CHCHD2 (Coiled-coil-helix-coiled-coil-helix domain containing 2) is a mitochondrial protein that plays crucial roles in:
Regulating mitochondrial respiration through complex IV stability
Maintaining mitochondrial cristae structure
Controlling apoptotic pathways through interaction with BCL-xL
Functioning in complex with CHCHD10
Its significance has dramatically increased since mutations in CHCHD2 were linked to autosomal dominant Parkinson's disease (PD) and potentially Alzheimer's disease/frontotemporal dementia. CHCHD2 shares 58% amino acid sequence identity with CHCHD10, which is associated with amyotrophic lateral sclerosis/frontotemporal dementia (ALS/FTD) when mutated .
CHCHD2 demonstrates dual localization patterns:
Primary localization in mitochondria (intermembrane space)
Secondary localization in the nucleus under certain conditions
When using immunofluorescence techniques, endogenous CHCHD2 appears in both mitochondria and nucleus, while overexpressed non-tagged CHCHD2 shows a similar pattern with less nuclear distribution. The protein shows little overlap with endoplasmic reticulum (ER, detected by KDEL antibody) or with the autophagy-lysosomal system (detected by LAMP1 antibody) . For optimal visualization of mitochondrial localization, super-resolution microscopy has been particularly valuable in identifying CHCHD2's proximity to MICOS (mitochondrial contact site and cristae organizing system) .
Current commercial CHCHD2 antibodies are validated for multiple applications:
| Application | Typical Dilution | Validated Sample Types |
|---|---|---|
| Western Blot (WB) | 1:500-1:2000 | Human (HT-1080, HEK-293, HepG2), mouse tissues |
| Immunoprecipitation (IP) | 0.5-4.0 μg for 1-3 mg lysate | HepG2 cells |
| Immunohistochemistry (IHC) | 1:50-1:500 | Human lung cancer tissue, mouse brain tissue |
| Immunofluorescence (IF/ICC) | 1:200-1:800 | HepG2 cells |
| Co-Immunoprecipitation (CoIP) | Application-specific | Various cell lines |
| ELISA | 1:100-1:1000 | Multiple sample types |
These applications have been validated across human and mouse samples, with observed molecular weight typically around 16-18 kDa .
To investigate CHCHD2-CHCHD10 interactions, researchers should consider:
Crosslinking approaches:
Use disuccinimidyl glutarate (DSG) or disuccinimidyl suberate (DSS) crosslinkers
Western blot analysis reveals two CHCHD2 immunoreactive bands near 37 kDa:
A CHCHD2 homodimer
A more abundant heterodimer composed of CHCHD2 and CHCHD10
Co-immunoprecipitation strategy:
Express tagged versions (e.g., CHCHD2-Flag) in stable cell lines
CHCHD2-Flag efficiently pulls down both endogenous CHCHD2 and CHCHD10
CHCHD10-Flag pulls down endogenous CHCHD2 but less endogenous CHCHD10 compared to CHCHD2-Flag
This interaction pattern indicates specific complex formation between these proteins and their potential functional redundancy .
Rigorous validation requires multiple controls:
CHCHD2 knockout cells/tissues:
The most definitive control showing absence of signal in WB, IF, and IHC
CRISPR-Cas9 generated CHCHD2 KO cell lines are ideal reference standards
Peptide competition assays:
Pre-incubate antibody with immunogenic peptide to block specific binding
Multiple antibody comparison:
Test antibodies raised against different epitopes (N-terminal vs. C-terminal)
Consistent results across antibodies confirm specificity
Positive controls:
Include known CHCHD2-expressing samples (HepG2, HEK-293, or mouse brain)
Expected molecular weight should be 16-18 kDa
Recombinant protein standards:
Use purified CHCHD2 protein as size reference
Several publications have validated CHCHD2 antibodies using knockout models, confirming specificity for detection of this protein in various experimental contexts .
For optimal CHCHD2 detection in tissue sections:
Fixation and embedding:
4% paraformaldehyde fixation followed by paraffin embedding preserves epitope accessibility
Antigen retrieval methods:
Primary recommendation: TE buffer pH 9.0
Alternative method: Citrate buffer pH 6.0
Blocking conditions:
5% skim milk in TBS-T for 1 hour at room temperature prior to primary antibody application
Primary antibody incubation:
Dilution range: 1:50-1:500
Incubate overnight at 4°C with continuous shaking
Use antibody diluent containing TBS-T buffer with 0.01% sodium azide
Detection systems:
HRP-conjugated secondary antibodies (1:1000) with ECL detection system
For fluorescent detection, use appropriate fluorophore-conjugated secondary antibodies
This protocol has been successfully utilized in studies of mouse brain tissue and human cancer samples .
