C16ORF53 Human

Chromosome 16 Open Reading Frame 53 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Overview of C16ORF53 Human

C16ORF53 (also known as PAGR1, PA1, or GAS) is a protein-coding gene located at chromosome 16p11.2 in humans. It encodes a 274-amino-acid protein with a molecular weight of 29.9 kDa (calculated), though observed weights on SDS-PAGE appear higher due to post-translational modifications or tags . The gene is evolutionarily conserved and plays roles in chromatin regulation, genome stability, and cellular proliferation .

Key identifiers:

AttributeValue
Gene SymbolC16ORF53, PAGR1
Entrez Gene ID79447
Chromosome Location16p11.2
Protein Length274 amino acids
Molecular Weight29.9 kDa (calculated)
Recombinant HostE. coli

Protein Structure

  • Domain Composition: Contains a glutamate-rich region central to its coactivator function .

  • Post-Translational Modifications: His-tagged recombinant versions are purified via chromatography and used in biochemical assays .

Functional Roles

C16ORF53 is a core component of Set1-like histone methyltransferase complexes, which catalyze H3K4 methylation – a mark of active transcription . Key interactions include:

  1. PAXIP1/PTIP: Direct interaction required for association with chromatin-modifying complexes .

  2. SRC1: Preferential binding to SRC1 (vs. GRIP1/AIB1) in oestrogen receptor (ERα)-mediated transcription .

  3. ERα: Participates in ERα-regulated gene expression and G1/S cell-cycle progression .

Neurodevelopmental and Disease Associations

  • 16p11.2 CNVs: Deletions/duplications in this region are linked to autism, schizophrenia, and neurodevelopmental disorders. C16ORF53 interacts with genes like MAPK3 and KCTD13 in modulating cell proliferation pathways .

  • Drosophila Models: Knockdown of homologs (coro/C16ORF53) disrupts neurogenesis and dendritic complexity, implicating conserved roles in neural development .

Cancer and Cellular Signaling

  • Estrogen Receptor Signaling: Enhances ERα-mediated transcription of target genes (e.g., pS2, c-Myc) and promotes cell-cycle progression in breast cancer cells .

  • H3K4 Methylation: While part of a methyltransferase complex, knockdown minimally affects H3K4 methylation at promoters, suggesting indirect regulatory mechanisms .

Genetic Interactions in 16p11.2

C16ORF53 interacts with genes in the 16p11.2 region to modulate phenotypes:

Interacting GeneFunctionPhenotypic Impact
MAPK3MAPK/ERK signalingRescues rough-eye phenotype in flies
KCTD13Cell cycle regulationAlters ommatidial size in flies
CHD8Chromatin remodelingSuppresses KCTD13 rough-eye phenotype

Potential Therapeutic Targets

  • Kinase Inhibitors: Rapamycin (mTOR) and sorafenib (RAF1) show promise in rescuing phenotypes linked to 16p11.2 deletions .

  • Epigenetic Modulators: Targeting H3K4 methylation or ERα pathways may address transcriptional dysregulation in cancer or neurodevelopmental disorders .

Recombinant Proteins and Tools

Product TypeFeaturesApplications
His-tagged C16ORF5329.9 kDa, >85% purity, E. coli-derivedSDS-PAGE, ELISA, biochemistry
Adenovirus VectorsCMV-driven overexpression, GFP/mCherry reporters optionalGene expression studies
AntibodiesRabbit polyclonal (WB, ELISA), validated in HEK-293 cells Protein detection, ChIP assays

