DRP1B Antibody

Shipped with Ice Packs
In Stock

Description

DRP1 Overview

DRP1 (DNM1L) is a dynamin-family GTPase critical for mitochondrial fission, synaptic vesicle recycling, and peroxisomal division . Dysregulation of DRP1 is implicated in neurodegenerative diseases, cancer, and metabolic disorders . Its activity is modulated by phosphorylation (e.g., Ser616 activation, Ser637 inhibition) and interactions with Aβ oligomers in Alzheimer’s disease .

DRP1 Rabbit Polyclonal Antibody (CAB16661)

  • Host Species: Rabbit

  • Reactivity: Human, Mouse, Rat

  • Immunogen: Amino acids 437–736 of human DNM1L

  • Applications: Western blot (WB)

  • Sequence Coverage: Includes residues critical for GTPase activity and mitochondrial localization .

DRP1 (D6C7) Rabbit mAb #8570

  • Host Species: Rabbit

  • Reactivity: Human, Mouse, Rat, Monkey

  • Applications: WB, Immunoprecipitation (IP), Immunofluorescence (IF) .

  • Validation: Detects endogenous DRP1 at 78–82 kDa .

DRP1 Antibody (AF6784)

  • Host Species: Rabbit

  • Reactivity: Human, Mouse, Rat

  • Immunogen: Synthesized peptide from human DRP1 (UniProt O00429) .

  • PTMs: Phosphorylation (Ser616, Ser637), SUMOylation (Lys558), ubiquitination .

Mitochondrial Dynamics

  • Role in Fission: DRP1 antibodies (e.g., Cell Signaling #8570) localize to mitochondrial division sites, as shown via IF . Mutations (e.g., K38A, G281D) disrupt fission, leading to hyperconnected mitochondria .

  • Disease Links:

    • Alzheimer’s Disease: DRP1 interacts with Aβ oligomers, correlating with mitochondrial fragmentation .

    • Cancer: EBV-LMP1 activates DRP1 (phospho-Ser616), enhancing glycolysis and survival in nasopharyngeal carcinoma .

Technical Validation

AntibodyValidation DataSource
ab184247 (Abcam)WB: 83–86 kDa bands in PC-12, NIH/3T3 lysates; IP in HeLa cells .
SCBT C-5Detects all DRP1 isoforms; used in apoptosis studies (mitochondrial translocation) .
NB110-55288IHC-P: Cytoplasmic staining in kidney tubules; WB: 81 kDa band .

Critical Epitopes and Functional Domains

  • GTPase Domain: Targeted by antibodies like AF6784 (residues 1–300) .

  • Phosphorylation Sites:

    • Ser616: Activated by CDK1/5, promotes fission .

    • Ser637: Inhibited by PKA, reduces fission .

  • Variable Isoforms: Isoform 1 (brain-specific), Isoform 5 (liver/heart), Isoform 6 (neurons) .

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
DRP1B antibody; ADL1B antibody; At3g61760 antibody; F15G16.150Dynamin-related protein 1B antibody; Dynamin-like protein B antibody
Target Names
DRP1B
Uniprot No.

Target Background

Function
DRP1B is a microtubule-associated force-producing protein that plays a crucial role in cytokinesis. It is specifically localized to the leading edges of the forming cell plate during this process. DRP1B exhibits GTPase activity.
Gene References Into Functions
  1. SIN2 GTPase, a homolog of DRP1B, has been shown to support a mitochondrial role in regulating diverse developmental pathways. Research indicates significant pleiotropic effects of SIN2 on both vegetative and floral growth processes. PMID: 16849600
Database Links

KEGG: ath:AT3G61760

STRING: 3702.AT3G61760.1

UniGene: At.34232

Protein Families
TRAFAC class dynamin-like GTPase superfamily, Dynamin/Fzo/YdjA family
Subcellular Location
Cytoplasm. Cytoplasm, cytoskeleton. Cytoplasm, cytoskeleton, phragmoplast.

Q&A

What is the functional significance of DRP1 in cellular processes?

DRP1 (dynamin-related protein 1), also known as DNM1L (dynamin-1-like protein), plays a crucial role in mitochondrial fission, which is essential for maintaining mitochondrial function and cellular homeostasis. This GTPase mediates the division of mitochondrial outer membranes through a process that requires GTP hydrolysis. The protein's activity is regulated through post-translational modifications including phosphorylation, SUMOylation, and ubiquitination, which affect its recruitment to mitochondria. Importantly, dysregulation of DRP1 has been associated with various pathological conditions, including neurodegenerative disorders and cancer, making it a significant target for research investigating mitochondrial dynamics in disease pathogenesis .

How do I determine the appropriate DRP1 antibody for my research application?

Selection of a suitable DRP1 antibody depends on several factors including the experimental technique, species of interest, and target epitope. For Western blot applications, polyclonal antibodies like the DNM1L Polyclonal Antibody (CAB16661) provide excellent sensitivity and can detect denatured protein effectively. Consider the host species (e.g., rabbit) and reactivity (human, mouse, rat) to ensure compatibility with your experimental system. Review the immunogen information to determine which region of DRP1 the antibody recognizes - for instance, antibodies targeting the amino acid sequence 437-736 of human DNM1L may be optimal for certain applications. Additionally, consult published literature to identify validated antibodies for specific techniques beyond Western blotting, such as immunohistochemistry, immunoprecipitation, or immunofluorescence microscopy .

What controls should be included when using DRP1 antibodies in experimental procedures?

Rigorous controls are essential for validating DRP1 antibody specificity and ensuring reliable results. Include a positive control (tissue or cell line known to express DRP1), negative control (tissue or cell line with low or no DRP1 expression), and technical controls (primary antibody omission, isotype control) in your experimental design. For more definitive validation, consider using DRP1 knockdown or knockout samples, which should show reduced or absent signal. When assessing DRP1 phosphorylation states, include controls treated with phosphatase inhibitors versus phosphatase enzymes. For subcellular localization studies, co-staining with established mitochondrial markers (e.g., TOM20, MitoTracker) is advisable to confirm DRP1 recruitment to mitochondria. These comprehensive controls will help distinguish specific signal from background and validate antibody performance in your specific experimental system .

How can I effectively distinguish between cytosolic and mitochondria-associated DRP1 populations using antibody-based techniques?

Differentiating cytosolic from mitochondria-associated DRP1 requires sophisticated approaches beyond standard immunostaining. Implement subcellular fractionation protocols to physically separate cytosolic and mitochondrial compartments before Western blot analysis with DRP1 antibodies. Validate fraction purity using compartment-specific markers such as GAPDH (cytosolic) and COX IV (mitochondrial). For higher resolution analysis, employ super-resolution microscopy techniques (STED, STORM, or PALM) with dual-labeling using mitochondrial markers and DRP1 antibodies to visualize recruitment dynamics. Proximity ligation assays (PLA) can detect DRP1 interactions with mitochondrial binding partners like MFF or Fis1 with nanometer-scale precision. For dynamic studies, combining FRAP (Fluorescence Recovery After Photobleaching) with antibody-based detection in fixed time-point analyses allows quantification of DRP1 trafficking between compartments. These approaches collectively provide a comprehensive understanding of DRP1 distribution and translocation dynamics under various experimental conditions .

What are the methodological considerations when investigating post-translational modifications of DRP1 using specific antibodies?

Investigating DRP1 post-translational modifications (PTMs) requires careful experimental design to preserve modification states and ensure specific detection. Begin by incorporating phosphatase inhibitors (for phosphorylation studies) or deubiquitinase inhibitors (for ubiquitination studies) during sample preparation. For phosphorylation analysis, use phospho-specific antibodies targeting known regulatory sites (Ser616, Ser637/Ser656) in combination with lambda phosphatase treatment controls to confirm specificity. When studying SUMOylation or ubiquitination, optimize immunoprecipitation conditions using antibodies against DRP1 followed by Western blotting with anti-SUMO or anti-ubiquitin antibodies. Consider enrichment strategies such as tandem ubiquitin binding entities (TUBEs) for ubiquitinated proteins prior to DRP1 detection. For comprehensive PTM profiling, combine immunoprecipitation with mass spectrometry analysis. Always validate PTM-specific antibodies using site-directed mutagenesis of the modified residues as negative controls. These approaches provide reliable assessment of DRP1 modification states under different physiological and pathological conditions .

