Uqcrq is encoded by the Uqcrq gene (HGNC:29594, NCBI Gene:27089) and is a low-molecular-weight ubiquinone-binding protein (~9.5 kDa) . Key features include:
Sequence and Structure: The rat protein (UniProt: Q7TQ16) shares 85% sequence identity with human and mouse orthologs. It contains a hydrophobic region critical for membrane integration and ubiquinone interaction .
Function:
Interactions: Binds cardiolipin via α-helices 2 and 3, contributing to mitochondrial membrane integrity and crista morphology .
Recombinant Uqcrq is produced using diverse systems, each with distinct advantages:
Partial Proteins: Truncated versions (e.g., aa 1–82) are used for antibody validation .
Tagged Proteins: His-tagged constructs (e.g., CSB-RP038554h) facilitate purification .
Electron Transport Chain (ETC) Analysis: Used to study CIII activity, proton pumping, and supercomplex dynamics .
Disease Modeling: Investigates mitochondrial complex III deficiencies linked to neurodegenerative disorders (e.g., Parkinson’s) .
Protein Interactions: Examines binding with cardiolipin and assembly factors like C11orf83 (UQCC3) .
Recombinant Uqcrq serves as a control for:
ELISA: Blocking experiments with antibodies (e.g., PA5-61285) .
Western Blot (WB): Verifies antibody specificity against CIII subunits .
Immunohistochemistry (IHC): Localizes Uqcrq in mitochondrial membranes .
Mitochondrial Morphology: Uqcrq depletion disrupts crista structure and reduces ATP levels, highlighting its role in respiratory chain efficiency .
Supercomplex Stabilization: Interacts with C11orf83 to maintain III₂/IV supercomplex integrity, critical for optimizing ETC flux .
Disease Links: Mutations in Uqcrq are associated with isolated complex III deficiency, underscoring its diagnostic relevance .
Folding Accuracy: Post-translational modifications (e.g., phosphorylation) may require mammalian systems for functional studies.
Therapeutic Potential: Targeting Uqcrq-stabilizing interactions (e.g., cardiolipin binding) could address mitochondrial disorders.
Rat Cytochrome b-c1 complex subunit 8 (Uqcrq) is a ubiquinone-binding protein of low molecular mass that functions as a subunit of mitochondrial complex III, an essential component of the electron transport chain. This protein plays a critical role in cellular energy production by facilitating electron transfer from ubiquinol to cytochrome c, contributing to the establishment of the proton gradient necessary for ATP synthesis .
Complex III consists of 11 subunits in mammals, with Uqcrq serving as one of the small core subunits that contribute to the structural stability and functional efficiency of the complex. Disruption of complex III subunits, as demonstrated with other subunits like UQCRC1, can lead to significant physiological consequences, highlighting the importance of each component including Uqcrq .
When expressing in bacterial systems, consideration should be given to:
Using a codon-optimized sequence for E. coli
Employing solubility-enhancing fusion tags (e.g., MBP, SUMO, or thioredoxin)
Adjusting induction conditions (lower temperature, 18-25°C, can improve folding)
Testing varying IPTG concentrations (0.1-1.0 mM range)
For more physiologically accurate studies, mammalian expression systems may be preferred, particularly when investigating protein-protein interactions or functional studies that require proper post-translational modifications.
A multi-step purification approach is recommended for obtaining high-purity recombinant Rat Uqcrq:
Initial capture: Affinity chromatography using His-tag, GST-tag, or other fusion tags
Intermediate purification: Ion exchange chromatography (typically anion exchange using Q-Sepharose)
Polishing step: Size exclusion chromatography to remove aggregates and obtain homogeneous protein
For structural studies, consider adding these additional steps:
Inclusion of reducing agents throughout purification to prevent oxidation of cysteine residues
Buffer optimization screening using differential scanning fluorimetry (DSF)
Limited proteolysis to identify stable domains if crystallization of full-length protein is challenging
On-column detergent exchange if the protein was solubilized with detergents
Final protein purity should be assessed by SDS-PAGE (aiming for >95% purity) and verified by mass spectrometry to confirm protein identity and integrity.
