UQCRQ Human

Ubiquinol-Cytochrome C Reductase Human Recombinant
Shipped with Ice Packs
In Stock

Description

Biochemical Characteristics

UQCRQ Human is a low-molecular-weight protein with distinct structural and functional properties:

PropertyDetails
Molecular Weight12.3 kDa
Amino Acid Sequence105 residues (1–82a.a) with a 23-amino-acid His-tag at the N-terminus
SourceRecombinant production in Escherichia coli
PurificationProprietary chromatographic techniques
Storage4°C for short-term use; -20°C for long-term storage with carrier proteins

The protein’s sequence includes conserved motifs critical for ubiquinone binding and interaction with other Complex III subunits. Its non-glycosylated nature reflects its mitochondrial localization .

Clinical Significance

Mutations in UQCRQ are associated with mitochondrial Complex III deficiency, a rare metabolic disorder. Key findings include:

Reported Mutations and Phenotypes

MutationInheritanceSymptomsSource
Ser45Phe (S45F)Autosomal recessiveHypoglycemia, lactic acidosis, hypotonia, severe neurological impairment
4-bp deletion (p.338-341)Autosomal recessiveSimilar to S45F; observed in consanguineous families

Patients present with neonatal-onset metabolic crises, elevated lactate levels, and impaired Complex III activity in fibroblasts. Notably, mutations disrupt Complex III stability, leading to reduced enzymatic activity and accumulation of TCA cycle intermediates .

Functional Insights

  • Role in Complex III Assembly: UQCRQ interacts with core subunits (e.g., UQCRC1/2) and stabilizes the complex’s structure. Defects impair electron transfer and proton translocation .

  • Pathogen Interactions: Oxidative modifications of UQCRQ have been observed in Trypanosoma cruzi-infected cardiac tissues, suggesting a role in mitochondrial dysfunction during infections .

Genetic Variants

The Leiden Open Variation Database (LOVD) reports four public variants in UQCRQ, including missense and frameshift mutations linked to Complex III deficiency. These variants are rare and predominantly found in consanguineous populations .

Tissue Expression

UQCRQ is ubiquitously expressed in human tissues, with cytoplasmic localization:

TissueExpression LevelRelevance
LiverHighCentral role in oxidative metabolism
Heart MuscleHighCritical for energy demand in cardiomyocytes
Skeletal MuscleModerateSupports ATP production during physical activity
Brain (Cerebral Cortex)ModerateImplicated in neurological symptoms of Complex III deficiency

Data from the Human Protein Atlas confirm its widespread distribution, aligning with its essential role in mitochondrial energy production .

Disease Associations

UQCRQ mutations cause Mitochondrial Complex III Deficiency, Nuclear Type 4 (MC3DN4), characterized by:

  • Biochemical Hallmarks: Reduced Complex III activity, decreased oxygen consumption in fibroblasts, and elevated urinary dicarboxylic acids .

  • Genetic Basis: Autosomal recessive inheritance, with homozygous mutations in UQCRQ .

Experimental Applications

Recombinant UQCRQ Human (Cat# ENZ-746) is used in:

  • Structural Studies: Analyzing interactions with ubiquinone and Complex III subunits .

  • Diagnostic Assays: Detecting mutations or post-translational modifications in patient samples .

Product Specs

Introduction
Ubiquinol-Cytochrome C Reductase (UQCRQ), a subunit of the ubiquinol-cytochrome c reductase complex III in the mitochondrial respiratory chain, is a small core-associated protein. It functions as a ubiquinone-binding protein with low molecular mass.
Description
Recombinant human UQCRQ, expressed in E. coli, is a single, non-glycosylated polypeptide chain. It consists of 105 amino acids (1-82a.a), including a 23 amino acid His-tag at the N-terminus, and has a molecular weight of 12.3 kDa. The protein is purified using proprietary chromatographic techniques.
Physical Appearance
A clear, sterile-filtered solution.
Formulation
The UQCRQ solution is provided at a concentration of 0.5 mg/ml in a buffer containing 20 mM Tris-HCl (pH 8.0), 10% glycerol, and 0.4 M urea.
Stability
For short-term storage (2-4 weeks), the product can be stored at 4°C. For long-term storage, it is recommended to store the protein at -20°C. The addition of a carrier protein (0.1% HSA or BSA) is recommended for long-term storage. Avoid repeated freeze-thaw cycles.
Purity
The purity of the protein is determined to be greater than 85% by SDS-PAGE analysis.
Synonyms
Cytochrome b-c1 complex subunit 8, Complex III subunit 8, Complex III subunit VIII, Ubiquinol-cytochrome c reductase complex 9.5 kDa protein, Ubiquinol-cytochrome c reductase complex ubiquinone-binding protein QP-C, Ubiquinol-Cytochrome C Reductase, UQCRQ.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSMGREFGN LTRMRHVISY SLSPFEQRAY PHVFTKGIPN VLRRIRESFF RVVPQFVVFY LIYTWGTEEF ERSKRKNPAA YENDK.