How can DRP1 antibodies be used to investigate the relationship between mitochondrial dysfunction and neurodegenerative diseases?

Investigating the nexus between DRP1, mitochondrial dysfunction, and neurodegeneration requires multi-level analytical approaches. Begin with comparative immunohistochemistry using DRP1 antibodies on post-mortem brain tissue from patients versus healthy controls to assess expression levels and subcellular distribution patterns specific to affected brain regions. Implement co-localization studies with markers of neurodegeneration (e.g., tau, α-synuclein, or Aβ) and mitochondrial fragmentation to establish spatial relationships. For mechanistic insights, utilize primary neuronal cultures or patient-derived iPSCs exposed to disease-relevant stressors, and monitor DRP1 phosphorylation status using phospho-specific antibodies to track activation. Complement these approaches with functional assays measuring mitochondrial parameters (membrane potential, respiration, ROS production) following DRP1 inhibition or activation. In animal models of neurodegeneration, chronological immunohistochemical analysis with DRP1 antibodies can reveal whether aberrant mitochondrial fission precedes or follows other disease markers. These methodologies collectively illuminate how DRP1-mediated mitochondrial dynamics contribute to the pathogenesis of neurodegenerative disorders .

What are the optimal fixation and permeabilization procedures for DRP1 immunocytochemistry?

Preserving DRP1 localization patterns while maintaining antigen accessibility requires careful optimization of fixation and permeabilization procedures. For immunocytochemistry, begin with 4% paraformaldehyde fixation for 15-20 minutes at room temperature, as this provides good structural preservation while maintaining DRP1 antigenicity. Avoid methanol fixation which can disrupt mitochondrial membranes and alter DRP1 distribution patterns. For permeabilization, 0.1-0.2% Triton X-100 for 10 minutes is generally effective, though gentler detergents like 0.1% saponin may better preserve fine mitochondrial morphology. When studying DRP1 phosphorylation states, include phosphatase inhibitors in all buffers. A critical comparison approach is recommended: perform parallel processing with different fixation methods (PFA at different concentrations and times, glutaraldehyde combinations) and different permeabilization agents (Triton X-100, saponin, digitonin) to determine optimal conditions for your specific cell type and antibody. Always validate staining patterns against subcellular markers and control for potential artifacts by comparing results with live-cell imaging of fluorescently-tagged DRP1 when possible .

How can I quantitatively assess DRP1 recruitment to mitochondria using immunofluorescence techniques?

Quantitative assessment of DRP1 recruitment to mitochondria requires rigorous image acquisition and analysis protocols. Begin with triple immunofluorescence labeling using antibodies against DRP1, a mitochondrial outer membrane marker (e.g., TOM20), and a cytosolic marker for reference. Acquire high-resolution z-stack images using confocal or super-resolution microscopy with consistent exposure parameters across all experimental conditions. For analysis, implement a multi-stage approach: first, create binary masks of mitochondrial networks using appropriate thresholding; second, quantify DRP1-positive puncta that overlap with these mitochondrial masks using co-localization algorithms (Manders' coefficient or Pearson's correlation); third, normalize mitochondria-associated DRP1 signal to total DRP1 expression and mitochondrial content. Employ advanced image analysis platforms like ImageJ with the JACoP plugin or CellProfiler with custom pipelines to automate this process across multiple cells and conditions. For dynamic studies, implement time-lapse imaging with photoactivatable or photoconvertible DRP1 constructs, complemented by fixed-timepoint antibody validation. This comprehensive approach provides robust quantitative data on DRP1 translocation under various experimental conditions .

What are the key considerations for using DRP1 antibodies in tissue-specific contexts?

