MAGOH Human

Mago-Nashi Homolog Human Recombinant
Shipped with Ice Packs
In Stock

Description

MAGOH Human Recombinant produced in E.Coli is a single, non-glycosylated, polypeptide chain containing 154 amino acids (1-146 a.a.) and having a molecular mass of 18.2 kDa. MAGOH protein is fused to a 8 amino acid His-Tag at C-terminus and purified by standard chromatography.

Product Specs

Introduction
Mutations in the MAGOH gene of Drosophila result in offspring with flawed germplasm assembly and germline development. MAGOH, the human counterpart of Drosophila mago nashi, is essential for embryo development. Unlike in Drosophila, where its expression is restricted to the germ plasm, mammalian MAGOH mRNA is ubiquitously expressed in adult tissues and is inducible by serum stimulation in quiescent fibroblasts.
Description
Recombinant human MAGOH protein, expressed in E. coli, is a non-glycosylated polypeptide chain comprising 154 amino acids (residues 1-146) and exhibiting a molecular weight of 18.2 kDa. An 8-amino acid His-tag is fused to the C-terminus of the MAGOH protein. Purification is achieved using standard chromatography techniques.
Physical Appearance
A clear, sterile solution.
Formulation
Human MAGOH is supplied in a solution containing 20mM Tris-HCl (pH 8), 2mM DTT, 0.1M NaCl, and 20% glycerol.
Stability
For short-term storage (2-4 weeks), maintain at 4°C. For extended storage, freeze at -20°C. The addition of a carrier protein (0.1% HSA or BSA) is recommended for long-term storage. Repeated freezing and thawing should be avoided.
Purity
Purity exceeds 90% as assessed by SDS-PAGE.
Synonyms
MAGOH1, MAGOHA, Protein mago nashi homolog, MAGOH, proliferation-associated (Drosophila).
Source
Escherichia Coli.
Amino Acid Sequence

MESDFYLRYY VGHKGKFGHE FLEFEFRPDG KLRYANNSNY KNDVMIRKEA YVHKSVMEEL KRIIDDSEIT KEDDALWPPP DRVGRQELEI VIGDEHISFT TSKIGSLIDV NQSKDPEGLR VFYYLVQDLK CLVFSLIGLH FKIKPILEHH HHHH.

Q&A

What is MAGOH and what is its primary function in human cells?

MAGOH (Protein mago nashi homolog) is a human protein encoded by the MAGOH gene located on chromosome 1. It functions as a core component of the exon junction complex (EJC), playing a critical role in RNA processing, specifically in post-splicing events. MAGOH forms a heterodimer with RBM8A (Y14), which is deposited approximately 24 nucleotides upstream of exon-exon junctions during splicing . This protein is ubiquitously expressed in adult tissues and can be induced by serum stimulation of quiescent fibroblasts . As part of the EJC, MAGOH participates in multiple RNA processing mechanisms including nonsense-mediated mRNA decay, mRNA export, and translational efficiency.

How is MAGOH expression regulated across different human tissues?

MAGOH shows variable expression patterns across different human tissues. According to GTEx data, MAGOH expression is generally higher than its paralog MAGOHB in normal tissues, with notable tissue-specific variation . Brain tissues display relatively lower expression levels of both MAGOH and MAGOHB compared to other tissue types . Interestingly, during brain development, MAGOH and MAGOHB show high expression in early stages of corticogenesis (enriched with neuronal precursor cells) followed by a sharp decrease as cortex morphogenesis progresses . This temporal regulation suggests a critical role for these proteins during neurogenesis and brain development.

What is the relationship between MAGOH and its paralog MAGOHB?

MAGOH and its paralog MAGOHB share remarkably high protein sequence identity (98.6%), despite having relatively lower nucleotide sequence similarity (86%) . Their nonsynonymous to synonymous substitution rate ratio (ω) of 0.0056 indicates strong purifying selection, suggesting functional redundancy between these paralogs . While both proteins are expressed in human tissues, MAGOH typically shows higher and more variable expression than MAGOHB . Both genes are conserved across vertebrates, highlighting their evolutionary importance . Despite their similarities, research indicates they may have some distinct functions, particularly in disease contexts like cancer, where both are frequently overexpressed .

What are the recommended approaches for studying MAGOH protein interactions?

