LMO7 antibodies are designed to detect the LMO7 protein, which contains LIM domains that mediate protein-protein interactions. These antibodies are available in both monoclonal (e.g., Santa Cruz’s B-7) and polyclonal forms (e.g., Thermo Fisher’s PA5-54281, Novus Biologicals’ NBP1-83915) and are validated for techniques such as Western blotting (WB), immunohistochemistry (IHC), and immunofluorescence (IF).
Monoclonal Antibodies: High specificity, often targeting distinct epitopes (e.g., amino acids 1321–1620 for Santa Cruz’s B-7) .
Polyclonal Antibodies: Broader epitope recognition, suitable for detecting denatured or modified proteins (e.g., Thermo Fisher’s PA5-54281) .
Applications:
WB: Detects LMO7 in lysates.
IHC: Localizes LMO7 in paraffin-embedded tissues.
IF: Visualizes subcellular distribution.
LMO7 has emerged as a critical oncogene in pancreatic cancer (PC) and other malignancies.
Overexpression: LMO7 is elevated in pancreatic ductal adenocarcinoma (PDAC) and pancreatic neuroendocrine tumors (PNETs), correlating with tumor progression and metastasis .
Mechanisms:
Cell Cycle Arrest and Apoptosis: Knockdown/knockout of LMO7 in PC cells induces G1/S phase arrest and apoptosis, reducing tumor growth and metastasis in vivo .
Epithelial-Mesenchymal Transition (EMT): LMO7 regulates transcription factors (e.g., Snail, ZEB1) that drive EMT, enhancing invasion and migration .
Immune Evasion: LMO7 promotes TGF-β and CCL5 secretion, enriching regulatory T cells (Tregs) and suppressing CD8+ T cells and natural killer (NK) cells in the tumor microenvironment .
Lung Cancer: LMO7 deficiency in mice increases susceptibility to lung cancer, suggesting context-dependent roles .
Prognostic Biomarker: Elevated LMO7 expression in PC is linked to poor survival and aggressive phenotypes .
Preclinical Studies: Inhibiting LMO7 in PC models reduces tumor burden and metastasis, suggesting potential as a therapeutic target .
Immune Modulation: LMO7 blockade may restore anti-tumor immunity by reducing TGF-β and CCL5, enhancing CD8+ T cell infiltration .
LMO7 is a multifunctional PDZ-LIM protein that interacts with various molecular partners and is found in several intracellular locations. The LMO7 gene is located on chromosome 13q14.11 in humans, encoding a protein that contains multiple domains including LIM domains (protein-protein interaction motifs), PDZ domains (interaction modules that bind to specific protein sequences), and CH domains (actin-binding motifs) . LMO7 has garnered significant research interest due to its involvement in diverse physiological processes including neuronal development, cardiovascular health, and cancer pathogenesis . For researchers, LMO7 represents an important target for studying signaling pathways related to cell growth, differentiation, cytoskeletal organization, and particularly cancer development.
LMO7 protein is predominantly expressed in cardiac and skeletal muscle tissues . Immunohistochemistry experiments have successfully detected positive LMO7 expression in mouse heart tissue and mouse skeletal muscle tissue . Additionally, Western blot analysis has confirmed LMO7 expression in A549 cells, HeLa cells, and mouse lung tissue . In vascular tissue, LMO7 is expressed at modest levels in uninjured vessels but becomes highly induced following injury, with expression peaking around day 10 post-injury while remaining elevated through 28 days . This tissue-specific expression pattern is important to consider when designing experiments and selecting appropriate positive control tissues for antibody validation.
The calculated molecular weight of LMO7 protein is 193 kDa, but the observed molecular weight in experimental contexts typically ranges from 140-160 kDa . This discrepancy between calculated and observed molecular weights can be attributed to several factors:
Post-translational modifications (phosphorylation, glycosylation, etc.)
Alternative splicing generating different isoforms
Proteolytic processing of the full-length protein
The presence of intrinsically disordered regions affecting migration patterns
Human LMO7 has at least 27 different predicted transcripts, nine of which are non-coding, while the others produce protein variants with potentially different molecular weights . Researchers should be aware of these variations when interpreting Western blot results and may need to validate which isoform they are detecting in their experimental system.
