GHITM regulates mitochondrial cristae structure and cytochrome c release. The antibody has been used to demonstrate GHITM’s role in maintaining mitochondrial integrity and delaying apoptosis by cross-linking cytochrome c to the inner mitochondrial membrane .
In kidney renal clear cell carcinoma (KIRC), GHITM downregulation correlates with poor prognosis. Key findings using this antibody include:
Proliferation Inhibition: GHITM overexpression reduced KIRC cell growth in vitro and in vivo (xenograft models) .
Metastasis Suppression: GHITM upregulation decreased migration, invasion, and lung metastasis in murine models .
Notch Signaling: GHITM suppresses Notch1, a pro-tumorigenic pathway, and enhances PD-1 blockade efficacy in KIRC .
WB: Detects GHITM in mouse liver, HEK-293, HeLa, and SH-SY5Y cells .
IHC: Localizes GHITM in mouse brain tissue with TE buffer pH 9.0 antigen retrieval .
Functional Studies: Confirmed GHITM’s role in mitochondrial hyperpolarization and ROS regulation .
GHITM, also known as MICS1, TMBIM5, or DERP2, is a mitochondrial protein that localizes to the inner membrane. It plays significant roles in maintaining mitochondrial homeostasis and morphology, particularly in specific cristae structures. GHITM is involved in the apoptotic release of cytochrome c from mitochondria, positioning it as an important regulatory protein in cell death pathways .
Recent research has revealed that GHITM functions in limiting mitochondrial hyperpolarization and reactive oxygen species (ROS) production . Additionally, studies have demonstrated that GHITM can impact the mitochondrial protein synthesis machinery to sustain the structure, shape, and function of mitochondria . These properties make GHITM a critical target for researchers investigating mitochondrial dynamics and cellular stress responses.
Methodologically, when studying GHITM's functions, researchers should consider using both gain-of-function (overexpression) and loss-of-function (knockdown/knockout) approaches to comprehensively characterize its biological effects in specific experimental contexts.
GHITM antibodies have been validated for multiple research applications, with varying levels of optimization required for each technique:
When selecting the appropriate application, researchers should consider:
The specific biological question being addressed
Sample type and availability
Required sensitivity and specificity
The need for quantitative (ELISA, WB) versus qualitative or localization data (IHC, IF)
Sample preparation varies by sample type and intended application. Follow these methodological guidelines for consistent results:
Serum/Plasma Preparation:
For serum: Allow samples to clot for 2 hours at room temperature or overnight at 2-8°C
Centrifuge at approximately 1000 × g (or 3000 rpm) for 15 minutes
Remove serum and assay immediately or aliquot and store at -20°C or -80°C
Tissue Homogenate Preparation:
Thoroughly rinse tissues in ice-cold PBS (0.02 mol/L, pH 7.0-7.2)
Mince tissues into small pieces and homogenize in PBS using a glass homogenizer on ice
Subject the suspension to ultrasonication or two freeze-thaw cycles to further break down cell membranes
Centrifuge for 15 minutes at 1500 × g (or 5000 rpm)
Remove the supernatant for immediate assay or aliquot and store at -20°C or -80°C
Cell Lysate Preparation:
For adherent cells: Detach with trypsin and collect by centrifugation
Proceed with appropriate lysis buffer containing protease inhibitors
For Western blotting applications, include phosphatase inhibitors if phosphorylation status is relevant
IHC Sample Preparation:
For paraffin-embedded samples, perform antigen retrieval preferably with TE buffer pH 9.0
Alternatively, use citrate buffer pH 6.0
Block with 10% normal goat serum for 30 minutes at room temperature
When performing Western blot analysis for GHITM, researchers should expect to observe bands at specific molecular weights depending on the sample type and protein modification status:
Calculated molecular weight: 37 kDa (based on 345 amino acids)
Observed molecular weight: primarily 42 kDa, with additional bands at 25-27 kDa
