PHOSPHO1 Human

Phosphatase Orphan-1 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Biochemical Characteristics

Catalytic Properties

SubstrateSpecific Activity (units/mg)
Phosphoethanolamine (PEA)4600 ± 582
Phosphocholine (PCho)2980 ± 335
Ribose 5-phosphate74.8 ± 6.2
p-Nitrophenyl phosphate64.5 ± 36.6
No activity detected for pyrophosphate, ATP, or phospho-L-serine .
  • Optimal activity at pH 6.7, with Mg²⁺ as a cofactor (Kₘ = 3.0 μM for PEA; 11.4 μM for PCho) .

  • Asp32, Asp34, Asp43, and Asp123 form the catalytic core .

Skeletal Mineralization

  • Generates inorganic phosphate (Pᵢ) via hydrolysis of PEA and PCho, enabling hydroxyapatite crystallization in bone and cartilage .

  • Localized to matrix vesicles in osteoblasts and chondrocytes .

Metabolic Regulation

  • Brown Adipose Tissue (BAT):

    • Depletion increases thermogenesis, mitochondrial biogenesis (UCP1, PGC1α), and cold tolerance .

    • Knockout (KO) mice resist diet-induced obesity and insulin resistance .

  • Phospholipid Homeostasis:

    • Regulates phosphatidylcholine (PC) and phosphatidylethanolamine (PE) levels, impacting membrane structure and lipid droplet dynamics .

Tissue Expression and Genetic Associations

  • High Expression: Bone, cartilage, and BAT .

  • Genetic Variants:

    • SNPs linked to obesity, type 2 diabetes, and cardiovascular disease .

    • DNA methylation at PHOSPHO1 loci correlates with HDL cholesterol and diabetes risk .

Therapeutic Implications

  • PHOSPHO1 Inhibition:

    • Improves glucose tolerance and reduces hepatic steatosis in murine models .

    • Phosphocholine supplementation mimics KO phenotypes, enhancing energy expenditure .

  • Safety: No adverse effects on adult bone mineralization or erythropoiesis observed .

Research Gaps and Future Directions

  • Mechanisms linking PHOSPHO1 to mitochondrial function remain unclear .

  • Tissue-specific KO models are needed to dissect BAT vs. systemic effects .

  • Development of PHOSPHO1-resistant PC analogs could enhance therapeutic utility .

Product Specs

Introduction
PHOSPHO1, involved in the mineralization process, plays a crucial role in bone and cartilage matrix mineralization. Its expression is evident at mineralization sites within these tissues, with high levels observed in the osteoblast cell line SaOS-2, known for producing a mineralized matrix. While Orphan-1, a collagen type-2, specifically targets cartilaginous tissues and is essential for normal skeletal embryonic development, linear growth, and cartilage's ability to withstand compressive forces, Phosphoethanolamine (2-O3POCH2CH2NH3) acts as a key intermediate in cephalin formation, occurring through ethanolamine phosphorylation in the liver and brain. PHOSPHO2 and PHOSPHO1, despite their similarities, exhibit subtle variations in charge distribution around the presumed substrate entry site and the positioning of potential H-bond donors. Distinguished by its high specific phosphoethanolamine and phosphocholine phosphatase activities, PHOSPHO1, a phosphatase enzyme with elevated expression in mineralizing cells, is involved in generating Pi for matrix mineralization - a critical process in skeletal development. As a member of the haloacid dehalogenase (HAD) superfamily of Mg2+-dependent hydrolases, PHOSPHO1 exhibits notable specific activities towards phosphoethanolamine (PEA) and phosphocholine (PCho).
Description
Recombinant Human Phospho1, produced in E.Coli, is a single, non-glycosylated polypeptide chain comprising 295 amino acids. With a molecular weight of 31.3 kDa, it features a 14 aa His tag at the N-Terminus. Purification of Human Phosphocholine Phosphatase is achieved through proprietary chromatographic techniques.
Physical Appearance
It appears as a filtered, lyophilized (freeze-dried) powder.
Formulation
The product is filtered (0.4µm) and lyophilized from a solution of 0.5mg/ml in 30mM acetate buffer with a pH of 4.
Solubility
To prepare a working stock solution of around 0.5mg/ml, add 0.1M Acetate buffer (pH 4) to the lyophilized pellet and allow it to dissolve completely. For conversion to a higher pH value, dilute the solution extensively with the appropriate buffer to a concentration of 10µg/ml. Note that the protein's solubility is limited at higher concentrations. The product is not sterile; filter it through a suitable sterile filter before using it in cell cultures.
Stability
Store the lyophilized protein at -20°C. After reconstitution, aliquot the product to minimize repeated freeze-thaw cycles. The reconstituted protein remains stable at 4°C for a limited duration.
Purity
The purity is determined to be greater than 95% through SDS-PAGE analysis.
Synonyms
Phosphoethanolamine/phosphocholine phosphatase, Phosphatase, Orphan 1, EC 3.1.3.75, Phospho1.
Source
Escherichia Coli.
Amino Acid Sequence
MRGSHHHHHH GMASMSGCFP VSGLRCLSRD GRMAAQGAPR FLLTFDFDET IVDENSDDSI VRAAPGQRLP ESLRATYREG FYNEYMQRVF KYLGEQGVRP RDLSAIYEAI PLSPGMSDLL QFVAKQGACF EVILISDANT FGVESSLRAA GHHSLFRRIL SNPSGPDARG LLALRPFHTH SCARCPANMC KHKVLSDYLR ERAHDGVHFE RLFYVGDGAN DFCPMGLLAG GDVAFPRRGY PMHRLIQEAQ KAEPSSFRAS VVPWETAADV RLHLQQVLKSC.

