TPI1 Human, Active

Triosephosphate Isomerase 1 Human Recombinant, Active
Shipped with Ice Packs
In Stock

Description

Functional Role in Glycolysis

TPI1 catalyzes the reversible isomerization of:
Dihydroxyacetone phosphate (DHAP)Glyceraldehyde 3-phosphate (G3P)\text{Dihydroxyacetone phosphate (DHAP)} \leftrightarrow \text{Glyceraldehyde 3-phosphate (G3P)}
This reaction is critical for glycolysis and gluconeogenesis, enabling efficient ATP production . The enzyme operates with near-perfect catalytic efficiency, achieving rate enhancements of 10910^9 compared to non-enzymatic reactions .

Oncogenic Implications in Lung Adenocarcinoma (LUAD)

Recent studies reveal non-canonical roles of TPI1 in cancer progression:

Key Findings from Preclinical Studies:

ObservationExperimental ModelImpact
Nuclear translocation under stressA549/H1299 lung cancer cellsEnhances chemoresistance
Knockdown effectsXenograft tumors60–70% reduction in tumor growth
Correlation with prognosisLUAD patient tissuesInverse survival correlation

Nuclear-localized TPI1 promotes tumorigenesis independent of its enzymatic activity, suggesting a moonlighting role in transcriptional regulation .

Clinical Relevance of TPI1 Dysregulation

Triosephosphate Isomerase Deficiency:

  • Genetic Cause: Autosomal recessive mutations in TPI1 (e.g., Glu104Asp) .

  • Symptoms:

    • Hemolytic anemia

    • Progressive neuromuscular degeneration

    • Cardiomyopathy

  • Pathology: Mutant enzymes exhibit reduced dimer stability, impairing glycolysis in erythrocytes and neurons .

Research Applications and Handling

Typical Use Cases:

  • Glycolytic pathway reconstitution assays

  • Enzyme kinetics studies

  • Cancer metabolism investigations

Product Specs

Introduction
TPI1, a member of the triosephosphate isomerase family, plays a crucial role in glycolysis and gluconeogenesis by catalyzing the interconversion of glyceraldehydes 3-phosphate (G3P) and dihydroxy-acetone phosphate (DHAP). Mutations in the TPI1 gene can lead to triosephosphate isomerase deficiency (TPI deficiency), an autosomal recessive disorder characterized by severe clinical manifestations such as neonatal jaundice, chronic hemolytic anemia, progressive neuromuscular dysfunction, cardiomyopathy, and increased vulnerability to infections.
Description
Recombinant TPI1, expressed in E. coli, is a single, non-glycosylated polypeptide chain with a molecular weight of 28.8 kDa. It consists of 269 amino acids (residues 1-249) and includes a 20 amino acid His-tag at the N-terminus. The protein is purified using proprietary chromatographic techniques.
Physical Appearance
A clear solution, sterile-filtered for purity.
Formulation
The TPI1 protein solution is provided at a concentration of 0.5 mg/ml in a buffer consisting of 20mM Tris-HCl (pH 8.0), 1mM DTT, and 10% glycerol.
Stability
For short-term storage (2-4 weeks), the protein can be stored at 4°C. For extended storage, it is recommended to store the protein frozen at -20°C. Adding a carrier protein such as HSA or BSA (0.1%) is recommended for long-term storage. To maintain protein integrity, avoid repeated freeze-thaw cycles.
Purity
The purity of TPI1 is determined to be greater than 95% using SDS-PAGE analysis.
Biological Activity
The specific activity of TPI1 is greater than 3000 units/mg. One unit is defined as the amount of enzyme required to convert 1.0 µmole of D-glyceraldehyde-3-phosphate to dihydroxyacetone phosphate per minute at a pH of 7.5 and a temperature of 25°C.
Synonyms
TPI, TIM, Triosephosphate Isomerase 1.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MAPSRKFFVG GNWKMNGRKQ SLGELIGTLN AAKVPADTEV VCAPPTAYID FARQKLDPKI AVAAQNCYKV TNGAFTGEIS PGMIKDCGAT WVVLGHSERR HVFGESDELI GQKVAHALAE GLGVIACIGE KLDEREAGIT EKVVFEQTKV IADNVKDWSK VVLAYEPVWA IGTGKTATPQ QAQEVHEKLR GWLKSNVSDA VAQSTRIIYG GSVTGATCKE LASQPDVDGF LVGGASLKPE FVDIINAKQ

Q&A

What is the basic enzymatic function of TPI1 in human cells?

