Recombinant Monachus schauinslandi Sex-determining region Y protein (SRY)

Shipped with Ice Packs
In Stock

Description

Definition and Biological Role

Recombinant Monachus schauinslandi Sex-Determining Region Y Protein (SRY) refers to the laboratory-synthesized version of the SRY protein derived from the Hawaiian monk seal (Neomonachus schauinslandi). SRY is a transcription factor encoded by the Y chromosome that initiates testis development by binding to DNA and activating downstream genes, such as SOX9, which drive male gonad differentiation . While recombinant SRY from humans and mice has been extensively studied, research on the Hawaiian monk seal’s SRY remains limited.

Genomic Context of SRY in Monachus schauinslandi

The Hawaiian monk seal’s genome was recently assembled at chromosomal resolution (NCBI ASM220157v2) . Key genomic features relevant to SRY include:

  • Chromosomal Location: SRY is located on the Y chromosome, though the exact coordinates in the monk seal remain unannotated.

  • Conservation: SRY’s High Mobility Group (HMG) box—a DNA-binding domain critical for function—is highly conserved across mammals . Comparative genomic analysis suggests the monk seal’s SRY HMG box shares structural homology with human and mouse orthologs.

  • Sequence Availability: While the monk seal’s SRY coding sequence has not been explicitly published, its genome assembly enables future cloning and expression studies .

Functional Insights from Related Species

Studies on human and murine SRY provide a framework for hypothesizing monk seal SRY activity:

FeatureHuman SRYMouse SryInferred for Monk Seal SRY
DNA BindingBinds AACAAAG motifs via HMG box Bends DNA to activate Sox9 Likely conserved HMG-mediated binding
TransactivationRequires partner proteins (e.g., SF1) Uses polyQ domain for Sox9 activation May depend on species-specific cofactors
Post-Translational ModificationsAcetylation regulates nuclear import Phosphorylation enhances DNA binding Potential lysine acetylation sites

Research Challenges and Opportunities

  • Cloning and Expression: The monk seal’s SRY gene could be amplified from genomic DNA (using primers targeting conserved HMG regions) and expressed in bacterial or mammalian systems .

  • Functional Assays: DNA-binding activity could be tested via electrophoretic mobility shift assays (EMSAs) using probes like AACAAAG . Transcriptional activation might be assessed using SOX9 reporter systems .

  • Structural Studies: Cryo-EM or X-ray crystallography could resolve the HMG box’s interaction with DNA, leveraging homology to human SRY .

Implications for Conservation Biology

The Hawaiian monk seal’s critically endangered status underscores the need to study its SRY:

  • Genetic Bottlenecks: Severe population declines (~23 founders post-bottleneck) may have reduced SRY allelic diversity, potentially affecting male fertility.

  • Disease Risks: Viral susceptibility in monk seal cell lines raises questions about SRY’s role in immune responses or developmental robustness.

Future Directions

  1. Gene Annotation: Prioritize SRY annotation in the monk seal genome to identify polymorphisms or deletions .

  2. Functional Characterization: Express recombinant SRY and compare its activity to human/mouse orthologs using in vitro assays .

  3. Population Genomics: Screen wild populations for SRY mutations linked to reproductive anomalies .

Product Specs

Form
Lyophilized powder
Note: While we prioritize shipping the available format, please specify your format preference in order notes for customized preparation.
Lead Time
Delivery times vary depending on the purchase method and location. Please consult your local distributor for precise delivery estimates.
Note: Standard shipping includes blue ice packs. Dry ice shipping requires advance notification 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 default glycerol concentration is 50% and can serve as a guideline.
Shelf Life
Shelf life depends on various factors including storage conditions, buffer composition, temperature, and protein stability. Generally, liquid formulations have a 6-month shelf life at -20°C/-80°C, while lyophilized forms have a 12-month shelf life at -20°C/-80°C.
Storage Condition
Upon receipt, store at -20°C/-80°C. Aliquot for multiple uses to prevent repeated freeze-thaw cycles.
Tag Info
Tag type is determined during manufacturing.
The tag type is determined during production. If a specific tag type is required, please inform us for preferential development.
Synonyms
SRY; TDF; Sex-determining region Y protein; Testis-determining factor
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
1-221
Protein Length
full length protein
Purity
>85% (SDS-PAGE)
Species
Neomonachus schauinslandi (Hawaiian monk seal) (Monachus schauinslandi)
Target Names
SRY
Target Protein Sequence
MFGVLNSSDH RAAVQQRNIP AFGRTSFELW TDNPTSNYRC ETGGNGRDSG QNRVRRPMNA FMVWSRDQRR RVALENPQMK NSEISKQLGY QWKMLTEAEK WPFFEEAQRL QAMHREKYPD YKYRPRPKAL PQKSDKLLPA ASSSMLCRQV LVDEKWYPFT YRDSCSRAAH PRMEDQLSSS QPVNIANSLL QQEHHYCSTS LRDSPETLAM HLSADPPFYP K
Uniprot No.

