Recombinant Human Spermatogenesis-associated protein 12 (SPATA12)

Shipped with Ice Packs
In Stock

Description

Introduction to Recombinant Human Spermatogenesis-associated protein 12 (SPATA12)

Recombinant Human Spermatogenesis-associated protein 12 (SPATA12) is a protein associated with spermatogenesis, a complex process of cell development and differentiation in the male reproductive system. SPATA12 is specifically expressed in germ cells and is believed to play a crucial role in the development of testicular cells. This article will delve into the characteristics, functions, and research findings related to SPATA12, highlighting its significance in both physiological and pathological contexts.

Characteristics and Expression of SPATA12

SPATA12 is identified as a stage-specific and germ cell-specific gene, suggesting its involvement in the development of testicular cells . It is primarily expressed in the testis, indicating its role in spermatogenesis. The protein's function is thought to be related to nuclear activities, given its predicted involvement in DNA/RNA binding and protein interactions .

Gene Expression and Regulation

Studies using microarray analysis have shown that SPATA12 influences the expression of numerous genes involved in various biological processes. Upon transfection of SPATA12 into GC-1 spg cells, 182 genes were upregulated and 104 genes were downregulated, with significant changes in genes related to immune responses, transcription regulation, and metabolic processes .

CategoryUpregulated GenesDownregulated Genes
Immune ResponseAntigen processing and presentation, T cell-mediated cytotoxicity-
Metabolic Processes-Carbohydrate metabolism, lipid metabolism, cholesterol biosynthesis
Transcription RegulationTranscription factors-

Pathway Analysis

Pathway analysis indicates that SPATA12 is involved in several signaling pathways, including the Wnt/β-catenin pathway. The downregulation of β-catenin and its downstream target cyclin E1 suggests that SPATA12 may negatively regulate cell cycle progression .

PathwayKey Genes Affected
Wnt/β-cateninβ-catenin, cyclin E1
Immune ResponseAntigen processing and presentation, T cell receptor signaling

Role in Spermatogenesis and Tumorigenesis

SPATA12 has been implicated as an inhibitor in both spermatogenesis and tumorigenesis. Its expression may influence the balance between cell proliferation and differentiation, potentially affecting cancer development .

Oxidative Damage and DNA Repair

Research suggests that SPATA12 might play a role in protecting against oxidative damage, which is crucial for maintaining genomic integrity during spermatogenesis .

Potential Therapeutic Targets

Understanding the function of SPATA12 could provide insights into novel therapeutic strategies for male infertility and cancer treatment. Its involvement in spermatogenesis and cell cycle regulation makes it a potential target for fertility control or cancer therapy.

Product Specs

Form
Lyophilized powder
Note: While we prioritize shipping the format currently in stock, please specify your preferred format in order notes for customized preparation.
Lead Time
Delivery times vary depending on purchasing method and location. Please contact your local distributor for precise delivery estimates.
Note: Standard shipping includes blue ice packs. Dry ice shipping requires prior arrangement and incurs additional charges.
Notes
Avoid repeated freeze-thaw cycles. Store working aliquots at 4°C for up to one week.
Reconstitution
Centrifuge the vial briefly before opening to consolidate the contents. Reconstitute the protein in sterile deionized water to a concentration of 0.1-1.0 mg/mL. For long-term storage, we recommend adding 5-50% glycerol (final concentration) and aliquoting at -20°C/-80°C. Our standard glycerol concentration is 50% and can serve as a reference.
Shelf Life
Shelf life depends on storage conditions, buffer composition, temperature, and protein stability. Generally, liquid formulations have a 6-month shelf life at -20°C/-80°C, while lyophilized formulations have a 12-month shelf life at -20°C/-80°C.
Storage Condition
Store at -20°C/-80°C upon receipt. Aliquot for multiple uses. Avoid repeated freeze-thaw cycles.
Tag Info
Tag type is determined during manufacturing.
The tag type is determined during production. If you require a specific tag, please inform us, and we will prioritize its development.
Synonyms
SPATA12; SRG5; Spermatogenesis-associated protein 12; Spermatogenesis-related protein 5
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
1-190
Protein Length
full length protein
Purity
>85% (SDS-PAGE)
Species
Homo sapiens (Human)
Target Names
SPATA12
Target Protein Sequence
MSSSALTCGS TLEKSGDTWE MKALDSSRLV PWPPRGLGSS TQHPNKPHCA LASCQGPGVL PGAASALPEL TFQGDVCQSE TCQRYLQAAI SLDIAVSQIN LLGRPSSPPA LLIQQGSCEQ VIHNSTPQFL GMEDGDNERT TGWLWRLCED IDAEPSSTGC SRSNQLTFTE GCFVRSLSTV YSNTHIHTHL
Uniprot No.

