NDUFAF2 Human

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

Description

Protein Characteristics

NDUFAF2 is a 20 kDa protein composed of 169 amino acids, encoded by the NDUFAF2 gene located on chromosome 5q12.1 . It functions as a molecular chaperone during the late stages of complex I assembly, associating with an 830 kDa subassembly to guide the integration of subunits .

PropertyDetail
GeneNDUFAF2 (aliases: B17.2L, NDUFA12L, mimitin)
Protein Size20 kDa (169 amino acids)
Subcellular LocationMitochondrial inner membrane
Primary RoleAssembly factor for complex I N-module stabilization

Complex I Assembly Mechanism

NDUFAF2 facilitates the attachment of the NADH-dehydrogenase (N) module to the nascent complex I structure. Structural studies reveal it acts as a flexible "lasso," capturing the N-module via its disordered regions and stabilizing it on the complex . This process is critical for electron transfer from NADH to ubiquinone and proton translocation across the inner mitochondrial membrane .

Mitochondrial Diseases

Mutations in NDUFAF2 are implicated in nuclear-type mitochondrial complex I deficiency, leading to severe neurological and systemic disorders. Key conditions include:

DisorderOMIMKey Features
Leigh Syndrome256000Progressive encephalopathy, brainstem lesions, elevated lactate
Progressive Encephalopathy618233Infantile-onset growth retardation, ophthalmological defects, apnea
Optic NeuropathyRetinal degeneration, optic nerve atrophy
Parkinson DiseaseAdult-onset neurodegeneration

Genetic Variants

The LOVD database reports 9 variants in NDUFAF2, including missense, nonsense, and deletion mutations. A recurrent 142-kb deletion (exons 2–4) causes loss of protein expression and severe infantile-onset brainstem neurodegeneration .

Cilia-Mitochondria Interplay

NDUFAF2 interacts with ARMC9, a centriole protein, to regulate primary cilia formation. In NDUFAF2-deficient cells:

  • Ciliary defects occur due to impaired CP110 removal and transition zone stabilization .

  • Mitochondrial dysfunction manifests as reduced oxygen consumption rate (OCR) and complex I activity .

Oxidative Stress and mtDNA Deletions

Ndufaf2 deficiency in murine models leads to:

  • Increased reactive oxygen species (ROS) .

  • Accumulation of mitochondrial DNA deletions, exacerbating cellular damage .

Protein Interactions and Partners

NDUFAF2 interacts with multiple mitochondrial and ciliary proteins, forming a network critical for organelle function:

Protein PartnerRole
ARMC9Regulates cilia formation via transition zone stabilization
CYB5BMitochondrial protein involved in electron transport
NDUFS4/6Competes with NDUFAF2 for N-module binding during assembly
SPG21Links mitochondria to ER via tethers (not directly studied in NDUFAF2)

Therapeutic Implications

Current therapies for NDUFAF2-related disorders are supportive (e.g., dietary management, respiratory support). Emerging strategies include:

  • Gene therapy: Re-expression of wild-type NDUFAF2 rescues ciliary and mitochondrial defects in vitro .

  • Chaperone therapy: Targeting complex I assembly to bypass NDUFAF2 deficiency.

Product Specs

Introduction
Mimitin, also known as NDUFAF2, belongs to the complex I NDUFA12 subunit family. This protein plays a crucial role in the mitochondrial electron transport chain as a component of NADH dehydrogenase. Located within the inner mitochondrial membrane, NADH dehydrogenase catalyzes the transfer of electrons from NADH to coenzyme Q (CoQ), initiating oxidative phosphorylation. Mimitin acts as a molecular chaperone, ensuring the proper assembly of mitochondrial complex I.
Description
Recombinant NDUFAF2 protein, expressed in E. Coli, is a single polypeptide chain devoid of glycosylation. It comprises 189 amino acids, with a sequence spanning from position 1 to 169, and possesses a molecular weight of 22kDa. For purification purposes, a 20 amino acid His-tag is fused to the N-terminus, and the protein is subsequently purified using proprietary chromatographic techniques.
Physical Appearance
The product is a sterile, filtered solution that appears colorless.
Formulation
The provided solution contains 0.5mg/ml of NDUFAF2 protein in a buffer consisting of 20mM Tris-HCl (pH 8.0), 1mM DTT, 20% glycerol, and 200mM NaCl.
Stability
For short-term storage (2-4 weeks), the product can be kept at 4°C. For extended storage, it is recommended to freeze the product at -20°C. The addition of a carrier protein (0.1% HSA or BSA) is advised for long-term storage. Avoid repeated freeze-thaw cycles to maintain protein integrity.
Purity
The purity of the NDUFAF2 protein is greater than 85.0%, as determined by SDS-PAGE analysis.
Synonyms

