Recombinant Rat Immunoglobulin superfamily member 10 (Igsf10), partial

Shipped with Ice Packs
In Stock

Product Specs

Form
Lyophilized powder
Note: While we prioritize shipping the format currently in stock, please specify your preferred format in order notes for customized preparation.
Lead Time
Delivery times vary depending on the purchasing method and location. Please contact your local distributor for precise delivery estimates.
Note: Standard shipping includes blue ice packs. Dry ice shipping requires advance notice 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 consolidate 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 serve as a reference.
Shelf Life
Shelf life depends on various factors including storage conditions, buffer components, temperature, and protein stability. Generally, liquid formulations have a 6-month shelf life at -20°C/-80°C, while lyophilized formulations have a 12-month shelf life at -20°C/-80°C.
Storage Condition
Store at -20°C/-80°C upon receipt. Aliquoting is essential for multiple uses. Avoid repeated freeze-thaw cycles.
Tag Info
Tag type is determined during the manufacturing process.
The tag type is determined during production. If you require a specific tag, please inform us, and we will prioritize its development.
Synonyms
Igsf10; Cmf608; Immunoglobulin superfamily member 10; IgSF10; Calvaria mechanical force protein 608; CMF608
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Protein Length
Partial
Purity
>85% (SDS-PAGE)
Species
Rattus norvegicus (Rat)
Target Names
Igsf10
Uniprot No.

Target Background

Function
This protein plays a role in regulating the early migration of gonadotropin-releasing hormone (GnRH) neurons and may contribute to maintaining the osteochondroprogenitor cell pool.
Database Links
Subcellular Location
Secreted.
Tissue Specificity
Expressed in bone. Main expression is present in mesenchymal osteochondroprogenitors with fibroblast-like morphology abundant in the regions of active bone modeling and remodeling.

Q&A

What is the molecular structure and function of rat IGSF10?

Rat IGSF10 is a large protein belonging to the immunoglobulin superfamily, characterized by multiple immunoglobulin-like domains. Similar to its human counterpart, rat IGSF10 likely has N-terminal and C-terminal functional regions that play distinct roles. The protein is expressed during embryonic development, particularly in nasal mesenchyme, where it guides GnRH neuronal migration to the hypothalamus .

Research methodology for studying IGSF10 structure typically involves:

  • Bioinformatic analysis using multiple sequence alignment to identify conserved domains

  • Structural prediction using crystallography or cryo-EM

  • Domain mapping using truncation mutants and functional testing

IGSF10 appears to function as a signaling protein that creates an extracellular environment conducive for proper GnRH neuronal migration. Knockdown experiments have demonstrated that IGSF10 deficiency leads to impaired migration of GnRH neurons, suggesting its crucial role in the development of the hypothalamic-pituitary-gonadal axis .

How does rat IGSF10 expression vary across developmental stages?

Rat IGSF10 shows temporally regulated expression patterns, with studies indicating strong expression in embryonic nasal mesenchyme during critical periods of GnRH neuronal migration to the hypothalamus . Understanding these expression patterns requires:

  • RT-qPCR analysis across multiple developmental timepoints

  • In situ hybridization to localize mRNA expression in tissue sections

  • Immunohistochemistry with anti-IGSF10 antibodies for protein localization

For RT-qPCR studies of rat IGSF10, researchers can utilize primers similar to those documented in breast cancer studies:

Forward primer (IGSF10): 5′-TTGGAGTTTGCCTGATGGAAC-3′
Reverse primer (IGSF10): 5′-CGCTACCCCAACTTTGTTGAAG-3′

Standardization against housekeeping genes like GAPDH is essential for accurate quantification:

Forward primer (GAPDH): 5′-GGAGCGAGATCCCTCCAAAAT-3′
Reverse primer (GAPDH): 5′-GGCTGTTGTCATACTTCTCATGG-3′

What expression systems are optimal for producing recombinant rat IGSF10?

The choice of expression system for recombinant rat IGSF10 depends on experimental needs:

  • Bacterial systems (E. coli):

    • Advantages: High yield, cost-effective, rapid production

    • Limitations: Lack of post-translational modifications, potential improper folding

    • Best for: Partial IGSF10 domains, structural studies of individual domains

  • Mammalian systems (HEK293, CHO cells):

    • Advantages: Proper folding, post-translational modifications similar to native protein

    • Limitations: Lower yield, higher cost, more complex protocols

    • Best for: Full-length functional studies, interaction analyses, secretion studies

  • Insect cell systems (Sf9, High Five):

    • Advantages: Higher yield than mammalian cells, some post-translational modifications

    • Limitations: Modifications not identical to mammalian cells

    • Best for: Balance between yield and functionality

For studying IGSF10 secretion pathways and mutations that affect protein trafficking, mammalian expression systems are essential, as demonstrated in studies of human IGSF10 variants that resulted in intracellular retention with failure in protein secretion .

How can researchers overcome challenges in expressing full-length versus partial rat IGSF10?

