Recombinant Mouse CCR4-NOT transcription complex subunit 4 (Cnot4)

Shipped with Ice Packs
In Stock

Description

Introduction to Recombinant Mouse CCR4-NOT Transcription Complex Subunit 4 (Cnot4)

Recombinant Mouse CCR4-NOT transcription complex subunit 4 (Cnot4) is a crucial component of the CCR4-NOT complex, a conserved multiprotein complex involved in regulating eukaryotic gene expression. The CCR4-NOT complex primarily functions through the shortening of poly(A) tails of messenger RNA (mRNA), a process known as deadenylation, which is essential for mRNA degradation and regulation of gene expression . Cnot4 serves as a regulatory subunit within this complex, playing roles in mRNA degradation, DNA damage repair, and meiosis progression during spermatogenesis .

mRNA Degradation and Regulation

Cnot4 acts as an mRNA adaptor for the CCR4-NOT complex, facilitating the targeting of specific mRNAs for deadenylation by subunits like CNOT7 . This process is critical for regulating mRNA stability and turnover, influencing gene expression across various cellular processes.

Role in Obesity Resistance

Recent studies have shown that heterozygous deletion of Cnot4 in mice leads to resistance against high-fat diet-induced obesity. This resistance is associated with reduced adipose tissue mass and hepatic fat depots, although it does not significantly affect lipid or glucose metabolism .

Involvement in Viral Replication

Cnot4 has been identified as a cellular ubiquitin ligase capable of ubiquitinating the nucleoprotein of Influenza A virus, thereby enhancing viral RNA replication .

Obesity Resistance

ParameterWild-Type MiceCnot4 Heterozygous Mice
Body WeightHigherLower (by approximately 13%)
Adipose TissueIncreasedReduced
Hepatic FatIncreasedReduced

Cnot4 heterozygous mice exhibit reduced body weight and adipose tissue mass compared to wild-type mice when fed a high-fat diet, indicating a role in obesity resistance .

mRNA Degradation

FunctionMechanismImpact
mRNA AdaptorTargets mRNAs for deadenylation by CNOT7Regulates mRNA stability and turnover
DeadenylationShortens poly(A) tails of mRNAsEnhances mRNA degradation

Cnot4's role in mRNA degradation is crucial for maintaining gene expression balance and preventing aberrant mRNA accumulation .

Viral Replication

Viral ComponentEffect of Cnot4Outcome
Influenza A Virus NucleoproteinUbiquitination by Cnot4Enhanced viral RNA replication

Cnot4's ubiquitination activity supports the replication of Influenza A virus by modifying its nucleoprotein .

References Yamaguchi T, et al. Cnot4 heterozygosity attenuates high fat diet-induced obesity in mice (2024). Reconstitution of recombinant human CCR4-NOT reveals molecular insights into regulated deadenylation (2019). CNOT4-Mediated Ubiquitination of Influenza A Virus Nucleoprotein (2017). Human CCR4-NOT is a global regulator of gene expression and is a novel silencer of retrotransposon activation (2024). Dai Xing-Xing, et al. The CNOT4 Subunit of the CCR4‐NOT Complex is Involved in mRNA Degradation, Efficient DNA Damage Repair, and XY Chromosome Crossover during Male Germ Cell Meiosis (2021). Roles of the CCR4‐Not complex in translation and dynamics of co‐translation events (2023). CNOT4 CCR4-NOT transcription complex subunit 4 (NCBI). The Regulatory Properties of the Ccr4–Not Complex (2020).

Product Specs

Form
Lyophilized powder
Note: While we prioritize shipping the format currently in stock, please specify your format preference in order notes for customized preparation.
Lead Time
Delivery times vary depending on purchasing method and location. Consult your local distributor for precise delivery estimates.
Note: Standard shipping includes blue ice packs. Dry ice shipping requires prior arrangement and incurs additional charges.
Notes
Avoid repeated freeze-thaw cycles. Store working aliquots at 4°C for up to one week.
Reconstitution
Centrifuge the vial briefly before opening to collect the contents. Reconstitute the protein in sterile, deionized water to a concentration of 0.1-1.0 mg/mL. For long-term storage, we recommend adding 5-50% glycerol (final concentration) and aliquoting at -20°C/-80°C. Our standard glycerol concentration is 50%, which can be used as a reference.
Shelf Life
Shelf life depends on storage conditions, buffer composition, temperature, and protein stability. Generally, liquid formulations have a 6-month shelf life at -20°C/-80°C, while lyophilized forms have a 12-month shelf life at -20°C/-80°C.
Storage Condition
Upon receipt, store at -20°C/-80°C. Aliquot to prevent repeated freeze-thaw cycles.
Tag Info
The tag type is determined during manufacturing.
If you require a specific tag, please inform us; we will prioritize its development.
Synonyms
Cnot4; Not4CCR4-NOT transcription complex subunit 4; EC 2.3.2.27; CCR4-associated factor 4; E3 ubiquitin-protein ligase CNOT4; Potential transcriptional repressor NOT4Hp; RING-type E3 ubiquitin transferase CNOT4
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
1-575
Protein Length
full length protein
Purity
>85% (SDS-PAGE)
Species
Mus musculus (Mouse)
Target Names
Cnot4
Target Protein Sequence
MSRSPDAKED PVECPLCMEP LEIDDINFFP CTCGYQICRF CWHRIRTDEN GLCPACRKPY PEDPAVYKPL SQEELQRIKN EKKQKQNERK QKISENRKHL ASVRVVQKNL VFVVGLSQRL ADPEVLKRPE YFGKFGKIHK VVINNSTSYA GSQGPSASAY VTYIRSEDAL RAIQCVNNVV VDGRTLKASL GTTKYCSYFL KNMQCPKPDC MYLHELGDEA ASFTKEEMQA GKHQEYEQKL LQELYKLNPN FLQLSTGSVD KNKNKVTPLQ RYDTPIDKPS DSLSIGNGDN SQQISNSDTP SPPPGLSKSN PVIPISSSNH SARSPFEGAV TESQSLFSDN FRHPNPIPSG LPPFPSSPQT PSDWPTAPEP QSLFTSETIP VSSSTDWQAA FGFGSSKQPE DDLGFDPFDV TRKALADLIE KELSVQDQPS LSPTSLQNAS SHTTTAKGPG SGFLHSAAPT NANSLNSTFS VLPQRFPQFQ QHRAVYNSFG FPGQAARYPW MAFPRNSIMH LNHTANPTSN SNFLDLNLPP QHNTGLGGIP IAGEEEVKVS TMPLSASSHS LQQGQQPTSL HTTVA
Uniprot No.