Target Background

Function
Recombinant Monachus schauinslandi Sex-determining region Y protein (SRY) is a transcriptional regulator controlling a genetic switch in male development. It is essential and sufficient for initiating male sex determination by directing the differentiation of supporting cell precursors (pre-Sertoli cells) into Sertoli cells rather than granulosa cells. SRY is involved in various aspects of gene regulation, including promoter activation and repression. It binds to the DNA consensus sequence 5'-[AT]AACAA[AT]-3'. The SRY HMG box recognizes DNA through partial intercalation in the minor groove, inducing DNA bending. Additionally, SRY is involved in pre-mRNA splicing. In the adult male brain, it contributes to maintaining the motor functions of dopaminergic neurons.
Protein Families
SRY family
Subcellular Location
Nucleus speckle. Cytoplasm. Nucleus.

Q&A

What is the SRY protein and what is its primary function?

The SRY protein is a transcription factor encoded by the sex-determining region Y gene found on the Y chromosome. It functions as the testis-determining factor (TDF) that initiates male sex determination in mammals. The protein contains a high mobility group (HMG)-box DNA-binding domain that allows it to bind and bend DNA, suggesting its role as a transcription factor that regulates the expression of genes involved in male development. The SRY protein causes a fetus to develop as a male through its regulatory actions on downstream genes in the sex determination pathway .

The SRY protein's primary function involves activation of male-specific developmental pathways while suppressing female developmental programs. It accomplishes this through direct DNA binding and recruitment of transcriptional machinery to target genes that initiate testis formation from the bipotential gonad. The timing of SRY expression is critically important, as reduced or delayed expression can impair testis development, highlighting the importance of its accurate spatiotemporal regulation .

How does the structure of SRY protein contribute to its function?

The SRY protein contains a central HMG-box domain that is responsible for its DNA-binding activity. This domain is present in a wide variety of proteins that bind and bend DNA, supporting its function as a transcription factor . The HMG-box allows the SRY protein to recognize specific DNA sequences and induce conformational changes in the DNA structure, facilitating the assembly of transcriptional complexes at target sites.

While the HMG-box sequences of SRY are reasonably conserved between species, sequences outside the HMG-box display a notable lack of sequence conservation . This structural organization suggests that the DNA-binding function is essential and conserved, while other regions may have evolved to accommodate species-specific regulatory mechanisms or protein-protein interactions. The sequence-specific DNA binding activity of SRY is critical for its function, as mutations that reduce this activity have been associated with XY females with gonadal dysgenesis .

How is SRY expression regulated during development?

SRY expression is tightly regulated through a complex network of transcription factors, epigenetic modifiers, and kinases. The expression pattern is precisely controlled in terms of timing and tissue specificity, with expression occurring in pre-Sertoli cells of the developing genital ridge during a critical window of development. Several key factors have been identified in the regulation of SRY expression:

  • Transcription factors: GATA4 is essential for SRY expression, as demonstrated by knockout studies showing that GATA4 is important for the earliest steps of male sex determination. GATA4 binds to specific regions of the SRY promoter and transactivates its expression. The interaction between GATA4 and its co-factor FOG2 (ZFPM2) is also required for SRY expression .

  • Nuclear receptors: NR5A1 (also known as SF-1) plays a central role in SRY regulation. NR5A1 binds to specific sites in the SRY promoter, and mutations in NR5A1 can result in XY sex reversal in humans .

  • Epigenetic regulation: Chromatin modifications and DNA methylation patterns influence SRY accessibility and expression. These epigenetic changes ensure that SRY is expressed at the right time and in the right cells.

  • Kinase signaling: MAP3K4 has been implicated in SRY regulation, as mice with mutations in this kinase show reduced SRY expression, partly due to decreased expression per cell .

