LUC7L2 Antibody

Shipped with Ice Packs
In Stock

Description

What is LUC7L2 Antibody?

LUC7L2 antibodies are immunodetection reagents targeting the LUC7-like 2 protein (LUC7L2), a component of the U1 small nuclear ribonucleoprotein (snRNP) complex. These antibodies are validated for techniques such as Western blotting (WB), immunohistochemistry (IHC), immunofluorescence (IF), and enzyme-linked immunosorbent assay (ELISA) .

Innate Immune Regulation

LUC7L2 antibodies have been instrumental in uncovering the protein’s role in suppressing DNA virus-triggered immune responses. Studies show that LUC7L2 binds to intron 3 of MITA/STING pre-mRNA, inhibiting splicing and promoting nonsense-mediated decay. This reduces MITA/STING protein levels, dampening antiviral signaling . Key findings include:

  • LUC7L2-deficient mice exhibit enhanced resistance to HSV-1 infection and reduced viral loads .

  • Loss of LUC7L2 increases MITA/STING expression, amplifying TBK1 and IRF3 phosphorylation .

Cancer Biology

In nasopharyngeal carcinoma (NPC), LUC7L2 antibodies revealed its role in radioresistance:

  • Overexpression: Promotes NPC cell survival post-irradiation .

  • Knockdown: Sensitizes NPC cells to radiation by reducing SQSTM1 (p62) levels and enhancing autophagy .

  • Clinical Relevance: High LUC7L2 expression correlates with shorter survival in NPC patients .

Table 1: Functional Insights from LUC7L2 Studies

Study FocusMethodologyOutcomeSource
Innate ImmunityCRISPR/Cas9 knockoutLUC7L2 deficiency enhances IFN-β/ISG56 transcription in HSV-1 infection
RadioresistanceProteomics/WBLUC7L2 stabilizes SQSTM1, suppressing autophagy in NPC cells
Metabolic ReprogrammingIP-MSLUC7L2 interacts with U1 snRNP to regulate alternative splicing

Validation and Quality Control

LUC7L2 antibodies are rigorously validated:

  • Western Blot: Detects bands at ~46 kDa in human cell lines (e.g., K-562, HeLa) .

  • Immunohistochemistry: Shows nuclear/cytoplasmic localization in NPC tissues .

  • Specificity: Confirmed via knockout cell lines and peptide blocking .

Future Directions

LUC7L2 antibodies will remain critical for exploring:

  • Mechanisms linking RNA splicing to immune evasion in viral infections.

  • Therapeutic targeting of LUC7L2 in radioresistant cancers.

  • Role in autoimmune diseases linked to MITA/STING dysregulation .

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Made-to-order (14-16 weeks)
Synonyms
CGI 59 antibody; CGI 74 antibody; FLJ10657 antibody; hLuc7B2 antibody; LC7L2_HUMAN antibody; LUC7 like 2 (S. cerevisiae) antibody; LUC7B2 antibody; LUC7L2 antibody; Putative RNA binding protein Luc7 like 2 antibody; Putative RNA-binding protein Luc7-like 2 antibody
Target Names
LUC7L2
Uniprot No.

Target Background

Function
LUC7L2 antibody may bind to RNA through its Arg/Ser-rich domain.
Gene References Into Functions
  1. Loss of LUC7L2 has been associated with myeloid neoplasms. PMID: 24429498
Database Links

HGNC: 21608

OMIM: 613056

KEGG: hsa:51631

STRING: 9606.ENSP00000347005

UniGene: Hs.731488

Protein Families
Luc7 family
Subcellular Location
Nucleus speckle. Nucleus, nucleoplasm.

Q&A

What is LUC7L2 and why is it important in research?