When investigating MAGOH protein interactions, researchers should consider multiple complementary approaches:

  • Co-immunoprecipitation (Co-IP): The primary method to verify direct protein interactions between MAGOH and other proteins like RBM8A and NXF1 . Use antibodies specific to MAGOH to pull down protein complexes, followed by Western blotting or mass spectrometry to identify binding partners.

  • Yeast two-hybrid screening: Useful for identifying novel interaction partners by expressing MAGOH as bait protein and screening against a library of potential interactors.

  • Proximity labeling techniques: BioID or APEX2 fusion proteins can identify proximal proteins in living cells, capturing even transient interactions within the exon junction complex.

  • Crosslinking mass spectrometry (XL-MS): For mapping the structural organization of MAGOH within the EJC by capturing dynamic interactions through chemical crosslinking.

  • Fluorescence resonance energy transfer (FRET): For visualizing MAGOH interactions in living cells by tagging MAGOH and potential partners with appropriate fluorophores.

When designing these experiments, it's critical to include appropriate controls and validate interactions through multiple independent methods to ensure biological relevance.

How can researchers effectively knockdown or overexpress MAGOH in experimental systems?

For manipulating MAGOH expression levels in experimental systems:

Knockdown approaches:

  • siRNA/shRNA: Design targeting sequences specific to MAGOH, avoiding regions with homology to MAGOHB to prevent off-target effects. For studying redundant functions, consider simultaneous knockdown of both MAGOH and MAGOHB .

  • CRISPR-Cas9: For more permanent knockout models, design guide RNAs targeting exons of MAGOH. Be aware that complete knockout may be lethal in some cell types due to MAGOH's essential functions.

  • Verification: Always confirm knockdown efficiency at both mRNA (qRT-PCR) and protein levels (Western blot), ideally achieving >70% reduction.

Overexpression approaches:

  • Plasmid-based expression: Use mammalian expression vectors with appropriate promoters (e.g., CMV for high expression, TET-inducible for controlled expression).

  • Stable cell line generation: Develop MAGOH-overexpressing cell lines like HEK-MAGO, which have shown two- to threefold increases in recombinant protein production compared to wild-type cells .

  • Tagged constructs: Consider adding epitope tags (FLAG, HA, etc.) or fluorescent proteins to facilitate detection while ensuring tags don't interfere with MAGOH function.

  • Verification: Confirm overexpression by qRT-PCR and Western blotting, and assess potential effects on cell viability and function.

What methods are available for studying MAGOH's role in RNA processing and splicing?

To investigate MAGOH's functions in RNA processing and splicing:

  • RNA-seq and splicing analysis: Following MAGOH knockdown or overexpression, perform RNA sequencing to identify global changes in splicing patterns. Focus analysis on alternative splicing events including exon skipping, intron retention, and alternative 5' or 3' splice sites .

  • RT-PCR validation: Design primers flanking alternatively spliced regions identified in RNA-seq to validate specific splicing events using RT-PCR.

  • RNA immunoprecipitation (RIP): Use antibodies against MAGOH to immunoprecipitate associated RNA molecules, followed by sequencing (RIP-seq) to identify direct RNA targets.

  • CLIP-seq methods: Employ crosslinking immunoprecipitation techniques (CLIP, iCLIP, or eCLIP) to map MAGOH binding sites on target RNAs with nucleotide resolution.

  • In vitro splicing assays: Develop minigene constructs containing exons and introns of interest to study the direct effect of MAGOH on specific splicing events in controlled conditions.

  • Polysome profiling: To study MAGOH's role in translation, analyze the distribution of mRNAs on polysomes after MAGOH manipulation.

When interpreting results, focus on the types of transcripts affected, particularly those involved in cell cycle, splicing, and cell division—biological processes most impacted by MAGOH/MAGOHB knockdown in previous studies .

How is MAGOH expression altered in cancer, and what are the implications?

MAGOH and MAGOHB show significant overexpression in multiple cancer types compared to corresponding normal tissues. This phenomenon has been observed in 14 different cancer types analyzed through TCGA data . The most pronounced expression difference between cancer and normal tissue was observed in high-grade glioma (HGG) .

Expression patterns in cancer:

Cancer TypeMAGOH/MAGOHB ExpressionStatistical Significance
High-grade gliomaHighest overexpressionp < 0.01
Other cancers (13 types)Significant overexpressionp < 0.01

Functional implications:

  • In glioblastoma cells, MAGOH/MAGOHB knockdown preferentially affects splicing of transcripts involved in cell cycle, cell division, and RNA processing .