TPI1 (Triosephosphate isomerase 1) functions as a key enzyme in the glycolytic pathway, catalyzing the reversible conversion of dihydroxyacetone phosphate (DHAP) to glyceraldehyde 3-phosphate (G3P). This isomerization reaction is critical for energy production, as it allows both three-carbon products of fructose 1,6-bisphosphate cleavage to continue through the glycolytic pathway toward ATP generation .

Methodologically, researchers investigate TPI1 enzymatic activity by measuring the conversion rate between DHAP and G3P using spectrophotometric assays that track changes in NADH concentration when coupled with glyceraldehyde-3-phosphate dehydrogenase. The enzyme exhibits remarkably high catalytic efficiency, with the reaction being bidirectional, though under physiological conditions, the equilibrium favors DHAP formation. Despite this equilibrium, the glycolytic pathway continues efficiently because G3P is rapidly consumed by subsequent enzymatic reactions.

What structural features are required for TPI1 activation?

TPI1 requires dimerization for full enzymatic activity. For the triosephosphate isomerase 1 enzyme to be turned on (active), it has to attach (bind) to another triosephosphate isomerase 1 enzyme, forming a two-enzyme complex called a dimer . Each TPI1 monomer contains a complete set of catalytic residues arranged in an α/β barrel structure, consisting of 8 exterior alpha helices surrounding 8 interior beta strands .

The importance of dimerization is highlighted in TPI1 deficiency cases, where mutations like Glu104Asp (E104D) destabilize the enzyme and impair its ability to form dimers . This mutation accounts for approximately 80% of triosephosphate isomerase deficiency cases and demonstrates how structural integrity is essential for proper enzymatic function.

Research methodologies to study TPI1 structure include X-ray crystallography to reveal the precise three-dimensional arrangement of the protein, and site-directed mutagenesis to identify crucial residues involved in dimerization and catalysis.

How do post-translational modifications affect TPI1 activity in different cellular contexts?

Post-translational modifications (PTMs) represent a critical regulatory layer for TPI1 function beyond its primary sequence. The most significant modifications include ubiquitination, phosphorylation, and acetylation, each affecting TPI1's stability, localization, and activity differently.

Research shows that in breast cancer cells, TPI1 undergoes ubiquitin-dependent proteasomal degradation mediated by the protein P62/SQSTM1 . This regulation mechanism was demonstrated using ubiquitination assays with MG132 proteasome inhibitor treatment, showing that "P62 promotes ubiquitin-dependent proteasome degradation of TPI1 in MDA-MB-231 cells" .

To study these modifications experimentally, researchers employ mass spectrometry-based proteomics to identify modification sites, site-directed mutagenesis to create non-modifiable variants, and in vitro enzymatic assays with purified modifying enzymes. Understanding these modifications is crucial as they determine TPI1's functional outcomes in different cellular environments and disease states.

How does TPI1 contribute to cancer progression, particularly in breast cancer?

TPI1 has been identified as a potential oncogene in multiple cancers, including breast cancer (BRCA). Studies reveal that "TPI1 promotes BRCA cell glycolysis, proliferation and metastasis in vitro and in vivo" . Mechanistically, TPI1 activates the PI3K/AKT/mTOR signaling pathway, which regulates epithelial-mesenchymal transformation (EMT) and aerobic glycolysis in cancer cells.

This activation occurs through TPI1's interaction with cell division cycle associated 5 (CDCA5) protein. According to research, "TPI1 activates phosphoinositide 3-kinase (PI3K)/AKT/mammalian target of rapamycin (mTOR) pathway to regulate epithelial–mesenchymal transformation (EMT) and aerobic glycolysis, which is positively mediated by cell division cycle associated 5 (CDCA5)" .

