TPPP3 Human

Tubulin Polymerization-Promoting Protein Family Member 3 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Molecular Profile

TPPP3 is a 21.4 kDa non-glycosylated protein containing 199 amino acids (1–176 residues) with a His-tag fusion for purification . Its amino acid sequence includes a conserved region critical for tubulin binding and microtubule stabilization .

PropertyDetails
Molecular Mass21.4 kDa (recombinant form in E. coli)
Amino Acid SequenceMGSSHHHHHHSSGLVPRGSH... (N-terminal His-tag included)
Expression SiteCytoplasmic
Key InteractionsTubulin, STAT3, Snail1, MMPs, β-catenin

Core Functions

  • Microtubule Organization: Promotes tubulin polymerization and stabilizes microtubule bundles, modulating mitotic processes .

  • Cell Cycle Regulation: Affects G1/S transition and apoptosis via interactions with STAT3, BCL-2, and Bax .

  • Pathway Activation: Modulates STAT3/Twist1 (migration/invasion) and β-catenin/NF-κB/COX-2 (decidualization) .

Oncogenic Roles

Cancer TypeMechanismClinical Correlation
NSCLCSTAT3 activation → Twist1 upregulation → EMT (migration/invasion) High expression → Poor OS/DFS
Colorectal Cancer (CRC)MMP-9/Rac-1 upregulation → Cell invasion; BCL-2 downregulation → Apoptosis High expression → Advanced TNM stage, lymph node metastasis
GlioblastomaSnail1-mediated EMT → Increased migration/proliferation High expression → Reduced survival

Tumor-Suppressive Roles

Cancer TypeMechanismClinical Correlation
Nasopharyngeal Carcinoma (NPC)Cell cycle arrest (DNA replication inhibition); MMP-2/9 downregulation → Reduced invasion Low expression → Poor prognosis

Key Signaling Pathways

  1. STAT3/Twist1 Axis:

    • TPPP3 overexpression → STAT3 phosphorylation → Twist1 upregulation → EMT (NSCLC) .

    • Inhibition by Cryptotanshinone (STAT3 inhibitor) → Reduced Twist1, MMP-2/9, and invasion .

  2. Snail1-Mediated EMT:

    • TPPP3 → Snail1 upregulation → E-cadherin ↓, N-cadherin ↑ → Mesenchymal transition (glioblastoma) .

  3. β-Catenin/NF-κB/COX-2 Cascade:

    • TPPP3 knockdown → β-catenin ↓, NF-κB nuclear translocation ↓ → COX-2 ↓ → Decidualization failure (endometrial cells) .

Prognostic Markers

Cancer TypeExpression PatternPrognostic Outcome
NSCLCHigh TPPP3 → High Twist1Poor OS/DFS
CRCHigh TPPP3 → Advanced stageShorter survival
GlioblastomaHigh TPPP3 → Snail1 ↑Poor survival
NPCLow TPPP3 → Cell cycle arrestPoor prognosis

Therapeutic Targets

  • Cancer-Specific Approaches:

    • NSCLC/CRC: STAT3 inhibitors (e.g., Cryptotanshinone) or TPPP3 siRNA .

    • Glioblastoma: Snail1-targeted therapies to disrupt EMT .

  • Non-Cancer: TPPP3 modulation for nerve regeneration or vascular injury .

Unresolved Questions

  1. Tissue-Specific Dual Roles: Why does TPPP3 act as an oncogene in NSCLC/CRC but a tumor suppressor in NPC?

  2. Epigenetic Regulation: Mechanisms driving TPPP3 downregulation in NPC vs. upregulation in other cancers .

  3. Immune Interactions: Role of TPPP3 in immune cell infiltration (e.g., CD8+ T cells, B memory cells) .

Emerging Avenues

  • Biomarker Development: Multiplex assays for TPPP3/Snail1/STAT3 in glioblastoma .

  • Preclinical Models: TPPP3 knockout mice for studying nerve regeneration and vascular repair .