LUC7L2 (LUC7-like 2) is an RNA-binding protein that functions as a component of the U1 small nuclear ribonucleoprotein (snRNP) complex involved in pre-mRNA splicing. It has gained significant research attention due to its role in regulating innate immune responses and potential implications in cancer biology. Recent studies have identified LUC7L2 as a negative regulator of DNA virus-triggered innate immune responses, specifically through its interaction with MITA (also known as STING) precursor mRNA . LUC7L2 directly binds to intron 3 of MITA, inhibiting its splicing and promoting nonsense-mediated decay, which results in downregulation of MITA protein levels . This mechanism represents an important feedback regulatory pathway in innate immunity. Additionally, LUC7L2 has been identified as playing a significant role in cancer radioresistance, particularly in nasopharyngeal carcinoma, making it a promising therapeutic target . The multifaceted functions of LUC7L2 in both immune regulation and cancer biology highlight its importance as a research target.

What types of LUC7L2 antibodies are available for research applications?

Currently, researchers have access to several types of LUC7L2 antibodies optimized for different experimental applications. The most common include:

  • Polyclonal antibodies: These include products like the rabbit polyclonal antibody (24202-1-AP) from Proteintech and the CAB13096 antibody from Assay Genie . These antibodies are generated against specific immunogens, such as the LUC7L2 fusion protein Ag21135 or recombinant proteins containing amino acid sequences corresponding to human LUC7L2 .

  • Application-specific antibodies: Depending on the experimental needs, researchers can select antibodies validated for specific applications including:

    • Western Blot (WB): Typically used at dilutions ranging from 1:500 to 1:3000

    • Immunofluorescence (IF)/Immunocytochemistry (ICC): Typically used at dilutions of 1:300 to 1:1200

    • ELISA: Various antibodies are validated for ELISA applications with appropriate dilution ranges

The selection of the appropriate antibody should be based on the specific experimental requirements, target species reactivity (most commonly human and sometimes mouse), and the application method being employed.

What is the molecular weight of LUC7L2 and how does this impact antibody detection?

LUC7L2 has a calculated molecular weight of 47 kDa (392 amino acids) and is typically observed at approximately 46 kDa in experimental conditions . This information is crucial for researchers interpreting Western blot results, as it helps confirm the specificity of antibody binding. When using any LUC7L2 antibody for Western blot applications, researchers should expect to see a band at approximately 46 kDa, which corresponds to the full-length LUC7L2 protein .

It's important to note that post-translational modifications or alternative splicing events may result in slight variations in the observed molecular weight. Additionally, when designing experiments involving protein detection of LUC7L2, researchers should use appropriate molecular weight markers that allow precise determination in the 40-50 kDa range. The consistent observation of LUC7L2 at 46 kDa across different cell lines, including K-562 cells and HeLa cells as reported in validation studies, provides confidence in antibody specificity when the expected band is observed .

How should LUC7L2 antibodies be stored to maintain optimal activity?

For optimal preservation of antibody activity, LUC7L2 antibodies should typically be stored at -20°C, where they remain stable for at least one year after shipment . The standard storage formulation includes PBS with 0.02% sodium azide and 50% glycerol at pH 7.3, which helps maintain antibody stability during freeze-thaw cycles .

For long-term storage considerations:

  • Aliquoting is generally unnecessary for -20°C storage of antibodies in glycerol formulations

  • Some product formulations (20μl sizes) may contain 0.1% BSA, which serves as a stabilizer

  • Avoid repeated freeze-thaw cycles which can lead to protein denaturation and loss of activity

  • Always centrifuge briefly before opening the vial to ensure the solution is at the bottom of the tube

  • Follow manufacturer-specific recommendations, as storage conditions may vary slightly between different antibody products

Proper storage is essential for maintaining antibody performance across multiple experiments and ensuring reproducible results in LUC7L2 detection assays.

How can LUC7L2 antibodies be used to study innate immune responses to DNA viruses?

LUC7L2 antibodies serve as crucial tools for investigating the regulatory role of LUC7L2 in innate immune responses to DNA viruses. Recent research has established that LUC7L2 functions as a negative regulator of DNA virus-triggered immune responses by modulating MITA/STING expression . Researchers can employ several strategic approaches using LUC7L2 antibodies:

  • Monitoring LUC7L2 induction during viral infection: Studies have shown that LUC7L2 is induced at both mRNA and protein levels following HSV-1 infection in monocytic THP-1 cells . By using LUC7L2 antibodies for Western blot analysis at different time points post-infection, researchers can track the temporal dynamics of LUC7L2 expression in response to viral challenge.