Q&A

What is the primary function of SPATA12 in human cells?

SPATA12 (Spermatogenesis-associated protein 12) primarily functions as an inhibitor in both spermatogenesis and tumorigenesis. Research has demonstrated that SPATA12 plays a significant role in maintaining genomic integrity. It responds to cellular stress conditions, particularly oxidative damage and UV-mediated DNA damage, suggesting its importance in cellular protective mechanisms .

The protein's inhibitory function in spermatogenesis indicates its role as a regulatory factor in male reproductive development, while its inhibitory effect in tumorigenesis suggests potential tumor suppressor activity that warrants further investigation in cancer research contexts .

How is SPATA12 expression regulated in cellular stress conditions?

SPATA12 expression is upregulated in response to oxidative stress, particularly through a transcription factor activator protein-1 (AP-1) mediated pathway. Studies have established cellular models of oxidative stress using H₂O₂ treatment and demonstrated that:

  • H₂O₂ significantly upregulates SPATA12 expression in a dose-dependent manner

  • The AP-1 binding site within the SPATA12 core promoter (77-302 bp) is critical for this response

  • The binding site encompasses the sequence 5′-TGAGTCA-3′, a core sequence in the AP-1 motif also known as the TPA responsive element (TRE)

Through luciferase reporter gene assays, researchers have confirmed that the TRE element within the SPATA12 promoter is crucial for responding to H₂O₂ treatment. When this element is mutated or deleted, the relative luciferase activity of the promoter is significantly reduced in cells exposed to H₂O₂, indicating that AP-1 is directly involved in the H₂O₂-induced transcriptional upregulation of SPATA12 .

Additionally, SPATA12 expression can be induced by UV-C radiation, with its promoter activity being upregulated in response to UV-C exposure through similar AP-1 mediated mechanisms .

What cellular localization patterns does SPATA12 exhibit?

SPATA12 primarily exhibits nuclear localization patterns, particularly when interacting with its binding partner CHD2 (chromodomain helicase DNA binding protein 2). This localization has been confirmed through:

  • Bimolecular fluorescence complementation (BiFC) assays

  • Subcellular co-localization experiments using confocal microscopy

  • Fluorescence microscopy studies with tagged proteins

These techniques have demonstrated that SPATA12 and CHD2 interact in the nucleus, suggesting that SPATA12's function may be closely related to nuclear processes such as transcription regulation and DNA damage response mechanisms .

What are the most effective methods for studying SPATA12-protein interactions?

Multiple complementary techniques have proven effective for investigating SPATA12 protein interactions:

Yeast Two-Hybrid Screening System:
This powerful genetic strategy successfully identified CHD2 as an interacting partner of SPATA12. The methodology involves:

  • Generating a bait construct by amplifying the SPATA12 open reading frame and inserting it into the pGBKT7 vector

  • Transforming the bait plasmid into yeast strain AH109

  • Screening a human testis cDNA library in the pGADT7-Rec vector

  • Identifying positive colonies through their ability to grow on selective media and verification by β-galactosidase assays

Bimolecular Fluorescence Complementation (BiFC):
This technique visualizes protein interactions in living cells by:

  • Creating fusion constructs of SPATA12 and potential interacting proteins with complementary fragments of a fluorescent protein

  • Transfecting cells with both constructs

  • Observing fluorescence reconstitution when the proteins interact

  • Analyzing using laser scanning confocal microscopy

Subcellular Co-localization:
Verification of protein interactions through:

  • Creating fluorescent protein-tagged constructs (e.g., pEGFP-SPATA12 and pDsRed-CHD2)

  • Co-transfecting cells with both constructs

  • Visualizing the localization patterns using confocal microscopy

  • Analyzing overlap in subcellular distribution

These methods have collectively established that SPATA12 interacts with CHD2 in the nucleus, providing insights into its potential role in transcriptional regulation and DNA damage response pathways .

What cellular models are most appropriate for studying SPATA12 function in oxidative stress?

