CYB5A Human

Cytochrome B5 Type A Human Recombinant
Shipped with Ice Packs
In Stock

Description

Overview of CYB5A (Cytochrome B5 Type A)

CYB5A (Cytochrome B5 Type A) is a human gene encoding a membrane-bound hemoprotein that functions as an electron carrier in redox reactions. It plays critical roles in lipid metabolism, steroidogenesis, and detoxification processes . CYB5A is implicated in diverse physiological and pathological processes, including cancer metastasis, rheumatoid arthritis (RA), and hereditary methemoglobinemia .

Genomic Localization

  • Chromosome: 18 (positions 74,250,847–74,292,016 bp, GRCh38) .

  • Gene IDs:

    DatabaseIdentifier
    HGNC2570
    EnsemblENSG00000166347
    MIM613218

Protein Characteristics

  • Isoforms: Two isoforms generated by alternative splicing:

    • Isoform 1: Membrane-bound to the endoplasmic reticulum (ER) via a C-terminal transmembrane domain.

    • Isoform 2: Cytoplasmic localization .

  • Function: Facilitates electron transfer to cytochrome P450 enzymes, enhancing steroid 17,20-lyase activity and androgen synthesis .

Tissue-Specific Expression

CYB5A is most abundant in metabolically active tissues:

  • High: Liver, kidney, adrenal gland .

  • Moderate: Lung, breast, endometrium .

Somatic Mutations in Cancer

CDS MutationProtein MutationAssociated Tissue
c.375G>Cp.L125FUrinary tract
c.232A>Cp.T78PBreast
c.383G>Ap.R128HStomach
c.339G>Ap.W113*Liver
(Source: Atlas of Genetics and Cytogenetics in Oncology and Haematology)

Hepatocellular Carcinoma (HCC)

  • Mechanism: CYB5A suppresses metastasis by inhibiting the JAK1/STAT3 pathway and inducing autophagy .

  • Clinical Correlation:

    • Low CYB5A expression correlates with advanced tumor stage (p<0.01p < 0.01) and poor survival .

    • Overexpression reduces HCC cell migration by 60% in vitro .

Rheumatoid Arthritis (RA)

  • Genetic Link: The rs1790834 SNP in CYB5A increases androgen synthesis, reducing RA risk in women (OR = 0.63, p=4.1×103p = 4.1 \times 10^{-3}) .

  • Functional Impact: The protective allele doubles CYB5A mRNA expression, enhancing 17,20-lyase activity 2–3 fold .

Hereditary Methemoglobinemia

  • Role: CYB5A mutations impair methemoglobin reduction, causing type IV hereditary methemoglobinemia .

CYB5A and Autophagy in Cancer

  • A 2022 study demonstrated that CYB5A overexpression increases autophagic flux (e.g., LC3-II/I ratio by 2.5-fold) under hypoxia, inhibiting intrahepatic and pulmonary metastasis in HCC xenografts .

Therapeutic Implications

  • RA: Targeting CYB5A-mediated androgen synthesis could offer sex-specific therapies .

  • HCC: CYB5A restoration via gene therapy is being explored to counteract metastasis .

Product Specs

Introduction
Cytochrome b5 isoform 1 (CYB5A) is a member of the cytochrome b5 family. It is a membrane-bound hemoprotein that acts as an electron carrier for several membrane-bound oxygenases. Genetic defects in CYB5A can lead to CYB5A-related methemoglobinemia (METHB-CYB5A). METHB-CYB5A is a type of methemoglobinemia, a blood disorder characterized by an excess of methemoglobin in red blood cells. This excess hinders the blood's oxygen-carrying capacity, resulting in cyanosis and hypoxia.
Description
Recombinant human CYB5A, produced in E. coli, is a single, non-glycosylated polypeptide chain. It consists of 132 amino acids (with amino acids 1 to 108 being the actual CYB5A sequence) and has a molecular weight of 14.9 kDa. A 24-amino acid His-tag is fused to the N-terminus of CYB5A for purification purposes, which is achieved using proprietary chromatographic techniques.
Physical Appearance
A clear, colorless solution that has been sterilized by filtration.
Formulation
The CYB5A protein solution is supplied at a concentration of 1 mg/ml. The solution is buffered with 20 mM Tris-HCl at a pH of 8.0 and also contains 20% glycerol, 0.1 M NaCl, and 1 mM DTT.
Stability
For short-term storage (2-4 weeks), the product can be kept at 4°C. For longer storage, the product should be frozen at -20°C. Adding a carrier protein (0.1% HSA or BSA) is recommended for long-term storage. Repeated freezing and thawing of the product should be avoided.
Purity
The purity of the CYB5A protein is greater than 95%, as determined by SDS-PAGE analysis.
Synonyms
Cytochrome b5, Microsomal cytochrome b5 type A, MCB5, CYB5A, CYB5.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSHMAEQSD EAVKYYTLEE IQKHNHSKST WLILHHKVYD LTKFLEEHPG GEEVLREQAG GDATENFEDV GHSTDAREMS KTFIIGELHP DDRPKLNKPP ETLITTIDSS SS.