  • Analyzing LUC7L2's impact on signaling pathways: Using LUC7L2 antibodies alongside antibodies against phosphorylated MITA, TBK1, and IRF3, researchers can assess how LUC7L2 affects the activation of these downstream signaling molecules. In LUC7L2-deficient cells, phosphorylation of these proteins is markedly enhanced following HSV-1 infection .

  • Comparative analysis between DNA and RNA virus responses: LUC7L2 specifically regulates DNA virus-triggered but not RNA virus-triggered innate immune responses . Antibodies can be used to examine LUC7L2's differential effects on signaling pathways activated by DNA viruses (like HSV-1) versus RNA viruses (like Sendai virus).

  • Cell type-specific expression patterns: LUC7L2 antibodies enable researchers to compare expression patterns across different cell types, including THP-1 cells, human foreskin fibroblasts (HFFs), bone marrow-derived macrophages (BMDMs), and bone marrow-derived dendritic cells (BMDCs), which is important for understanding tissue-specific immune regulation .

What role does LUC7L2 play in cancer radioresistance and how can antibodies help investigate this?

Recent research has identified LUC7L2 as a critical regulator of radioresistance in nasopharyngeal carcinoma (NPC) cells, opening new avenues for targeted radiotherapy enhancement . LUC7L2 antibodies provide several methodological approaches to investigate this phenomenon:

  • Expression correlation studies: Researchers can use LUC7L2 antibodies for immunohistochemistry to analyze expression levels in NPC tissues and correlate findings with patient survival data. Higher LUC7L2 expression has been associated with shorter survival in NPC patients .

  • Mechanistic investigations of radioresistance pathways: Through immunoblotting, researchers can compare LUC7L2 expression levels between radioresistant and radiosensitive NPC cell lines. After manipulation of LUC7L2 expression (overexpression or knockdown), antibodies can be used to confirm successful experimental intervention before assessing changes in cellular response to ionizing radiation .

  • Protein-protein interaction studies: Immunoprecipitation experiments using LUC7L2 antibodies have revealed that SQSTM1 (an autophagy receptor) is a potential binding partner of LUC7L2 . This technique allows researchers to identify novel protein interactions that may contribute to radioresistance mechanisms.

  • Autophagy regulation analysis: Evidence suggests that downregulation of LUC7L2 in NPC-radioresistant cells leads to reduced SQSTM1 expression and enhanced autophagy . Using LUC7L2 antibodies alongside autophagy markers helps elucidate the relationship between LUC7L2 expression, autophagy regulation, and radioresistance.

  • Combinatorial therapy assessment: When studying the effects of LUC7L2 knockdown in combination with autophagy inhibitors like chloroquine (CQ), antibodies provide critical validation of experimental conditions before cell death assays are conducted .

How can LUC7L2 antibodies be utilized to study mRNA splicing mechanisms?

LUC7L2 functions as a component of the U1 snRNP complex involved in pre-mRNA splicing, making LUC7L2 antibodies valuable tools for investigating splicing regulatory mechanisms:

  • RNA immunoprecipitation (RIP) assays: LUC7L2 antibodies can be used to immunoprecipitate LUC7L2-RNA complexes, allowing identification of direct RNA targets. This technique has revealed that LUC7L2 directly binds to intron 3 of MITA precursor mRNA . The protocol typically involves:

    • Crosslinking cells to preserve RNA-protein interactions

    • Cell lysis and immunoprecipitation using LUC7L2 antibodies

    • RNA extraction from immunoprecipitates

    • RT-PCR or RNA-seq analysis to identify bound RNA species

  • Analysis of intron retention events: LUC7L2 has been shown to affect the retention of specific introns in target transcripts. Researchers can use quantitative PCR with primers targeting specific intron-exon junctions to measure the levels of transcripts containing unspliced introns in LUC7L2-manipulated cells compared to controls .

