LHX2 Antibody

Shipped with Ice Packs
In Stock

Description

Definition and Overview of LHX2 Antibody

LHX2 antibodies are immunological tools designed to detect and study the LIM-homeobox 2 (LHX2) protein, a transcription factor critical for developmental processes, stem cell regulation, and cancer progression . LHX2 contains a LIM domain (cysteine-rich zinc-binding motif) and a homeodomain, enabling DNA binding and transcriptional regulation . Antibodies targeting LHX2 are widely used in research to investigate its expression patterns, molecular interactions, and functional roles in diseases such as cancer, neural development, and tissue regeneration .

Key Domains and Mechanisms

  • LIM domain: Mediates protein-protein interactions and subcellular localization .

  • Homeodomain: Facilitates DNA binding to regulate gene expression .

Biological Roles:

  • Cancer: Promotes tumor growth, metastasis, and immune evasion in breast cancer, nasopharyngeal carcinoma (NPC), and cervical cancer by activating pathways like PI3K/AKT/mTOR and FGF1-FGFR .

  • Neural development: Regulates neural progenitor differentiation and maintains stem cell pluripotency in human embryonic stem cells .

  • Tissue regeneration: Controls hair follicle stem cell activity and wound re-epithelization by modulating Sox9, Tcf4, and Lgr5 expression .

Applications of LHX2 Antibodies in Research

LHX2 antibodies are validated for diverse experimental applications:

ApplicationTechnique DetailsKey Studies
Western Blot (WB)Detects ~44 kDa LHX2 protein in human cell lysates .Breast cancer , NPC
Immunohistochemistry (IHC)Localizes LHX2 in tumor tissues (e.g., breast, cervical cancer) .Cervical cancer
Flow CytometryIdentifies LHX2-positive stem cells in hair follicles and neural progenitors .Stem cell studies
Chromatin Immunoprecipitation (ChIP)Maps LHX2 binding to promoters of FGF1, PAX6, and CER1 .NPC , retinal development

Cancer Biology

  1. Breast Cancer:

    • LHX2 overexpression correlates with poor prognosis, immune infiltration (Th1/Th2 cells), and PI3K/AKT/mTOR activation .

    • Promotes cell proliferation and metastasis while inhibiting apoptosis .

  2. Nasopharyngeal Carcinoma (NPC):

    • LHX2 upregulates FGF1, driving STAT3/ERK/AKT signaling and metastasis .

    • High LHX2 levels predict reduced survival (HR = 2.1, p < 0.01) .

  3. Cervical Cancer:

    • Radiation therapy reduces LHX2 methylation, increasing its pro-metastatic activity .

Developmental Biology

  • Neural differentiation: LHX2 regulates PAX6 and CER1 to direct neural progenitor fate .

  • Hair follicle stem cells: LHX2 maintains stemness by balancing Sox9 (proliferation) and Lgr5 (differentiation) .

Limitations and Future Directions

  • Limitations: Cross-reactivity with other LIM-homeobox proteins remains a concern .

  • Opportunities: Develop isoform-specific antibodies and explore LHX2’s role in immunotherapy resistance .

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Typically, we can ship products within 1-3 business days of receiving your order. Delivery times may vary depending on the shipping method and destination. Please consult your local distributor for specific delivery timeframes.
Synonyms
hLhx2 antibody; Homeobox protein LH-2 antibody; Homeobox protein LH2 antibody; LH2 antibody; LHX2 antibody; LHX2_HUMAN antibody; LIM homeobox 2 antibody; LIM homeobox protein 2 antibody; LIM HOX gene 2 antibody; LIM/homeobox protein Lhx2 antibody
Target Names
LHX2
Uniprot No.