Target Background

Function
Recombinant Mouse CCR4-NOT transcription complex subunit 4 (Cnot4) possesses E3 ubiquitin ligase activity, mediating the ubiquitination and subsequent degradation of target proteins. It is involved in JAK/STAT pathway activation and catalyzes the ubiquitination of methylated RBM15. Furthermore, it plays a role in the quality control of mitochondrial outer membrane-localized mRNA translation. As part of PINK1-regulated signaling, upon mitochondrial damage, it ubiquitinates ABCE1, recruiting autophagy receptors to the mitochondrial outer membrane and initiating mitophagy.
Database Links
Subcellular Location
Cytoplasm. Nucleus.

Q&A

What is the CCR4-NOT complex and what role does Cnot4 play within it?

The CCR4-NOT complex is a major mRNA deadenylase in eukaryotes that regulates gene expression by shortening poly(A) tails of messenger RNA. The complex comprises multiple subunits including the catalytic components CNOT6/6L and CNOT7/8, as well as regulatory subunits such as CNOT4 . While CNOT4 is not a constitutive component of human CCR4-NOT , it functions as a previously unrecognized mRNA adaptor of the complex by targeting specific mRNAs to CNOT7 for deadenylation of poly(A) tails, thereby mediating the degradation of a subset of transcripts . Cnot4's role is particularly significant during developmental processes and spermatogenesis, where it promotes appropriate gene expression by removing specific mRNAs during critical transitions.

How does mouse Cnot4 differ structurally and functionally from human CNOT4?

Mouse Cnot4 and human CNOT4 share significant structural homology, with both containing conserved functional domains that interact with components of the CCR4-NOT complex. The key difference relates to their association with the complex: while mouse Cnot4 interactions are not fully characterized in the provided literature, human CNOT4 is notably not a constitutive component of the CCR4-NOT complex . Despite this difference, both appear to function in mRNA regulation and ribosome interaction. In humans, CCR4-NOT is required for stable association of CNOT4, which then ubiquitinates ribosomes, likely signaling stalled translation elongation . This mechanism appears to be evolutionarily conserved, as CNOT4 serves similar functions in both species regarding targeted mRNA degradation during developmental processes.

What are the primary biological processes regulated by mouse Cnot4?

Mouse Cnot4 regulates several critical biological processes:

  • Embryonic development: Cnot4 is essential for post-implantation embryo development, with Cnot4-knockout embryos dying after implantation and before 11.5 days post coitus (dpc) .

  • Spermatogenesis: Cnot4 is critical for meiosis progression during spermatogenesis. Conditional knockout of Cnot4 in male germ cells leads to spermatogenic arrest in metaphase I (Stages IX–X) accompanied by apoptosis .

  • DNA damage repair: Cnot4 is required for normal DNA double-strand break repair during meiosis and efficient crossover between X and Y chromosomes .

  • mRNA stability regulation: Cnot4 mediates the degradation of a subset of transcripts, particularly during the zygotene to pachytene stage of spermatogenesis, which is crucial for the appropriate expression of genes involved in subsequent events .

What are the optimal expression systems for producing functional recombinant mouse Cnot4?

Based on experimental approaches with human CNOT4, which shares significant homology with mouse Cnot4, the optimal expression system appears to be E. coli bacterial expression for recombinant production . For mouse Cnot4 specifically, expression should be optimized with the following considerations:

  • Expression vector selection: Use vectors with strong inducible promoters (e.g., T7) and appropriate fusion tags (His6 or GST) to facilitate purification.

  • E. coli strain optimization: BL21(DE3) derivatives such as Rosetta or Arctic Express strains may improve folding of the complex protein.

  • Induction conditions: Low temperature induction (16-18°C) after reaching optimal culture density (OD600 ~0.6-0.8) with reduced IPTG concentration (0.1-0.5 mM) maximizes soluble protein yield.

  • Purification strategy: Sequential chromatography including affinity (Ni-NTA for His-tagged constructs), ion exchange, and size exclusion chromatography ensures high purity.

While E. coli is suitable for producing isolated Cnot4, reconstitution of the entire CCR4-NOT complex with Cnot4 requires a stepwise assembly of purified recombinant components, which has been successfully demonstrated for the human complex . For studies requiring mammalian post-translational modifications, insect cell (Sf9/Sf21) or mammalian expression systems may be preferable alternatives.

What are the key considerations when designing functional assays for recombinant mouse Cnot4?

