ENO2’s primary role is glycolysis, but it also participates in non-metabolic processes:
Converts 2-phosphoglycerate to phosphoenolpyruvate, enabling ATP production and lactate synthesis .
Upregulation drives the Warburg effect, a hallmark of cancer metabolism .
Neurotrophic Effects: Binds to neurons in a calcium-dependent manner, promoting survival .
Tumor Progression: Activates epithelial-mesenchymal transition (EMT) via YAP1 signaling in colorectal cancer .
Immune Modulation: Correlates with M2 macrophages and N2 neutrophils in renal cell carcinoma (RCC) .
ENO2 serves as a tumor biomarker and therapeutic target across malignancies:
Hypoxia Response: HIF-1α upregulates ENO2 in ccRCC, linking glycolysis to tumor microenvironments .
Prognostic Value: Serum ENO2 levels correlate with recurrence and survival in melanoma and pancreatic endocrine tumors .
EMT Regulation: Knockdown in RCC inhibits migration and invasion by suppressing EMT markers (e.g., N-cadherin, vimentin) .
Acetylation at K394: HDAC3 deacetylates ENO2, enhancing enzymatic activity, while PCAF-mediated acetylation inhibits it .
Glycolysis Activation: Promotes glucose uptake (via GLUT1) and lactate production (via LDHA) .
Knockdown Models: Silencing ENO2 reduces tumor growth in ccRCC and PDAC xenografts .
Ferroptosis Induction: Inhibition of ENO2 in ccRCC triggers lipid peroxidation and mitochondrial dysfunction .
Small-Molecule Inhibitors: ENO2 inhibitors (e.g., heterocyclic compounds) suppress glioblastoma growth .
Biomarker Potential: Serum ENO2 levels may predict response to immune checkpoint blockade in ccRCC .
Metabolic Reprogramming: Targeting ENO2-HIF-1α axis could disrupt hypoxia-driven glycolysis in solid tumors .
Context-Dependent Roles: ENO2’s dual glycolytic and signaling functions require tissue-specific studies.
Therapeutic Resistance: Compensatory mechanisms (e.g., ENO1 upregulation) may limit ENO2-targeted therapies.
Diagnostic Utility: Standardizing serum ENO2 thresholds for clinical use across cancers.
ENO2, a 433-amino acid protein, exists as two enolase isoenzymes (γγ and αγ) and functions as a critical glycolytic enzyme that catalyzes the conversion of β-glycerol phosphate to acetone dihydroxyphosphate . This conversion represents a key step in the glycolytic pathway, which is essential for cellular energy production. ENO2 is primarily expressed in neurons and neuroendocrine cells, although expression has also been documented in red blood cells, platelets, and tissues including breast, prostate, and uterus . Unlike its isoform ENO1, which is more widely expressed, ENO2 exhibits a more restricted tissue distribution pattern, indicating potential tissue-specific functions beyond its canonical role in glycolysis. The glycolytic function of ENO2 is particularly significant in cancer research, as enhanced glycolysis can supply energy required for tumor cell proliferation and survival under stress conditions .
For accurate quantification of ENO2 protein levels, several methodological approaches have been validated in research settings. Enzyme-linked immunosorbent assay (ELISA) represents a highly sensitive and specific method for ENO2 protein detection in plasma and other biological fluids. As demonstrated in studies on autism, commercially available ELISA kits (such as those from R&D Systems) can effectively quantify ENO2 levels with high precision . The microplate reader should be set to 450 nm for optimal detection, with readings taken within 30 minutes to ensure accuracy . For tissue samples, immunohistochemistry provides spatial information about ENO2 expression patterns. At the mRNA level, quantitative real-time PCR (RT-qPCR) using validated primer sets has been effectively employed, with β-actin serving as an appropriate internal control gene . The recommended primer sequences for ENO2 detection include ENO2-F (5′-GGGAACTCAGACCTCATCCTG-3′) and ENO2-R (5′-CTTGTTGCCAGCATGAGAGC-3′) . For optimal results, PCR conditions should follow a program of 95°C for 5 minutes, followed by 40 cycles of 95°C for 30 seconds and 59°C for 30 seconds, with fluorescent signal collection at 59°C .