Mimitin mitochondrial, B17.2-like, B17.2L, Myc-induced mitochondrial protein, MMTN, NADH dehydrogenase [ubiquinone] 1 alpha subcomplex assembly factor 2, NDUFA12-like protein, NDUFAF2, NDUFA12L, mimitin.

Source
Escherichia Coli.
Amino Acid Sequence

MGSSHHHHHH SSGLVPRGSH MGWSQDLFRA LWRSLSREVK EHVGTDQFGN KYYYIPQYKN WRGQTIREKR IVEAANKKEV DYEAGDIPTE WEAWIRRTRK TPPTMEEILK NEKHREEIKI KSQDFYEKEK LLSKETSEEL LPPPVQTQIK GHASAPYFGK EEPSVAPSST GKTFQPGSWM PRDGKSHNQ.

Q&A

What is NDUFAF2 and what is its primary function in human cells?

NDUFAF2, also known as mimitin, is a 24 kDa protein with a functional mitochondrial targeting sequence that acts as an assembly factor for mitochondrial Complex I (NADH:ubiquinone oxidoreductase) . It bears significant homology to the Complex I subunit NDUFA12, suggesting evolutionary and functional relationships between these proteins . NDUFAF2 plays a crucial role in the efficient assembly and function of Complex I, which is the first and largest enzyme complex in the mitochondrial respiratory chain responsible for electron transfer from NADH to ubiquinone . While traditionally classified as an assembly factor, research indicates that NDUFAF2 influences both the assembly kinetics and functional stability of Complex I rather than being absolutely required for assembly completion . Deficiency in NDUFAF2 leads to selective reduction in Complex I activity without preventing the assembly of a fully mature Complex I enzyme, though the assembly process occurs with reduced efficiency .

How does NDUFAF2 contribute to mitochondrial Complex I assembly?

NDUFAF2 associates with a specific Complex I subassembly containing the central subunits of the Q module (NDUFS2, 3, 7, and 8) during the assembly process . Contrary to traditional assumptions about assembly factors, NDUFAF2-deficient cells can still assemble a fully mature Complex I enzyme, albeit with reduced kinetics and without evidence of intermediate or incomplete assembly . Cryo-EM structural studies reveal that NDUFAF2 occupies the position that will later be filled by NDUFA12 in the fully assembled complex, with its C-terminal end anchoring the N module to the Q module . The assembly factor prevents binding of NDUFS6 while it occupies the NDUFA12 position, as evidenced by a long helix that clashes with a loop connecting two major domains of NDUFS6 . This structural arrangement explains why NDUFAF2 must be released through the concerted action of NDUFS4, NDUFS6, and NDUFA12 for proper complex completion . The role of NDUFAF2 appears to be providing a platform for the attachment of NDUFS1, which is then firmly anchored by the C-terminal end of the assembly factor .

What clinical manifestations are associated with NDUFAF2 deficiency?

Homozygous deletion of NDUFAF2 leads to severe juvenile onset encephalopathy involving degeneration of the substantia nigra and other sub-cortical regions, resulting in adolescent lethality . Five patients with homozygous deletion of NDUFAF2 have been described, all developing severe encephalopathy primarily affecting the midbrain and brainstem, leading to premature death during childhood . Interestingly, a heterozygous NDUFAF2 deletion has been observed in a patient with attention-deficit/hyperactivity disorder, suggesting a link between Complex I deficits and basal ganglia dysfunction . This is consistent with the midbrain pathology observed in homozygous NDUFAF2 patients and points to a potential spectrum of neurological phenotypes associated with varying degrees of NDUFAF2 deficiency . The pathogenic mechanism appears to involve increased oxidative stress and mitochondrial DNA deletion rather than simply reduced ATP production, a pattern consistent with other Complex I deficiency disorders .

How can researchers effectively model NDUFAF2 deficiency in laboratory settings?

