. These include:

  • BioID (proximity-dependent biotin identification): By creating HTATIP2-BirA* fusion proteins, researchers can identify proteins that come into close proximity with HTATIP2 in living cells. The BirA* biotin ligase attached to HTATIP2 biotinylates nearby proteins, which can then be purified using streptavidin and identified by mass spectrometry.

  • APEX2 (engineered ascorbate peroxidase): Similar to BioID but with faster labeling kinetics, HTATIP2-APEX2 fusions enable temporal mapping of protein interactions during specific cellular processes.

  • Antibody-based proximity proteomics: Using anti-HTATIP2 antibodies conjugated to peroxidases or photoactivatable crosslinkers to identify interacting proteins within their native cellular environment.

High-resolution imaging techniques coupling HTATIP2 antibodies with advanced microscopy include:

  • Super-resolution microscopy (STORM/PALM): Using fluorophore-conjugated anti-HTATIP2 antibodies to visualize HTATIP2 localization and co-localization with interacting partners at nanometer resolution.

  • FRET (Förster Resonance Energy Transfer): Employing antibody fragments labeled with compatible fluorophores to detect direct protein-protein interactions with HTATIP2 in real-time.

  • Expansion microscopy: Physically expanding specimens after antibody labeling to achieve super-resolution imaging on conventional microscopes.

Mass spectrometry-coupled approaches include:

  • Immunoprecipitation-mass spectrometry (IP-MS): Using highly specific anti-HTATIP2 antibodies to pull down HTATIP2 complexes followed by mass spectrometric identification of binding partners.

  • Cross-linking mass spectrometry (XL-MS): Combining chemical crosslinking with anti-HTATIP2 immunoprecipitation to capture transient interactions before MS analysis.

These innovative techniques collectively provide unprecedented insights into HTATIP2's protein interaction networks, helping to elucidate its roles in cancer progression, vascular remodeling, and autoimmune processes.

How can researchers resolve contradictory findings regarding HTATIP2 expression in different cancer types?

Resolving contradictory findings regarding HTATIP2 expression across cancer types requires systematic methodological approaches and careful data interpretation. Researchers should address these discrepancies through the following strategies:

  • Standardized detection methods: Implement consistent antibody-based detection protocols across studies, including:

    • Using validated antibodies with proven specificity for HTATIP2

    • Standardizing immunohistochemistry scoring systems using the three-category intensity scale (weak, moderate, strong)

    • Applying uniform thresholds for defining overexpression (e.g., ≥50% moderate-to-strong staining or ≥25% strong staining)

  • Comprehensive tissue analysis: Analyze multiple regions within tumor samples to account for intratumoral heterogeneity. For each cancer type, examine both primary tumors and metastatic sites to understand expression changes during disease progression.

  • Isoform-specific analysis: Different cancer types may express distinct HTATIP2 isoforms. Employ isoform-specific antibodies and primers to distinguish between variants that might have different or even opposing functions.

  • Contextual evaluation: Consider HTATIP2 expression in the context of:

    • Cancer subtype (e.g., intestinal-type vs. diffuse-type gastric cancer)

    • Histological grade and clinical stage

    • Molecular subtypes defined by comprehensive genomic profiling

    • Tumor microenvironment composition

  • Functional validation: Beyond expression studies, conduct functional assays to determine whether HTATIP2 has context-dependent roles:

    • Overexpression and knockdown studies in cell lines representing different cancer types

    • Migration and invasion assays to assess metastatic potential

    • Analysis of EMT markers (Snail, Slug) to understand underlying mechanisms

  • Multi-omics integration: Correlate HTATIP2 protein expression with:

    • Transcriptomic data to identify co-expressed genes

    • Mutation profiles to detect genetic alterations affecting HTATIP2 function

    • Epigenetic modifications that might regulate HTATIP2 expression

  • Survival analysis validation: Utilize multiple approaches like Kaplan-Meier survival analysis and PrognoScan database queries to verify prognostic implications across cancer types .

By implementing these approaches, researchers can reconcile contradictory findings and develop a more nuanced understanding of HTATIP2's cancer-specific roles, potentially revealing opportunities for targeted therapeutic interventions.

What are the emerging applications of HTATIP2 antibodies in targeted therapy development?

