IMPAD1 Mouse

Inositol Monophosphatase Domain Containing 1 Mouse Recombinant
Shipped with Ice Packs
In Stock

Description

Introduction to IMPAD1 in Mice

IMPAD1 is a Golgi-resident phosphatase that hydrolyzes phosphoadenosine phosphate (PAP) to adenosine monophosphate (AMP), preventing PAP accumulation, which inhibits sulfotransferases and disrupts glycosaminoglycan sulfation . In mice, Impad1 inactivation leads to skeletal dysplasia and joint abnormalities, mirroring human chondrodysplasia .

Molecular Functions and Biochemical Role

IMPAD1 exhibits 3'-nucleotidase activity, specifically targeting PAP . Key functions include:

  • Golgi Dynamics: Regulates vesicular trafficking and Golgi morphology via interaction with Syt11, influencing extracellular matrix (ECM) remodeling .

  • Metabolic Regulation: Inhibits mitochondrial Complex I, reducing reactive oxygen species (ROS) and activating AMPK-Notch1-HEY1 signaling to promote metastasis .

Table 1: Biochemical Properties of Mouse IMPAD1

PropertyDetailsSource
Substrate SpecificityHydrolyzes PAP to AMP; no activity toward inositol phosphates
Cellular LocalizationGolgi apparatus
Key InteractionsSyt11 (vesicular trafficking), mitochondrial Complex I

Role in Skeletal Development and Chondrodysplasia

Impad1 knockout mice exhibit severe chondrodysplasia, including:

  • Delayed Chondrocyte Maturation: Disorganized growth plates and ectopic interphalangeal joints .

  • Impaired Proteoglycan Sulfation: Reduced chondroitin sulfate levels, critical for cartilage integrity .

Table 2: Phenotypic Features of Impad1 Knockout Mice

PhenotypeMechanism/ConsequenceSource
Ectopic Joint FormationAberrant ECM sulfation due to PAP accumulation
Short Limb/TrunkDisrupted endochondral ossification

IMPAD1 in Cancer Metastasis

IMPAD1 drives lung cancer metastasis through:

  • EMT Regulation: Repressed by miR-200/miR-96 during epithelial-to-mesenchymal transition (EMT), enhancing invasion .

  • Secretome Modulation: Alters Golgi-mediated exocytosis to create a pro-metastatic tumor microenvironment .

  • AMPK Activation: AMP accumulation from PAP hydrolysis activates AMPK-Notch1-HEY1 signaling, promoting metastasis .

Research Tools: Recombinant Mouse IMPAD1 Protein

Recombinant mouse IMPAD1 (Catalog #7028-PD) is used to study its enzymatic activity :

  • Specifications: 6-His tagged, carrier-free formulation for assays requiring minimal interference .

  • Functional Assays: Used with malachite green phosphate detection kits to quantify PAP phosphatase activity .

Genetic Models and Phenotypic Findings

Key murine models include:

  • Gene-Trap Alleles: Exhibit "joints abnormal with splitting" (JAWS) phenotypes, validating IMPAD1’s role in skeletal development .

  • Syngeneic Tumor Models: Demonstrates IMPAD1’s immunosuppressive effects, reducing CD4+/CD8+ T cells in the tumor microenvironment .

Clinical Implications and Future Directions

IMPAD1’s dual role in development and cancer highlights its therapeutic potential:

  • Targeting AMPK-HEY1: Could mitigate metastasis in IMPAD1-overexpressing lung cancers .

  • Chondrodysplasia Therapies: Enzyme replacement strategies to restore PAP clearance in skeletal disorders .