Q&A

What is CYB5A and what cellular functions does it perform?

CYB5A, or cytochrome b5 type A, is a membrane-bound hemoprotein that functions as an electron carrier for several membrane-bound oxygenases. In humans, it consists of 134 amino acid residues with a molecular mass of approximately 15.3 kDa. The protein is primarily localized in the endoplasmic reticulum and cytoplasm, where it participates in various metabolic processes including fatty acid desaturation, steroid hormone biosynthesis, and cytochrome P450-mediated drug metabolism. Alternative splicing produces three different isoforms of the protein, allowing for functional diversity across different tissues . The protein's electron transfer capabilities make it a critical component in multiple redox reactions essential to cellular metabolism.

How is CYB5A structurally organized and what domains are crucial for its function?

The CYB5A protein contains a highly conserved heme-binding domain characteristic of the cytochrome b5 family. The canonical protein structure consists of a hydrophilic N-terminal domain that contains the heme-binding region and a hydrophobic C-terminal membrane anchor. The heme-binding domain adopts a characteristic fold with alpha helices and beta sheets that create a pocket for the heme prosthetic group, which is essential for its electron transfer capabilities. The membrane anchor domain facilitates proper localization to the endoplasmic reticulum. The three-dimensional structure of the protein enables specific protein-protein interactions with various partners, including cytochrome P450 enzymes, with which it participates in numerous metabolic pathways .

What are the key model organisms for studying CYB5A function?

Researchers investigating CYB5A commonly work with several model organisms where gene orthologs have been identified. These include mouse (Mus musculus), rat (Rattus norvegicus), bovine (Bos taurus), frog (Xenopus), zebrafish (Danio rerio), chimpanzee (Pan troglodytes), and chicken (Gallus gallus) . Each model organism offers distinct advantages for studying different aspects of CYB5A biology. Rodent models (mice and rats) are particularly valuable for metabolic and genetic studies due to their well-characterized genomes and physiological similarity to humans. Zebrafish models are useful for developmental studies, while bovine models may provide insights into metabolic functions in larger mammals. When selecting a model organism, researchers should consider the specific aspects of CYB5A function they wish to investigate and the conservation of relevant pathways across species.

What are the most effective methods for detecting and quantifying CYB5A protein in human tissues?

Detection and quantification of CYB5A protein in human tissues can be accomplished through several complementary techniques. Western blotting remains the gold standard for protein detection and semi-quantitative analysis, with numerous validated antibodies available commercially . For more precise quantification, enzyme-linked immunosorbent assay (ELISA) provides superior sensitivity and reproducibility. Immunohistochemistry (IHC) and immunofluorescence (IF) are valuable for visualizing the spatial distribution of CYB5A within tissues and cells, offering insights into subcellular localization patterns. For maximum sensitivity and specificity, researchers should consider using monoclonal antibodies targeting conserved epitopes in the CYB5A protein. Combining multiple detection methods provides the most comprehensive characterization of CYB5A expression patterns .

How can researchers effectively measure CYB5A enzymatic activity in experimental settings?

Measuring CYB5A enzymatic activity requires specialized assays that capture its electron transfer capabilities. The most common approach involves spectrophotometric methods that monitor the reduction of cytochrome c or other electron acceptors in the presence of NADH and cytochrome b5 reductase. This assay typically measures absorbance changes at 550 nm, corresponding to the reduced form of cytochrome c. For more complex analyses of CYB5A's role in specific metabolic pathways, researchers can employ reconstituted systems containing purified CYB5A, its reductase, and relevant partner enzymes such as cytochrome P450s. Mass spectrometry-based approaches can then be used to measure the formation of specific metabolic products. When designing these experiments, researchers must carefully control for pH, temperature, and the presence of detergents, as these factors significantly affect enzyme activity .