Validating the functional activity of recombinant Rat Uqcrq requires assessing its ability to incorporate into complex III and contribute to electron transport chain function. Methods include:
Complex III activity assay: Measuring the reduction of cytochrome c using spectrophotometric methods, similar to those used in studies of UQCRC1 . This typically involves monitoring absorbance changes at 550 nm in the presence of reduced ubiquinol.
Reconstitution studies: Incorporating purified Uqcrq into isolated mitochondria or liposomes containing other complex III components to assess restoration of function in complex III-deficient systems.
Binding assays: Using isothermal titration calorimetry (ITC) or surface plasmon resonance (SPR) to evaluate binding to other complex III subunits or to ubiquinone.
Protein-protein interaction studies: Co-immunoprecipitation or pull-down assays with other complex III components to verify proper protein-protein interactions.
When validating recombinant Uqcrq, comparison with native protein from rat mitochondria should be performed to ensure that the recombinant protein exhibits similar properties.
Dysregulation of complex III subunits, including potential dysregulation of Uqcrq, can significantly impact mitochondrial function and contribute to disease pathogenesis. Studies of other complex III subunits provide important insights:
Mitochondrial dysfunction contribution: Alterations in complex III subunits like UQCRC1 have been associated with decreased complex III formation, reduced complex III activity, and decreased ATP content . Similar consequences might be expected with Uqcrq dysregulation.
Neurological implications: Research with UQCRC1 heterozygous mice demonstrated increased vulnerability to brain ischemia, impaired learning and memory, reduced mitochondrial membrane potential, and increased free radical production . These findings suggest that Uqcrq disruption might similarly affect neurological function.
Cancer relevance: Multiple complex III subunits, including UQCRFS1 and UQCRC1, show downregulation in clear cell renal cell carcinoma (ccRCC), suggesting that alterations in complex III, potentially including Uqcrq, may contribute to cancer metabolism reprogramming .
Recombinant Uqcrq can be utilized to study these conditions through:
Reconstitution experiments in patient-derived mitochondria
Development of competitive inhibitors to model partial Uqcrq deficiency
Creation of biosensors to monitor complex III assembly in real-time
Screening for small molecules that stabilize complex III in the presence of mutant subunits
Based on studies with related complex III subunits, several gene editing approaches can be effectively applied to study Uqcrq function:
Conditional knockout models: Complete knockout of UQCRC1, another complex III subunit, resulted in embryonic lethality , suggesting that Uqcrq complete knockout might also be lethal. Therefore, conditional knockout systems (Cre-loxP) allowing tissue-specific or temporally controlled Uqcrq deletion would be more informative.
Heterozygous models: UQCRC1 heterozygous mice survived but exhibited phenotypic consequences including decreased complex III activity and poor performance in learning/memory tests . A similar approach with Uqcrq might reveal its function while avoiding embryonic lethality.
CRISPR-Cas9 point mutations: Introducing specific mutations that mimic disease-associated variants or target functional domains can provide insights into structure-function relationships without completely abolishing expression.
Knockin reporter systems: Tagging endogenous Uqcrq with fluorescent proteins or epitope tags can enable tracking of protein localization, turnover, and interactions in living cells or animals.
When designing gene editing studies, researchers should consider:
Potential compensatory mechanisms from other complex III subunits
The need for appropriate controls including wild-type and heterozygous animals
Confirmation of editing efficiency through genomic sequencing and protein expression analysis
Phenotypic characterization at multiple levels (molecular, cellular, physiological, behavioral)
Post-translational modifications (PTMs) likely play important roles in regulating Uqcrq function and complex III assembly, although specific data for rat Uqcrq is limited. Based on studies of the complex III family:
Phosphorylation: Phosphorylation of complex III subunits can modulate enzyme activity, protein stability, and interactions with other mitochondrial proteins. Mass spectrometry-based phosphoproteomic approaches can identify potential phosphorylation sites on Uqcrq.