SRY expression is initiated early in development, with studies in mice showing transcription as early as the two-cell stage, suggesting that mammalian sex determination starts prior to gonadal differentiation .

How does the SRY protein vary across different mammalian species?

The SRY protein exhibits significant variation across mammalian species, particularly in regions outside the conserved HMG-box domain. Comparative analysis of SRY proteins from 15 different species, including human, chimpanzee, dog, pig, rat, cattle, buffalo, goat, sheep, horse, zebra, frog, urial, dolphin, and killer whale, has revealed interesting patterns of conservation and divergence .

Sequence comparison between SRY protein sequences showed varying degrees of similarity between species. For example:

  • Orcinus orca (killer whale) and Tursiopsaduncus (dolphin) SRY proteins have the least genetic distance (0.33) among the 15 species studied, being 99.67% identical at the amino acid level.

  • Human and chimpanzee SRY proteins have a genetic distance of 1.35 and are 98.65% identical at the amino acid level.

  • The maximum genetic distance (89.98) was observed between Equuscabal (horse) and Musmusculus (mouse), with only 10.02% similarity at the amino acid level .

These variations suggest that SRY has evolved at different rates in different lineages. The evolutionary analysis of the SRY coding region among primates and rodents suggests rapid evolution, while data from wallaby and domestic ruminants indicate less rapid sequence evolution . These differences may reflect adaptations to species-specific mechanisms of sex determination or differences in the regulatory networks associated with SRY function.

What is known about the SRY protein in marine mammals, particularly Monachus schauinslandi?

While comprehensive information specifically about the Monachus schauinslandi (Hawaiian monk seal) SRY protein is limited in the provided search results, we can infer some characteristics based on what is known about SRY in other marine mammals and closely related species.

The search results mention that recombinant Monachus schauinslandi SRY protein is available as a full-length protein with >85% purity. This suggests that the protein has been successfully expressed and purified for research purposes, enabling various biochemical and functional studies.

Based on comparative data from other marine mammals like Orcinus orca (killer whale) and Tursiopsaduncus (dolphin), which show high sequence similarity (99.67%) in their SRY proteins , we might expect the Hawaiian monk seal SRY to share significant sequence identity with other marine mammals. This could potentially reflect common ancestral origins or similar selective pressures in the marine environment.

How can phylogenetic analysis of SRY inform our understanding of mammalian evolution?

Phylogenetic analysis of SRY proteins across various mammalian species provides a valuable tool for understanding evolutionary relationships and the divergence of sex determination mechanisms. The high degree of variation in SRY sequences outside the conserved HMG-box domain makes it particularly useful for inferring evolutionary distances between closely related species.

By constructing phylogenetic trees based on SRY sequence data, researchers can:

  • Trace the evolutionary history of sex determination mechanisms in mammals. The varying rates of SRY evolution in different lineages may reflect different selective pressures on sex determination systems.

  • Identify instances of convergent or divergent evolution in sex determination pathways. For example, the high similarity between killer whale and dolphin SRY proteins (99.67%) indicates strong conservation within marine mammals, potentially due to similar environmental pressures.

  • Correlate SRY sequence changes with speciation events. The 98.65% identity between human and chimpanzee SRY proteins aligns with their close evolutionary relationship.

  • Detect potential horizontally transferred segments or recombination events that might have influenced the evolution of sex determination systems.

  • Understand the molecular basis of sex determination disorders by comparing normal and pathogenic variations across species.

These phylogenetic analyses contribute to our broader understanding of mammalian evolution and can help resolve taxonomic relationships, particularly in cases where traditional morphological characteristics may be misleading or insufficient.

What are the preferred methods for expressing and purifying recombinant SRY protein?

For expressing and purifying recombinant SRY protein, including from species like Monachus schauinslandi, researchers typically employ several well-established methods:

  • Expression System Selection:

    • Bacterial expression systems (E. coli) are commonly used for their simplicity and high yield. The BL21(DE3) strain is preferred for protein expression due to its reduced protease activity.

    • For proteins requiring post-translational modifications, eukaryotic expression systems like yeast (S. cerevisiae, P. pastoris), insect cells (Sf9, Sf21 using baculovirus), or mammalian cells (HEK293, CHO) may be more appropriate.

  • Vector Design:

    • Vectors containing T7 or similar strong promoters are typically used.