  • High MAGOH/MAGOHB levels appear necessary to safeguard the splicing of genes that are in high demand in rapidly proliferating cells .

  • MAGOH/MAGOHB overexpression is potentially oncogenic, contributing to tumor progression through regulation of critical cell cycle and division genes.

These findings suggest that targeting MAGOH/MAGOHB could be a potential therapeutic strategy for certain cancers, particularly glioblastoma, as differentiated neuronal cells and glioblastoma cells have different requirements for MAGOH/MAGOHB function .

What role does MAGOH play in neurodevelopmental disorders?

MAGOH and the exon junction complex play critical roles in brain development, with implications for neurodevelopmental disorders:

  • Microcephaly connection: Members of the EJC, including MAGOH, are critical during brain development. Mutations in humans or knockout in mouse models result in microcephaly . This aligns with findings that common microcephaly-causing mutations occur in genes implicated in cell division (MCPH1, ASPM, CDK5RAP2, CENPJ, STIL, WDR62, and CEP152) .

  • Developmental expression pattern: MAGOH and MAGOHB show high expression in early stages of corticogenesis and reduced levels in differentiated neuronal cells . This temporal expression pattern suggests a crucial role in neuronal precursor cells and brain development.

  • Splicing regulation: MAGOH/MAGOHB knockdown affects splicing of transcripts implicated in cell cycle and cell division—processes that are essential for proper neurogenesis .

  • Potential mechanisms: Dysregulation of MAGOH likely affects neural progenitor self-renewal, differentiation, and survival through altered splicing of genes essential for brain development.

This connection between MAGOH and neurodevelopment suggests potential implications for understanding and treating microcephaly and other neurodevelopmental disorders with splicing defects as contributing factors.

How do MAGOH and MAGOHB isoforms differ functionally, and what are the implications for EJC assembly?

The MAGOH gene and its paralog MAGOHB produce multiple protein isoforms in humans through alternative splicing, raising important questions about their functional differences . Analysis of these isoforms reveals:

  • Conservation patterns: Both principal and alternate protein isoforms of MAGOH and MAGOHB are evolutionarily conserved across vertebrates, suggesting functional significance . The conservation of these isoforms correlates to the importance of MAGOH and MAGOHB throughout vertebrate evolution.

  • Structural differences: Comparison of amino acid sequences between the principal and alternate protein isoforms shows absence of key amino acid residues in the alternate isoforms . These differences likely affect protein-protein interactions within the EJC.

  • Species variation: While humans express multiple protein isoforms of both MAGOH and MAGOHB, mice produce multiple protein isoforms only for Magohb, not for Magoh . This species-specific difference may reflect evolutionary adaptations in RNA processing mechanisms.

  • Functional implications for EJC assembly:

    • Alternate isoforms lacking key residues may affect the stability or composition of the EJC

    • Isoform ratio changes could modulate EJC activity in different tissues or developmental stages

    • Isoforms might have distinct binding affinities for other EJC components like RBM8A

These findings suggest a complex regulatory system where the balance between different MAGOH and MAGOHB isoforms may fine-tune EJC function in different cellular contexts. Future research should focus on identifying the specific functions of each isoform and how their expression is regulated.

What mechanisms underlie the tissue-specific expression patterns of MAGOH and MAGOHB?

The differential expression of MAGOH and MAGOHB across tissues suggests sophisticated regulatory mechanisms:

  • Transcriptional regulation: Despite their functional similarity, MAGOH shows higher and more variable expression across tissues than MAGOHB . This suggests distinct transcriptional control mechanisms, potentially involving tissue-specific transcription factors.

  • Developmental programming: The high expression of MAGOH and MAGOHB in early neurogenesis followed by decreased expression in mature brain tissue indicates developmental stage-specific regulation . This pattern likely involves epigenetic mechanisms controlling chromatin accessibility at different developmental stages.

  • Feedback mechanisms: The correlation between MAGOH and MAGOHB expression levels (rho > 0.8) across various tissues suggests coordinated regulation , potentially through feedback mechanisms sensing the total MAGOH/MAGOHB protein levels.