  • Co-immunoprecipitation studies: LUC7L2 antibodies enable investigation of LUC7L2's interactions with other splicing factors and components of the spliceosome, providing insights into the composition of splicing regulatory complexes.

  • Cellular fractionation studies: By separating nuclear and cytoplasmic fractions and performing immunoblotting with LUC7L2 antibodies, researchers can determine the subcellular localization of LUC7L2, which is important for understanding its function in splicing.

  • Alternative splicing analysis: Following manipulation of LUC7L2 expression, researchers can use exon-specific primers and RT-PCR to analyze changes in alternative splicing patterns of target genes, with LUC7L2 antibodies providing confirmation of successful experimental manipulation.

What are the implications of LUC7L2's interactions with MITA/STING for autoimmune disease research?

The discovery that LUC7L2 negatively regulates MITA/STING-mediated innate immune responses has significant implications for autoimmune disease research . LUC7L2 antibodies can be instrumental in exploring these implications through several research approaches:

  • Expression analysis in autoimmune disease models: Researchers can use LUC7L2 antibodies to compare expression levels in tissues from autoimmune disease models versus healthy controls. Altered LUC7L2 expression could suggest dysregulation of MITA/STING-mediated immune responses in autoimmune conditions.

  • Mechanistic studies of cGAS-STING pathway regulation: The cGAS-STING pathway recognizes cytosolic DNA and triggers type I interferon production, a process implicated in several autoimmune diseases. LUC7L2 antibodies allow researchers to investigate how LUC7L2-mediated regulation of MITA/STING affects this pathway in different cell types relevant to autoimmunity.

  • Analysis of feedback regulatory mechanisms: LUC7L2 is induced following DNA virus infection, suggesting a feedback regulatory mechanism . Using LUC7L2 antibodies, researchers can investigate whether similar feedback mechanisms operate in response to self-DNA recognition in autoimmune contexts.

  • Therapeutic target validation: By manipulating LUC7L2 expression in autoimmune disease models and using antibodies to confirm intervention success, researchers can assess whether modulating LUC7L2 activity affects disease progression, potentially validating it as a therapeutic target.

  • Correlation studies in patient samples: Immunohistochemistry or Western blot analysis using LUC7L2 antibodies can help determine whether LUC7L2 expression correlates with disease severity or specific autoimmune disease subtypes in patient samples.

What are the optimal protocols for Western blot detection of LUC7L2?

For reliable and reproducible Western blot detection of LUC7L2, researchers should follow these optimized protocols based on validated antibody performance:

  • Sample preparation:

    • Lyse cells in RIPA buffer containing protease inhibitors

    • Sonicate briefly to shear DNA and reduce sample viscosity

    • Centrifuge at 14,000g for 15 minutes at 4°C to remove debris

    • Determine protein concentration using Bradford or BCA assay

  • Gel electrophoresis and transfer:

    • Load 20-30 μg of total protein per lane

    • Use 10% SDS-PAGE gels for optimal resolution around LUC7L2's 46 kDa size

    • Transfer to PVDF membranes at 100V for 90 minutes in cold transfer buffer

  • Antibody incubation:

    • Block membranes with 5% non-fat milk in TBST for 1 hour at room temperature

    • Incubate with primary LUC7L2 antibody at dilutions of 1:500-1:3000 in blocking buffer overnight at 4°C

    • Wash 3 times with TBST, 10 minutes each

    • Incubate with HRP-conjugated secondary antibody (anti-rabbit IgG) at 1:5000 dilution for 1 hour at room temperature

    • Wash 3 times with TBST, 10 minutes each

  • Detection and analysis:

    • Apply ECL substrate and capture images using a digital imaging system

    • Expected band size: 46 kDa

    • Positive control cell lines: K-562 cells, HeLa cells

  • Validation considerations:

    • Include positive control samples from validated cell lines

    • Consider using LUC7L2 knockdown or knockout samples as negative controls

    • For multiplex analysis, strip and reprobe membranes with antibodies against housekeeping proteins (e.g., GAPDH, β-actin) for loading control

This optimized protocol ensures specific detection of LUC7L2 while minimizing background signal and non-specific binding.

