RHOT2 antibodies target the RHOT2 gene product, a 68 kDa GTPase localized to the outer mitochondrial membrane. RHOT2 regulates mitochondrial transport, fusion-fission dynamics, and stress responses by interacting with motor/adaptor complexes (e.g., kinesin, dynein, TRAK1/2) and signaling pathways involving PINK1/Parkin . These antibodies enable detection of RHOT2 in diverse experimental models, aiding studies on cancer, neurodegenerative diseases, and mitochondrial biology.
RHOT2 is overexpressed in metastatic prostate cancer and correlates with poor survival .
Depleting RHOT2 impairs prostate cancer cell growth in vitro and in xenografts by disrupting the GCN1-GCN2-ATF4 signaling axis .
Mutant RHOT2 (159L) enhances GCN1 binding, promoting tumor progression .
Aberrant RHOT2 splicing events (e.g., RHOT2-32938-RI) correlate with renal cell carcinoma (KIRC) metastasis and poor prognosis .
RHOT2 dysregulation impacts sphingolipid metabolism and glycosylation pathways, suggesting novel therapeutic targets .
RHOT2 degradation by PINK1/Parkin halts mitochondrial transport during mitophagy, implicating it in Parkinson’s disease .
RHOT2 (Ras homolog gene family member T2), also known as MIRO-2 or hMiro-2, belongs to the mitochondrial Rho GTPase family. It is a crucial protein localized to the outer membrane of mitochondria and plays key roles in mitochondrial dynamics and transport. RHOT2 antibodies are essential tools for investigating mitochondrial function and dynamics due to their ability to specifically detect this protein in various experimental contexts. Current research indicates RHOT2 is involved in calcium-dependent regulation of mitochondrial movement and may have implications in neurodegenerative conditions such as Parkinson's disease . When designing experiments to study mitochondrial transport, RHOT2 antibodies enable visualization of this critical protein through multiple detection methods including Western blotting, immunofluorescence, and immunohistochemistry .
Monoclonal and polyclonal RHOT2 antibodies differ significantly in their experimental utility and application specificity:
Monoclonal RHOT2 antibodies (like 68469-1-Ig):
Recognize a single epitope on the RHOT2 protein, providing high specificity
Demonstrate consistent lot-to-lot reproducibility, critical for longitudinal studies
Typically show narrower reactivity across species (often human-specific)
Optimal for applications requiring precise epitope targeting, as in the case of 68469-1-Ig which is validated primarily for Western blot applications with dilution ranges of 1:5000-1:50000
Polyclonal RHOT2 antibodies (like 11237-1-AP):
Recognize multiple epitopes on the RHOT2 protein, offering higher sensitivity
Show broader cross-reactivity across species (human, mouse, rat)
Particularly effective for applications requiring signal amplification
Can be used across multiple applications including WB (1:1000-1:10000), IHC (1:50-1:500), IF/ICC (1:50-1:500), and IP (0.5-4.0 μg)
When designing experiments requiring detection of potentially low abundance RHOT2 in tissue samples, polyclonal antibodies often provide better sensitivity, while monoclonal antibodies offer advantages in experiments requiring consistent epitope recognition over time.
Proper storage and handling of RHOT2 antibodies is critical for maintaining their specificity and sensitivity in experimental applications. Based on manufacturer recommendations across multiple products:
Storage conditions:
Most formulations remain stable for one year after shipment when properly stored
Antibody 60464-3-PBS requires storage at -80°C due to its PBS-only buffer formulation
Avoid repeated freeze-thaw cycles which can lead to antibody degradation and loss of activity
Buffer formulations:
Most RHOT2 antibodies are provided in PBS with 0.02% sodium azide and 50% glycerol at pH 7.3
This buffer composition helps maintain stability during freeze-thaw cycles
For antibodies requiring conjugation (like 60464-3-PBS), azide-free and BSA-free formulations are available
For experimental methodologies requiring maximum antibody performance, consider the following research-validated approaches:
Aliquot antibodies upon receipt to minimize freeze-thaw cycles
Use sterile technique when handling to prevent microbial contamination
Centrifuge antibody vials briefly before opening to collect solution at the bottom of the vial
When working with diluted antibody solutions, prepare them fresh for each experiment
These handling procedures are essential for maintaining detection sensitivity, particularly in experiments targeting endogenous RHOT2 expression levels in primary cells or tissue samples.