Target Background

Function
LHX2 functions as a transcriptional activator, stimulating the promoter of the alpha-glycoprotein gene. This transcriptional regulatory protein plays a crucial role in controlling cell differentiation during the development of lymphoid and neural cell types.
Gene References Into Functions
  1. Research suggests that LHX2 can inhibit the proliferation of T-ALL-derived cells through both LMO2-dependent and -independent mechanisms. This finding proposes LHX2 as a potential new molecular tool for the development of anti-T-ALL drugs. PMID: 29278703
  2. Studies demonstrate that miR-1238 inhibits the proliferation of NSCLC cells, at least partially, through the repression of LHX2. This sheds light on the mechanistic interaction between miR-1238 and LHX2 in NSCLC carcinogenesis. PMID: 26189214
  3. Results indicate that LHX2 is overexpressed in pancreatic ductal adenocarcinoma cells and contributes to promoting cancer cell proliferation and enhanced tumor development. PMID: 25324171
  4. Lhx2 directly regulates tanycyte specification and differentiation in the hypothalamus. PMID: 25505333
  5. This study demonstrates that LHx2 regulates the development of the forebrain hem system. PMID: 23307637
  6. Data indicate a dual role for Lhx2 during EMT and tumor progression. By inducing the expression of PDGF-B, Lhx2 triggers an autocrine PDGF-B/PDGFRbeta loop necessary for cell migration, invasion, and metastatic dissemination. PMID: 24423492
  7. The findings suggest that LHX2 regulates the transcription of downstream intrinsic and extrinsic molecules that are essential for early neural differentiation. PMID: 23804753
  8. These results suggest that if LHX2 is involved in pituitary hormone deficiency associated with posterior pituitary and ocular defects, it would be a rare cause of this condition. PMID: 22535646
  9. Mutations in LHX2 are not a frequent cause of micro/anophthalmia. PMID: 21203406

Show More

Hide All

Database Links

HGNC: 6594

OMIM: 603759

KEGG: hsa:9355

STRING: 9606.ENSP00000362717

UniGene: Hs.696425

Subcellular Location
Nucleus.

Q&A

What are the primary validated applications for LHX2 antibodies?

LHX2 antibodies have been validated for multiple research applications, with varying degrees of optimization depending on the specific clone. The most commonly validated applications include:

  • Western Blotting (WB): Most commercially available LHX2 antibodies are validated for WB, typically at dilutions ranging from 1:500-1:2000 .

  • Immunohistochemistry (IHC): Both paraffin-embedded (IHC-P) and frozen sections (IHC-Fr) methodologies have been validated .

  • Immunofluorescence (IF): Multiple clones demonstrate reliable nuclear labeling of LHX2 protein .

  • Chromatin Immunoprecipitation (ChIP): Several antibodies are validated for ChIP applications, including ChIP-seq for genome-wide binding studies .

  • Immunoprecipitation (IP): Select antibodies have been verified for pulling down native LHX2 protein complexes .

For optimal results, researchers should select antibodies specifically validated for their intended application, as performance can vary significantly between techniques.

What is the expected molecular weight of LHX2 in Western blotting?

LHX2 is typically detected at 44-55 kDa in Western blot applications, though the exact position may vary depending on post-translational modifications and the specific tissue being analyzed:

  • Calculated molecular weight: 44 kDa (based on the 406 amino acid sequence)

  • Observed molecular weight: 50-55 kDa in most cell and tissue lysates

This discrepancy between calculated and observed molecular weight is likely due to post-translational modifications. When conducting Western blot analysis of LHX2, it is advisable to include positive control lysates from tissues known to express LHX2, such as cerebral cortex samples or neural progenitor cells .

What fixation and antigen retrieval methods are recommended for LHX2 immunohistochemistry?

Optimal detection of LHX2 in fixed tissues requires careful consideration of fixation and antigen retrieval protocols:

  • Fixation: 4% paraformaldehyde (PFA) for 30 minutes to 3 hours is commonly used for tissue sections .

  • Paraffin sections: Heat-mediated antigen retrieval with Tris/EDTA buffer pH 9.0 is recommended before commencing with IHC staining protocols .

  • For cultured cells: Fixation in 0.1% paraformaldehyde-PBS for 10 minutes on ice, followed by transfer to 90% methanol for 30 minutes on ice has been validated for optimal nuclear staining .

In dual or triple immunostaining protocols, the order of antibody application should be optimized, particularly when combining with BrdU detection .

How should I validate the specificity of an LHX2 antibody for my research?

Proper validation of LHX2 antibodies is crucial to ensure experimental rigor:

  • Positive controls: Use tissues with known LHX2 expression (e.g., olfactory epithelium, cerebral cortex, or neural progenitor cells) .

  • Negative controls: Include secondary antibody-only controls by substituting PBS for primary antibody .

  • Knockdown validation: Compare staining between wild-type samples and those with LHX2 knockdown (e.g., using shLHX2) .

  • Multiple antibody approach: When possible, validate key findings using antibodies from different suppliers or those targeting different epitopes .

  • Western blot verification: Confirm the antibody detects a band of the expected molecular weight (44-55 kDa) .

For ChIP applications specifically, include IgG controls and validate enrichment at known LHX2 binding sites through ChIP-qPCR before proceeding to genome-wide analyses .