How should immunofluorescence experiments with LUC7L2 antibodies be optimized?

Optimization of immunofluorescence (IF) experiments for LUC7L2 detection requires careful attention to several key parameters:

  • Cell preparation and fixation:

    • Culture cells on glass coverslips in appropriate growth medium

    • Wash cells twice with PBS prior to fixation

    • Fix cells with 4% paraformaldehyde for 15 minutes at room temperature

    • Permeabilize with 0.2% Triton X-100 in PBS for 10 minutes

  • Blocking and antibody incubation:

    • Block with 5% normal goat serum in PBS for 1 hour at room temperature

    • Incubate with primary LUC7L2 antibody at dilutions of 1:300-1:1200 in blocking solution overnight at 4°C

    • Wash 3 times with PBS, 5 minutes each

    • Incubate with fluorophore-conjugated secondary antibody (anti-rabbit) at 1:500 dilution for 1 hour at room temperature in the dark

    • Wash 3 times with PBS, 5 minutes each

    • Counterstain nuclei with DAPI (1 μg/ml) for 5 minutes

    • Mount coverslips with anti-fade mounting medium

  • Imaging parameters:

    • Use confocal microscopy for optimal resolution of nuclear structures

    • Capture images at 63× or 100× magnification for detailed subcellular localization

    • Employ appropriate filter sets for the selected fluorophores

    • Use sequential scanning to avoid bleed-through when performing multi-color imaging

  • Validation considerations:

    • Include positive control cells (A431 cells have been validated for LUC7L2 IF detection)

    • Perform secondary antibody-only controls to assess background fluorescence

    • Consider co-staining with markers of nuclear speckles (e.g., SC35) to confirm localization pattern of LUC7L2

    • For LUC7L2 knockdown validation, perform parallel IF and Western blot analysis

  • Expected localization pattern:

    • LUC7L2 typically shows a predominantly nuclear localization with a speckled pattern characteristic of splicing factors

    • Some diffuse nucleoplasmic staining may also be observed

These optimized protocols ensure specific detection and accurate subcellular localization of LUC7L2 in fixed cells.

What controls are essential when working with LUC7L2 antibodies in experimental design?

Rigorous experimental design requires appropriate controls to validate the specificity and reliability of LUC7L2 antibody applications:

  • Positive controls:

    • Cell lines with confirmed LUC7L2 expression (e.g., K-562 cells, HeLa cells for Western blot; A431 cells for immunofluorescence)

    • Recombinant LUC7L2 protein for antibody validation

    • Tissues with known LUC7L2 expression patterns for immunohistochemistry

  • Negative controls:

    • CRISPR/Cas9-mediated LUC7L2 knockout cell lines (as described in research using THP-1 cells)

    • siRNA or shRNA-mediated LUC7L2 knockdown cells

    • Primary antibody omission controls to assess secondary antibody specificity

    • Isotype controls using non-specific IgG from the same species as the LUC7L2 antibody

  • Expression validation controls:

    • Parallel analysis of LUC7L2 at both protein level (using antibodies) and mRNA level (using qPCR)

    • Multiple antibodies targeting different epitopes of LUC7L2 to confirm specificity

    • Correlation of protein detection with functional assays (e.g., RNA immunoprecipitation for LUC7L2)

  • Treatment controls:

    • Time-course samples following viral infection to demonstrate LUC7L2 induction (as observed with HSV-1 infection)

    • Dose-response samples for ionizing radiation exposure to assess LUC7L2's role in radioresistance

    • Combination treatment controls when studying LUC7L2 in conjunction with other interventions (e.g., autophagy inhibitors)

  • Species-specific considerations:

    • When working with mouse models, ensure the selected antibody has confirmed cross-reactivity with mouse LUC7L2

    • Include wild-type and Luc7l2-deficient mouse samples as definitive controls

Implementing these controls ensures robust and reproducible results when using LUC7L2 antibodies across different experimental contexts.