What are the optimal conditions for expressing and purifying recombinant human CYB5A protein?

Expressing and purifying recombinant human CYB5A requires careful optimization of experimental conditions. For bacterial expression systems, E. coli BL21(DE3) strains are commonly used with expression vectors containing strong inducible promoters like T7. Expression should be induced at lower temperatures (16-25°C) to enhance proper folding and heme incorporation. The soluble domain (amino acids 1-100) expresses more efficiently than the full-length protein containing the membrane anchor. For purification, a combination of immobilized metal affinity chromatography (using His-tags) followed by size exclusion chromatography yields high purity. The purification buffer should contain reducing agents like DTT or β-mercaptoethanol to prevent oxidation of cysteine residues. For functional studies, incorporation of the heme group is essential and can be verified by the characteristic absorbance at 413 nm. Researchers should verify protein quality through SDS-PAGE, Western blotting, and activity assays before proceeding with functional studies .

What role does CYB5A play in methemoglobinemia and how are mutations characterized?

CYB5A plays a critical role in methemoglobin reduction, converting the oxidized ferric (Fe³⁺) form of hemoglobin back to the functional ferrous (Fe²⁺) form. Inherited deficiencies in CYB5A lead to type II methemoglobinemia, characterized by elevated levels of methemoglobin in the blood that cannot effectively transport oxygen. Mutations in CYB5A causing this condition typically affect the protein's ability to bind heme or interact with NADH-cytochrome b5 reductase. Characterization of these mutations involves sequencing the CYB5A gene, followed by functional studies using site-directed mutagenesis to recreate the mutations in expression systems. Spectroscopic analysis of mutant proteins can reveal alterations in heme binding, while enzyme kinetic studies measure the impact on electron transfer capabilities. Clinical severity often correlates with the degree of functional impairment, with complete loss-of-function mutations causing more severe presentations .

How is CYB5A linked to disorders of sexual development and ambiguous genitalia?

CYB5A is implicated in disorders of sexual development through its role in steroid hormone biosynthesis, particularly in the synthesis of testosterone and dihydrotestosterone. The protein functions as an electron donor for several steroid metabolizing enzymes, including 17α-hydroxylase and 17,20-lyase, which are crucial for androgen production. Mutations that impair CYB5A function can lead to reduced androgen synthesis during critical developmental windows, resulting in ambiguous genitalia in genetic males . Research characterizing these conditions requires genetic sequencing to identify mutations, followed by in vitro enzyme assays to determine their impact on steroidogenic enzyme activities. Animal models with targeted CYB5A mutations have further elucidated its role in sexual development. The clinical presentation varies depending on the specific mutation and its effect on residual enzyme activity, with some patients showing isolated genital ambiguity while others have more complex endocrine abnormalities.

What evidence exists for CYB5A's involvement in metabolic disorders and obesity?

Recent research has uncovered compelling evidence linking CYB5A to metabolic disorders, particularly obesity. A significant study involving Southwest American Indian (SWAI) populations identified a 5' untranslated region variant in CYB5A (rs548402150) that associates with increased BMI and potentially contributes to obesity risk . Functional characterization demonstrated that this variant decreased luciferase expression by approximately 30% and correlated with decreased skeletal muscle CYB5A expression . Further analysis identified a specific haplotype associated with increased BMI and decreased 24-hour energy expenditure. These findings suggest that CYB5A may influence metabolic rate and energy homeostasis through mechanisms that are still being elucidated. The relationship between CYB5A expression levels and metabolic parameters indicates potential involvement in lipid metabolism, possibly through its interaction with fatty acid desaturases and elongases. This emerging area presents opportunities for research into novel therapeutic targets for obesity and related metabolic disorders .

How do population-specific CYB5A variants contribute to differential disease susceptibility?

Population-specific variants in CYB5A demonstrate significant contributions to differential disease susceptibility across ethnic groups. Research has identified functional variants that are enriched in specific populations, such as the Southwest American Indian (SWAI) communities, where the variant rs548402150 in the 5' untranslated region of CYB5A is associated with increased BMI . This variant exemplifies how population-specific genetic architecture can influence disease risk, as it appears to decrease CYB5A expression and consequently affect metabolic rate. When investigating such variants, researchers should employ both targeted genotyping and whole genome sequencing approaches, followed by functional validation through reporter assays, expression analysis in relevant tissues, and correlation with phenotypic data. Understanding the evolutionary history of these variants provides additional insights, potentially revealing selective pressures that maintained these alleles in specific populations. Researchers should design studies with adequate sample sizes from diverse populations to detect such population-specific effects and account for potential confounding factors such as linkage disequilibrium patterns .

