Recombinant Mouse Adipogenin (Adig)

Shipped with Ice Packs
In Stock

Product Specs

Form
Lyophilized powder
Note: We prioritize shipping the format currently in stock. However, if you have specific format requirements, please indicate them in your order notes. We will fulfill your request whenever possible.
Lead Time
Delivery time may vary depending on the purchasing method and location. For specific delivery times, please consult your local distributors.
Note: Our proteins are shipped with standard blue ice packs. If you require dry ice shipping, please inform us in advance. Additional fees will apply.
Notes
Repeated freezing and thawing is not recommended. Store working aliquots at 4°C for up to one week.
Reconstitution
We recommend briefly centrifuging the vial before opening to ensure the contents settle at the bottom. Please reconstitute the protein in deionized sterile 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 final glycerol concentration is 50%. Customers can use this as a reference.
Shelf Life
Shelf life is influenced by various factors, including storage conditions, buffer components, temperature, and the protein's inherent stability.
Generally, the shelf life of liquid form is 6 months at -20°C/-80°C. Lyophilized form has a shelf life of 12 months at -20°C/-80°C.
Storage Condition
Upon receipt, store at -20°C/-80°C. Aliquot for multiple uses. Avoid repeated freeze-thaw cycles.
Tag Info
The tag type will be determined during the manufacturing process.
The tag type is determined during production. If you have specific tag requirements, please inform us, and we will prioritize development according to your specifications.
Synonyms
Adig; Smaf1; Adipogenin; Small adipocyte factor 1; SMAF-1
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
1-80
Protein Length
full length protein
Species
Mus musculus (Mouse)
Target Names
Adig
Target Protein Sequence
MKYPLVPLVSDLTLSFLVFWLCLPVALLLFLTIVWLHFLLSQESKEDDSDLCFNWEPWSK RPSECGCEETFPGEEDGLHW
Uniprot No.

Target Background

Function
Adipogenin plays a significant role in stimulating adipocyte differentiation and development.
Gene References Into Functions
  1. Our research shows that Smaf1 is a novel small protein naturally found in adipocytes. Smaf1 expression is closely related to PPARgamma-mediated signals and the adipocyte phenotype. PMID: 26427354
  2. Adipogenin, an adipocyte-specific membrane protein, may be involved in adipogenesis as a regulator of adipose tissue development. PMID: 16132694
Database Links
Protein Families
Adipogenin family
Subcellular Location
Membrane; Single-pass membrane protein. Nucleus.
Tissue Specificity
Selectively expressed in adipose tissue where it is particularly enriched in brown adipose tissue. In adipose tissue, expressed exclusively in adipocytes and not in the stromal-vascular cell population. Expressed at much lower levels in heart, stomach and

Q&A

What is Adipogenin and where is it predominantly expressed?

Adipogenin (Adig) is an evolutionarily conserved microprotein that plays a crucial role in adipocyte development and function. It is predominantly expressed in adipose tissues and testis. The protein functions as a critical regulator for lipid droplet formation in adipocytes through direct interaction with seipin, forming a structurally rigid complex that facilitates lipid accumulation. This interaction is fundamental to understanding adipocyte biology and the mechanisms governing fat tissue development. When designing experiments involving Adig, researchers should consider its tissue-specific expression patterns to establish appropriate controls and experimental conditions .

How does Adipogenin interact with seipin at the molecular level?

Adipogenin interacts directly with seipin, specifically binding to dodecameric seipin complexes rather than undecameric forms. The interaction occurs through a specialized mechanism where the N-terminus of Adig embeds within seipin oligomers, positioning near the beginning segments of TM2 (transmembrane segment 2) from two adjacent seipin proteins. Cryo-EM structural analysis at 2.98Å resolution reveals that Adig bridges adjacent seipin monomers, with the TM segments from Adig forming critical contacts with seipin. This bridging function stabilizes the complex by suppressing thermal fluctuations and maintaining structural integrity. For researchers studying this interaction, it's essential to preserve the native conformation of both proteins during experimentation, as alterations to either protein's structure may disrupt this highly specific interaction .

What methods are recommended for expressing recombinant mouse Adipogenin in cellular systems?

For optimal expression of recombinant mouse Adipogenin in cellular systems, researchers should consider using mammalian expression systems that maintain proper protein folding and post-translational modifications. Doxycycline-inducible expression systems have proven effective, as demonstrated in transgenic mouse models. When designing expression constructs, attention should be paid to preserving the N-terminal region of Adig, which is critical for its interaction with seipin oligomers. For cellular studies, A431 cells containing seipin-degron-GFP have been successfully employed to study Adig function. This system allows for rapid and effective depletion of endogenous seipin, enabling researchers to observe the assembly of new complexes and evaluate Adig's contribution to this process. Visualization techniques using fluorescence microscopy can confirm successful expression and localization within adipocytes .

What is the functional relationship between Adipogenin and lipid droplet formation?

