SGTA Antibody

Small Glutamine-Rich Tetratricopeptide Repeat-Containing Protein Alpha, Mouse Anti Human
Shipped with Ice Packs
In Stock

Description

SGTA Antibody: Definition and Structure

SGTA antibodies target the 34–37 kDa SGTA protein, which contains three tetratricopeptide repeat (TPR) motifs, an N-terminal dimerization domain, and a C-terminal glutamine-rich region . These antibodies are critical for investigating SGTA's roles in:

  • Androgen receptor (AR) signaling

  • Viral particle assembly (e.g., HIV-1, parvovirus)

  • Protein quality control in neurodegenerative disorders

Common antibody formats include monoclonal (e.g., mouse IgG2b clones 3B10B11) and polyclonal (e.g., rabbit anti-SGTA) , validated for Western blot (WB), immunohistochemistry (IHC), and immunofluorescence (IF).

Viral Pathogenesis

SGTA antibodies have elucidated SGTA's role in HIV-1 replication:

ObservationMechanismCitation
SGTA overexpression reduces HIV-1 releaseInhibits Gag processing and virion assembly
SGTA binds HIV-1 Vpu via TPR domainDisrupts viral particle release
Depletion of SGTA (siRNA) shows no significant impact on tetherin levelsSGTA’s inhibitory effect is Vpu-independent

Neurodegenerative Diseases

SGTA antibodies identified SGTA as an aggregate-interacting protein in polyglutamine (polyQ) diseases:

DiseaseSGTA ColocalizationCase Study
Spinocerebellar ataxia (SCA1/2/3)Nuclear inclusions7/8 cases
Multiple system atrophy (MSA)Glial cytoplasmic inclusions4/4 cases
Huntington’s disease (HD)Reduced aggregates upon SGTA overexpressionIn vitro models

In HD model cells, SGTA overexpression reduced polyQ aggregates by 77% compared to controls (P=0.004P = 0.004) .

Applications of SGTA Antibodies

ApplicationProtocolRecommended DilutionProduct Example
Western BlotDetects endogenous SGTA (~34 kDa)1:5,000–1:50,000Proteintech #60305-1-Ig
ImmunohistochemistryIdentifies SGTA in liver cancer tissue1:20–1:200Prospec Bio PAT19E8AT
ImmunofluorescenceLocalizes SGTA in HeLa cell cytoskeleton1:20–1:200Antibodies-Online ABIN520164

Clinical and Therapeutic Implications

  • Cancer: Reduced SGTA expression correlates with prostate cancer progression .

  • Polycystic Ovary Syndrome (PCOS): SGTA dysregulation links to androgen excess .

  • Therapeutic Target: SGTA inhibition could mitigate HIV-1 replication , while its overexpression may reduce toxic aggregates in neurodegenerative disorders .

Product Specs

Introduction
SGTA is a ubiquitously expressed protein that contains three TPR protein-protein interaction duplicates. SGTA participates as a component of the androgen receptor (AR)-chaperone-cochaperone complex, functions as a cochaperone, and participates in androgen signaling. SGTA binds directly to HSC70 and HSP70 and mediates their ATPase activity. The SGTA gene encodes a protein that can interact with the chief nonstructural protein of parvovirus H-1 and 70-kDa heat shock cognate. Additionally, SGTA interacts with Vpu and Gag from HIV-1, SARS-CoV accessory protein 7a, DNAJC5, and DNAJC5B. Because this transcript is expressed universally in several tissues, SGTA serves a housekeeping function. As it is involved in apoptosis and androgen signaling, SGTA is a possible molecule for polycystic ovary syndrome, a disorder characterized by androgen excess, obesity, and menstrual disturbances.
Physical Appearance
Sterile filtered colorless solution.
Formulation
1mg/ml containing PBS, pH-7.4, 10% Glycerol and 0.02% Sodium Azide.
Storage Procedures
For periods up to 1 month store at 4°C, for longer periods, store at -20°C. Prevent freeze-thaw cycles.
Stability / Shelf Life
12 months at -20°C. 1 month at 4°C.
Applications
The antibody has been tested by ELISA, Western blot analysis, Flow cytometry, and ICC/IF to assure specificity and reactivity. Since application varies, each investigation should be titrated by the reagent to obtain optimal results.
Synonyms
alphaSGT, hSGT, SGT, Vpu-binding protein, UBP, Small glutamine-rich tetratricopeptide repeat-containing protein alpha, Alpha-SGT, SGT1.
Purification Method
SGTA antibody was purified from mouse ascitic fluids by protein-A affinity chromatography.
Type
Mouse Anti Human Monoclonal.
Clone
PAT19E8AT.
Immunogen
Anti-human SGTA mAb, is derived from hybridization of mouse F0 myeloma cells with spleen cells from BALB/c mice immunized with a recombinant human SGTA protein 1-313 amino acids purified from E. coli.
Ig Subclass
Mouse IgG1 heavy chain and k light chain.

Q&A

What is SGTA and what are its primary functions in cellular processes?

SGTA is a co-chaperone protein that plays crucial roles in cellular protein quality control mechanisms. It primarily functions by binding misfolded and hydrophobic patches-containing client proteins in the cytosol and mediating their targeting to the endoplasmic reticulum or regulating their sorting to the proteasome when targeting fails . SGTA also participates in tail-anchored/type II transmembrane protein membrane insertion as part of a targeting module with ASNA1 and the BAG6 complex . Additionally, it collaborates with the BAG6 complex to maintain hydrophobic substrates in non-ubiquitinated states, competing with RNF126 for interaction with BAG6 to prevent ubiquitination of client proteins .

In neurodegenerative diseases, SGTA has been identified as an aggregate-interacting protein. Research demonstrates that SGTA colocalizes with intracellular aggregates in Huntington's disease (HD) cell models and neurons of HD model mice . It also interacts with intranuclear inclusions in postmortem brains of patients with polyglutamine (polyQ) diseases including various spinocerebellar ataxias and dentatorubral-pallidoluysian atrophy .

How does SGTA structure relate to its function in protein quality control?

SGTA contains three distinct structural domains, each contributing to specific aspects of its function:

  • N-terminal domain: Mediates SGTA dimerization and interaction with the BAG6 complex, facilitating its role in protein quality control pathways

  • Central TPR domain: Contains tetratricopeptide repeat motifs that interact with Hsc70 and other chaperones, forming a functional chaperone complex

  • C-terminal domain: Binds directly to hydrophobic domains of client proteins, particularly crucial for interactions with polyQ aggregates and other misfolded proteins

This domain organization enables SGTA to function as a bridge between misfolded client proteins and the cellular protein quality control machinery. In vitro studies have shown that SGTA binds to polyQ aggregates specifically through its C-terminal domain, and SGTA overexpression can reduce intracellular aggregates, suggesting a protective role .

What are the most important considerations when selecting an SGTA antibody for neurodegenerative disease research?

When selecting an SGTA antibody for neurodegenerative disease research, researchers should consider:

  • Validated reactivity with aggregated proteins: Choose antibodies proven to detect SGTA in the context of intracellular aggregates. Research has shown that SGTA colocalizes with neuronal intranuclear inclusions in polyQ diseases and with glial cytoplasmic inclusions in multiple system atrophy (MSA) .

  • Cross-species compatibility: Select antibodies that recognize SGTA in both human tissue samples and relevant animal models (such as R6/2 mice for HD studies) to facilitate translational research .

  • Domain specificity: Consider antibodies targeting specific SGTA domains based on your research question. For studying interactions with polyQ aggregates, antibodies targeting the C-terminal domain may be most relevant .

  • Compatibility with multiple applications: Verify the antibody works in all required techniques (immunohistochemistry, western blotting, immunoprecipitation) and with different sample preparations (fixed tissue, protein lysates) .

  • Validation in specific disease contexts: Check if the antibody has been used successfully in your disease model of interest, as SGTA shows differential association with aggregates across neurodegenerative conditions (present in polyQ diseases and MSA, but not in Parkinson's disease or ALS) .

How does SGTA interact with polyglutamine aggregates in Huntington's disease and other polyQ disorders?

SGTA interacts with polyglutamine (polyQ) aggregates in Huntington's disease and other polyQ disorders through specific molecular mechanisms:

  • Immunohistochemistry studies have demonstrated that SGTA colocalizes with neuronal intranuclear inclusions (NIIs) in the CA1 region of HD model mouse brains, confirmed by double immunofluorescence staining with antibodies against SGTA and ubiquitin .

  • In postmortem human brain tissue from patients with polyQ diseases (SCA1, SCA2, SCA3, and DRPLA), SGTA localizes to NIIs in the pontine nucleus, verified through double immunofluorescence staining using anti-SGTA antibody and anti-expanded polyQ antibody (1C2) .

  • Molecular analysis reveals that SGTA binds to polyQ aggregates specifically through its C-terminal domain, which has an affinity for hydrophobic protein regions .

  • The interaction appears to occur with insoluble polyQ aggregates rather than soluble monomers or oligomers. Immunoprecipitation experiments with anti-GFP antibody failed to detect interaction with soluble tNhtt-polyQ, suggesting SGTA associates primarily with insoluble aggregated forms .

  • Notably, SGTA's binding partner BAG6 was not detected in the same aggregates, indicating that SGTA's interaction with polyQ aggregates occurs through alternative pathways distinct from its BAG6-mediated functions .

Why does SGTA colocalize with glial cytoplasmic inclusions in MSA but not with Lewy bodies in Parkinson's disease?

The differential association of SGTA with α-synuclein aggregates in different neurodegenerative diseases reveals important disease-specific mechanisms:

  • In MSA patient brains, SGTA strongly colocalizes with glial cytoplasmic inclusions (GCIs), a pathological hallmark of this disease. Double immunofluorescence staining confirmed that SGTA colocalizes with phosphorylated α-synuclein in these GCIs .

  • In contrast, in Parkinson's disease (PD) brains, SGTA is diffusely detected in the cytoplasm of neurons but does not colocalize with the phosphorylated α-synuclein-reactive portions of Lewy bodies. In some mature Lewy bodies with halo structure, SGTA antibody only weakly reacted with the core where no phosphorylated α-synuclein was detected .

  • Similarly, in ALS patient spinal cords, SGTA shows diffuse cytoplasmic distribution in neurons but does not associate with neuronal cytoplasmic inclusions containing phosphorylated TDP-43 .

This selective colocalization pattern suggests fundamental differences between aggregate types, potentially including:

  • Different conformational structures of α-synuclein in GCIs versus Lewy bodies

  • Cell-type specific processing mechanisms between oligodendrocytes (in MSA) and neurons (in PD)

  • Varying protein quality control systems in different cell populations

  • Distinct post-translational modifications affecting SGTA binding sites

What experimental evidence suggests SGTA might modify disease progression in polyQ disorders?

Several experimental findings suggest SGTA may modify disease progression in polyQ disorders:

  • Overexpression studies demonstrated that SGTA enhances the solubility of intracellular aggregates in HD model cells, as evidenced by filter trap assay results .

  • Flow cytometric analysis revealed that SGTA specifically reduces small and presumably insoluble polyQ aggregates (weak fluorescent signals), similar to the effect observed with the established chaperone Hdj1 .

  • SGTA binding to polyQ aggregates through its C-terminal domain suggests a direct molecular interaction with the pathological protein species .

  • The consistent colocalization of SGTA with polyQ-positive neuronal intranuclear inclusions across multiple polyQ diseases (SCA1, SCA2, SCA3, DRPLA) indicates a common interaction mechanism that may be therapeutically relevant .

  • SGTA's known roles in intracellular protein homeostasis, combined with its selective association with disease-specific aggregates, positions it as a potential modifier of aggregate formation and toxicity .

These findings collectively suggest that SGTA may function as part of an endogenous protective response to polyQ aggregation, potentially serving as a therapeutic target for modifying disease progression through enhancement of its activity .

What are the optimal conditions for immunohistochemical detection of SGTA in brain tissue samples?