When designing functional assays for recombinant mouse Cnot4, researchers should consider the following key aspects:

  • Ubiquitin ligase activity assessment: As Cnot4 possesses E3 ubiquitin ligase activity, in vitro ubiquitination assays should include:

    • Purified E1 and E2 enzymes along with recombinant Cnot4

    • Ubiquitin (preferably labeled for detection)

    • Potential substrates (ribosomes or specific proteins)

    • ATP regeneration system

    • Detection methods (western blotting, fluorescence-based assays)

  • mRNA targeting assays: To assess Cnot4's role in mRNA targeting:

    • Reconstitute minimal CCR4-NOT complex including Cnot7

    • Provide labeled target mRNAs with poly(A) tails

    • Monitor deadenylation activity with or without Cnot4

    • Compare deadenylation rates between different mRNA substrates

  • Ribosome binding assays: For evaluating interaction with translating ribosomes:

    • Utilize in vitro translation systems (rabbit reticulocyte lysate or reconstituted system)

    • Generate stalled ribosomes on specific mRNA templates

    • Include purified CCR4-NOT complex components

    • Analyze binding by sedimentation, pulldown assays, or cryoEM

  • Protein-protein interaction analysis: To map interaction networks:

    • Use reconstituted components for pulldown assays

    • Apply crosslinking mass spectrometry to identify precise interaction sites

    • Consider yeast two-hybrid or mammalian two-hybrid systems for validation

Control experiments should include catalytically inactive Cnot4 mutants and assays performed in the absence of key components to demonstrate specificity of activity.

How can researchers ensure that recombinant mouse Cnot4 maintains its native structure and activity?

Ensuring native structure and activity of recombinant mouse Cnot4 requires multiple validation approaches:

  • Structural validation:

    • Circular dichroism (CD) spectroscopy to confirm secondary structure elements

    • Size exclusion chromatography with multi-angle light scattering (SEC-MALS) to verify monodispersity and oligomeric state

    • Limited proteolysis to assess proper folding and domain accessibility

    • Thermal shift assays to evaluate protein stability

  • Functional validation:

    • E3 ubiquitin ligase activity assays using known substrates

    • Interaction assays with other CCR4-NOT components, particularly CNOT7 and CNOT3

    • RNA binding assays if studying direct RNA interactions

    • Complementation studies in Cnot4-deficient cells to restore function

  • Storage and handling considerations:

    • Determine optimal buffer conditions (pH, salt concentration, reducing agents)

    • Evaluate protein stability with freeze-thaw cycles

    • Consider addition of stabilizing agents (glycerol, specific cofactors)

    • Test activity retention over time under various storage conditions

  • Comparative analysis:

    • Compare activity of recombinant protein with native Cnot4 immunoprecipitated from mouse cells

    • Benchmark against published biochemical parameters when available

A combination of these approaches will help ensure that recombinant mouse Cnot4 closely resembles its native counterpart in structure and function.

How does Cnot4 regulate mRNA stability and what are its specific target transcripts?

Cnot4 plays a sophisticated role in regulating mRNA stability through multiple mechanisms:

  • Adaptor function: Cnot4 functions as an mRNA adaptor of the CCR4-NOT complex by targeting specific mRNAs to CNOT7 for deadenylation of poly(A) tails, thereby mediating the degradation of a subset of transcripts . Unlike general deadenylation factors, Cnot4 appears to confer specificity to the process.

  • Stage-specific regulation: During spermatogenesis, Cnot4-mediated mRNA removal is particularly active during the zygotene-to-pachytene transition, suggesting temporal control of target transcripts .

  • Target specificity: While the complete repertoire of Cnot4 target transcripts hasn't been fully characterized in the literature provided, studies suggest they include mRNAs involved in:

    • DNA damage repair pathways

    • Homologous recombination mechanisms

    • Sex chromosome pairing and crossover

    • Subsequent events of spermatogenesis

  • Regulatory mechanism: Cnot4 likely recognizes specific features in target mRNAs, potentially through:

    • Direct RNA binding

    • Interaction with RNA-binding proteins that recognize specific sequence motifs

    • Recognition of ribosomes translating specific mRNAs

Interestingly, studies with human CCR4-NOT suggest that the complex can recognize ribosomes stalled during translation elongation, with CNOT3 inserting a helical bundle into the empty E site of the ribosome and locking the L1 stalk in an open conformation to inhibit further translation . This mechanism could potentially link translation efficiency with mRNA stability, with Cnot4 playing a role in the recognition and ubiquitination of stalled ribosomes.

What is the molecular mechanism of Cnot4's interaction with the CCR4-NOT complex?

The molecular mechanism of Cnot4's interaction with the CCR4-NOT complex involves several key aspects:

  • Interaction partners: While recombinant human CNOT4 has been shown to associate directly with CNOT9 , the mouse Cnot4 interaction network within CCR4-NOT remains to be fully characterized. The human CCR4-NOT complex is required for stable association of CNOT4 , suggesting a cooperative binding mechanism.

  • Structural basis: Based on studies with human proteins, CNOT4 and CNOT11 bind in the vicinity of the E site of ribosomes recognized by CCR4-NOT . The CNOT3 subunit of CCR4-NOT recognizes translating ribosomes by inserting a helical bundle into the empty E site and locks the L1 stalk in an open conformation .

  • Functional integration: Cnot4 appears to function as a conditional component that joins the core CCR4-NOT complex under specific cellular conditions or in response to particular RNA substrates. Its E3 ubiquitin ligase activity adds an additional regulatory layer to CCR4-NOT function.