Adipogenin plays a key role in generating lipid droplets in adipocytes through its interaction with seipin complexes. Mechanistically, Adig improves the structural stability of dodecameric seipin complexes, which serve as essential scaffolds for lipid droplet formation at the endoplasmic reticulum. This enhanced stability is evidenced by reduced root mean square fluctuation (RMSF) in molecular dynamics simulations and improved conformational symmetry of the inner lumen of the seipin complex. Rather than directly increasing affinity for triacylglycerols (TAGs), Adig appears to maintain the structural integrity of the seipin complex during TAG sequestration. In experimental settings, visualization of lipid droplets can be achieved through standard techniques such as Oil Red O staining or BODIPY labeling, with quantitative analysis of droplet size and number providing valuable metrics for assessing Adig function .

How can researchers design effective gain-of-function studies for Adipogenin in mouse models?

To design effective gain-of-function studies for Adipogenin in mouse models, researchers should implement a Doxycycline-inducible system that enables adipocyte-specific Adig overexpression in adult animals. This approach allows for temporal control, separating the effects of Adig on mature adipocyte function from potential developmental effects. The transgenic model should be established with careful consideration of the promoter to ensure adipocyte specificity, with the adiponectin promoter being particularly effective. Following induction, a comprehensive analysis protocol should include: 1) Verification of Adig overexpression by western blotting in different adipose depots; 2) Assessment of body weight changes over time; 3) Analysis of adipose tissue mass in brown (BAT), subcutaneous white (sWAT), and epididymal white (eWAT) depots; 4) Histological examination using H&E staining to evaluate changes in lipid droplet morphology; 5) Quantification of triglyceride and cholesterol content in adipose tissues; and 6) Lipidomic analysis to determine specific changes in triglyceride profiles. For functional assessment, researchers should consider conducting triglyceride clearance tests and cold exposure experiments to evaluate the metabolic consequences of Adig overexpression .

What methodological approaches are most effective for studying the Adipogenin-seipin structural relationship?

For studying the Adipogenin-seipin structural relationship, a multi-modal approach incorporating cryo-electron microscopy, computational modeling, and molecular dynamics simulations has proven most effective. Cryo-EM at high resolution (2.98Å) can reveal the detailed architecture of the seipin/Adig complex, including the surprising finding that seipin can form two distinct oligomers (undecamers and dodecamers) with Adig selectively binding to the dodecameric form. This should be complemented with computational approaches such as AlphaFold 3 predictions, which can provide insights into how subunit interactions change with oligomer size. To assess the dynamic properties of these complexes, atomistic molecular dynamics simulations are invaluable, enabling measurement of parameters such as root mean square fluctuation (RMSF) and lumen diameter consistency over time. For analyzing TAG sequestration by the complex, coarse-grained molecular dynamics simulations with model ER bilayers enriched with TAG (approximately 1.25 mol%) can be employed. Researchers should also consider cellular imaging techniques using fluorescently tagged proteins to visualize complex formation in living cells, with recovery experiments after initial depletion particularly informative for studying assembly dynamics .

What are the recommended protocols for generating and validating adipocyte-specific Adipogenin knockout mice?

For generating adipocyte-specific Adipogenin knockout mice, the preferred approach is to establish an inducible system using Cre-lox technology. First, create Adig flox mice (Adigf/f) with loxP sites flanking critical exons of the Adig gene. Then, cross these mice with adiponectin-rtTA/TRE-Cre mice to generate inducible adipocyte-specific Adig knockout mice (Adig iAKO). This system allows temporal control of gene deletion, enabling researchers to induce Adig knockout in mature adipocytes (typically at 5-6 weeks of age) while avoiding developmental confounders. Validation of the knockout should include multiple steps: 1) Genomic PCR to confirm successful recombination in adipose tissues but not in other tissues; 2) RT-PCR and western blotting to verify the absence of Adig mRNA and protein specifically in adipose tissues; 3) Histological analysis of adipose tissues to assess morphological changes; 4) Lipidomic analysis to quantify changes in triglyceride species; and 5) Functional tests such as cold tolerance assessment. Control groups should include Adigf/f mice without Cre expression subjected to the same Doxycycline treatment to account for any effects of the antibiotic itself .

What analytical techniques should be employed to assess the effects of Adipogenin modulation on triglyceride metabolism?

To comprehensively assess the effects of Adipogenin modulation on triglyceride metabolism, researchers should employ a multi-tiered analytical approach. First, quantitative measurement of total triglyceride content in adipose tissues should be performed using colorimetric or enzymatic assays. This should be complemented with detailed lipidomic analysis using liquid chromatography-mass spectrometry (LC-MS) to identify changes in specific triglyceride species. The lipidomic data should be analyzed to determine if Adig modulation affects all triglyceride species uniformly or preferentially impacts certain molecular species. For in vivo assessment of triglyceride dynamics, triglyceride clearance tests should be conducted by administering a defined lipid load and measuring serum triglyceride levels at multiple time points post-administration. To examine the cellular uptake mechanisms, researchers can employ radioactively or fluorescently labeled fatty acids in combination with tissue-specific uptake inhibitors. For thermogenesis studies, particularly in brown adipose tissue, cold exposure protocols with core body temperature monitoring provide valuable functional data. Additionally, gene expression analysis of key lipid metabolism genes using qRT-PCR or RNA-seq can offer insights into the molecular mechanisms underlying the observed metabolic changes .