Based on published research, the following cellular models have proven effective for studying SPATA12 function in oxidative stress:

HeLa and MCF-7 Cell Lines:
These established human cancer cell lines have been successfully used to:

  • Create oxidative stress models using H₂O₂ treatment

  • Transfect with SPATA12 expression vectors

  • Evaluate the effects of SPATA12 on oxidative stress parameters

Oxidative Stress Parameters to Evaluate:
When establishing these models, researchers typically assess:

ParameterMeasurement MethodExpected Results with SPATA12
Superoxide dismutase (SOD) activityBiochemical assayNo significant change observed
Glutathione (GSH) contentBiochemical assayNo significant change observed
Malondialdehyde (MDA) levelsBiochemical assaySignificant reduction (p<0.05 in HeLa, p<0.01 in MCF-7)
Reactive oxygen species (ROS)DCFH-DA fluorescenceSignificant reduction compared to control

Induction Protocol:
For H₂O₂-induced oxidative stress, researchers typically:

  • Treat cells with varying concentrations of H₂O₂ (typically 20-100 μM)

  • Incubate for defined periods (typically 12-24 hours)

  • Verify oxidative stress induction by measuring decreased SOD activity, decreased GSH levels, and increased MDA levels

This model is particularly valuable because it allows direct observation of SPATA12's protective effects against oxidative damage through its ability to reduce ROS and MDA levels, even without altering SOD activity or GSH content .

How can researchers effectively measure SPATA12's impact on cellular response to DNA damage?

Several experimental approaches have been validated for measuring SPATA12's impact on cellular response to DNA damage:

Colony Formation Assay:
This technique assesses the effects of SPATA12 on cell proliferation and survival following DNA damage by:

  • Transfecting cells with SPATA12 expression vectors

  • Exposing cells to UV-C radiation or other DNA damaging agents

  • Allowing colonies to form over 1-2 weeks

  • Quantifying and comparing colony numbers and sizes between SPATA12-expressing and control cells

Host Cell Reactivation Assay:
This approach evaluates the influence of SPATA12 on DNA repair capacity by:

  • Damaging a reporter plasmid (typically containing luciferase gene) with UV-C radiation

  • Co-transfecting cells with the damaged reporter and SPATA12 expression vectors

  • Measuring reporter gene activity after allowing cells time for repair

  • Comparing repair efficiency between SPATA12-expressing and control cells

Flow Cytometry Analysis:
Flow cytometry provides insights into SPATA12's effects on cell cycle progression and apoptosis by:

  • Transfecting cells (H1299, MCF-7, HeLa) with SPATA12

  • Exposing cells to DNA damage

  • Staining with appropriate markers (e.g., Annexin V/PI for apoptosis)

  • Analyzing cell cycle distribution and apoptotic fractions

  • Comparing results between wild-type and p53-deficient cells to understand p53 dependency

Hoechst Staining:
This technique visualizes nuclear morphology to assess apoptosis by:

  • Treating cells with DNA damaging agents (e.g., 70 μM H₂O₂)

  • Staining with Hoechst dye

  • Observing chromatin condensation as an indicator of apoptosis

  • Comparing results between SPATA12-expressing and control cells

Research has shown that SPATA12 expression inhibits H₂O₂-induced apoptosis, as evidenced by decreased chromatin condensation in the superposition field of view following SPATA12 gene transfection . Additionally, colony formation assays demonstrate that SPATA12 leads to inhibition of cellular proliferation following UV-C-irradiated DNA damage .

How does SPATA12 interact with the DNA damage response pathway?

SPATA12 intersects with the DNA damage response pathway through several key mechanisms:

Interaction with CHD2:
SPATA12 directly interacts with CHD2 (chromodomain helicase DNA binding protein 2), which has been established through:

  • Yeast two-hybrid screening

  • Bimolecular fluorescence complementation

  • Subcellular co-localization assays

This interaction is particularly significant because CHD2 is a chromatin-remodeling factor required for genomic stability maintenance and is involved in the later stage of the DNA damage response pathway by influencing p53 transcriptional activity .

p53 Pathway Connection:
SPATA12 demonstrates a biological association with p53 in UV-C-irradiated DNA damage, as evidenced by:

  • Flow cytometry results showing differential effects in p53-positive vs. p53-negative cell lines

  • Investigations showing SPATA12 can upregulate p53 expression

The p53 connection is crucial, as p53 is a central coordinator of the cellular response to DNA damage, regulating cell cycle arrest, DNA repair, and apoptosis.

AP-1 Mediated Induction:
During DNA damage processes:

  • AP-1 is activated through phosphorylation pathways, primarily via c-Jun N-terminal kinase (JNK)

  • Activated AP-1 binds to the SPATA12 promoter region

  • This binding increases SPATA12 transcription

  • Elevated SPATA12 levels then contribute to cellular responses to DNA damage

The AP-1 binding site within the SPATA12 core promoter is critical for this response pathway, forming a regulatory axis connecting cellular stress, AP-1 activation, SPATA12 upregulation, and subsequent protective responses .