Researchers can employ several complementary models to study NDUFAF2 deficiency, including Ndufaf2-deficient human neuroblastoma cell lines and primary fibroblasts cultured from Ndufaf2 knock-out mice . These cellular models allow investigation of mitochondrial function, Complex I assembly kinetics, and pathological consequences at the cellular level . For genetic manipulation, site-directed mutagenesis techniques such as the QuickChange XL Site-Directed mutagenesis kit can be used to create specific mutations, such as changing the two methionine residues of NDUFAF2 to valine to study their functional significance . Expression constructs, including FLAG-tagged NDUFAF2 plasmids, are commercially available and can be used for overexpression studies or rescue experiments in deficient cells . When studying protein interactions, co-transfection approaches with tagged constructs followed by immunoprecipitation assays provide valuable insights into the NDUFAF2 interactome .

What techniques are most effective for studying NDUFAF2 protein interactions?

Immunoprecipitation assays represent a powerful approach for studying NDUFAF2 protein interactions in cellular contexts . Researchers can co-transfect cells (such as HEK293) with FLAG-NDUFAF2 and potential interaction partners tagged with reporters like turboGFP . After cell lysis with appropriate buffers (typically containing detergents like 1% NP-40), the lysates can be incubated with anti-FLAG antibodies followed by precipitation with Protein A beads . The precipitated complexes can then be analyzed by Western blotting using antibodies against the tags or specific proteins of interest . For identifying novel interaction partners, proximity labeling approaches using NDUFAF2 fused to enzymes like TurboID can be employed, allowing biotinylation of proximal proteins that can subsequently be purified and identified by mass spectrometry . For in vitro confirmation of direct interactions, recombinant his-tagged human NDUFAF2 protein can be produced using bacterial expression systems with vectors like pETDuet-1 .

How can researchers assess the impact of NDUFAF2 deficiency on Complex I assembly and function?

To evaluate Complex I assembly kinetics in NDUFAF2-deficient models, researchers can use blue native polyacrylamide gel electrophoresis (BN-PAGE) to visualize the formation of complex I and its intermediates over time . This technique allows for the detection of assembly intermediates that might accumulate in the absence of NDUFAF2 . For functional assessment, Complex I activity can be measured using spectrophotometric assays that monitor the oxidation of NADH, which is selectively reduced in NDUFAF2-deficient cells . Cryo-electron microscopy (cryo-EM) provides high-resolution structural insights into how NDUFAF2 deficiency affects Complex I architecture, revealing features such as the tilting of the N module or changes in solvent accessibility of iron-sulfur clusters . Electron paramagnetic resonance (EPR) spectroscopy can detect changes in the chemical environment of iron-sulfur clusters, which might explain altered electron transfer activities in mutant complexes .

What is the structural relationship between NDUFAF2 and the mature Complex I?

High-resolution cryo-EM structures have revealed that NDUFAF2 occupies the position that will later be filled by NDUFA12 in the fully assembled Complex I . The N-terminal core domain of NDUFAF2 in the assembly intermediate corresponds to the N-terminal core domain of NDUFA12 in wild-type complex I, confirming that NDUFA12 replaces NDUFAF2 during final assembly, as proposed based on evolutionary correlation . Beyond this domain, NDUFAF2 forms a long helix that points toward the distal part of the matrix arm, which would clash with a loop in NDUFS6, explaining why NDUFAF2 prevents binding of NDUFS6 while occupying the NDUFA12 position . Residues 122 to 223 of NDUFAF2 are not resolved in structural studies, suggesting this region may be disordered . The C-terminus of NDUFAF2 binds to subdomains 1 and 2 of the large C-terminal domain of NDUFS1, in the same position where the C-terminus of NDUFA12 binds in the native complex . This structural arrangement explains the assembly sequence where NDUFAF2 must be released through the concerted action of NDUFS4, NDUFS6, and NDUFA12 .

How does NDUFAF2 deficiency lead to oxidative stress and mitochondrial dysfunction?