Product Specs

Introduction
Inositol monophosphatase 3 (IMPAD1), a member of the inositol monophosphatase family, is localized in the Golgi apparatus. This enzyme plays a crucial role in catalyzing the conversion of phosphoadenosine phosphate (PAP) to adenosine monophosphate (AMP). Mutations in the IMPAD1 gene are associated with GRAPP type chondrodysplasia, a condition characterized by joint dislocations. Additionally, a pseudogene of IMPAD1 is present on chromosome 1's long arm.
Description
Produced in Sf9 Insect cells, IMPAD1 is a single, glycosylated polypeptide chain comprising 332 amino acids (34-356aa). It has a molecular mass of 36.2kDa, although on SDS-PAGE, it appears between 28-40kDa. The protein is expressed with a C-terminal 6-His tag and purified using proprietary chromatographic techniques.
Physical Appearance
The product is a sterile, colorless solution that has been filtered.
Formulation
The IMPAD1 protein solution is provided at a concentration of 0.5mg/ml in Phosphate Buffered Saline (pH 7.4) containing 10% glycerol.
Stability
For short-term storage (up to 4 weeks), the product can be stored at 4°C. For extended periods, it is recommended to store the protein frozen at -20°C. To ensure optimal stability during long-term storage, consider adding a carrier protein (0.1% HSA or BSA). Avoid repeated freeze-thaw cycles.
Purity
The purity of IMPAD1 is greater than 95.0% as determined by SDS-PAGE analysis.
Synonyms
Inositol monophosphatase 3, IMP 3, IMPase 3, Golgi 3-prime phosphoadenosine 5-prime phosphate 3-prime phosphatase, Golgi-resident PAP phosphatase, gPAPP, Inositol monophosphatase domain-containing protein 1, Inositol-1(or 4)-monophosphatase 3 Myo-inositol monophosphatase A3, Impad1, Impa3.
Source
Sf9 Insect cells.
Amino Acid Sequence
ADPGRFSLFG LGSEPAAGEA EVASDGGTVD LREMLAVAVL AAERGGDEVR RVRESNVLHE KSKGKTREGA DDKMTSGDVL SNRKMFYLLK TAFPNVQINT EEHVDASDKE VIVWNRKIPE DILKEIAAPK EVPAESVTVW IDPLDATQEY TEDLRKYVTT MVCVAVNGKP VLGVIHKPFS EYTAWAMVDG GSNVKARSSY NEKTPKIIVS RSHAGMVKQV ALQTFGNQTS IIPAGGAGYK VLALLDVPDM TQEKADLYIH VTYIKKWDIC AGNAILKALG GHMTTLNGEE ISYTGSDGIE GGLLASIRMN HQALVRKLPD LEKSGHHHHH HH.

Q&A

What is IMPAD1 and what is its primary function in mice?

IMPAD1 (Inositol Monophosphatase Domain-containing protein 1) is a Golgi-resident nucleotide phosphatase that hydrolyzes phosphoadenosine phosphate (PAP), which is a byproduct of sulfotransferase reactions, to produce AMP . In mouse models, IMPAD1 serves two primary functions depending on cellular context:

  • In developmental contexts: It plays a critical role in skeletal and joint development through regulation of proteoglycan sulfation pathways . It removes PAP after sulfate transfer from PAPS (3'-phosphoadenosine-5'-phosphosulfate) in the Golgi, preventing product inhibition of sulfotransferase reactions .

  • In cancer contexts: IMPAD1 functions as a mitochondrial electron transport inhibitor, specifically targeting Complex I activity, which reduces mitochondrial ROS levels and activates the AMPK-Notch1-HEY1 signaling pathway .

The protein has been detected in various mouse tissues, with notable expression in neuroblastoma cell lines (Neuro-2A) and mesenchymal stem cells differentiated into chondrocytes .

What phenotypes are observed in IMPAD1-deficient mice?

IMPAD1-deficient mice display a constellation of skeletal and developmental abnormalities that include:

  • Short limbs and trunk phenotype, classified as chondrodysplasia

  • Ectopic interphalangeal joints, leading to the designation of the phenotype as "JAWS" for "joints abnormal with splitting"

  • Delayed and disorganized maturation of growth-plate chondrocytes

  • Impaired sulfation of chondroitin and abnormal metabolism of chondroitin sulfate proteoglycan aggrecan

  • Marked reduction in sulfation of both chondroitin and heparan proteoglycans

These phenotypic manifestations bear similarities to human disorders associated with disturbed proteoglycan synthesis and sulfation, such as diastrophic dysplasia, recessive Larsen syndrome, and Desbuquois dysplasia .

How does IMPAD1 influence proteoglycan sulfation pathways?

