MRPL49 (mitochondrial ribosomal protein L49) is encoded by the nuclear gene MRPL49 (UniProt ID: Q13405) and functions in the 39S subunit of mitochondrial ribosomes . These ribosomes synthesize 13 essential oxidative phosphorylation (OXPHOS) proteins, making MRPL49 critical for cellular energy metabolism. The MRPL49 antibody specifically binds to this protein, facilitating its detection in experimental models.
MRPL49 antibodies are used to:
Assess OXPHOS complex deficiencies linked to MRPL49 dysregulation .
Study apoptosis pathways involving mitochondrial stability .
Cancer: Overexpression or depletion of MRPL49 correlates with tumor progression in breast and colon cancers .
Neurodegeneration: Reduced MRPL49 levels impair mitochondrial translation, contributing to disorders like Leigh syndrome .
Perrault Syndrome: Biallelic MRPL49 variants cause sensorineural hearing loss, ovarian insufficiency, and leukodystrophy .
Variants in MRPL49 (e.g., residues 88/92) destabilize mitochondrial ribosomes, reducing 16S rRNA stability .
Impaired mitochondrial translation leads to energy deficits, triggering apoptosis and tissue-specific pathologies .
| Application | Sample Type | Result | Source |
|---|---|---|---|
| Western Blot | HeLa, A549 cells | Clear band at 19 kDa | |
| IHC | Human colon | Strong cytoplasmic staining | |
| IP | HeLa lysates | Co-precipitation with mitoribosome proteins |
MRPL49 (Mitochondrial Ribosomal Protein L49) is an essential component of the 39S large subunit of the mitochondrial ribosome (mitoribosome). The mitoribosome is a 55S ribonucleoprotein complex composed of large and small subunits that coordinates the synthesis of the 13 proteins encoded by the mitochondrial genome . These proteins are vital components of the oxidative phosphorylation (OXPHOS) enzyme complexes.
Unlike bacterial, chloroplast, and cytosolic ribosomes, MRPL49 has no apparent homolog in these systems, suggesting a unique role in mitochondrial translation . Current research indicates that MRPL49:
May compensate for lost rRNA and stabilize bypass segments within the mt-LSU
Interacts intricately with MRPL4, MRPL15, MRPL57, and MRPL64, alongside the 16S rRNA
Is critical for maintaining the proper three-dimensional architecture of the mitoribosome
Has a homolog in yeast (Img2) that is required for mitochondrial genome integrity
The importance of MRPL49 is highlighted by the fact that biallelic variants in the MRPL49 gene cause combined oxidative phosphorylation deficiency (COXPD), a rare multisystem disorder .
When using MRPL49 antibodies in Western blot applications, you should expect to observe a band at approximately 19 kDa, which corresponds to the calculated molecular weight of the protein . This has been consistently observed across multiple antibody sources.
Regarding cellular localization:
In immunofluorescence studies, MRPL49 shows a punctate cytoplasmic pattern consistent with mitochondrial localization
In tissue sections from wild-type adult mice, MRPL49 has been observed in the mitochondria of outer hair cells, inner hair cells, and supporting cells of the inner ear
For optimal detection in immunohistochemistry applications, antigen retrieval with TE buffer pH 9.0 is suggested, although citrate buffer pH 6.0 may also be used as an alternative .
MRPL49 antibodies can be instrumental in investigating mitochondrial translation defects through several sophisticated approaches:
Complexome Profiling Method:
Complexome profiling is a quantitative mass spectrometry approach that can characterize deficiencies in respiratory complex genes . The methodology involves:
Isolating enriched mitochondrial fractions from patient-derived or experimental cell lines
Performing Blue Native electrophoresis (BNE)
Systematic dissection of the polyacrylamide gel
Tryptic digestion and tandem MS analysis
Visualization of protein abundance in each discrete section as heatmaps and line charts
This approach can reveal alterations in the content of mitoribosomes, as evidenced by changes in levels of individual mt-LSU and mt-SSU components. In fibroblasts from patients with MRPL49 variants, complexome profiling revealed:
Decreases of ~15-40% in mt-SSU levels
More pronounced reductions of ~60-70% in mt-LSU levels
Mitochondrial Protein Translation Assays:
To examine the impact of MRPL49 dysfunction on mitochondrial protein synthesis:
Measure the activity and levels of OXPHOS complexes using MRPL49 antibodies for immunoblotting
Compare complex I and IV subunit levels between control and experimental samples
Correlate changes in OXPHOS subunit levels with clinical phenotypes
16S:12S rRNA Ratio Analysis:
Previous studies investigating disease-associated variants in genes encoding mt-LSU proteins have consistently demonstrated a relative reduction in 16S rRNA levels . Using RT-qPCR in conjunction with MRPL49 antibody validation:
Extract RNA from control and experimental cells
Perform RT-qPCR to quantify 16S and 12S rRNA levels
Calculate the 16S:12S rRNA ratio
Correlate changes in this ratio with MRPL49 protein levels detected by antibodies
This approach can provide insights into how MRPL49 variants affect mitoribosome stability and function.