What is the relationship between SPATA12 and reactive oxygen species (ROS) in cellular damage?

SPATA12 exhibits a complex relationship with reactive oxygen species (ROS) that positions it as a key antioxidant factor:

Antioxidant Function:
Experimental evidence demonstrates SPATA12's antioxidant properties through:

  • Reduction of intracellular ROS levels as measured by DCFH-DA oxidation

  • Significant decrease in malondialdehyde (MDA) content, a marker of lipid peroxidation

  • Inhibition of H₂O₂-induced apoptosis

The relationship is bidirectional:

  • ROS (specifically H₂O₂) induces SPATA12 expression

  • SPATA12, once expressed, reduces ROS levels

Selective Antioxidant Mechanisms:
Interestingly, SPATA12 exhibits selective effects on oxidative parameters:

Oxidative ParameterEffect of SPATA12Statistical Significance
ROS levels (DCFH-DA)Decreasedp<0.05 vs. control group
MDA contentDecreasedp<0.05 in HeLa, p<0.01 in MCF-7
SOD activityNo significant change-
GSH contentNo significant change-

This selective pattern suggests that SPATA12 functions as an antioxidant not by enhancing conventional antioxidant systems (SOD, GSH) but through direct or indirect mechanisms that reduce ROS generation or enhance ROS neutralization through alternative pathways .

ROS-Mediated Signaling:
Beyond direct antioxidant effects, SPATA12 appears integrated in ROS-mediated signaling:

  • ROS activates JNK in cells

  • Activated JNK phosphorylates c-Jun and c-Fos proteins

  • This induces AP-1 transcriptional activity

  • AP-1 binds to the SPATA12 promoter, increasing its expression

  • Elevated SPATA12 then reduces ROS levels, potentially creating a negative feedback loop

This relationship positions SPATA12 as a potential therapeutic target for conditions characterized by oxidative stress and suggests its relevance in diverse pathological contexts beyond reproductive biology .

What are the contradictory findings in SPATA12 research and how can they be reconciled?

Several contradictory or seemingly paradoxical findings exist in SPATA12 research that require careful analysis:

Contradictory Finding 1: Inhibitory vs. Protective Roles
SPATA12 is described as an "inhibitor" in spermatogenesis and tumorigenesis, yet plays a "protective" role against oxidative damage and apoptosis.

Evidence for Inhibitory FunctionEvidence for Protective Function
Inhibits spermatogenesis processes Protects against H₂O₂-induced oxidative damage
Acts as an inhibitor of testicular tumorigenesis Reduces ROS and MDA levels in stressed cells
Inhibits cellular proliferation after DNA damage Inhibits H₂O₂-induced apoptosis

Reconciliation: These findings can be reconciled by understanding SPATA12 as a context-dependent regulator. Its inhibitory effects on proliferation and development likely represent protective mechanisms against genomic instability. The seemingly contradictory functions actually serve the unified purpose of maintaining cellular integrity under different conditions.

Contradictory Finding 2: Promoter Activity Variations
Research shows inconsistent activity patterns of the SPATA12 promoter regions:

ObservationContradictory Element
Full-length promoter (pGL3-958/302) activity increases with H₂O₂ Activity of pGL3-946/90 remains very low regardless of H₂O₂ treatment
Core promoter (pGL3-136/302) activity increases with H₂O₂ Suggests potential negative regulatory elements in specific regions

Reconciliation: Researchers hypothesize "that there may be a negative regulatory element located at the −946 to 90 bp region that may affect the transcriptional activity of the SPATA12 promoter" . This suggests a complex regulatory mechanism with multiple control elements that respond differently to cellular conditions.

Contradictory Finding 3: Relationship with p53
The relationship between SPATA12 and p53 shows some paradoxical elements:

ObservationApparent Contradiction
SPATA12 interacts with CHD2, which influences p53 activity The exact mechanism of how SPATA12 affects p53 function remains unclear
SPATA12 can upregulate p53 expression The pathway for this upregulation isn't fully established

Reconciliation: These contradictions highlight knowledge gaps rather than true contradictions. Future research should focus on the precise mechanisms by which SPATA12 influences p53 activity, potentially through its interaction with CHD2 or through independent pathways.