  • Ribosome interaction: The interaction involves recognition of ribosomes that are stalled during translation elongation, potentially due to non-optimal codons or other obstacles to efficient translation . This creates a mechanism to link translation efficiency with mRNA stability.

  • Ubiquitination targets: CNOT4 ubiquitinates the ribosome, likely to signal stalled translation elongation . The specific ubiquitination targets on the ribosome and the consequences of this modification require further investigation but may include specific ribosomal proteins involved in translation elongation.

This complex interaction mechanism enables CCR4-NOT to couple translation efficiency with mRNA deadenylation and decay, with Cnot4 playing a key regulatory role in this process.

How does Cnot4 contribute to spermatogenesis and what molecular pathways are involved?

Cnot4 plays a critical role in spermatogenesis through several molecular pathways:

  • Meiotic progression regulation:

    • Conditional knockout of Cnot4 in male germ cells leads to spermatogenic arrest in metaphase I (Stages IX–X)

    • Cnot4-null spermatocytes show increased apoptosis, as evidenced by cleaved caspase 3 immunostaining

    • While early meiotic prophase appears normal, with proper progression through leptotene, zygotene, pachytene, and diplotene stages, later stages are compromised

  • DNA damage repair pathways:

    • Cnot4 is required for normal DNA double-strand break repair during meiosis

    • Loss of Cnot4 leads to defective homologous recombination mechanisms

  • Sex chromosome dynamics:

    • Cnot4 is essential for efficient crossover between X and Y chromosomes

    • This function is critical for proper segregation of sex chromosomes during meiosis

  • Transcript regulation:

    • Cnot4 mediates the degradation of a subset of transcripts from the zygotene to pachytene stage

    • This mRNA removal is crucial for the appropriate expression of genes involved in subsequent events of spermatogenesis

  • Temporal coordination:

    • The activity of Cnot4 appears to be particularly important during specific developmental windows of spermatogenesis

    • The zygotene-to-pachytene transition represents a critical period where Cnot4-regulated mRNA decay shapes the transcriptome

The table below summarizes the phenotypic consequences of Cnot4 deletion in male germ cells:

PhenotypeWild-typeCnot4 conditional knockout
FertilityFertileInfertile
Sperm productionNormal sperm in epididymisNo normal sperm in epididymis
Meiotic progressionCompleteArrest at metaphase I
ApoptosisMinimalSignificant increase in apoptotic spermatocytes
DNA damage repairEfficientDefective
X-Y chromosome crossoverNormalDeficient
Testicular morphologyNormal seminiferous tubulesLarge cavities, few spermatocytes

These findings highlight Cnot4's essential role in coordinating mRNA stability, DNA repair, and chromosome dynamics during spermatogenesis.

What are the most effective strategies for generating Cnot4 knockout or conditional knockout mice?

Based on successful approaches documented in the literature, the most effective strategies for generating Cnot4 knockout or conditional knockout mice include:

  • Floxed allele generation:

    • The creation of a Cnot4-floxed mouse model has been successfully implemented by flanking critical exons (such as exon 2) with loxP sites

    • This strategy permits conditional deletion when crossed with tissue-specific Cre recombinase-expressing lines

    • Target exon 2 (266 bp) whose deletion causes a reading frame shift, ensuring complete functional knockout

  • Tissue-specific deletion:

    • For studying Cnot4's role in spermatogenesis, crossing Cnot4-floxed mice with Stra8-Cre transgenic mice has proven effective

    • Stra8-Cre drives expression specifically in pre-meiotic germ cells, allowing targeted deletion in the male germline

    • Other tissue-specific Cre lines can be selected based on research focus (e.g., Nestin-Cre for neural tissues)

  • Verification approaches:

    • PCR-based genotyping to confirm loxP site integration and subsequent deletion

    • Western blotting to verify absence of Cnot4 protein in targeted tissues

    • RT-PCR to confirm transcript changes

    • Histological analysis to assess phenotypic consequences

  • Alternative approaches:

    • CRISPR/Cas9-mediated generation of null alleles for constitutive knockout

    • Inducible knockout systems (e.g., tetracycline-controlled or tamoxifen-inducible Cre) for temporal control

    • Knock-in strategies for introducing point mutations to study specific functional domains

Key considerations:

  • Full constitutive knockout of Cnot4 results in embryonic lethality between implantation and 11.5 dpc , necessitating conditional approaches for studying postnatal functions

  • Heterozygous Cnot4+/- mice appear viable and can be maintained for breeding

  • Verification of knockout efficiency is essential, as partial deletion may result in hypomorphic phenotypes

What phenotypes have been observed in mouse models with Cnot4 mutations or deletions?

Mouse models with Cnot4 mutations or deletions exhibit distinct phenotypes depending on the nature and timing of the genetic manipulation:

  • Constitutive knockout phenotypes:

    • Embryonic lethality: Homozygous Cnot4-/- embryos die between implantation and 11.5 days post coitus (dpc)

    • Normal preimplantation development: Cnot4-/- embryos develop into blastocysts with normal morphology

    • Post-implantation failure: By 11.5 dpc, Cnot4-/- embryos show complete degeneration

  • Male germline-specific knockout (Cnot4fl/fl;Stra8-Cre) phenotypes:

    • Complete male infertility

    • Testicular abnormalities: Large cavities in seminiferous tubules and very few spermatocytes

    • Absence of sperm in epididymis

    • Spermatogenic arrest in metaphase I (Stages IX–X)

    • Increased apoptosis of spermatocytes (confirmed by cleaved caspase 3 immunostaining)

    • Normal distribution of early meiotic prophase stages (leptotene, zygotene, pachytene, and diplotene)

    • Defective DNA damage repair during meiosis

    • Impaired homologous crossover between X and Y chromosomes

  • Molecular phenotypes:

    • Altered transcriptome during spermatogenesis, particularly affecting the zygotene-to-pachytene transition

    • Dysregulation of genes involved in DNA repair and meiotic processes

    • Likely perturbation of translational regulation and ribosome-associated quality control mechanisms

These phenotypes highlight Cnot4's essential roles in embryonic development and spermatogenesis, with particularly critical functions in post-implantation embryonic development and meiotic progression in male germ cells.