Ubiquitination: This modification may regulate Uqcrq turnover and could be involved in quality control mechanisms ensuring proper complex III assembly. Proteasome inhibitors and ubiquitin mutants can be used to study this process.
Acetylation: Mitochondrial proteins are frequently regulated by acetylation/deacetylation cycles, often mediated by sirtuins. Acetylation could affect Uqcrq stability or its interaction with other complex III components.
Oxidative modifications: As part of the electron transport chain, complex III subunits including Uqcrq are exposed to reactive oxygen species, which can lead to oxidative modifications affecting protein function.
To study these PTMs, researchers can employ:
Site-directed mutagenesis of potential PTM sites in recombinant Uqcrq
Mass spectrometry to identify and quantify PTMs under different physiological conditions
Pharmacological or genetic manipulation of enzymes responsible for adding or removing PTMs
In vitro enzymatic assays to demonstrate the functional consequences of specific modifications
Based on studies of related complex III subunits, several factors likely influence Uqcrq expression in neural tissues:
Tissue-specific regulation: Different brain regions may express varying levels of Uqcrq based on their metabolic demands and vulnerability to oxidative stress. Studies of UQCRC1 demonstrated its abundant expression in neurons and astrocytes , suggesting that Uqcrq might have a similar expression pattern.
Developmental regulation: Expression patterns may change during different developmental stages, reflecting the varying energy demands during neural development and maturation.
Epigenetic regulation: DNA methylation has been shown to influence expression of other complex III subunits. For example, UQCRFS1 and UQCRC1 expression levels were inversely correlated with DNA CpG island hypermethylation in cancer tissues . Similar mechanisms may regulate Uqcrq expression in neural tissues.
Response to metabolic state: Expression may be modulated by cellular energy status, potentially through signaling pathways that sense ATP levels or redox state.
Methodological approaches to study these factors include:
RT-qPCR and Western blot analysis of different brain regions during development
Single-cell RNA sequencing to identify cell type-specific expression patterns
ChIP-seq to map transcription factor binding and chromatin modifications at the Uqcrq promoter
Reporter gene assays to identify regulatory elements controlling Uqcrq expression
Correlation of expression levels with functional parameters such as oxygen consumption rate, ATP production, and membrane potential
Recombinant Rat Uqcrq can serve as a valuable tool in high-throughput screening (HTS) for mitochondrial therapeutics through several approaches:
Assembly assays: Developing fluorescence-based assays to monitor complex III assembly in the presence of candidate compounds. This could involve FRET pairs on Uqcrq and interacting subunits to detect proper complex formation.
Activity-based screens: Utilizing recombinant Uqcrq in reconstituted systems to measure complex III activity when exposed to compound libraries. Activity can be monitored through cytochrome c reduction rates.
Binding assays: Screening for molecules that stabilize Uqcrq binding to other complex III components or that prevent pathological protein-protein interactions.
Cellular reporter systems: Creating cell lines with Uqcrq fused to luminescent or fluorescent reporters to monitor protein stability, localization, or function in response to compounds.