Optimizing immunohistochemical detection of SGTA in brain tissue requires careful consideration of several technical parameters:

Tissue Preparation:

  • Both formalin-fixed, paraffin-embedded sections and fresh-frozen tissue can be used for SGTA detection, though each requires specific processing .

  • Antigen retrieval is critical: Studies detecting SGTA in postmortem brain tissue from polyQ disease patients and MSA patients successfully utilized antigen retrieval techniques prior to immunostaining .

  • Section thickness of 5-7 μm provides optimal results for visualizing both diffuse cytoplasmic SGTA and its accumulation in pathological inclusions .

Antibody Conditions:

  • Antibody selection should consider the specific epitope—antibodies targeting the C-terminal domain may be particularly useful for detecting SGTA in polyQ aggregates .

  • For double immunofluorescence studies, compatibility with other primary antibodies (such as anti-expanded polyQ or anti-phosphorylated α-synuclein) should be verified .

  • Controls should include tissue sections from areas known to express SGTA diffusely (such as normal neuronal cytoplasm) as internal positive controls .

Visualization Methods:

  • Both chromogenic detection (DAB visualization) and fluorescence microscopy have been successfully used to detect SGTA in brain tissue .

  • For colocalization studies with aggregate markers, confocal microscopy with appropriate spectral separation is recommended to confirm true colocalization versus coincidental overlay .

Disease-Specific Considerations:

  • For polyQ diseases: Co-staining with 1C2 antibody (recognizing expanded polyQ) can confirm SGTA association with disease-specific inclusions .

  • For MSA: Anti-phosphorylated α-synuclein antibody is the preferred co-stain for confirming SGTA localization to GCIs .

  • For control studies in PD and ALS: Include appropriate disease-specific markers (phosphorylated α-synuclein for PD, phosphorylated TDP-43 for ALS) to confirm the specific association pattern .

How can researchers distinguish between non-specific binding and genuine SGTA-aggregate interactions?

Distinguishing genuine SGTA-aggregate interactions from non-specific binding requires rigorous controls and validation approaches:

Essential Controls:

  • Staining controls: Include sections stained with isotype-matched non-specific IgG and sections with primary antibody omitted to assess background staining .

  • Specificity controls: When possible, include tissue from SGTA knockout/knockdown models, or use competitive peptide blocking to confirm antibody specificity .

  • Multiple antibody verification: Confirm key findings using alternative SGTA antibodies targeting different epitopes to rule out epitope-specific artifacts .

Validation Approaches:

  • Biochemical correlation: Complement immunohistochemistry with biochemical fractionation studies to verify SGTA presence in insoluble protein fractions containing disease-specific aggregates .

  • Domain mapping: Use domain-specific antibodies or expression of domain deletion constructs to identify which SGTA regions mediate the interaction with aggregates, as demonstrated with the C-terminal domain binding to polyQ aggregates .

  • Reciprocal validation: Perform both immunoprecipitation of SGTA to detect aggregate proteins and immunoprecipitation of aggregate proteins to detect SGTA .

Experimental Design Considerations:

  • Quantitative assessment: Develop objective metrics for colocalization, such as calculating the percentage of aggregates containing SGTA signal or measuring the intensity ratio of SGTA in aggregates versus surrounding cytoplasm .

  • Cross-disease comparison: Include multiple neurodegenerative disease tissues (as done with polyQ diseases, MSA, PD, and ALS) to establish specificity patterns. The finding that SGTA associates with aggregates in polyQ diseases and MSA but not in PD or ALS provides strong evidence for specific rather than non-specific interactions .

  • Functional validation: Test whether modulating SGTA levels affects aggregate properties, as demonstrated by SGTA overexpression reducing polyQ aggregates in HD model cells .

What are the most effective experimental designs for studying SGTA's impact on protein aggregate formation?

Effective experimental designs for studying SGTA's impact on protein aggregate formation should incorporate multiple complementary approaches:

Cellular Models:

  • Expression systems: Use inducible systems to precisely control expression of both SGTA and aggregation-prone proteins. Studies in HD model cells demonstrating SGTA's effect on polyQ aggregation provide a methodological template .