The emerging applications of HTATIP2 antibodies in targeted therapy development represent a promising frontier in translational medicine, particularly in oncology and vascular diseases. These antibody-based approaches are moving beyond traditional diagnostic applications to therapeutic interventions based on HTATIP2's established biological roles.

In cancer therapeutics, emerging approaches include:

  • Antibody-drug conjugates (ADCs): For cancers where HTATIP2 is overexpressed, anti-HTATIP2 antibodies conjugated to cytotoxic payloads could deliver targeted therapy. While gastric cancer typically shows better prognosis with HTATIP2 overexpression , other cancer types might exhibit different patterns where targeted degradation could be beneficial.

  • Bispecific antibodies: Engineered antibodies that simultaneously target HTATIP2 and immune effector cells (T cells or NK cells) could enhance immune surveillance against cancer cells expressing altered HTATIP2 levels.

  • CAR-T cell therapy: Anti-HTATIP2 single-chain variable fragments (scFvs) are being investigated as targeting domains for chimeric antigen receptor T cells in cancers with aberrant HTATIP2 expression.

In vascular disease applications, therapeutic approaches focus on:

  • HTATIP2 modulation for angiogenesis: Given HTATIP2's inhibitory effect on neovascularization, antibodies that neutralize HTATIP2 function could potentially enhance angiogenesis in ischemic conditions like chronic limb threatening ischemia .

  • Cell-based therapy enhancement: Anti-HTATIP2 antibodies or siRNA approaches targeting HTATIP2 are being explored to improve the efficacy of autologous cell therapies by enhancing the pro-arteriogenic capacity of therapeutic cells .

  • Targeted nanoparticle delivery: Anti-HTATIP2 antibodies conjugated to nanoparticles carrying therapeutic cargo (siRNAs, small molecules) can direct treatment to specific cell populations like monocyte/macrophages where HTATIP2 modulation might improve vascular remodeling.

For autoimmune applications, particularly in type 1 diabetes where HTATIP2 influences T cell regulation , therapeutic antibodies are being developed to modulate HTATIP2 activity in immune cells, potentially correcting dysregulated immune responses.

These emerging therapeutic applications highlight HTATIP2's increasingly recognized importance across multiple disease contexts and underscore the versatility of antibody-based approaches in translating biological insights into potential clinical interventions.

What are the key quality control parameters for validating novel anti-HTATIP2 antibodies?

Rigorous quality control is essential when validating novel anti-HTATIP2 antibodies to ensure experimental reproducibility and data reliability. Researchers should implement the following comprehensive validation parameters:

  • Specificity assessment:

    • Western blot analysis demonstrating a single band at the expected molecular weight (~27 kDa for HTATIP2)

    • Immunoprecipitation followed by mass spectrometry confirmation

    • Parallel testing in HTATIP2 knockout/knockdown systems

    • Peptide competition assays showing signal reduction when antibody is pre-incubated with immunizing peptide

  • Sensitivity evaluation:

    • Limit of detection determination using purified recombinant HTATIP2 protein in concentration gradients

    • Signal-to-noise ratio calculation across multiple applications

    • Comparison with established commercial antibodies using standardized samples

  • Application-specific validation:

    • For Western blotting: Linearity of signal across protein concentration range

    • For immunohistochemistry: Consistent staining patterns across multiple tissue samples with known HTATIP2 expression levels

    • For immunofluorescence: Subcellular localization consistent with known HTATIP2 distribution

    • For flow cytometry: Clear separation between positive and negative populations

  • Reproducibility testing:

    • Lot-to-lot consistency evaluation

    • Inter-laboratory comparison when possible

    • Stability assessment under various storage conditions

  • Cross-reactivity profiling:

    • Testing against closely related proteins

    • Species cross-reactivity analysis for evolutionary studies

    • Evaluation in tissues known to be negative for HTATIP2 expression

  • Documentation requirements:

    • Complete antibody characteristics (host species, clonality, immunogen sequence, accession number reference)

    • Detailed validation protocols with positive and negative controls

    • Application-specific recommended conditions (dilutions, buffers, incubation times)

Implementing these validation parameters ensures that novel anti-HTATIP2 antibodies will generate reliable and reproducible results across diverse experimental applications, from basic research to potential clinical translation.

How does epitope accessibility affect HTATIP2 detection in different sample types?

Epitope accessibility significantly impacts HTATIP2 detection across various sample types, requiring tailored approaches to ensure reliable antibody-based detection. This accessibility can be affected by numerous factors related to both sample preparation and the intrinsic properties of HTATIP2 in different cellular contexts.