IMPAD1 plays a crucial role in maintaining efficient proteoglycan sulfation through its enzymatic activity as a phosphoadenosine phosphate (PAP) phosphatase in the Golgi apparatus . The mechanistic pathway involves:

  • During proteoglycan synthesis, sulfotransferases use PAPS (3'-phosphoadenosine-5'-phosphosulfate) as a sulfate donor to add sulfate groups to nascent proteoglycan chains

  • This reaction produces PAP as a byproduct, which can inhibit sulfotransferase activity if it accumulates

  • IMPAD1 hydrolyzes PAP to AMP and inorganic phosphate, preventing product inhibition of sulfotransferase reactions

  • This hydrolysis may also facilitate the reverse transport of AMP from the Golgi to the cytoplasm

When IMPAD1 function is compromised, the accumulation of PAP inhibits proper sulfation of proteoglycans, particularly chondroitin and heparan sulfate proteoglycans, leading to skeletal and joint developmental abnormalities .

What experimental approaches are most effective for studying IMPAD1 function in mouse models?

For comprehensive analysis of IMPAD1 function in mouse models, researchers should consider these methodological approaches:

  • Genetic manipulation techniques:

    • Gene-trap alleles as used by Mitchell et al. and Sohaskey et al. to study skeletal phenotypes

    • CRISPR/Cas9 for precise genetic modifications with tissue-specific promoters

  • Phenotypic characterization:

    • Radiographic examination to assess skeletal abnormalities, particularly metacarpals, phalanges, and vertebral irregularities

    • Joint histology to evaluate ectopic joint formation and chondrocyte organization

  • Biochemical assays:

    • In vitro enzymatic assays to measure PAP phosphatase activity as performed by Frederick et al.

    • Analysis of proteoglycan sulfation using metabolic labeling with radioactive sulfate

  • Cellular localization studies:

    • Immunofluorescence using specific antibodies (such as Anti-Mouse/Rat IMPAD1 Antibody) to determine subcellular localization

    • Western blot analysis for protein expression quantification in various tissues

  • Functional studies in tissue contexts:

    • Mesenchymal stem cell differentiation into chondrocytes to study IMPAD1's role in cartilage development

    • Analysis of growth plate development using histomorphometric techniques

These methodologies should be selected based on the specific aspect of IMPAD1 biology being investigated and can be combined for more comprehensive understanding.

How does IMPAD1 regulate cancer progression in mouse models?

IMPAD1 contributes to cancer progression through multiple mechanisms that can be studied in mouse models:

  • Enhancement of cellular invasion and metastasis:

    • IMPAD1 significantly increases invasion (32-fold increase) in transwell assays

    • It promotes cellular motility in scratch assays and invasive spheroid formation in 3D collagen/Matrigel matrices

    • In vivo studies show IMPAD1 overexpression results in increased metastasis without affecting primary tumor volume

  • Golgi-mediated secretion pathway:

    • IMPAD1 enhances Golgi-mediated secretion of matrix metalloproteases (MMPs), particularly MMPs 1a, 2, and 9

    • Treatment with the pan-MMP inhibitor Ilomastat suppresses IMPAD1-mediated invasion

  • Mitochondrial function modulation:

    • IMPAD1 inhibits mitochondrial Complex I activity, causing mitochondrial dysfunction

    • This inhibition reduces mitochondrial ROS levels

    • Accumulation of AMP increases phosphorylated AMPK (pAMPK), leading to Notch1 and HEY1 upregulation

    • The AMPK-Notch1-HEY1 signaling pathway activation promotes metastatic potential

Experimental validation through IMPAD1 knockdown shows reduced metastatic ability without affecting tumor growth, confirming its specific role in metastasis rather than proliferation .

What techniques are optimal for analyzing proteoglycan sulfation in IMPAD1-deficient mice?

To effectively analyze proteoglycan sulfation defects in IMPAD1-deficient mice, researchers should employ these specialized techniques:

  • Metabolic labeling:

    • Incorporate [35S]sulfate into newly synthesized proteoglycans

    • Analyze incorporation rates in different tissues and developmental stages

    • Compare wild-type and IMPAD1-deficient samples to quantify sulfation reduction

  • Biochemical characterization:

    • Isolate proteoglycans from cartilage and other tissues using guanidine extraction

    • Perform ion-exchange chromatography to separate based on charge density (reflective of sulfation levels)

    • Enzymatic digestion with chondroitinase ABC and heparinase to analyze specific glycosaminoglycan chains

  • Mass spectrometry:

    • Analyze the disaccharide composition after specific enzymatic digestion

    • Quantify the position and degree of sulfation on glycosaminoglycan chains

    • Compare sulfation patterns between wild-type and IMPAD1-deficient samples

  • Histological methods:

