PGK1 Human

Phosphoglycerate Kinase 1 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Clinical Implications

PGK1 dysfunction is implicated in diverse pathologies:

ConditionMechanismKey Findings
Phosphoglycerate Kinase DeficiencyReduced ATP production in red blood cells and neuronsChronic hemolytic anemia, neurological symptoms, early-onset Parkinson’s disease
Cancer MetabolismEnhanced glycolysis, angiogenesis, and chemoresistanceHigh PGK1 expression correlates with poor prognosis in ovarian and renal cancers
Parkinson’s DiseaseEnergy deficits in dopamine neuronsBoosting PGK1 activity with terazosin improves neuronal ATP production and synaptic function

Role in Neurodegeneration

PGK1 is a rate-limiting enzyme in neuronal glycolysis, particularly in dopamine neuron axons. A 2024 study demonstrated that PGK1 overexpression or terazosin (an α1-adrenergic blocker) rescues ATP depletion and synaptic dysfunction in Parkinson’s disease models :

  • Terazosin Mechanism: Enhances PGK1 activity, accelerating ATP recovery during low-glucose conditions.

  • DJ-1 Interaction: PGK1 forms a functional complex with DJ-1, a Parkinson’s-linked protein, to regulate energy metabolism .

Oncogenic and Metabolic Roles

PGK1 drives cancer progression via:

  • Glycolysis Upregulation: High PGK1 expression correlates with aggressive ovarian cancer and sorafenib resistance in renal cancer .

  • Mitochondrial Translocation: In hypoxic conditions, PGK1 translocates to mitochondria, phosphorylates PDHK1, and inhibits pyruvate dehydrogenase, suppressing TCA cycle activity and promoting lactate production .

Interactome Insights

PGK1 interacts with glycolytic enzymes and signaling proteins (data from STRING database ):

Protein PartnerFunctionInteraction Score
TPI1Triosephosphate isomerase0.999
GAPDHGlyceraldehyde-3-phosphate dehydrogenase0.999
ENO1Enolase (plasminogen receptor)0.996

Neuroprotection in Parkinson’s Disease

Terazosin, a repurposed drug, shows promise in preclinical models by enhancing PGK1 activity. Retrospective studies suggest prolonged terazosin use reduces Parkinson’s risk by ~37% .

Anti-Cancer Strategies

  • Glycolysis Inhibition: Targeting PGK1 in cancers with high glycolytic dependence (e.g., ovarian, renal) may improve treatment efficacy .

  • Combination Therapies: Inhibiting PGK1-mediated CXCR4/ERK signaling could overcome sorafenib resistance in renal cancer .

Experimental Tools and Resources

PGK1 research relies on advanced tools:

ResourceDetails
ORF ClonesMyc-DDK-tagged PGK1 clones (e.g., RC211172) for protein expression studies
AntibodiesWestern blot-validated reagents for quantifying PGK1 expression
BioinformaticsTISIDB, TIMER, and Metascape for analyzing PGK1-immune cell interactions

Future Directions

  1. Precision Therapies: Develop PGK1-targeted drugs for Parkinson’s and cancers.

  2. Biomarker Discovery: Validate PGK1 serum levels as prognostic markers in renal and ovarian cancers .

  3. Metabolic Reprogramming: Explore PGK1’s role in mitochondrial dynamics and redox balance.

Product Specs

Introduction
Phosphoglycerate kinase 1 (PGK1) is an enzyme encoded by the PGK1 gene located on the X chromosome. This enzyme plays a crucial role in the glycolytic pathway by catalyzing the reversible transfer of a phosphate group from 1,3-bisphosphoglycerate to ADP, yielding 3-phosphoglycerate and ATP. PGK1's function is essential for energy production in cells. Beyond its role in glycolysis, PGK1 may also function as a cofactor for polymerase alpha, contributing to DNA replication. Defects in the PGK1 gene can lead to chronic hemolytic anemia, often accompanied by mental retardation or muscular disorders like rhabdomyolysis. Elevated levels of PGK1 and its associated signaling pathways are linked to the progression and spread of diffuse primary gastric carcinomas. Additionally, studies have revealed altered expression patterns of PGK1 in specific brain regions of individuals with schizophrenia.
Description
Recombinant human PGK1, expressed in E. coli, is a non-glycosylated polypeptide chain containing 417 amino acids (corresponding to residues 1-417 of the native protein) with a 20 amino acid His-tag fused at the N-terminus. The protein has a molecular weight of 46.8 kDa and is purified to high purity using proprietary chromatographic techniques.
Physical Appearance
Clear, colorless solution, sterile-filtered.
Formulation
The PGK1 protein is supplied in a solution containing 20mM Tris buffer (pH 8.0), 10% glycerol, and 1mM DTT.
Stability
PGK1 is stable at 4°C for up to 1 week. For extended storage, it is recommended to store the protein below -18°C. The addition of a carrier protein like HSA or BSA (0.1%) is recommended for long-term storage. Avoid repeated freeze-thaw cycles.
Purity
The purity of PGK1 is determined to be greater than 95% as analyzed by SDS-PAGE.
Synonyms
Phosphoglycerate kinase 1, Primer recognition protein 2, Cell migration-inducing gene 10 protein, PRP 2, PGKA, MIG10, MGC8947, MGC117307, MGC142128, PGK1.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MSLSNKLTLD KLDVKGKRVV MRVDFNVPMK NNQITNNQRI KAAVPSIKFC LDNGAKSVVL MSHLGRPDGV PMPDKYSLEP VAVELKSLLG KDVLFLKDCV GPEVEKACAN PAAGSVILLE NLRFHVEEEG KGKDASGNKV KAEPAKIEAF RASLSKLGDV YVNDAFGTAH RAHSSMVGVN LPQKAGGFLM KKELNYFAKA LESPERPFLA ILGGAKVADK IQLINNMLDK VNEMIIGGGM AFTFLKVLNN MEIGTSLFDE EGAKIVKDLM SKAEKNGVKI TLPVDFVTAD KFDENAKTGQ ATVASGIPAG WMGLDCGPES SKKYAEAVTR AKQIVWNGPV GVFEWEAFAR GTKALMDEVV KATSRGCITI IGGGDTATCC AKWNTEDKVS HVSTGGGASL ELLEGKVLPG VDALSNI.

Q&A

What is the primary function of PGK1 in human cells?

PGK1 (phosphoglycerate kinase 1) primarily functions as a key enzyme in the glycolytic pathway, catalyzing the conversion of 1,3-diphosphoglycerate to 3-phosphoglycerate while generating one molecule of adenosine triphosphate (ATP). This reaction represents a critical energy-producing step during glycolysis, where glucose is broken down to produce cellular energy. PGK1 is ubiquitously expressed in cells and tissues throughout the human body, highlighting its fundamental importance in energy metabolism. The enzyme plays a crucial role in maintaining cellular energy homeostasis, particularly in tissues with high energy demands such as muscles and the nervous system. Beyond its canonical glycolytic function, emerging research suggests PGK1's involvement in additional cellular processes, though these secondary roles remain under active investigation .

How is the PGK1 gene structured and regulated in humans?

The human PGK1 gene is located on the X chromosome at position Xq13.3 and spans approximately 23 kilobases. The gene consists of 11 exons and 10 introns, encoding a 417-amino acid protein with a molecular mass of approximately 45 kDa. PGK1 is constitutively expressed in all somatic cells and premeiotic cells as a housekeeping gene. A separate gene, PGK2, exists on chromosome 6p12-21.1 and exhibits tissue-specific expression in spermatogenic cells. Interestingly, the PGK2 gene is intronless and displays characteristics of a retroposon. The regulation of PGK1 expression involves both transcriptional and post-transcriptional mechanisms, with its expression modulated in response to metabolic demands and cellular stress conditions. Understanding these regulatory mechanisms has important implications for both basic research and potential therapeutic interventions targeting PGK1-related pathways .

What experimental approaches are used to measure PGK1 enzymatic activity?

Researchers utilize several methodological approaches to measure PGK1 enzymatic activity. A commonly employed technique is spectrophotometric assay, which measures the conversion of 1,3-diphosphoglycerate to 3-phosphoglycerate by monitoring the oxidation of NADH to NAD+ when coupled with glyceraldehyde-3-phosphate dehydrogenase. Another approach involves radiometric assays using 32P-labeled substrates to track phosphate transfer. For more precise kinetic studies, researchers employ stopped-flow techniques to measure rapid reaction rates. In living cells, PGK1 activity can be assessed indirectly through measurements of glycolytic flux and ATP production rates. When investigating tissue samples from patients with suspected PGK1 deficiency, researchers typically perform enzyme activity assays on erythrocyte lysates, as red blood cells heavily depend on glycolysis for energy production. These methodological approaches provide complementary information about PGK1's catalytic function in various experimental contexts and clinical specimens .