Q&A

What is UQCRQ and what is its role in mitochondrial function?

UQCRQ is a 9.9 kDa protein composed of 82 amino acids that functions as a subunit of mitochondrial Complex III in the electron transport chain. It is encoded by the UQCRQ gene located on chromosome 5q31.1, spanning 2,217 base pairs. This transmembranous protein serves as a ubiquinone-binding component and plays a critical role in the electron transfer reaction between cytochrome c1 and cytochrome c, facilitating oxidative phosphorylation and cellular energy production . As a small core-associated protein within Complex III, UQCRQ contributes to maintaining the structural integrity and functional efficiency of the respiratory chain .

How does the molecular structure of UQCRQ relate to its function?

UQCRQ's structure features transmembrane domains with greater mass positioned on the matrix side of the inner mitochondrial membrane. This strategic positioning enables UQCRQ to participate effectively in ubiquinone binding and electron transfer processes. The protein's relatively small size (9.5 kDa) belies its importance in maintaining proper Complex III configuration and function. Research methodologies to investigate its structure-function relationship typically include X-ray crystallography, nuclear magnetic resonance spectroscopy, and molecular dynamics simulations. Researchers should consider using site-directed mutagenesis approaches to systematically analyze how specific structural domains contribute to electron transport efficiency .

What experimental approaches are most effective for measuring UQCRQ expression levels?

For quantitative assessment of UQCRQ expression:

  • Transcriptional analysis: RT-qPCR using primers targeting conserved regions of UQCRQ mRNA provides reliable quantification of gene expression.

  • Protein detection: Western blotting using specific anti-UQCRQ antibodies can determine protein abundance, while immunohistochemistry/immunofluorescence visualizes spatial distribution in tissues.

  • High-throughput approaches: RNA-sequencing captures expression patterns across multiple conditions, while proteomics using tandem mass spectrometry can measure absolute protein quantities and identify post-translational modifications.

  • Epigenetic profiling: Methylation-specific PCR and bisulfite sequencing are valuable for analyzing UQCRQ promoter methylation status, particularly relevant in cancer research where hypermethylation-induced downregulation has been observed .

What clinical manifestations are associated with UQCRQ mutations?

Mutations in the UQCRQ gene have been linked to Complex III deficiency, nuclear type 4, characterized by severe neurological disorders. Clinical features documented in patients with UQCRQ mutations include:

  • Neurological symptoms: Progressive encephalopathy, hypotonia, and developmental delays

  • Metabolic abnormalities: Recurrent hypoglycemia, lactic acidosis, and hyperammonemia

  • Other manifestations: Seizures and failure to thrive

In one documented set of cases involving twenty consanguineous patients with a Ser45Phe mutation in UQCRQ, the phenotype displayed an autosomal recessive inheritance pattern. Another reported variant was a homozygous 4-bp deletion at position p.338-341. These mutations resulted in mitochondrial Complex III deficiency with severe neurological symptoms, demonstrating the critical importance of UQCRQ in normal neurological development and function .

How do researchers distinguish between primary UQCRQ defects and secondary mitochondrial dysfunction?

Distinguishing primary from secondary defects requires a multi-faceted approach:

  • Genetic testing: Whole exome/genome sequencing identifies pathogenic variants in UQCRQ, with segregation analysis in families confirming inheritance patterns.

  • Biochemical assessment: Complex III activity measurement using spectrophotometric assays in affected tissues (muscle biopsies, fibroblasts) reveals specific deficiencies.

  • Supercomplex analysis: Blue native PAGE electrophoresis can identify abnormal Complex III assembly and aberrant supercomplex formation characteristic of UQCRQ deficiency.

  • Functional complementation: Lentiviral rescue experiments with wild-type UQCRQ in patient-derived cells can confirm pathogenicity of identified variants and rule out secondary defects.

  • Biomarker profiling: Comprehensive analysis of lactate, ammonia, and amino acid profiles provides biochemical signatures typical of UQCRQ deficiency .

What cellular mechanisms link UQCRQ dysfunction to tissue pathology?

The pathophysiological cascade connecting UQCRQ dysfunction to tissue damage involves:

  • Impaired electron transport: Defective UQCRQ disrupts Complex III function, reducing efficiency of electron transfer and decreasing ATP production.

  • Oxidative stress: Electron leakage from dysfunctional Complex III increases reactive oxygen species (ROS) production, damaging proteins, lipids, and DNA. Evidence from Trypanosoma cruzi infection studies shows that UQCRQ subunit undergoes oxidative modification in cardiac muscle tissue .

  • Metabolic derangement: Energy deficit shifts metabolism toward anaerobic glycolysis, increasing lactate production and disrupting ammonia clearance.

  • Mitochondrial membrane depolarization: Loss of membrane potential triggers mitochondrial quality control pathways and potentially apoptosis in severely affected cells.

  • Tissue-specific vulnerability: High-energy demanding tissues (brain, muscle, liver) show greater susceptibility to UQCRQ deficiency, explaining the predominant neurological phenotype in patients .

Advanced Experimental Models and Approaches

Researchers should consider the following cellular models when studying UQCRQ dysfunction:

  • Patient-derived fibroblasts: Primary cells from affected individuals provide physiologically relevant models that maintain patient-specific genetic background. These can be maintained in high glucose DMEM supplemented with uridine (200 μM) to support pyrimidine synthesis in cells with mitochondrial dysfunction .

  • CRISPR-engineered cell lines: Introduction of specific UQCRQ mutations in relevant cell types (neuronal cells, myoblasts, hepatocytes) allows systematic study of mutation-specific effects.

  • Inducible expression systems: Doxycycline-regulated expression (100-200 μg/ml for 72 hours) of wild-type UQCRQ in mutant backgrounds enables temporal control for rescue experiments and mechanistic studies .

  • Cancer cell models: KMRC2 renal cell carcinoma line demonstrates hypermethylation-induced UQCRQ extinction, making it suitable for studying UQCRQ's role in cancer metabolism. Ectopic overexpression of UQCRQ in this line restored mitochondrial membrane potential, increased oxygen consumption, and attenuated the Warburg effect .

  • iPSC-derived models: Patient-specific induced pluripotent stem cells differentiated into disease-relevant lineages provide platforms for developmental studies and drug screening.

What biochemical assays provide the most reliable assessment of UQCRQ-related Complex III function?

Comprehensive functional assessment requires multiple complementary approaches:

  • Enzymatic activity assays:

    • Spectrophotometric measurement of Complex III activity (ubiquinol-cytochrome c reductase) in isolated mitochondria or permeabilized cells

    • Polarographic determination of oxygen consumption using Clark-type electrodes or Seahorse XF analyzers with substrate-specific inhibitors

  • Respiratory chain complex analysis:

    • Blue native PAGE to assess Complex III assembly state and molecular weight of fully assembled holoenzyme

    • Identification of abnormal supercomplex formations (such as the large supercomplex SXL comprising one Complex I dimer and one Complex III dimer observed in UQCRH deficiency)

  • Mitochondrial integrity assessment:

    • Membrane potential measurement using potentiometric fluorescent dyes (TMRM, JC-1)

    • Mitochondrial morphology analysis through electron microscopy or confocal imaging

  • Metabolic flux analysis:

    • 13C-labeled substrate tracing to map metabolic rewiring

    • Lactate production measurements to quantify glycolytic shift

How does altered UQCRQ expression contribute to cancer metabolism?

UQCRQ downregulation plays a significant role in promoting the Warburg effect in renal cell carcinoma:

  • Epigenetic silencing mechanism: The Cancer Genome Atlas (TCGA) identified UQCRQ as the top-ranked gene showing inverse correlation between DNA hypermethylation and mRNA downregulation in clear cell renal cell carcinoma (ccRCC) .

  • Metabolic reprogramming: Loss of UQCRQ expression impairs mitochondrial membrane potential and reduces oxidative phosphorylation capacity, forcing cells to rely more heavily on glycolysis even in oxygen-rich conditions (Warburg effect) .

  • Experimental evidence: Ectopic overexpression of UQCRQ in the KMRC2 ccRCC cell line (which displays hypermethylation-induced UQCRQ extinction) demonstrated:

    • Restoration of mitochondrial membrane potential

    • Increased oxygen consumption

    • Attenuation of the Warburg effect

    • Higher apoptosis rates

    • Slower in vitro and in vivo tumor growth

  • Context-dependent effects: UQCRQ knockout in the 786O ccRCC cell line had minimal impact on metabolism and proliferation, suggesting UQCRQ's role becomes dispensable in cells that have entered a Warburg-like state through other mechanisms .

What methodologies are most appropriate for studying UQCRQ in cancer contexts?

Cancer-focused UQCRQ research requires specialized approaches:

  • Methylation analysis:

    • Bisulfite sequencing of UQCRQ promoter regions

    • Methylation-specific PCR for rapid screening

    • Chromatin immunoprecipitation to identify transcriptional regulators

  • Expression manipulation strategies:

    • Lentiviral vectors for stable UQCRQ overexpression in hypermethylated cell lines

    • CRISPR/Cas9-mediated knockout to assess the necessity of residual UQCRQ

    • Demethylating agents (5-azacytidine, decitabine) to reverse epigenetic silencing

  • Metabolic phenotyping:

    • Seahorse XF analysis to measure oxygen consumption rate and extracellular acidification rate

    • Glucose uptake and lactate production quantification

    • Mitochondrial function assessment through membrane potential and ROS production

  • In vivo cancer models:

    • Xenograft tumor growth assessment with UQCRQ-modulated cell lines

    • Patient-derived xenografts to maintain tumor heterogeneity

    • Metabolic imaging using PET tracers to assess glycolytic dependency

How can UQCRQ expression patterns inform cancer prognosis and treatment strategies?

UQCRQ expression analysis provides valuable clinical insights:

  • Prognostic value: Low UQCRQ expression correlates with shorter survival in ccRCC patients, suggesting its potential as a prognostic biomarker .

  • Patient stratification: UQCRQ expression patterns could identify metabolically distinct tumor subgroups that may respond differently to therapies targeting mitochondrial function or glycolysis.

  • Therapeutic implications:

    • Tumors with hypermethylation-induced UQCRQ silencing may be sensitive to epigenetic modifiers (DNA methyltransferase inhibitors)

    • Metabolic vulnerabilities created by UQCRQ deficiency could be exploited through glycolysis inhibitors

    • Restoration of UQCRQ function through gene therapy approaches might reverse the Warburg phenotype and slow tumor growth

  • Resistance mechanisms: Acquired resistance to therapies might involve compensatory metabolic adaptations that bypass UQCRQ-dependent pathways, necessitating combination approaches targeting multiple metabolic nodes.

How do mutations in UQCRQ compare with defects in other respiratory chain components?

Comparative analysis reveals important distinctions:

  • Phenotypic spectrum: UQCRQ mutations typically present with neurological disorders, hypoglycemia, lactic acidosis, and hypotonia . This contrasts with:

    • UQCRH defects, which cause episodic metabolic decompensation with lactic acidosis and hyperammonemia but less neurological involvement

    • UQCRC2 variants, characterized by episodes of metabolic crisis without significant neurological impairment

    • BCS1L mutations (the most common cause of Complex III deficiency), which present with diverse phenotypes ranging from isolated mitochondrial disease to GRACILE syndrome

  • Developmental impacts: Homozygous knockout of some Complex III components (UQCRC1, UQCRB) results in embryonic lethality in mouse models, whereas homozygous Uqcrh knockout mice are viable despite reduced birth rates (13%) .

  • Biochemical signatures: UQCRQ deficiency creates a characteristic pattern of Complex III assembly defects and supercomplex formation that differs from other component deficiencies, potentially affecting diagnosis and therapeutic approaches .

What systems biology approaches can reveal broader impacts of UQCRQ dysfunction?

