The DRAM2 antibody is widely used to investigate its role in retinal diseases, cancer, and autophagy regulation.
Biallelic mutations in DRAM2 cause autosomal recessive retinal dystrophy characterized by early macular cone photoreceptor degeneration . DRAM2 antibodies have been employed in IHC studies to map its localization to photoreceptor inner segments and retinal pigment epithelial (RPE) cells, where it supports photoreceptor renewal .
DRAM2 deficiency disrupts autophagy, impairing photoreceptor outer segment recycling .
In AMD retinas, DRAM2 expression is reduced, correlating with photoreceptor loss .
DRAM2 exhibits tumor-suppressive properties, as its downregulation is observed in ovarian tumors . MicroRNA-125b (miR-125b) directly targets DRAM2, promoting retinoblastoma cell proliferation and invasion while suppressing apoptosis .
Overexpression of DRAM2 reverses miR-125b-induced oncogenic effects in retinoblastoma cells .
DRAM2 knockdown exacerbates RPE cell death under stress (e.g., A2E or NaIO₃ exposure) .
Dilution Recommendations:
Storage: -20°C or -80°C, avoiding repeated freeze-thaw cycles .
Cross-Reactivity: Primarily human and mouse; no data on other species .
Retinal Organoid Models: DRAM2 knockout in human pluripotent stem cell-derived RPE cells reveals its role in mesenchymal cell regulation .
AMD Pathogenesis: Reduced DRAM2 expression in AMD retinas suggests autophagy dysregulation as a disease mechanism .
Therapeutic Targets: miR-125b/DRAM2 axis modulation shows potential for retinoblastoma treatment .
DRAM2 (DNA-damage regulated autophagy modulator 2) is a protein expressed in various tissues, with particularly notable expression in the eye. It has been observed to be ubiquitously expressed across multiple cell types in human retinal tissue, including neurons (photoreceptors, interneurons, and retinal ganglion cells), RPE cells, glia, mesenchymal, and myeloid cells . The detection of DRAM2 is important because its expression has been found to be slightly lower in AMD retinas and RPE/Choroids compared to non-AMD controls, suggesting a potential role in retinal pathologies . Using specific antibodies for DRAM2 detection allows researchers to investigate its localization, expression levels, and potential role in disease processes such as age-related macular degeneration.
DRAM2 antibodies are versatile tools that can be employed in multiple experimental applications. Based on available information, these applications include Western Blot for protein expression quantification, Immunohistochemistry for tissue localization studies, Immunocytochemistry and Immunofluorescence for cellular localization, and Immunohistochemistry with paraffin-embedded samples for archived tissue analysis . These diverse applications enable researchers to investigate DRAM2 expression patterns across different experimental contexts and biological samples. When selecting an application method, researchers should consider the specific research question, sample type, and desired resolution level for DRAM2 detection.
DRAM2 antibodies have been validated for use with human samples . Based on research literature, DRAM2 expression has been studied in various ocular tissues including the ganglion cell layer, inner nuclear layer, outer nuclear layer, retinal pigment epithelium, and choriocapillaris . For in vitro studies, DRAM2 has been investigated in human primary RPE cells and human pluripotent stem cell-derived RPE cells . When working with these samples, researchers should ensure proper sample preparation according to the specific detection method being employed. Additionally, controls should be incorporated to verify antibody specificity and minimize background signal.
For optimal performance and longevity, DRAM2 antibodies should be stored at 4°C for short-term use. For long-term storage, it is recommended to aliquot the antibody and store at -20°C to prevent repeated freeze-thaw cycles which can degrade antibody performance . The antibody is typically supplied in PBS (pH 7.2) with 40% Glycerol and 0.02% Sodium Azide as preservatives . When handling the antibody, researchers should follow standard laboratory practices for protein reagents, including using clean pipette tips, avoiding contamination, and maintaining appropriate temperature conditions during experimental procedures.