    • The SRY coding sequence is often cloned with affinity tags (His-tag, GST, MBP) to facilitate purification and potentially enhance solubility.

    • For the intronless SRY gene, direct PCR amplification from genomic DNA can be used, followed by restriction enzyme digestion and ligation into expression vectors.

  • Expression Optimization:

    • Expression conditions (temperature, IPTG concentration, induction time) are optimized to maximize soluble protein yield.

    • Lower temperatures (16-20°C) during induction often improve solubility of recombinant proteins.

    • Co-expression with chaperones may enhance proper folding of the SRY protein.

  • Purification Strategy:

    • Affinity chromatography (Ni-NTA for His-tagged proteins, glutathione-agarose for GST-fusion proteins) as the initial purification step.

    • Ion-exchange chromatography to remove contaminants based on charge differences.

    • Size-exclusion chromatography for final polishing and buffer exchange.

    • The purity of >85% achieved for the recombinant Monachus schauinslandi SRY protein suggests effective purification protocols have been established.

  • Quality Control:

    • SDS-PAGE analysis to assess purity and integrity of the purified protein.

    • Western blotting to confirm identity using specific antibodies.

    • Mass spectrometry for accurate molecular weight determination and sequence confirmation.

    • Functional assays such as DNA-binding tests to confirm biological activity of the purified SRY protein.

These methodologies can be adapted based on specific research requirements and the unique properties of SRY from different species.

What techniques are used to study SRY-DNA interactions and binding specificity?

Several sophisticated techniques are employed to study SRY-DNA interactions and binding specificity:

  • Electrophoretic Mobility Shift Assay (EMSA):

    • This technique has been used to demonstrate that NR5A1 binds to specific sites in the pig and human SRY promoters .

    • EMSA allows visualization of protein-DNA complexes based on their reduced mobility in a non-denaturing gel compared to unbound DNA.

    • Competitive binding assays using unlabeled DNA can determine binding specificity and relative affinities.

  • Chromatin Immunoprecipitation (ChIP):

    • ChIP has confirmed that GATA4 binds to DNA in specific regions of the mouse SRY promoter .

    • This technique identifies protein-DNA interactions within living cells, providing in vivo relevance.

    • ChIP-seq combines ChIP with next-generation sequencing to identify genome-wide binding sites.

  • DNase I Footprinting:

    • This method identifies specific DNA sequences protected from DNase I digestion by bound proteins.

    • It can map the precise nucleotides contacted by SRY in the regulatory regions of target genes.

  • Systematic Evolution of Ligands by Exponential Enrichment (SELEX):

    • SELEX can determine the consensus binding sequence for SRY by iteratively enriching for high-affinity binding sites from random DNA sequences.

  • Surface Plasmon Resonance (SPR):

    • SPR provides real-time measurements of protein-DNA binding kinetics and affinity constants.

    • It allows determination of association and dissociation rates for SRY-DNA interactions.

  • Fluorescence Anisotropy:

    • This solution-based technique measures changes in rotational diffusion of fluorescently labeled DNA upon protein binding.

    • It can determine binding constants under equilibrium conditions.

  • X-ray Crystallography and Nuclear Magnetic Resonance (NMR):

    • These structural techniques provide atomic-level details of SRY-DNA complexes.

    • They reveal the specific contacts between amino acid residues and nucleotides that determine binding specificity.

  • Directed Mutagenesis:

    • Site-directed mutagenesis of DNA binding sites can verify their importance in SRY binding.

    • This approach has been used to study NR5A1 binding sites in SRY promoters, showing that mutations lead to reduced SRY expression in cell-based reporter assays .

These techniques, often used in combination, provide comprehensive characterization of the DNA binding properties of SRY from different species, including sequence specificity, binding affinity, and the structural basis of recognition.

How can researchers effectively study the role of SRY in sex determination across different species?

To effectively study the role of SRY in sex determination across different species, researchers can employ a multi-faceted approach combining comparative genomics, functional assays, and developmental biology techniques:

  • Comparative Sequence Analysis:

    • Sequence comparison of SRY across species can identify conserved and divergent regions, providing insights into functional domains. For example, analysis of 15 species has revealed high conservation of the HMG-box but significant variation in other regions .

    • Bioinformatic tools can predict functional elements based on sequence conservation patterns and identify potential regulatory motifs.

