NDUFA2 Human

NADH Dehydrogenase 1 Alpha Subcomplex 2 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Genetic and Clinical Implications

Mutations in NDUFA2 are linked to severe mitochondrial disorders:

ConditionMechanismReferences
Leigh SyndromeExon 2 skipping, reduced Complex I activity, mitochondrial depolarization
Mitochondrial LeukoencephalopathyNovel recessive mutations causing cystic brain lesions
Alzheimer’s DiseaseBiomarker association (exact mechanism unclear)

Case Study: A novel NDUFA2 mutation (c.101G>A, p.Arg34Gln) disrupts Complex I assembly, leading to accumulation of 550 kDa subcomplexes and reduced oxygen consumption .

Research Findings and Assembly Dynamics

NDUFA2 is essential for Complex I assembly and stability:

  • Subcomplex Accumulation: Suppression of NDUFA2 leads to incomplete Complex I structures (815 kDa and 550 kDa subcomplexes) .

  • Interaction Networks: Co-purifies with assembly factors (C3orf1, TMEM126B) and chaperones (DNAJC11) .

  • Redox Regulation: May modulate Complex I activity via conformational changes during electron transfer .

Expression in Normal and Cancerous Tissues

The Human Protein Atlas reveals tissue-specific expression patterns:

Tissue TypeExpression LevelCancer AssociationSources
Brain, Liver, MuscleHighLow in colorectal, breast, and lung cancers
Blood, Cell LinesModerateCorrelation with survival in some cohorts

Cancer Prognosis: Elevated NDUFA2 mRNA in certain cancers (e.g., prostate) correlates with unfavorable outcomes, though data are inconsistent across tumor types .

Biochemical Properties and Recombinant Production

Recombinant NDUFA2 is produced in E. coli for research:

PropertyDetailsSources
Sequence122 aa (1–99 aa + His-tag)
Purity>95% (SDS-PAGE)
Storage-20°C for long-term; 4°C for short-term (with glycerol/DTT)

Future Directions

  1. Therapeutic Targets: Exploiting NDUFA2 interactions with assembly factors (e.g., TMEM126B) to treat Complex I deficiencies.

  2. Cancer Metabolism: Investigating NDUFA2’s role in mitochondrial reprogramming in tumors.

Product Specs

Introduction
NADH dehydrogenase [ubiquinone] 1 alpha subcomplex subunit 5 (NDUFA5) is a protein found in the inner mitochondrial membrane. It is a subunit of Complex I, which is responsible for transferring electrons from NADH to ubiquinone in the electron transport chain. The exact function of NDUFA5 is not fully understood, but it is part of the 'iron-sulfur protein' (IP) fraction of Complex I.
Description
Recombinant human NDUFA2 protein expressed in E.coli. This protein is a single, non-glycosylated polypeptide chain consisting of 122 amino acids (residues 1-99) with a molecular weight of 13.3 kDa. It includes a 23 amino acid His-tag at the N-terminus. The protein has been purified using proprietary chromatographic techniques.
Physical Appearance
Clear, colorless, and sterile filtered solution.
Formulation
The NDUFA2 is supplied as a 1 mg/ml solution in a buffer consisting of 20mM Tris-HCl (pH 8.0), 0.15M NaCl, 10% glycerol, and 1mM DTT.
Stability
For short-term storage (2-4 weeks), keep at 4°C. For extended periods, store frozen at -20°C. Adding a carrier protein (0.1% HSA or BSA) is recommended for long-term storage. Avoid repeated freezing and thawing.
Purity
Purity is greater than 95% as determined by SDS-PAGE analysis.
Synonyms
NADH dehydrogenase [ubiquinone] 1 alpha subcomplex subunit 2, Complex I-B8, CI-B8, NADH-ubiquinone oxidoreductase B8 subunit, NDUFA2, CD14, CIB8.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSMAAAAAS RGVGAKLGLR EIRIHLCQRS PGSQGVRDFI EKRYVELKKA NPDLPILIRE CSDVQPKLWA RYAFGQETNV PLNNFSADQV TRALENVLSG KA.