What is the correlation between PGK1 mutations and specific clinical manifestations?

The genotype-phenotype correlations in PGK1 deficiency present a complex pattern that researchers are still working to fully elucidate. The clinical presentation of PGK1 deficiency is remarkably pleomorphic, with patients exhibiting various combinations of hemolytic anemia, neurological dysfunction, and myopathy. Rarely do patients display all three clinical features simultaneously. Specific mutations appear to impact different tissues preferentially, though the molecular basis for this tissue specificity remains incompletely understood. For example, some mutations predominantly affect red blood cells, causing hemolytic anemia, while others primarily impact neural tissues or muscle function. The hemolytic anemia typically varies from mild to severe, with marked exacerbations during infections. Neurological manifestations can include mental retardation, progressive decline of motor function, developmental delay, seizures, epilepsy, ataxia, tremor, and hemiplegic migraines. Myopathy presents as a progressive condition characterized by exercise intolerance, muscle weakness, cramping, myalgia, and episodes of myoglobinuria during strenuous exercise or fever. The molecular mechanisms underlying this tissue specificity likely involve differences in energy metabolism, PGK1 expression levels, and potential non-glycolytic functions of PGK1 in different cell types .

What therapeutic approaches are being investigated for PGK1 deficiency?

Currently, no specific therapy exists for PGK1 deficiency, highlighting a significant unmet medical need. Current management approaches focus on symptom control rather than addressing the underlying enzyme deficiency. For severe anemia, red cell transfusions may be required, particularly in the first years of life or during intercurrent infections. Some patients have undergone splenectomy with varying degrees of success, which may improve red cell survival but does not correct the fundamental hemolytic process. Recent therapeutic innovations include allogeneic bone marrow transplantation, which has been attempted to arrest the development of neurological manifestations in some cases. Emerging research directions include enzyme replacement therapy, gene therapy approaches targeting the PGK1 gene, and small-molecule chaperones designed to stabilize mutant PGK1 protein and enhance residual enzyme activity. Metabolic bypass strategies that aim to supplement the energy deficit in affected tissues are also under investigation. These experimental approaches remain in preclinical stages, underscoring the need for continued research into novel therapeutic strategies for this rare genetic disorder .

How does PGK1 function as a protein kinase in cancer metabolism?

Recent groundbreaking research has revealed that PGK1 can function as a protein kinase, significantly expanding our understanding of this enzyme beyond its classical glycolytic role. Under hypoxic stress, EGFR activation, or oncogenic mutations (such as K-Ras G12V or B-Raf V600E), PGK1 undergoes ERK1/2-dependent phosphorylation at S203. This modification triggers isomerization by PIN1, exposing a mitochondrial targeting sequence that enables PGK1 translocation into mitochondria. Once in the mitochondria, PGK1 directly phosphorylates pyruvate dehydrogenase kinase isozyme 1 (PDHK1) at T338 using ATP as a phosphate donor. This phosphorylation activates PDHK1, enhancing its ability to phosphorylate pyruvate dehydrogenase E1α at S293, which subsequently inactivates the pyruvate dehydrogenase complex. This molecular mechanism effectively inhibits pyruvate oxidation in mitochondria while promoting lactate production in the cytosol - a metabolic shift known as the Warburg effect that supports cancer cell proliferation. Importantly, deficiency in mitochondrial PGK1 translocation dramatically reduces tumor growth in orthotopic brain tumor models, and clinical studies have shown that PGK1 pS203 and PDHK1 pT338 levels inversely correlate with survival duration in glioblastoma patients. These findings identify PGK1 as a crucial metabolic switch in cancer progression and a potential therapeutic target .

What methodologies are used to study PGK1 translocation to mitochondria?

Investigating PGK1 translocation to mitochondria requires sophisticated experimental approaches that integrate molecular biology, biochemistry, and advanced imaging techniques. Researchers typically begin with subcellular fractionation to isolate mitochondrial, cytosolic, and nuclear fractions, followed by Western blot analysis to detect PGK1 in each compartment. The purity of these fractions is verified using compartment-specific markers such as VDAC or COX IV for mitochondria. Immunofluorescence microscopy with co-localization analysis provides spatial resolution, often combining PGK1 antibodies with mitochondrial stains like MitoTracker. For studying the dynamics of translocation, live-cell imaging with fluorescently tagged PGK1 constructs allows real-time visualization of the process. Site-directed mutagenesis targeting key residues (particularly S203) helps establish the importance of specific phosphorylation events. CRISPR/Cas9-mediated knock-in of PGK1 S203A mutants provides definitive evidence for the role of this modification. Mass spectrometry is employed to verify phosphorylation states, while co-immunoprecipitation experiments demonstrate interactions with transport machinery components. Functional studies using mitochondrial respiration analysis and lactate production measurements connect translocation to metabolic outcomes. These complementary approaches collectively provide robust evidence for the regulated mitochondrial translocation of PGK1 and its functional consequences .

How does extracellular PGK1 promote neurite outgrowth?

Recent discoveries have revealed an unexpected role for extracellular PGK1 (ePGK1) in promoting neural regeneration through a specific receptor-mediated mechanism. Research demonstrates that ePGK1 interacts with the neural membrane protein Enolase-2 (Eno2), but not with Enolase-1 or Enolase-3, to stimulate neurite outgrowth in motor neurons. Specifically, the 380th-417th domain of PGK1 interacts with the 405th-431st domain of Enolase-2. This interaction reduces P38/Limk1/Cofilin signaling, a pathway known to regulate actin dynamics in growth cones. When neural cells are incubated with recombinant PGK1, they show a significant increase in the proportion of neurite-bearing cells and average neurite length. This effect is substantially enhanced when combined with Enolase-2 overexpression. Mechanistically, the addition of extracellular PGK1 decreases phosphorylated Cofilin levels, promoting actin dynamics necessary for neurite extension. Importantly, knockdown of Enolase-2 using siRNA abolishes this effect, confirming the specificity of the PGK1-Enolase-2 interaction. These findings have significant implications for neurodegenerative conditions, as low PGK1 expression has been linked to several neurodegenerative diseases, including Parkinson's disease and amyotrophic lateral sclerosis. Enhanced PGK1 activity has been shown to slow neurodegeneration in disease models, suggesting therapeutic potential for targeting this pathway in neural regeneration strategies .

What are the most effective methods for purifying recombinant human PGK1 for structural and functional studies?

Purification of recombinant human PGK1 for high-resolution structural and functional studies requires optimized protocols that preserve the enzyme's native conformation and activity. The most effective approach begins with cloning the human PGK1 cDNA into a bacterial expression vector (commonly pET systems) with an affinity tag such as 6xHis or GST. Expression in E. coli BL21(DE3) strains is typically induced with IPTG at lower temperatures (16-18°C) to enhance proper folding. For purification, a multi-step process yields the best results: initial capture using affinity chromatography (Ni-NTA for His-tagged constructs), followed by tag removal with a specific protease (TEV or thrombin), and subsequent purification steps including ion-exchange chromatography and size-exclusion chromatography to achieve >95% purity. Throughout the purification process, maintaining buffer conditions that stabilize PGK1 is critical - typically phosphate buffers at pH 7.4 with glycerol (5-10%) and reducing agents like DTT or β-mercaptoethanol to prevent oxidation of cysteine residues. For structural studies, especially crystallography, further optimization through limited proteolysis or surface entropy reduction may enhance crystallization propensity. Activity assays performed at each purification step ensure that the purified enzyme retains full catalytic function. When studying specific PGK1 mutations associated with disease, parallel purification of wild-type and mutant proteins under identical conditions allows direct comparison of structural and functional properties .

How can researchers differentiate between the glycolytic and protein kinase functions of PGK1?

Differentiating between PGK1's glycolytic and protein kinase functions requires sophisticated experimental approaches that can specifically isolate and measure each activity. For the glycolytic function, researchers typically use coupled enzyme assays that measure the conversion of 1,3-diphosphoglycerate to 3-phosphoglycerate through changes in NADH absorbance when linked to glyceraldehyde-3-phosphate dehydrogenase activity. In contrast, PGK1's protein kinase activity is assessed using in vitro kinase assays with purified PGK1 and potential substrate proteins (such as PDHK1), followed by detection of phosphorylation using phospho-specific antibodies or 32P-ATP incorporation. To distinguish between these functions in cellular contexts, researchers employ several strategies: subcellular fractionation to separate mitochondrial (where kinase activity predominates) from cytosolic PGK1 (primarily glycolytic); site-directed mutagenesis targeting residues specifically required for one function but not the other (e.g., T378P mutation abolishes kinase activity while preserving glycolytic function); and pharmacological approaches using inhibitors that preferentially affect one activity. CRISPR/Cas9-mediated gene editing to introduce specific mutations (like S203A that prevents mitochondrial translocation) helps establish the physiological significance of kinase activity. Metabolic flux analysis using isotope-labeled glucose further distinguishes the metabolic consequences of these distinct functions. These complementary approaches collectively enable researchers to parse the relative contributions of PGK1's dual functions in various physiological and pathological contexts .

What analytical techniques are most suitable for detecting PGK1 post-translational modifications?

Multiple analytical techniques are employed to comprehensively characterize PGK1 post-translational modifications (PTMs), each offering distinct advantages for specific research questions. Mass spectrometry (MS) remains the gold standard, with phosphoproteomics workflows using titanium dioxide enrichment particularly effective for detecting PGK1 phosphorylation at S203 and other sites. Modern MS approaches include multiple reaction monitoring (MRM) for targeted quantification of specific modified peptides and top-down proteomics for analyzing intact PGK1 proteoforms. Complementing MS, Western blotting with modification-specific antibodies (such as anti-phospho-S203-PGK1) provides a straightforward method for examining PTM status in cellular and tissue samples. For investigating acetylation, deacetylase inhibitors like trichostatin A or nicotinamide are often used prior to analysis. Proximity ligation assays offer enhanced sensitivity and specificity for detecting modified PGK1 in fixed cells or tissue sections. The dynamics of PTMs can be studied through pulse-chase experiments with metabolic labeling. For functional characterization, site-directed mutagenesis creating phosphomimetic (S→D/E) or phospho-deficient (S→A) mutants allows assessment of how specific modifications affect PGK1 activity, localization, and interactions. Increasingly, computational approaches integrating structural biology with molecular dynamics simulations help predict how PTMs affect PGK1 conformation and function. These diverse analytical approaches collectively provide a comprehensive view of the complex PTM landscape regulating PGK1's multifunctional roles .

How is PGK1 involved in the pathogenesis of neurodegenerative diseases?

Recent research has unveiled significant connections between PGK1 and neurodegenerative pathologies, suggesting potential therapeutic avenues. Low expression levels of PGK1 have been associated with several neurodegenerative conditions, notably Parkinson's disease. Parkinsonism patients exhibit PGK1 deficiency, suggesting that reduced glycolysis may contribute to nigrostriatal damage. Conversely, enhanced PGK1 activity, accompanied by increased glycolytic flux, has demonstrated neuroprotective effects, slowing neurodegeneration and symptom progression in Parkinson's disease and amyotrophic lateral sclerosis (ALS) models. PGK1 appears to function through multiple neuroprotective mechanisms: maintaining ATP production in neurons under stress conditions; reducing oxidative damage through NADPH generation via the pentose phosphate pathway; and promoting neural regeneration through its extracellular signaling role with Enolase-2. Extracellular PGK1 specifically interacts with neural membrane receptor Enolase-2 to reduce P38/Limk1/Cofilin signaling, promoting neurite outgrowth in motor neurons. This mechanism has proven effective in rescuing motor axon phenotypes in spinal muscular atrophy zebrafish models. These findings suggest that PGK1-targeted interventions could provide novel therapeutic strategies for neurodegenerative diseases characterized by energy metabolism deficits and impaired neural regeneration. The dual intracellular and extracellular functions of PGK1 in neural tissues represent an exciting frontier in neurodegenerative disease research .

What is the role of PGK1 in cancer progression and how might it be targeted therapeutically?

PGK1's multifaceted roles in cancer progression present both challenges and opportunities for therapeutic intervention. Beyond its classical glycolytic function, PGK1 contributes to cancer development through its protein kinase activity, particularly following mitochondrial translocation. This process is triggered by hypoxia, EGFR activation, or oncogenic mutations (K-Ras G12V, B-Raf V600E), leading to ERK1/2-dependent S203 phosphorylation and subsequent mitochondrial localization. Once in mitochondria, PGK1 phosphorylates PDHK1 at T338, activating it to phosphorylate pyruvate dehydrogenase E1α at S293. This modification inactivates the pyruvate dehydrogenase complex, shifting metabolism toward aerobic glycolysis (the Warburg effect) by inhibiting pyruvate oxidation in mitochondria and enhancing lactate production. Clinical evidence supports PGK1's oncogenic role, as PGK1 pS203 and PDHK1 pT338 levels inversely correlate with survival duration in glioblastoma patients. Several therapeutic strategies targeting PGK1 are under investigation: small molecule inhibitors that specifically block PGK1's protein kinase activity without affecting its glycolytic function; compounds that prevent PGK1 mitochondrial translocation by interfering with S203 phosphorylation or PIN1-mediated isomerization; and combinatorial approaches targeting both PGK1 and downstream effectors like PDHK1. CRISPR/Cas9-mediated gene editing studies have demonstrated that replacing endogenous PGK1 with a mitochondrial translocation-deficient mutant (S203A) dramatically reduces brain tumor growth in orthotopic mouse models, validating this pathway as a promising therapeutic target .

How do PGK1 interactions with other proteins influence its non-canonical functions?

PGK1's diverse non-canonical functions are increasingly understood to be mediated through specific protein-protein interactions that extend beyond its glycolytic role. As a moonlighting protein, PGK1 engages in functional interactions with multiple partners in different cellular compartments. In the mitochondria, following ERK1/2-mediated S203 phosphorylation, PGK1 interacts with Peptidyl-prolyl cis/trans isomerase NIMA-interacting 1 (PIN1), which catalyzes its isomerization and exposes the mitochondrial targeting sequence (38-QRIKAA-43). This sequence is recognized by the translocase of the outer membrane (TOM) complex, facilitating mitochondrial import. Once inside mitochondria, PGK1 directly interacts with and phosphorylates PDHK1, a key regulator of mitochondrial metabolism. In the extracellular environment, PGK1 specifically binds to Enolase-2 (but not Enolase-1 or Enolase-3) on neural cell membranes, with interaction domains precisely mapped to the 380th-417th domain of PGK1 and the 405th-431st domain of Enolase-2. This interaction triggers intracellular signaling cascades affecting P38/Limk1/Cofilin pathway activity, ultimately promoting neurite outgrowth. Additionally, nuclear PGK1 has been implicated in DNA replication and repair through interactions with DNA polymerases and other components of the replication machinery. These diverse protein interactions allow PGK1 to function as a metabolic sensor and signal transducer, coordinating cellular responses to changing environmental and metabolic conditions. Methodologically, these interactions are studied using co-immunoprecipitation, proximity ligation assays, FRET/BRET technologies, and hydrogen-deuterium exchange mass spectrometry to map interaction interfaces .

Product Science Overview

Gene and Protein Structure

PGK1 is encoded by the PGK1 gene located on the X chromosome. The gene is highly conserved across different species, indicating its essential role in cellular metabolism . The protein consists of 417 amino acids and has a molecular weight of approximately 44.5 kDa .

Biological Functions

Apart from its primary role in glycolysis, PGK1 has been identified as a moonlighting protein, meaning it performs multiple mechanistically distinct functions. For instance, PGK1 acts as a cofactor for polymerase alpha, playing a role in DNA replication . Additionally, PGK1 is involved in angiogenesis, the process of forming new blood vessels, by reducing disulfide bonds in the serine protease plasmin, leading to the release of angiostatin, a tumor blood vessel inhibitor .

Clinical Significance

Deficiency in PGK1 can lead to a variety of clinical phenotypes, including hemolytic anemia and neurological impairments . The enzyme’s dysregulation has also been observed in numerous malignancies, making it a potential target for cancer therapy . For example, PGK1 is upregulated in non-small cell lung cancer (NSCLC) and is associated with poor prognosis . Inhibition of PGK1 has been shown to reduce NSCLC cell proliferation, migration, and invasion .

Recombinant PGK1

Human recombinant PGK1 is produced using recombinant DNA technology, which involves inserting the human PGK1 gene into a suitable expression system, such as bacteria or yeast, to produce the protein in large quantities. This recombinant protein is used in various research applications, including studies on glycolysis, cancer metabolism, and enzyme kinetics .

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.