The ALS2CR12 Antibody is a research-grade rabbit polyclonal antibody designed to detect the human ALS2CR12 protein (UniProt ID: Q96Q35). This protein is primarily associated with male germ cell development and is localized to the sperm flagellum, specifically the fibrous sheath and outer dense fibers . The antibody is used in applications such as Western blot (WB), immunohistochemistry (IHC), and ELISA to study ALS2CR12’s role in spermatogenesis and cellular structures .
ALS2CR12 is critical for sperm motility. Studies using ALS2CR12 Antibody reveal:
Localizes to the sperm flagellum fibrous sheath, a structural component essential for sperm movement .
Expressed in mature sperm and germ cells during spermatogenesis .
GO Annotations: Associated with cytoplasm, outer dense fibers, and sperm flagellum .
To address concerns about antibody reliability, validation protocols include:
CRISPR Knockout (KO) Cell Lines: Comparing immunoblot signals between parental and KO cells to confirm specificity .
Blocking Experiments: Using a recombinant ALS2CR12 fragment to inhibit antibody binding .
Tissue Preparation: Optimal results require antigen retrieval (e.g., pH 9.0, 110°C) for paraffin-embedded samples .
Staining Patterns: ALS2CR12 Antibody detects punctate signals in sperm tails and specific brain regions (e.g., hippocampus, cerebellum) in KO-negative controls .
ALS2CR12 (amyotrophic lateral sclerosis 2 chromosome region candidate 12) is a gene of interest located in the chromosomal region associated with amyotrophic lateral sclerosis type 2. The gene is particularly notable for its expression pattern, as it has been identified as a spermatogenic cell-specific gene with significant roles in male germ cell development and potentially in sperm function. Research has shown that ALS2CR12 produces multiple transcript isoforms through alternative splicing, with at least one isoform being uniquely expressed in spermatogenic cells . The protein's localization in the sperm flagellum, specifically in association with the fibrous sheath, suggests its involvement in sperm motility and function, making it a valuable target for reproductive biology research .
Expression analysis has revealed that ALS2CR12 exhibits a highly specific expression pattern primarily restricted to male germ cells. In vitro and in silico analyses have demonstrated that while some isoforms may have broader expression patterns, at least one transcript isoform is uniquely expressed in spermatogenic cells . The protein product from the germ cell-specific Als2cr12 transcript is present in both mature sperm from the epididymis and developing germ cells within the testis . This tissue-specific expression pattern suggests a specialized role in spermatogenesis and potentially in sperm function. Researchers investigating this protein should consider these expression characteristics when designing experiments to understand its developmental regulation and functional significance.
Validating ALS2CR12 antibody specificity requires a multi-approach strategy to ensure experimental reliability. Begin with western blot analysis comparing tissues with known ALS2CR12 expression (testis and epididymal sperm) against negative control tissues to confirm the detection of appropriately sized bands corresponding to predicted ALS2CR12 protein variants . Immunoprecipitation followed by mass spectrometry can provide definitive identification of the protein recognized by the antibody. For spatial validation, immunolocalization studies should reveal the characteristic fibrous sheath localization pattern in sperm flagellum . Additionally, performing antibody specificity tests using recombinant ALS2CR12 protein and competitive binding assays can further validate specificity. Knockdown or knockout models, when available, offer the most stringent validation by demonstrating reduced or absent signal in samples where ALS2CR12 expression has been suppressed.
For comprehensive detection of ALS2CR12 splice variants, researchers should implement a coordinated approach combining multiple techniques. RT-PCR using primers spanning potential splice junctions has successfully identified two transcript isoforms of the mouse Als2cr12 gene . For more detailed analysis, 5' and 3' RACE (Rapid Amplification of cDNA Ends) can characterize the full transcript structures including untranslated regions. RNA-Seq provides a broader view of the transcriptome while enabling quantitative analysis of isoform expression across different tissues or developmental stages. For protein-level verification of splice variants, western blotting with antibodies targeting common and variant-specific epitopes can confirm translation of the different mRNA species. When analyzing data, particular attention should be paid to the spermatogenic cell-specific isoform, which has been established as having distinct expression patterns from other potential variants .