Researchers investigating TPI1's role in cancer progression employ multiple methodological approaches, including expression analysis through Western blot and immunohistochemistry, functional assays through knockdown and overexpression studies, pathway analysis, and in vivo tumor models. High TPI1 expression correlates with poor prognosis in breast cancer patients, associating with advanced clinical stages (p=0.002) and reduced survival rates .

What is the molecular mechanism behind TPI1 deficiency and its clinical manifestations?

TPI1 deficiency is a rare genetic disorder caused by mutations in the TPI1 gene. At least 12 mutations have been identified, with the Glu104Asp (E104D) mutation accounting for approximately 80% of cases .

Mechanistically, these mutations, particularly E104D, destabilize the TPI1 enzyme and impair its ability to form functional dimers, which are required for enzymatic activity. As a result, glycolysis is impaired, leading to decreased energy production in cells . The clinical manifestations include:

  • Hemolytic anemia (shortage of red blood cells)

  • Neurological problems, including movement disorders

  • Increased susceptibility to infections

  • Muscle weakness affecting cardiac and respiratory function

Red blood cells are particularly affected because they depend solely on glycolysis for energy production, lacking mitochondria for oxidative phosphorylation . When TPI1 activity is compromised, DHAP accumulates in cells, leading to cellular damage.

Research methodologies to study TPI1 deficiency include genetic sequencing to identify mutations, enzyme activity assays in patient samples, structural biology techniques to understand how mutations affect protein stability, and metabolomic profiling to measure glycolytic intermediates.

How can researchers distinguish between TPI1's enzymatic and non-enzymatic functions in experimental systems?

Distinguishing between enzymatic and non-enzymatic functions of TPI1 requires specialized experimental approaches that can separate its catalytic activity from protein-protein interactions:

  • Catalytically inactive mutants: Engineering TPI1 mutants with substitutions in catalytic residues that preserve protein structure but eliminate enzymatic activity allows researchers to determine which cellular effects depend on catalytic function versus protein-protein interactions.

  • Interactome analysis: Techniques like co-immunoprecipitation (co-IP) followed by mass spectrometry identify TPI1's interaction partners. In breast cancer research, this approach identified TPI1's interaction with CDCA5 and P62 .

  • Domain-specific mutations: Targeting regions of TPI1 involved in protein interactions rather than catalysis helps selectively disrupt non-enzymatic functions.

  • Substrate competition assays: Using competitive inhibitors that block enzymatic activity without affecting protein structure or interactions allows for functional separation.

Research has revealed that TPI1 exhibits non-enzymatic functions in cancer progression through interactions with proteins like CDCA5 and sequestosome-1 (SQSTM1/P62) . These interactions influence signaling pathways independently of TPI1's glycolytic function, suggesting potential therapeutic approaches targeting protein-protein interactions rather than enzymatic activity.

What are the most effective experimental approaches for investigating TPI1's role in glycolysis versus its signaling functions?

Investigating TPI1's dual roles in glycolysis and signaling requires targeted experimental designs that can differentiate between these functions:

  • Glycolytic function assessment:

    • Enzyme activity assays using purified protein or cell lysates

    • Metabolic flux analysis with isotope-labeled glucose

    • Measurement of glycolytic intermediates and end products

    • Extracellular acidification rate (ECAR) analysis

  • Signaling function assessment:

    • Protein-protein interaction studies (co-IP, proximity ligation assays)

    • Phosphoproteomic analysis to identify changes in signaling pathways

    • Use of catalytically inactive TPI1 mutants

    • Expression of TPI1 domains to identify regions involved in protein interactions

In breast cancer research, these approaches were combined by using knockdown and overexpression models to assess both glycolytic capacity and PI3K/AKT/mTOR pathway activation, performing co-IP experiments that identified TPI1's interaction with CDCA5 and P62, and conducting ubiquitination assays to determine how protein stability is regulated . These complementary methods allow researchers to distinguish which cellular phenotypes result from altered glycolysis versus changed signaling pathways.