How can researchers troubleshoot non-specific binding or weak signals when using LUC7L2 antibodies?

When encountering issues with LUC7L2 antibody performance, researchers can employ the following troubleshooting strategies:

  • For non-specific binding in Western blot:

    • Increase blocking stringency (try 5% BSA instead of milk, or increase blocking time)

    • Optimize primary antibody dilution (try a more dilute solution, e.g., 1:2000-1:3000)

    • Increase washing duration and number of washes

    • Add 0.1% Tween-20 to antibody dilution buffer to reduce non-specific interactions

    • Use freshly prepared buffers and reagents

    • Consider using alternative blocking agents (e.g., commercial blocking buffers)

  • For weak signals in Western blot:

    • Increase protein loading (up to 50 μg per lane)

    • Decrease primary antibody dilution (try 1:500)

    • Extend primary antibody incubation time (overnight at 4°C)

    • Use more sensitive detection methods (e.g., enhanced chemiluminescence substrates)

    • Optimize transfer conditions (e.g., longer transfer time for larger proteins)

    • Verify sample preparation procedure to ensure protein integrity

  • For immunofluorescence issues:

    • Optimize fixation conditions (try 4% PFA vs. methanol fixation)

    • Adjust permeabilization (try different detergents or concentrations)

    • Increase primary antibody concentration (use 1:300 dilution)

    • Extend primary antibody incubation time (overnight at 4°C)

    • Try antigen retrieval methods if working with fixed tissues

    • Use amplification systems (e.g., tyramide signal amplification) for weak signals

  • For inconsistent results across experiments:

    • Standardize protein extraction methods

    • Aliquot antibodies to avoid repeated freeze-thaw cycles

    • Maintain consistent incubation times and temperatures

    • Use the same lot number of antibody when possible

    • Include internal controls in each experiment

    • Verify antibody storage conditions (-20°C, with glycerol)

  • For cell/tissue-specific issues:

    • Verify LUC7L2 expression levels in your specific cell type using qPCR

    • Optimize protocols specifically for your cell type or tissue

    • Consider using alternative detection methods (e.g., if Western blot is problematic, try immunofluorescence)

    • For tissues, optimize fixation and processing methods

These troubleshooting approaches address the most common technical challenges encountered when working with LUC7L2 antibodies.

How should experiments be designed to study LUC7L2's role in viral immunity?

Based on current research findings, optimal experimental designs for investigating LUC7L2's role in viral immunity should incorporate multiple complementary approaches:

  • Expression analysis during viral infection:

    • Infect relevant cell types (e.g., THP-1 cells, primary immune cells) with DNA viruses (HSV-1) and RNA viruses (SeV) at different MOIs

    • Harvest cells at multiple time points post-infection (2, 4, 8, 12, 24 hours)

    • Analyze LUC7L2 expression at both mRNA level (qRT-PCR) and protein level (Western blot)

    • Compare induction patterns between DNA and RNA virus infections

  • Loss-of-function studies:

    • Generate LUC7L2 knockout cell lines using CRISPR/Cas9 technology

    • Alternatively, use siRNA or shRNA for transient knockdown

    • Confirm knockdown efficiency by Western blot using LUC7L2 antibodies

    • Challenge control and LUC7L2-deficient cells with viruses or synthetic nucleic acids

    • Measure antiviral gene induction by qRT-PCR (IFNB1, ISG56, IL6)

    • Assess signaling pathway activation by immunoblotting for phosphorylated MITA, TBK1, and IRF3

  • In vivo infection models:

    • Utilize Luc7l2-deficient mice and wild-type littermates

    • Challenge mice with lethal doses of HSV-1

    • Monitor survival rates

    • Measure serum cytokines (IFN-β, IL-6, CXCL10)

