PGAM2 antibodies are immunodetection reagents targeting the PGAM2 protein, encoded by the PGAM2 gene (NCBI Gene ID: 5224). PGAM2 is a 29 kDa glycolytic enzyme that catalyzes the conversion of 3-phosphoglycerate to 2-phosphoglycerate. Its dysregulation is implicated in metabolic myopathies, cancer progression, and cardiovascular diseases . Antibodies against PGAM2 are essential for studying its expression, localization, and interactions in both physiological and pathological contexts.
Protein Composition: 253 amino acids, forming homo- or heterodimers (e.g., MM in muscle, MB in heart) .
Key Residues: Lysine 100 (K100) in the active site is critical for catalytic activity and regulated by acetylation .
PGAM2 facilitates glycolysis via a two-step reaction:
Transfer of a phosphate group from phosphohistidine to 3-PGA, forming 2,3-bisphosphoglycerate.
Regeneration of phosphohistidine by transferring the C3 phosphate .
Observed Band Size: 25–29 kDa (matches predicted molecular weight) .
Tissue Specificity: Strong reactivity in skeletal muscle lysates .
Subcellular Distribution: Cytoplasmic and nuclear staining in human skeletal muscle and hepatocellular carcinoma (HCC) .
Clinical Correlation: Nuclear PGAM2 overexpression in HCC correlates with poor prognosis (HR = 1.82, P = 0.001) .
Cancer: PGAM2 inhibition sensitizes enzalutamide-resistant prostate cancer cells by suppressing BCL-xL .
Cardiology: PGAM2 knockdown reduces HSP90 stability, mitigating cardiac hypertrophy .
Model: Angiotensin II (Ang II)-induced rat cardiomyocyte hypertrophy.
Mechanism: PGAM2 competes with SYVN1 to stabilize HSP90, worsening hypertrophy. Knockdown improves LV mass and fibrosis .
Intervention: AAV9-mediated PGAM2 silencing reduces ANP/BNP expression (P < 0.01) .
CRISPR Screen: PGAM2 deletion overcomes enzalutamide resistance by activating BAD-mediated apoptosis .
Regulation: AR transcriptionally upregulates PGAM2, linking androgen signaling to metabolic adaptation .
Prognostic Marker: High nuclear PGAM2 correlates with reduced survival (log-rank P = 0.001) .
Metastasis: Nuclear PGAM2 promotes glycolysis and ROS generation in HCC cells .
PGAM2 (phosphoglycerate mutase 2) is a muscle-specific glycolytic enzyme that catalyzes the conversion of 3-phosphoglycerate to 2-phosphoglycerate in the glycolytic pathway. It is a 29 kDa protein consisting of 253 amino acid residues in humans . PGAM2 is predominantly expressed in heart and skeletal muscle tissues and represents a critical enzyme in cellular energy metabolism . Its importance extends beyond its metabolic role, as PGAM2 has been implicated in the Notch signaling pathway and spermatogenesis . Additionally, mutations in the PGAM2 gene have been associated with glycogen storage disease X (GSDX), making it an important target for both metabolic and genetic disease research . Recent studies have revealed non-glycolytic functions of PGAM2, including its interactions with regulatory proteins like 14-3-3ζ, highlighting its multifunctional nature in cellular processes .
PGAM2 antibodies are utilized across multiple immunodetection techniques in research settings. The most widely used application is Western Blotting (WB), with recommended dilutions typically ranging from 1:1000 to 1:6000 depending on the specific antibody . Other common applications include:
Application | Common Dilution Range | Notes |
---|---|---|
Western Blot (WB) | 1:1000-1:6000 | Most frequently cited application |
Immunohistochemistry (IHC) | 1:50-1:500 | Often requires antigen retrieval with TE buffer pH 9.0 |
Immunofluorescence (IF) | Variable | Used for subcellular localization studies |
Immunoprecipitation (IP) | Variable | For protein-protein interaction studies |
ELISA | Variable | For quantitative detection |
Researchers have successfully used these antibodies on various tissue types including human breast cancer tissue, mouse skeletal muscle, heart tissue, and brain tissue . The reactivity of commercially available antibodies commonly includes human, mouse, and rat samples .
Verifying antibody specificity is crucial for obtaining reliable experimental results. For PGAM2 antibodies, several validation approaches are recommended:
Positive control testing with tissues known to express high levels of PGAM2, such as skeletal muscle or heart tissue from human, mouse, or rat sources .
Western blot analysis to confirm detection of a band at the expected molecular weight of 29 kDa .
Comparative analysis between tissues with known differential expression (e.g., muscle tissue versus non-muscle tissue).
For advanced validation, using PGAM2 knockdown or knockout samples as negative controls.
Testing cross-reactivity with the homologous PGAM1 protein, which shares 79% sequence identity with PGAM2 .
When selecting validation methods, consider that some PGAM2 antibodies might show reactivity with orthologous proteins from different species including mouse, rat, bovine, frog, and chimpanzee .