Q&A

What is the structural characterization of human PHOSPHO1 protein?

PHOSPHO1 is a member of the haloacid dehalogenase (HAD) superfamily, containing four conserved motifs critical for enzyme catalysis. The human PHOSPHO1 protein features a Roseman folding structure with five parallel β-sheet structures surrounded by six α-helix structures, plus additional two β-sheets and four α-helices. The protein contains three conserved peptide motifs, with motif I comprising Thr and Val residues and two aspartic acids (Asp43 and Asp123) that are crucial for its catalytic activity .

The active site residues (Asp123, Asp32, Asp34, and Asp203) form a binding pocket for substrates like phosphocholine. The Mg²⁺ ion coordinates with these aspartic acid residues in an octahedral geometry, essential for the enzyme's function . Site-directed mutagenesis experiments revealed that mutation of Asp123 reduces the catalytic activity with phosphoethanolamine and phosphocholine by 20 and 60 times respectively, while mutation of Asp43 reduces activity with phosphoethanolamine and completely abolishes reactivity with phosphocholine .

How is the PHOSPHO1 gene organized and conserved across species?

The PHOSPHO1 gene shows remarkable conservation across multiple species. Human, chicken, and mouse PHOSPHO1 genes demonstrate conserved synteny, indicating they share the same evolutionary ancestor and are direct homologs . In humans, the PHOSPHO1 gene is located on chromosome HSA17, while in mice it is found in a region of conserved synteny .

Comparative analysis of amino acid sequences from human, pufferfish, drosophila, mouse, rat, chicken, zebrafish, and plants reveals that all possess the conserved motifs characteristic of the HAD superfamily, further confirming PHOSPHO1's ancient evolutionary origin across diverse species . This high degree of conservation suggests the fundamental importance of PHOSPHO1's function throughout evolutionary history.

What are the primary substrates and enzymatic activities of human PHOSPHO1?

Human PHOSPHO1 exhibits specific phosphatase activity toward phosphomonoester substrates, with highest specificity for phosphoethanolamine (PEA) and phosphocholine (PCho) . Using recombinant human PHOSPHO1 purified from the SaOS-2 osteosarcoma cell line, continuous spectrophotometric phosphate assays revealed that PHOSPHO1 efficiently hydrolyzes these substrates at an optimal pH range of 6.0 to 7.2 .

The enzyme catalyzes the hydrolysis of the phosphoester bond in these substrates, liberating inorganic phosphate (Pi) which is critical for biomineralization processes . Site-directed mutagenesis experiments disrupting the protein's active site dramatically decreased PEA and PCho hydrolysis to undetectable levels in some mutants, confirming the specificity of these substrates for PHOSPHO1 .

What is the role of PHOSPHO1 in bone mineralization and development?