    • Use sulfation-specific stains like Alcian blue at different pH values

    • Apply immunohistochemistry with antibodies recognizing specific sulfation patterns

    • Analyze tissue-specific differences in proteoglycan composition and distribution

  • Functional assays:

    • Assess growth factor binding capabilities of isolated proteoglycans

    • Measure mechanical properties of cartilage to correlate with sulfation defects

    • Evaluate cellular responses to undersulfated proteoglycans

These techniques provide complementary information about the nature and extent of sulfation defects caused by IMPAD1 deficiency, enabling comprehensive characterization of the molecular mechanisms underlying the observed phenotypes.

How should researchers design experiments to distinguish IMPAD1's developmental versus cancer-related functions?

When investigating the dual roles of IMPAD1 in development and cancer, researchers should implement the following experimental design strategies:

  • Temporal control systems:

    • Use doxycycline-inducible expression systems as demonstrated in previous research

    • Implement conditional knockout models with tissue-specific Cre recombinase expression

    • These approaches allow separation of developmental effects from adult/cancer phenotypes

  • Tissue-specific models:

    • Generate tissue-specific IMPAD1 knockout or overexpression models using:

      • Chondrocyte-specific promoters (Col2a1-Cre) for developmental studies

      • Lung epithelial promoters (SPC-Cre) for cancer studies

    • Compare phenotypes across different tissue contexts

  • Pathway-specific analysis:

    • For developmental studies: Focus on proteoglycan sulfation pathways and PAP metabolism

    • For cancer studies: Concentrate on AMPK-Notch1-HEY1 signaling and MMP secretion

    • Use pathway inhibitors to distinguish mechanisms (e.g., MMP inhibitor Ilomastat for cancer studies)

  • Comparative molecular profiling:

    • Perform RNA-seq and proteomics on developmental tissues versus cancer models

    • Identify context-specific binding partners and effectors

    • Map transcriptional networks in different cellular contexts

  • Rescue experiments:

    • Test whether specific enzymatic functions can rescue developmental defects versus cancer phenotypes

    • Evaluate domain-specific mutations that selectively impair either developmental or cancer-related functions

This multifaceted approach will help delineate the mechanistic distinctions between IMPAD1's roles in development and cancer progression.

What are the challenges in reconciling contradictory data from different IMPAD1 mouse models?

Researchers studying IMPAD1 mouse models face several challenges when interpreting potentially contradictory results:

  • Genetic background variations:

    • Different mouse strains may exhibit variable penetrance and expressivity of IMPAD1-related phenotypes

    • Solution: Backcross models to consistent genetic backgrounds and document strain-specific differences

  • Model generation methodology differences:

    • Gene-trap versus knockout approaches may result in different levels of gene inactivation

    • Some models may retain partial IMPAD1 function or express truncated proteins

    • Solution: Characterize protein expression and residual enzymatic activity in each model

  • Developmental timing considerations:

    • Embryonic versus postnatal phenotypes may differ significantly

    • Solution: Implement time-course analyses across developmental stages

  • Context-dependent functions:

    • IMPAD1 exhibits different functions in skeletal development versus cancer progression

    • Solution: Clearly define cellular context and avoid generalizing findings across tissues

  • Technical variability in phenotypic assessment:

    • Different methods for evaluating joint formation, chondrocyte maturation, or metastasis

    • Solution: Standardize analytical techniques and include multiple methodological approaches

  • Compensatory mechanisms:

    • Long-term IMPAD1 deficiency may trigger alternative pathways that mask or modify primary phenotypes

    • Solution: Use acute inactivation systems and analyze early consequences of IMPAD1 loss

When confronted with contradictory data, researchers should evaluate methodological differences, contextual factors, and potential technical artifacts before concluding genuine biological differences exist between models.

What detection methods are most reliable for IMPAD1 protein analysis in mouse tissues?

