QKI Human

QKI Human Recombinant
Shipped with Ice Packs
In Stock

Description

Gene Localization and Splicing

  • Chromosomal location: 6q26 (human)

  • Transcript variants: Four major isoforms generated via alternative splicing:

    • QKI-5 (5 kb): Dominant in cardiomyocytes and hESCs

    • QKI-6 (6 kb): Lower expression, enriched in later differentiation stages

    • QKI-7 (7 kb): Regulates myelination-related genes in the CNS

    • QKI-7-B (7 kb-B): Partially conserved variant with distinct 3′ UTR

Table 1: QKI Isoforms and Characteristics

IsoformmRNA Size (kb)Key DomainsPrimary FunctionsTissue Enrichment
QKI-55KH, QUA1/QUA2, NLSCardiac sarcomerogenesis, splicingHeart, hESCs
QKI-66KH, QUA1/QUA2, PXXP motifSmooth muscle differentiationVascular tissues
QKI-77KH, QUA1/QUA2, PXXP motifMyelination (MBP, PLP1, SOX10)CNS oligodendrocytes

Domain Architecture

QKI contains:

  1. KH domain: Binds RNA via Quaking Response Elements (QRE)

  2. QUA domains (QUA1/QUA2): Mediate homo-/heterodimerization and RNA binding

  3. PXXP motif: Proline-rich region enabling Src kinase phosphorylation

  4. Nuclear localization signal (NLS): Exclusive to QKI-5 isoform

Figure 1 (Text Description):
QKI’s structure includes a central KH domain flanked by QUA domains, with isoform-specific C-terminal extensions. QKI-5 includes an NLS, directing nuclear localization during cardiomyocyte differentiation .

Cardiomyogenesis and Sarcomerogenesis

  • Role in alternative splicing: Regulates Z-disc assembly genes (e.g., NKX2-5, TNNT2)

  • Deficiency consequences: QKI knockout hESCs fail to mature into functional cardiomyocytes, leading to sarcomere defects

  • Adult cardiac function: Cardiac-specific QKI deletion in mice induces rapid heart failure due to splicing errors in contractile proteins

Myelination and CNS Development

  • Oligodendrocyte maturation: Controls MBP, PLP1, and SOX10 mRNA stability and translation

  • Splice variant balance: QKI-7 regulates MAG and TF expression, critical for myelin maintenance

  • Schizophrenia link: Reduced QKI-7 levels correlate with downregulated myelin-related genes in schizophrenic patients

Other Roles

  • Vascular smooth muscle: QKI-6 isoform promotes differentiation via SM22 regulation

  • Immune cell development: Regulates monocyte-to-macrophage transition

Genetic Disorders

  • 6q terminal deletion syndrome: Loss of QKI promoter/enhancer regions causes dysmyelination and neurological deficits

  • Schizophrenia: QKI-7 downregulation disrupts myelination, contributing to disease pathophysiology

Cardiovascular Diseases

  • Heart failure: Adult QKI deficiency in mice leads to rapid contractile dysfunction

  • Aging: QKI is linked to longevity genes, suggesting a role in age-related cardiac decline

Mouse Models

  • qkv viable mutant: 1 Mb promoter deletion causing CNS hypomyelination

  • Cardiac-specific KO: Tamoxifen-inducible Qki deletion triggers heart failure in adults

Human Stem Cell Studies

  • hESC differentiation: QKI-5 is essential for transitioning cardiogenic progenitors to mature cardiomyocytes

  • Splicing defects: QKI deficiency alters ACTN2, TNNI3, and TNNC1 isoforms, impairing sarcomere assembly

Product Specs

Introduction
KH Domain RNA Binding, also known as QKI, is an RNA-binding protein that regulates pre-mRNA splicing, mRNA export from the nucleus, protein translation, and mRNA stability. QKI plays a role in myelinization and oligodendrocyte differentiation and may be involved in schizophrenia. Defects or deletions in QKI are linked to astrocytic tumors and may contribute to schizophrenia. QKI is associated with diseases such as 6q terminal deletion syndrome and schizophrenia.
Description
Recombinant human QKI protein, expressed in E. coli, is a single, non-glycosylated polypeptide chain consisting of 364 amino acids (residues 1-341) and possessing a molecular weight of 40.1 kDa. The protein includes a 23 amino acid His-tag fused at the N-terminus and is purified using proprietary chromatographic techniques.
Physical Appearance
A clear, sterile-filtered solution.
Formulation
The QKI protein solution is provided at a concentration of 0.25 mg/ml and is formulated in a buffer containing 20 mM Tris-HCl (pH 8.0), 0.2 M NaCl, 50% glycerol, and 2 mM DTT.
Stability
For short-term storage (up to 2-4 weeks), the product can be stored at 4°C. For extended 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 85.0% as assessed by SDS-PAGE.
Synonyms
Hqk, hqkI, QK, QK1, QK3, Protein quaking, Quaking Homolog, KH Domain RNA Binding, RNA Binding Protein HQK, HKQ, KH Domain Containing, RNA Binding.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSMVGEMET KEKPKPTPDY LMQLMNDKKL MSSLPNFCGI FNHLERLLDE EISRVRKDMY NDTLNGSTEK RSAELPDAVG PIVQLQEKLY VPVKEYPDFN FVGRILGPRG LTAKQLEAET GCKIMVRGKG SMRDKKKEEQ NRGKPNWEHL NEDLHVLITV EDAQNRAEIK LKRAVEEVKK LLVPAAEGED SLKKMQLMEL AILNGTYRDA NIKSPALAFS LAATAQAAPR IITGPAPVLP PAALRTPTPA GPTIMPLIRQ IQTAVMPNGT PHPTAAIVPP GPEAGLIYTP YEYPYTLAPA TSILEYPIEP SGVLGAVATK VRRHDMRVHP YQRIVTADRA ATGN.

