The MTCH2 Antibody is a polyclonal rabbit IgG antibody (UniProt ID: Q9Y6C9) designed to target human and rat MTCH2, a 33 kDa protein located on the mitochondrial outer membrane. It is widely used to investigate MTCH2's roles in mitochondrial function, metabolism, and cancer progression .
Non-Small Cell Lung Cancer (NSCLC): MTCH2 knockdown via CRISPR/Cas9 or shRNA reduced cell proliferation, migration, and mitochondrial respiration in NSCLC cells, highlighting its role in tumor progression .
Ovarian Cancer (OC): Silencing MTCH2 impaired ATP production, induced apoptosis, and suppressed metastasis by disrupting mitochondrial function and claudin-3/AIMP2 signaling .
MTCH2 regulates lipid transfer between the endoplasmic reticulum and mitochondria, influencing energy expenditure and obesity .
Skeletal muscle-specific MTCH2 deletion in mice increased energy expenditure and protected against diet-induced obesity .
MTCH2 acts as a receptor for the pro-apoptotic protein BID, mediating mitochondrial outer membrane permeabilization during apoptosis .
Mechanistic Insights:
Therapeutic Potential:
MTCH2 (mitochondrial carrier homolog 2) is a protein primarily localized to the mitochondrial outer membrane where it functions as a protein insertase that mediates insertion of transmembrane proteins. Specifically, MTCH2 catalyzes insertion of proteins with alpha-helical transmembrane regions, such as signal-anchored, tail-anchored, and multi-pass membrane proteins, but does not mediate insertion of beta-barrel transmembrane proteins . The protein has a calculated molecular weight of 33 kDa (303 amino acids) .
MTCH2 serves multiple critical cellular functions including:
Acting as a receptor for pro-apoptotic BH3-interacting domain death agonist (p15 BID), playing a critical role in apoptosis regulation
Regulating quiescence and cycling of hematopoietic stem cells (HSCs)
Functioning as a regulator of mitochondrial fusion, essential for naive-to-primed interconversion of embryonic stem cells
Contributing to lipid homeostasis and adipocyte differentiation
Supporting mitochondrial function and energy metabolism in various cell types
Research has demonstrated that MTCH2 dysregulation is implicated in several cancer types, including castration-resistant prostate cancer and ovarian cancer, highlighting its importance as a potential biomarker and therapeutic target .
MTCH2 antibodies have been validated for multiple research applications, each offering distinct advantages for specific experimental questions. Based on technical validation data, MTCH2 antibodies can be reliably used in the following applications:
It is important to note that optimal antibody concentrations should be determined for each experimental system through careful titration to achieve the best signal-to-noise ratio .
MTCH2 antibodies have been tested and validated in multiple species, with varying degrees of confirmed reactivity. When selecting an antibody for your research, it is essential to consider species compatibility:
For species not explicitly confirmed, researchers should consider protein homology between species. Many antibody manufacturers indicate that strong sequence homology between species may predict cross-reactivity, but experimental validation is always recommended before proceeding with extensive studies .
Recent bioinformatic analyses have revealed significant associations between MTCH2 expression and clinical parameters in various cancer types. In prostate cancer, MTCH2 overexpression is associated with critical clinical parameters, indicating its potential role as a prognostic marker . Single-cell sequencing data demonstrates elevated MTCH2 expression specifically in the prostate cancer epithelium .
In castration-resistant prostate cancer (CRPC):
MTCH2 is upregulated in locally treated CRPC tissue
Higher expression is observed in various primary human CRPC cells
MTCH2 expression correlates with disease progression and poor clinical outcomes
Research using genetic manipulation approaches (shRNA and CRISPR-mediated knockout) has demonstrated that MTCH2 depletion significantly impairs mitochondrial function in CRPC cells, resulting in:
Reduced oxygen consumption rate
Diminished complex I activity
Decreased ATP levels
Mitochondrial depolarization
These metabolic alterations led to inhibited cell viability, proliferation, and migration, with increased apoptosis in primary CRPC cells. Conversely, ectopic expression of MTCH2 enhanced ATP production and promoted cell proliferation and migration .
In ovarian cancer, MTCH2 has been shown to play roles in energy metabolism and metastatic potential. Experimental manipulation of MTCH2 expression levels demonstrably affects cell migration and invasion capabilities, with overexpression enhancing and knockdown reducing these cancer-associated behaviors .