For optimal detection of IMPAD1 protein in mouse tissues, researchers should consider these validated methodological approaches:

  • Western blot analysis:

    • Recommended antibodies: Sheep Anti-Mouse Inositol Monophosphatase 3/IMPAD1 Antigen Affinity-purified Polyclonal Antibody has been validated in mouse neuroblastoma (Neuro-2A) cell lines

    • Expected molecular weight: Approximately 42 kDa under reducing conditions

    • Optimal buffer systems: Immunoblot Buffer Group 1 has proven effective

    • Secondary antibody: HRP-conjugated Anti-Sheep IgG Secondary Antibody provides specific detection

  • Immunofluorescence microscopy:

    • Tissue preparation: Immersion fixation shows good preservation of IMPAD1 localization

    • Antibody concentration: 10 μg/mL with 3-hour room temperature incubation has shown reliable results

    • Secondary detection: NorthernLights™ 557-conjugated Anti-Sheep IgG Secondary Antibody provides strong signal

    • Counterstaining: DAPI for nuclear visualization complements cytoplasmic IMPAD1 staining

    • Cellular localization: Expect cytoplasmic staining pattern, with potential Golgi enrichment

  • Flow cytometry:

    • Cell preparation: Single-cell suspensions from relevant tissues (bone, cartilage, lung)

    • Fixation/permeabilization: Required for intracellular IMPAD1 detection

    • Controls: Include isotype controls and IMPAD1-knockout tissues for specificity validation

  • Immunohistochemistry:

    • Tissue sections: Paraffin-embedded or frozen sections from developmental tissues

    • Antigen retrieval: May be necessary for optimal detection

    • Visualization: DAB or fluorescence-based detection systems

When analyzing IMPAD1 expression, it is critical to include appropriate positive controls (such as mesenchymal stem cells differentiated into chondrocytes) and negative controls (IMPAD1-deficient tissues) to ensure specificity and reliability of the detection methods.

How can researchers effectively evaluate the impact of IMPAD1 on cancer metastasis in vivo?

For comprehensive evaluation of IMPAD1's role in cancer metastasis using mouse models, researchers should implement this methodological framework:

  • Experimental model selection:

    • Syngeneic models: WT 129/Sv mice with implanted 344SQ cells (either overexpressing IMPAD1 or with IMPAD1 knockdown)

    • Orthotopic models: Direct implantation into lung tissue for lung cancer studies

    • Genetic models: KRAS LA1/+;p53 R172HΔG/+ (KP) mice with IMPAD1 modifications

  • Cell line engineering:

    • Stable overexpression: Constitutive or doxycycline-inducible IMPAD1 expression systems

    • Knockdown approaches: Validated shRNA constructs targeting IMPAD1

    • Fluorescent tagging: GFP-labeled cells for tracking purposes

  • Implantation techniques:

    • Subcutaneous implantation: Allows for easy monitoring of primary tumor growth

    • Tail vein injection: Evaluates later stages of metastatic cascade

    • Intracardiac injection: Assesses systemic dissemination

  • Metastasis quantification methods:

    • Macroscopic lung nodule counting: Visual inspection and enumeration of surface metastases

    • Histological analysis: H&E staining of lung sections to confirm and quantify metastatic burden

    • Molecular quantification: qPCR-based methods to detect cancer cell-specific markers in target organs

    • In vivo imaging: Bioluminescence or fluorescence imaging for longitudinal tracking

  • Molecular validation:

    • Confirm IMPAD1 expression levels in primary tumors via RT-qPCR and Western blotting

    • Assess MMP expression patterns in tumor samples

    • Evaluate AMPK-Notch1-HEY1 pathway activation status

  • Therapeutic intervention studies:

    • MMP inhibitor treatment: Ilomastat administration to block IMPAD1-mediated invasion

    • ADORA1 inhibitor application: To modulate AMP-related signaling pathways

    • Combination approaches: Test synergistic effects with standard chemotherapeutics

This systematic approach allows for robust assessment of IMPAD1's contribution to metastatic progression while distinguishing effects on invasion from effects on primary tumor growth.

What are promising therapeutic targets in the IMPAD1 signaling pathway for skeletal disorders?