NDUFAF2 deficiency results in significant increases in oxidative stress and mitochondrial DNA deletion, consistent with contemporary hypotheses regarding the pathophysiology of inherited mutations in Complex I disorders . In NDUFAF2-deficient models, compromised Complex I function leads to inefficient electron transfer, which increases electron leakage and subsequent reactive oxygen species (ROS) production . The severity of inherited Complex I diseases often correlates better with increases in leaked electrons from the mutant Complex I than with decreased ATP production, suggesting that oxidative damage, rather than energy deficiency, may be the primary pathogenic mechanism . The structural basis for increased ROS production may relate to altered solvent accessibility of iron-sulfur clusters in the Complex I electron transfer chain . In wild-type Complex I, NDUFS4 points toward cluster N3 in NDUFV1, and in its absence (as may occur in NDUFAF2 deficiency scenarios where proper assembly is compromised), both cluster binding sites become exposed and solvent accessible . Solvent-accessible surface area calculations on cryo-EM structures show that regions around clusters N3 and N1b are more exposed to solvent in mutant than in wild type, which explains both the different EPR spectra and the increased ROS formation and decreased electron transfer activity of mutant complexes .

What protein interactions does NDUFAF2 participate in beyond Complex I assembly?

Recent research has revealed that NDUFAF2 interacts with methionine sulfoxide reductase proteins, specifically MSRA and all three MSRBs . These interactions were demonstrated through co-immunoprecipitation assays using FLAG-tagged NDUFAF2 and turboGFP-tagged MSR proteins . The functional significance of these interactions may relate to protection against oxidative stress, as methionine sulfoxide reductases repair oxidized methionine residues in proteins, which could be particularly important in the context of mitochondrial function where ROS production occurs . NDUFAF2 contains two methionine residues that could be substrates for these repair enzymes, and mutation of these residues to valine has been used to study their functional significance . Beyond these specific interactions, NDUFAF2 has been shown to associate with a subassembly of Complex I containing central subunits of the Q module (NDUFS2, 3, 7, and 8) during the assembly process . The assembly factor remains associated with Complex I in patients carrying mutations in NDUFS4 and in NDUFS4-deletion models, suggesting additional attachment sites in the absence of this subunit .

How might advances in structural biology enhance our understanding of NDUFAF2 function?

Recent advances in cryo-electron microscopy (cryo-EM) have revolutionized our understanding of Complex I structure and assembly, revealing high-resolution details of how NDUFAF2 interacts with assembly intermediates . Future applications of this technology, possibly at even higher resolutions or in different physiological states, could resolve currently unknown structural features, such as the disordered region of NDUFAF2 (residues 122-223) that remains uncharacterized . Integrating cryo-EM with other structural techniques like X-ray crystallography might provide complementary insights, especially since different detergents used for protein purification can result in different loop conformations observed by these methods . Time-resolved structural studies that capture intermediate states during Complex I assembly would be particularly valuable for understanding the dynamic processes controlled by NDUFAF2 . Advanced computational approaches, including molecular dynamics simulations and solvent-accessible surface area calculations, have already provided insights into how mutations affect the chemical environment of iron-sulfur clusters . Expanding these computational approaches could help predict the effects of disease-causing mutations and potentially guide therapeutic development .

What therapeutic strategies might address NDUFAF2 deficiency-related disorders?

Therapeutic strategies for NDUFAF2 deficiency should target the downstream consequences, particularly oxidative stress and mitochondrial dysfunction . Antioxidant therapies might mitigate the increased reactive oxygen species production observed in NDUFAF2-deficient cells and tissues, potentially slowing disease progression . Gene therapy approaches could aim to restore NDUFAF2 expression in affected tissues, with particular attention to the central nervous system given the neurological manifestations of NDUFAF2 deficiency . Alternatively, overexpression of compensatory factors, such as NDUFA12 (which shares homology with NDUFAF2), might partially rescue Complex I assembly and function . Research on the interaction between NDUFAF2 and methionine sulfoxide reductases (MSRs) suggests another potential therapeutic avenue . Enhancing MSR activity might protect against oxidative damage to mitochondrial proteins, including Complex I components . Small molecule screens could identify compounds that stabilize partially assembled Complex I or enhance the residual activity of Complex I in NDUFAF2-deficient cells . Understanding the supramolecular organization of respiratory complexes in NDUFAF2 deficiency might also guide therapeutic approaches, as interactions with Complex III have been proposed to mitigate structural disorder in the membrane arm of mutant Complex I .

What controls and validations are essential when studying NDUFAF2 function?