Q&A

What is the structural role of NDUFA2 in mitochondrial complex I?

NDUFA2 is a supernumerary (accessory) subunit of mitochondrial complex I with a compact structure resembling a thioredoxin fold that includes a flat four-stranded beta-sheet. The protein contains 99 amino acids with a mass of approximately 10.9 kDa after processing of the amino-terminal methionine. NDUFA2 binds uniquely to the matrix arm of complex I, making exclusive contacts with the core subunit NDUFS1. Specifically, it binds to C-terminal domains of NDUFS1 and is positioned at least 40 Å from any of the iron-sulfur (FeS) clusters . This positioning suggests it plays a structural role rather than directly participating in electron transfer.

The protein contains two cysteine residues (Cys24 and Cys58) located in the loops of the beta-sheet, though these do not appear to be oxidized in existing structures of mammalian complex I . The primary contacts with NDUFS1 occur through the beta-sheet region, including both cysteine residues, suggesting these interactions are crucial for proper complex I assembly.

How do mutations in NDUFA2 affect complex I assembly and function?

Mutations in NDUFA2 can significantly impair complex I assembly and function through various mechanisms:

  • Assembly disruption: Knockout cell lines lacking NDUFA2 show limited assembly of the N-module of complex I, specifically losing core subunits NDUFS1, NDUFV1, and NDUFV2, as well as supernumerary subunits NDUFS4, NDUFS6, NDUFA7, and NDUFV3 .

  • Activity reduction: NDUFA2 knockout cell lines demonstrate extremely low complex I activity, consistent with a missing NADH binding site . This suggests that while NDUFA2 does not directly participate in electron transfer, it is essential for proper assembly of components that do.

  • Interface disruption: Specific mutations like p.Lys45Thr affect residues at the interface with NDUFS1, likely destabilizing binding between these subunits. Lys45 has close contacts with several NDUFS1 residues including Gly376, Asp380, and Ser672 .

  • Ion pair disruption: The p.Glu57Ala mutation affects a residue that forms a possible ion pair with Arg382 of NDUFS1 and makes numerous nonbonding contacts with other NDUFS1 residues (Gly661, Ala662, Asn663, Tyr664, Leu381, Arg382, and Ser383) .

What clinical phenotypes are associated with NDUFA2 mutations?

Several clinical phenotypes have been associated with pathogenic NDUFA2 mutations:

Clinical PresentationMutationPatient OutcomeReference
Leigh syndrome with hypertrophic cardiomyopathyc.208+5G>A (homozygous, causing exon 2 skipping)Death from cardiovascular arrest at 11 months
Leukoencephalopathyp.Lys45Thr (homozygous)Focal epilepsy at 6 years, wheelchair-bound at 9 years
LeukoencephalopathyCompound heterozygous: p.Lys45Thr and c.225del (p.Asn76Metfs*4)Movement difficulties, evaluated at age 4
Leukoencephalopathy with microcephaly, seizures, and movement disordersp.Glu57Ala (homozygous)Evaluated at age 4
Breast cancer (significance uncertain)p.Asp50Asn (homozygous)Not reported

Leukoencephalopathy (abnormal white matter in the brain) appears to be a common manifestation, often accompanied by neurological symptoms including seizures and movement disorders .

What techniques are most effective for studying NDUFA2's role in complex I assembly?

To study NDUFA2's role in complex I assembly, researchers should consider the following methodological approaches:

  • Blue Native gel electrophoresis (BN-PAGE): This technique has proven valuable for analyzing complex I assembly in NDUFA2 knockout cell lines, revealing limited assembly of the N-module of complex I . Researchers should optimize sample preparation to preserve native protein complexes and consider using a gradient gel (3-12% or 4-16%) to achieve good separation of high molecular weight complexes.

  • Knockout/knockdown models: CRISPR-Cas9 or RNAi approaches can be used to generate NDUFA2-deficient cell lines. The search results indicate that NDUFA2 knockout cell lines show impaired complex I assembly, particularly affecting the N-module . When designing knockout strategies, researchers should consider potential compensatory mechanisms.