To address these contradictions, researchers should consider:

  • Conducting studies with tissue-specific or conditional SPATA12 knockout/knockin models

  • Performing detailed time-course analyses to understand temporal aspects of SPATA12 function

  • Utilizing proteomics approaches to identify the complete interactome of SPATA12

  • Employing ChIP-seq to identify all genomic regions affected by SPATA12 activity

How can resveratrol be utilized in SPATA12 research?

Resveratrol presents a valuable tool for SPATA12 research, offering a non-toxic approach to modulate SPATA12 expression:

Mechanism of Action:
Resveratrol enhances SPATA12 expression through:

  • Activation of AP-1 (activator protein-1)

  • Increased binding of AP-1 to the TRE element within the SPATA12 promoter

  • Upregulation of SPATA12 mRNA transcription

Experimental Applications:
Researchers can employ resveratrol in SPATA12 studies through several approaches:

Research ApplicationMethodologyExpected Outcome
SPATA12 activationTreat cells with 20 μM resveratrol for 12hSignificant increase in SPATA12 mRNA (p<0.01 in HeLa, p<0.05 in MCF-7)
Promoter activity analysisDual luciferase reporter assay with pGL3-136/302 promoter after resveratrol treatmentIncreased luciferase activity, which decreases when TRE is mutated/deleted
Antioxidant pathway studiesCombine resveratrol with oxidative stressorsEnhanced protective effects against oxidative damage

Advantages as a Research Tool:

  • Non-toxic activation: At 20 μM concentration, resveratrol shows no cytotoxicity while effectively increasing SPATA12 expression

  • Specificity: Works through the AP-1 pathway, providing a targeted approach

  • Accessibility: Widely available and well-characterized compound

  • Translational potential: Natural compound with established biological effects

Research Design Considerations:
When incorporating resveratrol in SPATA12 research:

  • Verify cytotoxicity profiles using MTT assays for each cell line

  • Establish dose-response curves (research indicates 20 μM for 12h as effective)

  • Include appropriate controls (AP-1 inhibitors, promoter constructs with mutated AP-1 sites)

  • Consider potential pleiotropic effects of resveratrol when interpreting results

This approach offers a valuable alternative to genetic overexpression, allowing for inducible and potentially reversible modulation of SPATA12 levels in experimental systems.

How can SPATA12 research contribute to understanding contradictions in genomic data?

SPATA12 research offers valuable insights into resolving contradictions in genomic data through several methodological approaches:

Resolving Expression Data Contradictions:
SPATA12 demonstrates context-dependent expression patterns that can help researchers understand:

  • How the same gene can show different expression profiles under various cellular conditions

  • The importance of experimental context when interpreting seemingly contradictory datasets

  • How stress-responsive genes may appear inconsistently expressed across datasets collected under different conditions

Methodological Framework for Resolving Contradictions:
Researchers studying contradictions in genomic data can apply lessons from SPATA12 research:

Contradiction TypeSPATA12 Research ApproachApplication to Other Genomic Research
Tissue-specific expression discrepanciesComprehensive analysis across multiple tissues and developmental stagesSystematic comparison of expression data across tissue types and conditions
Temporal expression variationsTime-course studies following stimulation (e.g., H₂O₂, UV-C)Temporal analysis of gene expression in response to various stimuli
Functional annotation conflictsMulti-method verification (yeast two-hybrid, BiFC, co-localization)Integration of multiple experimental approaches to verify functional annotations

Analytical Techniques from SPATA12 Research:
The statistical and analytical approaches used in SPATA12 research provide a template for addressing contradictions:

  • ANOVA with post hoc Dunnett's test to determine significant differences among groups

  • Expression of results as mean ± standard deviation

  • Use of SPSS software (version 17.0) with p<0.05 considered statistically significant

Application to Broader Contradiction Analysis:
SPATA12 research demonstrates how apparent contradictions can be systematically addressed:

  • Identification of specific regions (e.g., promoter regions) responsible for differential responses

  • Investigation of pathway-specific effects (e.g., AP-1 mediated vs. other pathways)

  • Context-dependent analysis (e.g., different effects in different cell types or under different stress conditions)

These approaches parallel established contradiction analysis frameworks like those developed by Stanford's Contradiction Corpora project, which employs a systematic methodology for identifying and classifying contradictions in text data .

What future research directions are most promising for SPATA12?