Product Specs

Introduction
C16ORF53, a component of the Set1-like multiprotein histone methyltransferase complex, interacts directly with PAXIP1/PTIP, facilitating its association with the PTIP complex. This interaction is vital for genome stability, chromatin condensation, and mitotic progression.
Description
Produced in E. coli, the non-glycosylated C16ORF53 polypeptide contains 274 amino acids (1-254 a.a.) with a molecular mass of 29.9 kDa. Note: SDS-PAGE analysis might show a higher molecular weight due to the 20 amino acid His-tag fused at the N-terminus. Purification is achieved using proprietary chromatographic techniques.
Physical Appearance
Sterile, colorless solution.
Formulation
C16ORF53 protein solution (1 mg/ml) in 20 mM Tris-HCl buffer (pH 8.0), 10% glycerol, 2 mM DTT, and 0.1 M NaCl.
Stability
For optimal storage, keep C16ORF53 Human Recombinant below -18°C. While it remains stable at 4°C for up to one week, repeated freeze-thaw cycles should be avoided.
Purity
Purity exceeds 85%, as determined by SDS-PAGE analysis.
Synonyms

PAXIP1-associated protein 1, PTIP-associated protein 1, PA1, C16orf53, GAS.

Source
Escherichia Coli.
Amino Acid Sequence

MGSSHHHHHH SSGLVPRGSH MSLARGHGDT AASTAAPLSE EGEVTSGLQA LAVEDTGGPS ASAGKAEDEG EGGREETERE GSGGEEAQGE VPSAGGEEPA EEDSEDWCVP CSDEEVELPA DGQPWMPPPS EIQRLYELLA AHGTLELQAE ILPRRPPTPE AQSEEERSDE EPEAKEEEEE KPHMPTEFDF DDEPVTPKDS LIDRRRTPGS SARSQKREAR LDKVLSDMKR HKKLEEQILR TGRDLFSLDS EDPSPASPPL RSSGSSLFPR QRKY.

Q&A

What is the current nomenclature for C16ORF53 and what are its basic molecular characteristics?

C16ORF53, now officially designated as PAXIP1 associated glutamate-rich protein 1 (PAGR1), is a component of a Set1-like multiprotein histone methyltransferase complex as described by Cho et al. in 2007 . The gene is also known by its synonym PA1 in some literature. PAGR1 is assigned NCBI Gene ID 79447 and encodes the human protein PAGR1_HUMAN .

Methodologically, researchers should be aware that database searches may require using all three designations (C16ORF53, PAGR1, and PA1) to ensure comprehensive literature coverage. When designing primers or probes, researchers should verify current reference sequences since nomenclature changes can affect research continuity and reproducibility.

Where is PAGR1 expressed in human tissues and at what developmental stages?

Expression data from the Allen Brain Atlas indicates that PAGR1 shows variable expression across different brain regions and developmental stages . The gene has over 4,000 functional associations with biological entities spanning 8 categories extracted from 84 datasets, including tissue expression profiles, suggesting a multifaceted role in cellular processes .

Methodologically, researchers can access tissue-specific expression data through resources like the Allen Brain Atlas, which provides both adult and developmental expression profiles across human and mouse brain tissues . When analyzing expression patterns, it's critical to normalize data appropriately and consider developmental trajectories when designing experiments targeting specific brain regions or developmental windows.

What is PAGR1's genomic context and its relation to 16p11.2 copy number variations?

PAGR1 is located within the proximal 16p11.2 region (BP4-BP5), which spans approximately 593 Kb and contains multiple genes frequently affected by copy number variations (CNVs) . Specifically, PAGR1 is among the genes within the BP4-BP5 region that includes approximately 25-30 genes such as KCTD13, TBX6, and MAPK3 . This region is susceptible to recurrent microdeletions and microduplications associated with neurodevelopmental disorders.

For research design, it's important to consider that studies targeting PAGR1 alone may not capture the full complexity of phenotypes associated with 16p11.2 CNVs, as neighboring genes likely contribute to the observed clinical presentations through genetic interactions.

What experimental approaches can effectively distinguish PAGR1's specific role from other genes in the 16p11.2 region?

Distinguishing PAGR1's specific contributions requires multi-layered experimental approaches:

  • CRISPR-based methodologies: Create precise deletions or mutations of PAGR1 while maintaining the integrity of surrounding genes. This can be complemented with rescue experiments reintroducing wild-type or mutant PAGR1.