  • Complex I activity assays: Spectrophotometric measurements of NADH:ubiquinone oxidoreductase activity are essential for assessing functional consequences of NDUFA2 deficiency. NDUFA2 knockout lines show extremely low complex I activity .

  • Subunit-specific antibodies: Immunoblotting with antibodies against various complex I subunits can help determine which components are affected by NDUFA2 mutations or absence. Focus on core subunits of the N-module (NDUFS1, NDUFV1, NDUFV2) as these are particularly affected by NDUFA2 deficiency .

  • Co-immunoprecipitation: This approach can help identify protein-protein interactions between NDUFA2 and other complex I subunits, particularly focusing on interactions with NDUFS1.

How can researchers effectively model NDUFA2 mutations for functional studies?

To model NDUFA2 mutations effectively:

  • Site-directed mutagenesis: Generate constructs expressing NDUFA2 with specific mutations identified in patients (e.g., p.Lys45Thr, p.Glu57Ala) . Use these for transfection into NDUFA2-null cell lines to assess rescue of complex I assembly and function.

  • Patient-derived cell lines: When available, fibroblasts or induced pluripotent stem cells (iPSCs) from patients with NDUFA2 mutations provide physiologically relevant models. These can be differentiated into neurons to study leukoencephalopathy phenotypes.

  • Structural analysis: Use the human complex I structure (PDB ID: 5xtd) to predict the impact of mutations on protein-protein interactions . For instance, analyzing how p.Lys45Thr might disrupt interactions with NDUFS1 residues Gly376, Asp380, and Ser672.

  • Protein stability assays: Pulse-chase experiments or cycloheximide chase assays can determine if mutations affect NDUFA2 protein stability.

  • Import assays: For mutations potentially affecting mitochondrial import (e.g., those in the transit peptide like p.Arg26Gln in NDUFV3), use in vitro import assays with isolated mitochondria to assess import efficiency .

  • RNA analysis: For mutations affecting splicing (e.g., c.208+5G>A), RT-PCR and sequencing of transcripts can verify aberrant splicing patterns .

What are the most reliable approaches for assessing NDUFA2 expression in tissue samples?

For reliable assessment of NDUFA2 expression:

  • qRT-PCR: Design primers spanning exon-exon junctions to avoid genomic DNA amplification. Consider analyzing all three exons of NDUFA2 separately, especially when investigating splicing mutations.

  • Western blotting: Use validated antibodies specific to NDUFA2. Consider using both N- and C-terminal antibodies when analyzing patient samples with mutations that might result in truncated proteins.

  • Immunohistochemistry: For tissue localization studies, validate antibodies thoroughly using positive and negative controls (e.g., NDUFA2 knockout tissues).

  • RNA-seq: For comprehensive expression analysis, use RNA-sequencing with appropriate depth (>30 million reads) to detect potential alternative splicing events.

  • Mass spectrometry: For protein-level confirmation, targeted mass spectrometry can provide quantitative data on NDUFA2 abundance and potential post-translational modifications.

When selecting methodology, consider that NDUFA2 mutations can result in complete absence of protein (null mutations) , making sensitive detection methods essential.

How do specific NDUFA2 mutations impact protein-protein interactions within complex I?

NDUFA2 mutations can disrupt protein-protein interactions within complex I through several mechanisms:

For experimental validation of these effects, researchers should consider using techniques such as hydrogen-deuterium exchange mass spectrometry (HDX-MS) or crosslinking mass spectrometry (XL-MS) to map interaction surfaces.

What is the current understanding of genotype-phenotype correlations in NDUFA2-related disorders?

The current understanding of genotype-phenotype correlations in NDUFA2-related disorders reveals several patterns:

  • Complete protein loss vs. missense mutations: Complete loss of NDUFA2 protein, as seen with the c.208+5G>A splicing mutation, appears to result in early-onset, severe phenotypes including Leigh syndrome and hypertrophic cardiomyopathy with death in infancy .