Based on current findings and knowledge gaps, several promising research directions for SPATA12 warrant exploration:

Therapeutic Applications in Oxidative Stress-Related Disorders:
Given SPATA12's protective role against oxidative damage, investigation into its potential therapeutic applications is warranted:

  • Evaluating SPATA12's protective effects in neurodegenerative disease models where oxidative stress is implicated

  • Exploring SPATA12's potential in reducing radiation-induced cellular damage

  • Investigating whether SPATA12 modulation could protect against age-related oxidative damage

Comprehensive Interactome Mapping:
Expanding our understanding of SPATA12's protein interaction network:

  • Employing advanced proteomics approaches to identify the complete set of SPATA12 interacting partners

  • Characterizing the dynamic changes in the SPATA12 interactome under various stress conditions

  • Developing a systems biology model of SPATA12's role in cellular stress responses

Detailed Structure-Function Analysis:
Understanding the structural basis of SPATA12 activity:

  • Determining the crystal structure of SPATA12 alone and in complex with key partners like CHD2

  • Identifying critical domains and residues required for its antioxidant and anti-apoptotic functions

  • Developing structure-based small molecule modulators of SPATA12 activity

In Vivo Models and Clinical Correlations:
Moving beyond cell culture to understand physiological relevance:

  • Developing and characterizing SPATA12 knockout and transgenic mouse models

  • Investigating SPATA12 expression patterns in human tissue samples from patients with disorders involving oxidative stress

  • Exploring correlations between SPATA12 genetic variants and susceptibility to oxidative stress-related diseases

Mechanistic Understanding of Selective Antioxidant Effects:
Elucidating how SPATA12 reduces ROS and MDA without affecting SOD and GSH:

  • Identifying direct ROS-scavenging capabilities through biochemical assays

  • Investigating potential regulation of non-canonical antioxidant pathways

  • Exploring possible mitochondrial effects that might reduce ROS generation

Data Science and Computational Biology Approaches:
Leveraging computational methods to advance SPATA12 research:

  • Applying machine learning to predict cellular responses to SPATA12 modulation under various conditions

  • Using network analysis to position SPATA12 within the broader cellular stress response network

  • Developing predictive models of SPATA12 activity based on cell type and stress conditions

These research directions would significantly advance our understanding of SPATA12 and potentially lead to novel therapeutic strategies for conditions involving oxidative stress and DNA damage.

What are the optimal parameters for analyzing SPATA12 expression in experimental settings?

Optimizing SPATA12 expression analysis requires attention to several critical parameters:

RNA-Based Expression Analysis:
Quantitative PCR (qPCR) has been effectively used for SPATA12 expression analysis with these considerations:

ParameterOptimal ConditionNotes
Reference genesGAPDH has been successfully usedConsider multiple reference genes for more accurate normalization
Primer designSpan exon-exon junctionsPrevents amplification of genomic DNA
RNA extraction timingCritical after stress inductionTime course analysis shows peak expression varies by cell type and stimulus
Statistical analysisOne-way ANOVA with Dunnett's testFor comparing multiple conditions to control

Protein-Based Expression Analysis:
Western blotting for SPATA12 protein detection requires:

  • Appropriate antibody selection and validation

  • Careful timing of protein extraction after stimulation

  • Proper normalization to loading controls

  • Consideration of subcellular fractionation to detect nuclear vs. cytoplasmic localization

Promoter Activity Analysis:
Dual luciferase reporter gene assays have proven effective with these parameters:

  • Use of pGL3-basic vector containing SPATA12 promoter fragments

  • Co-transfection with Renilla luciferase for normalization

  • Analysis of both full-length (pGL3-958/302) and core promoter (pGL3-136/302)

  • Inclusion of mutated AP-1 binding site constructs as controls

Experimental Design Considerations:
For optimal analysis of SPATA12 expression in stress response:

  • H₂O₂ concentration: 20-100 μM range has shown clear dose-dependent effects

  • UV-C exposure: Calibrated doses based on cell type sensitivity

  • Resveratrol treatment: 20 μM for 12h (after confirming non-toxicity via MTT assay)

  • Time points: Multiple time points post-treatment (e.g., 6h, 12h, 24h) to capture expression dynamics

Cell Type Considerations:
Expression patterns vary by cell type, with successful analysis performed in:

  • HeLa cells: Show strong SPATA12 induction with H₂O₂ and UV-C

  • MCF-7 cells: Exhibit similar response patterns to HeLa

  • H1299 cells: Useful for p53-independent studies

  • Primary testicular cells: Most physiologically relevant but technically challenging

These parameters provide a methodological framework for reliable and reproducible analysis of SPATA12 expression across various experimental conditions.

How should researchers approach contradictory data in SPATA12 studies?

