NDUFAF1 Human

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

Description

Molecular Structure and Localization

Mechanistic insights:

  • Binds ND3 subunit to lock its N-terminal helix, ensuring proper membrane arm conformation

  • Depletion reduces oxygen consumption rate (OCR) by 40–60% and increases ROS production

Clinical Implications of NDUFAF1 Mutations

Associated mitochondrial disorders:

Mutation TypePhenotypeKey Findings
Missense (c.278A>G)Fatal infantile cardiomyopathyLactic acidosis, complex I disassembly
Compound heterozygousLeukodystrophy, neuropathyWhite matter lesions, hyperreflexia
Biallelic lossLeigh syndrome, encephalopathyReduced ATP synthesis, developmental regression

Biomarker relevance:

  • Downregulated in pancreatic cancer (50% reduction in K-Ras-transformed cells)

  • Linked to metabolic reprogramming (Warburg effect) via NADH/ATP imbalance

Research Advancements

Key studies:

  1. Cryo-EM structural analysis revealed NDUFAF1’s role in preventing ND3 helix mispositioning, a defect linked to cardiomyopathy .

  2. siRNA knockdown models demonstrated:

    • 60% reduction in complex I activity

    • 2.5-fold ROS increase and glycolytic dependency

  3. K-Ras oncogene interaction: Suppresses NDUFAF1 expression, promoting tumor microenvironment adaptation .

Therapeutic targets under investigation:

  • Small molecules to enhance NDUFAF1-ECSIT interactions

  • Gene therapy vectors for NDUFAF1 missense mutations

Database and Pathway Integration

  • Pathway enrichment: Respiratory electron transport (GO:0022904), complex I biogenesis (R-HSA-6799198)

  • Variant database: 13 pathogenic variants cataloged in LOVD, including c.247G>A and c.278A>G

Product Specs

Introduction
NADH dehydrogenase [ubiquinone] 1 alpha subcomplex assembly factor 4 (NDUFAF4) plays a crucial role in assembling the mitochondrial NADH: ubiquinone oxidoreductase complex, also known as complex I. This protein is also involved in regulating cell proliferation and survival in hormone-dependent tumor cells. Additionally, NDUFAF4 may be implicated in the regulation of breast tumor cell invasion. Mutations in the NDUFAF4 gene are known to cause mitochondrial complex I deficiency.
Description
Recombinant human NDUFAF1, expressed in E. coli, is a single, non-glycosylated polypeptide chain. It consists of 326 amino acids (residues 25-327) and has a molecular weight of 37 kDa. The protein includes a 23 amino acid His-tag fused to its N-terminus and is purified using proprietary chromatographic techniques.
Physical Appearance
Clear, colorless, and sterile-filtered solution.
Formulation
The NDUFAF1 solution is provided at a concentration of 0.5 mg/ml. The solution is buffered with 20mM Tris-HCl at pH 8.0 and contains 0.15M NaCl, 10% glycerol, and 1mM DTT.
Stability
For short-term storage (2-4 weeks), the solution should be kept at 4°C. For extended storage, it is recommended to freeze the solution at -20°C. To further enhance long-term stability, consider adding a carrier protein such as HSA or BSA (0.1%). It is crucial to avoid repeated freeze-thaw cycles to maintain protein integrity.
Purity
The purity of NDUFAF1 is determined by SDS-PAGE analysis and is guaranteed to be greater than 90%.
Synonyms
Complex I intermediate-associated protein 30, mitochondrial, NADH dehydrogenase [ubiquinone] 1 alpha subcomplex assembly factor 1, NDUFAF1, CIA30, CGI-65, CGI65.
Source
Escherichia Coli.
Amino Acid Sequence

MGSSHHHHHH SSGLVPRGSH MGSYPFLGIR FAEYSSSLQK PVASPGKASS QRKTEGDLQG DHQKEVALDI TSSEEKPDVS FDKAIRDEAI YHFRLLKDEI VDHWRGPEGH PLHEVLLEQA KVVWQFRGKE DLDKWTVTSD KTIGGRSEVF LKMGKNNQSA LLYGTLSSEA PQDGESTRSG YCAMISRIPR GAFERKMSYD WSQFNTLYLR VRGDGRPWMV NIKEDTDFFQ RTNQMYSYFM FTRGGPYWQE VKIPFSKFFF SNRGRIRDVQ HELPLDKISS IGFTLADKVD GPFFLEIDFI GVFTDPAHTE EFAYENSPEL NPRLFK.

Q&A

What is NDUFAF1 and what is its primary function in human mitochondria?