PHOSPHO1 plays a fundamental role in skeletal mineralization by generating inorganic phosphate (Pi) within matrix vesicles, which are membrane-enclosed structures that serve as initial sites of mineral crystal formation. Through its phosphatase activity on phosphocholine and phosphoethanolamine, PHOSPHO1 contributes to the Pi pool necessary for hydroxyapatite crystal nucleation and growth .

Studies using PHOSPHO1 knockout mice (Phospho1-/-) have demonstrated its crucial importance in proper bone formation. These mice exhibit significant skeletal abnormalities including decreased bone mineral density, spontaneous fractures, bowed long bones, osteomalacia, and scoliosis . Interestingly, attempts to rescue this phenotype by cross-breeding with mice overexpressing tissue-nonspecific alkaline phosphatase (TNAP) were unsuccessful, indicating that PHOSPHO1's function in mineralization cannot be compensated by increased TNAP activity .

How do PHOSPHO1 and TNAP interact in the mineralization process?

PHOSPHO1 and tissue-nonspecific alkaline phosphatase (TNAP) collaborate in the biomineralization process through complementary mechanisms, though they function in distinct compartments. While PHOSPHO1 operates within matrix vesicles to generate Pi from phosphocholine and phosphoethanolamine, TNAP functions primarily on the cell membrane to hydrolyze pyrophosphate (PPi), an inhibitor of mineralization .

In Phospho1-/- mice, reduced plasma concentrations of TNAP and increased ectonucleotide pyrophosphatase/phosphodiesterase1 (NPP1) were observed, resulting in significantly higher PPi concentrations compared to wild-type controls . This mimics the biochemical profile seen in infantile hypophosphatasia in humans, where high serum PPi leads to inhibition of mineralization and subsequent rickets/osteomalacia .

The critical nature of this interaction is further demonstrated by PHOSPHO1-TNAP double knockout mice, which exhibit complete absence of skeletal mineralization and perinatal lethality, indicating the synergistic and non-redundant roles of these two phosphatases in the mineralization process .

What methodologies are used to assess PHOSPHO1 activity in mineralization studies?

Researchers employ several approaches to investigate PHOSPHO1's role in mineralization:

  • Continuous spectrophotometric phosphate assays: These are used to measure PHOSPHO1's enzymatic activity toward various phosphomonoester substrates, allowing for determination of substrate specificity and kinetic parameters .

  • Site-directed mutagenesis: By creating specific mutations in the active site residues, researchers can assess the importance of particular amino acids for enzymatic function .

  • Knockout mouse models: Phospho1-/- mice provide valuable insights into the in vivo function of PHOSPHO1 in bone development and mineralization .

  • Matrix vesicle isolation and analysis: Since PHOSPHO1 is present in matrix vesicles, isolation and analysis of these structures from mineralizing tissues helps understand its compartmentalized function .

  • Inhibitor studies: Specific PHOSPHO1 inhibitors have been developed to study its function without genetic manipulation, although interestingly, these inhibitors did not impair bone regeneration or influence bone mineralization in murine models, suggesting potential therapeutic applications that might not adversely affect skeletal integrity .

How does PHOSPHO1 influence brown adipose tissue thermogenesis?

PHOSPHO1 is highly enriched in both mouse and human brown adipose tissue (BAT) and acts as a negative regulator of thermogenesis . Several mechanisms have been identified:

  • Inverse relationship with UCP1: PHOSPHO1 appears to participate in UCP1-independent adipocyte respiration. Cold exposure and UCP1 activation increase PHOSPHO1 expression in adipose tissues, while knockdown of PHOSPHO1 by siRNA augments UCP1 expression in human brown adipocytes, suggesting a compensatory regulatory mechanism .

  • Regulation of thermogenic gene expression: PHOSPHO1 ablation induces the expression of thermogenic genes and mitochondria-related genes in BAT and subcutaneous white adipose tissue, enhancing cold tolerance and energy expenditure .

  • Phospholipid metabolism: Cold exposure increases total phosphatidylcholine (PC) and phosphatidylethanolamine (PE) content in mouse BAT, and since PHOSPHO1 expression is induced by cold exposure, there may be a regulatory feedback loop involving phospholipid metabolism .

Methodologically, these relationships have been studied using:

  • PHOSPHO1 knockout mice subjected to cold challenge tests

  • siRNA-mediated knockdown in clonal human brown adipocytes

  • Analysis of gene expression patterns in response to cold exposure

  • Measurement of energy expenditure and metabolic parameters

What is the relationship between PHOSPHO1 and glucose homeostasis/insulin sensitivity?