What transcriptional and post-transcriptional mechanisms regulate CYB5A expression?

Regulation of CYB5A expression involves complex transcriptional and post-transcriptional mechanisms that fine-tune protein levels across different tissues and physiological states. At the transcriptional level, the CYB5A promoter contains binding sites for several transcription factors involved in metabolic regulation, including members of the PPAR (peroxisome proliferator-activated receptor) family and SREBP (sterol regulatory element-binding protein), suggesting responsiveness to metabolic signals. Post-transcriptionally, CYB5A is regulated through alternative splicing that generates three distinct isoforms with potentially different functional properties or subcellular localizations . Additionally, 5' untranslated region variants like rs548402150 can significantly impact translation efficiency, as demonstrated by a 30% reduction in luciferase expression in reporter assays . MicroRNAs targeting the CYB5A transcript provide another layer of regulation, with several predicted binding sites in the 3' UTR. Researchers investigating these regulatory mechanisms should employ techniques such as chromatin immunoprecipitation (ChIP) for transcription factor binding, RNA-seq for alternative splicing analysis, and polysome profiling for translational efficiency assessment.

How does CYB5A interact with cytochrome P450 enzymes and influence drug metabolism?

CYB5A serves as an electron donor that modulates the activity of numerous cytochrome P450 (CYP) enzymes involved in drug metabolism, creating significant implications for pharmacogenomics and personalized medicine. The interaction between CYB5A and CYP enzymes occurs through specific protein-protein interfaces, with CYB5A typically enhancing the electron transfer efficiency of the CYP catalytic cycle. This enhancement can be substrate-specific and CYP isoform-dependent, resulting in complex effects on drug metabolism rates. Researchers studying these interactions should employ multiple complementary approaches: in vitro reconstitution systems with purified proteins allow precise mechanistic studies, while cell-based assays provide more physiologically relevant contexts. Structural studies using X-ray crystallography or cryo-electron microscopy can reveal the molecular details of these protein-protein interactions. Importantly, genetic variants in CYB5A that alter protein levels or function may contribute to inter-individual variability in drug metabolism, potentially affecting drug efficacy and toxicity. Research in this area should consider both genetic polymorphisms and environmental factors that might influence CYB5A-mediated electron transfer to drug-metabolizing enzymes .

What are the optimal experimental designs for studying CYB5A's role in energy expenditure and obesity?

Designing experiments to investigate CYB5A's role in energy expenditure and obesity requires multifaceted approaches spanning molecular, cellular, and whole-organism studies. Based on the association between CYB5A variants and 24-hour energy expenditure (24-h EE), researchers should consider metabolic chamber studies that precisely measure oxygen consumption, carbon dioxide production, and heat generation in human subjects or animal models with different CYB5A genotypes or expression levels . Tissue-specific knockout or overexpression models, particularly targeting skeletal muscle where CYB5A expression correlates with metabolic parameters, can reveal causative relationships. For cellular studies, primary myocytes or adipocytes with modified CYB5A expression enable investigation of substrate utilization patterns using radioactive tracers or Seahorse metabolic analyzers to measure oxygen consumption rates and extracellular acidification. In human studies, a combination of genetic analysis, expression profiling in muscle biopsies, and comprehensive metabolic phenotyping provides the most informative data . Researchers should control for confounding factors such as age, sex, diet, physical activity, and concurrent metabolic conditions to isolate CYB5A-specific effects.

How can CRISPR-Cas9 technology be optimized for studying CYB5A function?

CRISPR-Cas9 technology offers powerful approaches for investigating CYB5A function through precise genome editing. When designing CRISPR experiments for CYB5A, researchers should carefully select guide RNAs targeting conserved functional domains while minimizing off-target effects. For complete knockout studies, targeting early exons maximizes the likelihood of functional disruption. More subtle manipulations, such as introducing specific point mutations that mimic human variants (e.g., rs548402150), require homology-directed repair templates and strategies to enrich for edited cells . Cell type selection is critical: hepatocytes and myocytes express high levels of CYB5A and are relevant to its metabolic functions. For in vivo studies, conditional knockout models using Cre-lox systems combined with CRISPR allow tissue-specific and temporally controlled CYB5A disruption, preventing developmental compensation that might mask phenotypes. Validation of editing outcomes should include sequencing, protein expression analysis, and functional assays measuring electron transfer capacity. Advanced applications include CRISPR activation (CRISPRa) or interference (CRISPRi) systems to modulate CYB5A expression without altering the genomic sequence, allowing more nuanced studies of dosage effects .