What controls should I include when studying LHX2 in differentiation models?

When investigating LHX2's role in differentiation processes, particularly neural differentiation of stem cells, the following controls are essential:

  • Time-course controls: Include multiple time points to capture dynamic changes in LHX2 expression during differentiation .

  • Expression modulation controls:

    • Gain-of-function: Use doxycycline-inducible LHX2 overexpression (iLHX2) with and without doxycycline treatment .

    • Loss-of-function: Compare results using shLHX2 knockdown versus shLuc (luciferase) control .

  • Downstream target validation: Assess the expression of known LHX2 targets (e.g., PAX6, SOX1, CER1) to confirm functional activity .

  • Pluripotency markers: Monitor expression of pluripotency genes like NANOG to confirm differentiation status .

  • Cell-type specific markers: Include markers for specific lineages (e.g., NESTIN, FOXG1 for neural lineage) to validate differentiation trajectory .

These controls help distinguish between direct effects of LHX2 modulation and secondary consequences during complex differentiation processes.

How can I design experiments to investigate LHX2's transcriptional targets?

To study LHX2's role as a transcriptional regulator:

  • Chromatin occupancy analysis:

    • ChIP-seq can identify genome-wide binding sites of LHX2 using validated antibodies .

    • ChIP-qPCR can validate binding to specific regulatory regions of candidate target genes .

  • Target gene identification:

    • Combine ChIP data with transcriptome analysis (RNA-seq or microarray) after LHX2 modulation .

    • Focus on genes showing both LHX2 binding and expression changes after LHX2 overexpression or knockdown .

  • Functional validation:

    • Reporter assays with wild-type and mutated LHX2 binding sites can confirm direct regulation .

    • For targets like PAX6 and CER1, knockdown experiments can determine dependence on LHX2 .

  • Binding site analysis:

    • Identify conserved regions containing putative LHX2 binding sites through comparative genomics .

    • Use tools like VISTA browser to identify evolutionarily constrained regions .

This multi-faceted approach has successfully identified direct LHX2 targets such as SOX9, TCF4, and LGR5 in hair follicle stem cells, and PAX6 and CER1 in embryonic stem cells .

How do I troubleshoot inconsistent LHX2 detection in Western blot experiments?

Researchers experiencing variability in LHX2 Western blot results should consider:

  • Sample preparation:

    • Nuclear extraction may be necessary as LHX2 is a nuclear transcription factor .

    • Avoid repeated freeze-thaw cycles of samples .

  • Loading controls:

    • Use nuclear-specific loading controls when working with nuclear extracts.

    • β-Actin has been validated as a loading control in some LHX2 Western blot protocols .

  • Antibody optimization:

    • Titrate antibody concentration (recommended ranges: 1:500-1:2000) .

    • For monoclonal antibodies like LHX2A12G1 (sc-81311), 1:200-1:1000 dilutions have been validated .

    • For polyclonal antibodies like AB5756 (Chemicon), 1:2000 dilution has been effective .

  • Detection systems:

    • Enhanced chemiluminescence systems may provide improved sensitivity.

    • Consider using m-IgG Fc BP-HRP bundles for cleaner results with mouse monoclonal antibodies .

  • Blocking optimization:

    • 2% normal donkey serum in PBS has been effective for blocking non-specific binding .

    • 5% non-fat milk or BSA in TBST may be more appropriate for Western blot applications.

If weak signals persist, consider using antibodies targeting different epitopes or confirm protein expression at the mRNA level first .

What factors affect LHX2 detection in different neural populations?

LHX2 detection in neural tissues requires special considerations:

  • Developmental timing:

    • LHX2 expression is dynamically regulated during development, particularly in the telencephalon and olfactory epithelium .

    • Precise embryonic staging is crucial for reproducible results (e.g., E14 mouse embryos show strong expression) .

  • Cell type specificity:

    • LHX2 is expressed in specific neural progenitor populations and may be absent from differentiated neurons .

    • Co-staining with markers like NESTIN, PAX6, or SOX1 can help identify LHX2-expressing neural progenitors .

  • Subcellular localization:

    • Nuclear localization is expected and can be confirmed with DAPI co-staining .

    • Cytoplasmic detection may indicate antibody non-specificity or protein mislocalization in disease states.

  • Signal amplification:

    • Tyramide signal amplification may be necessary for detecting low-abundance expression.

    • Alexa Fluor secondary antibodies (Alexa488, Alexa594) have been validated for LHX2 immunofluorescence .