Ensuring antibody specificity is critical for obtaining reliable research results. For MRPL49 antibodies, consider implementing the following validation strategy:
Essential Controls:
Positive Controls:
Negative Controls:
MRPL49 knockdown or knockout cells (using siRNA or CRISPR-Cas9)
Secondary antibody-only control to assess non-specific binding
Isotype control to evaluate background
Peptide Competition Assay:
Advanced Validation Methods:
Orthogonal Validation:
Confirm results using at least two antibodies targeting different epitopes of MRPL49
Compare protein expression with mRNA levels using qPCR
Cross-Reactivity Assessment:
Test antibody against related mitochondrial ribosomal proteins, particularly those with similar molecular weights
Evaluate performance across multiple species if cross-reactivity is claimed
Application-Specific Validation:
Technical Considerations:
When using MRPL49 antibodies for Western blot, the sensitivity of different assays can vary. This has been demonstrated in fibroblasts from patients with MRPL49 variants, where complex I subunit deficiencies were detected in enzymatic assays but not always visible by Western blot .
MRPL49 dysregulation has been implicated in a spectrum of mitochondrial diseases, particularly combined oxidative phosphorylation deficiency (COXPD). Recent research has revealed fascinating correlations between MRPL49 expression patterns and disease phenotypes:
Phenotypic Spectrum Associated with MRPL49 Variants:
| Clinical Feature | Frequency in Affected Individuals | Notes |
|---|---|---|
| Hearing loss | 5/8 (62.5%) | Sensorineural type |
| Primary ovarian insufficiency (POI) | 3/4 (75%) of post-pubertal females | Part of Perrault syndrome presentation |
| Brain white matter changes | 6/6 (100%) | Progressive leukodystrophy |
| Learning disability | 8/8 (100%) | Universal feature |
| Microcephaly | 4/6 (66.7%) | Associated with neurodevelopmental phenotype |
| Retinal disease | Variable | Component of multi-system involvement |
| Hypoglycemia | Some cases | Metabolic manifestation |
| Renal disease | Some cases | Part of multi-organ involvement |
Genotype-Phenotype Correlations:
Interestingly, even within families harboring identical MRPL49 variants (e.g., homozygous His92Pro), there can be striking inter-familial phenotypic differences. For example:
Some affected females present with classical features of Perrault syndrome
Others with the same variant show no evidence of ovarian insufficiency or hearing loss
This suggests the presence of undefined genetic modifiers influencing the phenotypic expression of MRPL49 deficiency.
Correlation with Mitochondrial Function:
The severity of clinical presentation correlates with the degree of mitochondrial dysfunction as measured by:
Reduction in mt-LSU and mt-SSU levels
Decreased levels of OXPHOS enzyme complexes I and IV
For example, in fibroblasts from a severely affected individual (F5:II-1), complexome profiling revealed more pronounced reductions in mitoribosomal components (~40% in mt-SSU and 70% in mt-LSU) compared to a less severely affected individual (F1:II-1, ~15% in mt-SSU and 60% in mt-LSU) .
