LDHB

Lactate Dehydrogenase B Recombinant
Shipped with Ice Packs
In Stock

Description

Isoenzyme Distribution and Metabolic Roles

LDHB participates in five isoforms of LDH, each with distinct tissue distributions and metabolic functions :

IsoenzymeSubunit CompositionPrimary TissuesMetabolic Role
LDH-1 (4H)H₄Heart, Brain, RBCsLactate oxidation (Oxidative metabolism)
LDH-2 (3H1M)H₃M₁Reticuloendothelial systemBalanced glycolysis/oxidation
LDH-3 (2H2M)H₂M₂LungsLactate oxidation
LDH-4 (1H3M)H₁M₃Kidneys, Placenta, PancreasLactate production
LDH-5 (4M)M₄Liver, Striated MuscleAnaerobic glycolysis (Warburg effect)

LDHB-rich isoforms (LDH-1, LDH-2, LDH-3) are critical in oxidative environments, while LDHA-rich isoforms (LDH-4, LDH-5) dominate in glycolytic conditions .

LDHB in Ferroptosis and Glutathione Metabolism

LDHB regulates glutathione (GSH) synthesis by modulating SLC7A11 (cystine transporter) expression. Studies in KRAS-mutant lung cancer cells revealed:

  • LDHB knockdown reduces intracellular GSH, cysteine, glutamate, and γ-glutamylcysteine (γ-GC), impairing antioxidant defenses .

  • Mechanism: LDHB silencing downregulates SLC7A11 via STAT1 activation, increasing ferroptosis susceptibility .

  • Therapeutic Implication: Combined LDHB inhibition with SLC7A11/GPX4 blockade triggers synthetic lethality in cancer cells .

MetaboliteLDHB KD vs. ControlFunctional Impact
GSH/GSSG Ratio↓ (2.5-fold)Increased oxidative stress
SLC7A11 mRNA↓ (3.2-fold)Reduced cystine uptake

Selective Inhibitors of LDHB

Recent advancements in LDHB-targeted therapies include:

CompoundEC₅₀ (LDHB)Selectivity (LDHB vs. LDHA)MechanismSource
AXKO-004642 nM>100-foldUncompetitive (allosteric)
Tucatinib8.2 μMModerateUncompetitive
Capmatinib12.4 μMModerateUncompetitive

AXKO-0046, an indole derivative, binds to the LDHB tetramer interface, disrupting substrate binding. Its structure (N-({3-[2-(benzylamino)ethyl]-1H-indol-2-yl}methyl)cycloheptanamine) was resolved via X-ray crystallography, showing displacement of NADH and pyruvate .

Cancer Metabolism

LDHB is critical in cancer cell proliferation and survival:

  • Oxidative tumors (e.g., KRAS-mutant NSCLC): LDHB supports mitochondrial respiration and GSH synthesis, promoting resistance to ferroptosis .

  • Glycolytic tumors: LDHA dominates, but LDHB inhibition may synergize with LDHA-targeted therapies (e.g., AZD3965) .

Challenges and Future Directions

  • Bioavailability: Natural LDH inhibitors (e.g., berberine, carnosic acid) face poor solubility and stability .

  • Targeted Delivery: Development of prodrugs or nanoparticle formulations to enhance tumor-specific uptake.

  • Combination Therapies: Pairing LDHB inhibitors with ferroptosis inducers (e.g., erastin) or GSH biosynthesis blockers (e.g., buthionine sulfoximine) for enhanced anticancer efficacy .

Product Specs

Introduction
Lactate dehydrogenase (LDH) is an enzyme found in many organisms, such as plants and animals. It helps convert pyruvate to lactate and vice versa. This enzyme is important for energy production. In animals, there are different types of LDH.
Description
This product contains the LDH-B gene from chicken heart. The gene was obtained from a cDNA library.
Physical Appearance
Dry powder, free from bacteria.
Formulation
The protein was freeze-dried with a small amount of potassium phosphate. The final concentration is 1 mg/ml.
Solubility
To use, dissolve the LDH powder in sterile 18 megaohm-centimeter H2O to a concentration of at least 100 micrograms/ml. You can then dilute it further with other solutions as needed.
Stability
The LDH powder is stable at room temperature for 3 weeks. For long-term storage, keep it dry and below -18 degrees Celsius. After dissolving the powder, store the solution at 4 degrees Celsius for up to 7 days. For longer storage, keep it below -18 degrees Celsius. Avoid repeated freezing and thawing.
Purity
The purity is over 95%, confirmed by two methods: (a) RP-HPLC and (b) SDS-PAGE.
Biological Activity
The protein's specific activity is 258 units per milligram of protein.
Unit Definition
One unit of activity means 1 micromole of NAD+ is produced per minute at 25 degrees Celsius and pH 7.0. This includes both transaminase activities and a-hydroxyglutarate dehydrogenase activity.
Synonyms
Lactate Dehydrogenase, LDH.
Source
Escherichia Coli.

Q&A

What is the fundamental role of LDHB in cellular metabolism and how does it differ from LDHA?

LDHB (Lactate Dehydrogenase B) plays a pivotal role in cellular metabolism by catalyzing the conversion of lactate to pyruvate, with the concurrent reduction of NAD+ to NADH . Unlike LDHA, which preferentially converts pyruvate to lactate, LDHB has a higher affinity for lactate and preferentially drives the reverse reaction . This functional difference is associated with LDHB's net charge of +1, which contributes to its substrate preference .

How is LDHB expression regulated in normal and cancer cells?

LDHB expression is regulated through multiple mechanisms that vary significantly between normal and malignant tissues. At the transcriptional level, several key factors have been identified as positive regulators of LDHB expression, including mTORC1 (mammalian target of rapamycin complex 1) and STAT3 (signal transducer and activator of transcription 3) . STAT3, a widely recognized oncogenic driver, directly stimulates LDHB transcription, linking inflammatory signaling to metabolic reprogramming in cancer . Additionally, High-mobility group box 2 (HMGB2) participates in transcriptional activation of the LDHB gene .

Epigenetic regulation represents another critical control mechanism for LDHB expression. FGFR1 has been shown to suppress LDHB transcription by promoting DNA methylation in the LDHB promoter region through induction of Tet1, a DNA-binding protein . This promoter methylation is characterized as an epigenetic abnormality rather than a genomic alteration . Post-transcriptional regulation also occurs through miRNAs, with research demonstrating that miR-375 represses both LDHB mRNA and protein levels .

What methods are commonly used to measure LDHB activity in experimental settings?

Several robust methodological approaches have been developed to assess LDHB activity in experimental settings. Colorimetric assays represent a fundamental approach, with NBT (Nitro Blue Tetrazolium) and PMS (Phenazine Methosulfate) serving as primary components in reaction mixtures that enable measurement of LDH activity through spectrophotometric detection . This approach can be conducted as either an end-point assay or as a kinetic assay to monitor enzyme activity in real-time .

For kinetic assessment of LDHB activity, researchers typically employ a reaction mixture comprising CHES buffer (pH 9.6), NaCl, NBT, PMS, gelatin, NAD+, and sodium lactate . The reaction is initiated by adding purified LDHB, and absorbance measurements are taken at regular intervals (typically every 20 seconds) at 570 nm to track the production of NADH . Standard curves using known NADH concentrations enable quantitative determination of enzyme activity .

Zymogram analysis offers an additional technique for investigating LDHB activity and quaternary structure integrity. This NBT/PMS-based approach allows visualization of enzyme activity following native gel electrophoresis, providing insights into whether potential inhibitors disrupt the tetrameric structure of LDHB .

How does LDHB silencing affect DNA damage response pathways in cancer cells?