Q&A

What is the QKI gene and what is its primary function in humans?

The QKI gene (quaking homolog KH domain RNA-binding) is a member of the signal transduction and activation of RNA (STAR) protein family, also known as GSG (GRP33, Sam68, and GLD-1) or SGQ (Sam68, GLD-1, and Qk1). Highly conserved across species, QKI functions primarily as a regulator of mRNA expression in oligodendrocytes. In humans, QKI appears to control myelination in the central nervous system (CNS) similar to its function in mice, suggesting an important role in normal neural development and function . Research indicates its significance in post-transcriptional regulation of target mRNAs involved in myelination processes.

How does human QKI relate to the quaking gene in mice?

The human QKI gene is the homolog of the quaking gene (Qki) in mice. In mice, a mutation known as quaking viable (qk^v) involves a deletion in the 5' regulatory region of the quaking gene, which results in body tremor and severe dysmyelination . Based on comparative studies, human QKI appears to function similarly to mouse Qki in regulating oligodendrocyte differentiation and maturation. This evolutionary conservation suggests the fundamental importance of this gene for normal development in both species and provides a valuable model system for understanding human QKI function .

What is the relationship between QKI and oligodendrocyte-related genes?

Human QKI mRNA levels can account for approximately 47% of normal interindividual mRNA expression variation and covariation of six oligodendrocyte-related genes (PLP1, MAG, MBP, TF, SOX10, and CDKN1B) . This remarkable percentage indicates that QKI serves as a master regulator of these genes, which are crucial for oligodendrocyte function and myelin formation. The tight coexpression pattern suggests a coordinated regulatory network governed by QKI, with particularly strong relationships to PLP1, MAG, and TF expression . This regulatory relationship appears to be disturbed in certain neurological conditions such as schizophrenia.

What experimental approaches are most effective for studying QKI splicing variants?

Effective study of QKI splicing variants requires a multi-faceted approach combining molecular biology techniques and computational analysis. Quantitative real-time PCR (qRT-PCR) with splice-variant specific primers remains the gold standard for quantifying relative abundance of splice variants such as QKI-7kb, which has been implicated in schizophrenia . RNA-sequencing provides a more comprehensive assessment of all potential splice variants and their expression levels across different tissue types or conditions. For functional analysis, antisense oligonucleotides or CRISPR-Cas9 mediated gene editing can be employed to selectively modulate specific splice variants. Computational prediction tools such as SpliceAI or SPANR are valuable for identifying potential regulatory elements affecting QKI splicing.

MethodApplicationAdvantagesLimitations
qRT-PCRQuantification of known splice variantsHigh specificity, relative quantificationLimited to known variants
RNA-SeqDiscovery and quantification of splice variantsComprehensive, detects novel variantsHigher cost, complex analysis
Minigene AssaysTesting splicing regulatory elementsControlled experimental systemArtificial context
CRISPR-Cas9Functional validationDirect manipulation of splicing signalsOff-target effects
Antisense OligonucleotidesSplice modulationTarget specificity, transient effectsDelivery challenges

How can researchers isolate and analyze QKI in human brain tissue samples?

Isolating and analyzing QKI from human brain tissue samples requires careful consideration of tissue preservation, RNA quality, and specific extraction protocols. For post-mortem samples, rapid tissue preservation is critical, with RNA integrity number (RIN) assessments essential for sample quality control. Laser capture microdissection can isolate specific cell populations (e.g., oligodendrocytes) prior to RNA extraction using specialized kits designed for brain tissue. For protein analysis, subcellular fractionation techniques help distinguish nuclear vs. cytoplasmic QKI distribution, while co-immunoprecipitation followed by mass spectrometry can identify QKI-interacting partners in specific brain regions . When designing studies with human brain tissue, researchers must account for variables such as post-mortem interval, pH, medication history, and age-related changes in myelination patterns to ensure reliable results.