References

  1. ProSpecBio. TPPP3 Human Recombinant.

  2. PMC9564178. Modulatory Role of TPPP3 in Microtubule Organization.

  3. Karger. TPPP3 Promotes Cell Proliferation, Invasion, and Tumor Metastasis.

  4. ASCO. Knockdown of TPPP3 to Inhibit Cell Proliferation and Invasion in CRC.

  5. Ma'ayan Lab. TPPP3 Gene Functional Associations.

  6. J Cancer. Knockdown of TPPP3 in CRC.

  7. J Endocrinol. Inhibition of TPPP3 Attenuates β-Catenin/NF-κB/COX-2 Signaling.

  8. Nature. TPPP3 Promotes EMT via Snail1 in Glioblastoma.

  9. PMC. TPPP3 and Immune Infiltrates in HNSC.

  10. PMC. TPPP3 in NPC: Tumor Suppression via Cell Cycle Regulation.

Product Specs

Introduction
TPPP3, a member of the TPPP family, is located in the cytoplasm and plays a role in mitosis and cell proliferation. It exhibits microtubule bundling activity and binds to tubulin.
Description
Recombinant human TPPP3, expressed in E. coli, is a non-glycosylated polypeptide chain lacking any glycosylation modifications. It consists of 199 amino acids, with a sequence spanning from position 1 to 176, and has a molecular weight of 21.4 kDa. The protein is engineered with a 23 amino acid His-tag fused to its N-terminus to facilitate purification, which is achieved using proprietary chromatographic methods.
Physical Appearance
The product is a clear, colorless solution that has undergone sterile filtration.
Formulation
TPPP3 is supplied in a buffer solution containing 20 mM Tris-HCl (pH 8.0), 0.15 M NaCl, 1 mM DTT, and 10% glycerol.
Stability
For short-term storage (up to 2-4 weeks), the product can be stored at 4°C. For extended storage, it is recommended to store the product frozen at -20°C. To ensure optimal stability during long-term storage, adding a carrier protein (0.1% HSA or BSA) is advised. Repeated freezing and thawing should be avoided.
Purity
The purity of TPPP3 is determined to be greater than 95% using SDS-PAGE analysis.
Synonyms
Tubulin Polymerization-Promoting Protein Family Member 3, Brain Specific Protein, p25gamma, TPPP/p20, p20.
Source
Escherichia Coli.
Amino Acid Sequence

MGSSHHHHHH SSGLVPRGSH MGSMAASTDM AGLEESFRKF AIHGDPKASG QEMNGKNWAK LCKDCKVADG KSVTGTDVDI VFSKVKGKSA RVINYEEFKK ALEELATKRF KGKSKEEAFD AICQLVAGKE PANVGVTKAK TGGAVDRLTD TSRYTGSHKE RFDESGKGKG IAGRQDILDD SGYVSAYKNA GTYDAKVKK 

Q&A

What is TPPP3 and what is its primary function in human cells?

TPPP3 (Tubulin Polymerization Promoting Protein family member 3) is a 176-amino acid protein that functions primarily as a regulator of microtubule dynamics with demonstrated microtubule bundling activity . As a member of the TPPP family, it plays essential roles in microtubule organization, stabilization, and bundling by interacting with alpha and beta tubulins . Unlike its homolog TPPP1 (which contains an N-terminal extension), TPPP3 has a more compact structure but maintains similar functionality in microtubule regulation . Its expression is tissue-specific, with notable presence in various cell types including certain neural populations and progenitor cells in synovial/tendon sheath tissues .

Methodologically, researchers typically study TPPP3's microtubule-related functions through in vitro tubulin polymerization assays, microscopic visualization of cytoskeletal arrangements, and protein-protein interaction analyses using techniques such as co-immunoprecipitation and fluorescence complementation assays .

How is TPPP3 expression regulated in normal versus pathological conditions?

TPPP3 expression shows significant variation between normal tissues and pathological states, with particularly notable differences in cancer contexts. In glioblastoma multiforme (GBM), TPPP3 expression is significantly higher than in normal brain tissue (NBT), with expression levels increasing proportionally with glioma grade . Quantitative analysis demonstrates this progressive increase both at mRNA and protein levels, with Western blot experiments consistently showing elevated TPPP3 expression in glioblastoma cell lines compared to human normal astrocytes (NHA) .

For researchers investigating expression patterns, methodological approaches include:

  • Real-time fluorescence quantitative PCR for mRNA abundance analysis

  • Western blot for protein expression level quantification

  • Immunohistochemistry for spatial localization in tissues

  • Single-cell RNA sequencing for cell-specific expression profiling

When designing expression studies, researchers should include appropriate normal tissue controls and multiple pathological samples representing different disease stages for comprehensive analysis.

What are the known protein interactions of TPPP3 and their functional significance?