Applying DRP1 antibodies to diverse tissue contexts requires tissue-specific optimization and validation strategies. Begin by conducting species-compatibility assessment, as antibody epitope recognition may vary across species despite protein conservation. For each tissue type, optimize antigen retrieval methods—heat-induced epitope retrieval using citrate buffer (pH 6.0) is often effective for formalin-fixed paraffin-embedded tissues, while fresh-frozen sections may require milder permeabilization. Tissue-specific autofluorescence requires appropriate quenching methods (e.g., sodium borohydride treatment for brain tissue). Validation should include comparison of multiple DRP1 antibodies targeting different epitopes and correlation with DRP1 mRNA expression in the same tissue regions. For tissues with varying mitochondrial content (e.g., cardiac versus skeletal muscle), normalize DRP1 signal to mitochondrial mass using markers like VDAC or COX IV. Implement blocked peptide controls using the immunizing peptide to confirm specificity. When comparing diseased versus healthy tissues, account for potential changes in tissue architecture and mitochondrial content that might influence interpretation of DRP1 expression or localization patterns .

How can I address non-specific binding issues when using DRP1 antibodies in Western blotting?

Non-specific binding in Western blot applications can be systematically addressed through multiple optimization strategies. First, verify that you're using the appropriate DRP1 antibody concentration—typically 1:500 to 1:2000 dilution for polyclonal antibodies like CAB16661, with overnight incubation at 4°C providing optimal signal-to-noise ratio. Implement stringent blocking procedures using 5% non-fat milk or BSA in TBST for at least 1 hour at room temperature. Consider modifying washing conditions by increasing both the duration (5 washes of 5 minutes each) and stringency (0.1% to 0.3% Tween-20) in TBST buffers. For persistent background issues, incorporate additional blocking agents such as 5% normal serum from the same species as the secondary antibody or commercial background reducers. Validate specificity using DRP1 knockout/knockdown controls alongside positive controls. For high molecular weight non-specific bands, optimize SDS-PAGE conditions using gradient gels (4-15%) to improve separation. When detecting specific DRP1 isoforms or post-translationally modified forms, use purified recombinant proteins as reference standards. These systematic approaches will help distinguish specific DRP1 signal from background or cross-reactive bands .

What strategies can resolve discrepancies between DRP1 antibody-based detection methods and functional assays?

Resolving discrepancies between antibody detection and functional data requires systematic troubleshooting and integration of complementary approaches. First, determine whether the discrepancy relates to expression levels, localization, or activity of DRP1. For expression discrepancies, compare protein detection using multiple antibodies targeting different DRP1 epitopes alongside mRNA quantification via qRT-PCR. When localization data conflicts with functional outcomes, implement subcellular fractionation to biochemically validate immunofluorescence results, and consider that DRP1 activity may not correlate directly with localization due to post-translational regulation. For activity-related discrepancies, assess DRP1 GTPase activity using purified protein or immunoprecipitated complexes, comparing these results with indirect measures such as mitochondrial morphology analysis. Timing differences may explain apparent contradictions—monitor temporal dynamics using time-course experiments with both antibody detection and functional assays. Consider the effects of experimental conditions (cell confluence, serum levels, metabolic state) on both detection sensitivity and DRP1 function. Finally, integrate multiple methodologies by correlating antibody-based detection with functional assays at the single-cell level using microscopy-based approaches to directly link DRP1 status with mitochondrial phenotypes .

How do I validate DRP1 antibody specificity across diverse experimental systems?

Comprehensive validation of DRP1 antibody specificity across experimental systems requires a multi-faceted approach. Begin with genetic validation using CRISPR/Cas9-mediated DRP1 knockout or siRNA-mediated knockdown in each model system, confirming signal reduction or elimination. Implement epitope competition assays by pre-incubating the antibody with excess immunizing peptide, which should abolish specific binding. For cross-species applications, align the target epitope sequence across species to predict potential cross-reactivity and validate experimentally in each species model. When comparing different tissues or cell types, implement parallel detection methods including immunoblotting, immunofluorescence, and flow cytometry to confirm consistent target recognition. For absolute specificity assessment, perform immunoprecipitation followed by mass spectrometry to identify all proteins pulled down by the antibody. Create validation panels showing antibody performance across different detection methods, fixation conditions, and experimental systems, documenting optimal working conditions for each. This comprehensive validation approach ensures reliable DRP1 detection and facilitates accurate data interpretation across diverse experimental contexts .

