SLC25A35 belongs to the SLC25 family of mitochondrial carrier proteins, a group of transport proteins embedded in the inner mitochondrial membrane . It is a multi-pass membrane protein containing three Solcar repeats, which are characteristic structural features of this protein family . While the exact functions of SLC25A35 were previously unknown, recent research has revealed its significant role in mitochondrial metabolism, particularly in fatty acid oxidation and energy production .
SLC25A35 antibodies are immunological reagents specifically designed to bind to and detect the SLC25A35 protein in various experimental contexts. These antibodies have become essential tools for researchers investigating mitochondrial function, metabolism reprogramming in cancer, and potential therapeutic targets for metabolic disorders and cancer treatment.
Polyclonal antibodies against SLC25A35 are commonly produced in rabbits and recognize multiple epitopes of the protein. For instance, the Anti-SLC25A35 Rabbit Polyclonal Antibody from ProSci Inc. is purified by peptide affinity chromatography and lyophilized in PBS buffer with 2% sucrose . After reconstitution, the antibody concentration is 1 mg/mL, and it demonstrates reactivity against human, mouse, and rat SLC25A35 proteins .
Some antibodies target specific regions of the SLC25A35 protein:
N-Terminal antibodies: Target the amino-terminal region of the protein
Middle Region antibodies: Recognize epitopes in the central portion of the protein
Specific amino acid region antibodies: Such as the antibody targeting AA 120-168
Various conjugated forms of SLC25A35 antibodies are available for different detection methods:
Unconjugated: For standard applications requiring secondary antibody detection
FITC-conjugated: For fluorescent detection in imaging applications
Biotin-conjugated: For applications utilizing avidin-biotin complex detection
HRP-conjugated: For direct enzymatic detection in ELISA and similar applications
SLC25A35 antibodies have proven valuable in various research applications, allowing scientists to investigate the expression, localization, and function of this mitochondrial carrier protein.
Western blotting with SLC25A35 antibodies enables researchers to quantify protein expression levels in different tissues and cell lines. This application has been crucial in identifying differential expression of SLC25A35 in normal versus cancer tissues, particularly in hepatocellular carcinoma (HCC) .
IHC applications of SLC25A35 antibodies have facilitated the visualization of protein expression patterns in tissue sections. Recent studies have employed IHC to demonstrate upregulation of SLC25A35 in HCC tissues compared to adjacent non-tumor tissues, revealing its potential as a prognostic marker .
Immunoprecipitation using SLC25A35 antibodies allows for the investigation of protein-protein interactions, helping to elucidate the functional networks involving this mitochondrial carrier. These studies have contributed to understanding how SLC25A35 interacts with key metabolic regulators like PGC-1α .
SLC25A35 antibodies have been instrumental in knockdown and overexpression validation experiments, ensuring the specificity and efficiency of gene manipulation approaches used to study the functional consequences of altered SLC25A35 expression .
Recent research has revealed crucial roles of SLC25A35 in mitochondrial function and cellular metabolism, particularly in the context of cancer biology.
Studies using SLC25A35 antibodies to validate knockdown and overexpression have demonstrated that SLC25A35 significantly enhances mitochondrial oxidative respiration. Oxygen consumption rate (OCR) measurements reveal that SLC25A35 promotes mitochondrial metabolism without significantly affecting aerobic glycolysis .
SLC25A35 positively regulates ATP production while decreasing reactive oxygen species (ROS) levels. This unexpected finding challenges the conventional understanding that enhanced mitochondrial metabolism typically produces more ROS, suggesting that SLC25A35 may have unique mechanisms for controlling oxidative stress .
Mechanistic studies have revealed that SLC25A35 facilitates fatty acid oxidation (FAO) through upregulating peroxisome proliferator-activated receptor γ coactivator 1α (PGC-1α). This process involves increasing acetyl-CoA-mediated acetylation of PGC-1α, thereby enhancing its stability and activity .
Beyond metabolic regulation, SLC25A35 also enhances mitochondrial biogenesis, characterized by increased mitochondrial mass and DNA content. This function further amplifies the metabolic effects of SLC25A35 by expanding the mitochondrial network .
The discovery of SLC25A35's metabolic functions has led to important insights into its potential role in cancer development and progression.
Analysis using SLC25A35 antibodies has revealed significant upregulation of this protein in HCC tissues compared to normal liver tissues. This finding has been consistently observed in multiple patient cohorts and databases, including TCGA and independent clinical samples .
High SLC25A35 expression in HCC has been associated with adverse clinical parameters, including:
These correlations suggest that SLC25A35 may serve as a prognostic biomarker in HCC .
Functional studies have demonstrated that SLC25A35 promotes the proliferation and metastasis of HCC cells both in vitro and in vivo. Additionally, it enhances carcinogenesis in diethylnitrosamine (DEN)-induced HCC mouse models, further supporting its oncogenic role .