Recent studies have revealed that PGAM2, traditionally considered a cytoplasmic glycolytic enzyme, can also localize to the nucleus . When studying PGAM2 nuclear localization:
Use immunofluorescence techniques with verified PGAM2 antibodies, combined with nuclear staining (DAPI, Hoechst, etc.).
Consider measuring the nuclear-to-cytoplasmic fluorescence intensity ratio as a quantitative metric. Control conditions typically show a ratio of approximately 1.28 ± 0.3 .
Be aware that certain conditions can affect PGAM2 nuclear localization. Culturing cells in serum-depleted medium or treating with 1 μM wortmannin (an inhibitor of PI3K) has been shown to decrease nuclear PGAM2, reducing the nuclear-to-cytoplasmic ratio to 0.77 ± 0.19 and 0.9 ± 0.14, respectively .
When investigating residues involved in nuclear localization, site-directed mutagenesis studies have shown that the K146T mutation can decrease nuclear fluorescence intensity by approximately 30% (from 7.98 ± 2.61 in wild-type to 5.73 ± 1.45 in mutant cells) .
For protein-protein interaction studies related to nuclear localization, proximity ligation assays can be employed to detect PGAM2 interactions with proteins like 14-3-3ζ/δ .
PGAM2 has been implicated in cancer progression through both its metabolic and non-metabolic functions . Researchers investigating this relationship should consider:
PGAM protein and activity are upregulated in many cancerous tissues, making it an important cancer metabolism target .
PGAM2 has been shown to directly bind to 14-3-3ζ (tyrosine 3-monooxygenase/tryptophan 5-monooxygenase activation protein zeta), promoting its interaction with phosphorylated BAD, resulting in activation of BCL-xL and subsequent resistance to apoptosis in prostate cancer models .
Use co-immunoprecipitation (co-IP) with PGAM2 antibodies to study its interactions with 14-3-3ζ and other cancer-related proteins.
Combine IHC using PGAM2 antibodies with patient outcome data to evaluate the prognostic value of PGAM2 expression. High PGAM2 expression has been associated with shorter survival and rapid development of enzalutamide resistance in prostate cancer patients .
Consider that PGAM2 overexpression in transgenic mice has been shown to increase tumor size in chemically induced skin carcinogenesis models .
Employ PGAM2 antibodies in chromatin immunoprecipitation (ChIP) assays to investigate potential transcriptional regulation by androgen receptor (AR), as PGAM2 has been shown to be transcriptionally regulated by AR in prostate cancer models .
PGAM2 undergoes several post-translational modifications that regulate its activity and function . Researchers investigating these modifications should:
Be aware that PGAM2 activity is regulated by multiple post-translational modifications including ubiquitination, acetylation, phosphorylation, and sumoylation .
For sumoylation studies, PGAM2 contains two primary SUMO acceptor sites, lysine (K)49 and K176. Mutation of either K to arginine (R) abolishes PGAM2 sumoylation .
Use site-specific antibodies that recognize modified forms of PGAM2, or combine regular PGAM2 antibodies with antibodies against specific modifications (e.g., anti-SUMO antibodies).
Consider that the K176R mutation in PGAM2 impairs myogenic differentiation and affects glycolytic function, as measured by reduced proton efflux rate (PER), glycolytic PER (glycoPER), extracellular acidification rate (ECAR), and oxygen consumption rate (OCR) .
For functional studies of PGAM2 modifications, CRISPR-mediated homologous recombination can be used to generate cells with specific mutations at modification sites, followed by phenotypic analysis using PGAM2 antibodies .
Be aware that the p53/Mdm2 axis promotes proteolysis of PGAM during senescence-inducing stress, suggesting an important regulatory mechanism .
For optimal immunohistochemistry (IHC) results with PGAM2 antibodies:
Tissue preparation: Use formalin-fixed, paraffin-embedded (FFPE) sections or frozen sections depending on the antibody specifications.
Antigen retrieval: Most protocols recommend using TE buffer at pH 9.0 for optimal antigen retrieval, though citrate buffer at pH 6.0 may be used as an alternative depending on the specific antibody .
Dilution range: Typical dilutions for IHC applications range from 1:50 to 1:500, but optimal dilution should be determined empirically for each tissue and antibody combination .
Positive control tissues: Use human breast cancer tissue, mouse skeletal muscle tissue, mouse heart tissue, or mouse brain tissue as positive controls, as these have been validated for PGAM2 expression .
Visualization systems: Both chromogenic (DAB) and fluorescent secondary detection systems are compatible with most PGAM2 antibodies.
Counterstaining: Nuclear counterstains like hematoxylin for chromogenic detection or DAPI for fluorescent detection are recommended to provide cellular context.
Tissue-specific considerations: Be aware that expression levels differ significantly between tissues, with highest expression in skeletal muscle and heart tissues.
PGAM1 and PGAM2 share 79% sequence identity, which can present challenges for antibody specificity . To address these concerns:
Select antibodies raised against specific epitopes that differ between PGAM1 and PGAM2.
Validate antibody specificity using tissues with differential expression patterns: PGAM1 is broadly expressed (brain-form), while PGAM2 is predominantly expressed in muscle tissues (muscle-form) .