What techniques are most reliable for measuring TPI1 dimerization and how does this correlate with activity?

Measuring TPI1 dimerization requires specialized biophysical and biochemical techniques that can accurately assess protein oligomeric states:

  • Size exclusion chromatography (SEC): Separates proteins based on molecular weight, allowing quantification of monomeric versus dimeric TPI1.

  • Analytical ultracentrifugation (AUC): Provides precise measurements of protein oligomeric state and binding constants for dimerization.

  • Förster resonance energy transfer (FRET): By tagging TPI1 monomers with donor and acceptor fluorophores, researchers can monitor dimerization in real-time even in cellular contexts.

  • Cross-linking coupled with mass spectrometry: Identifies specific residues involved in the dimerization interface.

Correlation between dimerization and activity is typically assessed by parallel enzyme activity assays. The E104D mutation associated with TPI1 deficiency provides a natural example of this correlation, as this mutation "causes the enzyme to be unstable and impairs its ability to form a dimer and become active" .

Researchers often use site-directed mutagenesis to create variants with altered dimerization capacity, then measure both oligomeric state and catalytic activity to establish direct relationships between structure and function.

What are the current limitations in TPI1 research methodologies and how might they be overcome?

Current limitations in TPI1 research methodologies include several technical and conceptual challenges:

  • Difficulty distinguishing glycolytic from non-glycolytic functions:

    • Challenge: TPI1 modification affects both glycolysis and signaling simultaneously

    • Solution: Development of specific inhibitors that target protein-protein interactions without affecting catalytic activity; use of domain-specific mutations

  • Limited spatial and temporal resolution:

    • Challenge: Traditional biochemical assays provide population averages rather than single-cell dynamics

    • Solution: Implementation of live-cell imaging with fluorescent TPI1 reporters; development of sensors for real-time monitoring of TPI1 activity

  • Complexity of in vivo models:

    • Challenge: Systemic effects of TPI1 modification complicate interpretation

    • Solution: Tissue-specific and inducible genetic models; development of more physiologically relevant 3D culture systems

  • Translational barriers:

    • Challenge: Moving from mechanistic understanding to therapeutic applications

    • Solution: High-throughput screening for selective TPI1 modulators; structure-based drug design targeting specific TPI1 interactions

Advances in CRISPR-based technologies, single-cell omics, and computational modeling will likely play crucial roles in overcoming these limitations and advancing TPI1 research.

What is the significance of TPI1 subcellular localization for its various cellular functions?

While traditionally considered a cytosolic enzyme, research has revealed that TPI1 exhibits dynamic subcellular localization with functional implications:

  • Nuclear localization: TPI1 can translocate to the nucleus in certain cellular contexts. The search results indicate that in breast cancer cells, TPI1 was "expressed in cytoplasmic and nucleus of tumor cells" . This nuclear localization may involve roles in transcriptional regulation or nuclear metabolism.

  • Membrane association: TPI1 may associate with plasma membranes or organelle membranes in metabolically active cells, facilitating localized ATP production for energy-demanding processes.

  • Mitochondrial interaction: Despite glycolysis being a cytosolic pathway, TPI1 has been found to interact with mitochondrial proteins, suggesting a role in coordinating glycolytic and oxidative metabolism.

Research methodologies to study TPI1 localization include immunofluorescence microscopy, subcellular fractionation, proximity labeling techniques, and live-cell imaging with fluorescently tagged TPI1. Understanding the functional significance of diverse localizations requires correlation with local metabolic requirements, signaling environments, and protein interaction networks specific to each compartment.

What is the relationship between TPI1 and the Warburg effect in cancer cells?

The Warburg effect—cancer cells' preference for glycolysis even in the presence of oxygen—appears to be intimately connected to TPI1 function based on current research:

  • TPI1 overexpression in cancers: TPI1 is "highly expressed in BRCA tissue and cell lines" and has been "detected in several types of cancers, such as intrahepatic cholangiocarcinoma (ICC), gastric, lung, and prostate cancer" . This overexpression enhances glycolytic flux.