NDUFAF1 is a mitochondrial protein that functions as a complex I assembly factor. As part of the MCIA (mitochondrial complex I assembly) complex, it is essential for the assembly of the P module of complex I. The protein is the human homolog of Neurospora crassa CIA30 and is required for proper assembly of NADH-ubiquinone oxidoreductase (complex I), which catalyzes the transfer of electrons from NADH to ubiquinone in the first step of the mitochondrial respiratory chain . Experimental evidence shows that modulating the intramitochondrial amount of NDUFAF1 by knocking down its expression using RNA interference leads to reduced amount and activity of complex I, confirming its critical role in complex I biogenesis .

What is the evolutionary relationship between human NDUFAF1 and homologs in other species?

Human NDUFAF1 shares approximately 28% homology with the Neurospora crassa complex I assembly chaperone CIA30 . While the fungal ortholog CIA84 is found only in fungi, CIA30's ortholog in humans is NDUFAF1 . Research using recombinant NDUFAF1 protein has shown highest antigen sequence identity to mouse and rat orthologs (88% for both) . This conservation across species highlights the evolutionary importance of this assembly factor for mitochondrial function. The structural conservation between NDUFAF1 and carbohydrate-binding domain family 11 (CBM11) proteins suggests that NDUFAF1 evolved from a carbohydrate-binding protein, with its binding cleft being repurposed for peptide binding - an example of how structural scaffolds can assume new functions through evolution .

How is NDUFAF1 involved in complex I assembly?

NDUFAF1 is associated with two protein complexes of approximately 600 and 700 kDa in size, with the 700 kDa complex potentially representing a key step in the complex I assembly process . In humans, NDUFAF1 is a component of the early MCIA intermediate, which is the starting point for the step-by-step assembly of the P module of complex I . Studies with the yeast model Yarrowia lipolytica show that deletion of NDUFAF1 results in severe reduction of complex I activity (to 14% for NADH:hexaammineruthenium activity), with ubiquinone reductase activity falling below detection limits . Complementation with the NDUFAF1 gene restores complex I expression to levels similar to the parental strain, confirming its essential role in complex I assembly .

What is the structural basis for NDUFAF1 function in complex I assembly?

Cryo-electron microscopy studies of assembly intermediates from Yarrowia lipolytica have revealed that NDUFAF1 locks the ND3 subunit in a conformation that differs distinctly from its native structure in mature complex I . Specifically, NDUFAF1 embeds the TMH1-2 loop of ND3 in a binding cleft on its concave side. This binding cleft structurally resembles that found in carbohydrate-binding domain family 11 proteins, although the residues involved in binding differ . The architecture of NDUFAF1 includes a β-sandwich structure, with disease-related mutations clustering on β strands 6 and 9 of the outer leaflet and in an adjacent loop . By securing the ND3 loop, NDUFAF1 prevents the adjacent transmembrane helix TMH1 from moving freely during assembly, which could otherwise interfere with the addition of new subunits or result in incorrect positioning .

What methodologies are most effective for isolating and studying NDUFAF1-associated assembly intermediates?

For isolation and characterization of NDUFAF1-associated complexes, researchers should employ a combination of genetic manipulation and biochemical techniques. In model systems like Yarrowia lipolytica, homologous recombination can be used to delete the NDUFAF1 gene, followed by complementation with a tagged version (e.g., Twin-Strep-tag) for affinity purification . Purification of assembly intermediates should use detergent solubilization of mitochondrial membranes followed by affinity chromatography.

For structural studies, cryo-electron microscopy has proven invaluable for determining the structures of assembly intermediates at near-atomic resolution . This approach allows visualization of the interactions between NDUFAF1 and complex I subunits, particularly the binding of the ND3 loop in the NDUFAF1 binding cleft. Two-dimensional blue-native gel electrophoresis combined with immunodetection is effective for analyzing the distribution of NDUFAF1 between the 600 and 700 kDa complexes, particularly when comparing wild-type and patient samples with complex I deficiency .

How do the 600 kDa and 700 kDa NDUFAF1-associated complexes differ in function during complex I assembly?

The 600 kDa and 700 kDa NDUFAF1-associated complexes represent different stages in complex I assembly. Research indicates that the relative distribution of NDUFAF1 between these two complexes is altered in patients with complex I deficiency, suggesting they have distinct roles in the assembly process .

To investigate these complexes, researchers should use blue-native gel electrophoresis followed by second-dimension SDS-PAGE and immunoblotting to identify the protein components of each complex. RNA interference to modulate NDUFAF1 levels can help determine how this affects the distribution between the two complexes. Pulse-chase experiments with radiolabeled mitochondrial proteins can track the assembly kinetics and determine the temporal relationship between the two complexes. Cross-linking mass spectrometry could identify the spatial relationships of proteins within each complex, helping to construct a detailed model of the assembly pathway .

What is the molecular mechanism by which NDUFAF1 stabilizes the ND3 subunit during complex I assembly?

