LRIG1 is a 1093-amino acid type I transmembrane protein with extracellular leucine-rich repeats (LRRs) and intracellular signaling domains . In mice, it is expressed in diverse tissues, including:
LRIG1+ Treg cells exhibit enhanced suppressive activity compared to LRIG1− T cells. Mechanistically, LRIG1 modulates TGF-β1-dependent differentiation of induced Treg (iTreg) cells .
In the skin, LRIG1 identifies quiescent stem cells in hair follicle junctional zones. Genetic deletion of Lrig1 causes epidermal hyperplasia and reduced label-retaining cells, highlighting its role in stem cell dormancy .
LRIG1 inhibits BDNF/TrkB signaling, restricting hippocampal dendrite branching. Lrig1−/− mice exhibit:
Increased proximal dendritic complexity (Sholl analysis: higher branch points).
Social interaction deficits, correlating with altered CA1–CA3 circuitry .
During colon development, LRIG1+ cells generate clonal crypts. Inducible Lrig1 knockout (Lrig1-CreERT2/fl) leads to:
Epithelial hyperproliferation (Ki-67+ cells) by P14.
Normal differentiation, suggesting selective control over proliferation .
Lrig1−/− mice develop:
Corneal plaques and neovascularization by 24 months.
Pathological keratinization and stromal inflammation, linked to STAT3-dependent signaling .
LRIG1 physically interacts with RTKs (e.g., Ret, TrkB, ERBBs) to:
Block ligand binding (e.g., GDNF to Ret in DRG neurons).
Reduce receptor phosphorylation (e.g., ERBB2 in cancer models) .
In DRG neurons, LRIG1 and Lrig3 redundantly suppress Ret/GFRα signaling. Lrig1HT/Lrig3KO mice show:
Hypersensitivity to cold (4°C tail-flick test).
LRIG1 opposes oncogenic signaling via:
ERBB inhibition (e.g., ERBB2 in breast cancer).
MYC downregulation in prostate cancer models.
AR (androgen receptor) feedback regulation, restricting castration-resistant prostate cancer .
Enhanced dendrite branching in hippocampal neurons (vs. control) .
Reduced stem cell quiescence in epidermis (flow cytometry: fewer label-retaining cells) .
Overexpression in glioma stem cells (GSCs) reduces colony formation and size .
AR-driven LRIG1 expression inhibits TRAMP prostate tumor growth .
The protein LRIG1, short for Leucine Rich Repeats And Immunoglobulin Like Domains 1, plays a crucial role in regulating the EGFR signaling pathway. LRIG1 acts as a general inhibitor of this pathway. Research indicates that LRIG1 interacts with ErbB proteins and Met kinase, influencing their regulation through mechanisms like increased ubiquitination and degradation within lysosomes. This makes LRIG1 a tumor suppressor, as it controls cell proliferation by negatively regulating tyrosine kinase receptors in the EGF protein family. Notably, LRIG1 expression is often reduced in cancerous cells and serves as a potential indicator for cancer progression.
Produced in Sf9 Baculovirus cells, LRIG1 Mouse is a single, glycosylated polypeptide chain consisting of 770 amino acids (specifically, amino acids 35 to 796). It has a molecular weight of 84.8kDa. The protein is engineered with an 8 amino acid His tag at its C-terminus and undergoes purification using proprietary chromatographic techniques.
LRIG1 protein solution is provided at a concentration of 0.5mg/ml. It is prepared in a buffer containing 10% glycerol and Phosphate Buffered Saline (pH 7.4).
For short-term storage (up to 2-4 weeks), the solution can be stored at 4°C. For extended storage, it is recommended to freeze the solution at -20°C. The addition of a carrier protein like HSA or BSA (0.1%) is advisable for long-term storage to maintain protein stability. It is crucial to avoid repeated freeze-thaw cycles to prevent protein degradation.
Analysis by SDS-PAGE confirms that the purity of the LRIG1 Mouse protein is greater than 90%.
Leucine-rich repeats and immunoglobulin-like domains protein 1, LIG-1, Lrig1, LIG1, D6Bwg0781e, Img
Sf9, Baculovirus cells.