  • Enhanced glycolysis: TPI1 promotes "BRCA cell glycolysis, proliferation and metastasis in vitro and in vivo" , directly contributing to the Warburg phenotype.

  • Signaling integration: TPI1 "activates the PI3K/AKT/mTOR pathway to regulate epithelial–mesenchymal transformation (EMT) and aerobic glycolysis" , linking metabolic reprogramming to oncogenic signaling.

Research methodologies to investigate this relationship include metabolic flux analysis using isotope-labeled glucose, measurement of extracellular acidification rate (ECAR), analysis of glycolytic enzyme expression patterns in tumor samples, and determination of lactate production and glucose consumption rates.

Therapeutically, targeting TPI1 could potentially disrupt the Warburg effect, as research suggests that "targeting glycolytic multienzyme system and related pathways are hypothesized be a novel therapeutic strategy in BRCA" .

How is TPI1 regulated at the transcriptional and post-transcriptional levels in normal versus disease states?

Understanding the multi-layered regulation of TPI1 expression requires investigation of both transcriptional and post-transcriptional mechanisms:

  • Transcriptional regulation:

    • Promoter analysis to identify transcription factor binding sites

    • ChIP-seq to determine transcription factor occupancy at the TPI1 locus

    • Reporter assays to measure promoter activity under various conditions

    • Analysis of epigenetic modifications (DNA methylation, histone modifications)

  • Post-transcriptional regulation:

    • mRNA stability assessment through actinomycin D chase experiments

    • Identification of regulatory RNA-binding proteins through RNA immunoprecipitation

    • Investigation of microRNA-mediated regulation

    • Analysis of alternative splicing patterns

In breast cancer, TPI1 expression is significantly upregulated compared to normal tissues. Research shows that "TPI1 in primary BRCA was significantly higher than that in normal breast tissue" and "TPI1 was significantly upregulated in 10 pairs of BRCA samples compared with the matched adjacent normal tissues" .

The regulatory mechanisms behind this upregulation may involve oncogenic transcription factors, altered mRNA stability, or changes in microRNA expression profiles. Understanding these regulatory mechanisms could identify potential therapeutic targets for modulating TPI1 expression in disease states.

What controls are essential when designing experiments to study TPI1 in cancer models?

Rigorous experimental design for TPI1 cancer research requires multiple levels of controls to ensure reliable and interpretable results:

  • Genetic manipulation controls:

    • Empty vector controls for overexpression studies

    • Non-targeting shRNA/siRNA controls for knockdown studies

    • Rescue experiments with wild-type TPI1 to confirm specificity

    • Catalytically inactive TPI1 mutants to distinguish enzymatic from structural roles

  • Cell line considerations:

    • Matched normal and cancer cell lines (e.g., MCF-10A vs. breast cancer lines)

    • Panel of cancer cell lines with varying endogenous TPI1 expression

  • In vivo model controls:

    • The breast cancer study used several control groups: "The mice were randomly divided into three groups (n = 5/group). Two groups of mice were subcutaneously injected with MDA-MB-231 cells overexpressing TPI1-luciferase, while the remaining group was injected with MDA-MB-231-luciferase vector control cells" .

    • Additional controls included a drug treatment group: "Five mice from TPI1 overexpressing group were randomly selected to receive LY294002 (75 mg/kg) intragastric administration 3 times a week for 3 weeks" .

  • Pathway analysis controls:

    • Pathway inhibitors (like LY294002 for PI3K inhibition)

    • Monitoring of multiple pathway components to confirm consistent effects

  • Technical controls:

    • Loading controls for Western blots

    • Housekeeping genes for qPCR

    • Appropriate staining controls for IHC and IF

The implementation of these controls ensures that observed effects can be specifically attributed to TPI1 modulation rather than experimental artifacts or off-target effects.

How should researchers approach contradictory findings regarding TPI1's role in different cancer types?