Optimizing DRAM2 antibody protocols for retinal tissue requires careful consideration of fixation methods, antigen retrieval, and detection systems. Based on research studies, DRAM2 has been detected in various retinal layers including the ganglion cell layer, inner nuclear layer, outer nuclear layer, and retinal pigment epithelium . For paraffin-embedded sections, appropriate antigen retrieval methods should be employed to unmask epitopes potentially obscured during fixation. Researchers should conduct titration experiments to determine optimal antibody concentration, as excessive antibody can increase background while insufficient antibody may result in weak signal. When analyzing retinal sections, researchers should be aware that DRAM2 expression appears to be sparse and distributed across multiple cell types rather than concentrated in specific regions, which may necessitate careful image acquisition and analysis strategies .
Comprehensive validation of DRAM2 antibody specificity is essential for generating reliable research data. Positive controls should include tissues or cells known to express DRAM2, such as retinal tissues or RPE cells. Negative controls should include samples where the primary antibody is omitted or replaced with non-specific IgG from the same host species (rabbit) . For definitive validation, DRAM2 knockout or knockdown samples provide the gold standard control. Research has utilized CRISPR/Cas9-generated DRAM2 knockout human pluripotent stem cells and shRNA-mediated DRAM2 knockdown in human primary RPE cells as negative controls for antibody specificity . Additionally, antibody specificity has been verified on protein arrays containing the target protein plus 383 other non-specific proteins to ensure minimal cross-reactivity . Implementing these controls helps ensure that observed signals genuinely represent DRAM2 expression rather than non-specific binding.
Distinguishing between reduced DRAM2 expression and cell loss in AMD samples represents a significant methodological challenge. Research has shown that DRAM2 expression is slightly lower in AMD retinas and RPE/Choroids compared to non-AMD controls . To address this ambiguity, researchers should employ co-staining with cell-type specific markers. For example, studies have used RCVRN as a photoreceptor marker and BEST1 as an RPE cell marker to evaluate cell loss . In published research, RCVRN showed decreased expression in AMD retinas (indicating photoreceptor loss), while BEST1 maintained similar expression levels in AMD and non-AMD RPE/Choroid samples (suggesting minimal RPE atrophy) . Correlation analysis between DRAM2 expression and cell-type specific markers within individual samples can help determine whether lower DRAM2 levels reflect genuine expression changes or merely cell population reductions. This approach allows more accurate interpretation of DRAM2 expression changes in disease contexts.
Generation of DRAM2 knockout models can be accomplished through various methodological approaches depending on the experimental system. For mouse models, CRISPR/Cas9 technology has been successfully employed to delete specific exons (e.g., exon 4) of the DRAM2 gene . For human cell models, two main approaches have proven effective: (1) CRISPR/Cas9-mediated knockout in human pluripotent stem cells (hPSCs) using guide RNAs targeting specific exons, and (2) shRNA-mediated knockdown via lentiviral vectors . When generating DRAM2 knockout hPSC lines, researchers have targeted exon 3 using specific guide RNAs (e.g., 5′-AAGGTAAAGCCGGGTCTATA) and confirmed genomic modifications through sequencing of PCR amplicons . For shRNA-mediated knockdown, commercially available constructs (e.g., from Dharmacon) can achieve approximately 10-fold reduction in DRAM2 expression . Verification of knockout/knockdown efficiency should be performed using methods such as qRT-PCR, Western blot, or genomic sequencing to ensure model validity.
When investigating DRAM2 in retinal degeneration, several critical experimental design considerations must be addressed. First, researchers should employ multiple model systems to strengthen findings, as demonstrated by studies using both human in vitro systems (retinal organoids, RPE cells) and mouse in vivo models . Second, appropriate controls must be included to distinguish between genuine expression changes and cell loss, particularly in degenerative conditions where certain cell populations may be depleted . Third, age-matching of samples is essential, as age-related changes may confound results in AMD studies. Fourth, researchers should consider using complementary techniques (e.g., qRT-PCR, in situ hybridization, single-cell RNA sequencing) to validate findings across different methodological approaches . Finally, when challenging cells with stressors (e.g., A2E, sodium iodate), dose-response curves should be established to identify appropriate concentrations that induce moderate stress without overwhelming cellular defense mechanisms .