PHOSPHO1 serves as a significant regulator of glucose metabolism and insulin sensitivity, with knockout studies revealing protective effects against metabolic dysfunction . Mice lacking PHOSPHO1 demonstrate:

  • Improved basal glucose homeostasis: Phospho1-/- mice show better glucose tolerance under normal conditions .

  • Protection from diet-induced metabolic disorders: These mice are protected from high-fat diet (HFD)-induced obesity and diabetes .

  • Choline-dependent metabolic regulation: Interestingly, choline supplementation restores insulin sensitivity and adiposity in PHOSPHO1 knockout mice, suggesting that the metabolic benefits operate through altered choline metabolism pathways .

  • Amelioration of metabolic associated fatty liver disorder (MAFLD): Depletion of the PHOSPHO1 gene has been shown to improve MAFLD in mice, indicating its potential as a therapeutic target for fatty liver disease .

These findings position PHOSPHO1 as a potential therapeutic target for treating obesity and related metabolic disorders, with inhibition of its activity offering promise for improving metabolic health .

What experimental approaches should be used to study PHOSPHO1's role in stress erythropoiesis?

To investigate PHOSPHO1's function in stress erythropoiesis, researchers should consider the following approaches:

  • Phenylhydrazine-induced hemolytic anemia model: This established model reveals defects in stress erythropoiesis in PHOSPHO1 knockout mice, highlighting the enzyme's role in stress-related energy metabolism .

  • Metabolic profiling: Since PHOSPHO1 knockout mice switch to glycolysis for compensatory energy supply during stress erythropoiesis, comprehensive metabolic profiling can provide insights into altered metabolic pathways .

  • Hypoxia experiments: Given that PHOSPHO1 expression increases in blood samples from athletes undergoing high-altitude training (which induces hypoxia) and that hypoxia causes stress erythropoiesis, controlled hypoxia experiments can help elucidate PHOSPHO1's role in high-altitude adaptation-induced erythropoiesis .

  • Gene expression analysis: Assessing changes in PHOSPHO1 expression during different stages of erythropoiesis, particularly under stress conditions, can identify key regulatory points .

  • Red blood cell parameter measurements: Although adult PHOSPHO1 knockout mice show normal red blood cell properties (number, morphology, and osmotic fragility) under basal conditions, detailed hematological assessments under stress conditions are crucial for understanding PHOSPHO1's specific role in erythrocyte development and function .

How can we reconcile PHOSPHO1's dual roles in biomineralization and metabolic regulation?

The dual functionality of PHOSPHO1 in biomineralization and metabolism presents an intriguing research challenge. One approach to reconciling these roles is to examine the common biochemical pathways potentially linking these processes:

  • Phospholipid metabolism connection: PHOSPHO1's action on phosphocholine and phosphoethanolamine affects both matrix vesicle-mediated mineralization and cellular phospholipid homeostasis. Researchers should investigate whether alterations in phospholipid composition represent a mechanistic link between skeletal and metabolic phenotypes .

  • Tissue-specific expression and regulation: While PHOSPHO1 is expressed in multiple tissues, its regulation may differ significantly between bone cells and adipocytes. Comparative ChIP-seq and transcriptomic analyses across tissues could identify tissue-specific regulatory elements controlling PHOSPHO1 expression .

  • Protein interaction networks: Identifying tissue-specific protein interactors through co-immunoprecipitation followed by mass spectrometry could reveal how PHOSPHO1 achieves distinct functions in different cellular contexts .

  • Phosphometabolite profiling: Comprehensive analysis of phosphometabolites in various tissues of wild-type and PHOSPHO1 knockout mice could identify tissue-specific metabolic signatures that explain the diverse phenotypes observed .

  • Conditional knockout models: Generation of tissue-specific PHOSPHO1 knockout mice would help dissect the independent contributions of PHOSPHO1 in bone versus metabolic tissues, determining whether the metabolic phenotypes are secondary to skeletal changes or represent independent functions .

What are the implications of PHOSPHO1 inhibition for therapeutic applications in metabolic disorders?

Targeting PHOSPHO1 for metabolic disorders presents several promising avenues for therapeutic development, with several key considerations:

  • Safety profile regarding skeletal effects: Notably, PHOSPHO1 inhibitors did not impair bone regeneration or influence bone mineralization in murine models, suggesting they might regulate energy metabolism without causing skeletal impairment, at least in adulthood . This separation of effects is crucial for therapeutic development.

