AGXT2 (Alanine--glyoxylate aminotransferase 2) is a mitochondrial enzyme involved in amino acid metabolism, particularly in the conversion of alanine and glyoxylate to pyruvate and glycine. AGXT2 dysfunction is linked to metabolic disorders and oxidative stress-related pathologies.
| Parameter | Details |
|---|---|
| Target | AGXT2 (UniProt ID: Q9BYV1) |
| Host Species | Rabbit |
| Reactivity | Human, Mouse, Rat |
| Applications | Western Blot (WB), ELISA |
| Molecular Weight | ~57 kDa |
| Storage | -20°C long-term; 4°C for short-term use (avoid freeze-thaw cycles) |
| Key Research Areas | Metabolic disorders, neurodegenerative diseases, biomarker discovery |
Anti-AGXT2 antibodies (e.g., Boster Bio #A07716) are polyclonal IgG antibodies validated for detecting endogenous AGXT2 in human, mouse, and rat samples . These antibodies are critical for studying AGXT2's role in β-aminoisobutyric acid (BAIBA) metabolism, which influences insulin sensitivity and mitochondrial function .
AGTR2 is a G protein-coupled receptor that counterbalances AGTR1’s vasoconstrictive effects by promoting vasodilation, apoptosis, and anti-inflammatory responses. It is a therapeutic target in cardiovascular and renal diseases.
| Parameter | Details |
|---|---|
| Target | AGTR2 (UniProt ID: P50052) |
| Host Species | Rabbit, Mouse |
| Reactivity | Human, Mouse, Rat |
| Applications | Immunohistochemistry (IHC), Western Blot (WB), Flow Cytometry (FC) |
| Molecular Weight | ~41 kDa |
| Therapeutic Relevance | Hypertension, cancer, tissue repair |
AGTR2 antibodies (e.g., R&D Systems #MAB3659) are used to study receptor localization and signaling in pathologies such as hypertension and cancer . For example, AGTR2 activation induces nitric oxide release, which is cardioprotective . In oncology, AGTR2-targeted therapies are explored for their anti-angiogenic effects in colorectal carcinoma .
AGXT2 knockdown models show altered BAIBA levels, correlating with improved metabolic health .
Polymorphisms in AGXT2 are associated with cardiovascular risk, making these antibodies vital for biomarker studies .
Hypertension: AGTR2 antibodies validate receptor upregulation in hypertensive models, aiding drug development .
Cancer: AGTR2 inhibition reduces tumor growth in xenograft models, highlighting its role in apoptosis .
AGT2 (also known as AGXT2) encodes Alanine--glyoxylate aminotransferase 2, a mitochondrial protein with a molecular weight of approximately 56 kDa. This enzyme belongs to the class-III pyridoxal-phosphate-dependent aminotransferase family and catalyzes several important transamination reactions, including the conversion of glyoxylate to glycine using L-alanine as an amino donor. Its significance in research stems from its role as a regulator of methylarginines and involvement in blood pressure control in the kidney. Additionally, polymorphisms in this gene have been associated with methylarginine metabolism and carotid atherosclerosis, making it a target of interest for cardiovascular and renal research .
When selecting antibodies, researchers must be cautious not to confuse AGT2 (AGXT2) with AGT (angiotensinogen), as they are distinct proteins with different functions. AGT2 is a mitochondrial aminotransferase expressed primarily in the kidney, while AGT is a precursor in the renin-angiotensin-aldosterone system (RAAS) pathway. To avoid experimental errors, verify the specific gene target (AGXT2 vs. SERPINA8/AGT), molecular weight (AGT2 is approximately 56 kDa while AGT is approximately 53.2 kDa), and tissue expression patterns (AGT2 is primarily in kidney mitochondria, while AGT is mainly produced by the liver) . Always check the specificity information in the antibody documentation, including the precise epitope recognition region and validated reactivity.