Research Implications:
When using MRPL49 antibodies to study these correlations, researchers should:
Quantify MRPL49 protein levels in patient-derived cells or tissues
Correlate protein levels with disease severity and specific phenotypes
Consider the impact of genetic background on phenotypic expression
Complexome profiling is a powerful approach for analyzing mitochondrial protein complexes and has been successfully applied to study MRPL49-associated pathologies. Here is a detailed protocol optimized for MRPL49 antibody use in complexome profiling:
Sample Preparation:
Enrich mitochondrial fractions from cultured cells (fibroblasts or other cell types) :
Homogenize cells in isolation buffer (250 mM sucrose, 10 mM Tris-HCl pH 7.5, 1 mM EDTA)
Centrifuge at 1,000 × g for 10 minutes to remove nuclei and unbroken cells
Centrifuge supernatant at 10,000 × g for 10 minutes to pellet mitochondria
Wash mitochondrial pellet twice with isolation buffer
Solubilize mitochondrial proteins:
Resuspend mitochondrial pellet in 50 mM NaCl, 50 mM imidazole, 2 mM aminohexanoic acid, 1 mM EDTA, pH 7.0
Add digitonin or n-dodecyl-β-D-maltoside at a detergent-to-protein ratio of 4:1
Incubate on ice for 10 minutes
Centrifuge at 20,000 × g for 20 minutes to remove insoluble material
Blue Native Electrophoresis (BNE):
Load solubilized proteins onto 3-12% or 4-16% gradient native PAGE gels
Run at 100 V for 30 minutes, then increase to 300 V, maintaining current below 15 mA
Use Coomassie Blue G-250 as the charge carrier in cathode buffer
Sample Processing for Mass Spectrometry:
Cut gel lanes into approximately 60 equal pieces (1 mm each)
Process each gel piece separately:
Destain with 50% acetonitrile in 50 mM ammonium bicarbonate
Reduce with 10 mM DTT for 30 minutes at 56°C
Alkylate with 55 mM iodoacetamide for 30 minutes in the dark
Digest with trypsin overnight at 37°C
Extract peptides with 50% acetonitrile/0.1% formic acid
Dry extracts and reconstitute in 0.1% formic acid
Mass Spectrometry Analysis:
Analyze samples using nano-LC-MS/MS
Use a standard 90-minute gradient for peptide separation
Process data using MaxQuant or similar software for protein identification
Normalize protein intensities to allow comparison between samples
MRPL49 Antibody Validation During Complexome Profiling:
Run a parallel Western blot of the same BN-PAGE gel
Transfer proteins to PVDF membrane
Compare MRPL49 migration pattern with MS-based complexome profile
Use known mitoribosomal markers (e.g., MRPL4, MRPL15) as references
Data Analysis and Visualization:
Generate heatmaps and line charts to visualize protein abundance across gel slices
Specifically analyze:
MRPL49 distribution pattern
Co-migration with other mt-LSU components
Relative abundance compared to control samples
Correlation with OXPHOS complex profiles
Special Considerations:
Deposit MS data to repositories like ProteomeXchange Consortium via PRIDE
Include dataset identifiers in publications for reproducibility
For MRPL49 variant studies, analyze how variants affect migration patterns of interacting partners (MRPL4, MRPL15, MRPL57, MRPL64)
MRPL49 antibodies serve as valuable tools for elucidating the molecular mechanisms underlying mitochondrial diseases, particularly those involving mitoribosomal dysfunction. Here are methodological approaches for using these antibodies in mitochondrial disease research:
1. Characterizing Pathogenic Variants Impact:
To investigate how MRPL49 variants affect protein function and mitochondrial translation:
Structure-Function Analysis:
Mitoribosome Assembly Assessment:
Perform sucrose gradient centrifugation to separate ribosomal subunits
Use MRPL49 antibodies in Western blot to track protein distribution across fractions
Compare assembly profiles between wild-type and disease models
2. Translational Defect Quantification:
Pulse-Chase Labeling:
Perform [35S]-methionine labeling of newly synthesized mitochondrial proteins
Immunoprecipitate MRPL49 to assess its association with active translation
Quantify translation rates in the presence of wild-type vs. variant MRPL49
Polysome Profiling:
Isolate mitochondria and extract polysomes
Use MRPL49 antibodies to track mitoribosome distribution
Compare polysome profiles between control and disease models
3. Tissue-Specific Pathology Investigation:
Recent findings show that MRPL49 deficiency presents with tissue-specific manifestations . To investigate this:
Immunohistochemistry Protocol Optimization:
For brain tissue (to study leukodystrophy pathology):
Use antigen retrieval with TE buffer pH 9.0
Recommended antibody dilution: 1:50-1:200
Counterstain with DAPI to visualize nuclei
For inner ear tissue (to study hearing loss mechanism):
Decalcify temporal bones prior to processing
Use MRPL49 antibody at 1:50 dilution
Co-stain with markers for hair cells (Myo7a) and mitochondria (TOMM20)
For retinal tissue (to study retinal dystrophy):
Cryosection eyes at 10-12 μm thickness
Use MRPL49 antibody at 1:50 dilution
Co-stain with photoreceptor markers
4. Therapeutic Strategy Evaluation:
MRPL49 antibodies can be used to assess the efficacy of experimental treatments:
Gene Therapy Monitoring:
Quantify MRPL49 protein levels before and after gene supplementation
Assess restoration of mitoribosome assembly and function
Correlate with phenotypic rescue
Small Molecule Screening:
Use MRPL49 antibodies in high-throughput immunofluorescence assays
Screen compounds that stabilize mutant MRPL49 or enhance residual mitoribosome function
Validate hits with secondary assays measuring OXPHOS complex formation
5. Disease Progression Biomarkers:
MRPL49 antibodies may help identify biomarkers for disease progression:
Longitudinal Analysis:
Measure MRPL49 levels in accessible tissues (e.g., fibroblasts, blood cells)
Correlate with disease severity and progression
Establish threshold values for clinical significance
Methodological Guidance for Brain MRI Correlation:
MRPL49-associated pathologies show distinctive brain MRI findings, particularly symmetrical involvement of the globi pallidi and deep white matter changes . Researchers can correlate these imaging findings with molecular changes by:
Performing immunohistochemistry on post-mortem brain tissue from affected regions
Using MRPL49 antibodies to assess protein localization and abundance
Correlating findings with specific MRI patterns (e.g., T2 high signal, diffusion restriction, cystic change)
Comparing results across disease stages to understand progressive pathology
When facing inconsistent results with MRPL49 antibodies, a systematic troubleshooting approach is essential. Below are methodological solutions to common technical challenges:
1. Variability in Western Blot Detection:
Technical Note: The sensitivity of different assays can vary significantly. In fibroblasts from patients with MRPL49 variants, complex I subunit deficiencies were detected in enzymatic assays but not always visible by Western blot .