Based on validated antibody data, LMO7 antibodies can be successfully applied in several experimental contexts:
Application | Recommended Dilution | Notes |
---|---|---|
Western Blot (WB) | 1:2000-1:10000 | Effective for detecting LMO7 in A549 cells, HeLa cells, mouse lung tissue |
Immunohistochemistry (IHC) | 1:50-1:500 | Optimal results in mouse heart tissue, mouse skeletal muscle tissue |
ELISA | Application-dependent | Titration recommended for optimal results |
For immunohistochemistry applications, antigen retrieval with TE buffer pH 9.0 is suggested, though citrate buffer pH 6.0 may serve as an alternative . It is strongly recommended that researchers titrate the antibody in their specific testing system and sample type to obtain optimal results, as antibody performance can be sample-dependent.
When optimizing Western blot protocols for LMO7 detection, consider the following methodological approaches:
Sample preparation: Use strong lysis buffers (RIPA buffer with protease inhibitors) to ensure complete extraction of membrane-associated and nuclear LMO7 protein.
Gel percentage: Due to the high molecular weight of LMO7 (observed at 140-160 kDa), use lower percentage gels (6-8% acrylamide) to achieve better separation of high molecular weight proteins.
Transfer conditions: Employ longer transfer times (overnight at low voltage or 2+ hours at higher voltage) with added SDS in the transfer buffer (0.1%) to facilitate transfer of large proteins.
Blocking and antibody incubation:
Use 5% non-fat dry milk or BSA in TBST for blocking (1-2 hours at room temperature)
Incubate with primary antibody overnight at 4°C (using the recommended dilution of 1:2000-1:10000)
Wash thoroughly (4-5 times for 5-10 minutes each) before secondary antibody incubation
Controls: Include positive controls from tissues known to express LMO7 (heart, skeletal muscle, or lung tissue) .
Expected bands: Be prepared to observe bands between 140-160 kDa, and potentially additional bands representing alternative isoforms .
When troubleshooting, remember that due to LMO7's large size and potential post-translational modifications, transfer efficiency and detection sensitivity may require optimization.
For optimal immunohistochemical detection of LMO7:
Fixation: Use 10% neutral buffered formalin fixation for 24-48 hours, avoiding over-fixation.
Antigen retrieval: Heat-induced epitope retrieval with TE buffer pH 9.0 is recommended as the primary method, with citrate buffer pH 6.0 as an alternative .
Antibody dilution: Start with a mid-range dilution (1:200) from the recommended range (1:50-1:500) and adjust based on signal intensity and background.
Detection system: Use a high-sensitivity detection system, especially for tissues with lower LMO7 expression levels.
Positive control tissues: Include mouse heart tissue and skeletal muscle tissue as positive controls to validate staining specificity .
Negative controls: Include primary antibody omission controls and, if possible, tissues from LMO7 knockout models.
When analyzing results, pay attention to the expected subcellular localization patterns, as LMO7 can be found in multiple cellular compartments depending on the cell type and condition being studied.
LMO7 expression shows variability across cancer types, functioning as both a tumor suppressor and potential oncogene depending on the cancer context:
Upregulation in cancers:
Downregulation in cancers:
Prognostic significance:
Researchers studying LMO7 in cancer contexts should consider this tissue-specific and context-dependent expression pattern when designing experiments and interpreting results. Comparing expression across multiple cancer types using the same detection methods is recommended to understand these differential patterns.
LMO7 influences cancer progression through several key mechanisms:
Epithelial-Mesenchymal Transition (EMT) regulation:
LMO7 promotes EMT by regulating transcription factors such as snail, slug, and ZEB1
This regulation facilitates the loss of cell-cell adhesion and acquisition of invasive properties by cancer cells
Through activation of these transcription factors, LMO7 enables reprogramming of target genes involved in EMT
TGF-β signaling modulation:
LMO7 functions as a negative feedback regulator of TGF-β signaling
Loss of LMO7 enhances TGF-β signaling by upregulating TGF-β1 mRNA, TGF-β protein, and downstream effectors including Smad3 phosphorylation
Mechanistically, LMO7's LIM domain interacts with transcription factors c-Fos and c-Jun, promoting their degradation and interrupting TGF-β autoinduction
Immune evasion mechanisms:
In pancreatic ductal adenocarcinoma (PDAC), LMO7 drives immune evasion through regulatory T cell (Treg) enrichment
LMO7 promotes Treg cell differentiation and chemotaxis while inhibiting CD8+ T cells and natural killer cell cytotoxicity
Mechanistically, LMO7 binds and promotes the ubiquitination and degradation of Foxp1, which negatively regulates TGF-β and CCL5 expression
Understanding these mechanistic pathways provides potential targets for therapeutic intervention in cancers where LMO7 contributes to progression and immune evasion.
