MMAB Human

Methylmalonic Aciduria Type B Human Recombinant
Shipped with Ice Packs
In Stock

Description

Introduction to MMAB Human

The MMAB gene encodes ATP:cob(I)alamin adenosyltransferase, an enzyme critical for converting cob(I)alamin (a form of vitamin B12) into adenosylcobalamin (AdoCbl). This coenzyme is essential for the activity of methylmalonyl-CoA mutase (MMUT), an enzyme involved in the breakdown of proteins, lipids, and cholesterol . Mutations in MMAB cause methylmalonic acidemia (cblB-type), a rare metabolic disorder characterized by toxic accumulations of methylmalonic acid and related compounds .

Enzymatic Activity

MMAB facilitates the final step in AdoCbl biosynthesis, enabling MMUT to process methylmalonyl-CoA into succinyl-CoA. This pathway is critical for:

  • Propionate metabolism: Breakdown of odd-chain fatty acids and amino acids (e.g., valine, isoleucine) .

  • Cholesterol biosynthesis: Indirect regulation via SREBP2, a master transcription factor for cholesterol homeostasis .

Mechanistic Insight:

  • SREBP2 Regulation: MMAB expression is modulated by dietary cholesterol and statins. Low cholesterol increases SREBP2 activity, upregulating MMAB and MVK (mevalonate kinase) to inhibit HMGCR (3-hydroxy-3-methylglutaryl-CoA reductase), reducing intracellular sterols .

  • Cholesterol Feedback: MMAB knockdown reduces intracellular cholesterol, enhancing LDL receptor (LDLR) expression and LDL uptake .

Methylmalonic Acidemia (cblB-Type)

FeatureDetails
InheritanceAutosomal recessive
Key Mutations33 distinct variants identified (16 novel), including missense (61.9%) and truncating (18%) mutations
Common Variantsp.Arg186Trp (57 alleles), p.Arg191Trp (19 alleles), c.197-1G>T (22 alleles)
SymptomsMetabolic ketoacidosis, developmental delay, neurological/renal impairment
TreatmentOH-cobalamin (OHCbl) supplementation improves biochemical outcomes

Biochemical Impact: Mutations impair AdoCbl synthesis, leading to MMUT dysfunction and toxic metabolite accumulation .

Cholesterol Homeostasis

  • GWAS Links: MMAB/MVK loci associate with plasma LDL-C and HDL-C levels in humans .

  • Mechanistic Role: MMAB knockdown reduces intracellular cholesterol via increased methylmalonic acid, inhibiting HMGCR and cholesterol biosynthesis .

  • Therapeutic Potential: Targeting MMAB may modulate lipid metabolism in dyslipidemia .

Biomarker Applications

  • HIV Therapy: Efavirenz (NNRTI) increases mitochondrial MMAB expression, correlating with oxidative stress in liver tissue .

Table 1: Common MMAB Mutations in cblB-Type Methylmalonic Acidemia

Variant TypeVariantFrequencyEffectSource
Missensep.Arg186Trp57 allelesLoss of enzymatic activity
Missensep.Arg191Trp19 allelesDisrupted cob(I)alamin binding
Truncatingc.700C>T (p.Arg234*)22 allelesPremature termination
Splicingc.197-1G>T22 allelesExon skipping

Table 2: GWAS Associations of MMAB/MVK Loci

StudyPopulationAssociated TraitSignificanceSource
Multi-ancestry GWASGlobal cohortsLDL-C, HDL-CGenome-wide significance
Lipid metabolism GWASEuropean cohortsLDLR expressionSREBP2-mediated regulation