When encountering contradictory data in SPATA12 research, a systematic approach is essential:

Standardized Protocol for Addressing Contradictions:

  • Verification through Multiple Methodologies:

    • Confirm findings using complementary techniques (e.g., qPCR, Western blot, immunofluorescence)

    • Employ different experimental systems (cell lines, primary cells)

    • Use both overexpression and knockdown/knockout approaches

  • Context-Dependent Analysis:

    • Carefully document all experimental conditions (cell type, passage number, treatment protocols)

    • Consider cell-specific factors that might influence results

    • Evaluate the influence of culture conditions and serum factors

  • Statistical Rigor:

    • Apply appropriate statistical tests (ANOVA with post hoc analysis)

    • Ensure adequate sample sizes for statistical power

    • Consider biological vs. technical replicates in experimental design

  • Comprehensive Literature Integration:

    • Systematically compare findings with published literature

    • Identify potential methodological differences that might explain discrepancies

    • Consider theoretical frameworks that might accommodate seemingly contradictory results

Specific Approaches for Common SPATA12 Contradictions:

Contradiction TypeInvestigation ApproachExample from SPATA12 Research
Expression level discrepanciesTime-course analysis with frequent sampling pointsResolving apparent contradictions in SPATA12 induction timing after stress
Functional outcome variationsCell type-specific analysis with pathway inhibitorsUnderstanding why SPATA12 shows different effects in different cell lines
Promoter activity inconsistenciesSystematic deletion/mutation analysis of promoter regionsIdentifying negative regulatory elements in the −946 to 90 bp region

Contradiction Analysis Framework:
Stanford's Contradiction Corpora project offers a useful framework that can be adapted for SPATA12 research:

  • Identify the type of contradiction (direct negation, numerical mismatch, presupposition violation)

  • Determine if the contradiction is apparent or genuine

  • Apply appropriate resolution strategies based on contradiction type

Documentation and Reporting:
When publishing research with contradictory findings:

  • Explicitly acknowledge the contradictions

  • Present multiple possible interpretations

  • Suggest experiments that could resolve the contradictions

  • Consider data sharing through repositories to enable meta-analysis

What technical challenges should researchers anticipate when working with recombinant SPATA12?

Researchers working with recombinant human SPATA12 should anticipate and prepare for several technical challenges:

Expression and Purification Challenges:

ChallengeTechnical SolutionMethodological Considerations
Low expression levelsCodon optimization for expression systemDifferent optimization strategies needed for bacterial vs. mammalian expression
Protein solubility issuesFusion tags (His, GST, MBP) to enhance solubilityTag selection may affect protein function and should be validated
Purification complexityTandem affinity purification approachesMulti-step purification may be necessary to achieve high purity
Protein stability concernsBuffer optimization with stabilizing agentsSystematic screening of buffer conditions to enhance stability

Functional Analysis Challenges:

  • Activity Assessment:

    • Developing reliable assays for SPATA12's antioxidant activity

    • Standardizing ROS measurement methodologies (DCFH-DA assay optimization)

    • Establishing quantitative metrics for DNA damage protection

  • Interaction Studies:

    • Surface plasmon resonance (SPR) or isothermal titration calorimetry (ITC) optimization for binding kinetics

    • Pull-down assay conditions for confirming protein-protein interactions

    • BiFC assay optimization for visualizing interactions in living cells

  • Cellular Localization:

    • Ensuring recombinant protein maintains proper cellular localization

    • Validating that fusion tags don't interfere with nuclear localization

    • Establishing proper controls for subcellular fractionation studies

Experimental Design Considerations:

  • Controls and Validations:

    • Inclusion of catalytically inactive mutants as negative controls

    • Verification that recombinant protein exhibits expected interaction with CHD2

    • Demonstration that exogenous expression recapitulates endogenous functions

  • Data Analysis Approaches:

    • Application of appropriate statistical methods (one-way ANOVA with Dunnett's test)

    • Use of SPSS software (version 17.0) with p<0.05 considered statistically significant

    • Standardized presentation of results as mean ± standard deviation

  • Adapting Data Table Analysis:

    • Implementation of "What-If Analysis" with Data Table for systematically testing multiple variables

    • Side-by-side comparisons of how changes in experimental parameters affect results

    • Consideration of both one-variable and two-variable data tables for complex analyses

Alternative Approaches to Consider:
If direct work with recombinant SPATA12 proves challenging, researchers might consider:

  • Using resveratrol (20 μM) as a non-toxic activator of endogenous SPATA12

  • Employing CRISPR/Cas9 for endogenous tagging rather than recombinant expression

  • Developing cell lines with inducible SPATA12 expression systems

By anticipating these challenges and implementing appropriate technical solutions, researchers can enhance the efficiency and reliability of their SPATA12 studies.