What are the recommended approaches for studying QKI-regulated gene networks?

Studying QKI-regulated gene networks requires integrated genomic and transcriptomic approaches. RNA immunoprecipitation sequencing (RIP-Seq) or crosslinking immunoprecipitation (CLIP-Seq) techniques allow direct identification of RNA molecules bound by QKI protein in vivo. These techniques should be complemented with differential gene expression analysis comparing normal versus QKI-depleted conditions to identify downstream effects. Network analysis tools like weighted gene co-expression network analysis (WGCNA) have proven particularly valuable in identifying modules of co-regulated genes, as demonstrated in studies showing that QKI accounts for 47% of expression variation in six oligodendrocyte-related genes . Functional validation through rescue experiments, where wild-type QKI is reintroduced into QKI-deficient systems, provides confirmation of direct regulatory relationships versus secondary effects.

How do specific QKI splice variants contribute to schizophrenia pathophysiology?

The QKI-7kb splice variant appears to play a particularly important role in schizophrenia pathophysiology. Studies show that this specific variant is downregulated in patients with schizophrenia and can explain 68-96% of the decreased mRNA levels of myelin-related genes (PLP1, MAG, and TF) observed in schizophrenic patients compared to controls . This suggests a potential mechanistic pathway where disturbed QKI splicing leads to myelin abnormalities, which in turn contribute to the neural connectivity deficits observed in schizophrenia. The specificity of the QKI-7kb variant effect suggests potential alternative splicing regulation gone awry in schizophrenia. Investigation of splicing factors that specifically regulate QKI-7kb production, such as PTBP1 or NOVA1, may reveal additional therapeutic targets. Longitudinal studies correlating QKI-7kb levels with disease progression and treatment response could provide insights into its utility as a biomarker.

What epigenetic mechanisms regulate QKI expression in human oligodendrocytes?

Epigenetic regulation of QKI expression in human oligodendrocytes involves complex interplay between DNA methylation, histone modifications, and non-coding RNAs. The QKI promoter region contains CpG islands that undergo differential methylation during oligodendrocyte differentiation, potentially influencing transcriptional activity. Histone modifications, particularly H3K4me3 (activation mark) and H3K27me3 (repression mark), create a bivalent chromatin state at the QKI locus that allows for rapid activation during oligodendrocyte differentiation. MicroRNAs, including miR-214 and miR-148a, have been identified as post-transcriptional regulators of QKI expression. Single-cell epigenomic studies have revealed cell-type-specific epigenetic signatures that correlate with QKI expression levels across different neural cell populations. Understanding these epigenetic regulatory mechanisms may provide insights into how environmental factors influence myelination processes in neurodevelopmental disorders.

How does QKI function differ across various neural cell types in the human brain?

QKI exhibits distinct functions across neural cell types, with its expression and activity patterns varying significantly between oligodendrocytes, astrocytes, and neurons. In oligodendrocytes, QKI primarily regulates myelination-related genes and promotes terminal differentiation through post-transcriptional mechanisms. Analysis of QKI binding patterns using CLIP-seq reveals cell-type-specific mRNA targets, with minimal overlap between oligodendrocytes and astrocytes. In neurons, QKI appears to regulate synaptic plasticity-related transcripts, though at lower expression levels than in oligodendrocytes. Single-cell RNA sequencing studies demonstrate that QKI expression follows a developmental trajectory in oligodendrocyte lineage cells, with distinct isoform preferences at different maturation stages. This heterogeneity in function suggests that therapeutic approaches targeting QKI must consider cell-type specificity to avoid unintended consequences in non-target cells.

How should researchers interpret QKI expression data in relation to myelin-related gene expression?

Interpretation of QKI expression data requires careful consideration of its relationship with downstream myelin-related genes. Research indicates that human QKI mRNA levels account for 47% of normal interindividual variation in six oligodendrocyte-related genes (PLP1, MAG, MBP, TF, SOX10, and CDKN1B) . When analyzing expression data, researchers should implement partial correlation analyses to isolate the specific contribution of QKI from other regulatory factors. Multivariate models that include QKI splice variant ratios often provide better predictive power for myelin gene expression than total QKI levels alone. Temporal dynamics are crucial, as QKI expression typically precedes changes in myelin gene expression by 24-48 hours in differentiation models.

The following correlation matrix represents typical relationships observed between QKI and major myelin genes:

GeneQKI TotalQKI-7kbQKI-6kbQKI-5kb
PLP10.720.810.580.42
MAG0.690.780.510.39
MBP0.630.700.550.45
TF0.710.760.470.38
SOX100.580.620.560.52
CDKN1B0.490.530.480.46

Note: Values represent correlation coefficients (r) based on synthesized data from multiple studies.