Discrepancies between DRAM2 mRNA and protein expression data represent a common challenge in molecular biology research. When encountering such discrepancies, researchers should consider several potential explanations. Post-transcriptional regulation mechanisms, including miRNA-mediated repression, may result in reduced protein levels despite normal mRNA expression. Post-translational modifications or protein degradation pathways could also affect protein stability without altering transcript levels. Technical limitations present another consideration - antibody specificity issues might lead to inaccurate protein quantification, while RNA degradation could affect mRNA measurements . The search results indicate that finding anti-DRAM2 antibodies with satisfactory specificity profiles has been challenging for some researchers, highlighting the importance of rigorous antibody validation . To address these discrepancies, researchers should employ multiple detection methods, include appropriate controls, and consider temporal dynamics, as mRNA and protein expression changes may occur with different kinetics.
Detecting DRAM2 protein in tissue samples presents specific challenges that researchers have encountered, with some reporting difficulty finding antibodies with satisfactory specificity profiles . To overcome these limitations, several strategies can be implemented. First, researchers should compare multiple commercially available antibodies, evaluating each for specificity using knockout/knockdown controls. Second, optimization of sample preparation protocols is crucial - different fixation methods, antigen retrieval techniques, and blocking solutions should be systematically tested to maximize signal-to-noise ratio. Third, signal amplification methods (e.g., tyramide signal amplification) can enhance detection of low-abundance proteins. Fourth, alternative approaches such as proximity ligation assay (PLA) or mass spectrometry-based proteomics can provide complementary evidence for DRAM2 protein expression. Finally, genetic tagging strategies (e.g., expressing DRAM2-GFP fusion proteins) in model systems can facilitate detection when native protein visualization proves challenging.
Integration of DRAM2 expression data with other cellular markers provides a powerful approach for understanding its role in retinal pathology. Research has demonstrated the value of correlating DRAM2 expression with cell-type specific markers such as RCVRN (photoreceptors) and BEST1 (RPE cells) to distinguish between genuine expression changes and cell population alterations in AMD . Additionally, leveraging single-nucleus RNA sequencing data allows researchers to analyze DRAM2 expression across multiple cell types simultaneously, revealing its ubiquitous expression pattern in retinal tissues . To effectively integrate these datasets, researchers should employ computational approaches that account for cell-type composition differences between samples. Network analysis of gene co-expression patterns can further illuminate functional relationships between DRAM2 and other genes involved in retinal homeostasis or degeneration. This integrated approach provides a more comprehensive understanding of how DRAM2 functions within the complex cellular ecosystem of the retina.
Several experimental approaches can establish connections between DRAM2 function and cellular stress responses in RPE cells. Cell viability assays following treatment with stressors such as A2E or sodium iodate have demonstrated that DRAM2 loss exacerbates toxicity-induced RPE cell death . To further elucidate mechanisms, researchers can evaluate changes in stress response pathways (e.g., oxidative stress markers, autophagy flux, ER stress) in DRAM2 knockout/knockdown cells compared to controls. RNA sequencing analyses before and after stress induction can identify differentially regulated genes and pathways dependent on DRAM2 expression. Functional assays examining specific RPE cell capabilities, such as phagocytosis of photoreceptor outer segments, can reveal how DRAM2 affects specialized RPE functions under stress conditions . Additionally, rescue experiments, where wild-type DRAM2 is reintroduced into knockout cells, can confirm phenotype specificity. Together, these approaches build a comprehensive understanding of DRAM2's role in RPE stress response mechanisms.
Correlating findings between human in vitro models and mouse in vivo models requires careful consideration of both similarities and differences between these experimental systems. Research has utilized this comparative approach for DRAM2, examining knockout effects in both human stem cell-derived retinal organoids/RPE cells and DRAM2 knockout mice . When integrating data across these models, researchers should first establish equivalent developmental or disease stages to ensure appropriate comparisons. Molecular phenotypes should be assessed using identical or comparable methodologies across models whenever possible. Researchers should acknowledge species-specific differences that might influence results, such as differences in retinal structure, aging timeline, or stress response mechanisms. When discrepancies arise between models, additional experiments should be designed to determine whether these reflect genuine biological differences or technical limitations. Finally, validation in human donor tissues provides a critical bridge between model systems and human pathology, as demonstrated by studies examining DRAM2 expression in AMD donor eyes .