Product Specs

Introduction
The MMAB protein plays a crucial role in the final step of converting vitamin B12 into adenosylcobalamin (AdoCbl), a vital coenzyme for methylmalonyl-CoA mutase (MCM) that contains vitamin B12. Deficiencies in MMAB activity are linked to an inherited disorder known as vitamin B12-dependent methylmalonic aciduria, which is associated with the cblB complementation group.
Description
Recombinant Human MMAB, synthesized in E. coli, is a single, non-glycosylated polypeptide chain consisting of 239 amino acids (specifically, residues 33-250). With a molecular weight of 26.3 kDa, the MMAB protein is fused to a 21 amino acid His-Tag at its N-terminus. Purification is achieved through proprietary chromatographic methods.
Physical Appearance
The product is a clear solution that has been sterilized through filtration.
Formulation
The MMAB protein solution has a concentration of 1 mg/ml and contains 20 mM Tris buffer at pH 7.5 and 10% glycerol.
Stability
While Human MMAB remains stable at 4°C for a period of one week, it is recommended to store the protein below -18°C to ensure its long-term stability. It is crucial to avoid repeated freeze-thaw cycles.
Purity
SDS-PAGE analysis has determined the purity of the MMAB protein to be greater than 95.0%.
Synonyms
CBIB, Cob(I)alamin adenosyltransferase, EC 2.5.1.17.
Source
Escherichia Coli.
Amino Acid Sequence

MGSSHHHHHH SSGLVPRGSH MQSRGPQGVE DGDRPQPSSK TPRIPKIYTK TGDKGFSSTF TGERRPKDDQ VFEAVGTTDE LSSAIGFALE LVTEKGHTFA EELQKIQCTL QDVGSALATP CSSAREAHLK YTTFKAGPIL ELEQWIDKYT SQLPPLTAFI LPSGGKISSA LHFCRAVCRR AERRVVPLVQ MGETDANVAK FLNRLSDYLF TLARYAAMKE GNQEKIYKKN DPSAESEGL.

Q&A

What is the MMAB gene and what protein does it encode?

The MMAB gene (Metabolism of Cobalamin Associated B) provides instructions for making an enzyme that plays a critical role in the formation of adenosylcobalamin (AdoCbl), a compound derived from vitamin B12 (cobalamin). This enzyme functions as an ATP:Cob(I)alamin adenosyltransferase, catalyzing a key step in vitamin B12 metabolism . The enzyme is primarily active within mitochondria, where it not only facilitates the conversion of cob(I)alamin to AdoCbl but may also participate in delivering AdoCbl to methylmalonyl CoA mutase, an enzyme involved in breaking down certain proteins, lipids, and cholesterol .

Where is the MMAB gene located in the human genome?

The MMAB gene is located on chromosome 12 in the human genome . This chromosomal location is significant for genetic mapping and linkage analysis when studying inherited disorders related to MMAB mutations. Researchers investigating potential genetic associations or conducting family studies should note this location when designing genotyping strategies or analyzing inheritance patterns in affected families.

How does the MMAB enzyme function within mitochondria?

Within mitochondria, the MMAB enzyme functions as a critical component in vitamin B12 metabolism. After vitamin B12 has been transported into the mitochondria, the MMAB enzyme converts cob(I)alamin (a reduced form of vitamin B12) to adenosylcobalamin (AdoCbl) . This conversion involves an adenosyltransferase reaction where an adenosyl group from ATP is transferred to cob(I)alamin. Beyond this catalytic role, research suggests that the MMAB enzyme may have an additional function in delivering the synthesized AdoCbl directly to methylmalonyl CoA mutase, effectively creating a metabolic channeling system that ensures efficient utilization of this essential cofactor .

What alternative names have been used for MMAB in the scientific literature?

In scientific literature and databases, the MMAB gene and its protein product have been referenced under multiple alternative designations, which can complicate literature searches. These alternative names include: ATP:Cob(I)alamin Adenosyltransferase, ATR, cblB, CFAP23, cob(I)alamin adenosyltransferase, methylmalonic aciduria (cobalamin deficiency) cblB type, and methylmalonic aciduria (cobalamin deficiency) type B . When conducting literature reviews or database searches, researchers should include these alternative designations to ensure comprehensive retrieval of relevant publications.

What types of mutations in MMAB are associated with methylmalonic acidemia?

At least 25 different mutations in the MMAB gene have been identified in patients with methylmalonic acidemia . These mutations include several structural variations: some delete or duplicate small amounts of genetic material within the MMAB gene, while others are point mutations that change a single amino acid in the enzyme sequence . The diverse nature of these mutations highlights the genetic heterogeneity underlying this disorder. Understanding the specific mutation type is crucial for genotype-phenotype correlation studies and can inform prognosis and potential treatment approaches.