Several factors can explain molecular weight variations:
Post-translational modifications:
Phosphorylation or other modifications can alter electrophoretic mobility
Protein-protein interactions:
Incomplete processing:
As a mitochondrial protein, CHCHD2 may retain its transit peptide in some preparations
Crosslinking effects:
Splice variants:
The expected molecular weight range for monomeric CHCHD2 is 16-18 kDa, while dimeric forms appear around 37 kDa after crosslinking .
To achieve cleaner CHCHD2 immunofluorescence staining:
Increase blocking stringency:
Extend blocking time to 2 hours
Use 3-5% BSA with 0.1-0.3% Triton X-100 in PBS
Add 5-10% normal serum from the species of the secondary antibody
Optimize primary antibody conditions:
Titrate antibody concentration (1:200-1:800 dilution range)
Extend primary antibody incubation to overnight at 4°C
Include 0.01% sodium azide to prevent microbial growth during long incubations
Washing optimization:
Increase wash duration and number of washes (5-6 times for 5 minutes each)
Use PBS-T (PBS with 0.1% Tween-20) for more stringent washing
Fixation considerations:
For mitochondrial proteins, 4% paraformaldehyde for 15 minutes preserves structure while maintaining epitope accessibility
Avoid methanol fixation which can distort mitochondrial morphology
Confocal settings:
Adjust laser power and gain to minimize autofluorescence
Use spectral unmixing for tissues with high autofluorescence
Researchers have successfully visualized CHCHD2 in both mitochondria and nucleus using these optimized conditions .
A comprehensive experimental approach includes:
Generation of isogenic cell models:
Functional assays:
Structural studies:
Super-resolution microscopy to examine cristae morphology
Proximity to MICOS complex components
Interaction with cytochrome c
Biochemical analyses:
Co-immunoprecipitation to assess protein-protein interactions (e.g., with BCL-xL)
Blue native PAGE to examine native complex formation
Crosslinking studies to assess dimer formation
In vivo models:
Studies have demonstrated that CHCHD2 mutations impair mitochondrial function and can worsen behavioral deficits and dopaminergic neurodegeneration in animal models .
To investigate functional overlap between these paralogous proteins:
Genetic manipulation strategies:
Generate single knockouts (CHCHD2 KO, CHCHD10 KO)
Generate double knockouts (CHCHD2/10 DKO)
Perform rescue experiments with wild-type or mutant versions of either protein
Readouts for functional assessment:
Monitor steady-state levels of cytochrome c oxidase (COX) subunits, particularly COX2
Measure complex IV assembly using blue native PAGE
Assess mitochondrial cristae morphology
Evaluate apoptotic susceptibility
Interaction studies:
Use crosslinking agents (DSG or DSS) to stabilize protein complexes
Perform co-immunoprecipitation with tagged versions
Analyze migration patterns on western blots
Localization analysis:
Use super-resolution microscopy to examine co-localization patterns
Implement proximity ligation assays to confirm in situ interactions
Research has shown that CHCHD2 and CHCHD10 exhibit functional redundancy in maintaining COX2 levels and complex IV assembly, with double knockout cells showing more severe phenotypes than single knockouts .
To investigate CHCHD2's function in the ISR pathway:
Experimental design:
Generate CHCHD2 and/or CHCHD10 knockdown cells
Apply stress conditions (e.g., CCCP treatment for 24 hours)
Compare responses between control, single knockdown, and double knockdown conditions
Molecular readouts:
mRNA level analysis (RT-qPCR):
ATF3, ATF4, DDIT3, CHAC1
Protein level analysis (Western blotting):
ASNS, PCK2, PSPH, phosphorylated eIF2α
Stress induction methods:
Mitochondrial stress: CCCP (mitochondrial uncoupler)
ER stress: Tunicamycin or Thapsigargin
Oxidative stress: Hydrogen peroxide
Time course analysis:
Monitor changes at multiple timepoints (6h, 12h, 24h, 48h)
Assess both acute and chronic responses
Rescue experiments:
Re-express wild-type or mutant CHCHD2/CHCHD10 to determine which domains are critical for ISR regulation
Studies have demonstrated that CHCHD2 and CHCHD10 can regulate the integrated stress response under normal conditions and under mitochondrial stress induced by CCCP .