    • Calculating genetic distances between SRY proteins from different species (as done for killer whale, dolphin, human, chimpanzee, etc.) helps establish evolutionary relationships .

  • Transgenic and Knockout Models:

    • Creating transgenic animals where SRY from one species is expressed in another can test functional conservation.

    • CRISPR/Cas9-mediated gene editing can generate species-specific SRY knockouts or introduce specific mutations to assess functional consequences.

    • Conditional knockout systems allow time-specific inactivation of SRY, as demonstrated with GATA4 knockout at 10.5 dpc resulting in sex reversal .

  • Expression Analysis:

    • Quantitative RT-PCR can measure SRY expression levels during development, as used to show reduced expression in MAP3K4 mutant mice .

    • Immunofluorescence microscopy can visualize SRY protein localization in developing gonads.

    • Single-cell RNA sequencing provides cellular resolution of SRY expression and downstream effects.

    • In situ hybridization can map the spatiotemporal expression pattern of SRY during critical developmental windows.

  • Functional Genomics:

    • ChIP-seq identifies genome-wide binding sites of SRY in different species.

    • RNA-seq before and after SRY expression can elucidate downstream gene networks.

    • ATAC-seq reveals chromatin accessibility changes mediated by SRY.

    • CUT&RUN or CUT&Tag provide high-resolution mapping of SRY binding sites with lower cell numbers.

  • Cell-Based Reporter Assays:

    • Promoter constructs linked to reporter genes can assess SRY transcriptional activity across species. For example, GATA4 has been shown to transactivate SRY promoter constructs from mouse and pig, but not human .

    • Co-transfection experiments can identify species-specific co-factors required for SRY function.

  • Evolutionary Analysis:

    • Phylogenetic analysis of SRY sequences can correlate molecular evolution with phenotypic diversification of sex determination systems.

    • Tests for positive selection can identify amino acid positions under evolutionary pressure.

    • Comparative analysis of species with multiple SRY copies (e.g., rats with 11 non-identical copies) can provide insights into gene duplication and neo-functionalization .

  • Structural Biology:

    • Determining and comparing the three-dimensional structures of SRY proteins from different species can reveal mechanisms of functional conservation despite sequence divergence.

By integrating these approaches, researchers can develop a comprehensive understanding of how SRY function has evolved across different mammalian lineages while maintaining its central role in male sex determination.

How does SRY interact with other proteins in the sex determination pathway?

The SRY protein functions within a complex network of interacting proteins and transcription factors that collectively drive male sex determination. Understanding these interactions is crucial for elucidating the mechanistic basis of this developmental process:

  • Interaction with Transcriptional Co-factors:

    • SRY likely requires specific co-activators to effectively regulate target gene expression. While the search results don't directly identify these co-factors for SRY itself, the regulatory model suggests modularity in transcription factor networks.

    • The interaction between GATA4 and its co-factor FOG2 (ZFPM2) is required for SRY expression, suggesting similar co-factor requirements may exist for SRY function .

  • Regulatory Cascades and Feedback Loops:

    • SRY initiates a cascade of gene expression changes, working in concert with other sex-determining genes including SOX9, DMRT1, WNT1, AMH, SF1 (NR5A1), DAX1, GATA4, LIM1, Fra1, and aromatase .

    • The specific protein-protein interactions and regulatory relationships between SRY and these factors vary across species and can be studied using techniques such as co-immunoprecipitation, yeast two-hybrid assays, and proximity ligation assays.

  • Chromatin Remodeling Complexes:

    • SRY likely recruits chromatin-modifying enzymes and remodeling complexes to alter the epigenetic landscape at target loci.

    • The role of Polycomb group proteins in sex determination is suggested by the finding that the Polycomb group protein CBX2 regulates NR5A1 expression, which in turn affects SRY .

  • Integration with Signaling Pathways:

    • MAP3K4 and MAP3K1 signaling has been implicated in SRY regulation, with mutations in these kinases leading to human XY disorders of sexual development .

    • These kinase pathways likely modify SRY or its interacting partners through phosphorylation, affecting protein stability, nuclear localization, or DNA-binding activity.

  • Species-Specific Interaction Networks:

    • Despite the conservation of SRY's role in sex determination, the protein interaction networks may vary significantly between species.

    • The substantial sequence divergence outside the HMG-box domain suggests that species-specific protein interactions may have evolved to accommodate differences in developmental timing and regulatory mechanisms.