How can DRP1 antibodies be used to investigate the link between mitochondrial dynamics and cancer progression?

Investigating DRP1's role in cancer progression requires specialized antibody-based approaches across multiple experimental systems. Begin with comparative immunohistochemistry analysis of DRP1 expression and phosphorylation patterns in tumor versus adjacent normal tissue microarrays, correlating with clinical staging and patient outcomes. Implement spectral unmixing immunofluorescence to simultaneously visualize DRP1, mitochondrial morphology, proliferation markers, and metabolic sensors in cancer tissue sections. In cancer cell models, use phospho-specific DRP1 antibodies (particularly pSer616) to track activation status in response to oncogenic signaling, correlating with mitochondrial fragmentation analysis via high-content imaging. For functional validation, combine DRP1 knockdown or pharmacological inhibition with mitochondrial function assays (respiration, membrane potential) and cancer hallmark assessments (migration, invasion, resistance to apoptosis). In three-dimensional tumor spheroids or patient-derived xenografts, analyze spatial gradients of DRP1 activation in correlation with hypoxia markers and metabolic zonation. This multi-level approach connects DRP1-mediated mitochondrial dynamics with cancer cell adaptations to stress, providing insights into potential therapeutic targeting strategies .

What methodological approaches can assess DRP1 involvement in neurodegeneration using patient-derived samples?

Investigating DRP1's contribution to neurodegeneration using patient samples requires specialized methodological approaches that accommodate limited tissue availability and disease heterogeneity. For post-mortem brain tissue, implement multiplexed immunohistochemistry to simultaneously visualize DRP1, phospho-DRP1, mitochondrial markers, and disease-specific proteins (tau, α-synuclein, etc.) in affected brain regions. Quantify co-localization patterns using digital pathology platforms with machine learning algorithms to identify disease-specific signatures across multiple patients. From patient-derived fibroblasts or peripheral blood mononuclear cells, assess baseline differences in DRP1 expression, phosphorylation, and mitochondrial recruitment using flow cytometry and high-content imaging. Generate induced pluripotent stem cell (iPSC)-derived neurons from patients and controls to examine disease-relevant phenotypes, applying DRP1 antibodies in live-cell compatible formats to track dynamics in real-time. For functional validation, combine mitochondrial morphology analysis with bioenergetic profiling and stress response assays, correlating with DRP1 status. In all patient-derived systems, implement age and gender-matched controls and account for medication history. These approaches collectively provide mechanistic insights into DRP1's contribution to neurodegeneration while accommodating the complexities of human samples .

How can DRP1 antibodies be used to evaluate potential therapeutic approaches targeting mitochondrial dynamics?

Evaluating therapeutics targeting mitochondrial dynamics requires comprehensive antibody-based assessment protocols across multiple model systems. Develop a multi-parameter screening platform using high-content imaging with DRP1 antibodies to quantify drug effects on DRP1 expression, phosphorylation status, and mitochondrial localization. Implement dose-response and time-course analyses to establish pharmacodynamic relationships between compound exposure, DRP1 modulation, and downstream effects on mitochondrial morphology. For mechanistic validation, combine phospho-specific DRP1 antibodies with kinase activity assays to confirm target engagement of compounds affecting DRP1-regulatory pathways. In disease models, correlate therapeutic efficacy with normalization of DRP1 function using quantitative immunofluorescence or biochemical fractionation approaches. For in vivo validation, develop tissue-specific immunohistochemistry protocols to assess DRP1 modulation in target organs following compound administration, correlating with functional outcomes and biomarker responses. When developing therapeutic antibodies targeting DRP1 directly, use competition assays with detection antibodies to confirm binding to the intended epitopes. These methodological approaches provide robust assessment of compound effects on DRP1-mediated processes and facilitate translation of mitochondrial dynamics-targeting therapeutics into clinical applications .

What are the optimal conditions for using DRP1 antibodies in co-immunoprecipitation studies investigating protein interaction networks?