In fixed tissue samples, formaldehyde fixation creates protein cross-links that can mask HTATIP2 epitopes. The standard approach using 4% paraformaldehyde fixation requires optimization of antigen retrieval methods:

  • Heat-induced epitope retrieval (HIER) with citrate buffer (pH 6.0) typically works well for HTATIP2 detection

  • For challenging samples, alternative methods such as protease-induced epitope retrieval or high-pressure cooking may improve accessibility

  • Fixation time should be standardized (typically 24 hours) to prevent overfixation that permanently masks epitopes

In cell culture samples, detection varies by application:

  • For flow cytometry, membrane permeabilization protocols must be optimized as HTATIP2 has both cytoplasmic and nuclear localization

  • For immunofluorescence, different fixatives (methanol vs. paraformaldehyde) may reveal different HTATIP2 epitopes

  • For live-cell imaging, careful selection of antibodies targeting extracellular epitopes is necessary

In protein lysates for Western blotting:

  • Lysis buffer composition significantly affects epitope exposure; RIPA buffer is generally effective for HTATIP2 extraction

  • Sample denaturation conditions (temperature, detergent concentration) require optimization

  • Reducing agents are essential to break disulfide bonds that might obscure epitopes

Post-translational modifications of HTATIP2 can mask epitopes in context-dependent ways:

  • Phosphorylation states may alter antibody recognition

  • Protein-protein interactions in different cell types may obscure binding sites

  • Conformational changes under different cellular conditions may expose or hide epitopes

When working with clinical samples, additional considerations include:

  • Pre-analytical variables like ischemic time before fixation

  • Storage duration and conditions of archived samples

  • Decalcification procedures for bone-containing samples that may impact HTATIP2 epitope integrity

Addressing these epitope accessibility challenges requires systematic optimization for each sample type and application, often necessitating the use of multiple antibodies targeting different HTATIP2 epitopes to ensure comprehensive detection.

How can single-cell analysis techniques advance our understanding of HTATIP2 in heterogeneous tissues?

Single-cell analysis techniques offer unprecedented opportunities to elucidate HTATIP2's role in heterogeneous tissues, providing insights that bulk tissue analyses cannot capture. These technologies are particularly valuable for understanding HTATIP2's cell type-specific functions in complex microenvironments such as tumors, vascular tissues, and immune organs.

Single-cell RNA sequencing (scRNA-seq) enables researchers to:

  • Map HTATIP2 expression across diverse cell populations within heterogeneous tissues

  • Identify previously unrecognized cell types where HTATIP2 plays critical roles

  • Discover cell state-specific expression patterns during disease progression

  • Detect rare cell populations with unique HTATIP2 expression profiles that might be diluted in bulk analyses

For protein-level analysis, emerging technologies include:

  • Mass cytometry (CyTOF) using metal-conjugated anti-HTATIP2 antibodies to quantify protein expression alongside dozens of other markers

  • Single-cell Western blotting to directly measure HTATIP2 protein levels in individual cells

  • Imaging mass cytometry combining tissue morphology with single-cell HTATIP2 expression data

Spatial transcriptomics and proteomics add crucial contextual information by:

  • Preserving tissue architecture while measuring HTATIP2 expression

  • Revealing spatial relationships between HTATIP2-expressing cells and their microenvironment

  • Mapping HTATIP2 expression gradients that might influence disease progression

Functional single-cell approaches are also emerging:

  • CRISPR-based lineage tracing to track the fate of HTATIP2-expressing cells during development or disease

  • Single-cell secretome analysis to understand how HTATIP2 affects cellular communication

  • Multi-modal approaches combining transcriptomics, proteomics, and epigenomics at single-cell resolution

These technologies will help resolve contradictory findings regarding HTATIP2's role across different contexts, particularly in:

  • Gastric cancer, where HTATIP2 expression varies between intestinal and diffuse subtypes

  • Vascular diseases, where monocyte/macrophage heterogeneity influences HTATIP2-dependent angiogenic responses

  • Autoimmune conditions, where T cell subset-specific HTATIP2 functions may determine disease outcomes

The integration of these single-cell technologies promises to revolutionize our understanding of HTATIP2's context-dependent functions and potentially identify new therapeutic opportunities across multiple disease domains.