The enolase family consists of three distinct isoforms: ENO1 (α-enolase), ENO2 (γ-enolase or neuron-specific enolase), and ENO3 (β-enolase), each with unique tissue distribution patterns and functional characteristics . ENO1 is ubiquitously expressed across tissues and serves as the predominant isoform in most cell types. ENO2 is primarily expressed in neurons and neuroendocrine cells, while ENO3 shows enrichment in muscle tissues . These differential expression patterns necessitate isoform-specific approaches in research design. When studying ENO2, researchers must employ highly specific antibodies that can distinguish between the three isoforms to avoid cross-reactivity in protein detection methods. In cancer research, the relationship between ENO1 and ENO2 is particularly important due to the phenomenon of collateral lethality observed in glioblastoma (GBM), where homozygous deletion of ENO1 induces compensatory upregulation of ENO2 . This unique relationship indicates that experimental designs targeting ENO2 must consider the expression status of ENO1, especially in cancer models. In functional studies, isoform-specific knockdown using siRNA or shRNA targeting unique regions of ENO2 is essential to ensure specificity without affecting other enolase isoforms .
ENO2's role in cancer progression extends significantly beyond its canonical glycolytic function. Research has uncovered multiple non-glycolytic mechanisms through which ENO2 promotes cancer development and progression. In colorectal cancer (CRC), ENO2 has been shown to activate the YAP1-induced epithelial-mesenchymal transition (EMT) process, thereby promoting migration, invasion, and metastasis of CRC cells . This reveals that ENO2 can influence key signaling pathways that regulate cellular plasticity and invasive properties. In BRAF V600E-mutated CRC, ENO2 not only promotes proliferation and migration but also contributes to vemurafenib resistance, suggesting a role in drug resistance mechanisms . In glioblastoma, ENO2 is upregulated under cellular stress conditions such as serum starvation and hypoxia, and knockdown of ENO2 suppresses migration and sensitizes cells to hypoxia, radiotherapy, and chemotherapy . This indicates that ENO2 plays a crucial role in stress adaptation and therapy resistance. The C-terminal peptide of ENO2 has been reported to regulate cytoskeletal structure through RhoA kinase, influencing tumor cell metastasis . Additionally, in clear cell renal cell carcinoma (ccRCC), ENO2 has been linked to tumor progression through its effects on glucose metabolism, with knockdown experiments demonstrating reduced glucose utilization, lactate production, and intracellular ATP generation, ultimately inhibiting proliferation, migration, and invasion capabilities .
Epigenetic modifications, particularly DNA methylation, play a crucial role in regulating ENO2 expression in neurological disorders such as autism. Research has identified hypermethylation of the ENO2 gene promoter region as a significant epigenetic alteration in autistic samples . Methodologically, several complementary techniques can be employed to investigate these epigenetic modifications. Methylated DNA immunoprecipitation (MeDIP) chip analysis provides a genome-wide screening approach to identify differentially methylated regions . For validation and more detailed analysis, bisulfite sequencing PCR (BSP) represents the gold standard for methylation analysis, allowing for single-nucleotide resolution of methylation patterns across specific genomic regions . In the autism study, BSP confirmed hypermethylation of ENO2 within the promoter region in 14.5% (19/131) of autistic samples . To establish the functional consequences of these methylation changes, researchers should employ expression analysis using RT-qPCR for mRNA levels and ELISA for protein quantification . In the autism cohort, the mean ENO2 RNA level in hypermethylated samples was reduced by approximately 70% relative to controls, while protein expression was reduced by about 50% (15.18 ± 3.51 μg/l in autistic samples versus 33.86 ± 8.16 μg/l in controls) . For mechanistic insights, luciferase reporter assays using constructs containing the ENO2 promoter region can help establish a causal relationship between methylation status and transcriptional activity . Additionally, chromatin immunoprecipitation (ChIP) assays can be employed to investigate histone modifications associated with ENO2 regulation, providing a more comprehensive view of the epigenetic landscape.
ENO2 contributes to therapeutic resistance in cancer through multiple molecular mechanisms that extend beyond its primary metabolic functions. In glioblastoma (GBM), ENO2 is upregulated under cellular stress conditions such as hypoxia and serum starvation, and knockdown studies have demonstrated that ENO2 silencing sensitizes GBM cells to hypoxia, radiotherapy, and chemotherapy . This suggests that ENO2 plays a pivotal role in stress adaptation pathways that protect cancer cells from therapeutic insults. In BRAF V600E-mutated colorectal cancer, ENO2 has been specifically linked to vemurafenib resistance, with experimental evidence showing that BRAF V600E-mutated cells exhibit greater dependency on ENO2 compared to wild-type cells . Mechanistically, ENO2's role in glucose metabolism and ATP production appears to be critical for maintaining energy homeostasis in cancer cells during therapeutic stress. In clear cell renal cell carcinoma (ccRCC), knockdown of ENO2 inhibits glucose utilization, lactate production, and intracellular ATP generation, which are essential for cancer cell survival and proliferation under stress conditions . Additionally, ENO2's influence on cellular signaling pathways, including its activation of the YAP1-induced epithelial-mesenchymal transition (EMT) process in colorectal cancer, may contribute to therapy resistance by promoting cellular plasticity and survival mechanisms . Furthermore, ENO2's regulation of cytoskeletal structure through RhoA kinase may enhance cell motility and invasiveness, allowing cancer cells to evade therapeutic pressures . These diverse mechanisms highlight ENO2 as a multifaceted contributor to therapy resistance and suggest that targeting ENO2 may enhance the efficacy of existing cancer therapies across multiple cancer types.