What are the most informative experimental models for studying CYB5A in steroid hormone biosynthesis?

Investigating CYB5A's role in steroid hormone biosynthesis requires specialized experimental models that recapitulate the complexities of steroidogenic pathways. Cell-based models should include steroidogenic cell lines like H295R (adrenocortical) and Leydig cells that express the complete enzymatic machinery for steroid synthesis. CYB5A knockdown or overexpression in these systems, followed by comprehensive steroid profiling using liquid chromatography-tandem mass spectrometry (LC-MS/MS), can reveal specific steps in the pathway affected by CYB5A alterations. Reconstituted in vitro systems containing purified CYB5A, cytochrome P450c17 (17α-hydroxylase/17,20-lyase), and cytochrome P450 oxidoreductase allow precise kinetic studies of 17,20-lyase activity, which is particularly dependent on CYB5A . For in vivo studies, gonadal or adrenal-specific CYB5A knockout models generated through Cre-lox systems provide insights into tissue-specific functions while avoiding the potential lethality of global knockouts. In human studies, patients with disorders of sexual development and identified CYB5A mutations offer valuable opportunities for genotype-phenotype correlations. Researchers should measure multiple steroid intermediates rather than end products alone to identify specific enzymatic steps affected by CYB5A alterations.

What is the impact of CYB5A genetic variants on metabolic parameters?

Table 1: Key CYB5A Genetic Variants and Their Metabolic Effects

Variant IDLocationPopulation FrequencyAssociated PhenotypesFunctional EffectReference
rs5484021505' UTREnriched in SWAI populationsIncreased BMI (+2%)30% decrease in gene expression
Common haplotype*Multiple lociPrevalent across ethnic groupsLower BMI, lower body fat percentageIncreased CYB5A expression
Rare haplotype**Multiple loci incl. rs548402150Almost exclusively in SWAIIncreased BMI, increased body fatDecreased CYB5A expression

*Common haplotype includes splicing quantitative trait loci associated with higher expression
**Rare haplotype combines rs548402150 with two splicing quantitative trait loci

What experimental methods are most commonly used in CYB5A research?

Table 2: Experimental Methods for CYB5A Research

MethodApplicationAdvantagesLimitationsSpecial Considerations
Western BlotProtein detection and semi-quantificationWidely validated, specificSemi-quantitative onlyMultiple validated antibodies available
ELISAQuantitative protein measurementHigh sensitivity, quantitativeLimited spatial informationRequires careful antibody validation
ImmunohistochemistryTissue localizationPreserves tissue architectureLower sensitivity than IFAntigen retrieval often necessary
ImmunofluorescenceCellular localizationHigh resolution, co-localization studiesPhotobleachingMultiple antibodies available with validated reactivity
Luciferase Reporter AssayPromoter/UTR functionQuantitative, sensitiveArtificial contextSuccessfully used to characterize rs548402150
RNA-seqExpression profilingGenome-wide, isoform detectionRequires bioinformatic expertiseValuable for identifying splicing variants

How does CYB5A expression correlate with metabolic measures in human studies?

Table 3: Correlation Between CYB5A Expression and Metabolic Parameters

TissueMetabolic ParameterCorrelation DirectionStatistical SignificancePopulation StudiedReference
Skeletal MuscleCYB5A mRNA levelsNegative correlation with BMIp = 0.0008SWAI individuals
Skeletal MuscleCYB5A mRNA levelsPositive correlation with 24-h energy expenditurep < 0.05SWAI individuals
Skeletal MuscleCYB5A haplotypesAssociation with percentage body fatp = 0.0003SWAI individuals

What emerging technologies will advance our understanding of CYB5A biology?