    • Assess viral loads in target organs (brain)

    • Analyze immune cell populations in infected tissues

  • Mechanistic investigations:

    • Perform RNA immunoprecipitation (RIP) using LUC7L2 antibodies to identify viral and host RNA targets

    • Analyze intron retention patterns in MITA/STING mRNA using specific primers

    • Quantify MITA/STING protein levels in response to LUC7L2 manipulation

    • Assess nonsense-mediated decay of MITA/STING transcripts in the presence/absence of LUC7L2

  • Translational relevance:

    • Analyze LUC7L2 expression in patient samples with viral infections

    • Correlate expression levels with disease severity or outcomes

    • Investigate potential genetic variants in LUC7L2 that may affect antiviral responses

This comprehensive experimental approach enables thorough investigation of LUC7L2's role in viral immunity from molecular mechanisms to physiological significance.

What methodological approaches are recommended for studying LUC7L2 in cancer radioresistance?

Research into LUC7L2's role in cancer radioresistance requires specialized experimental approaches:

  • Expression profiling in cancer tissues:

    • Analyze LUC7L2 expression in tumor samples versus adjacent normal tissues using immunohistochemistry with validated LUC7L2 antibodies

    • Correlate expression levels with patient survival data and treatment outcomes

    • Compare expression across cancer types to identify those where LUC7L2 may be most relevant

  • Radiation response assays:

    • Generate stable LUC7L2 overexpression and knockdown in cancer cell lines

    • Confirm altered expression using Western blot with LUC7L2 antibodies

    • Expose cells to varying doses of ionizing radiation (0-10 Gy)

    • Assess cell viability using colony formation assays, which are the gold standard for measuring radioresistance

    • Perform cell cycle analysis to determine effects on radiation-induced cell cycle arrest

    • Measure DNA damage and repair using γH2AX foci formation assays

  • Molecular mechanism investigations:

    • Perform immunoprecipitation with LUC7L2 antibodies followed by mass spectrometry to identify binding partners (as identified with SQSTM1)

    • Confirm protein-protein interactions using reciprocal co-immunoprecipitation

    • Analyze autophagy markers (LC3-I/II conversion, p62/SQSTM1 levels) in cells with altered LUC7L2 expression

    • Assess autophagy flux using chloroquine or bafilomycin A1 treatment

    • Use RNA-seq to identify genes and pathways regulated by LUC7L2 in radioresistant cells

  • Combinatorial treatment approaches:

    • Test LUC7L2 knockdown in combination with autophagy inhibitors (e.g., chloroquine) or other radiosensitizers

    • Use clinically relevant fractionated radiation protocols rather than single-dose treatments

    • Assess both short-term (cell viability) and long-term (colony formation) outcomes

    • Analyze markers of cell death (apoptosis, necrosis, autophagy) to determine mechanism

  • In vivo validation:

    • Establish xenograft models using radioresistant cancer cells with LUC7L2 knockdown or overexpression

    • Apply fractionated radiation therapy using clinically relevant protocols

    • Monitor tumor growth kinetics and response to radiation

    • Analyze tumor samples for LUC7L2 expression, autophagy markers, and radiation response indicators

These methodological approaches provide a comprehensive framework for investigating LUC7L2's role in cancer radioresistance and evaluating its potential as a therapeutic target.

What are the emerging applications of LUC7L2 research in immunotherapy development?