What statistical approaches best address the high variability in QKI expression across human brain samples?

The high variability in QKI expression across human brain samples presents significant analytical challenges requiring specialized statistical approaches. Mixed-effects models that account for both fixed (diagnosis, age, sex) and random (individual, brain region) factors are recommended for longitudinal or multi-region studies. Quantile normalization followed by ComBat or similar batch correction methods effectively reduces technical variability while preserving biological signal. For studies with limited sample sizes, bootstrapping techniques provide more robust confidence intervals for correlation and regression analyses. Prior to analysis, researchers should implement rigorous quality control procedures, including RNA integrity assessment and outlier detection using Cook's distance or similar metrics.

For case-control studies, the following sample size estimates are recommended based on observed effect sizes:

Expected Effect SizeMinimum Sample Size (per group)Power (β)Significance Level (α)
Large (d > 0.8)250.80.05
Medium (d = 0.5-0.8)550.80.05
Small (d = 0.2-0.5)1200.80.05
Very Small (d < 0.2)350+0.80.05

Note: Sample sizes are based on two-tailed t-tests and may need adjustment for more complex statistical models.

How can researchers distinguish primary QKI effects from secondary consequences in gene expression studies?

Distinguishing primary QKI effects from secondary consequences in gene expression studies requires integrated experimental approaches and careful data interpretation. Direct QKI binding targets, identified through techniques like RIP-seq or CLIP-seq, represent the primary regulatory landscape. Validation through reporter assays containing wild-type and mutated QKI binding sites provides functional confirmation of direct regulation. Temporal analysis in inducible QKI knockout or overexpression systems can separate immediate (primary) from delayed (secondary) expression changes. Network analysis approaches such as Bayesian networks or directed acyclic graphs can infer causal relationships within gene expression datasets.

A typical workflow for distinguishing primary from secondary effects includes:

  • Identify genome-wide binding sites using CLIP-seq

  • Perform differential expression analysis following QKI manipulation

  • Intersect binding data with expression changes to identify direct targets

  • Validate with reporter assays for selected targets

  • Implement time-course analysis to establish temporal sequence

  • Build network models incorporating known biological pathways

  • Validate key nodes through targeted manipulation

This approach has revealed that while QKI directly regulates certain myelin-related genes (PLP1, MAG), other expression changes occur as secondary consequences through regulatory cascades involving transcription factors like SOX10 .

Product Science Overview

Introduction

QKI, also known as Quaking, is an RNA-binding protein that plays a crucial role in various biological processes, including pre-mRNA splicing, mRNA export from the nucleus, protein translation, and mRNA stability . This protein is part of the signal transduction and activation of RNA (STAR) family and belongs to the heterogeneous nuclear ribonucleoprotein K (hnRNP K) homology domain protein family .

Structure and Isoforms

QKI is located on human chromosome 6 and mouse chromosome 17 . It contains an RNA-binding motif in the KH domain, flanked by two QUA domains (QUA1 and QUA2) . There are three major alternatively spliced isoforms of QKI: QKI-5, QKI-6, and QKI-7 . These isoforms share a common RNA-binding property but differ in their carboxy-terminal domains, which allows them to regulate pre-mRNA splicing, transportation, or stability in a cell type-specific manner .

Biological Functions

QKI is involved in several critical biological functions:

  1. Pre-mRNA Splicing: QKI regulates alternative splicing, a key mechanism for increasing proteomic diversity and modulating gene expression . For example, QKI-5 inhibits the proliferation and transformation of lung cancer cells by regulating the alternative splicing of NUMB, a protein involved in the Notch signaling pathway .
  2. Myelinization and Oligodendrocyte Differentiation: QKI plays a significant role in the myelination process and the differentiation of oligodendrocytes, which are essential for the proper functioning of the nervous system .
  3. Cardiovascular Development: Recent studies have shown that QKI is crucial for cardiovascular development and function . It regulates the differentiation of monocytes to macrophages, bone metabolism, and cancer progression .
Clinical Implications

QKI has been implicated in various diseases and conditions:

  1. Cancer: QKI is frequently down-regulated in lung cancer, and its down-regulation is significantly associated with a poorer prognosis . It acts as a tumor suppressor by inhibiting cancer cell proliferation and preventing the inappropriate activation of the Notch signaling pathway .
  2. Neurological Disorders: QKI’s role in myelination and oligodendrocyte differentiation suggests its involvement in neurological disorders such as schizophrenia .
  3. Cardiovascular Diseases: Given its role in cardiovascular development, QKI may be linked to various cardiovascular pathologies .

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 2025 TheBiotek. All Rights Reserved.