How can researchers address the challenges of embryonic lethality when studying Cnot4 function?

Researchers studying Cnot4 face significant challenges due to the embryonic lethality of constitutive knockouts. Several strategies can effectively address this limitation:

  • Conditional knockout approaches:

    • Utilize the Cre-loxP system with tissue-specific or inducible Cre expression

    • Generate Cnot4-floxed mice (as successfully done in published studies )

    • Cross with appropriate Cre lines based on research questions:

      • Tissue-specific Cre lines (e.g., Stra8-Cre for male germline studies )

      • Inducible Cre systems (CreERT2) for temporal control of deletion

      • Developmental stage-specific Cre drivers for targeting specific embryonic tissues

  • Embryonic analysis techniques:

    • Preimplantation embryo isolation and culture:

      • Harvest 4.5 dpc embryos by flushing the uterus

      • Genotype and analyze blastocysts before lethality occurs

    • Ex vivo embryo culture systems to extend the developmental window

    • Single-cell RNA-seq of early embryos to identify molecular pathways affected

  • Hypomorphic alleles and specific mutations:

    • Design mutations that reduce but don't eliminate Cnot4 function

    • Target specific functional domains rather than creating null alleles

    • CRISPR/Cas9-mediated introduction of point mutations in critical residues

  • Embryonic stem cell (ESC) approaches:

    • Derive ESCs from Cnot4+/- intercrosses to obtain Cnot4-/- lines

    • Study cellular phenotypes in vitro

    • Create chimeric embryos with wild-type extraembryonic tissues

    • Differentiate ESCs into specific lineages to study tissue-specific effects

  • Tetraploid complementation:

    • Generate embryos where extraembryonic tissues are wild-type while embryonic tissues are Cnot4-/-

    • This approach can sometimes rescue embryonic lethality caused by extraembryonic defects

  • Transgenic rescue approaches:

    • Express wild-type Cnot4 or specific variants in Cnot4-/- background

    • Utilize conditional rescue systems to address specific developmental windows

    • Domain-specific rescue to map critical functional regions

Each of these approaches offers unique advantages for addressing different aspects of Cnot4 function while circumventing the limitations imposed by embryonic lethality.

How does mouse Cnot4 function compare with CNOT4 in other model organisms?

Mouse Cnot4 shows important functional similarities and differences when compared to CNOT4 in other model organisms:

  • Comparison with human CNOT4:

    • Both are involved in mRNA stability regulation and deadenylation processes

    • Human CNOT4 is not a constitutive component of CCR4-NOT , while mouse Cnot4's association status is less clearly characterized

    • Both appear to function in ribosome recognition and ubiquitination processes

    • Human CCR4-NOT requires CNOT3 for interaction with stalled ribosomes , which likely applies to mouse as well

    • Both are involved in developmental processes, though specific requirements may differ

  • Comparison with yeast Ccr4-Not:

    • Yeast Ccr4-Not binds and ubiquitinates ribosomes stalled on mRNAs with sub-optimal codons

    • This mechanism appears evolutionarily conserved, as human CCR4-NOT similarly recognizes translating ribosomes stalled during elongation

    • Both yeast and mammalian systems couple translation efficiency with mRNA stability

    • The specific molecular interactions may differ while preserving the core functional principle

  • Functional conservation across species:

    • The CCR4-NOT complex's role in deadenylation is highly conserved across eukaryotes

    • The ubiquitin ligase function of CNOT4 appears to be an ancient feature maintained throughout evolution

    • Ribosome recognition mechanisms show remarkable conservation, with the insertion of CNOT3's helical bundle into the ribosome's E site being a shared feature

  • Species-specific functions:

    • Mouse Cnot4 has essential roles in embryonic development and spermatogenesis

    • The requirement for Cnot4 in male fertility appears to be a mammalian-specific feature

    • The regulation of retrotransposon elements may vary across species, with human CCR4-NOT showing a role in suppressing LINE and other retrotransposon elements

The evolutionary conservation of core CNOT4 functions highlights its fundamental importance in eukaryotic gene expression regulation, while species-specific adaptations reflect the diverse roles it has acquired throughout evolution.

What is the relationship between Cnot4 and ribosome-associated quality control mechanisms?