Research methodologies to study these interactions include affinity purification coupled with mass spectrometry (AP-MS), BioID or APEX proximity labeling, fluorescence resonance energy transfer (FRET), and computational network analysis. These approaches can reveal the dynamic interactome of SRY during the critical window of sex determination and identify key differences in this network across species.

What are the methodological challenges in studying SRY function in non-model organisms like Monachus schauinslandi?

Studying SRY function in non-model organisms such as the Hawaiian monk seal (Monachus schauinslandi) presents numerous methodological challenges that require innovative approaches:

  • Limited Genetic and Genomic Resources:

    • Lack of a well-annotated genome assembly for Monachus schauinslandi complicates identification of the complete SRY gene sequence and its regulatory elements.

    • Absence of established genetic modification protocols makes functional studies challenging.

    • Solution approaches include whole genome sequencing and assembly, targeted capture of the SRY locus, and comparative genomics with better-characterized marine mammals.

  • Sample Acquisition and Ethical Considerations:

    • Hawaiian monk seals are endangered, severely limiting access to biological samples.

    • Working with protected species requires extensive permits and ethical approvals.

    • Non-invasive sampling methods, such as collection of shed skin cells or biopsy of stranded animals, can provide DNA for sequencing studies while minimizing impact.

  • Developmental Biology Limitations:

    • The inaccessibility of embryos during the critical period of sex determination prevents direct observation of SRY action.

    • The long gestation period and low reproductive rate of marine mammals further complicates developmental studies.

    • Alternative approaches include studying cultured cells derived from adult tissues or utilizing induced pluripotent stem cells (iPSCs) to model early developmental events.

  • Experimental System Development:

    • Lack of established cell lines or primary cell culture protocols for Monachus schauinslandi.

    • Absence of species-specific antibodies and molecular tools.

    • Cross-species approaches can be employed, such as expressing Monachus schauinslandi SRY in cell lines from other mammals and testing its ability to activate known SRY target genes.

  • Functional Assessment Challenges:

    • Inability to perform knockout or transgenic studies in the organism itself.

    • Limited knowledge of species-specific downstream targets and regulatory networks.

    • Heterologous systems, such as expressing the seal SRY in mouse gonadal ridge explants or in sex-reversed mouse models, can provide insights into functional conservation.

  • Comparative Analytical Complexity:

    • Interpreting functional differences between seal SRY and better-studied mammals requires sophisticated evolutionary analysis.

    • Distinguishing functional adaptations from neutral evolutionary changes.

    • Phylogenetic approaches comparing SRY across multiple marine and terrestrial mammals can help identify seal-specific adaptations.

  • Technical Challenges with Recombinant Protein:

    • Optimizing expression and purification of correctly folded seal SRY protein.

    • Ensuring the recombinant protein retains native DNA-binding properties.

    • The available recombinant Monachus schauinslandi SRY with >85% purity represents a valuable resource, but characterizing its functional properties requires specialized assays.

These challenges necessitate creative experimental designs that leverage comparative approaches, heterologous systems, and cutting-edge genomic technologies to gain insights into SRY function in non-model organisms like the Hawaiian monk seal.

How can advanced genomic and proteomic approaches enhance our understanding of SRY evolution and function?

Advanced genomic and proteomic approaches offer powerful tools to deepen our understanding of SRY evolution and function across species:

  • Long-Read Sequencing Technologies:

    • Oxford Nanopore and PacBio sequencing can resolve complex genomic regions around SRY, including repetitive elements that are difficult to assemble with short-read technologies.

    • This is particularly valuable for species like rats that have multiple SRY copies (11 non-identical copies) , enabling complete characterization of all variants.

    • Long-read sequencing can reveal structural variations and rearrangements that affect SRY expression or function across different lineages.

  • Comparative Epigenomics:

    • ChIP-seq for histone modifications can map the epigenetic landscape around SRY and its target genes during sex determination.

    • ATAC-seq reveals changes in chromatin accessibility induced by SRY binding.

    • DNA methylation profiling using bisulfite sequencing or EPIC arrays can identify epigenetic regulatory mechanisms controlling SRY expression.

    • These approaches can reveal how epigenetic regulation of SRY has evolved across species.

  • Single-Cell Multi-omics:

    • Single-cell RNA-seq can track cell-specific transcriptional changes during sex determination, revealing how SRY expression affects different cell populations.