AQAGPRAPCA AACTCAGDSL DCSGRGLATL PRDLPSWTRS LNLSYNRLSE IDSAAFEDLT NLQEVYLNSN ELTAIPSLGA ASIGVVSLFL QHNKILSVDG SQLKSYLSLE VLDLSSNNIT EIRSSCFPNG LRIRELNLAS NRISILESGA FDGLSRSLLT LRLSKNRITQ LPVKAFKLPR LTQLDLNRNR IRLIEGLTFQ GLDSLEVLRL QRNNISRLTD GAFWGLSKMH VLHLEYNSLV EVNSGSLYGL TALHQLHLSN NSISRIQRDG WSFCQKLHEL ILSFNNLTRL DEESLAELSS LSILRLSHNA ISHIAEGAFK GLKSLRVLDL DHNEISGTIE DTSGAFTGLD NLSKLTLFGN KIKSVAKRAF SGLESLEHLN LGENAIRSVQ FDAFAKMKNL KELYISSESF LCDCQLKWLP PWLMGRMLQA FVTATCAHPE SLKGQSIFSV LPDSFVCDDF PKPQIITQPE TTMAVVGKDI RFTCSAASSS SSPMTFAWKK DNEVLANADM ENFAHVRAQD GEVMEYTTIL HLRHVTFGHE GRYQCIITNH FGSTYSHKAR LTVNVLPSFT KIPHDIAIRT GTTARLECAA TGHPNPQIAW QKDGGTDFPA ARERRMHVMP DDDVFFITDV KIDDMGVYSC TAQNSAGSVS ANATLTVLET PSLAVPLEDR VVTVGETVAF QCKATGSPTP RITWLKGGRP LSLTERHHFT PGNQLLVVQN VMIDDAGRYT CEMSNPLGTE RAHSQLSILP TPGCRKDGTT VGVEHHHHHH
LRIG1 primarily functions as a negative regulator of receptor tyrosine kinases, particularly the ErbB family of receptors. In mouse models, LRIG1 has been shown to complex with all four ErbB receptors, promoting their ubiquitination and decreasing their numbers, thus dampening growth factor signaling . Additionally, LRIG1 maintains stem cell quiescence in multiple tissues, including the epidermis and intestine, by regulating pathways involved in proliferation . LRIG1 also forms an autoregulatory feedback loop with c-Myc, where LRIG1 decreases Myc levels, while Myc appears to influence LRIG1 expression .
LRIG1 is widely expressed across multiple mouse tissues. It appears on the surface of prostatic epithelium, endothelial cells, vascular and visceral smooth muscle, mammary epithelium, cardiac muscle, keratinocytes, and neurons . In the epidermis, LRIG1 expression defines a distinct multipotent stem cell population located in the hair follicle junctional zone adjacent to sebaceous glands . LRIG1 is also highly expressed by intestinal stem cells and in hippocampal neurons, where it regulates dendrite morphology .
LRIG1 knockout mice display several distinct phenotypes across different tissues:
Epidermis: LRIG1-null mice show epidermal hyperplasia with thicker tail epidermis, hair follicles that protrude at greater angles, and hyperproliferation (indicated by Keratin 6 expression) .
Stem cell compartments: Loss of LRIG1 decreases the number of quiescent (label-retaining) cells in all epidermal stem cell populations .
Intestine: LRIG1 KO mice show phenotypic changes related to increased ErbB signaling that can be rescued by both pharmacological and genetic modulation of ErbB activity .
Neural development: LRIG1-deficient mice exhibit enhanced primary dendrite formation and proximal dendritic branching of hippocampal neurons, resembling the effects of BDNF on these neurons .
Behavior: LRIG1-mutant mice display altered social behaviors, tending to be isolated from their littermates .
For identifying LRIG1-expressing cells in mouse tissues, researchers should employ multiple complementary techniques:
Immunohistochemistry/Immunofluorescence: Using antibodies such as Goat Anti-Mouse LRIG1 Alexa Fluor® 488-conjugated Antigen Affinity-purified Polyclonal Antibody . This approach allows visualization of LRIG1 expression patterns within tissue architecture.