When selecting an AGT2 antibody, consider these critical parameters:
Specificity: Ensure the antibody specifically recognizes AGT2/AGXT2 and not AGT/angiotensinogen, using validation data such as knockdown/knockout controls.
Application compatibility: Verify the antibody has been validated for your specific applications (WB, ELISA, IHC, etc.).
Species reactivity: Confirm the antibody recognizes AGT2 in your species of interest (human, mouse, rat, etc.).
Epitope location: Consider whether the antibody targets an epitope in a functional domain or a region affected by known polymorphisms.
Clonality: Polyclonal antibodies may offer broader epitope recognition, while monoclonal antibodies provide higher specificity for a single epitope.
Citation record: Check for successful use in peer-reviewed publications.
Sample compatibility: Ensure compatibility with your sample preparation methods (fixed tissues, protein lysates, etc.) .
For optimal Western blot results with AGT2 antibodies:
Sample preparation: Extract proteins from kidney tissue (primary expression site) using a mitochondrial isolation buffer containing protease inhibitors to preserve AGT2's native structure.
Protein loading: Load 20-40 μg of total protein per lane; for enriched samples, 10-15 μg may be sufficient.
Gel percentage: Use 10-12% polyacrylamide gels to properly resolve the 56 kDa AGT2 protein.
Transfer conditions: Transfer at 100V for 60-90 minutes using a wet transfer system with methanol-containing buffer.
Blocking: Block membranes with 5% non-fat milk or BSA in TBST for 1-2 hours at room temperature.
Primary antibody: Dilute AGT2 antibody according to manufacturer recommendations (typically 1:500-1:2000) and incubate overnight at 4°C.
Detection: Use appropriate secondary antibody and chemiluminescent or fluorescent detection systems.
Controls: Include positive control (kidney tissue lysate) and negative control (tissue with low AGT2 expression) .
Ensure mitochondrial markers are included to verify enrichment of the mitochondrial fraction where AGT2 is localized.
For optimal immunohistochemical detection of AGT2:
Tissue fixation: Use 4% paraformaldehyde or 10% neutral buffered formalin fixation for 24-48 hours.
Antigen retrieval: Perform heat-mediated antigen retrieval in citrate buffer (pH 6.0) for 15-20 minutes.
Endogenous peroxidase quenching: Block with 3% hydrogen peroxide for 10 minutes.
Blocking: Use 5-10% normal serum (from the species of secondary antibody) with 1% BSA for 1 hour.
Primary antibody: Apply AGT2 antibody at 1:100-1:500 dilution and incubate overnight at 4°C.
Secondary detection: Use biotinylated secondary antibody followed by streptavidin-HRP or polymer detection systems.
Counterstaining: Counterstain with hematoxylin to visualize tissue architecture.
Controls: Include kidney tissue as positive control and perform negative controls by omitting primary antibody .
For co-localization studies, consider double immunofluorescence with mitochondrial markers to confirm AGT2's subcellular localization.
To rigorously validate AGT2 antibody specificity:
Western blot analysis: Verify a single band at the expected molecular weight (56 kDa) in kidney tissue lysates.
Knockout/knockdown controls: Test the antibody on samples from AGT2 knockout mice or siRNA-treated cells to confirm absence of signal.
Peptide competition assay: Pre-incubate the antibody with the immunizing peptide to demonstrate signal reduction.
Multiple antibody approach: Use antibodies targeting different epitopes of AGT2 to confirm consistent staining patterns.
Mass spectrometry validation: Perform immunoprecipitation followed by mass spectrometry to confirm target identity.
Recombinant protein controls: Test antibody against purified recombinant AGT2 protein.
Cross-reactivity assessment: Test against related proteins, particularly AGT/angiotensinogen, to ensure specificity .
Document validation data systematically for publication and reproducibility purposes.
When analyzing AGT2 expression patterns:
Tissue-specific expression: While AGT2 is predominantly expressed in kidney tissue, lower expression levels may be detected in other tissues. Interpret these differences in the context of tissue-specific functions.