CHCHD2 exhibits context-dependent functions with important distinctions:
In cancer models:
Acts as an anti-apoptotic protein through BCL-xL interaction
Promotes cell proliferation and migration
Enhances mitochondrial respiration
Co-amplified with EGFR in non-small cell lung cancer
Associated with increased MMP2 expression and angiogenesis
Correlated with cancer progression in renal cell carcinoma
Expression correlates with poor prognosis in multiple cancer types
In neurodegenerative disease models:
Mutations (T61I, R145Q, Q126X) impair mitochondrial function
Mutation-bearing cells show abnormal cristae morphology
In PD models, mutations disrupt F1F0-ATPase assembly
May not interact with BCL-xL in stem cells, contrary to cancer cells
Loss of function leads to impaired neuroectodermal differentiation
This dual role makes CHCHD2 a fascinating target for both cancer and neurodegeneration research, requiring careful experimental design specific to each disease context .
When working with hPSCs (hESCs or iPSCs) for CHCHD2 research:
Culture adaptation monitoring:
Single-cell dissociation protocols:
Differentiation capacity assessment:
Genetic engineering considerations:
Specialized assays:
Research has demonstrated that CHCHD2's role in stem cells differs from cancer cells, particularly regarding BCL-xL interaction and effects on apoptosis .
To investigate CHCHD2's involvement in mtDNA regulation:
mtDNA copy number analysis:
mtDNA deletion detection:
Long-range PCR to identify large-scale deletions
Next-generation sequencing for comprehensive deletion mapping
Single-molecule PCR for low-frequency deletion detection
mtDNA transcription analysis:
In situ detection of respiratory chain deficiency:
Aging effects assessment:
Current research indicates that 12-month-old Chchd2-deficient mice accumulate COX-deficient colonic crypts, but show no changes in mtDNA copy number or mtDNA transcript levels in heart and skeletal muscle .
For developing CHCHD2-based biomarkers in PD:
Tissue-based approaches:
Immunohistochemical analysis of post-mortem brain sections
Evaluation of CHCHD2 expression patterns in PD vs. control brains
Assessment of CHCHD2 aggregation or mislocalization in disease states
Fluid biomarker development:
Detection of CHCHD2 levels in cerebrospinal fluid
Analysis of CHCHD2 in blood plasma/serum exosomes
Correlation with disease progression and severity
Multi-marker panels:
Combine CHCHD2 with other mitochondrial markers (CHCHD10, PINK1, Parkin)
Integrate with established PD biomarkers (α-synuclein, DJ-1)
Develop algorithms incorporating multiple protein measurements
Validation considerations:
Include antibodies targeting different CHCHD2 epitopes
Ensure consistent performance across patient cohorts
Establish sensitivity/specificity in distinguishing PD from other neurodegenerative conditions
Technical considerations:
Standardize sample collection and processing protocols
Implement highly sensitive detection methods (ELISA, digital ELISA, mass spectrometry)
Control for confounding factors (age, comorbidities, medications)
The association of CHCHD2 mutations with autosomal dominant PD makes this protein a promising candidate for biomarker development, though further validation studies are needed to establish clinical utility .
To discover therapeutic interventions targeting CHCHD2:
Drug screening platforms:
High-throughput screens using CHCHD2 mutant cell lines
Readouts: mitochondrial function, cell viability, protein aggregation
Repurposing FDA-approved compounds with mitochondrial targets
Therapeutic candidate examples:
Target validation approaches:
Genetic rescue experiments in disease models
Restoration of CHCHD2-CHCHD10 complex formation
Stabilization of mitochondrial cristae structure
Enhancement of respiratory chain complex assembly
Translational considerations:
Assess blood-brain barrier penetration
Evaluate safety in long-term administration
Develop biomarkers for treatment response
Design combinatorial approaches targeting multiple pathways
Model systems for evaluation:
Patient-derived iPSCs differentiated to neurons
CRISPR-engineered animal models expressing PD-linked CHCHD2 mutations
Combination with environmental stressors or aging to accelerate phenotypes
Research suggests the CHCHD2-CHCHD10 complex may represent a novel therapeutic target for PD and related neurodegenerative disorders, with potential benefit from mitochondria-targeted compounds like Elamipretide .
For comprehensive analysis of CHCHD2 mutations in patient samples:
Genetic screening methods:
Targeted sequencing of CHCHD2 coding regions
Inclusion in neurodegenerative disease gene panels
Whole exome/genome sequencing for novel variant discovery
Functional characterization:
Fibroblast isolation from mutation carriers
iPSC generation and neuronal differentiation
Assessment of mitochondrial parameters:
Respiration (OCR)
Membrane potential
ROS production
Cristae morphology
Protein analysis in patient samples:
Western blotting for expression levels and post-translational modifications
Immunohistochemistry in biopsy or autopsy tissue
Analysis of protein-protein interactions (co-IP with CHCHD10, BCL-xL)
Clinical correlation studies:
Genotype-phenotype correlation
Age of onset analysis
Progression rate assessment
Response to treatments
Population-specific considerations: