BDH1 Human

3-Hydroxybutyrate Dehydrogenase, Type 1 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Functional Role in Metabolism

BDH1 regulates ketone body metabolism, critical during fasting or high-fat diets. Its enzymatic activity is rate-limiting for βOHB interconversion, linking lipid catabolism to energy production .

Enzymatic Activity

SubstrateReaction DirectionRole in MetabolismSource
AcetoacetateReduction to βOHBKetogenesis (liver)
(R)-3-hydroxybutyrateOxidation to AcAcKetolysis (peripheral tissues)

Mechanistic Insight: BDH1-mediated βOHB metabolism modulates mitochondrial redox states and reactive oxygen species (ROS) levels, influencing cellular stress responses .

Hepatic Injury and Metabolic Dysfunction

BDH1 overexpression in db/db mice (model of non-alcoholic fatty liver disease) reverses liver damage by activating Nrf2, reducing ROS, and improving metabolic flux (βOHB-AcAc-succinate-fumarate) .

ParameterControl (db/db)BDH1 Overexpression
Hepatic FibrosisElevatedReduced
InflammationHighAttenuated
ApoptosisIncreasedDecreased

Source:

Acute Myeloid Leukemia (AML)

Low BDH1 expression correlates with poor prognosis in AML patients. BDH1 suppresses tumor growth by downregulating oncogenes (HOXA9, MEIS1) and upregulating TP53 .

GeneCorrelation with BDH1Role in AML
HOXA9NegativeOncogenic
MEIS1NegativeOncogenic
TP53PositiveTumor-suppressive

Source:

Diabetic Kidney Disease (DKD)

BDH1 is downregulated in DKD models and patients. Overexpression or βOHB supplementation reduces glucotoxicity/lipotoxicity in renal tubular cells .

ModelBDH1 ExpressionEffect of BDH1 Overexpression
db/db MiceReducedReversed fibrosis/apoptosis
HK-2 Cells (HG/PA)ReducedInhibited ROS/IL-1β secretion

Source:

Cardiac Protection

BDH1 transgenic mice show improved cardiac function under pressure overload (TAC-induced heart failure), with reduced ROS and apoptosis .

ParameterWild-Type (TAC)BDH1 Overexpression (TAC)
LVEFDecreasedPreserved
Lung WeightIncreasedReduced

Source:

Recombinant BDH1 Human Properties

PropertyDescriptionSource
Purity>85% (SDS-PAGE)
Stability4°C (2–4 weeks); -20°C (long-term)
Endotoxin Level<1 EU/µg

Amino Acid Sequence

The recombinant protein includes a His-tag (21 aa) and spans residues 47–343. Key motifs include the catalytic domain (residues 47–343) and lipid-binding regions .

Product Specs

Introduction
BDH1, a member of the short-chain dehydrogenase/reductase gene family, is a mitochondrial membrane enzyme. It functions as a homotetramer and requires lipids, particularly phosphatidylcholine, for optimal activity. BDH1 plays a crucial role in fatty acid catabolism by catalyzing the interconversion of acetoacetate and (R)-3-hydroxybutyrate, the two primary ketone bodies produced during this process.
Description
Recombinant BDH1 Human protein, expressed in E.coli, is a single, non-glycosylated polypeptide chain. It comprises 318 amino acids (residues 47-343) and has a molecular weight of 35.3kDa. The protein includes a 21 amino acid His-tag fused at the N-terminus and is purified using proprietary chromatographic methods.
Physical Appearance
Clear, colorless solution that has been sterilized by filtration.
Formulation
The BDH1 solution is supplied at a concentration of 0.25mg/ml and contains the following components: 20mM Tris-HCl buffer (pH 8.0), 30% glycerol, 0.2M NaCl, 2mM DTT, and 0.1mM PMSF.
Stability
For short-term storage (2-4 weeks), the BDH1 solution should be kept at 4°C. For extended storage, it is recommended to freeze the solution at -20°C. Adding a carrier protein (0.1% HSA or BSA) is advisable for long-term storage. Avoid repeated freeze-thaw cycles to maintain protein integrity.
Purity
The purity of BDH1 is greater than 85% as assessed by SDS-PAGE analysis.
Synonyms
D-beta-hydroxybutyrate dehydrogenase, mitochondrial, BDH, 3-hydroxybutyrate dehydrogenase, BDH1, SDR9C1.
Source
E.coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MASAAEPVGS KAVLVTGCDS GFGFSLAKHL HSKGFLVFAG CLMKDKGHDG VKELDSLNSD RLRTVQLNVC SSEEVEKVVE IVRSSLKDPE KGMWGLVNNA GISTFGEVEF TSLETYKQVA EVNLWGTVRM TKSFLPLIRR AKGRVVNISS MLGRMANPAR SPYCITKFGV EAFSDCLRYE MYPLGVKVSV VEPGNFIAAT SLYSPESIQA IAKKMWEELP EVVRKDYGKK YFDEKIAKME TYCSSGSTDT SPVIDAVTHA LTATTPYTRY HPMDYYWWLR MQIMTHLPGA ISDMIYIR.