LDHB silencing significantly impacts DNA damage response pathways, with implications for cancer therapy sensitivity. Transcriptomic analysis of LDHB-silenced NSCLC cell lines (A549 and H358) revealed dysregulation of 1789 genes, prominently including gene sets associated with cell cycle regulation and DNA repair mechanisms . This broad transcriptional reprogramming suggests LDHB functions extend beyond metabolic roles to influence genomic stability.

Mechanistically, LDHB silencing increases H2AX phosphorylation, a well-established surrogate marker of DNA damage . This effect appears to be p53-dependent, as LDHB silencing induces differential cell cycle arrest patterns based on p53 status - arresting at G1/S or G2/M checkpoints depending on the functional p53 status of the cancer cells . Long-term LDHB silencing not only increases basal levels of DNA damage but also sensitizes cancer cells to radiotherapy, resulting in augmented DNA damage and genomic instability as evidenced by elevated H2AX phosphorylation and micronuclei accumulation .

The combination of LDHB silencing with radiotherapy increases protein levels of the senescence marker p21, accompanied by increased phosphorylation of Chk2, indicating persistent unresolved DNA damage . These findings demonstrate that LDHB supports DNA damage repair capacity in cancer cells, while its inhibition creates vulnerabilities that can be exploited therapeutically.

What are the metabolic consequences of LDHB inhibition in cancer cells?

LDHB inhibition profoundly alters cancer cell metabolism beyond simply disrupting lactate-pyruvate conversion. Metabolomic analysis of tumor xenografts with LDHB silencing revealed decreased nucleotide metabolism, particularly affecting both purine and pyrimidine biosynthesis pathways . This metabolic deficiency creates vulnerabilities in cancer cells, especially when faced with DNA-damaging treatments like radiotherapy that require robust nucleotide pools for repair processes.

The metabolic dependency created by LDHB inhibition can be partially rescued through nucleotide supplementation, which attenuates DNA damage caused by combined LDHB silencing and radiotherapy . This finding indicates that LDHB's role in supporting nucleotide metabolism represents a critical mechanism through which it promotes cancer cell survival and therapeutic resistance.

What computational approaches can be used to identify and model specific LDHB inhibitors?

Advanced computational approaches offer powerful tools for identifying and modeling specific LDHB inhibitors. Proteochemometrics (PCM) represents an innovative methodology that simultaneously models the interaction between multiple proteins and multiple ligands, considering both protein and ligand properties in a unified framework . Unlike traditional virtual screening methods, PCM does not rely exclusively on a protein's three-dimensional structure, instead incorporating amino acid sequence information and protein descriptors .

To implement PCM for LDHB inhibitor discovery, researchers have utilized the camb package in the R Studio Server programming environment . This approach begins with retrieving activity data for candidate compounds from databases such as BindingDB, followed by curating the dataset to focus on human activity reported as IC50 values . For improved data distribution, negative logarithmic transformation of IC50 values (pIC50 = -log IC50 × 10-9) is typically applied .

Protein descriptors are generated by first aligning the crystal structure of target proteins (such as using LDHA structure with PDB ID 5W8J as reference for alignment with LDHB structure PDB ID 1I0Z) . Critical residues involved in protein-ligand interactions are identified, typically using a cutoff distance (e.g., 10 Å) from the center of known inhibitors . Machine learning algorithms, including gradient amplification models, random forests, and support vector machines, are then applied as regression methods to develop predictive models of inhibitor activity . These models can be further optimized through ensemble approaches (greedy and stacking optimization) to improve performance .

How do expression patterns of LDHB vary across different cancer types and what are the implications for targeted therapy?

LDHB expression exhibits remarkable heterogeneity across cancer types, with profound implications for targeted therapeutic strategies. In basal-like/triple-negative breast cancer, LDHB shows specific upregulation at both gene and protein levels compared to luminal cancers . This elevated expression is functionally significant, as LDHB inhibition abolishes cell proliferation in vitro and arrests tumor growth in vivo in these breast cancer subtypes .

In lung cancer, LDHB expression correlates with specific oncogenic driver mutations. LDHB is notably upregulated in lung cancer cell lines characterized by RAS pathway activation and is required for in vivo growth of KRAS-mutant lung tumors . High LDHB levels are also observed in other molecular subtypes, particularly those driven by c-MET (found in all examined cell lines) and EGFR (in approximately 38% of tested cell lines) . Clinically, high LDHB expression serves as a significant predictor of shorter survival in patients with lung adenocarcinomas, highlighting its potential as a prognostic biomarker .

Merkel cell carcinoma demonstrates a distinctive pattern, with elevated LDHB mRNA and protein levels in polyomavirus negative (MCPyV-) carcinoma cell lines compared to MCPyV+ cells . Conversely, hepatocellular carcinomas display significantly reduced LDHB levels compared to non-transformed tissues, with this decreased expression predicting unfavorable survival outcomes .

These diverse expression patterns suggest that LDHB-targeted therapies may require a precision medicine approach, focusing on cancer types and molecular subtypes where LDHB overexpression drives tumor progression. Tumor cell lines with high LDHB expression demonstrate significantly greater sensitivity to LDHB inhibition (p=0.00005) compared to those with low expression, indicating that LDHB expression levels could serve as a predictive biomarker for response to LDHB-targeted therapies .

What screening strategies are effective for identifying selective LDHB inhibitors?

Identifying selective LDHB inhibitors requires sophisticated screening strategies that integrate computational and experimental approaches. In silico screening represents a powerful initial approach, leveraging databases of commercially available compounds such as the Zinc database to identify potential inhibitors . This approach begins with a broad compound library (potentially exceeding 1.8 billion substances) that can be narrowed using relevant filters (such as bioactivity and commercial availability) .

Virtual screening with Autodock Vina software enables efficient evaluation of potential inhibitors against the human LDHB structure (such as PDB: 1T2F) . This process determines binding affinities and groups resulting poses based on conformations . Top candidates can be further evaluated through re-docking into predetermined receptor regions, with root mean square deviation (RMSD) calculations comparing the re-docked complex to reference co-crystallized complexes .

Complementary to structure-based approaches, ligand-based screening through libraries like CHEMBL_act (SwissSimilarity) identifies additional candidate molecules based on similarity to known bioactive compounds . Setting appropriate similarity thresholds (such as 0.9 probability of likeness) helps balance the breadth and specificity of the candidate pool .

Experimental validation follows computational screening, with colorimetric enzymatic assays enabling quantitative assessment of inhibitory potency through IC50 determination . GraphPad Prism software facilitates calculation of these values by fitting dose-response curves using non-linear regression analysis based on raw enzymatic assay data .

How can researchers effectively validate LDHB as a therapeutic target in specific cancer types?

Validating LDHB as a therapeutic target requires a multi-dimensional approach encompassing expression analysis, functional studies, and preclinical models. Clinical correlation studies represent an essential first step, examining LDHB expression levels in tumor versus normal tissues and correlating these with patient outcomes . These analyses should be stratified by molecular subtypes, as LDHB's significance varies across cancer genotypes, with particular relevance in certain contexts like KRAS-mutant lung cancers and triple-negative breast cancers .

Genetic manipulation through RNA interference or CRISPR-based approaches provides critical functional validation. Short-term and long-term LDHB silencing studies reveal differential effects on cancer cell proliferation, metabolism, and therapeutic sensitivity . These studies should assess phenotypic consequences across multiple cancer cell lines representing diverse molecular subtypes to identify contexts where LDHB dependency is strongest .

Metabolic profiling of LDHB-silenced cells offers mechanistic insights into how LDHB supports cancer progression. Metabolomic analysis of tumor xenografts has revealed that LDHB silencing decreases nucleotide metabolism, particularly affecting purine and pyrimidine biosynthesis pathways . These metabolic vulnerabilities can be further validated through rescue experiments, such as nucleotide supplementation studies that demonstrate partial attenuation of phenotypes caused by LDHB inhibition .

Combination therapy studies provide particularly compelling target validation, especially when LDHB inhibition sensitizes cancer cells to standard treatments. For example, LDHB silencing has been shown to enhance the effects of radiotherapy in lung cancer models by impairing nucleotide metabolism and promoting persistent DNA damage . Such synergistic interactions highlight the potential clinical utility of LDHB-targeted approaches.

What are the best experimental designs to investigate the differential roles of LDHA versus LDHB in cancer metabolism?

Investigating the differential roles of LDHA versus LDHB in cancer metabolism requires carefully designed experimental approaches that account for their potential functional redundancy. Single and double knockdown/knockout systems represent foundational tools for dissecting isoform-specific functions . While individual LDHA or LDHB knockouts may not strongly reduce lactate secretion due to compensatory mechanisms, double LDHA/B knockdowns enable comprehensive assessment of their collective contribution to glycolytic activity and lactic acid production .

Isoform-specific inhibitors provide complementary approaches to genetic manipulation. While several inhibitors target LDHA with or without activity against LDHB, the development of LDHB-specific inhibitors like AXKO-0046 has created new opportunities to dissect isoform-specific functions . Comparative studies using selective versus pan-LDH inhibitors can illuminate the relative contributions of each isoform to cancer cell metabolism and survival.

Metabolic flux analysis using isotope-labeled substrates offers dynamic insights into how LDHA and LDHB differentially affect metabolic pathways. By tracing the fate of labeled glucose, lactate, or pyruvate in cells with isoform-specific manipulations, researchers can determine how each enzyme influences carbon routing through glycolysis, the TCA cycle, and biosynthetic pathways.

Context-dependent studies across diverse cancer models are essential, as the relative importance of LDHA versus LDHB varies by cancer type and molecular subtype . For instance, LDHB is particularly critical in basal-like/triple-negative breast cancers and certain lung cancer subtypes, whereas other contexts may rely more heavily on LDHA . Comprehensive experimental designs should include multiple cell lines representing different cancer types and molecular subtypes to capture this heterogeneity.

What are the major technical challenges in developing selective LDHB inhibitors?

Developing selective LDHB inhibitors presents several significant technical challenges. The high sequence and structural similarity between LDHA and LDHB represents the foremost obstacle, with both enzymes sharing substantial homology in their active sites . This similarity complicates the design of compounds that selectively target LDHB without affecting LDHA, requiring sophisticated approaches to identify subtle structural differences that can be exploited for selectivity.

The limited availability of LDHB-specific crystal structures with bound inhibitors hampers structure-based drug design efforts . While proteochemometrics methods that incorporate protein descriptors beyond three-dimensional structure offer promising alternatives, they still face challenges in accurately modeling the interaction space between inhibitors and highly similar enzyme isoforms .

Another technical hurdle involves developing assays with sufficient sensitivity and specificity to differentiate LDHB inhibition from LDHA inhibition. Current colorimetric methods measuring LDH activity may not readily distinguish between isoform-specific effects without additional purification or separation steps . Zymogram analysis offers one approach to visualize isoform-specific inhibition, but higher-throughput methods would accelerate screening efforts .

The physicochemical properties required for LDHB inhibition present additional challenges. Many potential inhibitors identified through virtual screening may possess unfavorable pharmacokinetic properties or limited cell permeability . Optimizing these properties while maintaining selectivity for LDHB over LDHA requires extensive medicinal chemistry efforts guided by structure-activity relationship studies.

How does the metabolic interplay between LDHB and other enzymes affect cancer cell adaptation to therapy?