NDUFAF1 stabilizes ND3 by securing its long TMH1-2 loop in a binding cleft, preventing uncontrolled positioning of the N-terminal ND3 helix during assembly . Structural studies have revealed that NDUFAF1 locks ND3 in a conformation that differs distinctly from its native structure in mature complex I.

The binding interaction involves several specific molecular contacts: the strictly conserved Glu39 of ND3 forms an ion pair with Arg84 of NDUFAF1, while Phe80 of NDUFAF1 interacts with a short hydrophobic segment of the TMH1-2 loop of ND3 . This interaction prevents the adjacent TMH1 of ND3 from moving freely during assembly, which could otherwise interfere with the addition of new subunits. Additionally, by securing the loop, NDUFAF1 may protect the conserved Cys40 residue of ND3 from unwanted modifications during assembly, which is important as alkylation of this residue causes irreversible loss of complex I activity .

How do specific disease-causing mutations in NDUFAF1 affect its function at the molecular level?

Disease-causing mutations in NDUFAF1 associated with complex I deficiency and cardioencephalomyopathy cluster on β strands 6 and 9 of the outer leaflet of the β-sandwich structure and in an adjacent loop . Interestingly, these positions are not in direct contact with the binding site for the ND3 loop.

To study the effects of these mutations, researchers should use site-directed mutagenesis to introduce specific disease-associated mutations into expression constructs, followed by structural analysis using techniques like X-ray crystallography or cryo-EM to determine how the mutations affect protein folding. Functional assays should include binding studies with synthetic ND3 peptides to measure changes in binding affinity, in vitro complex I assembly assays to assess the ability of mutant NDUFAF1 to support complex I formation, and cellular complementation studies in NDUFAF1-deficient cell lines to measure rescue of complex I activity. Molecular dynamics simulations could provide insights into how these mutations affect the dynamics and stability of the NDUFAF1 structure .

What is the relationship between NDUFAF1 and tafazzin during complex I assembly?

To investigate this relationship, researchers should use co-immunoprecipitation with antibodies against NDUFAF1 to determine if tafazzin physically interacts with NDUFAF1. Proximity labeling techniques like BioID or APEX could identify the spatial relationship between these proteins in intact mitochondria. Functional studies should examine how tafazzin affects cardiolipin remodeling in the context of complex I assembly, potentially using lipidomic analysis of purified complex I assembly intermediates from wild-type and NDUFAF1-deficient cells .

How can cryo-EM be utilized to study conformational changes during NDUFAF1-mediated complex I assembly?

Cryo-electron microscopy has proven to be a powerful tool for elucidating the structures of complex I assembly intermediates associated with NDUFAF1 . To implement this methodology effectively, researchers should:

  • Purify assembly intermediates using affinity tags on NDUFAF1 or associated components, ensuring sample homogeneity through techniques like size exclusion chromatography.

  • Optimize grid preparation conditions to achieve uniform particle distribution and ice thickness using techniques like Quantifoil grids with thin carbon support.

  • Collect high-resolution data on modern cryo-EM instruments equipped with direct electron detectors and energy filters to maximize signal-to-noise ratio.

  • Process data using software packages like RELION, cryoSPARC, or EMAN2 for particle picking, 2D classification, 3D reconstruction, and refinement.

  • Perform focused refinement on specific regions, such as the NDUFAF1-ND3 interface, to obtain higher resolution in areas of interest.

  • Compare structures of assembly intermediates at different stages to track conformational changes, particularly in the ND3 subunit as it transitions from NDUFAF1-bound to its final position in mature complex I .

What experimental approaches can be used to investigate the role of NDUFAF1 in protecting the Cys40 residue of ND3 during assembly?

The conserved Cys40 residue in the TMH1-2 loop of ND3 is embedded in the binding cleft of NDUFAF1 during complex I assembly, potentially protecting it from unwanted modifications . Alkylation of this residue in mature complex I causes irreversible loss of ubiquinone reductase activity . To investigate this protective role:

  • Use site-directed mutagenesis to create Cys40 variants in ND3 and assess their effects on complex I assembly and activity in the presence and absence of NDUFAF1.

  • Employ selective cysteine-labeling reagents to assess the accessibility and redox state of Cys40 in different assembly intermediates.

  • Develop an in vitro system to test whether NDUFAF1 binding to ND3 peptides prevents chemical modification of Cys40.

  • Use mass spectrometry to identify post-translational modifications of Cys40 in wild-type versus NDUFAF1-deficient cells.

  • Perform structure-function analysis using cryo-EM to visualize the precise positioning of Cys40 in the NDUFAF1 binding cleft and how it changes during complex I assembly .

What approaches can be used to study the evolution of NDUFAF1's binding cleft from carbohydrate-binding to peptide-binding function?