TPPP3 engages in several key protein interactions that mediate its biological functions:

Interaction PartnerInteraction TypeFunctional SignificanceResearch Methods
α- and β-tubulinsDirect bindingFacilitates microtubule formation and stabilizationCo-IP, in vitro binding assays
TPPP1Homo/hetero-associationForms functional complexes affecting microtubule dynamicsBifunctional fluorescence complementation
β-cateninRegulatory interactionImplicated in embryo implantation and decidualizationCo-localization studies
Snail1Regulatory relationshipMediates epithelial-mesenchymal transition in cancer contextsExpression correlation analysis

Unlike TPPP1, TPPP3 shows virtually no binding affinity for alpha-synuclein (SYN), which may explain its potential protective role in Parkinson's disease models by counteracting the formation of TPPP1-SYN pathological complexes/aggregates . For comprehensive investigation of protein interactions, researchers should employ multiple complementary techniques including co-immunoprecipitation, proximity ligation assays, and fluorescence resonance energy transfer.

How does TPPP3 contribute to epithelial-mesenchymal transition (EMT) in glioblastoma progression?

TPPP3 promotes epithelial-mesenchymal transition in glioblastoma multiforme (GBM) primarily through regulation of Snail1 protein expression . Experimental evidence demonstrates a clear mechanistic relationship where:

  • TPPP3 upregulation in glioblastoma cells enhances migration, invasion, and proliferation while reducing apoptosis in vitro

  • Conversely, TPPP3 inhibition reduces migration, invasion, proliferation and induces apoptosis

  • Clinical data analysis confirms a positive correlation between TPPP3 and Snail1 protein expression levels in glioblastomas

  • Lower TPPP3 expression correlates with better survival expectations in GBM patients

To investigate this relationship methodologically, researchers should employ a multi-faceted approach:

  • RNA interference or CRISPR-based gene editing for TPPP3 knockdown/knockout

  • Overexpression systems using appropriate vectors for gain-of-function studies

  • Assessment of EMT markers (E-cadherin, N-cadherin, vimentin) via Western blot and immunofluorescence

  • Functional assays including wound healing, transwell invasion, and proliferation assays

  • Rescue experiments to confirm specificity of observed effects

The TPPP3-Snail1 axis represents a potential therapeutic target, requiring careful validation through in vivo models following initial in vitro characterization.

What is the therapeutic potential of TPPP3 in Parkinson's disease compared to other neurodegenerative conditions?

TPPP3 demonstrates significant therapeutic potential for Parkinson's disease through a unique anti-aggregative mechanism. Unlike its homolog TPPP1, which binds to alpha-synuclein (SYN) and promotes pathological aggregation, TPPP3 virtually does not bind to SYN . This distinctive property enables TPPP3 to counteract the formation of TPPP1-SYN pathological complexes by tightly associating with TPPP1 but not with SYN, thereby potentially inhibiting the aggregation process fundamental to Parkinson's pathology .

Research approaches for investigating this therapeutic potential include:

  • Comparative binding studies between TPPP3, TPPP1, and alpha-synuclein using purified recombinant proteins

  • Aggregation assays with thioflavin-T fluorescence or similar techniques

  • Cellular models of synucleinopathy with TPPP3 intervention

  • Structure-function analysis to identify specific TPPP3 fragments with optimal anti-aggregative properties

Recent research suggests TPPP3 may also have broader neurodegenerative applications, particularly in optic nerve regeneration. Studies demonstrate that TPPP3 overexpression in rodent optic nerve crush models enhances axon regeneration and improves retinal ganglion cell survival . These findings indicate TPPP3 might function through multiple neuroprotective and regenerative mechanisms, including:

  • Promotion of neurite outgrowth

  • Stabilization of microtubules critical for axon growth

  • Upregulation of pro-regenerative genes such as Bmp4

  • Modification of inflammatory pathways relevant to axonal repair

What molecular mechanisms explain the differential effects of TPPP3 in cancer progression across different tissue types?

TPPP3 exhibits context-dependent effects across different cancer types, functioning as either a promoter or inhibitor of malignancy depending on the tissue context. This duality presents a significant challenge for cancer research and therapeutic development.

In glioblastoma, TPPP3 clearly promotes malignant progression by:

  • Enhancing migration, invasion, and proliferation capabilities

  • Reducing apoptotic tendency

  • Promoting EMT through Snail1 regulation

  • Correlating with worse clinical outcomes

Conversely, in certain other cancers, TPPP3 appears to inhibit proliferation, invasion, and migration . This contextual dichotomy likely reflects tissue-specific molecular landscapes and signaling network configurations.