When designing experiments to study NDUFAF2, researchers should include several critical controls and validations . For genetic manipulation studies, both positive controls (wild-type NDUFAF2 expression) and negative controls (empty vector or irrelevant protein expression) should be included to ensure specificity of observed effects . When generating NDUFAF2-deficient models through knockout or knockdown approaches, researchers should validate the efficiency of NDUFAF2 depletion at both mRNA and protein levels using RT-qPCR and Western blotting, respectively . Functional validation of NDUFAF2 deficiency should include measurement of Complex I activity using standardized spectrophotometric assays, with other respiratory chain complexes (II-V) measured as specificity controls . For protein interaction studies, researchers should employ multiple complementary techniques (co-immunoprecipitation, proximity labeling, in vitro binding assays) to confirm interactions and include controls for non-specific binding, such as IgG controls for immunoprecipitation . When expressing tagged versions of NDUFAF2 (such as FLAG-NDUFAF2), researchers should confirm that the tag does not interfere with protein localization or function through subcellular fractionation and functional rescue experiments .

How can researchers distinguish between primary and secondary effects of NDUFAF2 deficiency?

Distinguishing primary from secondary effects of NDUFAF2 deficiency requires careful experimental design and multiple complementary approaches . Time-course studies can help establish the sequence of events following NDUFAF2 depletion, with early changes more likely representing primary effects and later changes reflecting secondary consequences . Rescue experiments in which wild-type NDUFAF2 is reintroduced into deficient cells provide strong evidence for effects directly attributable to NDUFAF2 function; effects that are reversed upon NDUFAF2 reintroduction are likely primary consequences of its deficiency . Domain-specific or point mutant versions of NDUFAF2 can help map specific functions to particular regions of the protein, with different mutants potentially affecting different aspects of NDUFAF2 function . Comparative studies across multiple model systems (different cell types, in vitro vs. in vivo models) can help distinguish conserved primary effects from context-dependent secondary consequences . Systems biology approaches, including transcriptomics, proteomics, and metabolomics, can provide comprehensive views of cellular responses to NDUFAF2 deficiency, with network analysis potentially revealing direct versus indirect effects on different pathways .

Product Science Overview

Function and Mechanism

NDUFAF2 acts as a molecular chaperone, facilitating 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 (coenzyme Q10), which is then reduced to ubiquinol . This process is essential for the production of ATP, the primary energy currency of the cell.

The reaction catalyzed by complex I can be summarized as follows:

NADH+H++CoQ+4Hin+NAD++CoQH2+4Hout+\text{NADH} + \text{H}^+ + \text{CoQ} + 4\text{H}^+_{\text{in}} \rightarrow \text{NAD}^+ + \text{CoQH}_2 + 4\text{H}^+_{\text{out}}

In this process, complex I translocates four protons across the inner mitochondrial membrane per molecule of oxidized NADH, helping to build the electrochemical potential difference used to produce ATP .

Biological Significance

NDUFAF2 is involved in the assembly of mitochondrial NADH:ubiquinone oxidoreductase complex (complex I) . This protein is essential for the normal functioning of cells, and mutations in its subunits can lead to a wide range of inherited neuromuscular and metabolic disorders . Defects in this enzyme are responsible for the development of several pathological processes, including ischemia/reperfusion damage (stroke and cardiac infarction), Parkinson’s disease, and other neurodegenerative disorders .

Clinical Implications

Mutations in the NDUFAF2 gene can cause mitochondrial complex I deficiency, nuclear type 10 (MC1DN10) . This condition is characterized by defective oxidative phosphorylation, which affects the production of ATP. Clinical manifestations of mitochondrial complex I deficiency can vary widely, ranging from lethal neonatal disease to adult-onset neurodegenerative disorders . Symptoms may include macrocephaly with progressive leukodystrophy, non-specific encephalopathy, cardiomyopathy, myopathy, liver disease, Leigh syndrome, Leber hereditary optic neuropathy, and some forms of Parkinson’s disease .

Research and Therapeutic Potential

Understanding the function and mechanism of NDUFAF2 is crucial for developing therapeutic strategies for diseases associated with mitochondrial dysfunction. Research on NDUFAF2 and its role in mitochondrial complex I assembly may lead to the development of targeted therapies for conditions such as Parkinson’s disease and other mitochondrial disorders .

In conclusion, NADH Dehydrogenase 1 Alpha Subcomplex, Assembly Factor 2 (Human Recombinant) is a vital protein involved in the assembly and function of mitochondrial complex I. Its role in electron transport and ATP production underscores its importance in cellular energy metabolism and its potential as a therapeutic target for mitochondrial diseases.

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.