LMX1A Antibody

Shipped with Ice Packs
In Stock

Description

Biological Role of LMX1A

LMX1A is a transcription factor encoded by the LMX1A gene on human chromosome 1. It plays pivotal roles in:

  • Dopaminergic Neuron Development: LMX1A regulates midbrain dopaminergic (mDA) neuron differentiation by controlling genes like Wnt1, Nurr1, and Pitx3 .

  • Mitochondrial Regulation: Sustained LMX1A expression is essential for maintaining mitochondrial function in adult mDA neurons, protecting against oxidative stress and DNA damage linked to Parkinson’s disease (PD) .

  • Cortical Hem Formation: LMX1A acts as a master regulator of the cortical hem, a signaling center critical for hippocampus development .

Research Applications

LMX1A antibodies have been instrumental in:

  • Developmental Studies:

    • Tracking granule cell migration in cerebellar rhombic lip progenitors .

    • Identifying Cajal-Retzius neuron deficits in cortical hem mutants .

  • Disease Models:

    • Linking LMX1A deficiency to PD-like pathology, including α-synuclein aggregation and mitochondrial dysfunction .

    • Validating Wnt1-Lmx1a autoregulatory loops in embryonic stem cell differentiation .

Table 1: LMX1A in Neurodevelopment and Disease

Study FocusKey InsightCitation
Dopaminergic NeuronsLMX1A ablation causes mitochondrial defects and progressive neuron loss, mimicking PD.
Cortical Hem DevelopmentLMX1A deficiency disrupts Wnt signaling and hippocampal boundary formation.
Insulin Gene RegulationLMX1A binds A/T-rich sequences in the insulin promoter, enhancing transcription.

Table 2: LMX1A Antibody Catalog

Product IDHostClonalityApplicationsSupplier
ab139726RabbitPolyclonalWB, IHC, IFAbcam
ABIN2779408RabbitPolyclonalWB, IHCantibodies-online
NBP2-41193RabbitPolyclonalWB, IHC, IFNovus Biologicals

Validation and Quality Control

LMX1A antibodies are rigorously validated:

  • Western Blot: Detects LMX1A at ~45 kDa in human, mouse, and rat skeletal muscle lysates .

  • Immunohistochemistry: Localizes LMX1A in paraffin-embedded brain tissues (e.g., meningioma, cerebellum) .

  • Knockout Validation: Reduced signal in Lmx1a mutant tissues confirms specificity .

Implications for Parkinson’s Disease Research

LMX1A antibodies have uncovered pathways connecting mitochondrial dysfunction and α-synuclein pathology. For example:

  • LMX1A-deficient mice exhibit Lewy neurite-like inclusions and respiratory chain defects .

  • Targeting LMX1A regulatory networks could mitigate dopaminergic neuron vulnerability in PD .

Limitations and Future Directions

  • Species Cross-Reactivity: Limited data for non-mammalian models .

  • Functional Assays: Further studies needed to map LMX1A’s post-translational modifications.

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 your order within 1-3 business days of receipt. Delivery times may vary depending on the shipping method and destination. Please consult your local distributor for specific delivery timelines.
Synonyms
HGNC:6653 antibody; LIM homeobox protein 1.1 antibody; LIM homeobox transcription factor 1 alpha antibody; LIM homeobox transcription factor 1-alpha antibody; LIM/homeobox protein 1.1 antibody; LIM/homeobox protein LMX1A antibody; LMX 1 antibody; LMX 1.1 antibody; LMX 1A antibody; LMX-1.1 antibody; LMX1 antibody; LMX1.1 antibody; LMX1A antibody; LMX1A_HUMAN antibody; MGC87616 antibody
Target Names
LMX1A
Uniprot No.