Subcellular localization: Always confirm mitochondrial localization using co-staining with mitochondrial markers to validate authentic AGT2 signal.
Quantitative analysis: Use appropriate normalization controls (housekeeping proteins for Western blot; internal tissue controls for IHC) when comparing expression levels across tissues.
Physiological state influence: Consider that expression may vary with physiological state, disease condition, or experimental interventions.
Antibody sensitivity: Different antibodies may have varying sensitivities, potentially leading to different detection profiles.
Alternative splicing: Be aware that alternative splicing may result in different isoforms that might be detected differently by various antibodies .
Present quantitative data with appropriate statistical analysis and consider biological significance beyond statistical significance.
Common non-specific staining issues and solutions:
Cross-reactivity with similar proteins: AGT2 antibodies may cross-react with AGT/angiotensinogen or other aminotransferases. Solution: Validate antibody specificity with knockout controls and peptide competition assays.
High background in Western blots:
Cause: Insufficient blocking or too high antibody concentration
Solution: Optimize blocking (try 5% BSA instead of milk), increase washing steps, and titrate antibody dilutions
Non-specific binding in IHC:
Cause: Endogenous biotin or peroxidase activity
Solution: Include appropriate blocking steps (avidin/biotin blocking for biotin-based detection systems)
Epitope masking:
Cause: Fixation affecting epitope accessibility
Solution: Test different antigen retrieval methods (citrate vs. EDTA buffers)
Non-mitochondrial staining:
Always include appropriate negative controls (no primary antibody, isotype controls) and positive controls (known AGT2-expressing tissues).
To address weak or absent AGT2 signals:
Sample preparation issues:
Verify protein integrity with a general protein stain
Ensure proper tissue collection and storage (flash-freezing)
Use fresh protease inhibitors in lysis buffers
Consider enriching for mitochondrial fraction
Technical optimization:
Increase protein loading (40-60 μg)
Reduce washing stringency (lower detergent concentration)
Extend primary antibody incubation time (overnight at 4°C)
Try different blocking agents (milk vs. BSA)
Optimize transfer conditions for high molecular weight proteins
Antibody considerations:
Verify antibody viability (avoid freeze-thaw cycles)
Test alternative AGT2 antibodies targeting different epitopes
Consider more sensitive detection systems (enhanced chemiluminescence)
Biological factors:
Document all optimization steps systematically to identify critical parameters affecting detection.
For investigating AGT2 in kidney disease contexts:
Expression analysis in disease models:
Perform quantitative Western blot and IHC analyses comparing AGT2 expression in normal vs. diseased kidney tissues
Use dual staining with nephron segment markers to identify segment-specific changes in expression
Functional studies:
Combine AGT2 antibodies with activity assays to correlate protein levels with enzymatic function
Use immunoprecipitation with AGT2 antibodies followed by activity measurements to assess functional changes
Co-localization studies:
Perform co-immunoprecipitation to identify disease-specific protein interaction partners
Use proximity ligation assays to detect altered protein interactions in situ
Post-translational modifications:
Use phospho-specific antibodies alongside general AGT2 antibodies to assess regulatory changes
Employ immunoprecipitation with AGT2 antibodies followed by mass spectrometry to identify disease-associated modifications
Intervention studies:
Present data in the context of pathophysiological mechanisms and potential therapeutic implications.
For successful AGT2 co-immunoprecipitation studies:
Lysis buffer optimization:
Use gentle, non-denaturing buffers (e.g., 1% NP-40 or 0.5% CHAPS) to preserve protein-protein interactions
Include stabilizers like glycerol (10%) and mild reducing agents
Adjust salt concentration (150-300 mM NaCl) to balance specificity with interaction preservation
Antibody selection:
Choose antibodies validated for immunoprecipitation
Consider epitope location to avoid disrupting interaction domains
Test both polyclonal and monoclonal antibodies, as each has advantages
Protocol considerations:
Pre-clear lysates with protein A/G beads to reduce non-specific binding
Cross-link antibody to beads to prevent antibody co-elution
Include appropriate controls (IgG control, input sample)
Consider mild wash conditions to preserve weak interactions
Detection strategy:
Use clean elution methods (peptide competition or low pH) rather than boiling in SDS
Employ sensitive detection methods for low-abundance interacting proteins
Consider mass spectrometry for unbiased interactome analysis
Validation approaches:
Document all experimental conditions precisely for reproducibility and method sharing.