The binding cleft of NDUFAF1 structurally resembles that found in carbohydrate-binding domain family 11 (CBM11) proteins, suggesting evolutionary repurposing of a carbohydrate-binding scaffold for peptide binding . To investigate this evolutionary transition:

  • Perform comprehensive phylogenetic analysis of NDUFAF1 homologs across diverse eukaryotic lineages, identifying key transitional forms.

  • Use structural bioinformatics to model ancestral sequences and predict their binding preferences.

  • Express and purify recombinant versions of evolutionary intermediates and test their binding to both carbohydrates and peptides using techniques like isothermal titration calorimetry or surface plasmon resonance.

  • Create chimeric proteins combining domains from CBM11 and NDUFAF1 to identify the minimal structural elements required for the functional transition.

  • Utilize protein design approaches to create artificial evolutionary intermediates with dual binding specificity.

  • Test the function of these proteins in complementation assays using NDUFAF1-deficient cell lines to assess their ability to support complex I assembly .

How can conditional expression systems be used to study the dynamics of NDUFAF1-mediated complex I assembly?

Conditional expression systems offer powerful tools for studying the temporal aspects of complex I assembly mediated by NDUFAF1. Researchers have successfully used these systems to show that the 700 kDa NDUFAF1-associated complex may represent a key step in the complex I assembly process . To implement this approach:

  • Develop inducible expression systems for NDUFAF1 using tetracycline-responsive promoters or similar conditional systems.

  • Create cell lines with endogenous NDUFAF1 knocked out (using CRISPR-Cas9) and replaced with the inducible construct.

  • Use time-course experiments following induction to track the formation of assembly intermediates using blue-native PAGE and western blotting.

  • Combine with pulse-chase labeling of mitochondrial proteins to track the incorporation of newly synthesized subunits into complex I under NDUFAF1-controlled conditions.

  • Implement live-cell imaging with fluorescently tagged NDUFAF1 and complex I subunits to visualize the assembly process in real-time.

  • Analyze the kinetics of complex I assembly after NDUFAF1 induction, determining rate-limiting steps and the temporal sequence of assembly events .

What therapeutic approaches show promise for NDUFAF1-related mitochondrial disorders?

For NDUFAF1-related mitochondrial disorders such as complex I deficiency, several therapeutic approaches warrant investigation:

  • Gene therapy approaches using adeno-associated viral vectors to deliver functional NDUFAF1 gene copies to affected tissues, particularly focusing on the heart and central nervous system for cardioencephalomyopathy patients.

  • Small molecule chaperones designed to stabilize mutant NDUFAF1 proteins, identified through high-throughput screening of compound libraries.

  • Bypass therapies that enhance alternative bioenergetic pathways, such as succinate-based approaches that bypass complex I and feed electrons directly to complex II.

  • Mitochondrially-targeted antioxidants to mitigate the oxidative stress resulting from complex I dysfunction.

  • Metabolic modulation through ketogenic diets or similar approaches that reduce dependence on complex I-mediated energy production.

  • Peptide mimetics based on the ND3 loop sequence that could facilitate complex I assembly in the presence of defective NDUFAF1.

  • mRNA-based therapies delivering translatable NDUFAF1 mRNA directly to affected tissues .

Product Science Overview

Introduction

NADH Dehydrogenase 1 Alpha Subcomplex, Assembly Factor 1 (NDUFAF1) is a crucial protein involved in the assembly of Complex I of the mitochondrial respiratory chain. This complex, also known as NADH:ubiquinone oxidoreductase, is the largest of the five complexes in the electron transport chain and plays a pivotal role in cellular respiration and energy production.

Gene and Protein Structure

The NDUFAF1 gene is located on chromosome 15q15.1 in humans . It encodes a protein that is composed of 327 amino acids and contains an N-terminal mitochondrial targeting sequence . The protein is moderately conserved among species, with the highest homology observed in the C-terminal region .

Function

NDUFAF1 acts as an assembly factor for Complex I, facilitating the correct assembly of this complex within the mitochondrial inner membrane . Complex I catalyzes the transfer of electrons from NADH to ubiquinone (coenzyme Q), which is the first step in the mitochondrial respiratory chain . This process results in the translocation of protons across the inner mitochondrial membrane, contributing to the proton gradient that drives ATP synthesis.

Biological Significance

The proper assembly and function of Complex I are essential for efficient cellular respiration and energy production. Mutations in the NDUFAF1 gene can lead to mitochondrial Complex I deficiency, a condition associated with a range of clinical manifestations, including neurodegenerative diseases and metabolic disorders .

Research and Clinical Implications

Understanding the role of NDUFAF1 in the assembly of Complex I has significant implications for research and clinical practice. Studies on NDUFAF1 can provide insights into the mechanisms underlying mitochondrial diseases and potentially lead to the development of therapeutic strategies for conditions associated with Complex I deficiency .

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.