  • Interface residue mutations: Mutations affecting residues at the interface with NDUFS1 (p.Lys45Thr, p.Glu57Ala) cause leukoencephalopathy with variable neurological manifestations including seizures and movement disorders . The severity seems to depend on the specific residue affected and its importance in the protein-protein interaction.

  • Non-interface mutations: The p.Asp50Asn mutation, which affects a residue not at the NDUFS1 interface, has uncertain clinical significance . It was found in a breast cancer patient, but the connection to cancer pathogenesis remains unclear.

  • Compound heterozygosity: One reported case with compound heterozygous mutations (p.Lys45Thr and c.225del) showed leukoencephalopathy with movement difficulties . This suggests that having one missense mutation along with one frameshift mutation may be sufficient to cause disease.

The relationship between specific mutations and clinical outcomes remains incompletely understood, partly due to the small number of reported cases and potential influence of genetic modifiers.

How does NDUFA2 dysfunction contribute to neurological manifestations in mitochondrial disorders?

NDUFA2 dysfunction can contribute to neurological manifestations through several pathophysiological mechanisms:

  • Energy deficiency: Neurons have high energy demands, and NDUFA2 mutations that impair complex I assembly and function lead to reduced ATP production. In cell culture models, NDUFA2 knockout results in extremely low complex I activity .

  • White matter vulnerability: Leukoencephalopathy (white matter abnormalities) is a common feature in patients with NDUFA2 mutations . Oligodendrocytes, which produce myelin in the central nervous system, are particularly vulnerable to mitochondrial dysfunction due to their high metabolic requirements for lipid synthesis.

  • Oxidative stress: Dysfunctional complex I can increase reactive oxygen species (ROS) production. The absence of properly assembled N-module components (NDUFV1, NDUFV2, NDUFS1) in NDUFA2-deficient cells may lead to electron leakage and increased ROS, damaging neuronal membranes and proteins.

  • Developmental impacts: NDUFA2 mutations present with developmental features such as microcephaly , suggesting impacts on neuronal proliferation or survival during brain development.

  • Seizure susceptibility: Several patients with NDUFA2 mutations develop epilepsy . Neurons have limited glycolytic capacity and rely heavily on oxidative phosphorylation, making them susceptible to energy deficiency when complex I is dysfunctional.

Research approaches to further elucidate these mechanisms should include studies in neuronal models, particularly those derived from patient iPSCs, and imaging techniques to visualize mitochondrial function in affected neural cells.

What are promising approaches for therapeutic targeting of NDUFA2-related disorders?

Several promising therapeutic approaches for NDUFA2-related disorders warrant investigation:

When pursuing these approaches, researchers should consider the tissue-specific manifestations of NDUFA2 deficiency and the challenge of delivering therapeutics across the blood-brain barrier given the prominent neurological phenotypes.

How might systems biology approaches enhance our understanding of NDUFA2 in mitochondrial function?

Systems biology approaches can significantly enhance our understanding of NDUFA2's role in mitochondrial function:

  • Multi-omics integration: Combining proteomics, transcriptomics, and metabolomics data from NDUFA2-deficient models can reveal compensatory pathways and secondary effects beyond complex I assembly. Meta-analysis approaches similar to those used for colonic disease signatures could identify robust gene expression changes.

  • Network analysis: Mapping protein-protein interaction networks centered on NDUFA2 may reveal unexpected connections to other cellular processes. The known interactions with NDUFS1 represent only the primary contacts, but secondary effects on mitochondrial pathways remain largely unexplored.

  • Flux balance analysis: Computational modeling of metabolic fluxes in NDUFA2-deficient cells can predict changes in metabolic pathway utilization and identify potential therapeutic targets.

  • Tissue-specific modeling: Developing tissue-specific models of NDUFA2 function could help explain the predilection for neurological manifestations and identify tissue-specific vulnerabilities and compensatory mechanisms.

  • Evolutionary analysis: Comparative genomics across species can identify conserved features of NDUFA2 and related proteins, potentially revealing functional domains not obvious from structural studies alone.