For studying AGT2 polymorphisms:
Antibody selection strategy:
Choose antibodies that recognize conserved regions unaffected by common polymorphisms for general detection
Develop or source polymorphism-specific antibodies that differentially recognize variant forms
Validate specificity using recombinant proteins expressing different variants
Expression analysis:
Compare protein expression levels across genotyped samples using quantitative Western blotting
Normalize to appropriate housekeeping proteins and analyze by genotype
Create a standardized protocol for sample collection and processing to minimize technical variation
Functional assessment:
Combine immunoprecipitation with activity assays to assess functional consequences of polymorphisms
Use immunoprecipitation to isolate variant proteins followed by structural analysis
Tissue localization studies:
Perform IHC on genotyped tissue samples to assess whether polymorphisms affect subcellular localization
Use high-resolution microscopy to detect subtle differences in distribution patterns
Interaction studies:
Investigate whether polymorphisms alter protein-protein interactions using co-immunoprecipitation
Consider proximity ligation assays for in situ interaction analysis in genotyped tissues
Experimental controls:
Present data in the context of genotype-phenotype correlations and potential clinical implications.
For investigating AGT2 in mitochondrial research contexts:
Subcellular fractionation approaches:
Optimize differential centrifugation protocols to isolate pure mitochondrial fractions
Validate fractionation quality using markers for mitochondria (VDAC, COX IV) and other compartments
Use AGT2 antibodies to track protein distribution across fractions
Functional correlation studies:
Combine AGT2 immunodetection with respirometry measurements
Correlate AGT2 levels with mitochondrial membrane potential using flow cytometry
Assess relationship between AGT2 expression and ROS production
Mitochondrial dynamics:
Use AGT2 antibodies in conjunction with markers of mitochondrial fusion/fission
Perform live-cell imaging with fluorescently tagged AGT2 antibody fragments
Stress response studies:
Monitor AGT2 expression changes during mitochondrial stress (OXPHOS inhibitors, etc.)
Assess mitochondrial localization changes under stress conditions
Super-resolution microscopy:
Employ STED or STORM microscopy with AGT2 antibodies to precisely localize within mitochondrial compartments
Perform multi-color imaging with markers for mitochondrial substructures
Metabolic flux analysis:
Present comprehensive data integrating protein expression, localization, and functional outcomes.
For multiplexed AGT2 detection in complex tissues:
Multiplexed immunofluorescence optimization:
Select compatible antibody pairs from different host species
Employ sequential staining protocols with careful antibody stripping/quenching between rounds
Use tyramide signal amplification for detecting low-abundance targets alongside AGT2
Validate spectral separation and absence of cross-reactivity
Imaging mass cytometry approaches:
Label AGT2 antibodies with rare earth metals
Combine with markers for cell types, organelles, and signaling states
Optimize staining protocols for metal-conjugated AGT2 antibodies
Develop comprehensive analysis pipelines for spatial data
Single-cell analysis integration:
Correlate AGT2 protein expression with single-cell transcriptomics
Develop protocols for antibody-based cell sorting followed by molecular analysis
Implement computational approaches to integrate protein and transcript data
Spatial context analysis:
Employ neighborhood analysis to identify cell types associated with high AGT2 expression
Quantify spatial relationships between AGT2-expressing cells and tissue structures
Validation strategies:
Design experiments with comprehensive tissue representation and sufficient statistical power for spatial analyses.