  • Interaction studies: Research has demonstrated genetic interactions between 16p11.2 homologs in Drosophila models, with 24 interactions identified between pairs of 16p11.2 homologs and 46 interactions between these homologs and other neurodevelopmental genes . Similar interaction screening approaches in human cellular models could distinguish PAGR1-specific interactions.

  • Comparative phenotyping: Studies in patients with atypical CNVs that include or exclude PAGR1 can help attribute specific phenotypes to this gene. For example, patient 7 in the clinical cohort presents with macrocephaly, intellectual disability, and speech delay with a deletion encompassing PAGR1, providing evidence for potential PAGR1 contributions to these phenotypes .

How can researchers effectively model and measure the epigenetic impacts of PAGR1 dysfunction?

As a component of histone methyltransferase complexes , PAGR1 likely influences epigenetic regulation. Researchers should consider:

  • ChIP-seq and CUT&RUN: These techniques can map genome-wide distribution of histone modifications affected by PAGR1 manipulation.

  • Combined RNA-seq and epigenomic profiling: This approach allows correlation between gene expression changes and alterations in chromatin states following PAGR1 manipulation.

  • Single-cell approaches: Given the likely cell type-specific effects, single-cell techniques provide resolution needed to detect subtle epigenetic changes in specific neural populations.

  • Time-course experiments: Since neurodevelopmental disorders reflect altered developmental trajectories, examining epigenetic changes across developmental timepoints is critical.

Methodologically, it's essential to include appropriate controls and validate findings across multiple experimental systems, as epigenetic effects can be highly context-dependent.

What is the current understanding of PAGR1's role in genetic interaction networks affecting neurodevelopment?

Evidence from Drosophila models suggests that PAGR1 (C16ORF53) participates in genetic interaction networks that modulate neurodevelopmental phenotypes . Research has identified specific interactions that suppress or enhance cell proliferation phenotypes, pointing to a complex role within developmental regulatory networks .

These genetic interactions are also enriched in human brain-specific networks, suggesting translational relevance to human neurodevelopment . The concept of "pervasive genetic interactions" supports a model where CNV genes interact with each other in conserved pathways to modulate phenotypic expression .

Methodologically, researchers should consider:

  • Network analysis approaches that incorporate protein-protein interactions, co-expression data, and functional genomics

  • Quantitative interaction screens in multiple model systems

  • Integration of human genetic data with experimental interaction data

How does PAGR1 dysfunction potentially contribute to the phenotypic spectrum observed in 16p11.2-related disorders?

The 16p11.2 region containing PAGR1 is associated with a wide phenotypic spectrum, including intellectual disability, speech impairment (70%), motor coordination difficulties (60%), autism spectrum disorder (20-25%), and seizures . Patient data from 16p11.2 deletion carriers shows:

PhenotypeFrequency in Patients with BP4-BP5 Deletion
Intellectual disability/speech impairment~70%
Motor coordination difficulties~60%
Autism spectrum disorder20-25%
SeizuresVariable
Head circumference abnormalitiesCommon (macrocephaly with deletion)

PAGR1's role in histone methyltransferase complexes suggests it may influence gene expression programs critical for neurodevelopment. The "mirror phenotypes" observed between deletions and duplications of the 16p11.2 region (e.g., macrocephaly vs. microcephaly) suggest dosage-sensitive mechanisms .

For clinical researchers, it's important to:

  • Collect detailed phenotypic data

  • Consider gene-dosage effects

  • Account for genetic background influences on expressivity

  • Investigate both cell-autonomous and non-cell-autonomous effects

What biomarkers might indicate altered PAGR1 functionality in patient samples?

Given PAGR1's role in histone methyltransferase complexes, potential biomarkers could include:

  • Epigenetic signatures: Altered patterns of specific histone modifications (particularly H3K4 methylation marks) in accessible patient samples like blood or induced pluripotent stem cells (iPSCs).

  • Transcriptomic changes: Expression changes in genes regulated by PAGR1-containing complexes.

  • Cellular phenotypes: In patient-derived cells, alterations in cell proliferation pathways would be consistent with findings from model systems showing PAGR1's involvement in these processes .