How do MMAB mutations impact enzyme function and lead to disease?

MMAB mutations typically result in the production of a nonfunctional enzyme that cannot properly convert cob(I)alamin to adenosylcobalamin (AdoCbl) . This enzymatic deficiency has cascading effects: without sufficient AdoCbl, the function of methylmalonyl CoA mutase is impaired, leading to incomplete breakdown of certain proteins, lipids, and cholesterol . The resulting accumulation of toxic metabolites in organs and tissues manifests as the clinical signs and symptoms of methylmalonic acidemia, including feeding difficulties, developmental delays, and long-term health complications . The severity of enzymatic dysfunction often correlates with the nature of the specific mutation and can influence disease presentation and progression.

What are the clinical manifestations of MMAB-related methylmalonic acidemia?

Methylmalonic acidemia resulting from MMAB mutations manifests with a spectrum of clinical features. The condition is typically characterized by feeding difficulties in infancy, which may be accompanied by lethargy, vomiting, and failure to thrive . As the disease progresses, patients often exhibit developmental delays and may experience metabolic crises triggered by catabolic stress (such as infections or prolonged fasting) . Long-term health complications can affect multiple organ systems, including the brain, kidneys, and heart. The severity and progression of symptoms can vary considerably between patients, even those sharing similar mutations, suggesting the influence of additional genetic or environmental modifiers.

How does long-term outcome in methylmalonic acidemia correlate with the underlying MMAB defect?

Long-term outcomes in methylmalonic acidemia are significantly influenced by the specific genetic defect underlying the condition. Research by Horster et al. has demonstrated that patients with cblB defects (MMAB mutations) show distinct clinical trajectories compared to those with other genetic causes of methylmalonic acidemia (mut0, mut-, or cblA) . These differences manifest in varying rates of metabolic decompensation, neurodevelopmental outcomes, and organ-specific complications. The correlation between genotype and long-term prognosis underscores the importance of precise molecular diagnosis in guiding clinical management and counseling affected families about expected disease course.

What techniques are used to identify and characterize MMAB mutations?

Multiple molecular techniques are employed to identify and characterize MMAB mutations in research and clinical settings. These include PCR amplification and Sanger sequencing of MMAB exons and exon-intron boundaries, next-generation sequencing panels targeting metabolic disorder genes, and in some cases, whole exome or genome sequencing . For larger structural variations, techniques such as multiplex ligation-dependent probe amplification (MLPA) or array comparative genomic hybridization (aCGH) may be necessary. Functional validation of novel variants often requires complementation studies in bacterial or mammalian cell systems to confirm pathogenicity. Researchers should select methodologies based on their specific research questions and available resources.

How is MMAB enzyme activity measured in experimental systems?

Measurement of MMAB enzyme activity is critical for functional studies and typically involves radioisotope-based or chromatographic assays. The standard approach quantifies the conversion of radiolabeled cob(I)alamin to adenosylcobalamin in the presence of ATP and appropriate reducing conditions . This can be performed using patient fibroblasts, lymphocytes, or recombinant protein systems. HPLC or mass spectrometry-based methods may also be employed to detect adenosylcobalamin formation. When designing such experiments, researchers must carefully control for factors such as oxygen exposure (which can oxidize cob(I)alamin), ATP concentration, and the presence of appropriate reducing agents to maintain physiological conditions.

What cellular and animal models are available for studying MMAB function?

Several model systems have been developed to study MMAB function and the consequences of its deficiency. Cellular models include patient-derived fibroblasts, lymphoblastoid cell lines, and engineered cell lines with CRISPR-Cas9 mediated MMAB knockout or specific mutations . Bacterial complementation systems using Escherichia coli with defects in their endogenous adenosyltransferase have been valuable for functional characterization of human MMAB variants . While specific MMAB-deficient mouse models have been limited, broader models of methylmalonic acidemia have provided insights into the systemic consequences of this metabolic defect. When selecting a model system, researchers should consider the specific aspect of MMAB biology under investigation and the translational relevance of their chosen model.