Target Background

Function
LMX1A acts as a transcriptional activator by binding to an A/T-rich sequence, the FLAT element, within the insulin gene promoter. It is essential for the development of the roof plate, which in turn plays a critical role in specifying dorsal cell fates in the central nervous system and developing vertebrae.
Gene References Into Functions
  1. Research has demonstrated an anti-metastatic function of LMX1A in gastric cancer. This effect is mediated by the negative regulation of beta-catenin signaling target genes. PMID: 27061089
  2. Polymorphisms in the dopamine pathway gene, LMX1A, have been linked to cognitive performance in individuals with bipolar disorder. PMID: 26534905
  3. LMX1A and LMX1B expression persists in mature dopaminergic neurons located in the substantia nigra pars compacta and the ventral tegmental area. [Review] PMID: 26526610
  4. Single nucleotide polymorphisms (SNPs) within the LMX1A gene may be associated with the susceptibility to congenital scoliosis and variations in clinical phenotypes among the Chinese Han population. PMID: 25099324
  5. To our knowledge, this is the first study demonstrating the successful use of TAT-LMX1A recombinant protein to enhance the differentiation of human embryonic stem cells (hESCs) into dopaminergic neurons, as evidenced by the expression of dopaminergic-specific markers. PMID: 25380985
  6. LMX1A exhibits reduced methylation levels in cervical adenocarcinomas compared to control samples, particularly in the metastatic stages of cervical cancer. PMID: 24407576
  7. Elevated LMX1A expression has been associated with more advanced tumor staging in Chinese patients diagnosed with pancreatic ductal adenocarcinomas. PMID: 23165334
  8. Research suggests that Sp1 and EZH2 may activate LMX1A expression during the development of cervical cancer in response to oncogenic stress. PMID: 24018208
  9. Higher immunostaining intensity for osteopontin (OPN) and LMX1A has been correlated with the World Health Organization (WHO) grades assigned to meningiomas and certain gliomas. However, unexpectedly, LMX1A staining in WHO grade IV gliomas was found to be weaker than in WHO grade III tumors. PMID: 22882568
  10. LMX1A can induce the desired neuronal lineages from most expressing neural progenitor cells through a mechanism resembling developmental binary cell-fate switching. PMID: 21624811
  11. A specific single nucleotide polymorphism (SNP) in LMX1A was strongly associated with the magnitude of training-related gains in verbal working memory. PMID: 21435346
  12. LMX1A may serve as a potential biomarker for gastric cancer. PMID: 21159062
  13. Genetic variations in LMX1A may increase the risk of developing schizophrenia. PMID: 20570600
  14. A study did not find evidence to support an association between any LMX1A SNPs and type 2 diabetes mellitus (T2DM) in Pima Indians, suggesting that LMX1A does not significantly contribute to T2DM etiology in this population. PMID: 12062816
  15. LMX1A expression is crucial for the expression of bone morphogenetic protein (BMP), and for the normal generation and differentiation of the dorsal-most spinal cord neurons, the dl1 interneurons. PMID: 15183721
  16. LMX1A may be critical to the development of midbrain dopamine neurons from human embryonic stem cells. PMID: 18832589
  17. Research has shown that three single nucleotide polymorphisms (SNPs) in LMX1A and one in LMX1B are associated with Parkinson's disease. PMID: 19189040

Show More

Hide All

Database Links

HGNC: 6653

OMIM: 600298

KEGG: hsa:4009

STRING: 9606.ENSP00000294816

UniGene: Hs.667312

Subcellular Location
Nucleus.
Tissue Specificity
Isoform 1 is expressed in many tissues. Not found in heart, liver, spleen and testis. Relatively highly expressed in fetal brain. Isoform LMX1A-4AB is expressed in testis.

Q&A

What is LMX1A and why is it significant in neurodevelopmental research?

LMX1A (LIM homeobox transcription factor 1 alpha) functions as a transcriptional activator by binding to an A/T-rich sequence known as the FLAT element in gene promoters. This 42.7 kilodalton protein plays a crucial role in the differentiation and development of dopamine-producing neurons essential for proper nervous system function . LMX1A operates within a complex transcriptional network alongside other key transcription factors such as NURR1 and PITX3 to regulate genes necessary for dopamine synthesis and neuron maintenance . Additionally, it is required for the development of the roof plate and the subsequent specification of dorsal cell fates in the central nervous system (CNS) and developing vertebrae . The significance of studying this protein extends to understanding neurodevelopmental disorders, particularly those affecting dopaminergic systems.