Flow cytometry: This method enables quantitative analysis of LRIG1 expression in single-cell suspensions. Protocols for staining membrane-associated proteins should be followed, as demonstrated with D3 mouse embryonic stem cell lines .
Reporter mouse models: Utilizing Lrig1-eGFP-ires-CreERT2 knock-in mice allows for identification of LRIG1-expressing cells through GFP fluorescence, enabling live cell tracking and lineage tracing experiments .
RT-qPCR: For quantitative measurement of LRIG1 mRNA expression levels in sorted cell populations or tissue samples, often used to validate findings from other methods .
When analyzing LRIG1-expressing cells, co-staining with other markers (such as Lgr5, Ascl2, and Msi1 for intestinal stem cells) can provide additional context about their identity and functional state .
Generating and validating LRIG1 knockout mouse models requires careful consideration of genetic background and thorough validation:
Genetic background consideration: Previous work with LRIG1 knockout mice was initially conducted on outbred genetic backgrounds. For more consistent results, it's advisable to maintain the line on inbred backgrounds such as FVB/N .
Breeding strategy: Heterozygous breeding (LRIG1+/- × LRIG1+/-) is recommended to generate littermate controls (wild-type, heterozygous, and knockout) for experimental comparisons .
Validation methods:
Genotyping: PCR-based genotyping to confirm the genetic modification
Protein expression: Western blotting to confirm the complete absence of LRIG1 protein in tissues of interest
Phenotypic validation: Examining known phenotypes such as epidermal hyperplasia or altered dendrite morphology
Molecular validation: Assessing increased expression of known LRIG1 targets, such as upregulation of Myc protein and mRNA in the skin of LRIG1-null mice
Rescue experiments: Crossing LRIG1 KO animals with mice carrying mutations that affect downstream pathways (e.g., hypomorphic Egfr wa-2 mice) can confirm specificity of phenotypes through genetic rescue .
To study LRIG1's effects on stem cell quiescence in mouse models, the following protocols are recommended:
BrdU label-retention assays:
Pulse-chase studies using BrdU incorporation are effective for determining the proliferative status of LRIG1-expressing cells
Administer single or repeated intraperitoneal injections of BrdU (10mg/mL, 100μL) every 12 hours for pulse labeling
For chase periods, monitor label retention after defined periods (e.g., 1-week chase) to identify quiescent stem cell populations
Analyze by flow cytometry for specific populations (e.g., CD34 positive, Sca1 negative, α6 high for bulge cells)
Quantification methods:
In vitro colony formation assays:
Compare clonogenic potential of LRIG1-positive versus LRIG1-negative cells
Assess impact of LRIG1 knockout or overexpression on colony-forming efficiency and size
Cell cycle analysis:
Flow cytometric analysis of DNA content and cell cycle markers
Assessment of cell cycle regulators by Western blotting or qPCR
The combination of these approaches provides comprehensive insights into how LRIG1 regulates stem cell quiescence in various tissues .
LRIG1 mouse models offer sophisticated platforms for studying ErbB receptor regulation through several approaches:
Genetic interaction studies:
Cross LRIG1 knockout mice with Egfr wa-2 mice (carrying a missense mutation in the kinase domain of Egfr) to study genetic interactions between LRIG1 and EGFR
Quantify rescue effects of reduced EGFR signaling on LRIG1 knockout phenotypes, which has demonstrated approximately 40% phenotype rescue in heterozygous Egfr wa-2 backgrounds
Pharmacological intervention:
Biochemical analysis:
Perform co-immunoprecipitation studies to analyze LRIG1-ErbB physical interactions in different tissues
Examine receptor ubiquitination, internalization, and degradation rates in the presence or absence of LRIG1
Quantify activation of downstream signaling molecules (phospho-ERK, phospho-AKT) in response to EGF stimulation
Cell-specific analysis:
Use conditional knockout models to ablate LRIG1 in specific cell types expressing various ErbB receptor profiles
Compare the impacts on receptor levels, distribution, and signaling dynamics across different cellular contexts
These approaches collectively enable researchers to dissect the molecular mechanisms by which LRIG1 regulates ErbB receptor function in vivo, providing insights that cannot be obtained from in vitro systems alone .