Cnot4 plays a significant role in ribosome-associated quality control (RQC) mechanisms, connecting translation efficiency with mRNA stability:

  • Recognition of stalled ribosomes:

    • Based on studies with human CCR4-NOT, the complex specifically recognizes ribosomes that are stalled during translation elongation

    • CNOT3, a component of CCR4-NOT, inserts a helical bundle into the empty E site of the ribosome and locks the L1 stalk in an open conformation to inhibit further translation

    • This mechanism enables detection of ribosomes translating mRNAs with suboptimal codons or other translation obstacles

  • Ubiquitination function:

    • CNOT4 ubiquitinates the ribosome, likely to signal stalled translation elongation

    • This ubiquitination may serve as a molecular tag for subsequent quality control steps

    • The specific targets of ubiquitination on the ribosome and the ubiquitin chain topology remain areas for further investigation

  • Coupling translation to mRNA decay:

    • The recognition of stalled ribosomes by CCR4-NOT and the subsequent recruitment of CNOT4 establish a direct link between translation efficiency and mRNA stability

    • This mechanism allows cells to selectively degrade mRNAs that are inefficiently translated, potentially due to defects in the mRNA sequence or structure

    • CNOT4 functions as a previously unrecognized mRNA adaptor of CCR4-NOT by targeting mRNAs to CNOT7 for deadenylation

  • Integration with other quality control pathways:

    • The Cnot4-mediated RQC likely interfaces with other quality control mechanisms including:

      • No-Go Decay (NGD) for mRNAs with strong secondary structures or damaged bases

      • Non-Stop Decay (NSD) for mRNAs lacking stop codons

      • Nonsense-Mediated Decay (NMD) for mRNAs with premature termination codons

  • Developmental consequences:

    • In mouse spermatogenesis, Cnot4's role in RQC appears particularly important during the zygotene-to-pachytene transition

    • This function ensures appropriate expression of genes involved in subsequent meiotic events

    • Disruption of this quality control mechanism leads to defective DNA repair and chromosome crossover

The integration of Cnot4 into ribosome-associated quality control represents a sophisticated regulatory mechanism that ensures the integrity of gene expression by eliminating inefficiently translated mRNAs.

What are the emerging therapeutic implications of targeting Cnot4 in disease models?

While the search results don't directly address therapeutic applications of targeting Cnot4, emerging implications can be inferred from its molecular functions:

  • Potential in male infertility treatment:

    • Cnot4's critical role in spermatogenesis suggests it as a potential therapeutic target or biomarker for specific forms of male infertility

    • Modulating Cnot4 activity might help address infertility cases related to meiotic defects

    • Diagnostic applications could include screening for CNOT4 mutations or expression abnormalities in infertile patients

  • Cancer biology applications:

    • Dysregulation of mRNA stability and translation is a hallmark of many cancers

    • As a regulator of gene expression at multiple levels, Cnot4 could represent a novel target for cancer therapeutics

    • Modulating Cnot4 activity might help reestablish normal gene expression patterns in transformed cells

    • The connection between CCR4-NOT and suppression of retrotransposon elements suggests potential roles in genomic stability relevant to cancer

  • Developmental disorders:

    • Given Cnot4's essential role in embryonic development , mutations affecting its function might contribute to developmental disorders

    • Therapeutic approaches might focus on restoring normal Cnot4 activity or compensating for its loss

  • RNA-based therapeutics:

    • Understanding Cnot4's role in mRNA stability could inform the design of RNA-based therapeutics with improved stability profiles

    • Manipulating Cnot4 activity might enhance the efficacy of mRNA vaccines or therapeutic mRNAs by modulating their degradation kinetics

  • Translational regulation disorders:

    • Cnot4's involvement in ribosome-associated quality control makes it relevant to diseases involving defective translation

    • Targeting the Cnot4-ribosome interaction could potentially address conditions characterized by ribosome stalling or inefficient translation

  • Methodological considerations for therapeutic development:

    • Small molecule inhibitors targeting Cnot4's E3 ubiquitin ligase activity

    • Peptide-based approaches to disrupt specific protein-protein interactions

    • RNA aptamers to modulate Cnot4's interaction with target mRNAs

    • Gene therapy approaches for cases of CNOT4 deficiency

These potential therapeutic applications remain speculative and would require extensive validation through additional research, including studies in appropriate disease models and eventual clinical investigations.

What are the common technical challenges when working with recombinant mouse Cnot4 and how can they be addressed?

Researchers working with recombinant mouse Cnot4 commonly encounter several technical challenges that require specific strategies to overcome:

  • Protein solubility and stability issues:

    • Challenge: Cnot4, like many E3 ubiquitin ligases, can be prone to aggregation and insolubility.

    • Solutions:

      • Express as fusion proteins with solubility-enhancing tags (MBP, SUMO, or GST)

      • Optimize buffer conditions (increased salt, mild detergents, or stabilizing agents)

      • Lower expression temperature (16-18°C) and reduce inducer concentration

      • Consider co-expression with interaction partners or chaperones

      • Explore truncated constructs focusing on specific functional domains

  • Functional activity assessment:

    • Challenge: Confirming that recombinant Cnot4 retains physiologically relevant activity.

    • Solutions:

      • Design robust activity assays incorporating proper controls (including catalytically inactive mutants)

      • Use multiple complementary assays to verify function (ubiquitination, RNA binding, protein-protein interaction)

      • Compare activity of recombinant protein with native Cnot4 immunoprecipitated from mouse cells

      • Ensure proper cofactors and interaction partners are present in functional assays

  • Reconstitution with CCR4-NOT complex:

    • Challenge: Integrating Cnot4 with other components of the CCR4-NOT complex for functional studies.

    • Solutions:

      • Adopt stepwise assembly approach as demonstrated for human CCR4-NOT

      • Consider co-expression of multiple subunits in insect cells or mammalian systems

      • Optimize stoichiometry of components (estimated cellular concentrations of CCR4-NOT are approximately 20 nM)

      • Verify complex formation by size exclusion chromatography or native PAGE

  • Species-specific considerations:

    • Challenge: Extrapolating from human CNOT4 studies to mouse Cnot4.