  • Primary cells and relevant models: Validate findings in models closely representing the disease context, such as primary neurons for polyQ diseases or oligodendrocytes for MSA, given SGTA's differential interaction with aggregates across cell types .

Manipulation Strategies:

  • Bidirectional modulation: Both overexpression and knockdown/knockout approaches should be employed to fully characterize SGTA's role. Research has shown that SGTA overexpression enhances solubility of polyQ aggregates .

  • Domain manipulation: Generate SGTA constructs with mutations or deletions in specific domains (particularly the C-terminal domain identified as crucial for polyQ binding) to determine structure-function relationships .

Quantification Methods:

  • Filter trap assay: Quantify insoluble aggregates retained on cellulose acetate membranes, a technique successfully used to demonstrate SGTA's effect on polyQ aggregate solubility .

  • Flow cytometry: Assess aggregate size distribution in large cell populations, allowing differentiation between effects on small versus large aggregates .

  • Microscopy analysis: Combine fluorescence microscopy with image analysis to quantify aggregate number, size, and SGTA colocalization .

Analytical Framework:

  • Time-course studies: Monitor aggregate formation kinetics to distinguish effects on initial formation versus later stages of aggregation or clearance .

  • Biochemical characterization: Analyze protein solubility in multiple fractions (soluble, oligomeric, insoluble) to fully characterize SGTA's effects on aggregate properties .

  • Mechanistic investigation: Examine whether SGTA acts alone or requires other chaperones/co-chaperones, noting that BAG6 was not detected in aggregates, suggesting SGTA may function through alternative pathways in this context .

This multi-faceted approach enables comprehensive characterization of SGTA's role in aggregate dynamics across different disease contexts.

How might the interaction between SGTA and protein aggregates be exploited for therapeutic development?

The interaction between SGTA and protein aggregates presents several promising therapeutic development strategies:

Target Enhancement of Endogenous SGTA Function:

  • Small molecule screening: Develop high-throughput assays to identify compounds that enhance SGTA's binding to aggregates or increase its chaperone activity. The experimental finding that SGTA overexpression reduces intracellular aggregates provides proof-of-concept that enhancing SGTA function may be beneficial .

  • Gene therapy approaches: Design viral vectors to increase SGTA expression specifically in affected cell populations, with consideration for the differential involvement across diseases (neurons in polyQ diseases, oligodendrocytes in MSA) .

  • Domain-specific targeting: Focus on compounds that enhance the function of SGTA's C-terminal domain, which has been identified as the critical region mediating interaction with polyQ aggregates .

SGTA as a Therapeutic Delivery Vehicle:

  • Bifunctional molecules: Design constructs that leverage SGTA's aggregate-binding capacity to deliver therapeutic cargo (such as disaggregases or degradation-enhancing factors) directly to pathological inclusions .

  • Target discrimination: Exploit SGTA's selective binding to different aggregate types (binding to polyQ and MSA inclusions but not PD or ALS inclusions) to develop disease-specific therapeutic approaches .

Biomarker Applications:

  • Diagnostic tools: Develop assays detecting SGTA-aggregate complexes in accessible biospecimens as potential disease biomarkers .

  • Treatment response monitoring: Measure changes in SGTA-aggregate interaction as an indicator of therapeutic efficacy .

Considerations and Challenges:

  • Specificity concerns: Since SGTA plays multiple roles in cellular protein quality control, therapeutic modulation must avoid disrupting essential functions .

  • Cell-type specific delivery: Different approaches may be required for neuronal (polyQ diseases) versus glial (MSA) pathologies based on SGTA's differential association patterns .

  • Timing of intervention: SGTA-targeting therapies might be most effective at early disease stages before extensive aggregate formation .

What structural features of SGTA determine its selective interaction with different types of protein aggregates?

The selective interaction of SGTA with different types of protein aggregates is determined by specific structural features:

Domain-Specific Contributions:

  • C-terminal domain: Research has demonstrated that SGTA binds to polyQ aggregates specifically through its C-terminal domain, which contains glutamine-rich regions that may facilitate interactions with glutamine-rich substrates like expanded polyQ tracts .