Optimizing DRP1 co-immunoprecipitation requires careful consideration of buffer composition and experimental conditions to preserve physiologically relevant protein interactions. Begin with gentle lysis buffers containing 0.5-1% non-ionic detergents (NP-40 or Digitonin) rather than stronger detergents like SDS that may disrupt protein-protein interactions. Include 150-300 mM NaCl to reduce non-specific binding while maintaining specific interactions. Incorporate protease inhibitors, phosphatase inhibitors, and GTPγS (a non-hydrolyzable GTP analog) to preserve DRP1's interaction state. Pre-clear lysates with appropriate control beads and pre-immune serum to reduce background. For the immunoprecipitation step, use polyclonal antibodies like CAB16661 at 2-5 μg per mg of protein lysate, with overnight incubation at 4°C under gentle rotation. For detecting transient or weak interactions, implement crosslinking with membrane-permeable crosslinkers prior to lysis. Validate interactions using reciprocal immunoprecipitation and assess specificity using DRP1-knockout controls. When investigating interactions affected by post-translational modifications, use phospho-specific antibodies for the immunoprecipitation step to enrich specifically modified DRP1 complexes. These optimized conditions will facilitate detection of the dynamic interactome surrounding DRP1 under various cellular conditions .

How can I implement super-resolution microscopy techniques with DRP1 antibodies to visualize mitochondrial fission events?

Implementing super-resolution microscopy with DRP1 antibodies requires specialized sample preparation and imaging protocols to capture fission events below the diffraction limit. Begin with optimal fixation using 4% paraformaldehyde supplemented with 0.1% glutaraldehyde for 15 minutes to preserve ultrastructural details without compromising antigenicity. For Structured Illumination Microscopy (SIM), use high-quality primary antibodies at optimized concentrations (typically 1:100 to 1:500) and fluorophore-conjugated secondary antibodies with brightness and photostability suitable for super-resolution (Alexa Fluor 488, 568, or 647). For STORM/PALM applications, use secondary antibodies conjugated to photoswitchable fluorophores and implement appropriate imaging buffers containing oxygen scavenging systems and reducing agents. Co-label with outer mitochondrial membrane markers positioned distinctly from DRP1 binding sites to prevent steric hindrance. For live-cell super-resolution, consider implementing genetically encoded tags (HaloTag, SNAP-tag) fused to DRP1 with cell-permeable fluorescent ligands as alternatives to antibodies. Optimize acquisition parameters including exposure time, laser power, and frame rate to balance resolution with photobleaching. For data analysis, implement advanced segmentation algorithms to track DRP1 puncta formation at constriction sites and correlate with subsequent fission events. This approach enables nanoscale visualization of DRP1 assembly and function during mitochondrial division .

What strategies can be employed for multiplexed detection of DRP1 and other mitochondrial dynamics proteins?

Implementing multiplexed detection of mitochondrial dynamics proteins requires specialized approaches to overcome antibody compatibility limitations and spectral overlap constraints. For immunofluorescence, employ sequential staining protocols using antibodies from different host species (rabbit anti-DRP1, mouse anti-Mfn1/2, goat anti-Fis1) with species-specific secondary antibodies. When antibodies derive from the same species, implement tyramide signal amplification (TSA) with sequential rounds of staining, detection, and inactivation. For fixed tissue analysis, consider cyclic immunofluorescence (CycIF) with iterative antibody application, imaging, and stripping to detect >10 targets on the same specimen. Implement spectral unmixing algorithms when using fluorophores with overlapping emission spectra to increase multiplexing capacity. For mass cytometry applications, conjugate DRP1 antibodies with rare earth metals and combine with other metal-labeled antibodies against mitochondrial proteins for highly multiplexed single-cell analysis. When examining subcellular localization patterns, combine antibody-based detection with organelle-specific dyes or genetically encoded markers in spectrally distinct channels. For proximity-based interaction studies, implement proximity ligation assays (PLA) using antibody pairs targeting DRP1 and potential interaction partners. These multiplexed approaches provide comprehensive insights into the interplay between DRP1 and other components of the mitochondrial dynamics machinery .