Methodologically, researchers investigating these differential effects should:

  • Perform comparative transcriptomic and proteomic analyses across multiple cancer types with TPPP3 manipulation

  • Identify tissue-specific TPPP3 interaction partners using techniques like BioID or proximity labeling

  • Map TPPP3-responsive signaling pathways through phosphoproteomics

  • Analyze epigenetic regulation of TPPP3 and its target genes in different cellular contexts

  • Employ CRISPR-based screens to identify synthetic lethal interactions in TPPP3-high versus TPPP3-low cancers

Understanding these context-dependent mechanisms is crucial for developing targeted therapeutic approaches that exploit TPPP3's functions without triggering adverse effects in unintended tissues.

What are the optimal methods for studying TPPP3 expression and localization in human tissues?

For comprehensive characterization of TPPP3 expression and localization in human tissues, researchers should employ complementary approaches:

TechniqueApplicationAdvantagesLimitations
RT-qPCRmRNA quantificationHigh sensitivity, quantitativeNo spatial information, post-transcriptional modifications not detected
Western blotProtein expressionSemi-quantitative, detects specific isoformsLimited spatial information
ImmunohistochemistryTissue localizationPreserves tissue architecture, spatial contextLower quantitative precision
ImmunofluorescenceSubcellular localizationHigh-resolution visualization, co-localization studiesPotential autofluorescence interference
Single-cell RNA-seqCell-type specific expressionReveals heterogeneity, discovers rare populationsTechnically challenging, costly
Mass spectrometryProtein identification, PTMsUnbiased detection, identifies modificationsSample preparation challenges

When studying TPPP3 in human tissues, particular attention should be paid to antibody validation, as cross-reactivity with other TPPP family members can confound results. The use of knockout/knockdown controls and multiple antibodies targeting different epitopes is recommended .

For clinical samples, optimized fixation protocols are essential as improper fixation can affect TPPP3 immunoreactivity. Fresh-frozen tissue analysis in parallel with fixed specimens can provide complementary data when feasible.

How can researchers effectively manipulate TPPP3 expression in experimental models?

Effective manipulation of TPPP3 expression requires selection of appropriate genetic tools based on research objectives:

For TPPP3 knockdown:

  • Short hairpin RNA (shRNA) has been successfully employed to reduce TPPP3 expression in glioblastoma cells

  • siRNA offers transient knockdown suitable for short-term experiments

  • CRISPR-Cas9 provides more complete and stable knockout for long-term studies

For TPPP3 overexpression:

  • Plasmid vectors (e.g., pcDNA-TPPP3) have been validated for TPPP3 upregulation

  • Viral delivery systems (lentivirus, adenovirus) offer higher transduction efficiency in difficult-to-transfect cells

  • Inducible expression systems allow temporal control of TPPP3 levels

For in vivo studies, conditional knockout or transgenic models using Cre-loxP systems provide tissue-specific manipulation. Recent studies have employed Tppp3 inducible reporter mice in combination with other lineage reporters to track cell fate in heterotopic ossification models .

Key methodological considerations include:

  • Verification of altered expression by multiple methods (RT-qPCR, Western blot, immunofluorescence)

  • Use of appropriate controls (empty vector, scrambled siRNA)

  • Titration of expression levels to avoid non-physiological effects

  • Assessment of potential off-target effects

What high-throughput approaches can identify novel TPPP3-associated signaling pathways?

To comprehensively map TPPP3-associated signaling networks, researchers should consider these advanced high-throughput approaches:

  • Transcriptomic profiling:

    • RNA-seq of tissues/cells with manipulated TPPP3 expression has revealed TPPP3 upregulates pro-regenerative genes like Bmp4 and modulates inflammatory pathways

    • Single-cell RNA-seq can further dissect cell type-specific responses to TPPP3 modulation

  • Proteomic approaches:

    • Proximity labeling techniques (BioID, APEX) can identify proteins in close spatial proximity to TPPP3

    • Phosphoproteomics can reveal signaling cascades activated or suppressed following TPPP3 manipulation

    • Co-immunoprecipitation coupled with mass spectrometry for identifying direct interaction partners

  • Functional genomics:

    • CRISPR screens to identify synthetic lethal interactions or genetic dependencies related to TPPP3 function

    • Pooled shRNA libraries for pathway component identification

  • Systems biology integration:

    • Network analysis combining transcriptomic, proteomic, and interaction data

    • Pathway enrichment analysis to identify overrepresented functional categories

Recent studies using these approaches have begun to elucidate TPPP3's role in the BMP signaling pathway, though further research is needed to determine whether this interaction occurs through canonical or alternative pathways .

How might TPPP3-targeted therapies be developed for neurodegenerative disorders?