The role of LUC7L2 in regulating MITA/STING-mediated immune responses opens several promising avenues for immunotherapy research:

  • Enhancement of anti-tumor immunity:

    • STING activation is crucial for anti-tumor immune responses, particularly through type I interferon production

    • By manipulating LUC7L2 expression or function in tumor microenvironments, researchers could potentially enhance STING activation and improve cancer immunotherapy efficacy

    • Experimental approaches might include:

      • Localized LUC7L2 silencing in tumors combined with STING agonist therapy

      • Development of inhibitors targeting LUC7L2-MITA interactions

      • Screening for small molecules that modulate LUC7L2 splicing activity

  • Viral vaccine adjuvant development:

    • Given LUC7L2's role in regulating immune responses to DNA viruses , transient inhibition of LUC7L2 could enhance vaccine efficacy

    • Researchers could investigate:

      • LUC7L2 inhibition as an adjuvant strategy for DNA-based vaccines

      • Temporal dynamics of LUC7L2 inhibition for optimal immune enhancement

      • Tissue-specific targeting to minimize systemic effects

  • Autoimmune disease interventions:

    • Excessive STING activation contributes to several autoimmune conditions

    • LUC7L2 enhancement could potentially dampen pathological STING activation

    • Research directions might include:

      • Cell type-specific LUC7L2 overexpression in autoimmune disease models

      • Development of stabilizers for LUC7L2-MITA interactions

      • Identification of natural compounds that enhance LUC7L2 expression or function

  • Infectious disease applications:

    • While LUC7L2 inhibition might enhance innate immune responses to acute infections, controlled modulation could help prevent cytokine storms in severe infections

    • Researchers could explore:

      • Temporal inhibition/enhancement of LUC7L2 at different disease stages

      • Combination therapies with existing antivirals

      • Biomarkers for identifying patients who might benefit from LUC7L2-targeted interventions

  • RNA-based therapeutic approaches:

    • Given LUC7L2's role in RNA splicing, RNA-based technologies might be particularly effective

    • Potential approaches include:

      • Antisense oligonucleotides targeting LUC7L2 mRNA

      • siRNA delivery systems for localized silencing

      • CRISPR-based transcriptional modulation of LUC7L2 expression

These emerging applications represent promising areas for translational research that builds upon fundamental discoveries about LUC7L2's immunoregulatory functions.

How might advances in single-cell technologies impact future LUC7L2 research?

Single-cell technologies are poised to revolutionize our understanding of LUC7L2 biology:

  • Single-cell RNA sequencing (scRNA-seq):

    • Enables characterization of LUC7L2 expression patterns across heterogeneous cell populations

    • Can reveal cell type-specific roles of LUC7L2 in immune responses or cancer

    • Allows identification of rare cell populations with distinctive LUC7L2 expression

    • Potential applications include:

      • Mapping LUC7L2 expression across immune cell subsets during viral infection

      • Identifying cancer cell subpopulations with elevated LUC7L2 expression that may contribute to radioresistance

      • Correlating LUC7L2 expression with cell state transitions during disease progression

  • Single-cell proteomics:

    • Provides protein-level confirmation of LUC7L2 expression patterns observed in transcriptomic data

    • Enables analysis of post-translational modifications that may regulate LUC7L2 function

    • Can detect protein-protein interactions at single-cell resolution

    • LUC7L2 antibodies will be critical reagents for developing validated single-cell proteomic assays

  • Spatial transcriptomics and proteomics:

    • Preserves tissue context while providing single-cell resolution data

    • Allows visualization of LUC7L2 expression patterns in relation to tissue microenvironment

    • Particularly valuable for understanding LUC7L2's role in complex tissues such as tumors or inflamed tissues

    • Immunofluorescence with LUC7L2 antibodies can be integrated with spatial transcriptomics for multi-omic analyses

  • Single-cell epigenomics:

    • Enables investigation of chromatin accessibility and histone modifications at the LUC7L2 locus

    • Can reveal regulatory mechanisms controlling LUC7L2 expression in different cell types

    • May identify targetable epigenetic mechanisms for modulating LUC7L2 expression

  • Integrated multi-omic approaches:

    • Combining transcriptomic, proteomic, and epigenomic data from the same cells provides comprehensive understanding

    • Can reveal disconnects between mRNA and protein levels that might indicate post-transcriptional regulation

    • Helps establish causal relationships between LUC7L2 expression and cellular phenotypes

These single-cell approaches will provide unprecedented resolution in understanding LUC7L2's diverse functions across different cellular contexts and disease states.

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.