Proper validation of MTCH2 antibody specificity is crucial for experimental reliability. Based on published research methodologies, the following controls should be included:
Genetic manipulation controls:
Detection controls:
Technique-specific controls:
Published studies have successfully used MTCH2 siRNA to reduce protein expression, confirming antibody specificity when the signal decreases proportionally to the knockdown efficiency . Similarly, CRISPR-sgRNA approaches provide definitive validation of antibody specificity when the signal is absent in knockout cell lines .
MTCH2 interacts with multiple proteins as part of its various cellular functions. Research has demonstrated several approaches to investigate these interactions:
Co-immunoprecipitation (Co-IP):
Successfully used to identify interactions between MTCH2 and other proteins
Published studies have documented interactions between MTCH2, claudin-3, and AIMP2 (aminoacyl tRNA synthetase-interacting multifunctional protein 2)
Recommended protocol: Use 0.5-4.0 μg of MTCH2 antibody for 1.0-3.0 mg of total protein lysate
Proximity ligation assays:
Allows visualization of protein interactions in situ
Can detect endogenous protein interactions without overexpression
Functional interaction studies:
Research has revealed that MTCH2 interacts with apoptotic factors like p15 BID, suggesting a role in apoptotic signaling pathways . Additional interaction partners include proteins involved in mitochondrial fusion and lipid homeostasis, consistent with MTCH2's multiple cellular functions .
MTCH2 is primarily localized to the mitochondrial outer membrane, but studying its distribution and function requires careful consideration of subcellular fractionation and localization techniques:
Subcellular fractionation:
Mitochondrial isolation procedures should be optimized to maintain outer membrane integrity
Differential centrifugation with gradient purification is recommended
Western blot analysis should include compartment-specific markers:
Outer mitochondrial membrane: TOM20, VDAC
Inner mitochondrial membrane: Complex IV subunits
Matrix: HSP60
Cytosolic: GAPDH or β-actin
Immunofluorescence microscopy:
Co-staining with mitochondrial markers (MitoTracker or TOM20) is essential
Fixation method can significantly impact results:
4% paraformaldehyde preserves structure while maintaining antigenicity
Cold methanol fixation may be preferred for membrane proteins
Functional assays for mitochondrial compartments:
Outer membrane permeabilization: Cytochrome c release assays
Respiration studies: Oxygen consumption measurements
Membrane potential: JC-1 or TMRE staining
ATP production: Luminescence-based assays
Research has shown that MTCH2 depletion impacts mitochondrial function across multiple parameters, including oxygen consumption rate, complex I activity, ATP levels, membrane potential, and reactive oxygen species production .
Western blotting is one of the most common applications for MTCH2 antibodies. The following protocol has been validated for optimal results:
Sample preparation:
Gel electrophoresis and transfer:
10-12% SDS-PAGE is suitable for resolving the 33 kDa MTCH2 protein
Transfer to PVDF membrane is preferred over nitrocellulose
Use wet transfer method for better efficiency with membrane proteins
Antibody incubation:
Detection and analysis:
This protocol has been successfully used in multiple published studies investigating MTCH2 expression in various cell types and tissues .
Immunohistochemistry (IHC) allows visualization of MTCH2 expression in tissue samples. The following protocol is recommended based on validated methods:
Sample preparation:
Fix tissues in 10% neutral buffered formalin
Embed in paraffin and section at 4-5 μm thickness
Mount sections on positively charged slides
Antigen retrieval:
Staining procedure:
Controls and analysis:
IHC studies have successfully detected MTCH2 in various tissues including human liver , prostate cancer samples , and ovarian cancer specimens , with primarily mitochondrial and membranous staining patterns.
Co-immunoprecipitation (Co-IP) is valuable for studying MTCH2 protein interactions. The following protocol has been validated for MTCH2 Co-IP experiments:
Cell lysis and preparation:
Use mild lysis buffer (e.g., 25 mM Tris-HCl pH 7.4, 150 mM NaCl, 1% NP-40, 1 mM EDTA, 5% glycerol)
Add protease and phosphatase inhibitors freshly
Clear lysate by centrifugation (14,000 × g, 10 min, 4°C)
Pre-clear lysate with protein A/G beads to reduce non-specific binding
Immunoprecipitation:
Analysis:
Validation approaches:
Reverse Co-IP: Immunoprecipitate with antibody against suspected interacting protein
Cross-linking prior to lysis can capture transient interactions
Proximity ligation assay as independent confirmation
Successful Co-IP experiments have demonstrated interactions between MTCH2 and both claudin-3 and AIMP2, revealing a tripartite interaction among these proteins that may regulate mitochondrial function .