    • Single-cell ATAC-seq identifies cell-specific chromatin accessibility changes in response to SRY.

    • Multi-modal approaches combining transcriptomics, epigenomics, and proteomics at single-cell resolution can provide comprehensive views of SRY function.

    • These techniques can identify species-specific differences in cellular responses to SRY expression.

  • Structural Proteomics:

    • Cryo-electron microscopy can visualize SRY-containing complexes bound to DNA.

    • Hydrogen-deuterium exchange mass spectrometry (HDX-MS) can map protein-protein interaction surfaces.

    • Cross-linking mass spectrometry (XL-MS) can identify proximity relationships between SRY and interacting proteins.

    • These approaches can reveal how structural differences in SRY proteins from different species affect their function.

  • Systems Biology Approaches:

    • Network analysis integrating transcriptomic, proteomic, and epigenomic data can model the regulatory networks controlled by SRY.

    • Mathematical modeling of gene regulatory networks can predict species-specific differences in sex determination timing or robustness.

    • Genome-scale CRISPR screens can identify novel factors affecting SRY function.

  • Genome Editing Technologies:

    • CRISPR/Cas9-mediated precise editing can introduce species-specific SRY variants into model organisms.

    • Base editors or prime editors can create specific amino acid substitutions to test evolutionary hypotheses.

    • These approaches can directly test the functional significance of sequence differences between species.

  • Protein-DNA Interaction Mapping:

    • ChIP-seq and CUT&RUN can map genome-wide binding sites of SRY from different species expressed in the same cellular background.

    • DNA shape analysis algorithms can predict how sequence variations affect DNA structure and SRY binding.

    • These approaches can identify species-specific differences in target gene selection by SRY.

By integrating these advanced technologies, researchers can develop a comprehensive understanding of how SRY structure, function, and regulatory networks have evolved across mammalian lineages while maintaining the essential role in male sex determination.

What are common challenges in interpreting SRY functional data across species?

Interpreting functional data for SRY across different species presents several significant challenges:

To address these challenges, researchers should employ multiple complementary approaches, carefully design controls that account for species differences, and interpret data within the appropriate evolutionary and developmental context. Collaborative efforts that standardize methodologies across species can also help resolve apparent contradictions in the literature.

What quality control measures are essential when working with recombinant SRY proteins?

When working with recombinant SRY proteins, including those from Monachus schauinslandi, implementing rigorous quality control measures is essential to ensure reliable experimental results:

  • Protein Purity Assessment:

    • SDS-PAGE analysis to verify protein size and assess purity. The >85% purity reported for recombinant Monachus schauinslandi SRY protein should be independently verified.

    • Silver staining for detecting low-abundance contaminants that may not be visible with Coomassie staining.

    • Mass spectrometry to identify any co-purifying proteins that might affect functional assays.

    • Size-exclusion chromatography to assess aggregation state and homogeneity of the protein preparation.

  • Protein Identity Confirmation:

    • Western blotting with specific antibodies against SRY or epitope tags.

    • Mass spectrometry for peptide mass fingerprinting to confirm protein identity.

    • N-terminal sequencing to verify the correct start of the protein.

    • For tagged proteins, verification that the tag doesn't interfere with function through comparative assays with untagged versions.

  • Structural Integrity Verification:

    • Circular dichroism (CD) spectroscopy to assess secondary structure content and proper folding.

    • Thermal shift assays to determine protein stability.

    • Limited proteolysis to probe for correctly folded domains resistant to digestion.

    • Nuclear magnetic resonance (NMR) spectroscopy for detailed structural characterization if feasible.

  • Functional Activity Testing:

    • DNA binding assays (EMSA, fluorescence anisotropy) using known SRY target sequences.

    • Comparison of binding affinities with previously characterized SRY proteins from other species.

    • Testing for sequence-specific DNA bending, a characteristic property of SRY.

    • Cell-based reporter assays to verify transcriptional regulatory activity.

  • Batch-to-Batch Consistency:

    • Standardized production and purification protocols to ensure reproducibility.

    • Reference standards for comparison between batches.

    • Lot-specific activity measurements to account for potential variations.

    • Storage stability tests to determine optimal conditions and shelf-life.

  • Contaminant Testing:

    • Endotoxin testing, particularly important for proteins produced in bacterial systems.