The metabolic interplay between LDHB and other enzymes creates complex adaptation mechanisms that influence cancer therapeutic response. LDHB functions within a broader metabolic network where inhibition of one enzyme often leads to compensatory changes in related pathways . Understanding these adaptive responses is critical for designing effective therapeutic strategies targeting cancer metabolism.

LDHB's role in supporting mitochondrial metabolism creates interdependencies with the tricarboxylic acid (TCA) cycle and electron transport chain . When LDHB is inhibited, cancer cells may activate alternative metabolic pathways to maintain redox balance and energy production. These adaptations can involve upregulation of glutaminolysis, fatty acid oxidation, or enhanced glucose uptake through alternative transporters .

The relationship between LDHB and nucleotide metabolism represents a particularly important metabolic node affecting therapeutic response . LDHB silencing decreases nucleotide metabolism, particularly purine and pyrimidine biosynthesis, creating vulnerabilities when cancer cells face DNA-damaging treatments like radiotherapy . This metabolic deficiency impairs DNA damage repair capacity, leading to persistent DNA damage and enhanced therapeutic efficacy .

Metabolic adaptation to LDHB inhibition likely varies across cancer genetic backgrounds. Cell lines with certain oncogenic drivers (like KRAS mutations) show particular dependency on LDHB , suggesting that genetic context influences the metabolic wiring and adaptive capacity of cancer cells. Comprehensive metabolomic profiling across diverse genetic backgrounds would provide deeper insights into these context-dependent adaptations and identify potential combination strategies to prevent resistance.

What research approaches can resolve contradictory findings regarding LDHB's role in different cancer types?

Resolving contradictory findings regarding LDHB's role across cancer types requires integrated research approaches that account for biological context and methodological differences. Molecular subtyping represents a critical first step, as LDHB's function appears highly context-dependent . Comprehensive genomic and proteomic profiling should accompany LDHB functional studies to identify molecular signatures that predict LDHB dependency, moving beyond broad cancer type classifications to more precise molecular taxonomies.

Standardized experimental methodologies would facilitate more direct comparisons across studies. This includes consistent approaches for measuring LDHB expression (distinguishing between mRNA and protein levels), enzyme activity assays with clear specificity for LDHB over LDHA, and well-defined functional endpoints when assessing phenotypic consequences of LDHB manipulation .

Meta-analyses of existing data, combining results across multiple studies with careful attention to methodological differences, can help identify patterns that explain seemingly contradictory findings. Statistical approaches that account for interstudy heterogeneity and publication bias would strengthen these analyses and potentially resolve apparent contradictions in the literature.

Product Science Overview

Structure and Function

LDHB is one of the isoforms of lactate dehydrogenase, which exists as a tetramer composed of different combinations of two subunits: LDHA and LDHB. The B isoform is predominantly found in the heart and red blood cells, where it facilitates the conversion of lactate to pyruvate, a key step in the Cori cycle. This cycle is vital for maintaining glucose homeostasis in the body.

Recombinant Production

Recombinant LDHB is produced using genetic engineering techniques. The gene encoding LDHB is cloned into an expression vector, which is then introduced into a host organism, typically E. coli. The host cells are cultured, and the recombinant protein is expressed and subsequently purified. This method allows for the production of large quantities of LDHB with high purity and activity .

Applications

Recombinant LDHB has several applications in research and clinical diagnostics:

  1. Cancer Research: LDHB is a marker for certain types of cancer. Its expression levels can provide insights into the metabolic state of cancer cells, which often exhibit altered glycolysis pathways.
  2. Cardiac Health: Given its abundance in the heart, LDHB levels can be indicative of cardiac health and are used in the diagnosis of myocardial infarction.
  3. Biochemical Studies: LDHB is used in various biochemical assays to study enzyme kinetics and metabolic pathways.
Clinical Significance

The altered metabolism in cancer cells, known as the Warburg effect, often involves a shift to aerobic glycolysis with increased lactate production. LDHB plays a significant role in this process, making it a valuable target for cancer diagnostics and therapeutics .

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.