Research has identified that SLC25A35 upregulation in HCC is caused, at least partially, by decreased expression of miR-663a. This finding reveals a regulatory axis that could potentially be targeted for therapeutic intervention .
The emerging understanding of SLC25A35's functions and clinical significance opens several avenues for future research and potential therapeutic applications.
Given its differential expression in HCC and correlation with clinical outcomes, SLC25A35 detection using specific antibodies may have potential as a diagnostic or prognostic biomarker. Further validation in larger clinical cohorts is needed to establish its utility in this context.
The oncogenic role of SLC25A35 in HCC suggests that it could be an attractive therapeutic target. Strategies to inhibit SLC25A35 expression or function may offer novel approaches for cancer treatment, particularly for tumors exhibiting metabolic reprogramming .
Current research highlights the need for continued development of more specific and diverse SLC25A35 antibodies. Particularly valuable would be:
Antibodies targeting specific functional domains
Therapeutic antibodies capable of modulating SLC25A35 function
Antibodies suitable for live-cell imaging to study real-time localization and dynamics
SLC25A35 (Solute Carrier Family 25, Member 35) is a member of the mitochondrial carrier family that functions as a multi-pass membrane protein in the inner mitochondrial membrane . It contains three Solcar repeats, a characteristic feature of this protein family . Recent research has established SLC25A35 as a significant metabolic regulator that enhances fatty acid oxidation (FAO) and mitochondrial biogenesis, making it relevant for understanding mitochondrial metabolism in both normal and disease states . The protein has emerged as a potential oncogene in hepatocellular carcinoma (HCC), where it reprograms mitochondrial metabolism to support cancer cell proliferation and metastasis .
Several types of SLC25A35 antibodies are available for research, targeting different regions of the protein:
These antibodies are predominantly rabbit polyclonal antibodies and are available in various conjugated and unconjugated forms, including:
Most commercially available SLC25A35 antibodies demonstrate cross-reactivity across multiple species, though with varying degrees of sequence homology. The predicted reactivity based on sequence conservation includes :
Human: 100%
Mouse: 100%
Rat: 100%
Dog: 100%
Horse: 100%
Pig: 100%
Rabbit: 100%
Cow: 86%
Guinea Pig: 93%
Zebrafish: 91%
This broad cross-reactivity makes these antibodies versatile tools for comparative studies across different model organisms .
For Western blot analysis using SLC25A35 antibodies, researchers should consider the following methodological approach:
Sample preparation: Isolate proteins from cells or tissues using RIPA buffer supplemented with protease inhibitors.
Protein quantification: Use Bradford or BCA assay to normalize protein loading.
Gel electrophoresis: Separate 20-50 μg of protein on 10-12% SDS-PAGE gels.
Transfer: Transfer proteins to PVDF membranes (0.45 μm pore size preferred).
Blocking: Block membranes in 5% non-fat milk or BSA in TBST for 1 hour at room temperature.
Primary antibody incubation: Dilute SLC25A35 antibody at 1:500-1:2000 in blocking buffer and incubate overnight at 4°C .
Secondary antibody: Use appropriate HRP-conjugated secondary antibody (typically anti-rabbit IgG) at 1:5000-1:10000 dilution.
Detection: Develop using ECL substrate and visualize using a digital imaging system.
The middle region targeting antibody (ABIN5517415) has been validated specifically for Western blot applications, making it a reliable choice for protein expression analysis .
Proper storage and handling of SLC25A35 antibodies is critical for maintaining their activity and specificity:
Storage temperature: Store at -20°C for up to 1 year from receipt date .
Avoid freeze-thaw cycles: Aliquot antibodies upon first thaw to minimize repeated freeze-thaw cycles .
Reconstitution: For lyophilized antibodies, reconstitute in distilled water to a final concentration of 1 mg/mL .
Storage buffer: Some antibodies are provided in PBS containing 50% glycerol, 0.5% BSA, and 0.02% sodium azide to enhance stability .
Working dilution preparation: Prepare working dilutions on the day of the experiment and discard after use.
Transportation: Transport on ice when moving between storage and experimental areas.
Following these guidelines will help maintain antibody stability and experimental reproducibility .
Validating the specificity of SLC25A35 antibodies is crucial for reliable experimental outcomes. Researchers should consider implementing the following validation approaches:
Positive controls: Use cell lines or tissues known to express SLC25A35, such as HCC cell lines (SNU-368, HLE, SNU739, SNU-354, HLF, HUH-7) .
Negative controls:
Peptide competition assay: Pre-incubate antibody with the immunizing peptide before application to samples.
Multiple antibody verification: Use different antibodies targeting distinct epitopes of SLC25A35 to confirm observations.
Cross-reactivity testing: Evaluate potential cross-reactivity with other SLC25 family members through sequence alignment analysis.
Immunoprecipitation followed by mass spectrometry: To confirm the identity of the protein being recognized.
The antibody targeting amino acids 154-204 has been confirmed to detect endogenous levels of SLC25A35 in human and mouse samples , providing a validated option for research applications.
Recent studies have revealed that SLC25A35 plays a crucial role in reprogramming mitochondrial metabolism in cancer cells . Researchers can utilize SLC25A35 antibodies to investigate this process through:
Co-localization studies: Combine SLC25A35 antibodies with mitochondrial markers to confirm localization and distribution patterns in cancer versus normal cells.
Metabolic pathway analysis: Use SLC25A35 antibodies in combination with antibodies against fatty acid oxidation (FAO) components to study pathway interactions.
Bioenergetic profiling: Correlate SLC25A35 expression levels (determined by immunoblotting) with oxygen consumption rate (OCR) and ATP production measurements.
Post-translational modification analysis: Investigate how acetylation affects SLC25A35 function using specific antibodies against acetylated proteins.
Mitochondrial biogenesis assessment: Quantify relationship between SLC25A35 expression and mitochondrial mass/DNA content using immunofluorescence and qPCR.
Research has demonstrated that SLC25A35 enhances mitochondrial function characterized by increased oxygen consumption rate, ATP production, and decreased ROS levels through fatty acid oxidation enhancement , making these investigations particularly valuable.
SLC25A35 has been shown to facilitate fatty acid oxidation and mitochondrial biogenesis through regulation of PGC-1α . To investigate this relationship, researchers can:
Co-immunoprecipitation: Use SLC25A35 antibodies to pull down protein complexes and probe for PGC-1α interactions.
Acetylation status analysis: Employ acetyl-CoA and acetylated-PGC-1α antibodies in conjunction with SLC25A35 antibodies to explore SLC25A35's role in PGC-1α acetylation.
Chromatin immunoprecipitation (ChIP): Investigate how SLC25A35-mediated changes in PGC-1α affect binding to target gene promoters.
Proximity ligation assay (PLA): Visualize and quantify interactions between SLC25A35 and PGC-1α in situ.
Transcriptional reporter assays: Measure PGC-1α transcriptional activity in the presence of varying SLC25A35 levels.
This methodological approach can help elucidate the mechanistic pathway through which SLC25A35 upregulates PGC-1α via increased acetyl-CoA-mediated acetylation, ultimately enhancing mitochondrial function .
When facing contradictory results regarding SLC25A35 function or expression across different cancer types, researchers should consider:
Tissue-specific expression profiling: Use SLC25A35 antibodies for immunohistochemical analysis across diverse cancer tissue microarrays to establish tissue-specific expression patterns.
Isoform-specific analysis: Employ antibodies targeting different epitopes to identify potential isoform-specific functions.
Context-dependent signaling analysis: Investigate SLC25A35 in conjunction with tissue-specific transcription factors and signaling pathways.
Multi-omics integration: Correlate protein expression data (obtained using antibodies) with transcriptomics and metabolomics data.
Patient stratification: Analyze SLC25A35 expression in patient cohorts stratified by clinical parameters, genetic alterations, or metabolic profiles.
While SLC25A35 has been identified as an oncogene in hepatocellular carcinoma , research suggests that the broader SLC25 family may have varied prognostic value across different cancer types, including colon cancer , necessitating careful comparative studies.
SLC25A35 has emerged as a critical oncogene in hepatocellular carcinoma with significant therapeutic implications:
Elevated expression: SLC25A35 is highly expressed in HCC tissues compared to adjacent non-tumor tissues, as demonstrated in a comprehensive study of 238 pairs of HCC and adjacent non-tumor tissues .
Clinical correlation: High SLC25A35 expression correlates with adverse patient survival outcomes, suggesting its value as a prognostic marker .
Functional significance: SLC25A35 promotes:
Molecular mechanism: SLC25A35 reprograms mitochondrial metabolism by enhancing fatty acid oxidation and mitochondrial biogenesis through PGC-1α upregulation .
Regulatory insights: SLC25A35 upregulation in HCC is partly caused by decreased miR-663a expression .
These findings position SLC25A35 as a promising therapeutic target for HCC treatment, where antibodies could serve as valuable tools for patient stratification and response monitoring .
Researchers working with patient-derived xenograft models can utilize SLC25A35 antibodies to develop personalized approaches to cancer therapy:
Expression profiling in PDX tissue: Use immunohistochemistry with SLC25A35 antibodies to characterize expression patterns in individual patient tumors grown in PDX models.
Therapy response correlation:
Quantify SLC25A35 expression before and after treatment to identify expression changes
Correlate expression levels with response to metabolic-targeting therapies
Heterogeneity mapping: Apply multicolor immunofluorescence with SLC25A35 and other markers to map intratumoral heterogeneity.