The search results highlight contradictory findings regarding TPI1's role in cancer: while it appears oncogenic in most cancers studied, "in HCC, TPI1 was identified as a tumor suppressor gene" . Addressing such contradictions requires systematic approaches:

  • Context-dependent analysis:

    • Compare experimental conditions, cell types, and model systems used in contradictory studies

    • Investigate tissue-specific factors that might influence TPI1 function

    • Examine genetic background differences that could modify TPI1 effects

  • Mechanistic reconciliation:

    • Determine if contradictory findings involve different molecular mechanisms

    • Investigate whether TPI1 engages different signaling partners in different contexts

    • Consider whether post-translational modifications differ between systems

  • Technical validation:

    • Replicate key experiments from contradictory studies using standardized protocols

    • Perform side-by-side comparisons in multiple cell lines

    • Employ complementary methodologies to verify findings

  • Integrated analysis:

    • Conduct meta-analysis of existing datasets (e.g., TCGA, METABRIC)

    • Design experiments that directly test hypotheses explaining contradictions

What methodological approaches are most effective for translating basic TPI1 research findings into potential therapeutic applications?

Translating basic TPI1 research into therapeutic applications requires methodological approaches that bridge fundamental science and clinical development:

  • Target validation strategies:

    • Genetic validation using CRISPR-Cas9 in relevant cell lines and animal models

    • Pharmacological validation with available tool compounds

    • Clinical correlation studies analyzing TPI1 expression/activity in patient samples

  • Drug discovery approaches:

    • Structure-based drug design targeting TPI1's catalytic site or protein interaction domains

    • High-throughput screening of compound libraries against TPI1

    • Fragment-based approaches to identify initial chemical matter

    • Computational approaches including virtual screening and molecular dynamics simulations

  • Therapeutic modality selection:

    • Small molecule inhibitors for enzymatic function

    • Protein-protein interaction disruptors for non-enzymatic functions

    • RNA-based therapeutics for expression modulation

    • PROTAC approaches for targeted degradation

  • Preclinical development:

    • In vitro efficacy in panel of cancer cell lines

    • In vivo efficacy in xenograft and genetic mouse models

    • Toxicity assessment in normal cells and tissues

    • Pharmacokinetic and pharmacodynamic studies

The breast cancer study suggests potential therapeutic applications: "TPI1 promotes tumor development and progression, which may serve as a therapeutic target for BRCA" , and demonstrates efficacy of targeting the PI3K/AKT/mTOR pathway downstream of TPI1 using LY294002 in mouse models.

Product Science Overview

Structure and Function

TPI1 is a homodimeric enzyme, meaning it consists of two identical subunits. Each subunit contains 247 amino acids and forms a structure known as a TIM barrel, characterized by eight alpha helices surrounding eight beta strands . This structure is essential for the enzyme’s catalytic activity, which involves the transfer of a hydrogen atom from carbon 1 to carbon 2, facilitating the isomerization of a ketose to an aldose .

The enzyme’s active site is located in the lower loop regions of the TIM barrel, where it binds to the phosphate group of the substrate. TPI1’s efficiency is remarkable, providing a rate enhancement of 10^9 times compared to the nonenzymatic reaction .

Recombinant Human TPI1

Recombinant human TPI1 is produced using Escherichia coli expression systems, ensuring high purity and biological activity. This recombinant protein is often tagged with a His tag at the N-terminus to facilitate purification . The recombinant form retains the full-length sequence of the human enzyme, making it suitable for various applications, including SDS-PAGE, functional assays, and mass spectrometry .

Biological Significance

TPI1 is vital for cellular energy production. By catalyzing the interconversion of DHAP and G3P, it ensures a continuous supply of intermediates for ATP generation in glycolysis. Additionally, TPI1 is involved in other metabolic pathways, such as the pentose phosphate shunt and fatty acid biosynthesis .

Mutations in the TPI1 gene can lead to triosephosphate isomerase deficiency, a rare genetic disorder characterized by hemolytic anemia and neurological dysfunction . This highlights the enzyme’s importance in maintaining normal cellular function.

Applications and Research

Recombinant human TPI1 is widely used in research to study its structure, function, and role in metabolic diseases. It is also employed in high-throughput screening assays to identify potential inhibitors or activators that could modulate its activity for therapeutic purposes .

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.