What complementation studies have been used to validate MMAB mutations?

Complementation studies have been instrumental in confirming the pathogenicity of MMAB variants and understanding their functional consequences. These typically involve introducing wild-type or mutant MMAB cDNA into cells from patients with methylmalonic acidemia or into bacterial systems with defective adenosyltransferase activity . Rescue of adenosylcobalamin synthesis or methylmalonyl-CoA mutase activity indicates functional complementation. Studies by Leal et al. and Zhang et al. have employed such approaches to systematically characterize multiple MMAB mutations and correlate their biochemical impact with clinical presentations . When designing complementation experiments, researchers should include appropriate positive and negative controls and consider the potential influence of endogenous enzyme activity.

How does the MMAB enzyme interact with other proteins in vitamin B12 metabolism?

The MMAB enzyme functions within a complex network of proteins involved in intracellular vitamin B12 metabolism. Research by Leal et al. has demonstrated that MMAB interacts with methionine synthase reductase, suggesting potential cross-regulation between the cytosolic and mitochondrial branches of cobalamin metabolism . Additionally, evidence indicates that MMAB may directly interact with methylmalonyl CoA mutase to facilitate targeted delivery of its adenosylcobalamin cofactor, creating a metabolic channeling system . These protein-protein interactions may be influenced by mutations or post-translational modifications, potentially contributing to the phenotypic variability observed in patients. Advanced techniques such as co-immunoprecipitation, proximity labeling, or structural studies can further elucidate these interaction networks.

What is known about the structure-function relationship of the MMAB protein?

Understanding the structure-function relationship of MMAB provides critical insights into how specific mutations impact enzyme activity. Structural analyses have identified key catalytic residues involved in ATP binding, cob(I)alamin positioning, and adenosyl transfer . Mutations affecting these residues typically result in complete loss of enzyme function, while variants in peripheral regions may cause more subtle effects on protein stability or interaction capabilities. Research by Zhang et al. has correlated the location of specific mutations with their biochemical consequences, demonstrating how structural alterations translate to functional deficits . Researchers employing molecular modeling or structural biology approaches should pay particular attention to the conservation of crucial catalytic domains across species and potential conformational changes induced by substrate binding.

What therapeutic approaches target MMAB dysfunction in methylmalonic acidemia?

Current therapeutic strategies for MMAB-related methylmalonic acidemia primarily focus on supportive management, including dietary protein restriction, carnitine supplementation, and management of acute metabolic crises . High-dose vitamin B12 supplementation shows variable response depending on the specific mutation and residual enzyme activity. Emerging experimental approaches include enzyme replacement therapy, gene therapy targeting MMAB, and liver or combined liver-kidney transplantation for severe cases . Cell-based therapies using genetically corrected autologous cells are also under investigation. Researchers developing novel therapeutic strategies should consider the mitochondrial localization of MMAB, the need for appropriate cofactor (ATP) availability, and potential interactions with other proteins in the vitamin B12 metabolic pathway.

What are the current challenges in understanding MMAB regulation in different tissues?

Despite advances in characterizing MMAB function, significant knowledge gaps remain regarding its tissue-specific regulation and expression patterns. The enzyme appears to be differentially regulated across tissues, which may contribute to the organ-specific manifestations of methylmalonic acidemia . Current challenges include understanding the transcriptional and post-transcriptional mechanisms governing MMAB expression, identifying potential tissue-specific isoforms, and characterizing how metabolic states influence enzyme activity. Research approaches combining tissue-specific transcriptomics, proteomics, and metabolomics could provide valuable insights into these regulatory mechanisms and potentially reveal tissue-tailored therapeutic approaches.

How might CRISPR-Cas9 gene editing be applied to MMAB mutations?