How does LMX1A contribute to inner ear development?

Recent research has established that LMX1A is essential for marginal cell differentiation and stria vascularis development in the inner ear . Expression analysis shows that LMX1A is detected as early as embryonic day 8.5 (E8.5) and by E10.5 is expressed throughout almost the entire otocyst . As development progresses, LMX1A expression becomes concentrated in the endolymphatic sac and the lateral side of the cochlear duct . Studies using LMX1A knockout (KO) mice have demonstrated that absence of LMX1A leads to severe defects in the cellular architecture of the stria vascularis, including the absence of key proteins such as KCNQ1 and BSND (barttin) in marginal cells . These findings highlight the critical role of LMX1A in the proper formation and function of cochlear and vestibular structures, suggesting its importance in hearing development and potentially in addressing congenital hearing disorders.

What alternative nomenclature exists for LMX1A in the scientific literature?

When conducting literature searches and interpreting research papers, it's essential to recognize that LMX1A may be referenced under several alternative designations. The protein is also known as LMX1, LMX1.1, and LIM/homeobox protein 1.1 . In database searches and literature reviews, researchers should include these alternative names to ensure comprehensive coverage of relevant studies. Additionally, gene naming conventions may vary between species, so orthologous genes in model organisms like mouse (Lmx1a) might be formatted differently in publications . Understanding this nomenclature diversity is critical for thorough literature analysis and avoiding duplication of research efforts.

What criteria should guide the selection of an appropriate LMX1A antibody for specific experimental applications?

Selection of an appropriate LMX1A antibody should be guided by several experimental considerations. First, determine the specific application requirements (Western blot, immunohistochemistry, immunofluorescence, or ELISA) as antibodies often perform differently across techniques . For example, some LMX1A antibodies are specifically validated for Western blot and immunocytochemistry/immunofluorescence but may not perform optimally in other applications . Second, consider species reactivity—ensure the antibody recognizes LMX1A in your experimental model organism (human, mouse, rat, etc.) . Third, evaluate the recognition site—antibodies targeting different epitopes (N-terminal, C-terminal, or internal regions) may produce varying results depending on protein conformation and potential post-translational modifications . Finally, review publication citations using the antibody to assess its reliability in experimental contexts similar to yours . For challenging applications, consider antibodies with published validation data showing specificity and sensitivity in your application of interest.

What are the advantages and limitations of polyclonal versus monoclonal LMX1A antibodies in different research applications?

Polyclonal LMX1A Antibodies:

Advantages:

  • Recognize multiple epitopes of LMX1A, potentially increasing signal strength in applications like Western blot and immunohistochemistry

  • Often more tolerant of minor protein denaturation or conformational changes

  • Generally less expensive and easier to produce

  • May be more effective for detecting low-abundance LMX1A expression

Limitations:

  • Batch-to-batch variability can complicate long-term studies

  • Higher potential for cross-reactivity with related proteins

  • May produce higher background in some applications

Monoclonal LMX1A Antibodies:

Advantages:

  • Consistent performance across batches for reproducible results in longitudinal studies

  • Higher specificity for a single epitope, reducing non-specific binding

  • Often preferred for applications requiring precise epitope mapping

  • Superior for distinguishing between closely related protein isoforms

Limitations:

  • May have lower sensitivity if the single epitope is masked or modified

  • Production is more complex and costly

  • The single epitope may be lost during certain sample processing methods

For studying LMX1A in developmental contexts where protein conformation may vary across differentiation stages, researchers might benefit from using polyclonal antibodies that can detect multiple epitopes . Conversely, for highly specific detection of LMX1A in complex tissue samples like neural tissue, monoclonal antibodies may provide cleaner results with less background .

How can researchers validate the specificity of LMX1A antibodies to ensure reliable experimental results?