Based on current understanding of IMPAD1 function in skeletal development, several promising therapeutic targets emerge for skeletal disorders associated with proteoglycan sulfation defects:

  • PAP metabolism modulation:

    • Development of small molecules that enhance PAP clearance from the Golgi

    • Design of PAP-resistant sulfotransferases to maintain activity despite PAP accumulation

    • Targeted delivery of PAP phosphatase activity to chondrocytes

  • Proteoglycan sulfation enhancement:

    • PAPS synthetase activators to increase sulfate donor availability

    • Sulfotransferase overexpression or activation in cartilage tissue

    • Engineered proteoglycans with pre-sulfated glycosaminoglycan chains

  • Downstream signaling pathway intervention:

    • Modulation of growth factor signaling pathways affected by undersulfated proteoglycans

    • Targeting of chondrocyte differentiation pathways to correct maturation defects

    • Regulation of ECM organization to compensate for proteoglycan abnormalities

  • Cell-based therapies:

    • Gene-corrected mesenchymal stem cells for cartilage regeneration

    • IMPAD1-overexpressing chondrocyte transplantation

    • Engineered exosomes delivering functional IMPAD1 or downstream effectors

  • Combination approaches:

    • Pairing sulfation enhancement with joint preservation strategies

    • Targeting both developmental defects and inflammatory consequences

    • Personalized approaches based on specific IMPAD1 mutations

These therapeutic avenues require further validation in animal models before clinical translation, but they represent rational targets based on our understanding of IMPAD1's role in skeletal development and proteoglycan biology.

How might IMPAD1 mouse models inform our understanding of human skeletal disorders?

IMPAD1 mouse models provide valuable insights that can be translated to human skeletal disorders through multiple approaches:

  • Genotype-phenotype correlations:

    • Comparison of mouse phenotypes with human chondrodysplasias associated with IMPAD1 mutations

    • Identification of modifier genes that influence phenotypic severity across species

    • Creation of humanized IMPAD1 mouse models carrying patient-specific mutations

  • Developmental trajectory analysis:

    • Detailed characterization of joint formation abnormalities in mice as models for human joint disorders

    • Timeline comparison of skeletal element ossification between mouse models and human patients

    • Identification of critical developmental windows for therapeutic intervention

  • Therapeutic testing platform:

    • Evaluation of proteoglycan sulfation enhancement strategies in mouse models before human application

    • Preclinical testing of drugs targeting PAP metabolism or downstream pathways

    • Development of biomarkers for monitoring treatment efficacy

  • Comparative molecular pathology:

    • Multi-omics analysis of affected tissues in both mouse models and human patient samples

    • Identification of conserved and divergent disease mechanisms between species

    • Discovery of compensatory pathways that might explain phenotypic differences

  • Translational research applications:

    • Development of diagnostic criteria based on molecular fingerprints identified in mouse models

    • Design of minimally invasive monitoring techniques for disease progression

    • Personalized medicine approaches based on specific IMPAD1 variants

By systematically comparing IMPAD1-deficient mouse phenotypes with human skeletal disorders, researchers can identify conserved disease mechanisms and develop targeted therapeutic strategies with higher translational potential.

Product Science Overview

Gene and Protein Structure

The IMPAD1 gene is located on chromosome 8q12.1 in humans . The mouse IMPAD1 protein has a short N-terminal tail, a transmembrane domain, and an N-glycosylation site . The recombinant mouse IMPAD1 protein is often produced in Chinese Hamster Ovary (CHO) cells and includes an N-terminal 6-His tag for purification purposes .

Function and Activity

IMPAD1 is a PAP-specific phosphatase that removes the 3’-phosphate from PAP, which is cytotoxic, to form non-toxic AMP . This activity is crucial for the regulation of sulfur metabolism, as PAP can inhibit many sulfotransferases . The recombinant mouse IMPAD1 protein has been shown to have robust 3’-nucleotidase activity towards PAP, and its activity can be inhibited by lithium in a noncompetitive manner .

Biological Significance

IMPAD1 is expressed in various tissues, including the brain, spinal cord, lung, kidney, and costal cartilage . It is particularly important in cartilage development, as evidenced by studies on Gpapp -/- mice (the mouse equivalent of IMPAD1). These mice exhibit severe respiratory distress, dwarfism, and abnormal cartilage morphology, highlighting the protein’s role in skeletal development .

Clinical Relevance

Mutations in the IMPAD1 gene have been associated with a condition characterized by short stature, chondrodysplasia with brachydactyly, congenital joint dislocations, micrognathia, cleft palate, and facial dysmorphism . This condition is inherited in an autosomal recessive manner, and several loss-of-function mutations have been identified in affected individuals .

Research and Applications

Recombinant mouse IMPAD1 protein is used in various research applications, including enzyme activity assays and studies on sulfur metabolism . The protein is available in both carrier-free and BSA-containing formulations, depending on the specific requirements of the experimental setup .

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.