The discrepancy between calculated and observed molecular weights likely results from post-translational modifications or alternative splicing. To ensure specific detection:
Always include positive controls such as HEK-293 cells, HeLa cells, SH-SY5Y cells, or mouse liver tissue, which have been validated for GHITM expression
Consider running a gradient gel (8-15%) to improve separation of bands in the 25-45 kDa range
Validate antibody specificity using GHITM-overexpressing and GHITM-knockdown samples when possible
For loading controls, consider mitochondrial markers when examining mitochondrial fractions
Proper storage and handling of GHITM antibodies is critical for maintaining their reactivity and specificity:
Storage Conditions:
Buffer Composition:
Stability:
Handling Recommendations:
Bring antibody to room temperature before opening
Gently mix by inverting the tube or gentle pipetting (avoid vortexing)
Centrifuge briefly before opening to collect contents at the bottom of the tube
Return to -20°C immediately after use
Work in a clean environment to prevent contamination
GHITM has emerged as a potential biomarker and therapeutic target in kidney renal clear cell carcinoma (KIRC). Recent research has revealed several key aspects of GHITM's role in KIRC that researchers should consider:
Expression Pattern and Clinical Correlation:
GHITM is downregulated in KIRC compared to normal tissues
Aberrant GHITM downregulation correlates with clinicopathological features
Low GHITM expression is associated with unfavorable prognosis in KIRC patients
Diagnostic Value:
GHITM shows significant diagnostic accuracy with AUC = 0.964 in TCGA-KIRC dataset
For early-stage (Stage I) patients, GHITM maintained high diagnostic value:
Methodological Approaches for GHITM Analysis in KIRC:
Transcriptomic analysis: Compare GHITM mRNA levels between tumor and adjacent normal tissues using RT-qPCR
Protein expression analysis: Perform IHC staining on tissue microarrays to correlate GHITM levels with clinicopathological parameters
Survival analysis: Use Kaplan-Meier curves to evaluate the relationship between GHITM expression and patient outcomes
ROC curve analysis: Assess GHITM's potential as a diagnostic biomarker for early KIRC detection
When designing studies to investigate GHITM in KIRC or other cancers, researchers should consider both in vitro cell line models and clinical tissue samples to establish comprehensive evidence of its role.
Investigating GHITM's effects on tumor cell phenotypes requires a multi-faceted experimental approach. Based on recent research, the following methodological strategies are recommended:
Establishing GHITM-Modified Cell Lines:
Generate stable GHITM-overexpressing (GHITM-OE) and control (GHITM-EV) cell lines using lentiviral transduction
Confirm expression levels via Western blot analysis using validated antibodies
Cell Proliferation Assays:
CCK-8 assay: Measure metabolic activity at 24, 48, 72, and 96 hours post-seeding
Colony formation assay: Evaluate long-term proliferative capacity (14-21 days)
EdU incorporation assay: Assess DNA synthesis as a measure of proliferation
Migration and Invasion Assays:
Wound healing assay: Monitor cell migration into scratch area over 24-48 hours
Transwell assay: Quantify invasive capacity through Matrigel-coated membranes
Angiogenesis Assessment:
Tube formation assay: Evaluate the effect of conditioned medium from GHITM-modified cells on endothelial cell tube formation
Morphological analysis: Observe changes in cell shape and structure using phase-contrast microscopy
In Vivo Tumor Models:
Subcutaneous xenograft model: Inject GHITM-OE and control cells into immunodeficient mice to assess tumor growth
Metastasis model: Deliver cells via tail vein injection to evaluate pulmonary metastasis formation
Tumor analysis: Perform IHC staining of xenografts for proliferation markers (Ki-67) and pathway components (e.g., Notch1)
Recent research has shown that GHITM overexpression inhibits KIRC cell proliferation, migration, and invasion both in vitro and in vivo, supporting its potential tumor-suppressive role .