Studies examining LHX2's role in neural differentiation have successfully used combinations of these approaches to characterize its expression and function in specific neural populations .

How do I optimize dual immunostaining protocols involving LHX2 antibodies?

For co-localization studies with LHX2 and other markers:

  • Antibody compatibility:

    • Select primary antibodies raised in different host species (e.g., rabbit anti-LHX2 with mouse anti-PAX6) .

    • If using same-species antibodies, consider directly conjugated antibodies or sequential staining protocols.

  • Validated antibody combinations:

    • Rabbit anti-LHX2 (AB5756, Chemicon) with Mouse anti-PAX6 (Hybridoma bank) at 1:200 and 1:50 dilution respectively .

    • Rabbit anti-LHX2 with Mouse anti-NESTIN (AB5326, Chemicon) at 1:100 dilution .

    • Rabbit anti-LHX2 with Mouse anti-Tuj1 (MAB1637, Chemicon) at 1:200 dilution .

  • Secondary antibody selection:

    • Use highly cross-adsorbed secondary antibodies to minimize cross-reactivity.

    • Validated combinations include Alexa594-donkey anti-mouse with Alexa488-donkey anti-rabbit .

  • Protocol optimization:

    • Sequential staining may be necessary for certain combinations.

    • For triple staining (e.g., LHX2, L1, and BrdU), process first for LHX2 and L1, then fix in 4% PFA before BrdU staining .

This approach has been successfully employed to demonstrate co-expression of LHX2 with stem cell markers like PAX6 and SOX9 in various developmental contexts .

How can LHX2 antibodies be utilized to study stem cell dynamics in tissue regeneration?

LHX2 antibodies have proven valuable for investigating stem cell activity during tissue regeneration, particularly in skin:

  • Wound healing studies:

    • LHX2-positive cells represent the majority of proliferating cells in hair follicle bulge and secondary hair germ that respond to skin injury .

    • Immunostaining for LHX2 combined with BrdU or Ki67 can identify activated stem cells during wound repair .

  • Hair follicle cycling:

    • LHX2 differentially regulates stem cell markers SOX9, TCF4, and LGR5 in hair follicle stem cells .

    • Antibody detection can track changes in LHX2-expressing populations during the transition from telogen to anagen phases .

  • Lineage regulation:

    • LHX2 promotes wound re-epithelialization via positive regulation of SOX9 and TCF4 in bulge cells .

    • LHX2 simultaneously inhibits hair follicle cycling through negative regulation of LGR5 in the secondary hair germ .

  • Genetic model validation:

    • Compare LHX2 antibody staining patterns between wild-type and heterozygous LHX2 knockout mice to correlate with phenotypic differences in wound healing rates .

These applications demonstrate how LHX2 antibodies can help elucidate the molecular mechanisms controlling tissue-specific stem cell activation during regenerative processes.

What approaches can be used to study LHX2's role in neural development?

To investigate LHX2's functions in neural development:

  • Neural differentiation models:

    • Track LHX2 expression during embryonic stem cell differentiation toward neural lineages .

    • Combined with PAX6 staining, LHX2 antibodies can identify early neural rosette structures .

  • Axon guidance studies:

    • LHX2 regulates thalamocortical axonal guidance through modulation of Robo1 and Robo2 receptors .

    • Co-staining with axonal markers can elucidate LHX2's role in circuit formation .

  • Regional patterning analysis:

    • LHX2 is critical for telencephalon patterning and cortical development .

    • Antibody staining in brain sections can reveal regional expression domains .

  • Transcriptional regulation:

    • ChIP using LHX2 antibodies followed by qPCR for genes like PAX6, SOX1, and FOXG1 can reveal direct regulatory relationships .

    • Combined with reporter assays, this approach has identified CER1 as a direct LHX2 target in neural precursors .

  • Experimental manipulation:

    • Compare staining patterns in gain-of-function (iLHX2 + doxycycline) and loss-of-function (shLHX2) models .

    • This approach revealed that LHX2 overexpression promotes neural differentiation of human embryonic stem cells .

These methodologies have established LHX2 as a key regulator of neural development, particularly in controlling the transition from pluripotent to neural fates.

What are the key differences between available LHX2 antibody clones?