Validating LMX1A antibody specificity requires a multi-faceted approach to ensure experimental rigor. First, perform positive and negative control experiments—compare staining in tissues or cells known to express high levels of LMX1A (such as developing neural tissue or dopaminergic neurons) versus those with minimal expression . Second, conduct knockout validation by comparing antibody staining between wild-type and LMX1A knockout models; complete absence of signal in knockout samples strongly supports antibody specificity . Third, use knockdown approaches with siRNA or shRNA targeted against LMX1A to demonstrate signal reduction proportional to knockdown efficiency. Fourth, perform Western blotting to confirm the antibody detects a single band of the expected molecular weight (approximately 42.7 kDa) . For additional confidence, employ peptide competition assays where pre-incubation of the antibody with the immunizing peptide should abolish specific staining. Finally, compare results across multiple antibodies targeting different epitopes of LMX1A—concordant results increase confidence in specificity. Documentation of these validation steps is essential for publication and reproducibility of research findings.

What are the optimal fixation and immunostaining protocols for detecting LMX1A in different tissue samples?

Optimal fixation and immunostaining protocols for LMX1A detection vary depending on the tissue type and experimental application. For neural tissues and inner ear samples, 4% paraformaldehyde fixation for 12-24 hours is typically effective . When working with embryonic tissues, shorter fixation times (4-8 hours) may better preserve antigenicity. For immunofluorescence applications in cultured cells, formaldehyde fixation followed by permeabilization with 0.3% Triton X-100 has been successfully used with LMX1A antibodies . Antigen retrieval methods may be necessary for certain fixed tissues—citrate buffer (pH 6.0) heat-induced retrieval often enhances LMX1A epitope accessibility. For blocking, 3% BSA or 30% normal goat serum has proven effective in reducing background . Primary antibody incubation should be conducted at 4°C overnight at dilutions typically ranging from 1:100 to 1:500 depending on the specific antibody . For visualization, Alexa Fluor secondary antibodies (e.g., Alexa Fluor 488) at 1:1,000 dilution provide good signal-to-noise ratio . When performing double or triple immunostaining, careful selection of antibodies from different host species is essential to avoid cross-reactivity. Each new tissue type or developmental stage may require protocol optimization to balance signal intensity with background reduction.

How can LMX1A antibodies be effectively used in analyzing neurodevelopmental processes?

LMX1A antibodies serve as powerful tools for analyzing neurodevelopmental processes, particularly in studying dopaminergic neuron differentiation and roof plate development. For developmental time course studies, researchers can use LMX1A antibodies in conjunction with stage-specific markers to track the progression of neural specification and differentiation . In neural stem cell cultures, LMX1A immunocytochemistry helps identify cells committed to dopaminergic lineage, especially when combined with other markers like NURR1 and PITX3 . Quantitative assessment of LMX1A expression through fluorescence intensity measurements can reveal how manipulation of signaling pathways affects dopaminergic specification. For spatial analysis of expression patterns in developing neural tube, immunohistochemistry on cryosections using LMX1A antibodies can be combined with roof plate markers to understand dorsal-ventral patterning mechanisms . In disease models, such as Parkinson's disease, LMX1A antibodies can evaluate the success of protocols designed to generate dopaminergic neurons from stem cells. When performing these experiments, appropriate controls are essential, including wild-type vs. knockout tissue comparisons and developmental stage-matched samples . Researchers should also consider using phospho-specific LMX1A antibodies when available to investigate how post-translational modifications affect transcription factor activity during critical developmental windows.

What troubleshooting approaches are recommended when LMX1A antibody staining yields inconsistent results?

When encountering inconsistent LMX1A antibody staining, systematic troubleshooting is essential. First, verify antibody quality and storage conditions—LMX1A antibodies may lose activity if subjected to repeated freeze-thaw cycles or stored improperly . Second, optimize fixation parameters—overfixation can mask epitopes while underfixation may compromise tissue morphology; try a range of fixation times to determine optimal conditions . Third, enhance antigen retrieval—if using paraffin sections, test different antigen retrieval methods (heat-induced epitope retrieval with citrate buffer or EDTA) and durations . Fourth, adjust antibody concentration—perform a dilution series (e.g., 1:100, 1:200, 1:500) to identify the optimal signal-to-noise ratio for your specific sample type . Fifth, extend primary antibody incubation—overnight incubation at 4°C often yields more consistent results than shorter incubations at room temperature . Sixth, modify blocking conditions—test different blocking agents (BSA, normal serum, commercial blocking reagents) to reduce background . Finally, consider tissue-specific factors—expression levels of LMX1A vary significantly across developmental stages and cell types, so ensure appropriate positive controls are included . For particularly challenging samples, signal amplification systems like tyramide signal amplification may enhance detection of low-abundance LMX1A expression. Document all optimization steps methodically to establish a reliable protocol for future experiments.

How can LMX1A antibodies be utilized in studying inner ear development and related disorders?

LMX1A antibodies offer sophisticated approaches for investigating inner ear development and related disorders. For comprehensive analysis of stria vascularis development, researchers can implement multi-label immunofluorescence combining LMX1A antibodies with markers for specific cellular compartments such as KCNQ1 and BSND for marginal cells, and CD44 for intermediate cells . Time-course studies using LMX1A antibodies at defined embryonic stages (E8.5, E10.5, E18.5) can track the dynamic expression patterns during critical periods of cochlear development . For mechanistic investigation of LMX1A function, researchers should consider chromatin immunoprecipitation (ChIP) using validated LMX1A antibodies to identify direct transcriptional targets regulating inner ear development. In disease models, such as mouse strains with hearing deficits, comparative immunohistochemistry with LMX1A antibodies can reveal structural abnormalities in cochlear development. For single-cell resolution analysis, combine LMX1A immunostaining with laser capture microdissection to isolate specific cell populations for transcriptomic or proteomic profiling. When analyzing human temporal bone specimens from patients with congenital hearing disorders, optimized protocols for LMX1A immunohistochemistry may reveal patterns of developmental disruption. These approaches should incorporate appropriate controls, including LMX1A knockout mouse tissues as negative controls and wild-type littermates as positive controls . The integration of LMX1A antibody-based techniques with genetic manipulation models provides powerful insights into the molecular mechanisms of inner ear development and pathogenesis of hearing disorders.

What methodological considerations apply when using LMX1A antibodies in studies of dopaminergic neuron differentiation?

When using LMX1A antibodies to study dopaminergic neuron differentiation, several methodological considerations are critical for robust experimental design. First, implement time-resolved analysis—LMX1A expression precedes terminal dopaminergic differentiation, so design experiments to capture sequential expression of LMX1A followed by NURR1, PITX3, and finally tyrosine hydroxylase (TH) . Second, employ co-labeling strategies combining LMX1A antibodies with markers for neural progenitors (SOX2, Nestin) and dopaminergic neurons (TH, DAT) to precisely identify transitional cell states . Third, consider subcellular localization—use confocal microscopy with nuclear counterstains to confirm the expected nuclear localization of this transcription factor, as cytoplasmic staining may indicate experimental artifacts . Fourth, incorporate functional validation by correlating LMX1A immunostaining with dopamine production measured by HPLC or dopaminergic neuron functionality assessed by electrophysiology. Fifth, when manipulating LMX1A expression (overexpression or knockdown), use antibodies targeting epitopes not affected by your genetic modifications . Finally, for translational studies using human iPSC-derived dopaminergic neurons, validate that your LMX1A antibody maintains specificity across species . For quantitative analyses, develop standardized image acquisition parameters and automated analysis workflows to minimize subjective interpretation. These methodological refinements enhance the reliability of LMX1A antibody-based investigations in dopaminergic differentiation research, which has significant implications for Parkinson's disease and regenerative medicine.

How might LMX1A antibodies contribute to understanding the role of this transcription factor in tumor biology?