  • Effects on erythropoiesis: Similarly, adult PHOSPHO1 knockout mice do not exhibit hemolytic anemia or alterations in red blood cell properties, indicating PHOSPHO1 inhibitors should not impair normal erythropoiesis .

  • Mechanism of action: PHOSPHO1 inhibition leads to accumulation of its substrate phosphocholine, which itself induces cold tolerance and metabolic benefits in mice . Understanding whether therapeutic benefits derive from PHOSPHO1 inhibition directly or from phosphocholine accumulation will guide optimal drug development strategies.

  • Tissue-specific targeting: Given PHOSPHO1's multiple functions, developing delivery systems that target inhibitors specifically to adipose tissue could maximize metabolic benefits while minimizing potential side effects in other tissues.

  • Potential for combination therapy: Since choline supplementation restores insulin sensitivity and adiposity in PHOSPHO1 knockout mice , combination therapies that modulate both PHOSPHO1 activity and choline metabolism might offer synergistic benefits for metabolic disorders.

What methodological approaches should be employed to explore PHOSPHO1's subcellular localization and trafficking?

Understanding PHOSPHO1's subcellular distribution is critical for elucidating its function in different contexts. Several methodological approaches should be considered:

  • Advanced microscopy techniques:

    • Super-resolution microscopy (STORM, PALM, or STED) to visualize PHOSPHO1 with nanometer precision

    • Live-cell imaging with fluorescently tagged PHOSPHO1 to track its dynamics

    • Correlative light and electron microscopy (CLEM) to combine functional data with ultrastructural context

  • Subcellular fractionation and proteomics:

    • Isolation of organelles and membrane compartments followed by Western blotting

    • Proximity labeling methods (BioID or APEX) to identify proteins in close proximity to PHOSPHO1

    • Quantitative mass spectrometry of subcellular fractions to determine enrichment patterns

  • Localization signal analysis:

    • Computational prediction of localization signals

    • Creation of truncation mutants to identify domains responsible for specific localization patterns

    • Site-directed mutagenesis of predicted localization signals

  • Trafficking studies:

    • Pulse-chase experiments with PHOSPHO1 fusion proteins

    • Inhibition of specific trafficking pathways using pharmacological agents

    • Co-localization studies with markers of secretory and endocytic pathways

  • Matrix vesicle isolation and characterization:

    • Differential ultracentrifugation to isolate matrix vesicles

    • Immunogold electron microscopy to visualize PHOSPHO1 within vesicles

    • Proteomic and lipidomic analysis of matrix vesicles in wild-type versus PHOSPHO1 knockout models

What are the critical gaps in our understanding of PHOSPHO1 regulation?

Despite significant advances, several aspects of PHOSPHO1 regulation remain poorly understood:

  • Transcriptional regulation: Although PHOSPHO1 expression is known to increase during terminal erythropoiesis and in response to cold exposure in BAT, the specific transcription factors and regulatory elements controlling its expression in different tissues remain largely unidentified .

  • Post-translational modifications: Information about how PHOSPHO1 activity might be regulated through phosphorylation, ubiquitination, or other post-translational modifications is lacking.

  • Substrate availability and transport: While PHOSPHO1 is likely located in the cytosol according to database predictions, it remains unclear how its substrates (phosphocholine and phosphoethanolamine) are transported or diffused to where PHOSPHO1 is located for efficient catalysis . Understanding substrate compartmentalization is crucial for complete mechanistic insights.

  • Feedback regulation: The compensatory regulation observed between PHOSPHO1 and UCP1 suggests complex feedback mechanisms that require further elucidation .

  • Tissue-specific regulatory networks: The diverse phenotypes observed in different tissues of PHOSPHO1 knockout mice suggest tissue-specific regulatory networks that remain to be mapped.

How should researchers approach the design of PHOSPHO1 inhibitors for metabolic applications?