What are the key unresolved questions in SPATA12 research?

Despite significant advances in understanding SPATA12 function, several critical questions remain unresolved:

Mechanistic Questions:

  • What is the precise molecular mechanism by which SPATA12 reduces ROS and MDA levels without affecting SOD activity or GSH content?

  • How does the interaction between SPATA12 and CHD2 influence transcriptional regulation during DNA damage response?

  • What is the complete set of transcription factors beyond AP-1 that regulate SPATA12 expression under different cellular conditions?

  • How does SPATA12 influence p53 activity, and is this relationship direct or indirect through CHD2?

Structural and Functional Questions:

  • What is the three-dimensional structure of SPATA12, and which domains are critical for its antioxidant and anti-apoptotic functions?

  • Are there post-translational modifications that regulate SPATA12 activity under different stress conditions?

  • Does SPATA12 have enzymatic activity, or does it function primarily as a scaffold/adaptor protein?

  • What explains the apparent negative regulatory element located at the −946 to 90 bp region of the SPATA12 promoter?

Physiological and Pathological Relevance:

  • How does SPATA12 function differ between reproductive tissues and other cell types?

  • What is the physiological significance of SPATA12's dual role in spermatogenesis inhibition and cellular protection?

  • Does SPATA12 dysfunction contribute to pathological conditions characterized by oxidative stress or genomic instability?

  • Could SPATA12 serve as a biomarker for cellular stress responses or DNA damage in clinical settings?

Therapeutic Potential:

  • Can modulation of SPATA12 expression or activity be therapeutically beneficial in conditions characterized by oxidative stress?

  • How might resveratrol's effect on SPATA12 contribute to the compound's reported health benefits?

  • Are there small molecules beyond resveratrol that can selectively modulate SPATA12 activity?

  • Could targeting the SPATA12-CHD2 interaction represent a novel approach to modulating DNA damage responses?

Addressing these questions will require interdisciplinary approaches combining structural biology, biochemistry, molecular biology, and translational research, and would significantly advance our understanding of SPATA12's biological functions and potential clinical applications.

How might advances in SPATA12 research impact broader fields of biology?

Advances in SPATA12 research have the potential to impact multiple biological fields in significant ways:

Cancer Biology:
SPATA12's role as an inhibitor in tumorigenesis and its connection to DNA damage response pathways suggests substantial implications for:

  • Understanding novel tumor suppressor mechanisms beyond canonical pathways

  • Identifying potential therapeutic targets in the stress response network

  • Developing biomarkers for cellular responses to DNA-damaging cancer therapies

  • Exploring the intersection between oxidative stress and genomic instability in cancer progression

Reproductive Biology:
As a spermatogenesis-associated gene, SPATA12 research contributes to:

  • Advancing our understanding of molecular regulators in spermatogenesis

  • Providing insights into male infertility causes and potential treatments

  • Elucidating how cellular stress response mechanisms are integrated with reproductive functions

  • Understanding the role of oxidative stress in reproductive disorders

Stress Response Biology:
SPATA12's role in oxidative stress protection expands our understanding of:

  • Non-canonical antioxidant mechanisms that complement traditional systems

  • How cells integrate multiple stress response pathways

  • The relationship between transcriptional regulation and stress adaptation

  • Molecular determinants of cell fate decisions under stress conditions

Translational Medicine:
Applied aspects of SPATA12 research may impact:

  • Development of novel antioxidant therapies based on SPATA12's mechanisms

  • Strategies for protecting normal tissues during cancer treatments

  • Approaches to reducing oxidative damage in age-related disorders

  • Biomarker development for assessing cellular stress in clinical samples

Bioinformatics and Data Science:
The methodological approaches used in SPATA12 research offer models for:

  • Integrating multiple data types to resolve apparent contradictions

  • Applying systematic analytical frameworks to complex biological questions

  • Using "What-If Analysis" with Data Table for systematically testing multiple experimental variables

  • Developing predictive models of cellular responses to stress conditions

Systems Biology:
SPATA12 research exemplifies how:

  • Single proteins can have context-dependent functions within complex networks

  • Cellular responses are coordinated through interconnected regulatory mechanisms

  • Seemingly contradictory functions can serve unified biological purposes

  • Multiple levels of regulation (transcriptional, post-translational, protein-protein interactions) integrate to determine cellular outcomes

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 2025 TheBiotek. All Rights Reserved.