The connection between GHITM and Notch signaling represents an important area for cancer research. To investigate this relationship, researchers should consider these methodological approaches:
Expression Analysis of Notch Pathway Components:
Perform Western blot analysis to detect Notch receptors (Notch1-4) in GHITM-overexpressing and control cells
Use validated antibodies such as:
Rescue Experiments:
Co-express GHITM and Notch1 to determine if Notch1 overexpression rescues the phenotypes induced by GHITM upregulation
Measure endpoints including cell proliferation, migration, and invasion
Notch Pathway Activity Assessment:
Evaluate expression of Notch target genes using RT-qPCR
Use luciferase reporter assays with Notch-responsive elements to measure pathway activation
Assess Notch intracellular domain (NICD) levels by Western blot as an indicator of active Notch signaling
In Vivo Validation:
Perform IHC staining of xenograft tumors to examine the correlation between GHITM and Notch1 expression
Analyze human tumor samples for GHITM and Notch pathway component expression patterns
Mechanistic investigations have revealed that GHITM overexpression induces downregulation of Notch1, which acts as an oncogene in KIRC. Furthermore, the inhibitory effects of GHITM upregulation can be effectively rescued by Notch1 overexpression, confirming the functional relationship between these proteins .
GHITM has recently been identified as a potential modulator of immunotherapy response, particularly related to the PD-1/PD-L1 pathway. Researchers investigating this aspect should consider these methodological strategies:
PD-L1 Expression Analysis:
Perform Western blot analysis using anti-PD-L1 antibodies (e.g., Proteintech, 28076-1-AP) in GHITM-modified cells
Conduct flow cytometry to quantify cell surface PD-L1 expression
Use immunofluorescence to visualize PD-L1 localization patterns
Combination Treatment Studies:
Evaluate the combined effects of GHITM modulation and PD-1 blockade in preclinical models
Test GHITM overexpression in combination with other targeted therapies (e.g., sunitinib)
T Cell Co-culture Experiments:
Co-culture GHITM-modified tumor cells with T cells
Measure T cell activation markers, proliferation, and cytokine production
Assess tumor cell killing in the presence of PD-1 blocking antibodies
In Vivo Immunotherapy Models:
Establish syngeneic tumor models in immunocompetent mice
Treat with anti-PD-1 antibodies alone or in combination with GHITM modulation
Monitor tumor growth, survival, and immune infiltration
Recent research has demonstrated that GHITM can regulate PD-L1 protein abundance and that ectopic overexpression of GHITM enhances the antitumor efficiency of PD-1 blockade in KIRC. These findings suggest that GHITM may serve as a valuable target for improving immunotherapy outcomes .
Understanding the transcriptional regulation of GHITM is important for comprehensive characterization of its role in normal and disease states. Research has identified YY1 as a transcriptional regulator of GHITM, and the following approaches can be used to study this and other regulatory mechanisms:
Promoter Analysis:
Perform in silico analysis of the GHITM promoter region to identify potential transcription factor binding sites
Generate luciferase reporter constructs containing the GHITM promoter region
Test the effects of candidate transcription factors (e.g., YY1) on reporter activity
Chromatin Immunoprecipitation (ChIP):
Use ChIP assays to confirm direct binding of transcription factors to the GHITM promoter
Apply ChIP-seq for genome-wide analysis of transcription factor binding patterns
Transcription Factor Modulation:
Overexpress or knock down candidate transcription factors (e.g., YY1)
Measure changes in GHITM mRNA and protein levels
Use Western blot with anti-GHITM (e.g., Proteintech, 16296-1-AP) and anti-YY1 (e.g., Proteintech, 22156-1-AP) antibodies
Epigenetic Analysis:
Investigate DNA methylation patterns in the GHITM promoter region
Examine histone modifications associated with active/repressed transcription
Test the effects of epigenetic modifying drugs on GHITM expression
Recent research has demonstrated that YY1 can decrease GHITM levels by binding to its promoter. This finding suggests that transcriptional regulation by YY1 may be an important mechanism controlling GHITM expression in cancer contexts .