What are the implications of HTATIP2 post-translational modifications for antibody selection and experimental design?

HTATIP2 undergoes various post-translational modifications (PTMs) that significantly impact its function, localization, and protein interactions. These modifications have critical implications for antibody selection and experimental design when studying HTATIP2 biology.

Phosphorylation represents a major regulatory mechanism for HTATIP2. Multiple serine, threonine, and tyrosine residues can be phosphorylated, potentially altering protein conformation and function. When selecting antibodies, researchers should consider:

  • Phospho-specific antibodies that recognize specific phosphorylated residues

  • Phosphorylation-independent antibodies that detect HTATIP2 regardless of phosphorylation status

  • Combination approaches using both types to comprehensively analyze HTATIP2 regulation

Experimental designs should include:

  • Phosphatase treatments to determine if antibody recognition is phosphorylation-dependent

  • Kinase inhibitors to evaluate dynamic phosphorylation changes

  • Site-directed mutagenesis of key phosphorylation sites to assess functional impacts

Acetylation of HTATIP2 may also occur, potentially affecting its interactions with DNA or other proteins. For acetylation studies:

  • Acetylation-specific antibodies are needed to detect this modification

  • Experimental designs should include HDAC inhibitors to preserve acetylation status

  • Mass spectrometry validation should confirm specific acetylation sites

Ubiquitination and SUMOylation can regulate HTATIP2 stability and localization. Considerations include:

  • Antibodies recognizing total HTATIP2 may give misleading results if ubiquitination masks epitopes

  • Proteasome inhibitors should be included when studying HTATIP2 stability

  • Denaturing conditions may be necessary to detect highly ubiquitinated forms

For comprehensive PTM analysis, researchers should:

  • Perform immunoprecipitation with anti-HTATIP2 antibodies followed by mass spectrometry

  • Use multiple antibodies targeting different epitopes to ensure comprehensive detection

  • Include appropriate controls for each PTM (phosphatase treatment, HDAC inhibitors, etc.)

  • Consider cell type-specific PTM patterns, as modifications may vary between tissues

When studying HTATIP2 in disease contexts like cancer or vascular disorders , PTM analysis becomes especially important as aberrant modifications may contribute to pathogenesis. Experimental designs should therefore incorporate these considerations to accurately characterize HTATIP2's dynamic regulation and develop more targeted therapeutic approaches.

What are the most significant unresolved questions in HTATIP2 antibody research?

Despite substantial progress in understanding HTATIP2's biological roles, several critical questions remain unresolved in HTATIP2 antibody research. These knowledge gaps represent important opportunities for future investigations that could significantly advance both basic science and translational applications.

First, the tissue-specific functions of HTATIP2 require further elucidation. While HTATIP2's role as a favorable prognostic marker in gastric cancer is established , and its involvement in vascular remodeling and T cell regulation in diabetes has been identified, the mechanistic basis for these seemingly diverse functions remains unclear. Developing antibodies that can distinguish between different functional states of HTATIP2 would help resolve this question.

Second, the relationship between HTATIP2 structure and function needs deeper investigation. Current antibodies typically target linear epitopes, but developing conformation-specific antibodies could reveal how HTATIP2's structure changes in different cellular contexts or disease states. This approach might explain why HTATIP2 can function as a tumor suppressor in some cancers while potentially playing different roles in vascular and immune cells.

Third, the temporal dynamics of HTATIP2 expression during disease progression remain poorly understood. Most studies provide static snapshots rather than longitudinal analyses. Developing antibody-based imaging approaches for tracking HTATIP2 expression in real-time would provide valuable insights into its role during disease initiation and progression.

Fourth, the exact mechanisms by which HTATIP2 regulates epithelial-mesenchymal transition in cancer cells and neovascularization in ischemic tissues need further investigation. Antibodies that can block specific protein-protein interactions involving HTATIP2 would help dissect these pathways.

Fifth, the therapeutic potential of targeting HTATIP2 remains largely unexplored. Developing therapeutic antibodies or antibody-drug conjugates targeting HTATIP2 could provide novel treatment approaches for conditions where HTATIP2 dysfunction contributes to pathology.

Finally, standardization of HTATIP2 detection methodologies across different research groups would address the current challenge of contradictory findings. Establishing reference standards and validated protocols for HTATIP2 antibody applications would enhance data reproducibility and accelerate research progress in this promising field.

