Recombinant Trachemys scripta Doublesex- and mab-3-related transcription factor 1 (DMRT1)

Shipped with Ice Packs
In Stock

Description

Overview of Recombinant Trachemys scripta Doublesex- and mab-3-related transcription factor 1 (DMRT1)

Recombinant Trachemys scripta DMRT1 is a synthetically produced protein derived from the Dmrt1 gene of the red-eared slider turtle (Trachemys scripta), a species with temperature-dependent sex determination (TSD). This protein plays a pivotal role in male sex determination by initiating testicular differentiation and regulating downstream pathways. Its recombinant form enables functional studies to dissect molecular mechanisms of TSD and evolutionary conservation of sex determination pathways .

Gene and Protein Structure

  • Gene sequence: The T. scripta Dmrt1 cDNA spans 2,448 bp, encoding a 369-amino-acid protein with a conserved DM DNA-binding domain .

  • Conservation: Shares 80.7% amino acid identity with chicken DMRT1 and 75.3% with mice, highlighting evolutionary conservation .

  • Expression specificity: Testis-specific mRNA and protein localization in Sertoli cells, absent in ovaries or somatic tissues .

Recombinant Production Methods

Recombinant DMRT1 is generated using lentiviral vectors for in vivo and in vitro applications:

  • Knockdown: Lentivirus-mediated RNA interference (shRNA) reduces endogenous Dmrt1 by 73.7% in MPT (male-producing temperature) embryos .

  • Overexpression: Electroporation of cultured FPT (female-producing temperature) gonads with Dmrt1 ORF induces male-specific gene activation .

Functional Role in Sex Determination

ExperimentOutcomeCitation
Dmrt1 knockdown in MPT embryosFeminized gonads; ectopic aromatase expression, germ cell cortical migration
Dmrt1 overexpression in FPT gonadsTesticular cord formation; upregulation of Sox9 and Amh
Temperature-shift assaysDmrt1 expression responds within 2 days, preceding Sox9 and Amh changes

Thermosensitivity and Epigenetic Regulation

  • Temperature-dependent expression: Dmrt1 mRNA is detectable at MPT (male-producing temperature) as early as developmental stage 15, but absent at FPT (female-producing temperature) .

  • DNA methylation: Promoter methylation dynamics correlate with temperature shifts, suggesting epigenetic regulation of Dmrt1 .

Molecular Mechanisms in TSD

Recombinant DMRT1 studies reveal its role as a master regulator:

  1. Upstream trigger: Acts before gonadal differentiation (stage 14–17), initiating medullary Sertoli cell differentiation .

  2. Pathway activation: Directly upregulates Sox9 and Amh while suppressing Cyp19a1 (aromatase) and Foxl2 .

  3. Cross-species conservation: Functional parallels exist with Dmrt1 in chicken, medaka, and mammals, despite mechanistic differences in sex determination systems .

Research Applications

  • TSD mechanism elucidation: Study temperature-epigenetic-gene interactions using recombinant protein .

  • Conservation biology: Model for understanding evolutionary transitions between TSD and genotypic sex determination (GSD) .

Technical Challenges and Innovations

  • Lentiviral delivery: Achieved 55% transfection efficiency in turtle embryos, enabling robust in vivo gene modulation .

  • In vitro gonad culture: Maintains gonadal viability for 30 days, facilitating long-term functional studies .

Comparative Insights Across Species

SpeciesRole of DMRT1Sex Determination System
Trachemys scriptaMaster male-determining gene; TSDTemperature-dependent
ChickenNecessary for testis maintenance; GSDGenotypic (ZZ/ZW)
HumanCritical for postnatal testis developmentGenotypic (XX/XY)
MedakaDmy (DMRT1 paralog) drives male fateGenotypic (XX/XY)

Table 2: Evolutionary roles of DMRT1 across vertebrates .