Development of TPPP3-based therapeutic strategies for neurodegenerative disorders, particularly Parkinson's disease, should follow a structured translational pathway:

  • Therapeutic modality selection:

    • Recombinant TPPP3 protein delivery or selected fragments with anti-aggregative properties

    • Gene therapy approaches to express TPPP3 in affected tissues

    • Small molecules that mimic TPPP3's anti-aggregative function or enhance endogenous TPPP3 activity

  • Delivery optimization:

    • Blood-brain barrier penetration strategies (nanoparticles, viral vectors, peptide conjugation)

    • Targeted delivery to specific neural populations

    • Controlled release formulations for sustained effect

  • Efficacy validation progression:

    • In vitro aggregation models → cellular models → animal models → clinical trials

    • Assessment of both neuroprotection and symptom amelioration

    • Long-term safety evaluation, particularly important given TPPP3's roles in cancer contexts

  • Biomarker development:

    • TPPP3 levels in cerebrospinal fluid or blood as potential diagnostic or prognostic indicators

    • Imaging approaches to visualize TPPP3-targeted engagement in vivo

For optic nerve regeneration applications, TPPP3-based approaches should focus on promoting long-distance axon regeneration with functional connectivity, as current research demonstrates primarily short-distance regeneration . Combination therapies incorporating TPPP3 with other regenerative factors may yield synergistic effects.

What preclinical validation is needed before pursuing TPPP3 as a therapeutic target in glioblastoma?

  • Target validation requirements:

    • Confirmation of TPPP3 overexpression in diverse patient cohorts

    • Prognostic significance verification in multiple independent datasets

    • Demonstration that TPPP3 inhibition reduces tumor growth in patient-derived xenograft models

    • Evaluation of effects on radiation and chemotherapy sensitivity

  • Inhibition strategy development:

    • siRNA/shRNA for proof-of-concept studies

    • Small molecule inhibitors targeting TPPP3-tubulin or TPPP3-Snail1 interactions

    • Aptamers or peptide-based approaches for specific targeting

    • Antibody-drug conjugates for targeted delivery to tumor cells

  • Combination therapy assessment:

    • TPPP3 inhibition + standard-of-care treatments (temozolomide, radiation)

    • TPPP3 inhibition + other targeted therapies (e.g., anti-angiogenic agents)

    • Evaluation for synergistic or additive effects

  • Resistance mechanism characterization:

    • Identification of potential bypass pathways

    • Monitoring for compensatory upregulation of other TPPP family members

    • Development of biomarkers for therapy response prediction

Careful toxicity profiling is particularly important given TPPP3's roles in normal cellular functions and potential beneficial effects in neurodegenerative contexts .

How can TPPP3 expression patterns inform precision medicine approaches in cancer?

TPPP3 expression patterns offer potential for personalized medicine approaches in cancer management:

  • Prognostic stratification:

    • In glioblastoma, low TPPP3 expression correlates with improved survival expectations

    • Expression levels could be incorporated into multi-factor prognostic models

    • Monitoring TPPP3 expression changes during treatment may provide early indicators of therapeutic response

  • Predictive biomarker development:

    • TPPP3 expression may predict response to specific therapeutic interventions

    • Rational combination therapy selection based on TPPP3 status

    • Resistance mechanism identification through correlative analysis with treatment outcomes

  • Patient selection for clinical trials:

    • Enrichment strategies based on TPPP3 expression levels

    • Companion diagnostic development for TPPP3-targeted therapies

    • Stratification of patients for different treatment arms

  • Therapeutic targeting considerations:

    • Context-dependent roles require careful tissue-specific approaches

    • Dual roles in cancer versus neurodegenerative disease necessitate targeted delivery systems

    • Potential for repurposing TPPP3-modulatory compounds across indications

Product Science Overview

Structure and Function

TPPP3 is a protein-coding gene that enables tubulin binding activity. It is involved in microtubule bundle formation and decidualization, which is the process by which the endometrial cells undergo structural and functional changes to support pregnancy . The protein colocalizes with microtubule bundles and the perinuclear region of the cytoplasm .

Role in Disease

TPPP3 has been associated with various diseases, including Creutzfeldt-Jakob Disease and Dementia . It has also been studied in the context of cancer. For instance, overexpression of TPPP3 has been shown to inhibit cell proliferation and invasion in nasopharyngeal carcinoma (NPC) cells . This suggests that TPPP3 may act as an antioncogene, playing a significant role in the occurrence and progression of NPC .

Research and Applications

Research has demonstrated that TPPP3 is a specific marker of the developing musculoskeletal system through its role in inducing tubulin polymerization . Its expression is increased in lung cancer and/or chronic obstructive pulmonary disease (COPD) groups, indicating its potential as a target for early diagnosis of lung cancer .

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.