  • Central TPR domain: Although primarily involved in chaperone interactions (e.g., with Hsc70), this domain may contribute to spatial positioning that affects aggregate binding specificity .

  • N-terminal domain: Mediates SGTA dimerization and interaction with the BAG6 complex, potentially influencing which aggregate types SGTA can engage .

Substrate Selectivity Patterns:

  • PolyQ aggregate recognition: SGTA shows strong association with aggregates in multiple polyQ diseases (SCA1, SCA2, SCA3, DRPLA), suggesting a common recognition mechanism for this aggregate type .

  • MSA GCI recognition: SGTA colocalizes with α-synuclein in oligodendrocyte GCIs, but notably not with α-synuclein in neuronal Lewy bodies in PD patients, indicating specific recognition determinants beyond simply the presence of α-synuclein .

  • Lack of interaction with ALS inclusions: SGTA does not associate with TDP-43 inclusions in ALS, suggesting it lacks affinity for the structural features of these aggregates .

Molecular Basis of Specificity:

  • Hydrophobicity profile: SGTA's C-terminal domain binds hydrophobic substrates, suggesting the exposure of hydrophobic patches in aggregates may be a key determinant .

  • Aggregate conformation: Different conformational states of the same protein (e.g., α-synuclein in MSA versus PD) likely affect SGTA binding, potentially explaining disease-specific interactions .

  • Cellular context: Cell-type specific factors in oligodendrocytes versus neurons may contribute to the differential binding pattern observed between MSA and PD inclusions .

Understanding these structural determinants could facilitate the development of agents that modulate specific SGTA-aggregate interactions for therapeutic purposes.

How does SGTA's role in protein aggregate dynamics compare to other molecular chaperones?

SGTA's role in protein aggregate dynamics shows both similarities and important distinctions compared to other molecular chaperones:

Functional Comparisons:

Substrate Specificity Patterns:

  • Selective aggregate engagement: SGTA shows remarkable selectivity in the types of aggregates it associates with (polyQ diseases and MSA but not PD or ALS), whereas some chaperones like Hsp70 have been found in a broader range of inclusion types .

  • Unique binding mechanism: SGTA binds to polyQ aggregates through its C-terminal domain, which contains glutamine-rich regions that may facilitate interactions with polyQ tracts—a specificity mechanism distinct from that of heat shock proteins .

Integration in Chaperone Networks:

  • Cooperative versus independent action: While SGTA often works with the BAG6 complex in protein quality control, its association with disease aggregates occurs independently of BAG6, suggesting context-dependent mechanisms .

  • Regulation of protein degradation: SGTA can have both pro-degradation and anti-degradation effects depending on context. It helps target misfolded proteins to the ER but can also prevent ubiquitination of client proteins by competing with RNF126 for BAG6 binding .

Therapeutic Implications:

  • Complementary targeting: The specific aggregate types SGTA associates with (polyQ and MSA inclusions) suggest it could complement other chaperones in therapeutic approaches targeting specific neurodegenerative diseases .

  • Functional enhancement: Like other chaperones, SGTA overexpression shows potential protective effects against protein aggregation, suggesting similar therapeutic potential through enhancement of chaperone activity .

This comparative understanding helps position SGTA within the broader protein quality control network and identifies its unique contributions to cellular proteostasis in health and disease.

How should researchers address contradictory findings regarding SGTA's role in aggregate formation versus clearance?

Addressing contradictory findings regarding SGTA's role in aggregate dynamics requires systematic analysis of experimental variables:

Methodological Factors to Consider:

  • Expression level effects: SGTA's impact may differ between physiological expression, moderate overexpression, and high overexpression. Filter trap assay results showing SGTA overexpression reduces polyQ aggregates should be interpreted in the context of expression level .

  • Aggregate detection methods: Different techniques (filter trap, microscopy, flow cytometry) may detect different aggregate species. Flow cytometric analysis indicated SGTA specifically reduces small, presumably insoluble polyQ aggregates but not large, mature aggregates, which may explain discrepancies between assay types .

  • Temporal dynamics: SGTA may have different effects at different stages of aggregate formation. Time-course experiments are essential to distinguish effects on formation versus clearance .