Since RHOT2 is a mitochondrial outer membrane protein, sample preparation must preserve its native localization while allowing antibody accessibility. The following methodological approaches have been validated in research settings:
For Western blot applications:
Cell lysis should be performed using mild detergents (like 1% Triton X-100) that preserve membrane protein integrity
Include protease inhibitors in lysis buffers to prevent degradation
Enrichment of mitochondrial fractions may enhance detection sensitivity
The observed molecular weight of RHOT2 ranges from 68-80 kDa depending on post-translational modifications
For immunofluorescence/immunocytochemistry:
Mild fixation (4% paraformaldehyde for 10-15 minutes) preserves RHOT2 epitopes
Brief permeabilization (0.1-0.2% Triton X-100 for 5-10 minutes) allows antibody access without extracting membrane proteins
Co-staining with established mitochondrial markers (e.g., TOM20, MitoTracker) confirms specificity of RHOT2 localization
RHOT2 antibodies like 11237-1-AP have been validated for IF/ICC at dilutions of 1:50-1:500
For immunohistochemistry on fixed tissue:
Antigen retrieval is critical; both citrate buffer (pH 6.0) and TE buffer (pH 9.0) methods have been validated
For 11237-1-AP and NBP2-93326 antibodies, high-pressure antigen retrieval with 10mM citrate buffer (pH 6.0) is recommended
Optimal dilutions range from 1:50-1:500 depending on the specific antibody and tissue type
When performing co-localization studies, these methodological considerations are essential for accurate determination of RHOT2 interactions with other mitochondrial or cytoskeletal proteins.
Phosphosite-specific antibodies represent an advanced approach for studying RHOT2 regulation through post-translational modifications. Building on methodologies developed for other membrane proteins like dopamine receptors , researchers can employ similar techniques for RHOT2:
Methodological approach:
Identification of putative phosphorylation sites through phosphoproteomic analysis or prediction algorithms
Generation of phospho-specific antibodies using double phosphorylated peptides corresponding to specific regions of RHOT2
Validation using phosphatase treatments to confirm specificity for phosphorylated versus non-phosphorylated forms
Application in Western blotting to monitor changes in phosphorylation state under different cellular conditions
For RHOT2 regulation studies, researchers can adapt the approach used for D2R phosphorylation detection:
Use λ-phosphatase treatment as a negative control to confirm phospho-specificity
Compare basal versus stimulated phosphorylation states
Employ kinase inhibitors to identify responsible kinases
A sample experimental design based on D2R phosphorylation study methodologies :
| Treatment | Detection Method | Expected Outcome | Control |
|---|---|---|---|
| Basal conditions | Phospho-RHOT2 antibody | Baseline signal | Total RHOT2 |
| Calcium influx | Phospho-RHOT2 antibody | Increased signal | Total RHOT2 |
| Kinase inhibitor + Calcium | Phospho-RHOT2 antibody | Reduced signal | Total RHOT2 |
| λ-phosphatase | Phospho-RHOT2 antibody | Abolished signal | Total RHOT2 |
This methodological framework enables researchers to investigate how RHOT2 phosphorylation states affect mitochondrial dynamics and transport in response to calcium signaling and other cellular pathways.