How can researchers differentiate between the direct effects of Adipogenin on lipid droplet formation versus secondary metabolic adaptations?

To differentiate between direct effects of Adipogenin on lipid droplet formation and secondary metabolic adaptations, researchers should implement a comprehensive experimental design with appropriate temporal controls. In cellular systems, acute manipulation of Adig expression followed by immediate assessment of lipid droplet dynamics (within 24-48 hours) can help isolate direct effects before secondary adaptations occur. The use of A431 cells containing seipin-degron-GFP allows rapid depletion of endogenous seipin and subsequent monitoring of complex reassembly in the presence or absence of Adig. For in vivo studies, inducible systems with short induction periods (2-3 days) can help distinguish primary from secondary effects. Tissue-specific analyses should examine not only adipose tissues but also liver, muscle, and circulation to identify systemic changes that may represent secondary adaptations. Molecular dynamics simulations that examine the immediate structural consequences of Adig binding to seipin provide valuable mechanistic insights into direct effects. Additionally, in vitro reconstitution experiments using purified components can establish the minimal requirements for Adig-mediated effects on lipid droplet formation. Finally, transcriptomic and proteomic analyses at multiple time points following Adig modulation can help establish the temporal sequence of molecular events, distinguishing immediate targets from downstream adaptive responses .

What are the optimal conditions for purifying functional recombinant mouse Adipogenin?

For optimal purification of functional recombinant mouse Adipogenin, researchers should consider its properties as a microprotein that interacts with membrane-associated complexes. Expression systems should preserve the native conformation, with mammalian cell-based expression systems generally preferred over bacterial systems. When designing expression constructs, include affinity tags (such as His6 or FLAG) positioned to avoid interference with the N-terminal region critical for seipin interaction. Lysis conditions should employ mild detergents (such as CHAPS or DDM) to solubilize the protein while preserving its native structure. Purification protocols typically involve affinity chromatography followed by size exclusion chromatography to isolate properly folded protein. Throughout the purification process, maintain physiological pH (7.2-7.4) and include glycerol (10-15%) to stabilize the protein. For functional validation, in vitro binding assays with purified seipin can confirm the retention of binding capacity. Additionally, circular dichroism spectroscopy can verify proper secondary structure formation. Researchers should also consider the option of co-purifying Adig with seipin to maintain the native complex, particularly for structural studies. Finally, storage conditions should include cryoprotectants and aliquoting to minimize freeze-thaw cycles, with functionality tests performed after storage to confirm protein stability .

What imaging techniques provide the most informative data on Adipogenin's role in lipid droplet dynamics?

For investigating Adipogenin's role in lipid droplet dynamics, a multi-modal imaging approach yields the most comprehensive insights. Confocal microscopy with fluorescently labeled Adig and seipin provides spatial resolution to visualize their co-localization at nascent lipid droplet formation sites. Live-cell imaging using GFP-tagged Adig in combination with lipid dyes (such as BODIPY or Nile Red) enables real-time monitoring of lipid droplet nucleation, growth, and remodeling. For higher resolution analysis, super-resolution microscopy techniques like Structured Illumination Microscopy (SIM) or Stimulated Emission Depletion (STED) microscopy can resolve subcellular details of the Adig-seipin complex. Cryo-electron microscopy is essential for structural studies at the molecular level, while electron tomography can bridge the gap between molecular and cellular scales by providing 3D reconstructions of lipid droplet formation sites. For functional analysis in tissue samples, histological techniques combined with digital image analysis allow quantification of lipid droplet size distribution, number, and morphology. In mice with altered Adig expression, whole-body imaging techniques such as EchoMRI can track changes in fat mass non-invasively over time. Each imaging modality should be selected based on the specific research question, with appropriate controls and quantification methods to ensure reproducible and statistically significant results .

What experimental controls are essential when investigating Adipogenin's interaction with the seipin complex?

When investigating Adipogenin's interaction with the seipin complex, several essential controls must be implemented to ensure experimental validity. First, protein specificity controls should include experiments with structurally similar but functionally distinct proteins to confirm the specificity of the Adig-seipin interaction. Second, domain-specific mutants of both Adig and seipin should be tested to map critical interaction regions, with particular attention to the N-terminus of Adig that embeds in seipin oligomers. Third, concentration-dependent binding assays should determine whether the interaction follows expected biochemical kinetics. Fourth, competition assays with unlabeled proteins can confirm binding specificity and relative affinities. Fifth, negative controls using cell lines that lack endogenous expression of either protein help establish baseline signals. Sixth, positive controls with known interaction partners of seipin validate the experimental system. Seventh, subcellular localization controls should confirm that observed interactions occur in physiologically relevant cellular compartments. Eighth, for structural studies, proper folding controls using circular dichroism or limited proteolysis ensure the proteins are in their native conformations. Ninth, for in vivo studies, appropriate genetic background controls minimize strain-specific effects. Finally, when using inducible systems, both non-induced and empty vector controls should be included to account for leaky expression or vector-related effects .

How can researchers accurately quantify changes in lipid droplet morphology resulting from Adipogenin manipulation?

To accurately quantify changes in lipid droplet morphology resulting from Adipogenin manipulation, researchers should implement a standardized multi-parameter analysis workflow. Begin with high-resolution imaging using confocal or differential interference contrast microscopy of fixed cells stained with lipid-specific dyes such as BODIPY, Oil Red O, or LipidTOX. For tissue sections, use H&E staining complemented with specific lipid stains. Acquire multiple images across different fields to ensure representative sampling. Image analysis should be performed using specialized software (such as ImageJ with appropriate plugins, CellProfiler, or commercial solutions) with automated detection and measurement protocols to minimize bias. Quantitative parameters should include: 1) Lipid droplet diameter/area distribution; 2) Number of lipid droplets per cell; 3) Total lipid droplet volume per cell; 4) Circularity index to detect morphological abnormalities; and 5) Spatial distribution patterns within the cell. For brown adipose tissue, which typically contains multilocular lipid droplets, additional metrics should assess the degree of multilocularity versus unilocularity. Statistical analysis should employ appropriate tests for distribution comparisons, with data visualized as frequency distributions rather than simple averages. For validation, orthogonal techniques such as flow cytometry of isolated adipocytes using lipid-specific fluorescent dyes can provide population-level data. Finally, longitudinal live-cell imaging can capture dynamic changes in lipid droplet formation, fusion, and growth that may be missed in fixed samples .

How might Adipogenin research contribute to understanding metabolic disorders?

Research on Adipogenin has significant potential to advance our understanding of metabolic disorders through several key mechanisms. The observed role of Adig in promoting adipose tissue expansion and lipid droplet formation directly impacts lipid storage capacity, a critical factor in metabolic health. Dysfunctional adipose tissue expansion is associated with lipid spillover, ectopic fat deposition, and subsequent insulin resistance. By enhancing our understanding of how Adig regulates triglyceride sequestration via seipin complex stabilization, researchers can identify novel therapeutic targets for conditions like obesity, type 2 diabetes, and non-alcoholic fatty liver disease. The finding that Adig overexpression increases triglyceride uptake and elevates thermogenesis during cold exposure suggests potential applications in brown adipose tissue activation for metabolic benefit. Furthermore, the aberrant lipid droplet formation observed in Adig knockout mice parallels pathologies seen in lipodystrophies, indicating Adig's relevance to these rare but metabolically severe conditions. For translational researchers, investigating Adig expression and function in human adipose tissue samples from patients with various metabolic conditions could reveal whether alterations in Adig contribute to pathogenesis or represent adaptive responses. Additionally, the Adig-seipin interaction represents a model system for studying how microproteins can regulate complex cellular processes, potentially informing broader principles of metabolic regulation .

What approaches can be used to translate findings from mouse Adipogenin studies to human applications?

To translate findings from mouse Adipogenin studies to human applications, researchers should implement a multi-faceted approach that bridges species differences while leveraging conserved biology. First, comparative genomic and proteomic analyses should establish the degree of conservation between mouse and human Adig, with particular attention to functional domains such as the N-terminus that interacts with seipin. Second, structural studies should determine whether human Adig interacts with seipin in a manner similar to the mouse protein, ideally using cryo-EM or other high-resolution techniques. Third, ex vivo studies using primary human adipocytes or adipose tissue explants can validate whether manipulation of Adig levels produces effects comparable to those observed in mouse models. Fourth, development of human cell line models (such as differentiated human preadipocytes or iPSC-derived adipocytes) with controlled Adig expression enables mechanistic studies in a human cellular context. Fifth, analysis of human adipose tissue samples from various metabolic states (lean, obese, insulin-resistant, diabetic) can establish correlations between Adig expression/function and metabolic parameters. Sixth, genetic association studies examining whether Adig variants correlate with metabolic phenotypes in human populations can provide population-level insights. Finally, development of humanized mouse models expressing human Adig can serve as a bridge between species, allowing in vivo testing of human-specific interventions. Throughout this translational process, researchers should remain attentive to species-specific differences in adipose tissue biology, such as the relative importance and distribution of brown/beige adipose tissue .

How can structural insights about the Adipogenin-seipin complex inform drug discovery efforts?

The high-resolution structural insights about the Adipogenin-seipin complex offer valuable opportunities for rational drug discovery efforts targeting metabolic disorders. The cryo-EM structure at 2.98Å resolution provides detailed information about interaction interfaces, particularly how Adig selectively binds to dodecameric seipin complexes and bridges adjacent seipin subunits. This selectivity presents an opportunity to design compounds that could either enhance or disrupt this specific interaction. Computational approaches can leverage this structural data to perform virtual screening of compound libraries against the Adig binding pocket on seipin or the seipin interaction surface on Adig. Structure-based drug design efforts should focus on: 1) Compounds that mimic Adig's ability to stabilize seipin dodecamers, potentially beneficial in conditions with impaired lipid storage; 2) Molecules that modulate the rigidity of the complex, which appears crucial for proper lipid droplet formation; and 3) Allosteric modulators that affect the conformational dynamics of the seipin complex. For assay development, researchers can establish high-throughput screening systems using fluorescence resonance energy transfer (FRET) between tagged Adig and seipin components to monitor complex assembly or stability. Molecular dynamics simulations of the complex with potential drug candidates can predict binding affinities and effects on complex stability prior to experimental validation. Additionally, the finding that Adig affects thermogenesis during cold exposure suggests that modulators of this pathway could have applications in metabolic disorders characterized by reduced energy expenditure .

What are the most promising techniques for studying the temporal dynamics of Adipogenin activity during adipocyte development?

For studying the temporal dynamics of Adipogenin activity during adipocyte development, researchers should implement a comprehensive approach combining time-resolved imaging, molecular profiling, and functional assays. Live-cell imaging using fluorescently tagged Adig in adipocyte precursors undergoing differentiation provides real-time visualization of protein localization and dynamics. This approach can be enhanced with photoactivatable or photoconvertible fluorescent tags to track specific protein populations over time. Complementary to imaging, inducible expression systems enable precise temporal control of Adig levels at defined stages of differentiation. For molecular profiling, time-series RNA-seq and proteomics during differentiation can map the temporal relationship between Adig expression and downstream molecular changes. Single-cell technologies are particularly valuable, as they can capture heterogeneity in differentiation trajectories and correlate this with Adig activity. Chromatin immunoprecipitation sequencing (ChIP-seq) at multiple time points can identify dynamic changes in the regulatory landscape associated with Adig expression. For functional assessment, metabolic flux analysis using isotope tracers provides insights into how lipid metabolism changes temporally in response to Adig activity. Finally, computational modeling based on time-series data can generate testable hypotheses about the kinetics of Adig-mediated processes. When designing these experiments, researchers should include appropriate time-matched controls and collect data at sufficient temporal resolution to capture both rapid and gradual changes in Adig activity and its downstream effects .

What are common pitfalls in recombinant Adipogenin expression systems and how can they be addressed?

Common pitfalls in recombinant Adipogenin expression systems include protein misfolding, aggregation, and low yield, which can be addressed through systematic optimization. For bacterial expression systems, the hydrophobic nature of Adig's transmembrane domain often leads to inclusion body formation. This can be mitigated by: 1) Using specialized E. coli strains designed for membrane protein expression; 2) Lowering induction temperature to 16-18°C; 3) Adding solubility-enhancing fusion tags such as SUMO or thioredoxin; or 4) Including mild detergents in lysis buffers. For mammalian expression systems, which generally provide better folding of Adig, common issues include variable expression levels and cytotoxicity. These can be addressed by: 1) Using inducible promoters to control expression timing and level; 2) Screening multiple cell lines for optimal expression; and 3) Co-expressing chaperone proteins that assist with folding. For both systems, codon optimization based on the expression host can improve translation efficiency. When purifying Adig, protein loss often occurs during buffer exchanges or concentration steps. This can be minimized by including glycerol (10-15%) and appropriate detergents throughout the purification process. For functional validation, activity assays should be developed based on Adig's known interaction with seipin, with proper controls to distinguish specific from non-specific binding. Finally, storage stability is crucial; researchers should determine optimal conditions through stability testing and consider flash-freezing aliquots in liquid nitrogen for long-term storage .

How can researchers address variability in phenotypes when studying Adipogenin in mouse models?

To address variability in phenotypes when studying Adipogenin in mouse models, researchers should implement rigorous experimental design and controls. First, genetic background significantly influences adipose tissue biology, so using genetically homogeneous strains and littermate controls is essential. For transgenic models, backcrossing for at least 6-10 generations ensures genetic homogeneity. Second, sex differences substantially impact adipose tissue function; studies should either include both sexes with sufficient numbers for separate analysis or provide clear justification for studying only one sex. Third, age standardization is critical, as adipose tissue undergoes significant changes during development and aging. Fourth, environmental factors including housing conditions, diet composition, and feeding regimens should be strictly controlled, with detailed reporting of these parameters. Fifth, for inducible models, doxycycline administration protocols should be standardized, accounting for potential off-target effects of the antibiotic itself. Sixth, phenotyping protocols should be comprehensive and standardized, including body composition analysis, glucose homeostasis assessment, and adipose tissue-specific measurements. Seventh, variability can be reduced by increasing sample sizes, determined through proper power calculations based on preliminary data. Eighth, when assessing cold tolerance or other stress responses, standardized acclimation periods are essential. Ninth, for molecular analyses, sample collection timing should account for circadian variations in adipose tissue metabolism. Finally, advanced statistical approaches such as multivariate analysis can help identify patterns in variable datasets and determine which factors contribute most significantly to phenotypic differences .

What quality control measures should be implemented when working with recombinant Adipogenin preparations?

Comprehensive quality control measures for recombinant Adipogenin preparations should verify both molecular integrity and functional activity. For molecular characterization, SDS-PAGE with Coomassie staining or western blotting should confirm the expected molecular weight and purity, with acceptance criteria typically requiring >90% purity. Mass spectrometry analysis provides precise molecular weight verification and can identify post-translational modifications or truncations. Circular dichroism spectroscopy should assess secondary structure content, ensuring proper protein folding. For functional validation, binding assays with purified seipin components should confirm interaction with expected affinity constants. Size exclusion chromatography or analytical ultracentrifugation can verify that Adig remains monomeric rather than forming non-physiological aggregates. Thermal shift assays evaluate protein stability and can be used to optimize buffer conditions. For lot-to-lot consistency, researchers should establish reference standards and acceptance criteria for key parameters. When Adig is intended for cellular studies, sterility testing and endotoxin measurement are essential, with endotoxin levels kept below 0.1 EU/mg protein. For activity testing, cellular assays measuring lipid droplet formation in Adig-null cells reconstituted with the recombinant protein provide functional validation. Finally, stability studies should determine appropriate storage conditions and shelf-life, with activity testing at multiple time points. Detailed documentation of all quality control results should be maintained, establishing specifications for critical quality attributes based on the intended experimental applications .

How can researchers standardize lipid droplet analysis protocols across different experimental systems?

Standardizing lipid droplet analysis protocols across different experimental systems requires establishing consistent methodologies for sample preparation, imaging, quantification, and data analysis. For sample preparation in cellular systems, standardize fixation procedures (typically 4% paraformaldehyde for 15 minutes at room temperature) and lipid staining protocols (concentration, incubation time, and washing steps). For tissue samples, standardize sectioning thickness (typically 5-10 μm), staining procedures, and antigen retrieval methods if immunostaining is performed. Imaging parameters should be rigorously controlled, including microscope settings (objective magnification, numerical aperture, exposure time, gain), z-stack parameters, and acquisition of multiple fields per sample to ensure representative sampling. For quantitative analysis, implement automated image analysis workflows using open-source software such as ImageJ/Fiji with specialized plugins or CellProfiler, with detailed documentation of all parameters and thresholds. Standardized metrics should include: 1) Lipid droplet size distribution (diameter or area); 2) Number of lipid droplets per cell; 3) Total lipid content per cell; and 4) Morphological features such as circularity. To facilitate cross-laboratory comparison, include reference standards such as cell lines with known lipid droplet characteristics in each experimental batch. For data reporting, provide detailed methodology sections in publications, including all parameters and thresholds used in image analysis. Consider depositing raw image data in public repositories with appropriate metadata. Finally, participate in inter-laboratory standardization efforts and proficiency testing to ensure consistency across research groups .

How can transcriptomic approaches be utilized to understand Adipogenin's regulatory network?

Transcriptomic approaches offer powerful tools for elucidating Adipogenin's regulatory network across multiple experimental contexts. RNA sequencing (RNA-seq) of adipose tissues or adipocytes with modulated Adig expression (overexpression, knockout, or knockdown) provides comprehensive insights into gene expression changes. This approach should include multiple time points following Adig manipulation to distinguish between primary and secondary transcriptional responses. For greater cellular resolution, single-cell RNA-seq can identify cell type-specific responses to Adig modulation within heterogeneous adipose tissue, potentially revealing differential responses in mature adipocytes versus preadipocytes or immune cells. To identify direct transcriptional effects, researchers can combine RNA-seq with chromatin immunoprecipitation sequencing (ChIP-seq) for transcription factors affected by Adig, or with ATAC-seq to map changes in chromatin accessibility. For mechanistic insights, RNA-seq can be performed under various physiological challenges such as cold exposure, high-fat diet, or fasting/refeeding to determine how Adig influences adaptive transcriptional responses. Network analysis tools such as weighted gene co-expression network analysis (WGCNA) can identify modules of co-regulated genes associated with Adig function. Validation of key findings should include qRT-PCR and protein-level analysis, focusing on pathways relevant to lipid metabolism, adipocyte differentiation, and thermogenesis. Finally, comparative transcriptomic analysis between mouse models and human adipose samples can identify conserved regulatory networks with translational relevance .

What specialized techniques are available for studying the biophysical properties of the Adipogenin-seipin complex?

Studying the biophysical properties of the Adipogenin-seipin complex requires specialized techniques that can capture structural details and dynamic behaviors at multiple scales. Cryo-electron microscopy has proven invaluable, achieving resolutions of 2.98Å for the seipin/Adig complex and revealing unexpected features such as the existence of both undecameric and dodecameric seipin complexes. For dynamic properties, hydrogen-deuterium exchange mass spectrometry (HDX-MS) can map regions of differential flexibility or solvent accessibility upon Adig binding. Surface plasmon resonance or isothermal titration calorimetry provides quantitative binding kinetics and thermodynamic parameters. Fluorescence resonance energy transfer (FRET) with strategically placed fluorophores can monitor conformational changes in the complex in response to various conditions or ligands. Single-molecule techniques such as atomic force microscopy or optical tweezers can measure mechanical properties and forces involved in complex assembly or lipid interactions. For membrane-associated properties, supported lipid bilayers with reconstituted complexes allow controlled manipulation of lipid composition while monitoring complex behavior. Small-angle X-ray scattering (SAXS) provides information about complex shape in solution under near-physiological conditions. Molecular dynamics simulations at atomistic resolution can predict dynamic behaviors and have already revealed that Adig reduces thermal fluctuations in the seipin complex and maintains circular symmetry of the inner lumen. For studying lipid interactions, native mass spectrometry can identify specific lipid species that associate with the complex. These complementary approaches provide a multi-scale understanding of how Adig influences seipin complex structure and function .

How can CRISPR-Cas9 genome editing be optimized for studying Adipogenin function?

Optimizing CRISPR-Cas9 genome editing for studying Adipogenin function requires careful consideration of targeting strategy, delivery methods, and validation approaches. For guide RNA (gRNA) design, researchers should target conserved functional domains of Adig, particularly the N-terminal region critical for seipin interaction. Multiple gRNAs should be designed and validated to identify those with highest editing efficiency and specificity. For cellular models, adipocyte precursor cell lines such as 3T3-L1 or primary preadipocytes can be edited, followed by differentiation to assess effects on mature adipocyte function. Delivery methods should be optimized for each cell type, with lipofection typically effective for cell lines while electroporation may be preferable for primary cells. For in vivo editing, adeno-associated viral (AAV) vectors with adipose-specific promoters provide tissue selectivity. When creating knockout models, researchers should design strategies that ensure complete loss of function, typically by targeting early exons to create frameshift mutations. For more sophisticated modifications, homology-directed repair templates can introduce specific mutations or tags for tracking endogenous Adig. Validation of editing should be comprehensive, including genomic DNA sequencing, mRNA expression analysis, and protein-level verification. Off-target analysis is critical, with whole-genome sequencing recommended for cell lines intended for extensive study. For phenotypic characterization, edited cells should undergo comprehensive assessment of lipid droplet formation, adipocyte differentiation, and metabolic function. Finally, complementation experiments reintroducing wild-type or mutant Adig can confirm specificity of observed phenotypes and establish structure-function relationships .

What are the most effective methods for studying Adipogenin's role in thermogenesis?

To effectively study Adipogenin's role in thermogenesis, researchers should implement a multi-level approach spanning molecular, cellular, and physiological analyses. In vivo cold exposure experiments provide direct functional assessment, with core body temperature monitoring during acute (4-6 hours) and chronic (5-7 days) cold challenge at 4-8°C. For maximum sensitivity, implantable temperature probes or thermal imaging cameras offer continuous data collection. At the tissue level, histological analysis of brown adipose tissue before and after cold exposure can reveal changes in lipid droplet morphology and distribution. Infrared thermography of interscapular regions can non-invasively assess BAT activation in rodents. Metabolic cage studies measuring oxygen consumption, carbon dioxide production, and heat production provide whole-body energy expenditure data, particularly when combined with β3-adrenergic agonist (CL-316,243) stimulation to specifically activate BAT. At the cellular level, seahorse extracellular flux analysis of isolated brown adipocytes can measure changes in oxygen consumption rate in response to norepinephrine or other activators. For molecular characterization, quantitative PCR and western blotting should assess thermogenic gene expression (UCP1, PGC1α, PRDM16) and protein levels. Lipidomic analysis of BAT before and after cold exposure can reveal how Adig affects mobilization of specific lipid species for thermogenesis. Importantly, triglyceride clearance tests during cold exposure can directly assess how Adig influences the uptake of circulating lipids for thermogenic fuel. Finally, calcium imaging in brown adipocytes can monitor adrenergic signaling, a key pathway in thermogenic activation .

How should researchers design experiments to investigate potential sex differences in Adipogenin function?

To rigorously investigate sex differences in Adipogenin function, researchers should implement a comprehensive experimental design that accounts for hormonal influences, developmental timing, and depot-specific effects. First, experimental groups must include both males and females with sufficient sample sizes (typically n≥8-10 per sex) to detect sex-specific effects, with power calculations based on preliminary data or literature. Second, age-matching is critical, with consideration of developmental stages particularly during puberty, reproductive maturity, and aging. Third, estrous cycle staging in females should be performed using vaginal cytology, as adipose tissue biology varies across cycle phases. Fourth, hormone manipulation studies (gonadectomy with or without hormone replacement) can establish causality between sex hormones and observed differences in Adig function. Fifth, analysis should encompass multiple adipose depots (subcutaneous, visceral, brown) as sex differences often manifest in a depot-specific manner. Sixth, molecular analyses should include assessment of Adig expression, localization, and protein-protein interactions across sexes and depots. Seventh, functional studies should evaluate lipid droplet formation, thermogenic capacity, and metabolic parameters in both sexes under basal and challenged conditions (such as cold exposure or high-fat diet). Eighth, for mechanistic insights, chromatin immunoprecipitation can identify sex-specific transcription factor binding at the Adig promoter. Ninth, in vitro studies using adipocytes derived from males and females can distinguish cell-autonomous from systemic effects. Finally, translational relevance should be assessed by analyzing human adipose samples from both sexes for correlations between Adig expression/function and metabolic parameters .