Biological Context Variations:

  • Cell-type specific effects: SGTA's differential association with aggregates in neurons versus glia suggests cell type is a critical variable. The observation that SGTA colocalizes with GCIs in oligodendrocytes but not with similar α-synuclein aggregates in neurons highlights this context-dependency .

  • Disease-specific patterns: SGTA associates with aggregates in polyQ diseases and MSA but not in PD or ALS, indicating disease-specific mechanisms that should be considered when interpreting apparently contradictory findings .

  • Substrate-specific interactions: SGTA binds polyQ aggregates through its C-terminal domain, suggesting substrate-specific mechanisms that may explain different outcomes with different aggregate types .

Reconciliation Strategies:

  • Unified theoretical framework: Develop models incorporating both pro-clearance and pro-formation activities based on concentration, timing, and cellular context .

  • Domain-specific function analysis: Since SGTA functions through distinct domains, determine if contradictions stem from domain-specific effects by utilizing truncation or mutation constructs .

  • Interaction partner analysis: Consider the involvement of SGTA's binding partners. The finding that BAG6 was not detected in aggregates suggests SGTA may function through alternative pathways in aggregate biology compared to its canonical roles .

By systematically addressing these variables, researchers can transform apparently contradictory data into a more nuanced understanding of SGTA's context-dependent functions in protein aggregate dynamics.

What controls are essential for validating antibody specificity when studying SGTA in neurodegenerative disease models?

Validating antibody specificity is crucial when studying SGTA in neurodegenerative disease contexts, requiring multiple essential controls:

Negative Controls for Specificity:

  • Genetic validation: When possible, include SGTA knockout/knockdown samples as the gold standard negative control .

  • Peptide competition: Pre-incubate the SGTA antibody with excess recombinant SGTA protein or immunizing peptide to block specific binding sites .

  • Non-specific IgG: Include isotype-matched non-specific IgG control antibodies to establish background staining levels .

  • Primary antibody omission: Process tissue sections identically but omit the primary antibody to assess secondary antibody specificity .

Positive Controls for Validation:

  • Multiple antibody verification: Confirm key findings using alternative SGTA antibodies targeting different epitopes. The consistent pattern of SGTA localization to specific inclusion types strengthens confidence in specificity .

  • Known expression patterns: Validate that the expected cytoplasmic distribution of SGTA is observed in normal cells, as demonstrated in control mouse brain showing diffuse cytoplasmic SGTA distribution in neurons .

  • Overexpression systems: Include samples from cells overexpressing SGTA as positive controls for antibody sensitivity and specificity .

Application-Specific Controls:

  • For immunohistochemistry: Include both disease-affected and unaffected brain regions to compare SGTA's normal distribution versus pathological localization, as shown in studies comparing SGTA distribution in various neurological disease tissues .

  • For co-localization studies: Include single-stained sections to rule out bleed-through when performing double immunofluorescence, as was done in studies co-staining for SGTA and aggregate markers like ubiquitin, expanded polyQ, or phosphorylated α-synuclein .

  • For biochemical studies: Perform western blots to confirm antibody specificity at the expected molecular weight before using for other applications, and include multiple protein fractions to assess specificity across different solubility states .

Disease Context Controls:

  • Cross-disease comparison: Include tissues from multiple neurodegenerative conditions as internal controls. The finding that SGTA associates with aggregates in polyQ diseases and MSA but not in PD and ALS provides strong internal validation of specific recognition patterns .

  • Cross-species validation: Confirm findings across species when possible, as demonstrated by similar SGTA localization patterns in HD model mice and human polyQ disease tissues .

These comprehensive controls ensure that observed SGTA patterns represent genuine biological phenomena rather than technical artifacts.

How can researchers establish causality versus correlation in studies of SGTA and protein aggregation?

Establishing causality versus correlation in SGTA-aggregate studies requires multifaceted experimental approaches:

Intervention Studies:

  • Bidirectional manipulation: Both overexpression and knockdown/knockout studies are necessary. Research has shown that SGTA overexpression reduces intracellular aggregates in HD model cells, suggesting a causal relationship .