Product Specs

Form
Lyophilized powder. We will ship the in-stock format preferentially. For specific format requirements, please note them during order placement.
Lead Time
Delivery times vary based on purchasing method and location. Consult local distributors for specific delivery times. All proteins are shipped with standard blue ice packs. For dry ice shipping, contact us in advance; additional fees apply.
Notes
Avoid repeated freeze-thaw cycles. Working aliquots are stable at 4°C for up to one week.
Reconstitution
Briefly centrifuge the vial before opening. Reconstitute protein in sterile deionized water to 0.1-1.0 mg/mL. Add 5-50% glycerol (final concentration) and aliquot for long-term storage at -20°C/-80°C. Our default final glycerol concentration is 50%.
Shelf Life
Shelf life depends on storage conditions, buffer components, storage temperature, and protein stability. Liquid form is generally stable for 6 months at -20°C/-80°C. Lyophilized form is generally stable for 12 months at -20°C/-80°C.
Storage Condition
Store at -20°C/-80°C upon receipt. Aliquot for multiple uses. Avoid repeated freeze-thaw cycles.
Tag Info
The tag type will be determined during production. If you require a specific tag, please inform us and we will prioritize its development.
Synonyms
DMRT1Doublesex- and mab-3-related transcription factor 1; Fragment
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
1-205
Protein Length
full length protein
Purity
>85% (SDS-PAGE)
Species
Trachemys scripta (Red-eared slider turtle) (Pseudemys scripta)
Target Names
DMRT1
Target Protein Sequence
GHKRFCMWRD CQCKKCSLIA ERQRVMAAQV ALRRQQAQEE ELGISHPIPL PSAPKLFVKK ENNGGSSCLL LESSSPTHST NTATTASTTP SEGRMLIQDI PSITSRGHLE STSDLVVDST YYSSFYQPSL YPYYNNLYNY SQYQMAVASD SSSSDMGGTL AGSPVKNSLR SLPATYMSSQ SGNQWQMKST ESRHAMSSQY RMHSY
Uniprot No.

Target Background

Function
May be involved in testis development.
Protein Families
DMRT family
Subcellular Location
Nucleus.

Q&A

What is the molecular structure and function of DMRT1 in Trachemys scripta?

DMRT1 is a dose-sensitive transcription factor containing a conserved zinc finger-like DNA-binding motif known as the DM domain. This domain represents an ancient, conserved component of the vertebrate sex-determining pathway that regulates male development across diverse taxonomic groups from nematodes to vertebrates . Unlike the SOX9 transcription factor, which induces DNA bending when bound, DMRT1 attaches to the DNA minor groove without causing significant conformational changes .

In T. scripta, DMRT1 functions as a primary regulator of male sex determination and testicular development. The protein is predominantly expressed in the testicular cord and Sertoli cells, with lower levels detected in germ cells . Functional analyses demonstrate that DMRT1 is both necessary and sufficient to initiate male development in this species, positioning it as an upstream regulator of the male developmental pathway .

How does temperature influence DMRT1 expression in TSD species?

DMRT1 expression in T. scripta exhibits a clear temperature-dependent pattern that correlates with sexual outcomes. At male-producing temperatures, DMRT1 shows significantly higher expression compared to female-producing temperatures . This sexually dimorphic expression precedes morphological gonadal differentiation, suggesting its role as an early molecular switch in the sex determination cascade.

The responsiveness of DMRT1 to temperature is remarkably dynamic. Experimental evidence shows that DMRT1 can respond rapidly to temperature shifts during the thermosensitive period of embryonic development . When embryos initially incubated at female-promoting temperatures are shifted to male-promoting temperatures, DMRT1 expression increases accordingly, and vice versa, demonstrating the plasticity of this regulatory system .

The mechanism linking temperature to DMRT1 expression appears to involve epigenetic regulation. DNA methylation dynamics of the DMRT1 promoter region show strong correlation with incubation temperature and could serve as the molecular bridge mediating temperature's influence on gene expression . This epigenetic regulation may represent a conserved mechanism for environmental sex determination across reptilian lineages.

What methodologies are recommended for isolating and characterizing T. scripta DMRT1?