How can DRP1 antibodies be integrated with emerging spatial transcriptomics approaches to correlate protein localization with gene expression patterns?

Integrating DRP1 protein detection with spatial transcriptomics requires innovative methodological approaches bridging antibody-based imaging with nucleic acid analysis. Implement sequential immunofluorescence and in situ hybridization protocols, first detecting DRP1 and mitochondrial markers using antibodies, followed by fixation and detection of relevant transcripts (DNM1L, MFN1/2, FIS1) using RNAscope or single-molecule FISH. For tissue sections, combine multiplexed immunofluorescence for DRP1 with spatial transcriptomics platforms like Visium or GeoMx DSP, which preserve spatial context while enabling region-specific transcriptomic analysis. Develop computational pipelines to register immunofluorescence images with spatial transcriptomics data, allowing correlation between DRP1 protein distribution and expression patterns of genes involved in mitochondrial dynamics. For single-cell resolution, implement CITE-seq approaches using oligonucleotide-tagged DRP1 antibodies in combination with single-cell RNA sequencing to simultaneously profile protein levels and transcriptome in individual cells. These integrated approaches provide unprecedented insights into the relationship between transcriptional regulation and protein-level changes in DRP1 and associated factors across different tissue regions and cell types, particularly valuable for understanding disease-related alterations in spatially complex tissues like brain or tumor samples .

What are the considerations for developing and validating phospho-specific DRP1 antibodies for dynamic studies of mitochondrial fission regulation?

Developing phospho-specific DRP1 antibodies requires rigorous design and validation strategies to ensure specificity and sensitivity for dynamic regulation studies. Begin with careful epitope selection, focusing on key regulatory phosphorylation sites (Ser616, Ser637/Ser656) with surrounding sequences that are unique within the proteome. Generate phospho-peptides containing the modified residue plus 7-10 flanking amino acids for immunization, and implement dual-purification strategies: first affinity-purify against the phospho-peptide, then deplete antibodies recognizing the non-phosphorylated form. Validate specificity using multiple approaches: Western blotting of samples treated with kinase activators versus inhibitors, lambda phosphatase treatment controls, and cells expressing phospho-mimetic (S→D) versus phospho-deficient (S→A) DRP1 mutants. For dynamic studies, optimize antibody performance in multiple applications beyond Western blotting, including immunofluorescence, flow cytometry, and ELISA-based quantification. Develop standardized protocols with careful consideration of fixation methods that preserve phosphorylation status (include phosphatase inhibitors throughout). Create validation panels showing antibody performance across various stimuli known to modulate the specific phosphorylation site. These rigorously validated phospho-specific antibodies enable precise monitoring of DRP1 activation states in response to various physiological and pathological stimuli .

How can computational image analysis be optimized for quantitative assessment of DRP1 distribution and function using antibody-based detection methods?

Optimizing computational image analysis for DRP1 studies requires specialized algorithms and validation protocols to extract biologically meaningful quantitative data. Develop automated pipelines incorporating machine learning-based segmentation of mitochondrial networks from markers like TOM20, followed by detection and classification of DRP1 puncta based on size, intensity, and morphology. Implement 3D analysis incorporating z-stack information to account for the complex three-dimensional organization of mitochondrial networks. For temporal studies, develop tracking algorithms that follow individual DRP1 puncta over time, correlating their dynamics with subsequent mitochondrial fission events. Quantify population-level metrics including DRP1 cluster density per mitochondrial surface area, percentage of mitochondria with DRP1 recruitment, and spatial distribution patterns of DRP1 along mitochondrial networks. Implement correlation analyses between DRP1 recruitment and local mitochondrial parameters such as membrane potential, ROS production, or calcium levels using multiplexed imaging data. For reproducibility and standardization, develop open-source analysis modules with comprehensive documentation and validation datasets. Validate computational measurements against manual quantification by expert observers across diverse experimental conditions and cell types. These advanced analysis approaches transform antibody-based imaging from qualitative observation to quantitative assessment of DRP1 function in mitochondrial dynamics regulation .

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.