GHITM is known to play a significant role in maintaining mitochondrial homeostasis and morphology. To investigate these functions, researchers should consider these methodological approaches:
Mitochondrial Morphology Analysis:
Perform live-cell imaging using mitochondrial-specific dyes (e.g., MitoTracker)
Use confocal microscopy to visualize mitochondrial network structure
Quantify morphological parameters (length, branching, fragmentation) using specialized software
Mitochondrial Membrane Potential Assessment:
Use potential-sensitive dyes (e.g., TMRM, JC-1) to measure mitochondrial membrane potential
Evaluate changes in mitochondrial hyperpolarization in response to GHITM modulation
Reactive Oxygen Species (ROS) Measurement:
Employ fluorescent probes (e.g., DCFDA, MitoSOX) to quantify cellular and mitochondrial ROS levels
Assess the impact of GHITM on ROS production under basal and stressed conditions
Cytochrome c Release Assays:
Fractionate cells into cytosolic and mitochondrial components
Detect cytochrome c distribution by Western blot
Evaluate how GHITM modulation affects apoptotic release of cytochrome c
Mitochondrial Protein Synthesis:
Use pulse-chase labeling with radioactive amino acids to measure mitochondrial protein synthesis rates
Analyze the composition of respiratory complexes by blue native PAGE
Assess the impact of GHITM on mitochondrial translation machinery
Previous research has demonstrated that GHITM can limit mitochondrial hyperpolarization and ROS production. Interestingly, in KIRC cell lines, GHITM overexpression did not significantly affect ROS generation or the proportion of apoptotic cells, suggesting context-dependent functions that warrant further investigation .
When working with samples that may have low GHITM expression levels, researchers can employ several strategies to enhance detection sensitivity:
Western Blot Optimization:
Increase protein loading (up to 50-80 μg per lane)
Use higher primary antibody concentration (1:200 dilution)
Extend primary antibody incubation time (overnight at 4°C)
Switch to more sensitive detection systems (e.g., chemiluminescent substrates with enhanced sensitivity)
Use PVDF membranes instead of nitrocellulose for improved protein binding
IHC Sensitivity Enhancement:
Optimize antigen retrieval (TE buffer pH 9.0 is recommended)
Increase primary antibody concentration (1:20-1:50 dilution)
Employ signal amplification systems (e.g., tyramide signal amplification)
Use polymer-based detection systems instead of traditional ABC methods
ELISA Optimization:
Increase sample volume or concentration
Extend incubation times
Optimize buffer conditions
Use high-sensitivity substrates with extended development time
Sample Enrichment Strategies:
Perform subcellular fractionation to concentrate mitochondrial proteins
Use immunoprecipitation to enrich for GHITM before detection
Consider tissue or cell types with known higher GHITM expression as positive controls
When optimizing detection methods, always include appropriate positive and negative controls to ensure that enhanced sensitivity does not come at the cost of specificity.
Researchers working with GHITM antibodies and studying GHITM function may encounter several challenges. Here are common pitfalls and their solutions:
Antibody Cross-Reactivity:
Problem: Non-specific binding leading to false-positive signals
Solution: Validate antibody specificity using GHITM knockout/knockdown controls
Alternative approach: Use multiple antibodies targeting different epitopes
Inconsistent Molecular Weight Detection:
Problem: Variable band patterns in Western blots (42 kDa vs. 25-27 kDa)
Solution: Use gradient gels for better separation and characterize band patterns in positive control samples
Note: The observed molecular weight (42 kDa) differs from the calculated weight (37 kDa), likely due to post-translational modifications
Mitochondrial Localization Challenges:
Problem: Difficulty in distinguishing GHITM from other mitochondrial proteins
Solution: Perform co-localization studies with established mitochondrial markers
Alternative approach: Use super-resolution microscopy for detailed localization analysis
Context-Dependent Function Interpretation:
Problem: GHITM's functions may vary across different cell types or disease contexts
Solution: Include multiple cell lines and primary samples in functional studies
Alternative approach: Use inducible expression/knockdown systems to study acute versus chronic effects
Reproducibility Across Model Systems:
Problem: Findings in cell lines may not translate to in vivo models
Solution: Validate key findings in multiple model systems (cell lines, patient-derived xenografts, clinical samples)
Alternative approach: Consider species differences when using mouse models, as GHITM antibodies may have different reactivity patterns
By anticipating these challenges and implementing appropriate controls and validation steps, researchers can enhance the reliability and reproducibility of their GHITM-related investigations.