Ex vivo drug sensitivity testing: Use SLC25A35 antibodies in combination with viability assays to correlate expression with drug sensitivity in PDX-derived organoids or slice cultures.
Combinatorial therapy development: Identify potential synergistic targets that interact with SLC25A35-mediated metabolic pathways.
This approach could help identify patient subgroups likely to benefit from metabolism-targeting therapies, particularly those that interfere with fatty acid oxidation or mitochondrial biogenesis that are enhanced by SLC25A35 .
Understanding the metabolic consequences of SLC25A35 inhibition is essential for evaluating its potential as a therapeutic target:
These metabolic alterations following SLC25A35 inhibition provide a mechanistic basis for the observed reduction in cancer cell proliferation and metastasis, supporting the therapeutic potential of targeting this protein .
Researchers can implement SLC25A35 antibody-based high-throughput screening strategies to identify potential therapeutic compounds:
Cell-based immunofluorescence assays: Develop automated imaging platforms using SLC25A35 antibodies to screen compound libraries for molecules that alter SLC25A35 expression, localization, or post-translational modifications.
AlphaLISA/HTRF assays: Design homogeneous antibody-based assays to detect changes in SLC25A35 protein interactions or modifications following compound treatment.
Reporter systems: Generate cells with fluorescent tags linked to SLC25A35 regulatory elements and validate with antibodies to screen for transcriptional modulators.
Functional metabolic screening: Combine SLC25A35 expression analysis (using antibodies) with Seahorse-based metabolic profiling to identify compounds that specifically disrupt SLC25A35-mediated metabolic reprogramming.
Phenotypic correlation: Correlate compound-induced changes in SLC25A35 expression/function (detected by antibodies) with cancer cell proliferation, migration, and invasion.
This approach could yield novel therapeutic candidates that specifically target the oncogenic functions of SLC25A35 in mitochondrial metabolism reprogramming .
Advanced techniques for investigating SLC25A35's interactions within the mitochondrial environment include:
Proximity-dependent biotin identification (BioID): Fuse BioID to SLC25A35 and use streptavidin pull-down followed by mass spectrometry to identify proximal proteins, validating key interactions with SLC25A35 antibodies.
APEX2-based proximity labeling: Apply APEX2-tagged SLC25A35 for temporal mapping of protein neighborhoods within mitochondria.
Cross-linking mass spectrometry (XL-MS): Use chemical crosslinkers to capture transient interactions involving SLC25A35, followed by mass spectrometry identification and antibody validation.
Super-resolution microscopy: Combine SLC25A35 antibodies with super-resolution techniques like STORM or PALM to visualize nanoscale organization within mitochondria.
Mitochondrial interactome mapping: Apply affinity purification with SLC25A35 antibodies coupled with quantitative proteomics to map comprehensive interaction networks.
These emerging techniques could reveal novel SLC25A35 interaction partners and mechanisms involved in mitochondrial metabolism regulation and cancer progression .
Researchers beginning work with SLC25A35 antibodies should consider these essential points:
Antibody selection: Choose antibodies targeting specific epitopes based on your research question; middle region antibodies (AA 120-168) have been well-validated for Western blot applications .
Validation strategy: Implement multiple validation approaches including positive and negative controls, particularly using SLC25A35 knockdown or knockout models .
Application-specific optimization: Optimize conditions for each application (WB, IHC, IF) as dilution ranges vary from 1:500-1:2000 depending on the application and specific antibody .
Species considerations: Verify cross-reactivity for your model organism, though most antibodies show good conservation across mammals .
Storage and handling: Aliquot antibodies to avoid freeze-thaw cycles and store at -20°C for maximum stability .
Following these guidelines will help ensure reliable and reproducible results when studying SLC25A35 in various research contexts.
The field of SLC25A35 research is poised for significant advances in several directions:
Expanded cancer applications: Beyond HCC, investigation of SLC25A35's role in other cancer types will likely emerge, building on findings of SLC25 family members' prognostic value in colon cancer .
Therapeutic development: Development of specific inhibitors targeting SLC25A35 or its downstream effectors may yield novel cancer therapeutics, particularly for metabolically active cancers .
Biomarker potential: Validation of SLC25A35 as a prognostic or predictive biomarker for specific cancer subtypes could improve patient stratification and treatment selection.
Systems biology integration: Integration of SLC25A35 into broader mitochondrial metabolic networks will enhance understanding of its regulatory role in health and disease.
Structure-function studies: Determination of SLC25A35's 3D structure and transport mechanism will facilitate rational drug design approaches.
As research progresses, SLC25A35 antibodies will remain essential tools for characterizing expression patterns, protein interactions, and functional consequences of this emerging mitochondrial metabolism regulator .