For investigating LRIG1's role in neural development using mouse models, the following experimental designs are optimal:
Morphological analysis of neuronal architecture:
Molecular pathway analysis:
Investigate LRIG1 interaction with TrkB (BDNF receptor) through:
Co-immunoprecipitation studies to confirm physical interaction
Phosphorylation analysis of TrkB and downstream signaling molecules
Compare BDNF-induced morphological changes in wild-type versus LRIG1-deficient neurons
Perform gain and loss of function assays to determine how LRIG1 restricts BDNF-induced dendrite morphology
Behavioral assessment:
These experimental approaches allow for comprehensive investigation of how LRIG1 influences neural development from molecular interactions to behavioral outcomes, establishing LRIG1 as a critical regulator of neurotrophin-induced dendritic arborization .
For effective lineage tracing of LRIG1-positive stem cells across different tissues, researchers should implement the following comprehensive approach:
Generation of appropriate genetic tools:
Utilize Lrig1-eGFP-ires-CreERT2 knock-in mouse models that enable both visualization of LRIG1-expressing cells (through GFP) and inducible genetic manipulation (through CreERT2)
Cross these mice with reporter lines (e.g., Rosa26-loxP-stop-loxP-tdTomato) to permanently label LRIG1-expressing cells and their progeny
Temporal control of lineage tracing:
Administer tamoxifen at specific developmental timepoints or in adult mice to initiate labeling
For short-term lineage tracing, analyze tissues 1-7 days post-induction
For long-term studies, extend analysis to several months to assess long-term stem cell potential and contribution to tissue maintenance
Tissue-specific considerations:
Epidermis: Assess contribution to interfollicular epidermis, sebaceous glands, and hair follicles separately
Induce retinoic acid stimulation to test the bipotent nature of LRIG1-positive cells in contributing to sebaceous gland and interfollicular epidermis
Perform skin reconstitution assays to test the ability of LRIG1-positive cells to give rise to all adult epidermal lineages
Quantitative analysis:
Calculate the percentage of labeled cells in different compartments over time
Track proliferation rates using co-labeling with BrdU or Ki67
Analyze expression of differentiation markers to assess the fate of LRIG1-positive cell progeny
Perturbation studies:
This methodological framework enables comprehensive analysis of LRIG1-positive stem cell behavior in homeostasis, development, and response to perturbations across different tissue contexts .
When facing conflicting data on LRIG1 function across different tissues, researchers should implement a systematic approach to data reconciliation:
Contextual analysis framework:
Create a comparative table documenting LRIG1 functions across tissues, noting specific molecular interactions, phenotypes, and experimental conditions
Identify tissue-specific expression levels of LRIG1 interaction partners (particularly ErbB family members and TrkB), as interaction dynamics may depend on relative abundance
Consider developmental timing, as LRIG1 functions may vary during embryonic development versus adult homeostasis
Methodological standardization:
Compare experimental methodologies used across studies (e.g., constitutive versus conditional knockout models)
Account for genetic background differences, as LRIG1 knockout phenotypes can vary between outbred and inbred strains
Standardize analytical techniques to ensure comparable measurements across tissue types
Molecular mechanism investigation:
Determine whether LRIG1 interacts with different receptor targets in different tissues (e.g., ErbB in epidermis and intestine versus TrkB in neurons )
Investigate tissue-specific post-translational modifications of LRIG1 that might alter its function
Examine potential cross-talk between LRIG1 and other LRIG family members (e.g., LRIG3 can oppose LRIG1 function)
Validation through complementary approaches:
Confirm findings using both in vivo and in vitro models
Implement rescue experiments with tissue-specific re-expression of LRIG1 in knockout backgrounds
Use pharmacological approaches targeting downstream pathways to validate mechanism-based hypotheses
By systematically addressing these aspects, researchers can develop nuanced models of LRIG1 function that account for tissue-specific variations while identifying conserved regulatory mechanisms .