For initial characterization of DMRT1 in T. scripta samples, RT-PCR and quantitative RT-PCR (qRT-PCR) using specific primers targeting conserved regions of the DMRT1 gene are effective approaches. Based on published data, the following primer sequences have been successfully employed for DMRT1 amplification in chelonian species:

PrimerSequence (5'–3')
DMRT1-forwardAAR AAG TGC AGC CTG ATC GC
DMRT1-reverseCAT ATA TGT GGC TGG GAG GC
qmDMRT1-forward[species-specific sequence required]
qmDMRT1-reverse[species-specific sequence required]

These primers amplify a fragment containing portions of the DM domain, which can be subsequently sequenced for confirmation . For protein detection, Western blot analysis using antibodies recognizing conserved epitopes of DMRT1 can verify expression levels across different tissues and developmental stages .

Immunohistochemistry and immunofluorescence techniques are valuable for localizing DMRT1 protein within gonadal tissues. Studies in related species have demonstrated that DMRT1 protein is predominantly localized in the nucleus of Sertoli cells surrounding spermatogonia in testes .

How can recombinant T. scripta DMRT1 be efficiently produced for functional studies?

Production of recombinant DMRT1 protein requires careful consideration of expression systems to ensure proper folding and functionality of this transcription factor. While the search results don't provide specific protocols for T. scripta DMRT1 production, successful approaches for related transcription factors typically involve:

  • Cloning the full-length coding sequence or functional domains (particularly the DM domain) into appropriate expression vectors

  • Selecting expression systems that facilitate proper folding of zinc finger-containing proteins (mammalian or insect cell systems often yield better results than bacterial systems)

  • Including affinity tags (His, GST, or FLAG) positioned to minimize interference with DNA binding capacity

  • Optimizing purification protocols to maintain protein stability and activity

For functional characterization, DNA-binding assays such as electrophoretic mobility shift assays (EMSAs) or chromatin immunoprecipitation (ChIP) can determine the binding specificity and genomic targets of recombinant DMRT1.

What approaches are effective for manipulating DMRT1 expression in T. scripta embryos?

Both loss-of-function and gain-of-function approaches have proven effective for investigating DMRT1 function in turtle embryos. Based on successful studies in T. scripta and related species:

RNA interference (RNAi):
Lentiviral vector-mediated RNA interference has been successfully applied to knockdown DMRT1 in reptilian embryos . This approach involves:

  • Designing short hairpin RNAs (shRNAs) targeting conserved regions of DMRT1 mRNA

  • Packaging the constructs into lentiviral vectors for efficient delivery

  • Microinjecting viral particles into developing embryos at specific developmental stages

Studies in P. sinensis demonstrated that DMRT1 knockdown in ZZ embryos resulted in male-to-female sex reversal, characterized by gonadal feminization, downregulation of testicular markers (Amh and Sox9), and upregulation of ovarian regulators (Cyp19a1 and Foxl2) . Similar approaches are applicable to T. scripta research.

Overexpression studies:
Ectopic expression of DMRT1 in genetic females can be achieved through lentiviral vectors carrying the DMRT1 coding sequence under control of a strong promoter. In P. sinensis, this approach led to masculinization of genetic females, including induction of male-specific markers and downregulation of female pathway genes . These methodologies can be adapted for T. scripta with appropriate sequence modifications.

How does DMRT1 function in the context of temperature-dependent sex determination differ from its role in genetic sex determination?

DMRT1 exhibits remarkable functional conservation across diverse sex-determining systems while showing specific adaptations to different mechanisms:

In TSD species (T. scripta):

  • DMRT1 expression is directly responsive to temperature cues

  • DNA methylation at the DMRT1 promoter correlates with temperature and may mediate environmental effects on gene expression

  • DMRT1 functions as a master male sex determinant that integrates environmental signals

In GSD species (P. sinensis with ZZ/ZW system):

  • DMRT1 shows male-specific embryonic expression

  • It is located on sex chromosomes in some GSD species

  • Its expression is typically less environmentally plastic but still crucial for male development

These comparative patterns suggest that DMRT1 represents an evolutionarily conserved component of vertebrate sex determination that has been repeatedly recruited during transitions between environmental and genetic sex-determining mechanisms. The temperature sensitivity of DMRT1 expression in TSD species likely evolved through modifications to its regulatory regions, particularly with respect to epigenetic mechanisms like DNA methylation.

What critical factors should be considered when designing temperature-shift experiments to study DMRT1 regulation?