2. Immunohistochemistry Optimization:
MRPL49 detection in tissues requires careful optimization:
Fixation Effects:
Overfixation in formalin can mask epitopes
Test multiple fixation times (6, 12, 24 hours)
Compare frozen vs. FFPE sections for epitope preservation
Antigen Retrieval Comparison:
Antibody Incubation Optimization:
3. Cross-Reactivity Assessment:
MRPL49 has pseudogenes located on chromosomes 5q and 8p , which may affect antibody specificity:
Perform BLAST analysis of the immunogen sequence
Test antibody in cells with MRPL49 knockout
Compare results with antibodies targeting different epitopes
4. Sample-Specific Considerations:
Different sample types require specific handling for optimal MRPL49 detection:
Cell Lines:
Tissue Samples:
Fresh tissues should be fixed within 30 minutes of collection
Consider regional variation in mitochondrial content
Use FFPE samples less than 5 years old for consistent results
Patient-Derived Samples:
Account for genetic background variations
Consider disease state and medication effects
Compare with age-matched controls
5. Complexome Profiling Challenges:
For researchers using MRPL49 antibodies in complexome profiling studies:
Ensure complete solubilization of mitochondrial membranes
Optimize detergent type and concentration (digitonin vs. n-dodecyl-β-D-maltoside)
Use a mitochondrial marker protein as an internal standard
Consider native vs. denaturing conditions based on research question
6. Advanced Analytical Approach:
When troubleshooting particularly difficult cases:
Quantify mitochondrial DNA content to normalize for mitochondrial mass
Measure 16S:12S rRNA ratio to assess mt-LSU stability
Perform RT-qPCR to compare mRNA and protein levels
Consider post-translational modifications or alternative splicing
Recent research has revealed intriguing tissue-specific manifestations of MRPL49 deficiency, suggesting differential regulation of mitochondrial translation across tissues. MRPL49 antibodies can be instrumental in exploring these tissue-specific mechanisms:
Methodological Approach for Tissue Comparison:
Multi-tissue Expression Analysis:
Perform Western blot analysis of MRPL49 across tissue panels
Quantify relative expression levels normalized to tissue-specific housekeeping proteins
Compare with other mitoribosomal proteins to identify tissue-specific patterns
Recent immunofluorescence studies in wild-type adult mice showed MRPL49 expression in the mitochondria of outer hair cells, inner hair cells, and supporting cells of the inner ear . This tissue-specific expression pattern provides insights into the hearing loss phenotype observed in some patients with MRPL49 variants.