Recent evidence suggests LMX1A may function as a biomarker or risk factor in tumor aggressiveness, opening a new frontier for LMX1A antibody applications in cancer research . For tumor classification studies, researchers can develop immunohistochemistry panels incorporating LMX1A antibodies to identify specific molecular subtypes with distinct prognostic outcomes. Quantitative image analysis of LMX1A immunostaining in tumor microarrays can establish correlations between expression levels and clinical parameters such as survival rates, metastatic potential, and treatment response . For mechanistic investigations, chromatin immunoprecipitation sequencing (ChIP-seq) using validated LMX1A antibodies can map the transcriptional networks regulated by this factor in different tumor contexts. Researchers should implement dual immunofluorescence approaches combining LMX1A with markers of tumor stemness or differentiation to characterize the cellular states associated with LMX1A expression. In tumor models, spatial analysis of LMX1A expression at tumor-normal tissue interfaces may reveal roles in invasion and metastasis. For translational applications, develop protocols for LMX1A detection in liquid biopsies using techniques like proximity extension assays with LMX1A antibodies. When conducting these studies, appropriate controls are essential, including normal adjacent tissue, isotype controls, and comparative analysis across diverse tumor types. These advanced applications require highly specific antibodies with validated performance in complex tissue microenvironments. As this research area develops, correlation of LMX1A expression patterns with genomic alterations and epigenetic profiles will provide comprehensive insights into its role in oncogenesis and potential as a therapeutic target.

What quantitative approaches are recommended for analyzing LMX1A expression patterns in developmental studies?

When analyzing LMX1A expression patterns in developmental studies, several quantitative approaches enhance data reliability and interpretability. First, implement developmental staging normalization—precisely define embryonic stages (e.g., E8.5, E10.5, E18.5) using standardized morphological criteria to ensure comparable analysis across specimens . Second, employ semi-quantitative scoring systems for immunohistochemistry—develop 0-3+ or 0-4+ scales based on staining intensity and distribution patterns, with multiple blinded observers scoring samples to reduce subjectivity . Third, utilize digital image analysis—convert immunofluorescence images to quantifiable data using software like ImageJ or CellProfiler to measure parameters such as nuclear/cytoplasmic ratios of LMX1A, intensity distributions, and co-localization coefficients with other markers . Fourth, apply spatial transcriptomics approaches—correlate LMX1A protein expression with its mRNA distribution using techniques like RNAscope combined with immunofluorescence to understand transcriptional and post-transcriptional regulation. Fifth, perform comparative quantification across mutant models—standardize quantification methods when comparing LMX1A expression between wild-type and knockout or transgenic models . For comprehensive developmental analysis, generate quantitative expression maps showing LMX1A levels across anatomical structures throughout developmental timepoints. These approaches should be accompanied by appropriate statistical analyses, including tests for normal distribution and selection of parametric or non-parametric methods accordingly. The integration of these quantitative methods provides robust assessment of LMX1A expression dynamics during critical developmental windows.

How should researchers address discrepancies between LMX1A antibody results and mRNA expression data?

Discrepancies between LMX1A protein detection using antibodies and mRNA expression data represent a complex challenge requiring systematic investigation. First, consider temporal dynamics—LMX1A mRNA and protein expression may be temporally offset due to transcriptional, translational, and post-translational regulation; design time-course experiments with narrow intervals to capture these differences . Second, evaluate post-transcriptional regulation—assess microRNA targeting of LMX1A through bioinformatic prediction tools and experimental validation using reporter assays. Third, investigate protein stability factors—LMX1A may undergo regulated degradation in specific contexts; consider proteasome inhibition experiments to determine if protein turnover explains discrepancies. Fourth, examine epitope accessibility issues—certain protein modifications or interactions may mask antibody epitopes without affecting protein levels; test multiple antibodies targeting different LMX1A regions . Fifth, validate antibody specificity in the specific experimental context—perform Western blots of the tissue or cells being studied to confirm the antibody detects a band of the expected size . Sixth, assess technical variables—RNA preservation may differ from protein preservation in certain protocols; optimize and standardize sample collection procedures. When reporting such discrepancies in publications, present both datasets transparently with detailed methodological descriptions and discuss potential biological or technical explanations. These approaches transform apparent contradictions into opportunities for discovering novel regulatory mechanisms controlling LMX1A expression and function.

What are the key considerations when interpreting LMX1A expression in the context of neurological disease models?