Designing effective PHOSPHO1 inhibitors for metabolic applications requires a multi-faceted approach:

  • Structure-based drug design:

    • Utilize the AlphaFold-predicted structure of PHOSPHO1 for virtual screening of compound libraries

    • Focus on competitive inhibitors that target the substrate-binding pocket formed by Asp123, Asp32, Asp34, and Asp203 residues

    • Design transition-state analogs based on the reaction mechanism

  • High-throughput screening:

    • Develop robust biochemical assays measuring phosphate release from PHOSPHO1 substrates

    • Screen diverse chemical libraries using purified recombinant human PHOSPHO1

    • Validate hits in cellular assays measuring phosphocholine accumulation

  • Selectivity profiling:

    • Test lead compounds against other HAD family members to ensure specificity

    • Assess effects on related phosphatases like TNAP to avoid unintended consequences on bone mineralization

    • Perform comprehensive phosphoproteomic analyses to identify off-target effects

  • Pharmacokinetic optimization:

    • Design compounds with appropriate tissue distribution, particularly enrichment in adipose tissue

    • Optimize metabolic stability and half-life

    • Consider prodrug approaches for targeted delivery

  • Therapeutic index determination:

    • Establish dose-response relationships for metabolic benefits versus potential skeletal effects

    • Conduct long-term safety studies in adult and developing animals

    • Evaluate effects on stress erythropoiesis under various challenge conditions

What future research directions could advance our understanding of PHOSPHO1 in human health and disease?

Several promising research directions could significantly advance our understanding of PHOSPHO1:

  • Human genetic studies:

    • Analysis of PHOSPHO1 variants in populations with metabolic syndrome or bone disorders

    • Genome-wide association studies (GWAS) to identify correlations between PHOSPHO1 polymorphisms and disease risk

    • Functional characterization of identified variants

  • Single-cell analyses:

    • Single-cell RNA sequencing to identify cell populations expressing PHOSPHO1 in various tissues

    • Spatial transcriptomics to map PHOSPHO1 expression patterns within tissues

    • Single-cell metabolomics to understand cell-specific metabolic changes in response to PHOSPHO1 modulation

  • Overexpression studies:

    • Generation of PHOSPHO1 overexpression models to complement existing knockout studies

    • Analysis of tissue-specific effects of PHOSPHO1 overexpression

    • Investigation of dose-dependent effects on metabolism and bone mineralization

  • Integration with systems biology:

    • Multi-omics approaches combining transcriptomics, proteomics, and metabolomics

    • Network analysis to position PHOSPHO1 within metabolic and signaling pathways

    • Mathematical modeling of PHOSPHO1's role in phospholipid homeostasis

  • Translational research:

    • Development of biomarkers for PHOSPHO1 activity in clinical samples

    • Preclinical testing of PHOSPHO1 inhibitors in animal models of obesity and diabetes

    • Exploration of natural compounds that modulate PHOSPHO1 activity for potential nutritional interventions

Product Science Overview

Structure and Production

PHOSPHO1 is a member of the haloacid dehalogenase (HAD) superfamily of Mg2±dependent hydrolases. The human recombinant version of PHOSPHO1 is produced in Escherichia coli (E. coli) and is a single, non-glycosylated polypeptide chain containing 295 amino acids with a molecular mass of 31.3 kDa. It is fused to a 14 amino acid His tag at the N-terminus .

Function and Activity

PHOSPHO1 exhibits high specific activities toward phosphoethanolamine (PEA) and phosphocholine (PCho), which are key intermediates in the formation of cephalins. These activities are crucial for the generation of inorganic phosphate (Pi) necessary for matrix mineralization, a process central to skeletal development .

Role in Metabolism

Recent studies have identified PHOSPHO1 as a potential regulator of energy metabolism. Research involving Phospho1 mutant mice has shown that the absence of PHOSPHO1 leads to improved basal glucose homeostasis and resistance to high-fat-diet-induced weight gain and diabetes. This suggests that PHOSPHO1 could be a potential therapeutic target for treating obesity and diabetes .

Expression and Localization

PHOSPHO1 is highly expressed at sites of mineralization in bone and cartilage, particularly in osteoblast cell lines such as SaOS-2, which produce a mineralized matrix. It is also specific for cartilaginous tissues, indicating its specialized role in skeletal development .

Stability and Storage

The recombinant PHOSPHO1 protein is typically stored as a lyophilized (freeze-dried) powder and should be reconstituted in an appropriate buffer before use. It is recommended to store the lyophilized protein at -20°C and to aliquot the product after reconstitution to avoid repeated freezing and thawing cycles .

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.