For robust analysis of morphological changes in LRIG1-deficient tissues, the following statistical approaches are recommended:
Quantitative morphometric analysis:
Epidermal measurements:
Neuronal morphology:
Appropriate statistical tests:
Use unpaired t-tests for simple comparisons between wild-type and knockout samples
Implement ANOVA with post-hoc tests for multi-group comparisons (e.g., wild-type, heterozygous, and knockout)
Apply repeated measures ANOVA for parameters measured across multiple time points or spatial positions
Use non-parametric alternatives (Mann-Whitney, Kruskal-Wallis) when data do not meet normality assumptions
Sample size and power considerations:
Conduct power analysis to determine adequate sample sizes
For dendrite analysis, examine at least 20-30 neurons per condition across multiple animals
For epidermal analyses, sample multiple skin regions from at least 3-5 animals per genotype
Advanced analytical methods:
Apply hierarchical linear modeling to account for nested data structures (e.g., multiple cells within animals)
Use principal component analysis to identify patterns in complex morphological datasets
Implement machine learning approaches for unbiased classification of morphological phenotypes
Visualization techniques:
Generate heatmaps of morphological changes across tissue regions
Create violin or box plots showing distribution of measurements
Use representative images alongside quantitative data to illustrate key findings
Differentiating between direct and indirect effects of LRIG1 deletion in complex phenotypes requires a multi-faceted experimental strategy:
Temporal manipulation studies:
Implement inducible knockout systems (e.g., CreERT2) to delete LRIG1 at specific timepoints
Monitor the sequence of molecular and phenotypic changes, with direct effects typically manifesting earlier than indirect consequences
Compare acute versus chronic LRIG1 deletion to identify adaptive responses
Pathway dissection approaches:
Perform detailed signaling analysis to establish direct molecular targets:
Use small molecule inhibitors to block specific pathways:
Genetic interaction studies:
Cell-autonomous versus non-cell-autonomous effects:
Create mosaic models with focal LRIG1 deletion to determine whether effects are cell-autonomous
Perform tissue-specific knockout studies to identify potential systemic or paracrine effects
Use co-culture systems to assess how LRIG1-deficient cells influence neighboring wild-type cells
Molecular rescue experiments:
Re-express LRIG1 with specific domain mutations to identify which protein regions are essential for phenotype rescue
Implement downstream pathway activation/inhibition to bypass LRIG1 and directly test mechanistic hypotheses
This comprehensive approach enables researchers to construct mechanistic models that distinguish primary effects of LRIG1 loss from secondary consequences, facilitating interpretation of complex phenotypes .
Working with LRIG1 antibodies presents several technical challenges that can be addressed through specific optimization strategies:
Antibody specificity issues:
Challenge: Cross-reactivity with other LRIG family members (LRIG2, LRIG3) due to structural similarities
Solution: Validate antibody specificity using LRIG1 knockout tissues as negative controls
Always include isotype control antibodies in flow cytometry experiments
Use multiple antibodies targeting different LRIG1 epitopes to confirm findings
Detection sensitivity limitations:
Challenge: Low endogenous expression levels in certain tissues
Solution: Implement signal amplification methods such as tyramide signal amplification for immunohistochemistry
Optimize antibody concentration through titration experiments
For flow cytometry, use bright fluorophores like Alexa Fluor 488 for better detection sensitivity
Fixation and sample preparation issues:
Challenge: Epitope masking during fixation, particularly for membrane proteins
Solution: Test multiple fixation protocols (4% PFA, methanol, acetone)
Optimize antigen retrieval methods (heat-induced versus enzymatic)
For flow cytometry of membrane-associated LRIG1, follow specific staining protocols designed for membrane proteins
Storage and handling considerations:
Protocol optimization guidelines:
Start with manufacturer's recommended dilutions and adjust based on signal-to-noise ratio
Include positive control tissues known to express high LRIG1 levels
For challenging applications, consider using LRIG1 reporter mice instead of antibody-based detection
These strategies significantly improve detection reliability and experimental reproducibility when working with LRIG1 antibodies across various applications .