  • Protein complex integrity: Analysis of protein complexes in patient cells might reveal altered assembly or composition of histone methyltransferase complexes containing PAGR1.

Methodologically, researchers should establish reliable normative data from matched controls and consider both technical and biological variability when evaluating potential biomarkers.

How can variable expressivity in patients with PAGR1-affecting CNVs be reconciled with molecular mechanisms?

The clinical data reveals significant phenotypic variability even within families carrying identical CNVs affecting PAGR1 . This suggests several mechanisms that researchers should consider:

  • Genetic modifiers: Additional genetic variants may interact with PAGR1 or other 16p11.2 genes to modify phenotypic expression.

  • Complex interaction networks: As demonstrated in Drosophila models, genes within the 16p11.2 region participate in extensive interaction networks . Variation in these networks could contribute to phenotypic differences.

  • Environmental factors: Epigenetic mechanisms influenced by environmental exposures may modify the impact of PAGR1 dysfunction.

  • Developmental compensation: Compensatory mechanisms during development may buffer against genetic perturbations to varying degrees.

Researchers investigating variable expressivity should employ comprehensive approaches including whole-genome sequencing to identify potential modifiers, detailed environmental history collection, and longitudinal studies to capture developmental trajectories.

What cellular and animal models are most appropriate for studying PAGR1 function in neurodevelopment?

Selecting appropriate models requires consideration of research questions and translational goals:

  • Cellular models:

    • Neural progenitor cells and differentiated neurons derived from human iPSCs

    • Isogenic cell lines with PAGR1 mutations or deletions

    • 3D brain organoids to capture more complex developmental processes

  • Animal models:

    • Drosophila: Already demonstrated utility for genetic interaction studies

    • Mouse models: For more complex behavioral and developmental phenotyping

    • Zebrafish: Useful for high-throughput screening and live imaging of neurodevelopmental processes

Each model system offers distinct advantages, but researchers should be aware of species-specific differences in PAGR1 function and the genomic context of the 16p11.2 region.

How can researchers effectively measure PAGR1's impact on histone methylation patterns?

To assess PAGR1's role in histone methylation, researchers should consider:

  • Genome-wide approaches:

    • ChIP-seq targeting specific histone methylation marks (particularly H3K4me3)

    • CUT&RUN or CUT&Tag for higher resolution profiling

    • ChIP-MS to identify protein complexes associated with specific genomic loci

  • Locus-specific approaches:

    • ChIP-qPCR at candidate genes

    • CRISPRi-based recruitment assays to test direct effects

  • Functional readouts:

    • Reporter assays linked to methylation-sensitive promoters

    • Mass spectrometry quantification of histone modifications

When designing these experiments, controls should include manipulation of known histone methyltransferase components and careful normalization for cellular heterogeneity.

What approaches can detect interaction effects between PAGR1 and other genes in the 16p11.2 region?

Building on the genetic interaction studies in Drosophila , researchers can apply several approaches to human systems:

  • Combinatorial genetic perturbations:

    • CRISPR-based multiplexed gene editing

    • shRNA/siRNA combinatorial knockdowns

    • Overexpression combined with knockdown

  • Protein-protein interaction studies:

    • Proximity labeling (BioID, APEX)

    • Co-immunoprecipitation followed by mass spectrometry

    • Yeast two-hybrid or mammalian two-hybrid screens

  • Functional genomic screens:

    • CRISPR screens in sensitized backgrounds

    • Synthetic lethality/sickness screens

    • Modifier screens in model organisms

  • Computational approaches:

    • Network analysis of existing datasets

    • Machine learning to predict genetic interactions

    • Integration of multi-omics data

Data interpretation should focus on identifying synergistic or antagonistic effects that deviate from expected additive effects of single-gene perturbations.

How should researchers analyze RNA-seq data to identify downstream targets of PAGR1 activity?