Q&A

What is the basic biochemical function of BDH1 in human metabolism?

BDH1 catalyzes the reversible reduction of acetoacetate (AcAc) to 3-hydroxybutyrate (3HB), representing the last enzyme in hepatic ketogenesis and the first enzyme in ketolysis. The enzyme functions as a homotetrameric lipid-requiring enzyme located in the mitochondrial membrane and has a specific requirement for phosphatidylcholine for optimal enzymatic activity . BDH1 belongs to the short-chain dehydrogenase/reductase gene family and plays a crucial role in fatty acid catabolism by facilitating the interconversion of the two major ketone bodies .

Methodologically, researchers investigating BDH1 function should consider using isotope-labeled substrates such as [1-14C] β-hydroxybutyrate to measure ketone body oxidation rates in isolated tissues. Studies have demonstrated that this approach can quantify changes in ketone metabolism, as evidenced by the 2.2-fold upregulation of ketone body oxidation in transverse aortic constriction hearts .

What is the molecular structure of human BDH1 protein and how does it affect experimental design?

Human BDH1 is a full-length protein spanning amino acids 1 to 343. The recombinant form of the protein typically includes a His-tag (MGSSHHHHHH) for purification purposes . The complete amino acid sequence reveals multiple functional domains that contribute to its enzymatic activity.

For experimental design considerations, researchers should note that:

  • The protein forms a homotetrameric structure

  • Requires phosphatidylcholine for optimal activity

  • Contains NAD+/NADH binding sites for redox reactions

When designing studies involving recombinant BDH1, researchers should verify protein purity (>95% is standard for commercial preparations) and ensure low endotoxin levels (<1 EU/μg) to avoid experimental artifacts, particularly in cell culture systems .

How is BDH1 expression regulated in different tissues and under various physiological conditions?

BDH1 expression varies significantly across tissues and can be dramatically altered by physiological conditions. In the liver, BDH1 expression decreases in fatty liver disease models, including transgenic db/db mice . During fasting, wild-type mice show differential regulation of BDH1 compared to fed states, highlighting the importance of nutritional status in BDH1 expression .

In heart failure models, BDH1 expression increases significantly—microarray analysis and mitochondrial proteomics revealed 2.5-fold and 2.8-fold increases, respectively, after transverse aortic constriction . This metabolic adaptation appears to be a protective mechanism against energy starvation during cardiac stress.

Methodologically, researchers should consider multiple approaches for assessing BDH1 expression:

  • qRT-PCR for mRNA quantification

  • Western blotting for protein levels

  • Enzymatic activity assays to confirm functional changes

  • Single-cell analysis to detect tissue heterogeneity in expression patterns

What is the relationship between BDH1 expression and liver cancer prognosis?

BDH1 expression has significant prognostic value in liver cancer. Clinical data analysis has revealed that BDH1 mRNA expression levels correlate with several clinical parameters in liver cancer patients:

Clinical ParameterCorrelation with BDH1P-value
GenderSignificant correlation0.0175
Histologic gradeSignificant correlation0.0001
StageSignificant correlation< 0.001
T classificationSignificant correlation< 0.001

Patients with high BDH1 expression demonstrate longer average relapse-free survival (P = 0.00022). This advantage is particularly pronounced in patients with G1/G2 classification (P = 0.00084) and stage I/II disease (P = 0.0082) .

For researchers investigating BDH1 in cancer contexts, these findings suggest the importance of stratifying patient cohorts by BDH1 expression levels and incorporating multiple clinical parameters in study designs.

What phenotypes are observed in BDH1-deficient animal models?

  • Very low blood 3HB levels despite moderately higher acetoacetate levels

  • Significantly lower total ketone body levels compared to wild-type mice after 16, 24, and 48 hours of fasting

  • Greater hepatic fat content after 24 hours of fasting

These findings indicate that BDH1 deficiency compromises the body's ability to utilize ketone bodies as alternative energy sources during fasting, resulting in hepatic lipid accumulation. The phenotype highlights the critical role of ketogenesis in maintaining lipid energy balance in the liver .

For researchers designing studies with BDH1-deficient models, it's essential to include fasting conditions to reveal the metabolic phenotype, as fed conditions may not expose the underlying defects.

How does BDH1 overexpression affect liver disease progression in metabolic dysfunction-associated fatty liver disease (MAFLD)?

BDH1 overexpression provides significant hepatoprotective effects in MAFLD models. In db/db mice (a model of MAFLD), adeno-associated virus (AAV)-mediated BDH1 overexpression successfully reversed:

  • Abnormal hepatic function indices

  • Fibrosis

  • Inflammation

  • Apoptosis in fatty livers

Mechanistically, BDH1-mediated β-hydroxybutyrate (βOHB) metabolism inhibits reactive oxygen species (ROS) overproduction through activation of Nrf2. This occurs via enhancement of a metabolic flux pathway composed of βOHB-AcAc-succinate-fumarate .

In vitro experiments demonstrated that BDH1 knockdown led to ROS overproduction and ROS-induced inflammation and apoptosis in LO2 cells. Conversely, BDH1 overexpression protected these cells from lipotoxicity by inhibiting ROS overproduction .

These findings identify BDH1 as a potential therapeutic target for MAFLD, suggesting that enhancing ketone body metabolism could be a viable treatment strategy.

What are the optimal methods for measuring BDH1 enzymatic activity in tissue samples?

Measuring BDH1 enzymatic activity requires careful consideration of its reaction kinetics and cofactor requirements. The recommended approach includes:

  • Tissue preparation:

    • Isolation of mitochondria from fresh tissue samples

    • Gentle homogenization to preserve enzyme integrity

    • Suspension in phosphate buffer containing phosphatidylcholine

  • Enzymatic activity assay:

    • Spectrophotometric measurement of NAD+/NADH conversion at 340 nm

    • Reaction mixture containing 3-hydroxybutyrate or acetoacetate substrate

    • Inclusion of phosphatidylcholine to ensure optimal activity

  • Validation methods:

    • Use of [1-14C] β-hydroxybutyrate to measure 14CO2 release, as employed in cardiac studies

    • Parallel measurement of other ketone metabolizing enzymes

    • Confirmation of specificity using BDH1 inhibitors

The rate-limiting nature of BDH1 in ketone metabolism makes activity measurements particularly valuable for understanding metabolic flux through this pathway.

What gene manipulation approaches have been successfully used to study BDH1 function?

Several genetic approaches have proven effective for studying BDH1 function:

  • Global knockout models:

    • Bdh1-deficient mice (Bdh1 KO) have been successfully generated to study systemic effects of BDH1 deficiency

    • These models are particularly useful for studying fasting responses

  • Tissue-specific overexpression:

    • Cardiac-specific Bdh1-overexpressing transgenic mice have been developed to study ketone metabolism in heart failure

    • These models showed 1.7-fold increase in ketone body oxidation

  • Viral vector-mediated overexpression:

    • Adeno-associated virus (AAV)-mediated Bdh1 overexpression has been used to reverse hepatic dysfunction in db/db mice

    • This approach allows for temporal control of overexpression

  • In vitro manipulation:

    • Adenovirus-mediated Bdh1 overexpression in cell culture systems

    • RNA interference-based knockdown approaches

Each approach offers distinct advantages for investigating specific aspects of BDH1 biology, from systemic metabolic effects to tissue-specific functions.

What analytical techniques are most appropriate for detecting changes in ketone body levels in BDH1 research?

Accurate measurement of ketone bodies is essential for BDH1 research. The following analytical approaches are recommended:

  • Clinical chemistry analyzers:

    • Enzymatic assays for 3-hydroxybutyrate and acetoacetate

    • Allow for high-throughput analysis of plasma samples

    • Limited sensitivity compared to mass spectrometry

  • Mass spectrometry-based methods:

    • LC-MS/MS for precise quantification of 3-hydroxybutyrate and acetoacetate

    • Can be coupled with isotope tracers for flux analysis

    • Requires specialized equipment but offers superior sensitivity

  • Nuclear magnetic resonance (NMR) spectroscopy:

    • Non-destructive analysis of ketone bodies in complex matrices

    • Allows simultaneous measurement of multiple metabolites

    • Useful for tissue extract analysis

  • Isotope tracing:

    • Use of [1-14C] β-hydroxybutyrate for measuring oxidation rates

    • Collection and quantification of 14CO2 provides direct measure of ketone utilization

    • As demonstrated in studies measuring 2.2-fold increase in ketone oxidation in cardiac tissue

Researchers should select methods based on their specific experimental questions, sample types, and available equipment.

How does BDH1 influence gene expression programs related to metabolic adaptation?

BDH1 expression levels significantly influence various signaling pathways and gene expression programs. Gene Set Enrichment Analysis (GSEA) of liver cancer patients with high versus low BDH1 expression revealed:

Enriched Pathways in BDH1-High SamplesEnriched Pathways in BDH1-Low Samples
SpermatogenesisAdipogenesis
Epithelial mesenchymal transition (EMT)Oxidative phosphorylation
Apical junctionFatty acid metabolism
Mitotic spindle
E2F targets
G2M checkpoint
MYC signaling
Wnt/β-catenin signaling

These findings suggest that BDH1 may influence fundamental cellular processes beyond its direct role in ketone metabolism . The enrichment of pathways related to cell cycle regulation, differentiation, and signaling in BDH1-high samples points to broader regulatory functions.

For advanced research, investigating the mechanistic link between ketone metabolism and these signaling pathways could reveal novel therapeutic targets. Potential approaches include:

  • ChIP-seq to identify transcription factor binding patterns

  • RNA-seq time course during BDH1 expression changes

  • Metabolomic profiling to connect metabolic shifts with gene expression

What is the role of BDH1 in mediating the crosstalk between ketone metabolism and oxidative stress pathways?

BDH1 plays a critical role in modulating oxidative stress, particularly through its influence on reactive oxygen species (ROS) production and antioxidant defenses.

In cardiac tissue, BDH1 overexpression protected against rotenone-induced ROS production and hydrogen peroxide-induced apoptosis . Similarly, in liver cells, BDH1 overexpression ameliorated oxidative damage through Nrf2 activation .

The molecular mechanisms connecting BDH1 to oxidative stress regulation include:

  • Enhancement of metabolic flux through the βOHB-AcAc-succinate-fumarate pathway

  • Activation of Nrf2, a master regulator of antioxidant response

  • Upregulation of antioxidant enzyme expression

  • Decrease in protein carbonylation and oxidative damage

  • Modulation of histone acetylation patterns

For researchers exploring this connection, experimental approaches should include:

  • Measurement of ROS using flow cytometry with appropriate fluorescent probes

  • Assessment of protein carbonylation as a marker of oxidative damage

  • Analysis of antioxidant enzyme expression and activity

  • Investigation of Nrf2 nuclear translocation and target gene activation

  • Evaluation of mitochondrial function and respiratory chain activity

How might ketone flux through BDH1 contribute to tissue-specific metabolic remodeling in different disease states?

Emerging evidence suggests that BDH1-mediated ketone metabolism contributes significantly to tissue-specific metabolic remodeling in various disease states.

In heart failure, energy substrate preference shifts from fatty acids to glucose, but recent research has uncovered the importance of ketone bodies in this metabolic adaptation. Microarray and proteomics analyses revealed 2.5-fold and 2.8-fold increases in BDH1 expression in hearts after transverse aortic constriction . This upregulation corresponded with a 2.2-fold increase in ketone body oxidation, suggesting that failing hearts increasingly rely on ketones as an alternative fuel source.

In liver disease, particularly MAFLD, BDH1 expression is downregulated, contributing to hepatic dysfunction. Restoration of BDH1 expression ameliorates disease progression, indicating that ketone metabolism is crucial for liver health during metabolic stress .

Recent research also suggests a role for BDH1 in skeletal muscle metabolic remodeling , although the detailed mechanisms require further investigation.

For researchers investigating tissue-specific metabolic remodeling, key considerations include:

  • Differential expression patterns of BDH1 across tissues

  • Tissue-specific responses to metabolic challenges

  • Integration of ketone metabolism with other metabolic pathways

  • Temporal dynamics of metabolic adaptation during disease progression

The development of tissue-specific BDH1 transgenic or knockout models, combined with comprehensive metabolomic profiling, will be crucial for advancing this field.

What are the contradictions in current BDH1 research data and how might they be reconciled?

Several apparent contradictions exist in current BDH1 research:

  • BDH1 expression in cancer vs. metabolic disease:

    • In liver cancer, high BDH1 expression correlates with better prognosis

    • In fatty liver disease, BDH1 expression is downregulated

    Reconciliation approach: Investigate the differential roles of BDH1 in proliferating cancer cells versus metabolically stressed hepatocytes. Cancer cells may leverage ketone metabolism differently than normal cells under metabolic stress.

  • Protective vs. pathological ketone metabolism:

    • BDH1-mediated ketone metabolism protects cardiac tissue from oxidative stress

    • Yet chronic ketosis is often associated with metabolic acidosis

    Reconciliation approach: Examine dose-dependent effects and temporal dynamics of ketone body utilization to determine optimal therapeutic windows.

  • Tissue-specific effects:

    • BDH1 overexpression is protective in heart and liver

    • Effects in other tissues like brain and kidney remain unclear

    Reconciliation approach: Develop tissue-specific models to systematically compare BDH1 function across different organ systems.

  • Genetic vs. environmental regulation:

    • BDH1 expression responds to nutritional states (fasting)

    • Genetic determinants of BDH1 expression/activity are less well characterized

    Reconciliation approach: Conduct genome-wide association studies to identify genetic variants influencing BDH1 expression and correlate with metabolic phenotypes.

Researchers addressing these contradictions should employ multi-omics approaches, combining transcriptomics, proteomics, and metabolomics to construct a more comprehensive understanding of BDH1 biology.

What emerging technologies might advance our understanding of BDH1 function and regulation?

Several cutting-edge technologies hold promise for deepening our understanding of BDH1:

  • CRISPR-based screening approaches:

    • CRISPR activation/interference screens to identify regulators of BDH1 expression

    • Base editing to study the impact of specific mutations on BDH1 function

    • Prime editing for precise modification of BDH1 regulatory elements

  • Single-cell metabolomics:

    • Analysis of ketone metabolism at the single-cell level

    • Identification of cellular heterogeneity in BDH1 expression and activity

    • Correlation with other metabolic parameters

  • In situ metabolic imaging:

    • Development of fluorescent sensors for ketone bodies

    • Real-time visualization of metabolic flux in living tissues

    • Spatial resolution of ketone metabolism in complex tissues

  • Structural biology approaches:

    • Cryo-EM studies of BDH1 protein complexes

    • Structure-based drug design for BDH1 modulators

    • Investigation of allosteric regulation mechanisms

  • Organoid models:

    • Development of liver, heart, and brain organoids for BDH1 research

    • Modeling of genetic disorders affecting ketone metabolism

    • High-throughput screening of therapeutic compounds

These technologies will enable researchers to address fundamental questions about BDH1 function with unprecedented precision and comprehensiveness.

What therapeutic strategies targeting BDH1 show the most promise for treatment of metabolic diseases?

Based on current research, several therapeutic strategies targeting BDH1 show promise:

  • Gene therapy approaches:

    • AAV-mediated BDH1 overexpression has shown efficacy in reversing hepatic dysfunction in db/db mice

    • Tissue-specific delivery systems could optimize therapeutic effects while minimizing off-target consequences

  • Small molecule activators:

    • Development of compounds that enhance BDH1 enzymatic activity

    • Potential for oral administration and dose-dependent modulation

  • Metabolic supplementation:

    • Exogenous ketone supplementation combined with BDH1 modulation

    • Optimization of ketone formulations to maximize therapeutic benefit

  • Dietary interventions:

    • Ketogenic diets to increase substrate availability for BDH1

    • Intermittent fasting regimens to stimulate endogenous ketone production

  • Combination therapies:

    • BDH1 modulation combined with antioxidant therapy

    • Integration with existing treatments for metabolic disorders

The most promising approaches will likely involve personalized strategies based on individual genetic profiles, disease state, and tissue-specific considerations.

How might BDH1 research connect to broader questions in metabolic adaptation and disease resilience?

BDH1 research intersects with several fundamental questions in metabolic biology:

  • Metabolic flexibility and energy homeostasis:

    • BDH1-mediated ketone metabolism represents an adaptive response to energy stress

    • Understanding how organisms switch between energy substrates during various challenges

  • Evolutionary biology of metabolism:

    • Conservation of ketone utilization pathways across species

    • Adaptive advantages of ketone metabolism during periods of resource scarcity

  • Aging and metabolic resilience:

    • Role of ketone bodies as signaling molecules in aging processes

    • Potential for BDH1-targeted interventions to promote healthy aging

  • Metabolic-epigenetic interactions:

    • Influence of ketone bodies on histone acetylation and gene regulation

    • Long-term consequences of altered ketone metabolism on epigenetic programming

  • Systems biology of metabolic networks:

    • Integration of BDH1 function within broader metabolic networks

    • Identification of critical nodes and feedback mechanisms in metabolic regulation

Product Science Overview

Introduction

3-Hydroxybutyrate Dehydrogenase, Type 1 (BDH1), is an enzyme encoded by the BDH1 gene. This enzyme plays a crucial role in the metabolism of ketone bodies, which are produced during the breakdown of fatty acids. BDH1 is a member of the short-chain dehydrogenase/reductase (SDR) family and is primarily located in the mitochondrial membrane .

Structure and Function

BDH1 forms a homotetrameric lipid-requiring enzyme that is embedded in the mitochondrial membrane . It has a specific requirement for phosphatidylcholine to achieve optimal enzymatic activity . The primary function of BDH1 is to catalyze the interconversion of acetoacetate and ®-3-hydroxybutyrate, the two major ketone bodies produced during fatty acid catabolism .

Genetic Information

The BDH1 gene is located on chromosome 3q29 in humans . It encodes a protein that is involved in various metabolic pathways, including ketone body metabolism and general metabolism . The gene has several aliases, including SDR9C1 and D-beta-hydroxybutyrate dehydrogenase .

Clinical Significance

Mutations or dysregulation of the BDH1 gene can be associated with metabolic disorders. For instance, disorders of ketone body transport and chromosome 2Q31.1 duplication syndrome have been linked to BDH1 . The enzyme’s role in ketone body metabolism makes it a potential target for therapeutic interventions in metabolic diseases.

Recombinant BDH1

Recombinant BDH1 refers to the enzyme produced through recombinant DNA technology, which allows for the expression of the human BDH1 gene in various host systems. This recombinant form is used in research to study the enzyme’s function, structure, and potential therapeutic applications.

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.