Addressing genetic background influences in LRIG1 mouse studies requires systematic approaches to ensure reproducible and interpretable results:
Background standardization protocol:
Challenge: LRIG1 knockout phenotypes vary between outbred and inbred genetic backgrounds
Solution: Backcross LRIG1 mutant lines to well-characterized inbred strains (e.g., C57BL/6J, FVB/N) for at least 10 generations to achieve >99.9% genetic homogeneity
Document the specific background used in all publications (e.g., FVB/N as used in some LRIG1 studies)
Littermate control implementation:
Background-specific phenotype characterization:
Challenge: Certain phenotypes may be prominent only on specific backgrounds
Solution: Compare LRIG1 knockout phenotypes across multiple inbred backgrounds
Create a standardized phenotyping pipeline to systematically document strain-specific variations
Consider using F1 hybrids between different inbred strains to mitigate strain-specific effects
Genetic quality control measures:
Challenge: Genetic drift in mouse colonies over time
Solution: Implement regular genetic quality control testing
Reestablish colonies from cryopreserved embryos periodically
Consider single nucleotide polymorphism (SNP) panel screening to confirm strain identity
Statistical approaches for background effects:
Challenge: Distinguishing LRIG1-specific effects from background influences
Solution: Include strain background as a variable in statistical analyses
Consider using mixed-effect models when combining data across different backgrounds
Report effect sizes rather than just statistical significance to better gauge biological relevance
By implementing these approaches, researchers can minimize confounding factors related to genetic background and enhance the reproducibility and translational relevance of findings from LRIG1 mouse studies .
To minimize variability in stem cell behavior studies using LRIG1 mouse models, researchers should implement the following comprehensive strategies:
Standardized isolation protocols:
Challenge: Technical variations in stem cell isolation affecting experimental outcomes
Solution: Develop rigorous protocols for isolating LRIG1-positive cells:
Experimental design optimization:
Challenge: Biological variability in stem cell behavior
Solution: Control for key variables:
Age-match animals precisely (within 3-5 days)
Consider sex as a biological variable and analyze males and females separately or ensure balanced groups
Standardize housing conditions, including diet, light cycles, and cage density
Control for circadian effects by performing experiments at consistent times of day
Quantitative assay standardization:
Challenge: Variability in functional assays of stem cell behavior
Solution: Implement rigorous controls for stem cell assays:
Technical replication framework:
Challenge: Distinguishing technical from biological variability
Solution: Implement nested experimental designs:
Technical replicates within biological replicates
Multiple tissue sections per animal
Multiple fields of view per section
Appropriate statistical modeling to account for nested data structure
Reporting standards implementation:
Challenge: Incomplete methods reporting hindering reproducibility
Solution: Adopt comprehensive reporting guidelines:
Document all animal characteristics (age, sex, weight, health status)
Report exclusion criteria and any excluded samples/animals
Provide raw data distributions rather than just means
Share detailed protocols through repositories or supplementary materials
The LRIG1 gene is a protein-coding gene that plays a significant role in various biological processes. The protein encoded by this gene contains multiple leucine-rich repeats and immunoglobulin-like domains, which contribute to its function in cellular signaling and interaction . The LRIG1 protein is a type-I transmembrane protein with 1092 amino acids and a molecular weight of approximately 120 kDa .
LRIG1 acts as a feedback negative regulator of signaling by receptor tyrosine kinases. It enhances receptor ubiquitination and accelerates intracellular degradation, thereby modulating signaling pathways that are crucial for cell growth and differentiation . This regulatory function is essential for maintaining cellular homeostasis and preventing uncontrolled cell proliferation.
LRIG1 is ubiquitously expressed in various tissues, including the skin, nervous system, and gastrointestinal tract . It is predominantly located in the plasma membrane but can also be found in the extracellular matrix and extracellular space . The expression of LRIG1 is tightly regulated and can be induced in response to specific cellular signals.
LRIG1 has been identified as a tumor suppressor gene in various types of cancer. It is highly expressed in tumor-infiltrating lymphocytes (TILs) and may play a role in modulating T cell activation . The expression of LRIG1 in TILs suggests that it could be involved in regulating the anti-tumor immune response, making it a potential target for cancer immunotherapy .
The study of LRIG1 has significant implications for understanding cancer biology and developing therapeutic strategies. By elucidating the mechanisms by which LRIG1 regulates receptor tyrosine kinase signaling and immune responses, researchers can identify new targets for cancer treatment and improve existing therapies .