Researchers should consider several factors when selecting among available LHX2 antibody clones:

Clone/Catalog NumberHost/TypeImmunogenValidated ApplicationsReactivityNotable Features
EPR20449 (ab184337)Rabbit monoclonalRecombinantIP, WB, IHC-Fr, IHC-PHuman, Mouse, RatCited in 12 publications
LHX2A12G1 (sc-81311)Mouse monoclonal IgG1Human originWB, IP, IFHumanDetects LHX2 of human origin
6G2 (sc-517243)Mouse monoclonal IgG1 κNot specifiedWB, IP, IF, ELISAMouse, Rat, HumanAvailable in multiple conjugated forms
18133-1-APRabbit polyclonalLHX2 fusion proteinWB, ELISAHuman, Mouse, RatObserved at 50-55 kDa
CL6137 (ab243030)Mouse monoclonalSynthetic peptide (aa 200-250)IHC-P, WBMouse, HumanCited in 3 publications
AB5756 (Chemicon)Rabbit polyclonalNot specifiedWB, IF, ChIPHumanUsed at 1:2000 for WB, 1:200 for IF

The choice between monoclonal and polyclonal antibodies depends on specific research requirements:

  • Monoclonal antibodies offer higher specificity for a single epitope but may be more sensitive to epitope masking.

  • Polyclonal antibodies recognize multiple epitopes, potentially providing stronger signals but with increased background risk .

How should researchers interpret varying molecular weights of LHX2 in experimental results?

Variations in the detected molecular weight of LHX2 may reflect important biological information:

  • Expected range:

    • Calculated molecular weight: 44 kDa based on the amino acid sequence

    • Observed range: 50-55 kDa in most cell and tissue lysates

  • Potential explanations for variations:

    • Post-translational modifications (phosphorylation, SUMOylation, etc.)

    • Alternative splicing variants

    • Cell type-specific processing

    • Experimental conditions (reducing vs. non-reducing, denaturing conditions)

  • Validation approaches:

    • Use multiple antibodies targeting different epitopes to confirm specificity

    • Include positive control lysates from tissues known to express LHX2 (cerebral cortex, neural progenitors)

    • Consider phosphatase treatment to determine if phosphorylation contributes to mobility shifts

  • Documentation:

    • Always report both predicted and observed molecular weights

    • Note the specific tissue/cell type and extraction method used

    • Document the specific antibody clone and detection system employed

These considerations can help researchers properly interpret LHX2 Western blot results and potentially gain additional insights into the protein's regulation in different cellular contexts.

How can LHX2 antibodies contribute to our understanding of disease mechanisms?

LHX2 antibodies are increasingly valuable for investigating disease mechanisms:

  • Cancer research:

    • LHX2 promotes growth and metastasis of nasopharyngeal carcinoma by regulating Wnt signaling .

    • LHX2 plays a vital role in breast cancer progression and prognosis .

    • Immunohistochemical analysis with LHX2 antibodies could help identify patient subgroups and correlate with prognosis.

  • Neurodevelopmental disorders:

    • Given LHX2's critical role in telencephalon patterning and cortical development , antibodies can help investigate developmental abnormalities.

    • Analysis of LHX2 expression patterns in model systems of neurodevelopmental disorders may reveal pathogenic mechanisms.

  • Regenerative medicine:

    • LHX2's role in regulating stem cell populations during wound healing suggests potential applications in developing regenerative therapies.

    • Antibodies can help track transplanted cells and monitor their differentiation status.

  • Drug development:

    • Identifying compounds that modulate LHX2 expression or function could lead to novel therapeutic approaches.

    • LHX2 antibodies would be essential tools for validating drug effects in preclinical models.

The continued development and validation of high-specificity LHX2 antibodies will be crucial for advancing these research areas.

What methodological advances might improve LHX2 antibody applications?

Several emerging technologies and approaches could enhance LHX2 antibody applications:

  • Single-cell protein analysis:

    • Adapting LHX2 antibodies for mass cytometry (CyTOF) or single-cell Western blotting could reveal population heterogeneity.

    • These approaches would be particularly valuable for studying mixed neural progenitor populations.

  • Spatial transcriptomics integration:

    • Combining LHX2 immunostaining with spatial transcriptomics could correlate protein expression with transcriptional states at single-cell resolution.

    • This integration would provide insights into LHX2's context-specific functions in development and disease.

  • Live-cell imaging:

    • Development of non-disruptive labeling methods, such as nanobodies derived from existing LHX2 antibodies, could enable live tracking of LHX2 dynamics.

    • This approach would be valuable for studying real-time regulation during differentiation processes.

  • Proximity labeling:

    • Adapting LHX2 antibodies for proximity labeling techniques (BioID, APEX) could identify novel protein interaction partners in different cellular contexts.

    • This would expand our understanding of LHX2's function beyond its direct transcriptional targets.

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.