    • Nuclease activity assays to ensure DNA-binding studies aren't affected by contaminating nucleases.

    • Protease activity assays to confirm absence of proteolytic contaminants.

    • Testing for co-purifying DNA that might affect DNA-binding assays.

  • Post-Translational Modification Analysis:

    • Phosphorylation analysis using mass spectrometry or phospho-specific antibodies.

    • Verification that any observed modifications match those expected in the native protein.

    • Assessment of how modifications affect protein function.

  • Storage and Handling Validation:

    • Freeze-thaw stability testing to determine the impact of multiple freeze-thaw cycles.

    • Buffer optimization to maintain protein stability during experiments.

    • Temperature sensitivity studies to establish appropriate handling conditions.

    • Long-term storage tests to determine shelf-life under various conditions.

Implementing these quality control measures ensures that functional studies with recombinant SRY proteins yield reliable and reproducible results, enabling valid comparisons across species and experimental conditions.

How can researchers distinguish between causative and correlative relationships when studying SRY in sex determination?

Distinguishing between causative and correlative relationships is a fundamental challenge in sex determination research. When studying SRY, researchers can apply several methodological approaches to establish causality:

  • Genetic Manipulation Approaches:

    • Loss-of-function studies: Targeted deletion or mutation of SRY in model organisms provides direct evidence of its necessity for male sex determination.

    • Gain-of-function studies: Introduction of functional SRY into XX embryos resulting in male development demonstrates sufficiency for initiating male sex determination.

    • Precise genome editing: CRISPR/Cas9-mediated introduction of specific mutations can establish causal relationships between sequence features and functional outcomes.

    • These approaches have established SRY as essential for triggering male development, with mutations causing XY disorders of sexual development (DSD) .

  • Temporal Manipulation:

    • Conditional expression/deletion systems: Using inducible promoters to control when SRY is expressed or deleted helps establish critical windows for its action.

    • Developmental time-course analyses: Careful staging of embryos combined with precise sampling can reveal the temporal sequence of molecular events following SRY expression.

    • This approach has shown that GATA4 knockout at 10.5 dpc, but not at later times, results in sex reversal, indicating its importance for the earliest steps of male sex determination .

  • Dose-Response Relationships:

    • Quantitative expression modulation: Varying SRY levels through controlled expression systems can establish dose-dependency.

    • Hypomorphic alleles: Mutations that reduce but don't eliminate SRY function can reveal threshold effects.

    • Studies have shown that reduced or delayed SRY expression impairs testis development, highlighting the importance of its accurate spatiotemporal regulation .

  • Molecular Mechanism Elucidation:

    • Direct binding demonstration: ChIP experiments confirm physical interaction of SRY with target DNA sequences in vivo.

    • Mutational analysis of binding sites: Directed mutagenesis of predicted binding sites can confirm their functional importance, as shown for NR5A1 sites in the SRY promoter .

    • Protein-protein interaction verification: Techniques like co-immunoprecipitation or proximity ligation can establish direct physical interactions between SRY and other proteins.

  • Pathway Validation:

    • Epistasis analysis: Determining whether the effects of manipulating one gene depend on the status of another gene helps establish their relative positions in a pathway.

    • Rescue experiments: Testing whether expression of downstream genes can rescue SRY deficiency helps confirm directional relationships.

    • Combined perturbations: Simultaneous manipulation of multiple genes can reveal redundancies or synergies in the pathway.

  • Natural Experiments:

    • Analysis of human DSD cases: Correlating specific SRY mutations with phenotypic outcomes in patients with disorders of sex development.

    • Comparative studies across species: Examining how natural variations in SRY sequence correlate with differences in sex determination mechanisms.

    • These approaches have identified that mutations in SRY can cause XY females with gonadal dysgenesis (Swyer syndrome), while translocation of part of the Y chromosome containing SRY to the X chromosome causes XX male syndrome .

  • Systems-Level Approaches:

    • Network inference: Computational analysis of large-scale gene expression data to infer causal relationships.

    • Mathematical modeling: Development of predictive models that can be experimentally validated.

    • Integration of multiple data types: Combining genomic, transcriptomic, and proteomic data to build causal models of sex determination.

By combining these approaches and cross-validating findings through multiple independent methods, researchers can establish robust causal relationships between SRY and downstream events in sex determination, moving beyond merely correlative observations.

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.