Temperature-shift experiments provide valuable insights into the plasticity and regulation of DMRT1 expression during the thermosensitive period of development. Key considerations include:

Timing of temperature shifts:

  • Identify the precise thermosensitive period for your study species (typically mid-embryonic development in T. scripta)

  • Design shift experiments to cover different windows within this period to determine critical windows for DMRT1 regulation

Temperature parameters:

  • Use temperatures known to produce near 100% males (e.g., 26°C in T. scripta) and near 100% females (e.g., 31°C in T. scripta)

  • Monitor actual temperatures in incubators continuously to account for fluctuations

Sampling strategy:

  • Collect samples at multiple timepoints following temperature shifts (e.g., 6h, 24h, 72h post-shift)

  • Include appropriate controls (embryos maintained at constant temperatures)

  • Process samples for both mRNA expression analysis and protein detection

Analytical approaches:

  • Quantify DMRT1 expression using qRT-PCR with appropriate reference genes

  • Assess downstream targets (Amh, Sox9) and antagonistic pathway genes (Cyp19a1, Foxl2)

  • Evaluate epigenetic modifications at the DMRT1 promoter through bisulfite sequencing

What are effective strategies for analyzing contradictory data in DMRT1 expression studies across different turtle species?

Studies of DMRT1 expression across turtle species have revealed both similarities and differences. When encountering contradictory results, consider these analytical approaches:

  • Developmental timing standardization: Ensure precise staging of embryos across studies. Expression differences may reflect slight variations in developmental timing rather than true species differences.

  • Methodological harmonization: Evaluate whether discrepancies arise from methodological differences:

    • Sample preparation (whole gonad vs. isolated cell populations)

    • Detection method sensitivity (traditional RT-PCR vs. qRT-PCR)

    • Normalization approaches (different reference genes)

  • Phylogenetic context: Interpret differences in light of evolutionary relationships:

SpeciesSex Determination MechanismDMRT1 Expression PatternReference
T. scriptaTSDGradual upregulation at male-producing temperatures over stages 15-23; consistently low at female-producing temperatures
L. olivaceaTSDConsistently higher at male-producing temperature; gradual downregulation over development at female-producing temperatures
C. pictaTSDDifferent pattern from T. scripta (specific details not provided)
P. sinensisGSD (ZZ/ZW)Early male-specific embryonic expression
  • Tissue specificity analysis: Determine if expression differences reflect distinct spatial patterns within the developing gonad.

  • Functional conservation testing: Despite expression differences, functional assays (knockdown/overexpression) may reveal conserved roles in sex determination.

What controls should be included when conducting DMRT1 knockdown and overexpression studies?

Robust experimental design for DMRT1 functional studies requires comprehensive controls:

For knockdown studies:

  • Scrambled shRNA controls (non-targeting sequences with similar properties)

  • Multiple shRNA constructs targeting different regions of DMRT1 to confirm specificity

  • Rescue experiments (co-delivery of shRNA-resistant DMRT1 constructs)

  • Quantitative assessment of knockdown efficiency at both mRNA and protein levels

  • Monitoring of known DMRT1 target genes to confirm functional consequences

For overexpression studies:

  • Empty vector controls

  • Overexpression of mutated DMRT1 lacking functional domains

  • Dose-response experiments with varying levels of DMRT1 expression

  • Temporal controls (expression at different developmental stages)

For both approaches:

  • Appropriate controls for delivery method toxicity

  • Uninjected embryos as baseline controls

  • Monitoring of developmental progression to ensure normal development outside the gonad

  • Assessment of both early (gene expression) and late (gonadal histology) phenotypes

How can issues with recombinant DMRT1 protein solubility and stability be addressed?