  • Rescue experiments: Demonstrate that reintroducing SGTA after knockdown restores the original phenotype, and that this rescue depends on specific functional domains (particularly the C-terminal domain identified as critical for polyQ binding) .

  • Structure-function analysis: The finding that SGTA binds to polyQ aggregates specifically through its C-terminal domain provides mechanistic insight supporting causality. Creating domain deletion or point mutation constructs can further establish which specific features are causally linked to observed effects .

Temporal Relationship Analysis:

  • Time-course studies: Determine whether SGTA association precedes changes in aggregate properties or vice versa. Longitudinal studies following aggregate formation and SGTA recruitment over time can help establish temporal precedence .

  • Inducible systems: Use conditional expression systems to control the timing of SGTA expression relative to aggregate formation .

Dose-Dependency Assessment:

  • Titration experiments: Demonstrate that varying levels of SGTA produce proportional effects on aggregate measures. Flow cytometric analysis showing SGTA reduces small polyQ aggregates provides quantitative data supporting a functional relationship .

  • Threshold effects: Identify whether specific SGTA expression levels are required for effects on aggregation, which would strengthen evidence for causality .

Mechanistic Validation:

  • Interaction requirements: The observation that SGTA does not interact with soluble monomers or oligomers of tNhtt-polyQ but does bind insoluble aggregates provides mechanistic specificity supporting a causal relationship .

  • Pathway intervention: Test whether blocking specific downstream pathways prevents SGTA's effects on aggregates .

  • Partner dependency: The finding that BAG6 does not colocalize with polyQ aggregates in HD model mouse brains and postmortem polyQ disease brains suggests SGTA's effect occurs through alternative pathways, providing mechanistic insight .

Control for Confounding Factors:

  • Cell-type specificity: Control for cell-type dependent factors that might explain SGTA's differential association with aggregates in oligodendrocytes versus neurons (as seen in MSA versus PD) .

  • Disease-specific factors: Account for disease-specific variables when comparing SGTA's role across conditions. The specific association with aggregates in polyQ diseases and MSA but not in PD or ALS suggests disease-specific mechanisms rather than a universal effect .

By integrating these approaches, researchers can move beyond correlative observations to establish mechanistic understanding of SGTA's causal role in protein aggregation processes.

Product Science Overview

Introduction

Small Glutamine-Rich Tetratricopeptide Repeat-Containing Protein Alpha (SGTA) is a 35 kDa protein involved in various biological processes. It is known for its role as a co-chaperone and regulator of androgen and growth hormone receptor signaling. SGTA has been studied in the context of several diseases, including polycystic ovary syndrome (PCOS) and non-small-cell lung cancer (NSCLC).

Structure and Function

SGTA contains tetratricopeptide repeat (TPR) motifs, which are known for mediating protein-protein interactions. These motifs allow SGTA to interact with other proteins, including androgen receptors (AR) and growth hormone receptors (GHR). SGTA acts as a molecular co-chaperone, assisting in the proper folding and functioning of these receptors .

Role in Polycystic Ovary Syndrome (PCOS)

SGTA has been identified as a candidate gene for PCOS, a complex genetic disease characterized by hyperandrogenism, menstrual dysfunction, and polycystic ovarian morphology. Studies have shown that SGTA may play a role in androgen signaling as a co-chaperone. Polymorphisms in the SGTA gene have been associated with PCOS, suggesting a potential role in its pathogenesis .

Role in Non-Small-Cell Lung Cancer (NSCLC)

SGTA has also been implicated in the tumorigenesis of NSCLC. It is highly expressed in NSCLC tissues and is significantly correlated with histological differentiation, clinical stage, and proliferation markers such as Ki-67. Suppression of SGTA expression has been shown to reduce the proliferation of NSCLC cells, indicating its potential as a therapeutic target .

Other Biological Roles

SGTA is involved in various other biological processes, including endocytosis, viral replication, mitosis, apoptosis, and cell division. It interacts with intracellular aggregates in neurodegenerative diseases, such as Huntington’s disease, highlighting its diverse functional roles .

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2024 Thebiotek. All Rights Reserved.