    • Solutions:

      • Carefully align sequences to identify conserved and divergent regions

      • Design constructs based on documented functional domains from both species

      • Test cross-species compatibility of interactions where appropriate

      • Validate findings with native mouse proteins whenever possible

  • Ribosome interaction studies:

    • Challenge: Reconstituting and analyzing the Cnot4-ribosome interaction.

    • Solutions:

      • Utilize established in vitro translation systems with programmed stalling

      • Include CCR4-NOT complex components necessary for stable CNOT4 association

      • Consider structural approaches (cryoEM) for detailed interaction analysis

      • Use crosslinking mass spectrometry to map precise interaction sites

These strategies can significantly improve the success rate when working with this challenging but biologically significant protein, enabling more robust and reproducible research outcomes.

How can researchers design experiments to distinguish Cnot4-specific effects from general CCR4-NOT complex functions?

Designing experiments that distinguish Cnot4-specific effects from general CCR4-NOT complex functions requires careful experimental planning:

  • Genetic manipulation approaches:

    • Comparative knockouts/knockdowns: Compare phenotypes of Cnot4 deletion with other CCR4-NOT subunits (e.g., Cnot1, Cnot7)

      • Example: Cnot7 knockout male mice are infertile, while Cnot6/6l double knockout mice remain fertile , demonstrating distinct functions

    • Rescue experiments: Test whether Cnot4 deficiency can be rescued by other CCR4-NOT components or only by Cnot4 itself

    • Domain-specific mutations: Target specific functional domains of Cnot4 (e.g., E3 ligase domain) rather than complete knockout

  • Biochemical separation strategies:

    • Sequential immunoprecipitation: Deplete CCR4-NOT complexes containing Cnot4 and analyze remaining complexes

    • Size exclusion chromatography: Separate Cnot4-containing and Cnot4-free CCR4-NOT complexes

    • Density gradient centrifugation: Separate different complex populations based on composition

  • Functional assays with isolated components:

    • Reconstitution experiments: Compare activity of CCR4-NOT with and without Cnot4

      • Example: Deadenylation assays using purified components to determine Cnot4's specific contribution

    • Substrate specificity analysis: Identify mRNAs or proteins that are specifically affected by Cnot4 vs. general CCR4-NOT function

    • Ubiquitination assays: Test E3 ligase activity unique to Cnot4

  • Temporal and spatial resolution:

    • Inducible systems: Use temporal control of gene expression/deletion to distinguish immediate vs. secondary effects

    • Cell-type specific analysis: Examine effects in different cell types with varying dependency on Cnot4

    • Developmental stage analysis: Compare effects at different developmental stages when dependency on specific components may vary

  • Molecular interaction mapping:

    • Protein-protein interaction screens: Identify Cnot4-specific interaction partners not shared with other CCR4-NOT components

    • RNA-binding analysis: Determine whether Cnot4 has RNA targets distinct from the core CCR4-NOT complex

    • Crosslinking mass spectrometry: Map precise interaction sites of Cnot4 with other components or substrates

  • Experimental design table:

QuestionControl conditionExperimental conditionExpected outcome if Cnot4-specificExpected outcome if general CCR4-NOT
mRNA stabilityCnot1 knockoutCnot4 knockoutDifferent subset of affected mRNAsSame subset of affected mRNAs
Ribosome bindingCCR4-NOT without Cnot4CCR4-NOT with Cnot4Enhanced binding to specific ribosomesNo change in binding pattern
UbiquitinationE3-dead Cnot4 mutantWild-type Cnot4Loss of specific ubiquitination eventsNo effect on general CCR4-NOT functions
DevelopmentConditional Cnot1 KOConditional Cnot4 KODistinct developmental defectsSimilar developmental defects

These approaches enable researchers to dissect the specific contributions of Cnot4 to cellular processes beyond the general functions of the CCR4-NOT complex.

What are the most promising research directions for understanding Cnot4's role in transcriptional regulation?

Based on current knowledge and emerging insights, several promising research directions can advance our understanding of Cnot4's role in transcriptional regulation:

  • Genome-wide analysis of Cnot4's impact on transcription:

    • Apply RNA-seq and nascent RNA sequencing (e.g., GRO-seq, PRO-seq) in Cnot4 knockout/knockdown models

    • Studies indicate that inactivating CCR4-NOT leads to increased transcription and activation of LINE and other retrotransposon elements

    • Map Cnot4 genomic binding sites using ChIP-seq or CUT&RUN to identify direct targets

    • Integrate with epigenomic data (histone modifications, chromatin accessibility) to understand context-dependent effects

  • Mechanistic studies of Cnot4's interaction with the transcriptional machinery:

    • Investigate potential interactions with RNA polymerase II complex components

    • Examine relationships with transcription factors and cofactors

    • Explore roles in transcription initiation, elongation, and termination

    • Investigate connections between Cnot4's E3 ubiquitin ligase activity and transcriptional regulation

  • Connection between translational control and transcriptional feedback:

    • Investigate how Cnot4's role in ribosome recognition might influence transcriptional outcomes

    • Explore potential feedback mechanisms between translation efficiency and transcriptional regulation

    • Examine how ubiquitination of ribosomal or nuclear proteins by Cnot4 affects gene expression

  • Retrotransposon regulation mechanisms:

    • Further investigate the novel function for CCR4-NOT in maintaining genome integrity by suppressing retrotransposon activation

    • Explore the relationship between Cnot4 and KZFP gene expression, which appears downregulated upon CCR4-NOT inactivation

    • Characterize Cnot4-dependent mechanisms that prevent mobilization of transposable elements

  • Development and tissue-specific transcriptional programs:

    • Analyze Cnot4's contribution to developmental transcriptional programs in embryogenesis

    • Investigate stage-specific roles during spermatogenesis, particularly in meiotic gene expression

    • Compare Cnot4 functions across diverse tissues and cell types to identify context-dependent activities

These research directions would significantly advance our understanding of how Cnot4 contributes to transcriptional regulation beyond its established roles in post-transcriptional control, potentially revealing new paradigms in the coordination of gene expression.