Full-length IGSF10 expression presents significant challenges due to its large size. Methodological approaches include:

  • For full-length expression:

    • Use expression vectors with strong promoters (CMV for mammalian cells)

    • Optimize codon usage for the host system

    • Consider adding secretion signal sequences for improved trafficking

    • Use fusion tags (His, GST) that can be later removed by protease cleavage

  • For partial IGSF10 expression:

    • Analyze domain boundaries through bioinformatics

    • Focus on functional domains (N-terminal domains implicated in neuronal migration)

    • Express multiple overlapping fragments to ensure coverage of functional regions

Based on human IGSF10 studies, particular attention should be paid to the N-terminal region containing mutations p.Arg156Leu and p.Glu161Lys, which have been associated with delayed puberty and demonstrated strong segregation with autosomal dominant inheritance patterns .

What assays can effectively measure rat IGSF10 activity in neuronal migration?

Several methodological approaches can be employed to assess IGSF10's impact on neuronal migration:

  • In vitro migration assays:

    • Boyden chamber/Transwell assays with GnRH neurons

    • Time-lapse microscopy to track neuronal movement

    • Gap closure/wound healing assays for migratory cell populations

  • Ex vivo approaches:

    • Explant cultures from embryonic nasal regions

    • Slice cultures with labeled GnRH neurons

  • Quantification methods:

    • Track distance traveled by neurons over time

    • Measure directionality and persistence of migration

    • Analyze cellular morphology and leading-edge formation

IGSF10 knockdown has been shown to cause reduced migration of immature GnRH neurons in vitro, providing a basis for experimental design . These assays should include appropriate controls, including scrambled siRNA for knockdown studies and vector-only controls for overexpression experiments.

How can researchers investigate the signaling pathways associated with rat IGSF10?

Based on GSEA (Gene Set Enrichment Analysis) data from IGSF10 studies, several pathways have been associated with IGSF10 function:

  • Pathway analysis methodology:

    • Phosphoproteomic analysis before and after IGSF10 stimulation

    • RNA-seq to identify transcriptional changes

    • Western blotting for key pathway components

    • Pharmacological inhibition of suspected pathways

  • Key pathways to investigate:

    • PI3K/Akt/mTOR signaling pathway

    • mTORC1 signaling

    • TGF-β signaling pathway

    • EMT (Epithelial-Mesenchymal Transition)

    • TNF signaling pathway via NFκB

  • Interaction studies:

    • Co-immunoprecipitation to identify binding partners

    • Proximity ligation assays for in situ interaction detection

    • Yeast two-hybrid screening for novel interactors

Studies have indicated that IGSF10 expression positively correlates with several cancer-related biological processes, including DNA repair (HALLMARK_DNA_REPAIR), cell cycle (HALLMARK_G2M_CHECKPOINT), and glycolysis (HALLMARK_GLYCOLYSIS) pathways , suggesting multiple signaling functions beyond neuronal migration.

What approaches are most effective for generating and validating rat models with IGSF10 mutations?

Creating rat models with IGSF10 mutations requires carefully designed strategies:

  • Generation methods:

    • CRISPR-Cas9 gene editing for precise mutations

    • Transgenic overexpression of mutant IGSF10

    • Conditional knockout systems (Cre-loxP) for tissue-specific studies

  • Target mutations to consider:

    • Homologous mutations to human variants with known phenotypes:

      • p.Arg156Leu (c.467G>T)

      • p.Glu161Lys (c.481G>A)

      • p.Glu2264Gly (c.6791A>G)

      • p.Asp2614Asn (c.7840G>A)

  • Validation approaches:

    • Genotyping using PCR and sequencing

    • Expression analysis (mRNA and protein levels)

    • Functional validation through phenotyping (timing of puberty, fertility)

    • Histological analysis of GnRH neuronal positioning

When designing CRISPR-Cas9 targeting strategies, researchers should consider the conservation of targeted regions between rat and human IGSF10 sequences to ensure mutation of functionally relevant residues.

How can researchers phenotypically characterize IGSF10 mutant rat models?

Comprehensive phenotypic characterization should include:

  • Developmental timing assessment:

    • Monitor pubertal onset markers (vaginal opening, first estrus in females; preputial separation in males)

    • Track growth curves and body weight development

    • Record timing of sexual maturation markers

  • Reproductive axis evaluation:

    • Measure gonadotropin levels (LH, FSH)

    • Assess sex steroid production (testosterone, estradiol)

    • Evaluate fertility and fecundity

  • Neuroanatomical analysis:

    • Immunohistochemistry for GnRH neurons in the hypothalamus

    • Tract tracing to assess migration paths

    • Quantification of GnRH neuron numbers and distribution

The following table outlines key phenotypic parameters to assess in IGSF10 mutant models, based on human studies:

ParameterMeasurement MethodExpected Phenotype Based on Human Studies
Puberty onsetPhysical examinationDelayed (similar to human DP phenotype)
Bone ageRadiographic examinationDelayed relative to chronological age
Hormone levelsELISA/RIADelayed rise in gonadotropins and sex steroids
GnRH neuron migrationImmunohistochemistryAltered distribution pattern
Body compositionDEXA scanPotential impact on BMI (as observed in humans)

How does rat IGSF10 compare functionally to human IGSF10?