Cell Type-Specific Translation Assessment:
Use immunofluorescence co-localization with cell type-specific markers
Combine with mitochondrial protein synthesis assays
Quantify translation efficiency in different cell populations
Developmental Regulation Analysis:
Track MRPL49 expression during embryonic and post-natal development
Correlate with mitochondrial maturation milestones
Identify critical periods for tissue-specific mitochondrial biogenesis
Experimental Design for Tissue-Specific Studies:
For investigating the role of MRPL49 in tissue-specific pathologies:
Neurological Manifestations:
Compare MRPL49 levels in different brain regions (cerebellum, basal ganglia, white matter)
Correlate with region-specific vulnerability in leukodystrophy
Use primary neuronal cultures to assess cell autonomous effects
Reproductive System:
Analyze MRPL49 expression in ovarian tissue at different developmental stages
Correlate with markers of ovarian reserve and folliculogenesis
Investigate the mechanistic link to primary ovarian insufficiency
Sensory Systems:
Compare MRPL49 distribution in cochlear vs. retinal tissues
Assess mitochondrial content and activity in these specialized cells
Explore relationships with tissue-specific energy demands
Advanced Single-Cell Applications:
To dissect cell type-specific roles of MRPL49:
Combine MRPL49 immunostaining with laser capture microdissection
Perform single-cell proteomics on isolated populations
Correlate MRPL49 levels with mitochondrial function at single-cell resolution
The mitochondrial ribosome plays a crucial role in mitochondrial stress responses, and MRPL49 may serve as a key regulatory component. MRPL49 antibodies can help elucidate these mechanisms:
Methodological Framework for Stress Response Studies:
Stress Induction Protocols:
Oxidative stress: Treatment with hydrogen peroxide or paraquat
Protein misfolding stress: Treatment with tunicamycin or thapsigargin
Mitochondrial translation stress: Treatment with doxycycline or chloramphenicol
Energy depletion: Glucose deprivation or oligomycin treatment
MRPL49 Response Assessment:
Monitor MRPL49 protein levels by Western blot at different time points post-stress
Track subcellular localization changes by immunofluorescence
Assess post-translational modifications using specialized antibodies
Integration with Stress Response Pathways:
Co-immunoprecipitation of MRPL49 with stress response mediators
Analysis of MRPL49 association with quality control machinery
Investigation of potential regulatory interactions
Experimental Design for Patient-Derived Cells:
For cells harboring MRPL49 variants, stress response studies can provide insights into disease pathomechanisms:
Expose control and patient fibroblasts to graded stressors
Monitor activation of mitochondrial stress responses:
Mitochondrial unfolded protein response (UPRmt)
Integrated stress response (ISR)
Mitophagy and quality control pathways
Correlate stress susceptibility with clinical severity
Based on complexome profiling data, fibroblasts from severely affected individuals show more pronounced reductions in mitoribosomal components compared to less severely affected individuals . This suggests differential stress response capabilities that could be further explored using MRPL49 antibodies.
The variable clinical presentations associated with MRPL49 variants highlight the complex interplay between genotype and phenotype in mitochondrial diseases. MRPL49 antibodies can play a crucial role in unraveling this heterogeneity:
Methodological Strategy for Heterogeneity Analysis:
Genotype-Phenotype Correlation:
Quantify MRPL49 protein levels in patient-derived cells
Correlate with specific variant types and positions
Relate to clinical severity and organ involvement
Modifier Detection:
Develop co-immunoprecipitation protocols to capture MRPL49 interactors
Identify variant-specific interaction patterns
Screen for genetic or environmental modifiers that alter MRPL49 function
Threshold Effect Investigation:
Establish dose-response relationships between MRPL49 levels and mitochondrial function
Determine tissue-specific thresholds for functional impairment
Create models for predicting phenotypic severity
Experimental Approach for Variability Studies:
The striking inter-familial differences observed even with identical MRPL49 variants (e.g., homozygous His92Pro) suggest the presence of undefined genetic modifiers . To investigate this:
Create a panel of cell lines with controlled genetic backgrounds
Introduce identical MRPL49 variants
Compare protein levels, stability, and function using MRPL49 antibodies
Correlate with mitochondrial translation efficiency and OXPHOS assembly
This approach can help identify factors contributing to the phenotypic variability observed in MRPL49-associated disorders, ranging from classical Perrault syndrome to severe childhood-onset leukodystrophy .
As our understanding of mitochondrial biology continues to evolve, MRPL49 antibodies will play an increasingly important role in advancing several frontier areas of research:
Single-cell mitochondrial heterogeneity studies:
Combining MRPL49 antibodies with single-cell technologies to understand cell-to-cell variation in mitochondrial translation
Correlating mitoribosome composition with functional states in individual cells
Therapeutic monitoring in mitochondrial disease trials:
Using MRPL49 antibodies as pharmacodynamic biomarkers for treatments targeting mitochondrial translation
Developing standardized assays for clinical trial endpoints
Spatiotemporal regulation of mitochondrial translation:
Applying super-resolution microscopy with MRPL49 antibodies to visualize translation sites within mitochondria
Tracking dynamic changes in mitoribosome distribution during cellular stress
Integration with multi-omics approaches:
Combining MRPL49 immunoprecipitation with RNA-seq to identify associated transcripts
Correlating proteomics and translatomics data to build comprehensive models of mitochondrial translation regulation