Consider using genetic models (knockouts or knockdowns of either isoform) to validate antibody specificity.
Employ Western blotting with recombinant PGAM1 and PGAM2 proteins as controls to assess cross-reactivity.
When studying tissues that express both isoforms, consider using isoform-specific PCR in parallel to confirm expression patterns.
For absolute confirmation, mass spectrometry-based approaches can be used to identify which isoform is being detected in your experimental system.
When investigating PGAM2 interactions using antibody-based methods:
Be aware that proximity ligation assays for PGAM2 interaction with proteins like 14-3-3ζ/δ may detect signals in both nuclear and cytoplasmic compartments .
Not all interactions are detectable by all methods. For example, in one study, proximity ligation assay did not detect the RPLP0-PGAM2 interaction, which may reflect methodological limitations rather than absence of interaction .
Consider that the distance between antibodies interacting with binding partners might be too long for hybridization of nucleic acids associated with the antibodies in proximity ligation assays .
For co-IP studies, optimize lysis conditions carefully, as some PGAM2 interactions may be sensitive to detergent types and concentrations.
Control conditions that affect PGAM2 localization (serum starvation, PI3K inhibition) may also affect its protein-protein interactions .
For studying dynamic interactions, consider live-cell imaging approaches using tagged PGAM2 constructs, validated with antibody-based methods.
Recent research has expanded our understanding of PGAM2's involvement in various diseases:
Glycogen Storage Disease X (GSDX): Naturally occurring mutations in PGAM2 have been etiologically linked to GSDX, highlighting its importance in muscle metabolism .
Cancer progression: PGAM2 has emerging roles in cancer beyond its glycolytic function. In prostate cancer, PGAM2 contributes to enzalutamide resistance through its interaction with 14-3-3ζ, which promotes anti-apoptotic signaling .
Myogenic differentiation: Sumoylation-deficient PGAM2 (K176R mutation) impairs myogenic differentiation in muscle cells, suggesting a role in muscle development and potentially in muscle disorders .
Metabolic reprogramming: PGAM2 supports anti-oxidative defense not only by reducing mitochondrial reactive oxygen species but also via activation of the pentose phosphate pathway .
These diverse roles make PGAM2 antibodies valuable tools for studying multiple disease mechanisms beyond basic glycolysis.
Cutting-edge approaches for investigating PGAM2 include:
CRISPR-mediated homologous recombination: Used to generate precise mutations in PGAM2, such as the K176R knock-in cells, enabling detailed functional studies of specific amino acid residues .
Glycolytic and mitochondrial stress assays: The XF96 Extracellular Flux analyzer can measure parameters like proton efflux rate (PER), glycolytic PER (glycoPER), extracellular acidification rate (ECAR), and oxygen consumption rate (OCR) to assess the metabolic impact of PGAM2 mutations or inhibition .
Genome-wide CRISPR-Cas9 library screens: These have identified PGAM2 as a potential therapeutic target in conditions like enzalutamide-resistant prostate cancer .
High-resolution crystallography: Recent studies have presented the crystal structure of PGAM2, enabling structure-function analyses of this enzyme .
Combined metabolomic and proteomic approaches: These provide comprehensive insights into how PGAM2 alterations affect both metabolic pathways and protein interaction networks.
As PGAM2 emerges as a potential therapeutic target, especially in cancer, antibodies play a crucial role in target validation:
For prostate cancer research, PGAM2 inhibition has been shown to overcome enzalutamide resistance both in vivo and in vitro. PGAM2 antibodies can be used to monitor protein levels in response to treatment and validate target engagement .
In mechanistic studies, PGAM2 antibodies help elucidate how PGAM2 inhibition triggers apoptosis by decreasing levels of the antiapoptotic protein BCL-xL and increasing activity of the proapoptotic protein BAD .
Immunohistochemistry with PGAM2 antibodies on patient-derived xenografts or tissue microarrays can help correlate PGAM2 expression with treatment response.
Antibodies can be used in high-throughput screening assays to identify compounds that disrupt specific PGAM2 interactions, such as with 14-3-3ζ.
For patient stratification studies, PGAM2 antibodies in IHC can help identify patients with high PGAM2 expression who might benefit from targeted therapies in future clinical trials.
The PGAM2 gene is located on chromosome 7 in humans and on chromosome 11 in mice . It consists of three exons and two introns. The gene encodes a dimeric enzyme that exists in different isoforms depending on the tissue type. The muscle-specific isoform is known as PGAM-M, while the brain-specific isoform is PGAM1 .
PGAM2 is essential for efficient energy production in muscle cells. It is involved in the glycolytic pathway, which is a series of reactions that break down glucose to produce energy in the form of adenosine triphosphate (ATP). The enzyme’s activity is crucial for maintaining the energy balance in muscle tissues, especially during intense physical activity .
Mutations in the PGAM2 gene can lead to a condition known as muscle phosphoglycerate mutase deficiency, also referred to as glycogen storage disease type X. This condition is characterized by exercise intolerance, muscle cramps, and myoglobinuria. Patients with this deficiency have a reduced ability to produce ATP through glycolysis, leading to muscle fatigue and weakness .