Determining the precise subcellular localization of ALS2CR12 in sperm cells requires multiple complementary imaging approaches. High-resolution immunofluorescence microscopy with co-staining for known flagellar compartment markers is essential, as research has shown ALS2CR12 associates with the fibrous sheath in the sperm flagellum . For more definitive localization, immunogold electron microscopy offers nanometer-scale resolution to pinpoint the exact structural association within the flagellum. Biochemical fractionation of sperm cells followed by western blot analysis of the isolated fractions (membrane, cytoskeletal, and soluble components) can confirm the association with specific subcellular structures. Additionally, proximity labeling techniques such as BioID or APEX can identify neighboring proteins, providing functional context for ALS2CR12's localization. When conducting these studies, researchers should consider the dynamic nature of sperm development and examine localization patterns throughout spermatogenesis as well as in mature sperm from the epididymis .
Distinguishing the functional roles of different ALS2CR12 isoforms requires strategic experimental design that specifically targets individual variants. Begin with isoform-specific knockdown or knockout models using CRISPR-Cas9 with guide RNAs targeting unique exons or splice junctions of each variant. Phenotypic analysis should focus on sperm development, morphology, and motility parameters, given ALS2CR12's association with the sperm flagellum . For complementation studies, rescue experiments using constructs expressing individual isoforms can confirm functional specificity. Interactome analysis through co-immunoprecipitation or yeast two-hybrid screening with isoform-specific bait proteins may reveal unique binding partners. Temporal expression analysis during spermatogenesis can provide insights into stage-specific functions, particularly comparing the spermatogenic cell-specific isoform to any other variants . Lastly, computational structural prediction followed by targeted mutagenesis of isoform-specific domains can establish structure-function relationships unique to each variant.
Given ALS2CR12's localization to the fibrous sheath of the sperm flagellum and its specific expression in spermatogenic cells, its dysfunction could potentially contribute to male infertility through several mechanisms . The fibrous sheath provides structural support to the flagellum and serves as a scaffold for proteins involved in signal transduction and energy production essential for sperm motility. Dysregulation of ALS2CR12 might therefore lead to structural abnormalities in the flagellum or disrupted signaling pathways controlling sperm movement. Researchers investigating this connection should consider: (1) screening infertile male populations, particularly those with asthenozoospermia (reduced sperm motility), for ALS2CR12 mutations or expression abnormalities; (2) detailed ultrastructural analysis of sperm flagella in cases with identified ALS2CR12 variations; and (3) functional studies in animal models with targeted disruption of ALS2CR12. Correlative studies examining ALS2CR12 expression levels or protein modifications in relation to sperm motility parameters could provide additional insights into how this protein contributes to male fertility.
Developing highly specific antibodies against different ALS2CR12 epitopes presents several technical challenges that researchers must address strategically. First, the existence of alternative splice variants necessitates careful epitope selection to either distinguish between isoforms or recognize common regions . Researchers should perform detailed sequence analysis to identify regions unique to each isoform and regions conserved across variants. Second, the high specificity of ALS2CR12 expression to male germ cells may complicate antibody validation in standard cell lines, requiring specialized testicular cell cultures or transgenic expression systems . Third, potential sequence homology with related proteins must be thoroughly assessed to prevent cross-reactivity. For monoclonal antibody development, researchers should consider using multiple immunization strategies with different antigen formats (peptides, recombinant protein fragments, and full-length protein) to maximize the diversity of the antibody repertoire. Finally, comprehensive validation through techniques including western blotting, immunoprecipitation, and immunohistochemistry using both positive (testicular tissue) and negative control tissues is essential to confirm specificity before application in critical research contexts.