What emerging technologies could enhance our understanding of Cnot4's molecular functions?

Several cutting-edge technologies show particular promise for advancing our understanding of Cnot4's molecular functions:

  • Advanced structural biology approaches:

    • CryoEM and integrative structural biology: Building on existing cryoEM structures of human CCR4-NOT with ribosomes , apply these techniques to mouse Cnot4 complexes

    • Single-particle cryo-electron tomography: Visualize Cnot4-containing complexes in cellular contexts

    • Time-resolved structural methods: Capture dynamic conformational changes during Cnot4's functional cycle

    • AlphaFold2 and other AI structure prediction: Model Cnot4 interactions with partners and substrates

  • Spatially-resolved transcriptomics and proteomics:

    • Spatial transcriptomics: Map Cnot4-dependent gene expression changes in tissues with spatial context

    • Proximity labeling proteomics (BioID, APEX): Identify Cnot4 interaction partners in specific cellular compartments

    • Single-cell multi-omics: Integrate transcriptomic, proteomic, and epigenomic data at single-cell resolution in Cnot4 models

  • Advanced functional genomics:

    • CRISPR screens with single-cell readouts: Identify genetic interactions with Cnot4

    • Base and prime editing: Introduce precise mutations to dissect domain-specific functions

    • CRISPR activation/repression: Modulate Cnot4 expression with spatiotemporal precision

    • Long-read sequencing: Characterize complex transcriptional effects including isoform changes

  • Live-cell imaging and dynamics:

    • Live-cell single-molecule tracking: Monitor Cnot4 dynamics during cellular processes

    • Optogenetics: Control Cnot4 activity with light to examine acute effects

    • Biosensors: Develop tools to monitor Cnot4-dependent ubiquitination in real-time

    • Super-resolution microscopy: Visualize Cnot4 localization and interactions at nanoscale resolution

  • Translational efficiency and ribosome profiling:

    • Ribosome profiling with computational advances: Map translation efficiency changes in Cnot4 models

    • Selective ribosome profiling: Identify specific mRNAs affected by Cnot4-mediated quality control

    • Translation complex purification: Isolate and characterize Cnot4-associated translation complexes

  • Systems biology integration:

    • Network analysis: Position Cnot4 within gene regulatory networks

    • Multi-scale modeling: Connect molecular functions to cellular and organismal phenotypes

    • Machine learning approaches: Predict Cnot4 targets and functional outcomes from multi-omic data

These emerging technologies, particularly when applied in combination, have the potential to reveal unprecedented insights into Cnot4's diverse molecular functions and their integration into cellular physiology.

How might our understanding of Cnot4 inform broader principles of post-transcriptional gene regulation?

Our evolving understanding of Cnot4 is poised to inform several fundamental principles of post-transcriptional gene regulation:

  • Integration of multiple regulatory layers:

    • Cnot4 exemplifies how a single factor can participate in transcriptional control, mRNA stability regulation, and translational quality control

    • This integration challenges traditional divisions between discrete gene regulatory steps

    • Suggests a model where regulatory proteins serve as "connectors" between different expression layers

    • Future research may reveal additional regulatory proteins that, like Cnot4, coordinate across multiple levels

  • Specificity in RNA decay pathways:

    • Cnot4 functions as a previously unrecognized mRNA adaptor of CCR4-NOT by targeting specific mRNAs to CNOT7 for deadenylation

    • This reveals how general deadenylation machinery achieves transcript specificity

    • Suggests a broader principle where adaptor proteins confer target selectivity to core decay machinery

    • May lead to identification of additional adaptors for other RNA decay pathways

  • Translation-coupled mRNA quality control:

    • The connection between Cnot4, ribosome recognition, and mRNA decay illustrates a sophisticated surveillance system

    • Reveals how cells monitor translation efficiency and couple it to mRNA stability

    • Expands our understanding of ribosome-associated quality control beyond traditional no-go decay

    • May inspire identification of similar mechanisms for other classes of problematic mRNAs

  • Developmental context-dependent regulation:

    • Cnot4's critical role in embryonic development and spermatogenesis highlights how post-transcriptional regulators function in specific developmental windows

    • Illustrates the principle that RNA regulation is dynamically tuned during development

    • Suggests a model where the composition and activity of regulatory complexes shift during cellular differentiation

    • May lead to identification of developmental stage-specific regulatory principles

  • Genome integrity and transposon control:

    • CCR4-NOT's role in suppressing retrotransposon activation reveals connections between post-transcriptional regulation and genome integrity

    • Indicates that RNA decay factors contribute to genomic stability beyond their canonical roles

    • Suggests a broader principle where post-transcriptional mechanisms are integral to defense against mobile genetic elements

    • May inspire exploration of other RNA regulatory factors in transposon control

These principles emerging from Cnot4 research suggest that post-transcriptional regulation is more integrated, specific, and context-dependent than previously appreciated, potentially reshaping our conceptual framework for understanding gene expression control.

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.