LMO7 genes undergo extensive alternative splicing, generating multiple isoforms that can significantly impact experimental results:
Isoform diversity across species:
Structural and functional differences:
Experimental considerations:
Antibodies may detect multiple isoforms depending on the epitope location
PCR primers should be designed with isoform specificity in mind
When performing functional studies, researchers should verify which isoform(s) they are working with
To address isoform variability, researchers should:
Use isoform-specific primers when possible
Consider epitope locations when selecting antibodies
Validate which isoforms are expressed in their experimental system using sequencing or isoform-specific detection methods
Report which isoforms were studied in publications to improve reproducibility
Given LMO7's role as a multidomain protein that interacts with various partners, several methodological approaches are recommended:
Co-immunoprecipitation (Co-IP):
Use antibodies against LMO7 to pull down protein complexes
Consider using domain-specific antibodies to determine which domains mediate specific interactions
Include appropriate controls (IgG controls, input controls) and validate interaction specificity
Example: Co-IP confirmed interaction between LRIG3 and LMO7, showing co-localization and co-immunoprecipitation
Proximity ligation assays (PLA):
Useful for detecting protein-protein interactions in situ with subcellular resolution
Particularly valuable for LMO7 given its presence in multiple cellular compartments
Domain-based interaction mapping:
Functional validation:
After identifying interactions, validate their functional significance through mutagenesis studies
Target key residues within interaction domains
Assess the effect of mutations on downstream signaling pathways like TGF-β signaling
Computational analysis:
When reporting interaction studies, researchers should specify the isoform used, the cellular context, and validation methods to ensure reproducibility.
Recent research has identified the LMO7-Foxp1-TGF-β/CCL5 signaling axis as a promising therapeutic target, particularly in pancreatic ductal adenocarcinoma (PDAC):
Mechanistic basis for targeting:
LMO7, through its LIM domain, directly binds and promotes the ubiquitination and degradation of Foxp1
Foxp1 negatively regulates TGF-β and CCL5 expression by binding to specific regulatory sites
Elevated TGF-β and CCL5 levels contribute to regulatory T cell enrichment, inducing immune evasion in PDAC
Therapeutic approaches:
Combined treatment with TGF-β/CCL5 neutralizing antibodies along with LMO7 inhibition effectively reverses immune evasion in PDAC
This combined approach activates immune responses and prolonged mouse survival in experimental models
Targeting specific domains (particularly the LIM domain) of LMO7 may disrupt its interaction with Foxp1
Research methodology considerations:
When studying this axis, researchers should monitor:
LMO7 and Foxp1 expression levels
TGF-β and CCL5 production
Regulatory T cell infiltration and function
CD8+ T cell and NK cell cytotoxicity
Translational potential:
This axis represents a novel immunotherapeutic strategy for PDAC, an immunologically "cold" tumor with limited responsiveness to current immunotherapies
The heterogeneity of PDAC necessitates characterizing patient-specific immune microenvironment profiles to identify those who may benefit from targeting this pathway
Researchers exploring this therapeutic axis should employ combinatorial approaches that target multiple aspects of the pathway simultaneously while monitoring immune cell populations and their functional status within the tumor microenvironment.