Designing and validating robust ENO2 knockdown or overexpression models requires careful consideration of several methodological aspects. For transient knockdown, small interfering RNA (siRNA) targeting ENO2 has been successfully employed in multiple studies . When designing siRNA sequences, researchers should target regions unique to ENO2 to avoid cross-reactivity with other enolase isoforms (ENO1 and ENO3). For stable knockdown, which is preferable for long-term studies and in vivo experiments, small hairpin RNA (shRNA) delivered via lentiviral vectors represents an effective approach . In a GBM study, researchers successfully established intracranial GBM xenograft models using GBM cells harboring ENO2 or scrambled shRNA, demonstrating the feasibility of this approach for in vivo studies . For overexpression models, the full-length ENO2 coding sequence should be cloned into appropriate expression vectors with strong promoters. Validation of knockdown or overexpression efficiency is critically important and should include both mRNA and protein-level assessments. For mRNA validation, RT-qPCR should be performed using validated primer pairs, with recommended sequences including ENO2-F (5′-GGGAACTCAGACCTCATCCTG-3′) and ENO2-R (5′-CTTGTTGCCAGCATGAGAGC-3′) . At the protein level, Western blot and ELISA provide quantitative measures of ENO2 expression changes . Functional validation should include assays relevant to ENO2's known functions, such as glucose consumption, lactate production, and ATP generation assays . Additionally, phenotypic assays such as proliferation (CCK8), migration, and invasion (Transwell) should be performed to confirm the functional impact of ENO2 modulation . For in vivo validation, subcutaneous or orthotopic tumor models in appropriate mouse strains should be considered, with careful monitoring of tumor growth parameters, as demonstrated in the ccRCC study where ENO2 knockdown significantly suppressed tumor weight and volume in nude mice .
Selecting appropriate in vivo models for ENO2 research requires careful consideration of the specific research questions and disease context. For cancer studies, xenograft models using immunodeficient mice represent a well-validated approach. In ccRCC research, male athymic nude mice (aged 5 weeks) were successfully used to establish subcutaneous tumor models using A498 cells with stable ENO2 knockdown or control cells . For this model, researchers should monitor tumor growth by regularly measuring dimensions using calipers, with tumor volume calculated using the formula V = 0.5 × length × width² . For more physiologically relevant models, orthotopic implantation may be preferable. In glioblastoma research, researchers effectively implanted GBM cells harboring ENO2 or scrambled shRNA into the right frontal cortex of NOD/SCID mice to establish intracranial GBM xenograft models, demonstrating that ENO2 knockdown significantly extended mouse survival by 7.2% . For studying ENO2 in neurodevelopmental contexts, transgenic mouse models with conditional ENO2 knockout or overexpression in specific neural cell populations would provide valuable insights. When designing in vivo experiments, researchers should carefully consider sample size calculations to ensure adequate statistical power, with the ccRCC study utilizing 12 mice divided between experimental and control groups . Endpoint analyses should be comprehensive, including not only tumor volume measurements but also histological assessment, immunohistochemical analysis of ENO2 expression, and metabolic profiling. For translational relevance, patient-derived xenograft (PDX) models may provide advantages over cell line-based models, particularly for evaluating ENO2-targeted therapies. All procedures involving animals must comply with institutional ethical guidelines and animal welfare regulations, as noted in the ccRCC study where procedures were approved by the Hubei Provincial Experimental Animal Research Centre .
Research has provided compelling evidence for ENO2 as a potential biomarker in autism spectrum disorder (ASD). A key study identified hypermethylation of the ENO2 gene promoter region in 14.5% (19/131) of autistic samples, confirmed through bisulfite sequencing PCR (BSP) . This epigenetic alteration was functionally significant, as the mean ENO2 RNA level in the 19 hypermethylated autistic samples was reduced by approximately 70% compared to controls . At the protein level, ELISA analysis revealed that the average ENO2 protein expression in these autistic samples (15.18 ± 3.51 μg/l) was about half of that observed in controls (33.86 ± 8.16 μg/l) . These findings suggest that reduced ENO2 expression may serve as a biomarker for a specific subset of children with autism . The study included a diverse cohort of 131 autistic subjects (96 males and 30 females) across various age groups (from 3 to 12 years), enhancing the generalizability of the findings . From a methodological perspective, researchers employed a comprehensive approach including initial screening with methylated DNA immunoprecipitation (MeDIP) chip analysis, validation with BSP, and functional confirmation with RT-qPCR and ELISA . This multi-level validation strengthens the evidence for ENO2 as a potential biomarker. The specificity of this biomarker for a subset (14.5%) rather than all autistic children highlights the heterogeneity of autism and suggests that ENO2 might help identify a specific molecular subtype of ASD . For clinical application, further research is needed to determine whether ENO2 alterations correlate with specific clinical features, severity, or treatment response in autism. Additionally, longitudinal studies would be valuable to assess whether ENO2 alterations are stable over time or change with development or intervention.