When analyzing transcriptomic data from PAGR1 perturbation experiments:

  • Differential expression analysis:

    • Apply appropriate statistical methods accounting for multiple testing

    • Consider both magnitude of change (fold change) and statistical significance

    • Validate key findings with qRT-PCR

  • Pathway and network analysis:

    • Gene set enrichment analysis (GSEA)

    • Weighted gene co-expression network analysis (WGCNA)

    • Integration with protein-protein interaction databases

  • Integration with epigenomic data:

    • Correlate expression changes with alterations in histone modifications

    • Identify direct targets through integration with ChIP-seq data

  • Cell-type specific considerations:

    • Single-cell RNA-seq to resolve cell-type specific effects

    • Deconvolution approaches for bulk RNA-seq

Careful experimental design including appropriate time points and replicates is essential for meaningful interpretation of transcriptomic changes.

What statistical approaches are most appropriate for analyzing genetic interactions involving PAGR1?

Based on interaction studies in model systems , researchers should consider:

  • Quantitative interaction metrics:

    • Multiplicative vs. additive models for expected combined effects

    • Deviation from expected combined effects as measure of interaction strength

    • Statistical tests specific for interaction effects in linear models

  • Multiple testing correction:

    • Appropriate FDR control for large-scale interaction screens

    • Hierarchical testing strategies to improve power

  • Bayesian approaches:

    • Prior probability incorporation based on existing knowledge

    • Network-informed priors for interaction likelihood

  • Visualization techniques:

    • Interaction heat maps

    • Network visualization with interaction strength encoding

    • Genetic interaction profiles as signatures for functional similarity

Researchers should be transparent about interaction definitions and statistical thresholds used to identify significant interactions.

How can researchers reconcile contradictory findings regarding PAGR1 function across different experimental systems?

Contradictory findings may arise from biological or technical factors:

  • Biological factors:

    • Cell type-specific effects

    • Developmental stage differences

    • Species-specific functions

    • Genetic background influences

  • Technical considerations:

    • Differences in knockdown/knockout efficiency

    • Off-target effects

    • Differences in assay sensitivity

    • Cellular stress responses to manipulation

Reconciliation approaches include:

  • Direct comparison studies using standardized methodologies

  • Meta-analysis of published findings

  • Collaboration between labs with contradictory findings

  • Use of multiple complementary approaches to address the same question

Researchers should avoid overinterpreting findings from single experimental systems and seek convergent evidence across multiple approaches.

Product Science Overview

Introduction

Chromosome 16 Open Reading Frame 53 (C16orf53) is a gene located on the 16th chromosome in humans. This gene encodes a protein that is involved in various cellular processes. The recombinant form of this protein, produced through biotechnological methods, is used in research to understand its function and potential applications in medicine.

Chromosome 16 Overview

Chromosome 16 is one of the 23 pairs of chromosomes in humans. It spans approximately 90 million base pairs and represents just under 3% of the total DNA in cells . This chromosome contains a significant number of genes, including those involved in metabolic pathways, immune response, and developmental processes.

Gene Structure and Function

The C16orf53 gene is one of the many open reading frames (ORFs) on chromosome 16. An open reading frame is a sequence of DNA that has the potential to be translated into a protein. The C16orf53 gene encodes a protein that consists of 274 amino acids and has a molecular mass of approximately 29.9 kDa . This protein is non-glycosylated and is produced in Escherichia coli (E. coli) for research purposes.

Recombinant Protein Production

The recombinant form of C16orf53 is produced using E. coli expression systems. The gene encoding C16orf53 is inserted into a plasmid vector, which is then introduced into E. coli cells. These cells are cultured under specific conditions to express the protein. The recombinant protein is then purified using chromatographic techniques to ensure its purity and functionality .

Applications in Research

The recombinant C16orf53 protein is used in various research applications, including:

  • Functional Studies: Researchers use the recombinant protein to study its role in cellular processes and its interaction with other proteins.
  • Disease Research: Understanding the function of C16orf53 can provide insights into its involvement in diseases and potential therapeutic targets.
  • Biotechnological Applications: The recombinant protein can be used in the development of diagnostic tools and therapeutic agents.

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.