  • Post-transcriptional control: The generation of multiple RNA transcripts from both genes, not all of which code for proteins, suggests important post-transcriptional regulatory mechanisms .

  • Evolutionary constraints: The strong purifying selection observed (ω = 0.0056) indicates evolutionary pressure to maintain both paralogs , suggesting they fulfill subtly different but essential functions that cannot be completely compensated by the other paralog.

Understanding these regulatory mechanisms could provide insights into how cells balance MAGOH/MAGOHB expression to meet tissue-specific requirements for RNA processing and ensure proper development and cellular function.

How might MAGOH function be targeted therapeutically in cancer while minimizing neurodevelopmental side effects?

The differential requirements for MAGOH/MAGOHB between cancer cells (particularly glioblastoma) and differentiated neuronal cells present both an opportunity and a challenge for therapeutic development:

  • Selective targeting strategies:

    • Threshold-based approach: Target MAGOH/MAGOHB to levels that impair cancer cell function but remain sufficient for normal cells, exploiting the higher dependency of cancer cells .

    • Isoform-specific targeting: Design therapies targeting cancer-specific isoforms or expression patterns while sparing those essential for neuronal function .

    • Context-dependent inhibition: Develop inhibitors that specifically disrupt MAGOH interactions with cancer-promoting factors while preserving essential functions.

  • Therapeutic approaches:

    • Antisense oligonucleotides: Design these to modulate specific splicing events affected by MAGOH/MAGOHB overexpression in cancer.

    • Small molecule inhibitors: Develop compounds that disrupt specific protein-protein interactions of MAGOH within the cancer context.

    • Synthetic lethality: Identify and target genes that become essential specifically in MAGOH/MAGOHB-overexpressing cancers.

  • Potential biomarkers for patient selection:

    • MAGOH/MAGOHB expression levels could serve as biomarkers for patient stratification .

    • Survival analyses from TCGA and CGGA cohorts stratifying patients based on MAGOH/MAGOHB expression levels could identify patients most likely to benefit from such therapies .

  • Developmental considerations:

    • Due to the critical role of MAGOH in neurodevelopment, therapeutic approaches must consider developmental stage-specific effects.

    • Treatment strategies would need careful design to avoid neurodevelopmental side effects, particularly in pediatric patients.

This therapeutic approach is particularly promising for glioblastoma, where MAGOH/MAGOHB are significantly overexpressed, and their inhibition would impact cancer cells while potentially sparing differentiated neuronal cells .

What is the role of MAGOH in optimizing recombinant protein production in mammalian cells?

MAGOH overexpression has emerged as a promising strategy for enhancing recombinant protein production in mammalian expression systems:

  • Enhanced translational efficiency: Tethering MAGOH to specific mRNA sequences has been shown to significantly increase mRNA translational efficiency . This occurs through MAGOH's role in the exon junction complex, which influences mRNA export, surveillance, and translation.

  • HEK-MAGO cell line development: A HEK293 cell line overexpressing MAGOH (HEK-MAGO) has been developed specifically for therapeutic protein production . This represents a significant advance in expression technology for biopharmaceutical applications.

  • Production enhancements: The HEK-MAGO cell line and derived clones have demonstrated two- to threefold increases in recombinant human erythropoietin (rhEPO) production compared to wild-type cells .

  • Promoter-independent effect: The enhancement of protein expression in MAGOH-overexpressing cells is promoter-independent , highlighting the versatility of this expression platform for various expression constructs.

  • Advantages over CHO cells: While Chinese Hamster Ovary (CHO) cells remain the most commonly used host for therapeutic protein expression, they produce glycans not present in human cells that are potentially immunogenic . Human-derived cells like HEK-MAGO offer potential advantages for producing proteins with human-compatible post-translational modifications.

This application of MAGOH represents an important advancement in biopharmaceutical production technology, potentially enabling more efficient production of complex therapeutic proteins while maintaining human-compatible post-translational modifications.

What are the key experimental controls needed when studying MAGOH function?

When designing experiments to study MAGOH function, researchers should implement these critical controls:

  • Paralog considerations: Due to the high sequence similarity and potential functional redundancy between MAGOH and MAGOHB, experiments should:

    • Include separate knockdowns of MAGOH and MAGOHB alongside combined knockdowns

    • Verify specificity of antibodies and nucleic acid probes for distinguishing between paralogs

    • Consider complementation experiments where one paralog is expressed in the absence of the other

  • Isoform-specific controls:

    • When studying a specific MAGOH isoform, include controls for other isoforms to determine isoform-specific effects

    • Design experiments to distinguish between effects of protein-coding and non-coding transcripts from MAGOH genes

  • Splicing analysis validation:

    • Include both computational analysis and experimental validation (RT-PCR) of splicing changes

    • Use multiple splicing detection algorithms to avoid method-specific biases

    • Validate key splicing events in independent biological samples

  • Phenotypic rescues:

    • Perform rescue experiments with wild-type MAGOH to confirm phenotypes are specifically due to MAGOH loss

    • Consider cross-species rescue (e.g., Drosophila mago nashi) to assess functional conservation

  • Tissue-specific considerations:

    • Include appropriate tissue-specific controls when studying MAGOH in different contexts, as expression and function vary across tissues

    • For cancer studies, include both matched normal tissue and cancer samples to account for tissue-specific variations

These controls are essential for distinguishing between MAGOH-specific effects, paralog-redundant functions, and potential off-target effects in experimental systems.

What are the most sensitive methods for detecting changes in MAGOH-regulated splicing events?

Detecting MAGOH-regulated splicing events requires sensitive and specific methodologies:

  • High-throughput approaches:

    • RNA-seq with sufficient depth: At least 50-100 million paired-end reads per sample to capture low-abundance transcripts and subtle splicing changes

    • Junction-centric analysis: Focus on exon-exon junction reads rather than whole transcript abundance

    • Specialized algorithms: MISO, rMATS, MAJIQ, or VAST-TOOLS specifically designed to detect and quantify alternative splicing events

  • Targeted validation methods:

    • RT-PCR with isoform-specific primers: Design primers spanning exon-exon junctions to amplify specific splice variants

    • Droplet digital PCR (ddPCR): For absolute quantification of splice variants with high sensitivity

    • Minigene splicing assays: For direct testing of specific splicing events in controlled contexts

  • Visualization and quantification:

    • Sashimi plots: To visualize RNA-seq reads supporting specific splicing events

    • Percent Spliced In (PSI): Calculate the proportion of transcripts including a specific exon or splice site

    • ∆PSI thresholds: Consider changes of ≥10-15% in PSI values with statistical significance as biologically relevant

  • Functional categorization:

    • Perform Gene Ontology (GO) analysis on genes with altered splicing to identify enriched biological processes

    • Previous studies have shown MAGOH/MAGOHB knockdown preferentially affects splicing of genes involved in cell division/cycle, RNA processing, and translation

For comprehensive analysis, researchers should employ multiple complementary methods and focus on splicing events consistently detected across different analytical approaches to minimize false positives.

Product Science Overview

Introduction

The Mago-Nashi homolog, often referred to as MAGOH, is a protein that plays a crucial role in various biological processes. It is the human homolog of the Drosophila mago nashi protein, which is essential for normal germplasm development in the Drosophila embryo . In humans, MAGOH is involved in the exon junction complex (EJC), which is critical for mRNA processing and regulation.

Gene and Protein Structure

The MAGOH gene encodes a protein that consists of 146 amino acids and has a molecular weight of approximately 18 kDa . The protein is expressed ubiquitously in adult tissues and can be induced by serum stimulation of quiescent fibroblasts . The recombinant human MAGOH protein is often expressed in E. coli and purified using conventional chromatography techniques .

Biological Functions

MAGOH is a component of the exon junction complex (EJC), which is deposited on messenger RNAs (mRNAs) during splicing . The core of the EJC consists of eIF4A3, MLN51, Y14, and MAGOH . This complex plays a vital role in mRNA surveillance and nonsense-mediated decay (NMD), a process that degrades mRNAs containing premature stop codons .

Clinical Significance

Diseases associated with MAGOH include metaphyseal chondrodysplasia, Schmid type, and primary autosomal recessive microcephaly . The protein’s involvement in mRNA processing and regulation makes it a significant target for research in various genetic disorders and developmental abnormalities.

Recombinant Human MAGOH

Recombinant human MAGOH protein is typically produced with a polyhistidine tag at the C-terminus to facilitate purification . The protein is supplied in a buffer solution and is stable for up to twelve months when stored at -20°C to -80°C . It is used in various research applications to study its function and role in different biological processes.

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.