Transcription factors containing zinc finger domains like DMRT1 often present challenges in recombinant expression. Common issues include protein insolubility, misfolding, and aggregation. These challenges can be addressed through:

  • Expression strategy optimization:

    • Use fusion tags that enhance solubility (MBP, SUMO, or TRX)

    • Express individual domains separately (particularly the DM domain)

    • Employ eukaryotic expression systems (insect cells, mammalian cells) instead of bacterial systems

  • Buffer optimization:

    • Include zinc in purification buffers to maintain DM domain structure

    • Test various pH conditions (typically pH 7.5-8.5 works best for DNA-binding proteins)

    • Add stabilizing agents (10% glycerol, low concentrations of reducing agents)

  • Protein refolding protocols:

    • Gradual dialysis from denaturing to native conditions

    • On-column refolding during purification

    • Chaperone co-expression to facilitate proper folding

  • Storage condition optimization:

    • Determine optimal storage buffer composition

    • Test stability at different temperatures (-80°C, -20°C, 4°C)

    • Evaluate freeze-thaw stability and consider single-use aliquots

What approaches can resolve challenges in detecting endogenous DMRT1 in early embryonic samples?

Early embryonic gonadal tissue presents challenges for protein detection due to limited material and potentially low expression levels. Researchers can overcome these limitations through:

  • Sample pooling strategies:

    • Pool multiple embryonic gonads from the same developmental stage and temperature treatment

    • Carefully document the number of samples pooled for accurate normalization

  • Signal amplification techniques:

    • Employ tyramide signal amplification (TSA) for immunohistochemistry

    • Use highly sensitive chemiluminescent substrates for Western blotting

    • Consider proximity ligation assays for detecting protein interactions with high sensitivity

  • Enrichment approaches:

    • Implement laser capture microdissection to isolate specific cell populations

    • Use nuclear extraction protocols to concentrate nuclear transcription factors

    • Apply immunoprecipitation before Western blotting to concentrate the target protein

  • Alternative detection methods:

    • Utilize droplet digital PCR for absolute quantification of low-abundance transcripts

    • Implement single-cell transcriptomics when protein detection is challenging

    • Consider reporter constructs driven by the DMRT1 promoter to monitor activity indirectly

What emerging technologies could advance our understanding of DMRT1 function in temperature-dependent sex determination?

Several cutting-edge approaches show promise for elucidating DMRT1 function in TSD systems:

  • CRISPR/Cas9 genome editing:

    • Generate targeted mutations in the DMRT1 coding sequence or regulatory regions

    • Create reporter knock-ins to monitor endogenous DMRT1 expression in live tissues

    • Perform epigenome editing to manipulate methylation status of the DMRT1 promoter

  • Single-cell multi-omics:

    • Apply single-cell RNA-seq to identify cell-specific expression patterns during gonadal differentiation

    • Combine with ATAC-seq to correlate chromatin accessibility with DMRT1 expression

    • Implement spatial transcriptomics to map DMRT1 expression in the context of developing gonadal architecture

  • Chromatin conformation analysis:

    • Use Hi-C or related techniques to examine temperature-dependent changes in chromatin organization around the DMRT1 locus

    • Identify long-range interactions between DMRT1 and other regulatory elements

  • Organoid models:

    • Develop gonadal organoid culture systems to study DMRT1 function under controlled conditions

    • Test temperature responsiveness in vitro with precise manipulation of culture conditions

How might comparative studies of DMRT1 across reptilian lineages inform our understanding of sex determination evolution?

Comparative evolutionary studies offer valuable insights into the conservation and divergence of DMRT1 function:

  • Sequence evolution analysis:

    • Compare coding sequences to identify conserved functional domains and species-specific adaptations

    • Analyze promoter regions to identify temperature-responsive elements in TSD species

  • Expression pattern comparisons:

    • Systematically compare DMRT1 expression across TSD and GSD reptiles

    • Identify correlations between expression patterns and specific sex-determining mechanisms

  • Functional conservation testing:

    • Perform cross-species complementation experiments (e.g., express alligator DMRT1 in turtle embryos)

    • Test whether temperature responsiveness is intrinsic to the DMRT1 protein or its regulatory context

  • Ancestral state reconstruction:

    • Combine molecular data with phylogenetic analyses to reconstruct the evolutionary history of DMRT1 function

    • Identify molecular signatures associated with transitions between TSD and GSD mechanisms

Through these comparative approaches, researchers can trace the evolutionary history of DMRT1 function and understand its role in the repeated transitions between environmental and genetic sex determination mechanisms observed in reptilian lineages.

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.