Integrative methodologies provide comprehensive understanding:

  • Multi-omics integration:

    • Combined analysis of transcriptomics, proteomics, and metabolomics data from UQCRQ-deficient models

    • Network analysis to identify perturbed pathways beyond direct respiratory chain effects

    • Temporal profiling to distinguish primary from secondary consequences

  • Computational modeling:

    • Constraint-based metabolic modeling (flux balance analysis) to predict metabolic rewiring

    • Dynamic models of electron transport chain function incorporating UQCRQ parameters

    • Machine learning approaches to identify biomarker signatures from multi-dimensional data

  • Tissue crosstalk analysis:

    • Investigation of how UQCRQ deficiency in one tissue affects distant organs through metabolite or exosome-mediated signaling

    • Integrated physiology approaches in animal models to capture systemic effects

  • Evolutionary analysis:

    • Comparative genomics across species to identify conserved functional domains and species-specific adaptations

    • Natural selection patterns in UQCRQ to understand evolutionary constraints on mitochondrial function

What future research directions might expand our understanding of UQCRQ biology?

Promising research frontiers include:

  • Structural biology advances:

    • Cryo-electron microscopy to determine high-resolution structures of UQCRQ within intact Complex III and supercomplexes

    • Hydrogen-deuterium exchange mass spectrometry to map dynamic protein interactions

  • Regulatory mechanisms:

    • Investigation of post-translational modifications affecting UQCRQ function

    • Identification of transcriptional and epigenetic regulators controlling UQCRQ expression

    • MicroRNA and long non-coding RNA interactions with UQCRQ expression

  • Therapeutic development:

    • Small molecule screens for compounds that can bypass or compensate for UQCRQ deficiency

    • Gene therapy approaches using adeno-associated viral vectors for tissue-specific UQCRQ restoration

    • Metabolic interventions tailored to the specific defects caused by UQCRQ dysfunction

  • Novel pathogenic mechanisms:

    • Investigation of UQCRQ's potential roles beyond electron transport

    • Examination of potential immunological consequences of mitochondrial dysfunction

    • Exploration of interactions between UQCRQ deficiency and environmental stressors

Product Science Overview

Introduction

Ubiquinol-Cytochrome C Reductase, also known as Complex III or Cytochrome b-c1 complex, is a crucial component of the mitochondrial electron transport chain. This enzyme complex plays a pivotal role in cellular respiration, facilitating the transfer of electrons from ubiquinol to cytochrome c, which ultimately contributes to the production of ATP, the primary energy currency of the cell.

Structure and Composition

Ubiquinol-Cytochrome C Reductase is a multisubunit transmembrane complex located in the inner mitochondrial membrane. It consists of several subunits, including core proteins such as UQCRC1 and UQCRC2. The human recombinant form of this enzyme is produced through recombinant DNA technology, allowing for the study and utilization of this complex in various research and therapeutic applications.

Function

The primary function of Ubiquinol-Cytochrome C Reductase is to facilitate the transfer of electrons within the mitochondrial electron transport chain. This process is essential for oxidative phosphorylation, where the energy derived from electron transfer is used to pump protons across the inner mitochondrial membrane, creating an electrochemical gradient. This gradient drives the synthesis of ATP by ATP synthase.

Mechanism of Action

The enzyme complex operates by accepting electrons from ubiquinol (reduced form of coenzyme Q) and transferring them to cytochrome c. This electron transfer is coupled with the translocation of protons from the mitochondrial matrix to the intermembrane space, contributing to the proton motive force required for ATP synthesis. The overall reaction can be summarized as follows:

[ \text{Ubiquinol} + 2 \text{Cytochrome c (oxidized)} + 2 \text{H}^+{\text{matrix}} \rightarrow \text{Ubiquinone} + 2 \text{Cytochrome c (reduced)} + 4 \text{H}^+{\text{intermembrane space}} ]

Clinical Significance

Defects in Ubiquinol-Cytochrome C Reductase can lead to mitochondrial disorders, which are often characterized by impaired energy production. One such disorder is mitochondrial complex III deficiency, which can result in a range of symptoms including muscle weakness, neurological deficits, and metabolic abnormalities. Understanding the structure and function of this enzyme complex is crucial for developing therapeutic strategies to address these conditions.

Research and Applications

The human recombinant form of Ubiquinol-Cytochrome C Reductase is widely used in research to study mitochondrial function and pathology. It serves as a valuable tool for investigating the mechanisms of electron transport and oxidative phosphorylation. Additionally, it has potential applications in drug development, particularly in the design of therapies targeting mitochondrial dysfunction.

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.