Interpreting LMX1A expression in neurological disease models requires nuanced analytical approaches. First, establish appropriate baseline expression—characterize LMX1A expression patterns in healthy controls matched for age, sex, and genetic background, recognizing that normal expression varies across brain regions and developmental stages . Second, differentiate between causative and compensatory changes—determine whether altered LMX1A expression represents a primary pathogenic mechanism or a secondary response to disease processes through time-course studies and intervention experiments. Third, evaluate cell type-specific effects—use dual immunolabeling with cell type-specific markers to determine whether LMX1A alterations occur globally or in specific neural populations . Fourth, consider regional vulnerability—systematically analyze multiple brain regions to identify selective vulnerability patterns that may explain disease phenotypes. Fifth, correlate with functional outcomes—establish relationships between LMX1A expression patterns and functional metrics such as dopamine levels, electrophysiological properties, or behavioral phenotypes . Sixth, examine gene-environment interactions—assess how environmental factors relevant to the disease model (oxidative stress, inflammation, toxin exposure) modulate LMX1A expression. For translational relevance, compare findings from animal models with human post-mortem samples using carefully validated antibodies that work across species . These analytical frameworks help distinguish meaningful disease-associated changes from experimental artifacts or epiphenomena, leading to more accurate interpretations of LMX1A's role in neurological disease pathogenesis and potential as a therapeutic target.

What are the typical working dilutions and experimental conditions for commonly used LMX1A antibodies?

ApplicationAntibody TypeRecommended DilutionIncubation ConditionsSample TypePositive Control
Western BlotPolyclonal1:500 - 1:1000Overnight at 4°CCell/tissue lysateNeural stem cells
IHC-ParaffinPolyclonal1:100 - 1:200Overnight at 4°CFFPE sectionsDeveloping neural tube
ICC/IFPolyclonal1:200 - 1:50016 hours at 4°CFixed cellsMouse neural stem cells
ELISAPolyclonal/Monoclonal1:1000 - 1:50001-2 hours at RTPurified proteinRecombinant LMX1A
ChIPPolyclonal5-10 μg per IPOvernight at 4°CCross-linked chromatinDopaminergic cell lines
Flow CytometryMonoclonal1:50 - 1:10030-60 min at 4°CFixed/permeabilized cellsLMX1A-expressing cell lines

These parameters serve as starting points and may require optimization based on specific experimental conditions, sample types, and individual antibody characteristics . For new applications or sample types, a dilution series is recommended to determine optimal signal-to-noise ratio. Document all optimization parameters for consistent results across experiments and enable reproducibility by other researchers.

What are the expected staining patterns of LMX1A in different tissue and cell types?

Tissue/Cell TypeSubcellular LocalizationExpression PatternDevelopmental StageCo-expression MarkersNotes
Neural stem cellsNuclearHomogeneous in committed populationsEarly differentiationSOX2, Nestin Strong expression in cells committed to dopaminergic fate
Developing neural tubeNuclearDorsal midline (roof plate)E8.5-E18.5GDF7, BMP6 Expression decreases ventrally following concentration gradient
Dopaminergic neuronsNuclearHomogeneous in mature neuronsPost-mitoticTH, NURR1, PITX3 Maintained in mature dopaminergic neurons of substantia nigra
Inner ear/OtocystNuclearAlmost entire otocyst by E10.5E8.5-E18.5KCNQ1, BSND Later restricted to endolymphatic sac and lateral cochlear duct
Stria vascularisNuclearMarginal cell layerE18.5-P2KCNQ1, BSND Absent in Lmx1a knockout mice
CerebellumNuclearRestricted to specific neuronal populationsPostnatalATOH1Lower expression compared to dopaminergic regions

This table provides expected staining patterns that can serve as reference points for validating antibody specificity and interpreting experimental results. Deviations from these patterns may indicate technical issues with staining protocols or reveal novel biological insights depending on experimental context .

What specific molecular weight and banding patterns should be expected in Western blot analysis of LMX1A protein?

Expected banding patterns by tissue type:

  • Neural stem cells: Strong single band at 42.7 kDa

  • Developing neural tissue: Primary band at 42.7 kDa with possible phosphorylated forms at slightly higher molecular weights

  • Inner ear tissue: Primary band at 42.7 kDa, may be weaker in adult compared to embryonic samples

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.