CRISPR-Cas9 technology offers promising approaches for addressing MMAB mutations at the genomic level. Potential applications include precise correction of pathogenic variants in patient-derived cells, creation of isogenic cell lines for mechanistic studies, and development of improved animal models . For clinical translation, ex vivo gene editing of hematopoietic stem cells or hepatocytes followed by autologous transplantation represents a potential therapeutic strategy. Researchers pursuing CRISPR-based approaches should consider mutation-specific editing strategies, potential off-target effects, and delivery methods capable of reaching mitochondria-rich tissues such as liver and muscle. Validation of functional rescue should include assessment of adenosylcobalamin synthesis and methylmalonyl-CoA mutase activity.

What is the significance of MMAB in conditions beyond methylmalonic acidemia?

Emerging evidence suggests that MMAB may have implications beyond its established role in methylmalonic acidemia. Altered MMAB function could potentially influence mitochondrial metabolism more broadly, affecting energy production, one-carbon metabolism, and oxidative stress responses . The interaction between MMAB and methionine synthase reductase suggests possible connections to homocysteine metabolism and associated cardiovascular or neurological disorders . Additionally, given the role of adenosylcobalamin in methylmalonyl-CoA mutase function, MMAB may indirectly influence branched-chain amino acid metabolism and fatty acid oxidation. Researchers investigating these broader implications should employ systems biology approaches to map the extended metabolic networks influenced by MMAB activity.

How can multi-omics approaches advance our understanding of MMAB-related disorders?

Integration of multiple omics platforms offers powerful approaches for comprehensively characterizing the molecular consequences of MMAB dysfunction. Combining genomics (to identify mutations), transcriptomics (to assess compensatory gene expression changes), proteomics (to map protein interaction networks), and metabolomics (to profile metabolic disturbances) can provide unprecedented insights into disease mechanisms . Such integrated approaches may reveal novel biomarkers for disease monitoring, identify potential therapeutic targets, and help explain phenotypic variability among patients with similar mutations. Researchers employing multi-omics strategies should develop appropriate computational pipelines for data integration and consider both targeted and untargeted approaches to maximize discovery potential while ensuring biological relevance.

Product Science Overview

Introduction

Methylmalonic Aciduria (MMA) is a rare metabolic disorder characterized by the accumulation of methylmalonic acid in the blood and urine. This condition is caused by a deficiency in the enzyme methylmalonyl-CoA mutase, which is essential for the metabolism of certain amino acids and fatty acids . MMA can be classified into different types based on the underlying genetic mutations and the specific metabolic pathways affected.

Methylmalonic Aciduria Type B (cblB)

Methylmalonic Aciduria Type B, also known as cblB type, is a specific form of MMA caused by mutations in the MMAB gene. This gene encodes an enzyme called adenosyltransferase, which is involved in the conversion of vitamin B12 into its active form, adenosylcobalamin (AdoCbl) . AdoCbl is a crucial coenzyme for the proper functioning of methylmalonyl-CoA mutase.

Human Recombinant MMAB

Human recombinant MMAB is a laboratory-produced version of the MMAB enzyme. It is typically produced in Escherichia coli (E. coli) bacteria and purified using chromatographic techniques . The recombinant enzyme is used in research and therapeutic applications to study the biochemical pathways involved in MMA and to develop potential treatments for the disorder.

Biochemical Pathways

The MMAB enzyme catalyzes the final step in the conversion of vitamin B12 into AdoCbl. This process is essential for the proper metabolism of certain amino acids (such as threonine, methionine, isoleucine, and valine) and odd-chain fatty acids . In individuals with cblB type MMA, the deficiency of MMAB leads to the accumulation of methylmalonic acid, resulting in various clinical symptoms.

Clinical Symptoms and Diagnosis

Patients with MMA, including the cblB type, may present with a range of symptoms, including developmental delays, feeding difficulties, vomiting, lethargy, and metabolic acidosis . Diagnosis is typically based on biochemical tests that measure the levels of methylmalonic acid in the blood and urine, as well as genetic testing to identify mutations in the MMAB gene.

Treatment and Management

The treatment of MMA is complex and requires a multidisciplinary approach. Management strategies may include dietary modifications to reduce the intake of certain amino acids, supplementation with vitamin B12, and regular monitoring of metabolic parameters . In some cases, liver or kidney transplantation may be considered as a treatment option.

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.