Key considerations for developing HTS assays include:
Assay robustness with Z' factors >0.5
Miniaturization to 384- or 1536-well format
Implementation of counter-screens to eliminate false positives
Secondary assays in more physiologically relevant systems to validate hits
Structure-activity relationship studies to optimize lead compounds
Aggregation of recombinant Uqcrq during expression and purification is a common challenge given its hydrophobic nature as a membrane-associated protein. To minimize aggregation:
Expression optimization:
Lower induction temperature (16-20°C)
Reduce inducer concentration
Use specialized E. coli strains designed for membrane proteins (C41(DE3), C43(DE3))
Consider co-expression with chaperones (GroEL/GroES, DnaK/DnaJ)
Purification strategies:
Include appropriate detergents throughout purification (DDM, LMNG, or Fos-choline)
Maintain reducing conditions with DTT or TCEP
Add glycerol (10-20%) to stabilize the protein
Consider purifying in the presence of lipids or amphipols
Implement on-column refolding if necessary
Analytical methods to monitor aggregation:
Dynamic light scattering
Size exclusion chromatography with multi-angle light scattering (SEC-MALS)
Negative-stain electron microscopy to visualize protein particles
By implementing these strategies, researchers can increase the yield of properly folded, functional recombinant Uqcrq suitable for downstream applications.
For studying Uqcrq interactions with other Complex III components, researchers should consider these complementary approaches:
In vitro binding assays:
Pull-down assays using recombinant tagged proteins
Surface plasmon resonance (SPR) for kinetic and affinity measurements
Isothermal titration calorimetry (ITC) for thermodynamic parameters
Microscale thermophoresis (MST) for interactions in solution
Structural approaches:
X-ray crystallography of reconstituted subcomplexes
Cryo-electron microscopy of intact complex III
Hydrogen-deuterium exchange mass spectrometry (HDX-MS) to map interaction interfaces
Crosslinking coupled with mass spectrometry to identify proximity relationships
Cellular approaches:
Bioluminescence resonance energy transfer (BRET) or fluorescence resonance energy transfer (FRET)
Proximity ligation assay (PLA) to visualize interactions in situ
Co-immunoprecipitation from mitochondrial fractions
Split-reporter systems (BiFC, split-luciferase) for detecting interactions in living cells
Researchers should employ multiple complementary methods to validate interactions and characterize their functional significance in the context of complex III assembly and function.
Single-cell technologies offer unprecedented opportunities to understand Uqcrq function in heterogeneous tissues like brain, where cell type-specific effects may be masked in bulk analyses. Future directions include:
Single-cell transcriptomics (scRNA-seq) to:
Map Uqcrq expression patterns across diverse cell types
Identify correlations between Uqcrq and other mitochondrial genes
Discover cell populations particularly vulnerable to Uqcrq dysregulation
Track expression changes during development or disease progression
Single-cell proteomics to:
Quantify Uqcrq protein levels in individual cells
Identify cell type-specific post-translational modifications
Correlate protein expression with functional parameters
Spatial transcriptomics/proteomics to:
Preserve spatial context of Uqcrq expression patterns
Identify regional variations within tissues
Correlate expression with microenvironmental factors
Single-cell functional analyses:
Measure mitochondrial membrane potential in Uqcrq-expressing vs. non-expressing cells
Assess ROS production at single-cell resolution
Correlate ATP levels with Uqcrq expression
These approaches would help elucidate cell type-specific roles of Uqcrq and identify the most vulnerable cell populations in disease states, potentially revealing new therapeutic targets.
Based on studies of related complex III subunits, Uqcrq likely plays an important role in neurodegeneration and brain ischemia protection. Future research directions should investigate:
Neuroprotective mechanisms:
The role of Uqcrq in maintaining mitochondrial function during oxidative stress
Whether Uqcrq overexpression can protect against neuronal death similar to the protective effects observed with UQCRC1 overexpression against oxygen-glucose deprivation
The interaction between Uqcrq and mitochondrial quality control pathways
Translational applications:
Development of small molecules that stabilize Uqcrq or enhance its function
Cell-penetrating peptides derived from Uqcrq that might preserve complex III function
Gene therapy approaches to upregulate Uqcrq in vulnerable brain regions
Biomarker potential:
Whether Uqcrq levels in biofluids correlate with neurodegeneration or ischemic vulnerability
If Uqcrq modifications (oxidation, phosphorylation) indicate mitochondrial stress
Genetic associations:
Analyzing whether Uqcrq polymorphisms associate with stroke outcomes or neurodegenerative disease risk