Therapeutic strategies targeting ENO2 are being explored across multiple pathological conditions, with cancer being the primary focus. Several approaches show promise for clinical development. Direct inhibition of ENO2 enzymatic activity represents one strategy, with small molecule inhibitors being developed to block its glycolytic function . These inhibitors could potentially disrupt cancer cell metabolism, particularly in highly glycolytic tumors that depend on ENO2 for energy production . The collateral lethality phenomenon observed in glioblastoma offers a precision medicine approach, where ENO2 inhibition could be selectively lethal to tumors with ENO1 deletion while sparing normal tissues . Preclinical evidence supports this strategy, as selective knockdown of ENO2 effectively inhibited the growth and survival of ENO1-deleted GBM cells but not ENO1-intact cells . In vivo validation in GBM xenograft models demonstrated that ENO2 knockdown extended mouse survival by 7.2%, providing proof-of-concept for this approach . Beyond direct inhibition, targeting ENO2's non-glycolytic functions represents another strategy. In colorectal cancer, ENO2 promotes cancer progression by activating the YAP1-induced EMT process, suggesting that disrupting this interaction could inhibit metastasis . For BRAF V600E-mutated colorectal cancer, combining ENO2 inhibitors with vemurafenib might overcome resistance to BRAF inhibitors . In neurological conditions like autism, where ENO2 is hypermethylated and underexpressed, epigenetic modifiers that reduce methylation might restore normal ENO2 expression . For clinical development, companion diagnostics will be essential to identify patients most likely to benefit from ENO2-targeted therapies. For example, ENO1 deletion status could identify GBM patients suitable for ENO2 inhibition therapy . Additionally, monitoring ENO2 levels during treatment could serve as a pharmacodynamic marker to assess therapeutic efficacy .
Addressing contradictory findings regarding ENO2 expression and function requires a systematic approach to reconcile discrepancies and understand context-dependent effects. One key strategy is to thoroughly consider tissue and disease specificity. While ENO2 is upregulated in various cancers, including lung cancer, ccRCC, and colorectal cancer , it is hypermethylated and downregulated in autism . These opposing patterns suggest that ENO2 regulation is highly context-dependent and may serve different functions in different pathological conditions. Researchers should explicitly acknowledge these contradictions and design experiments to test hypotheses about the underlying mechanisms driving these differences. Methodological differences can contribute significantly to contradictory findings. Variations in sample processing, detection methods, and analytical approaches can yield seemingly conflicting results. To address this, researchers should employ multiple complementary techniques (e.g., RT-qPCR, Western blot, immunohistochemistry, ELISA) to validate expression findings . In the autism study, researchers confirmed hypermethylation through both MeDIP chip analysis and BSP, while validating expression changes at both mRNA (RT-qPCR) and protein (ELISA) levels . The heterogeneity within disease populations also contributes to contradictory findings. In autism, ENO2 hypermethylation was found in 14.5% of cases, not all patients , while in cancer, ENO2 expression may vary based on tumor subtype, stage, or molecular features . Researchers should employ detailed subgroup analyses and consider molecular stratification to identify patterns within heterogeneous populations. Meta-analyses and systematic reviews can help synthesize conflicting findings across studies, while replication in independent cohorts strengthens evidence for specific observations. Ultimately, contradictions often reflect the complex biology of ENO2, which likely has distinct and sometimes opposing functions depending on cellular context, developmental stage, and disease state.
Gamma-enolase is a phosphopyruvate hydratase that plays a crucial role in the glycolytic pathway. It catalyzes the conversion of 2-phosphoglycerate to phosphoenolpyruvate, a key step in glycolysis . This reaction is essential for the production of ATP, the primary energy currency of the cell .
In addition to its role in glycolysis, gamma-enolase has several other functions:
Research on gamma-enolase continues to uncover its multifunctional roles in the central nervous system. Its neuroprotective properties and involvement in neuronal survival make it a promising target for therapeutic interventions in neurodegenerative diseases . However, the effectiveness of enolase inhibitors as a therapeutic strategy is still under debate .