What experimental approach is recommended for investigating Adipogenin's potential role beyond adipose tissue?

To investigate Adipogenin's potential roles beyond adipose tissue, researchers should implement a systematic approach that begins with comprehensive expression profiling and extends to functional studies in non-adipose contexts. First, quantitative PCR and western blotting should survey Adig expression across multiple tissues under various physiological conditions (fasting, cold exposure, inflammatory challenge) to identify tissues with significant expression or regulation. Second, single-cell RNA sequencing of tissues showing Adig expression can identify specific cell types and contexts associated with Adig. Third, immunohistochemistry with validated antibodies can confirm protein localization and cellular distribution. Fourth, tissue-specific conditional knockout or overexpression models should be developed for tissues with meaningful Adig expression, prioritizing those with lipid metabolism functions (liver, muscle, intestine). Fifth, for tissues like testis, where Adig is known to be expressed, detailed phenotyping should assess morphology, lipid content, and reproductive function in models with altered Adig expression. Sixth, for potential non-cell-autonomous effects, parabiosis experiments between wild-type and Adig-modified mice can determine whether Adig influences distant tissues through circulating factors. Seventh, for tissues that rely on lipid metabolism (brain, heart), functional assessments should include lipid uptake, oxidation, and storage. Eighth, for potential developmental roles, inducible models allowing temporal control of Adig deletion or overexpression are essential. Ninth, unbiased interactome studies using proximity labeling or co-immunoprecipitation followed by mass spectrometry can identify tissue-specific Adig interaction partners. Finally, comparative studies across species can provide evolutionary insights into conserved non-adipose functions .

How can researchers design experiments to distinguish between Adipogenin's roles in white versus brown adipose tissues?

To distinguish between Adipogenin's roles in white versus brown adipose tissues, researchers should implement a comparative experimental design with tissue-specific interventions and functional readouts. First, depot-specific gene expression analysis should quantify baseline Adig expression across classical brown adipose tissue (interscapular BAT), subcutaneous white adipose tissue (inguinal WAT), and visceral white adipose tissue (epididymal or perigonadal WAT), including assessment of browning/beiging markers in white depots. Second, conditional knockout or overexpression models with depot-specific targeting can be achieved using promoters selective for brown adipocytes (UCP1-Cre) versus white adipocytes (Adiponectin-Cre with BAT deletion using UCP1-Cre prior to Adiponectin-Cre activation). Third, for cellular studies, isolated brown and white adipocytes should be cultured under identical conditions to compare intrinsic responses to Adig modulation. Fourth, browning/beiging potential can be assessed by exposing Adig-modified mice to cold or β3-adrenergic agonists, with subsequent analysis of UCP1 induction and multilocular lipid droplet formation in white depots. Fifth, functional differences can be examined through metabolic cage studies measuring depot-specific contributions to whole-body energy expenditure. Sixth, lipidomic analysis comparing lipid species profiles between brown and white depots with altered Adig expression can reveal depot-specific lipid handling. Seventh, transcriptomic and proteomic analyses can identify depot-specific molecular pathways regulated by Adig. Eighth, for thermogenic assessment, infrared thermography can localize heat production to specific depots. Ninth, transplantation of Adig-modified brown or white adipose tissue into recipient mice can determine depot-specific systemic effects. Finally, for translational relevance, human samples from both brown and white depots should be analyzed for correlations between Adig expression and depot-specific functions .

What approach should be used to investigate potential interactions between Adipogenin and other known regulators of lipid metabolism?

To investigate potential interactions between Adipogenin and other known regulators of lipid metabolism, researchers should implement a systematic approach combining molecular, cellular, and in vivo methods. First, computational analysis using protein-protein interaction databases and structural modeling can identify potential interaction partners based on sequence or structural complementarity. Second, unbiased proteomic approaches such as proximity labeling (BioID or APEX) or co-immunoprecipitation followed by mass spectrometry can identify the Adig interactome in adipocytes. Third, direct protein-protein interactions can be confirmed using techniques such as co-immunoprecipitation, pull-down assays, or fluorescence resonance energy transfer (FRET). Fourth, for genetic interactions, combinatorial gene manipulation (double knockouts, combined overexpression and knockout) can reveal synergistic or antagonistic relationships. Fifth, epistasis experiments, where the effects of manipulating one factor are tested in the presence or absence of another, can establish hierarchical relationships between Adig and other regulators. Sixth, transcriptomic analysis in models with altered Adig expression can identify convergent or divergent pathways with other lipid metabolism regulators. Seventh, lipid flux studies using isotope tracers can determine whether Adig modulates specific metabolic pathways regulated by factors such as PPAR𝛾, SREBP, or LXR. Eighth, for pharmaceutical relevance, testing how Adig modulation affects responses to established metabolic drugs (such as thiazolidinediones or fibrates) can uncover clinically relevant interactions. Ninth, for pathway analysis, phosphoproteomic studies can identify signaling cascades connecting Adig to known regulatory networks. Finally, temporal analysis of protein complex formation during adipocyte differentiation or in response to metabolic challenges can reveal dynamic interactions under physiologically relevant conditions .

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.