When researchers encounter conflicting results between different detection methods using RHOT2 antibodies, systematic troubleshooting and validation approaches are essential:
Epitope accessibility differences:
Western blot detects denatured proteins while IHC/IF detect proteins in their native conformation
Use antibodies targeting different epitopes for cross-validation
The RHOT2 antibody 11237-1-AP (targeting fusion protein Ag1752) may detect different epitopes than 68469-1-Ig (targeting fusion protein Ag29690)
Validation through genetic approaches:
Cross-platform validation methodology:
| Detection Method | Sample Preparation | Epitope Concern | Validation Approach |
|---|---|---|---|
| Western Blot | Denatured protein | Linear epitopes | RHOT2-KO control lysate |
| Immunofluorescence | Fixed cells/tissues | Conformational | Co-localization with mitochondrial markers |
| Immunohistochemistry | Fixed, embedded tissue | Masked epitopes | Antigen retrieval optimization |
| Immunoprecipitation | Native protein | Accessible surface | Pre-clearing optimization |
Application-specific considerations:
For Western blot: The observed molecular weight may vary (68-80 kDa) based on post-translational modifications
For IHC: Antigen retrieval methods significantly impact epitope accessibility; both citrate buffer (pH 6.0) and TE buffer (pH 9.0) have been validated
For IP applications: Pre-clearing lysates and optimizing antibody concentrations (0.5-4.0 μg for 1.0-3.0 mg of protein) are critical
By implementing these systematic validation strategies, researchers can resolve discrepancies and ensure reliable RHOT2 detection across experimental platforms.
RHOT2 has emerged as a potential factor in Parkinson's disease (PD) pathogenesis through its role in mitochondrial transport and quality control. Researchers can employ RHOT2 antibodies in PD research through the following methodological approaches:
Genetic association studies have investigated RHOT2 variation in PD risk:
The search results indicate that RHOT2 genetic variation has been analyzed in relation to PD risk, though cumulative effects were inconsistent across different statistical tests (N variants = 40; CMC p = 0.044, Zeggini p = 0.166, MB p = 0.404, SKAT p = 0.125, SKAT-O p = 0.210, Fp p = 0.659)
This genetic foundation provides rationale for protein-level investigations
Experimental design for RHOT2 protein analysis in PD models:
Compare RHOT2 expression levels between control and PD patient-derived samples using validated antibodies
Analyze RHOT2 localization in relation to damaged mitochondria in neurotoxin-based or genetic PD models
Investigate RHOT2 interactions with PINK1/Parkin pathway components using co-immunoprecipitation with antibodies validated for this application like 11237-1-AP
Tissue-specific analysis strategy:
The RHOT2 antibody 11237-1-AP has been validated for detection in multiple human tissues including colon, kidney, heart, and lung cancer tissues
RHOT2 antibody NBP2-93326 has been validated for IHC in rat kidney and mouse brain tissues, making it suitable for neurological research
These validations enable comparative studies between brain regions affected in PD versus unaffected regions
Cell-type specific analysis:
RHOT2 antibodies have been validated in multiple cell lines including neuronal models
Use immunofluorescence to compare RHOT2 distribution in dopaminergic versus non-dopaminergic neurons
Combine with markers for mitochondrial fragmentation to assess correlation with disease pathology
This methodological framework enables researchers to investigate whether RHOT2 dysfunction contributes to the mitochondrial transport defects observed in PD pathogenesis, potentially revealing new therapeutic targets.
When investigating RHOT2 protein interactions in disease contexts, rigorous controls are essential for ensuring experimental validity and reproducibility:
Antibody specificity controls:
Co-immunoprecipitation controls:
The 11237-1-AP antibody has been validated for co-immunoprecipitation (CoIP) applications
Include IgG-matched isotype controls for non-specific binding assessment
Reverse Co-IP (using antibodies against the interacting partner to pull down RHOT2)
Input controls (5-10% of lysate used for IP) to verify protein expression
Disease-specific experimental design:
| Disease Context | Positive Control | Negative Control | Validation Method |
|---|---|---|---|
| Parkinson's Disease | PINK1/Parkin interactions | RHOT2 knockdown | Reciprocal Co-IP |
| Mitochondrial dysfunction | Known RHOT2 interactors (e.g., TRAK1/2) | Competition with excess peptide | Proximity ligation assay |
| Neurodegeneration | Age-matched healthy tissue | Non-neuronal tissue | Mass spectrometry validation |
Methodological controls for phosphorylation studies:
Cell type and subcellular localization controls:
Include mitochondrial markers (TOM20, MitoTracker) to confirm RHOT2 localization
Use mitochondrial fractionation to enrich for RHOT2 and its interacting partners
Compare disease-relevant cell types with non-relevant cells as biological controls
By implementing these systematic controls, researchers can ensure that observed RHOT2 interactions are specific, reproducible, and relevant to the disease context under investigation.