How might advances in antibody technology reshape HTATIP2 research in the next decade?

Emerging antibody technologies are poised to revolutionize HTATIP2 research over the next decade, potentially transforming our understanding of this protein's role in disease pathways and enabling novel therapeutic approaches. These technological advances will likely impact HTATIP2 research in several transformative ways.

Synthetic antibody development using phage display and yeast display technologies will enable the generation of highly specific anti-HTATIP2 antibodies with precisely engineered properties. These technologies allow for the selection of antibodies against specific conformational states or post-translational modifications of HTATIP2, providing unprecedented insights into its functional dynamics. Additionally, the ability to engineer antibodies that recognize specific epitopes will help resolve contradictory findings about HTATIP2's role in different tissues and disease states.

Nanobodies and single-domain antibodies derived from camelids or sharks will transform intracellular HTATIP2 research. Their small size allows better penetration into cells and access to sterically hindered epitopes. When expressed as intrabodies, they can track and potentially modulate HTATIP2 function in living cells, offering real-time visualization of HTATIP2 dynamics during cellular processes like migration, invasion, and angiogenesis .

Multispecific antibodies targeting HTATIP2 alongside other proteins will enable more sophisticated functional studies. Bispecific antibodies could simultaneously target HTATIP2 and its interaction partners to elucidate signaling networks, while trispecific formats could additionally recruit effector cells or deliver therapeutic payloads in HTATIP2-expressing tissues.

Antibody-based proximity labeling will map the HTATIP2 interactome with unprecedented detail. By conjugating enzymes like BirA* or APEX2 to anti-HTATIP2 antibodies, researchers can identify proteins that interact with HTATIP2 in specific cellular compartments or under particular disease conditions, potentially revealing new therapeutic targets in cancer , vascular diseases , and autoimmune conditions .

CRISPR-based antibody screening platforms will accelerate the discovery of functional antibodies against HTATIP2. These high-throughput approaches will identify antibodies that not only bind HTATIP2 but also modulate its function, potentially leading to therapeutic candidates for conditions where HTATIP2 dysregulation contributes to pathology.

Smart antibody delivery systems using nanoparticles, cell-penetrating peptides, or exosomes will enhance the delivery of anti-HTATIP2 antibodies to specific tissues and cellular compartments. This will be particularly valuable for studying HTATIP2's role in difficult-to-access tissues like the central nervous system or in specific immune cell populations.

Product Science Overview

Introduction

HIV-1 Tat Interactive Protein 2 (HTATIP2), also known as TIP30, CC3, or SDR44U1, is a protein encoded by the HTATIP2 gene. This protein plays a crucial role in various cellular processes, including tumor suppression, apoptosis, and angiogenesis regulation .

Gene and Protein Structure

HTATIP2 is a protein-coding gene located on chromosome 11 in humans. The protein itself is approximately 30 kDa in size and belongs to the short-chain dehydrogenases/reductases (SDR) family . It has multiple isoforms, with Isoform 1 acting as a metastasis suppressor and Isoform 2 having antiapoptotic effects .

Function and Mechanism

HTATIP2 is involved in several critical cellular functions:

  • Tumor Suppression: HTATIP2 acts as a tumor suppressor by inhibiting nuclear import and competing with nuclear import substrates for binding to nuclear transport receptors . It also induces the expression of apoptosis-related genes such as Bad and Siva .
  • Apoptosis and Angiogenesis: The protein plays a role in the positive regulation of programmed cell death and transcription by RNA polymerase II. It also has antiangiogenic properties, which help in inhibiting the formation of new blood vessels in tumors .
  • Redox Sensor: HTATIP2 may act as a redox sensor linked to transcription through the regulation of nuclear import .
Clinical Significance

Mutations or defects in the HTATIP2 gene are associated with various diseases, including cervical cancer and osteogenesis imperfecta, type V . The protein’s role in tumor suppression and apoptosis makes it a potential target for antitumor therapies .

Mouse Anti-Human HTATIP2 Antibody

The mouse anti-human HTATIP2 monoclonal antibody is used in research to study the protein’s function and its role in various diseases. This antibody is derived from hybridization of mouse myeloma cells with spleen cells from BALB/c mice immunized with recombinant human HTATIP2 . It is used in various applications, including ELISA, Western Blot, Flow Cytometry, Immunocytochemistry, and Immunofluorescence .

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.