To correlate AGT2 protein levels with enzymatic activity:
Combined immunodetection and activity assays:
Perform parallel Western blot and enzyme activity measurements from the same samples
Calculate protein-to-activity ratios to identify samples with altered specific activity
Develop standardized normalization protocols for cross-sample comparisons
Sequential analysis methods:
Immunoprecipitate AGT2 using validated antibodies
Measure enzyme activity directly from the immunoprecipitated protein
Include controls for antibody interference with enzymatic function
In situ approaches:
Combine immunohistochemistry with histochemical enzyme activity staining on sequential sections
Develop protocols for combined fluorescent immunostaining and fluorogenic substrate assays
Implement image analysis for co-localization quantification
Advanced protein analysis:
Use antibodies to quantify post-translational modifications that might affect enzyme activity
Develop assays to measure AGT2 oligomerization states (monomer vs. homotetramer)
Assess cofactor (pyridoxal phosphate) binding using specialized antibodies
Experimental design considerations:
Present comprehensive data tables showing protein levels, activity measurements, and derived parameters across experimental conditions.
| Parameter | AGT2 (AGXT2) Antibodies | AGT (Angiotensinogen) Antibodies | Implications for Research |
|---|---|---|---|
| Target Protein | Alanine--glyoxylate aminotransferase 2 (mitochondrial) | Angiotensinogen (secreted) | Must verify target identity in publications |
| Molecular Weight | ~56 kDa | ~53.2 kDa | Similar sizes require careful interpretation |
| Primary Expression Site | Kidney | Liver | Tissue source affects experimental design |
| Subcellular Localization | Mitochondria | Secreted (also found in plasma) | Different fractionation approaches needed |
| Function | Amino acid metabolism, methylarginine regulation | RAAS pathway precursor | Different functional assays required |
| Common Applications | WB, ELISA, IHC-P | WB, ELISA, IHC-P, plasma assays | AGT often measured in circulation |
| Cross-reactivity Risk | May cross-react with other aminotransferases | May cross-react with angiotensin peptides | Validation with specific controls critical |
| Polymorphism Impact | Multiple functional polymorphisms | A-20C and M235T well-studied | Genotype may affect antibody binding |
| Research Focus | Metabolic pathways, kidney function | Hypertension, cardiovascular disease | Different research communities |
When using either antibody type, researchers should:
Confirm antibody specificity using appropriate positive and negative controls
Validate expected molecular weight and subcellular localization
Consider the possibility of cross-reactivity, particularly in tissues expressing both proteins
Clearly specify which protein (AGT2/AGXT2 or AGT/angiotensinogen) is being studied in all publications
When comparing AGT2 across model systems:
Antibody validation in each species:
Verify epitope conservation across species using sequence alignment
Perform Western blot validation in each species to confirm expected molecular weight
Consider developing species-specific antibodies for highly divergent regions
Standardization approaches:
Use recombinant proteins from each species as standards
Implement consistent sample preparation protocols across species
Develop normalization strategies accounting for species differences
Expression pattern considerations:
Document tissue-specific expression patterns in each species
Note developmental differences in expression timing
Consider sex-specific differences in expression
Technical parameters table:
| Parameter | Mouse | Rat | Human | Considerations |
|---|---|---|---|---|
| Antibody Dilution | 1:500-1:1000 | 1:500-1:1000 | 1:500-2000 | May require species-specific optimization |
| Expected MW | 53-56 kDa | 54-56 kDa | 56 kDa | Minor species variations may exist |
| Sample Preparation | Flash-frozen tissue | Flash-frozen tissue | Surgical/biopsy samples | Human samples may have variable quality |
| Primary Expression | Kidney | Kidney | Kidney | Consistent across mammals |
| Fixation for IHC | 4% PFA, 24h | 4% PFA, 24h | 10% NBF, variable time | Human samples often have variable fixation |
| Background Issues | Low in most tissues | Moderate in liver | Variable across tissues | Human samples may require additional blocking |
Functional correlation:
Document all methodological details comprehensively to enable proper interpretation of cross-species differences.