Emerging technologies are poised to significantly expand our understanding of CYB5A biology across multiple scales. Single-cell multi-omics approaches combining transcriptomics, proteomics, and metabolomics will reveal cell-type-specific CYB5A functions and regulatory networks with unprecedented resolution. Spatial transcriptomics and proteomics technologies will map CYB5A expression patterns within tissues, potentially uncovering microenvironmental influences on its function. For protein interaction studies, proximity labeling methods like BioID or TurboID coupled with mass spectrometry will identify the CYB5A interactome in native cellular environments. Advanced structural biology techniques, including cryo-electron microscopy and AlphaFold-based predictions, will provide insights into CYB5A's interactions with partner proteins at atomic resolution. Metabolic flux analysis using stable isotope tracers combined with high-resolution mass spectrometry will elucidate CYB5A's contribution to specific metabolic pathways in real-time. Finally, human-derived organoids and induced pluripotent stem cell (iPSC) models carrying specific CYB5A variants will bridge the gap between cellular studies and whole-organism physiology, offering platforms for personalized medicine applications .

How can systems biology approaches be applied to understand CYB5A's role in metabolic networks?

Systems biology approaches offer powerful frameworks for understanding CYB5A's role within complex metabolic networks. Multi-omics integration combining transcriptomics, proteomics, and metabolomics data from tissues with varying CYB5A expression levels can identify correlated changes across biological scales. Constraint-based metabolic modeling, particularly genome-scale metabolic models incorporating CYB5A-dependent reactions, can predict systemic effects of CYB5A perturbations on metabolic flux distributions. Network analysis identifying hub metabolites affected by CYB5A activity may reveal previously unrecognized functions. For experimental validation of these computational predictions, metabolic flux analysis using stable isotope-labeled substrates tracked through CYB5A-influenced pathways provides direct evidence of altered metabolic routing. Importantly, integration of genetic association data with phenotypic measurements and molecular profiles can establish causal relationships between CYB5A variants and metabolic outcomes. Researchers applying these approaches should consider tissue-specific metabolic environments, circadian variations in metabolism, and potential compensatory mechanisms that may mask CYB5A effects in steady-state measurements .

What are the most promising therapeutic applications targeting CYB5A pathways?

The involvement of CYB5A in multiple metabolic and endocrine pathways suggests several promising therapeutic applications. For obesity and metabolic disorders, compounds that enhance CYB5A expression or activity could potentially increase energy expenditure based on the positive correlation between CYB5A levels and 24-hour energy expenditure . Small molecules that stabilize CYB5A-cytochrome P450 interactions might enhance fatty acid metabolism in tissues like skeletal muscle. In disorders of sexual development related to CYB5A deficiency, gene therapy approaches delivering functional CYB5A to steroidogenic tissues represent a potential curative strategy. For type II methemoglobinemia caused by CYB5A mutations, pharmacological chaperones that stabilize mutant CYB5A proteins and preserve residual function could ameliorate symptoms. Drug metabolism applications include developing CYB5A modulators that could selectively enhance the metabolism of specific drugs, potentially improving efficacy or reducing side effects. When developing these therapeutic strategies, researchers should consider tissue-specific delivery methods, potential off-target effects due to CYB5A's involvement in multiple pathways, and individual genetic variation that might influence response to CYB5A-targeted interventions .

Product Science Overview

Structure and Isoforms

Cytochrome B5 Type A has two isoforms produced by alternative splicing:

  1. Isoform 1: This isoform is bound to the cytoplasmic side of the endoplasmic reticulum and contains a C-terminal transmembrane alpha-helix .
  2. Isoform 2: This isoform is found in the cytoplasm .

The protein’s structure includes a heme-binding domain, which is essential for its electron transfer function. The heme group is coordinated by two histidine residues, providing the fifth and sixth ligands to the iron atom in the heme .

Biological Functions

Cytochrome B5 Type A is involved in several critical biological processes:

  • Electron Transport: It acts as an electron donor to various oxygenases, facilitating oxidative reactions .
  • Fatty Acid Metabolism: It is essential for the desaturation and elongation of fatty acids, which are vital for maintaining cell membrane integrity and producing signaling molecules .
  • Cholesterol Biosynthesis: Cytochrome B5 Type A is involved in the synthesis of cholesterol, a crucial component of cell membranes and precursor for steroid hormones .
  • Drug Metabolism: It participates in the metabolism of various drugs and xenobiotics by cytochrome P450 enzymes .
Clinical Significance

Defects in the CYB5A gene can lead to type IV hereditary methemoglobinemia, a condition characterized by an abnormal increase in methemoglobin levels in the blood. This condition can result in cyanosis and other related symptoms .

Recombinant Production

Human recombinant Cytochrome B5 Type A is typically produced using Escherichia coli expression systems. The recombinant protein is often tagged with histidine to facilitate purification . This recombinant form is used in various research applications to study its structure, function, and interactions with other proteins.

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.