Advanced imaging techniques offer unprecedented opportunities to visualize ALS2CR12 dynamics throughout spermatogenesis, revealing functional insights beyond static localization. Super-resolution microscopy techniques (STORM, PALM, or STED) can overcome the diffraction limit to visualize ALS2CR12 organization within the fibrous sheath at nanometer resolution, potentially revealing organizational patterns invisible to conventional microscopy . Live-cell imaging using fluorescently tagged ALS2CR12 in ex vivo testicular explants or organoids can track its recruitment to developing flagellar structures in real time. For studying protein dynamics, fluorescence recovery after photobleaching (FRAP) or photoactivation can measure ALS2CR12 mobility and turnover rates within the flagellum. Expansion microscopy physically enlarges biological specimens, potentially revealing finer details of ALS2CR12 integration into the fibrous sheath. Correlative light and electron microscopy (CLEM) combines the molecular specificity of fluorescence with ultrastructural context from electron microscopy. These techniques could collectively establish a comprehensive timeline of ALS2CR12 incorporation into the developing flagellum and identify any potential redistribution during sperm maturation or capacitation events.
Investigating ALS2CR12 binding partners in the sperm flagellum requires specialized approaches that account for the unique structure and protein composition of this cellular compartment. Proximity-dependent biotin labeling methods (BioID or TurboID) are particularly valuable, as they can identify nearby proteins in living cells by expressing ALS2CR12 fused to a biotin ligase that tags proximal proteins for subsequent purification and identification . For more direct interaction analysis, cross-linking mass spectrometry can covalently link interacting proteins before digestion and analysis, preserving transient or weak interactions. Targeted approaches such as co-immunoprecipitation from purified flagellar fractions followed by mass spectrometry can identify stable interactors. Complementary biophysical methods like microscale thermophoresis or surface plasmon resonance using recombinant ALS2CR12 domains can verify direct binding and determine interaction affinities. Functional validation of identified interactions should include co-localization studies in sperm cells and assessment of how disrupting these interactions affects flagellar structure or function. Given ALS2CR12's association with the fibrous sheath , particular attention should be paid to potential interactions with known structural components of this flagellar element and signaling proteins that regulate sperm motility.
Single-cell transcriptomics represents a powerful approach to decipher the precise temporal regulation of ALS2CR12 expression throughout the complex process of spermatogenesis. This technology can reveal stage-specific expression patterns across the continuum from spermatogonia to mature spermatozoa with unprecedented resolution. Researchers can map the exact timing of upregulation for each ALS2CR12 splice variant, potentially identifying critical developmental windows when the spermatogenic cell-specific isoform becomes dominant . Integrating single-cell transcriptomics with parallel measurements of chromatin accessibility (scATAC-seq) can identify regulatory elements controlling stage-specific expression. Trajectory analysis algorithms applied to single-cell data can reconstruct the developmental progression of germ cells, placing ALS2CR12 expression changes within the broader context of transcriptional programs driving spermatogenesis. Co-expression network analysis may identify transcription factors or regulatory RNAs that coordinate ALS2CR12 expression with other flagellar components. This approach could be particularly valuable for understanding the regulatory mechanisms governing the alternative splicing that generates the germ cell-specific isoform of ALS2CR12 , potentially revealing therapeutic targets for male contraception or fertility enhancement.
The highly specific expression pattern of ALS2CR12 in spermatogenic cells and its localization to a critical functional component of sperm—the flagellar fibrous sheath—positions it as a potential target for male contraceptive development . ALS2CR12 antibodies could contribute to this field through several research and development pathways. Initially, these antibodies serve as essential tools for validating ALS2CR12 as a contraceptive target by characterizing its exact function in sperm motility and fertilization competence. For contraceptive development strategies, researchers might explore vaccination approaches that induce endogenous ALS2CR12 antibody production to interfere with sperm function without affecting hormone levels or other physiological processes. Alternatively, engineered antibody fragments or single-domain antibodies could be developed as topical contraceptives that bind to ALS2CR12 on sperm surfaces, potentially immobilizing sperm or preventing capacitation. Research should include detailed assessment of reversibility by monitoring sperm function recovery after antibody clearance. Safety evaluation must be comprehensive, examining whether anti-ALS2CR12 approaches affect other tissues despite its apparently highly specific expression pattern . Cross-reactivity testing with human tissues beyond the reproductive system would be essential before clinical development of any antibody-based contraceptive targeting this protein.