Accurate quantification of MTCH2 expression is critical for comparative studies. Several validated approaches include:
Western blot densitometry:
Capture images within linear dynamic range
Use software like ImageJ, Image Lab, or similar
Normalize to appropriate loading controls
For mitochondrial proteins, consider normalizing to:
Total protein (Ponceau S or REVERT stain)
Mitochondrial markers (VDAC, TOM20)
Present data as fold change relative to control
qRT-PCR for transcript analysis:
Design primers specific to MTCH2 transcript
Use reference genes appropriate for tissue/treatment
Calculate relative expression using 2^(-ΔΔCt) method
Validate protein changes with Western blot
Immunohistochemistry quantification:
Digital pathology approaches using color deconvolution
Score staining intensity (0-3+) and percentage of positive cells
Calculate H-score or Quick score
Use automated image analysis software when possible
Flow cytometry (for cell studies):
Requires cell permeabilization for intracellular staining
Use fluorochrome-conjugated secondary antibodies
Include isotype control
Present as mean fluorescence intensity (MFI)
These methods have been successfully employed to demonstrate differential MTCH2 expression in normal versus cancer tissues, and to quantify changes following genetic manipulation .
Researchers may encounter several technical challenges when working with MTCH2 antibodies. Here are evidence-based solutions to common issues:
Low or no signal in Western blot:
Increase protein loading (40-60 μg total protein)
Optimize antibody concentration (try higher concentration within 1:50-1:500 range)
Extend primary antibody incubation (overnight at 4°C)
Try alternative antigen retrieval methods for fixed samples
Check sample preparation (include protease inhibitors)
Consider more sensitive detection methods (ECL Plus or Femto)
Multiple bands or non-specific binding:
Increase blocking stringency (5% BSA instead of milk)
Add 0.1-0.3% Triton X-100 to reduce hydrophobic interactions
Increase washing duration and number of washes
Validate specificity with knockdown/knockout controls
Use fresh antibody aliquots (avoid freeze-thaw cycles)
High background in immunofluorescence:
Optimize fixation protocol (try 4% PFA vs. methanol)
Extend blocking time (2 hours at room temperature)
Use Image-iT FX Signal Enhancer before antibody incubation
Include 0.1% Tween-20 in antibody dilution buffer
Prepare fresh mounting medium with anti-fade reagent
Poor immunoprecipitation efficiency:
Researchers have successfully addressed these challenges by optimizing antibody concentrations and incubation conditions, as evidenced by published studies using MTCH2 antibodies in various applications .
Proper validation of MTCH2 genetic manipulation models is essential for functional studies. The following approaches have been successfully used in published research:
siRNA/shRNA knockdown validation:
Western blot confirmation of protein reduction
Multiple siRNA sequences targeting different regions
Include scrambled/non-targeting control
Quantify knockdown efficiency (typically 70-90% reduction)
Check for off-target effects on related proteins
Monitor expression over time for transient knockdown
CRISPR-Cas9 knockout validation:
Western blot confirmation of complete protein absence
PCR and sequencing of targeted genomic region
Multiple sgRNA designs to control for off-target effects
Single-cell cloning and screening
Functional validation of mitochondrial phenotypes
Overexpression model validation:
Western blot confirmation of increased protein levels
Include empty vector control
Verify subcellular localization using fractionation or IF
Monitor potential toxicity from overexpression
Functional validation:
Mitochondrial function assays (oxygen consumption, ATP production)
Apoptosis measurements
Cell proliferation and migration assays
Phenotype rescue experiments
Published studies have demonstrated that genetic silencing via shRNA and knockout through CRISPR-sgRNA approaches effectively deplete MTCH2, leading to impaired mitochondrial function and altered cellular behaviors . Similarly, ectopic expression of MTCH2 enhances ATP production and promotes cancer cell proliferation and migration .