Detecting endogenous versus overexpressed RHOT2 requires different optimization strategies due to expression level differences. The following methodological guidelines address common challenges:
For endogenous RHOT2 detection:
Antibody selection and dilution:
Sample preparation:
Enrich for mitochondrial fractions to concentrate RHOT2 content
Load higher protein amounts (40-80 μg) for tissue samples
Include phosphatase inhibitors to preserve post-translational modifications
Detection system:
For overexpressed RHOT2 detection:
Antibody dilution:
Sample preparation:
Load less protein (10-20 μg) to prevent signal saturation
Include untransfected controls for comparison
Short exposure times to prevent oversaturation
Special considerations:
Monitor for potential aggregation or degradation of overexpressed protein
Verify subcellular localization matches endogenous pattern through fractionation
Common troubleshooting approach:
These methodological approaches ensure accurate detection of RHOT2 regardless of expression level context.
Detecting RHOT2 in primary neurons requires careful optimization due to the complex morphology and sensitivity of neuronal cells. Based on validated antibody protocols and neuronal imaging best practices:
Fixation and permeabilization parameters:
Mild fixation: 4% paraformaldehyde for 10-15 minutes at room temperature
Gentle permeabilization: 0.1% Triton X-100 for 5-10 minutes to preserve mitochondrial membrane integrity
Alternative: 100% ice-cold methanol for 10 minutes for simultaneous fixation and permeabilization
Antibody selection and optimization:
Neuronal-specific considerations:
Block with 5-10% normal serum plus 1% BSA to reduce non-specific binding
Include cytoskeletal markers (MAP2, Tau) to visualize neuronal processes
Co-stain with mitochondrial markers (TOM20, MitoTracker) to confirm RHOT2 localization
Image both cell bodies and distal neurites to assess mitochondrial distribution
Advanced imaging parameters:
Perform z-stack acquisition (0.3-0.5 μm steps) to capture three-dimensional distribution
Use deconvolution algorithms to improve resolution of individual mitochondria
Consider super-resolution techniques for detailed mitochondrial morphology analysis
Critical controls:
Include RHOT2 siRNA knockdown neurons as negative controls
Compare with fibroblasts where RHOT2 detection has been well-established
Process control samples with primary antibody omission
Methodological validation table for neuronal RHOT2 imaging:
| Parameter | Recommendation | Rationale | Troubleshooting |
|---|---|---|---|
| Antibody dilution | Start at 1:50-1:100 | Higher concentration needed for neuronal detection | If high background, increase dilution gradually |
| Blocking duration | 1-2 hours at room temperature | Reduces non-specific binding in neurons | Extend to overnight at 4°C for problematic samples |
| Primary antibody incubation | Overnight at 4°C | Improves penetration and specific binding | Can extend to 48 hours for thick sections |
| Secondary antibody | Low cross-reactivity, highly adsorbed | Reduces background in sensitive neuronal preparations | Pre-adsorb if necessary |
| Mounting medium | Anti-fade with DAPI | Preserves signal during extended imaging sessions | Allow complete curing before imaging |
By carefully optimizing these parameters, researchers can achieve specific and sensitive detection of RHOT2 in primary neurons, enabling detailed analysis of mitochondrial dynamics in neuronal compartments.