  • Dynamic modeling: Simulating the assembly process of complex I with and without functional NDUFA2 could provide insights into the sequential steps disrupted by NDUFA2 mutations and identify potential intervention points.

These approaches should ideally be integrated with traditional biochemical and cell biological methods to validate predictions and generate new hypotheses.

What is the potential role of NDUFA2 in aging and neurodegenerative diseases?

NDUFA2's potential role in aging and neurodegenerative diseases represents an emerging area of research:

  • Mitochondrial theory of aging: Complex I dysfunction and resulting oxidative stress are implicated in aging processes. NDUFA2's role in complex I assembly suggests it may influence age-related mitochondrial decline. The observation that NDUFA2 deficiency affects N-module assembly , which contains the NADH binding site, suggests potential impacts on NADH/NAD+ ratios that influence many aging-related pathways.

  • Neurodegeneration connection: The leukoencephalopathy observed in NDUFA2 mutation carriers shares features with other neurodegenerative conditions. Investigating NDUFA2 expression and function in models of Alzheimer's, Parkinson's, and other neurodegenerative diseases could reveal whether it contributes to pathogenesis.

  • Oxidative stress resistance: The thioredoxin-like fold of NDUFA2 suggests potential redox-related functions that might influence cellular responses to oxidative stress, a common feature in neurodegenerative diseases.

  • Mitochondrial quality control: NDUFA2 dysfunction leads to complex I assembly defects , which might trigger mitochondrial quality control mechanisms including mitophagy. Dysregulation of these processes is implicated in both aging and neurodegeneration.

  • Somatic mutations: Investigating whether somatic mutations in NDUFA2 accumulate with age, particularly in post-mitotic tissues like neurons, could provide insights into tissue-specific aging processes.

Research methodologies should include longitudinal studies of NDUFA2 expression and function in aging tissues, analysis of NDUFA2 variants in neurodegenerative disease cohorts, and investigation of NDUFA2 interactions with known aging-related factors and pathways.

Product Science Overview

Introduction

NADH Dehydrogenase 1 Alpha Subcomplex 2, also known as NDUFAF2, is a protein encoded by the NDUFAF2 gene in humans. This protein is a crucial component of the mitochondrial respiratory chain complex I, which plays a vital role in cellular energy production. The recombinant form of this protein is often used in research to study its function and role in various diseases.

Structure and Function

NDUFAF2 acts as a molecular chaperone for the assembly of mitochondrial complex I . Complex I, also known as NADH:ubiquinone oxidoreductase, is the first enzyme in the mitochondrial electron transport chain. It catalyzes the transfer of electrons from NADH to ubiquinone, a process that is essential for the production of ATP, the primary energy currency of the cell .

The protein is located in the mitochondrial inner membrane and is involved in the transfer of electrons from NADH to the respiratory chain. The immediate electron acceptor for the enzyme is believed to be ubiquinone . The proper assembly and function of complex I are critical for maintaining cellular energy homeostasis.

Biological Significance

NDUFAF2 is essential for the normal functioning of cells. Mutations in the NDUFAF2 gene can lead to mitochondrial complex I deficiency, a condition characterized by defective oxidative phosphorylation. This deficiency is the most common biochemical signature of mitochondrial disorders, which can manifest in various clinical phenotypes, including neurodegenerative disorders, cardiomyopathy, and liver disease .

Recombinant Protein Production

The recombinant form of NDUFAF2 is produced using genetic engineering techniques. The gene encoding NDUFAF2 is cloned into an expression vector, which is then introduced into a suitable host cell, such as Escherichia coli or yeast. The host cells are cultured under conditions that promote the expression of the recombinant protein. The protein is then purified using various chromatographic techniques to obtain a high-purity product suitable for research purposes.

Applications in Research

Recombinant NDUFAF2 is widely used in research to study the assembly and function of mitochondrial complex I. It is also used to investigate the molecular mechanisms underlying mitochondrial disorders and to develop potential therapeutic strategies. Additionally, the recombinant protein can be used in structural studies to gain insights into the protein’s three-dimensional structure and its interactions with other components of the electron transport chain.

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.