Designing comparative studies between rat and human IGSF10 requires:

  • Sequence and structure comparison:

    • Perform multiple sequence alignment to identify conserved regions

    • Compare protein domain organization and key functional motifs

    • Analyze conservation of known mutation sites

  • Cross-species functional assays:

    • Test rat IGSF10 functionality in human cell lines

    • Assess human IGSF10 in rat primary cultures

    • Measure species-specific interaction partners

  • Complementation studies:

    • Determine if rat IGSF10 can rescue phenotypes in human cells with IGSF10 mutations

    • Test if human IGSF10 variants affect rat GnRH neuronal migration

Human IGSF10 mutations have been identified in individuals with delayed puberty and hypothalamic amenorrhea, with four key variants (p.Arg156Leu, p.Glu161Lys, p.Glu2264Gly, and p.Asp2614Asn) showing significant clinical associations . Comparative studies should focus on these regions to determine conservation of function across species.

What experimental approaches help translate rat IGSF10 findings to human applications?

Translational research requires methodological rigor:

  • Parallel experimentation:

    • Conduct identical experiments in rat and human cells

    • Use matched tissue samples when possible

    • Apply consistent analytical methods across species

  • Validating conservation:

    • Confirm that molecular mechanisms are conserved

    • Verify that phenotypic outcomes are similar

    • Test whether pharmacological interventions have cross-species efficacy

  • Clinical correlation:

    • Associate rat model findings with human patient data

    • Analyze IGSF10 variants in human cohorts with relevant phenotypes

    • Develop biomarkers based on rat studies that can be applied to human diagnostics

Human genetic studies have demonstrated that IGSF10 variants follow an autosomal dominant inheritance pattern with variable penetrance , suggesting that rat models should be designed to investigate haploinsufficiency and dominant-negative effects rather than complete loss-of-function.

How can researchers address contradictory data in IGSF10 functional studies?

Resolving contradictions requires systematic approaches:

  • Methodological standardization:

    • Use consistent experimental conditions across studies

    • Standardize protein preparation and storage protocols

    • Employ the same assay systems for comparative analysis

  • Variable isolation:

    • Test one variable at a time (cell type, species, mutation)

    • Control for post-translational modifications

    • Account for potential splice variants

  • Multi-method verification:

    • Confirm findings using orthogonal techniques

    • Combine in vitro, ex vivo, and in vivo approaches

    • Validate with both gain- and loss-of-function studies

For example, when studying IGSF10's role in cell migration, researchers should combine transwell assays, time-lapse microscopy, and in vivo migration tracking to build a consensus understanding of the protein's function.

What cutting-edge technologies can enhance rat IGSF10 research?

Emerging technologies offer new insights into IGSF10 function:

  • Single-cell technologies:

    • scRNA-seq to identify IGSF10-responsive cell populations

    • Single-cell proteomics to map signaling cascades

    • Spatial transcriptomics to visualize expression patterns in tissue context

  • Advanced imaging:

    • Super-resolution microscopy for subcellular localization

    • Light-sheet microscopy for whole-embryo imaging of migration

    • Intravital imaging for real-time migration in developing rats

  • Organoid and microphysiological systems:

    • Hypothalamic organoids to study GnRH neuron development

    • Microfluidic systems to model migration gradients

    • Organ-on-chip approaches for complex tissue interactions

These technologies can help resolve the complex role of IGSF10 in neuronal migration and potentially uncover new functions beyond those currently described in the literature.

What are essential controls for recombinant rat IGSF10 experiments?

Rigorous controls ensure reliable results:

  • Expression controls:

    • Empty vector transfection

    • Irrelevant protein of similar size and structure

    • Wild-type IGSF10 (when studying mutations)

  • Functional controls:

    • Known regulators of GnRH migration (positive controls)

    • Scrambled siRNA (for knockdown studies)

    • Heat-inactivated IGSF10 (for binding studies)

  • Specificity controls:

    • Pre-absorption of antibodies with recombinant protein

    • Multiple antibodies targeting different epitopes

    • Gene editing validation with multiple guide RNAs

For studying IGSF10 variants, researchers should include both wild-type controls and previously characterized variants with known effects, such as the four variants (p.Arg156Leu, p.Glu161Lys, p.Glu2264Gly, and p.Asp2614Asn) identified in human studies .

How should researchers standardize IGSF10 detection methods?

Standardization approaches include:

  • Antibody validation:

    • Verify specificity using knockout/knockdown controls

    • Test on recombinant protein of known concentration

    • Compare multiple commercial antibodies

  • Expression quantification:

    • Use absolute quantification with standard curves

    • Include multiple reference genes for RT-qPCR

    • Develop standardized protocols for immunohistochemistry scoring

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2025 TheBiotek. All Rights Reserved.