When faced with weak or absent LMO7 signal in Western blot applications, consider these methodological solutions:
Sample preparation issues:
Protein amount and transfer problems:
Increase total protein amount loaded (start with 50-80 μg for tissues with lower expression)
For high molecular weight proteins like LMO7 (140-160 kDa), use extended transfer times and lower percentage gels
Add 0.1% SDS to transfer buffer to facilitate migration of large proteins
Consider using PVDF membrane instead of nitrocellulose for better protein retention
Antibody-related factors:
Increase primary antibody concentration (try 1:2000 if 1:10000 yields no signal)
Extend primary antibody incubation time (overnight at 4°C)
Verify antibody storage conditions (aliquot to avoid repeated freeze-thaw cycles)
Check antibody expiration date and consider a new lot if necessary
Detection system optimization:
Use more sensitive detection reagents (enhanced ECL substrates)
Increase exposure time during imaging
Consider alternative detection methods (fluorescent secondary antibodies may offer better sensitivity)
Expression level considerations:
Successful detection of LMO7 in immunohistochemistry experiments can be influenced by several key factors:
Tissue preparation and fixation:
Overfixation can mask epitopes; limit fixation to 24-48 hours in 10% neutral buffered formalin
Proper tissue handling and embedding techniques are essential
Section thickness should be consistent (4-5 μm recommended)
Antigen retrieval optimization:
Antibody dilution and incubation:
Start in the middle of the recommended range (1:200) and adjust based on results
Extended incubation times (overnight at 4°C) may improve sensitivity
Use humidity chambers to prevent section drying
Detection system considerations:
Polymer-based detection systems often provide better sensitivity than biotin-based systems
For tissues with lower expression, amplification steps may be necessary
Optimize chromogen development time based on signal intensity
Counterstaining and mounting:
Overstaining with hematoxylin can obscure weak positive signals
Use appropriate mounting media that doesn't interfere with signal preservation
When troubleshooting, always include positive control tissues (mouse heart and skeletal muscle) alongside your experimental samples to confirm that technical aspects of the protocol are working properly .
LMO7's function as a negative feedback regulator of TGF-β signaling presents important methodological considerations for fibrosis research:
Experimental design considerations:
LMO7 expression is induced by TGF-β1 in a concentration- and time-dependent manner, with peak protein expression at 0.5 ng/ml TGF-β1 for up to 48 hours in human coronary artery smooth muscle cells
LMO7 induction follows the initiation of TGF-β signaling (as indicated by p-SMAD3) in vascular injury models
Global or smooth muscle cell-specific LMO7 deletion enhances neointimal formation, TGF-β signaling, ECM deposition, and proliferation in vascular injury models
Methodological approaches:
When studying fibrotic processes, monitor both LMO7 and TGF-β pathway components (TGF-β1, p-SMAD3, CTGF) together
Include time course analyses to capture the temporal relationship between TGF-β signaling activation and LMO7 induction
Consider using LMO7 knockout/knockdown models to assess enhanced fibrotic responses
Technical recommendations:
For in vitro studies, standardize TGF-β1 concentrations (0.5 ng/ml recommended) and exposure times
For immunostaining, use multiple markers (p-SMAD3, TGF-β, CTGF, and LMO7) to comprehensively assess pathway activity
When quantifying results, measure both the extent and intensity of staining/expression
Translational implications:
LMO7's role suggests potential therapeutic targets for fibrotic diseases
Research methodologies should include assessment of whether interventions restore normal feedback regulation of TGF-β signaling
Understanding this negative feedback loop is crucial for interpreting experimental results in fibrosis research, as LMO7 deficiency may exacerbate TGF-β-driven fibrotic responses in multiple tissue contexts.
To effectively investigate LMO7's emerging role in immune modulation, particularly its influence on regulatory T cells and immune evasion in cancer:
Single-cell approaches:
Spatial analysis techniques:
Spatial proteomics and multiplex immunohistochemistry/immunofluorescence are recommended to visualize the relationship between LMO7-expressing tumor cells and infiltrating immune cells
These methods preserve tissue architecture while providing cellular resolution information
Functional immunology assays:
Mechanistic studies:
In vivo models:
Syngeneic mouse models with LMO7 manipulation to study immune infiltration dynamics
Combine with immune checkpoint inhibitors to assess potential synergistic effects
Monitor survival outcomes alongside immune parameter changes
These methodological approaches should be integrated to develop a comprehensive understanding of how LMO7 regulates immune cell function, particularly in the context of cancer immunotherapy research.