OSR2 Human

Odd-Skipped Related Transcription Factor 2 Human Recombinant
Shipped with Ice Packs
In Stock

Description

OSR2 Human Recombinant produced in E.Coli is a single, non-glycosylated polypeptide chain containing 299 amino acids (1-276 a.a) and having a molecular mass of 33kDa.
OSR2 is fused to a 23 amino acid His-tag at N-terminus & purified by proprietary chromatographic techniques.

Product Specs

Introduction
Odd-Skipped Related Transcription Factor 2, also known as OSR2, is a member of the Odd C2H2-type zinc-finger protein family. It is a mammalian counterpart to the Drosophila odd-skipped family of transcription factors.
Description
Recombinant human OSR2 protein, expressed in E. coli, is a single, non-glycosylated polypeptide chain. It comprises 299 amino acids (with amino acids 1 to 276 representing OSR2) and has a molecular weight of 33 kDa. A 23-amino acid His-tag is fused to the N-terminus of the OSR2 protein. Purification is achieved using proprietary chromatographic methods.
Physical Appearance
Clear, colorless solution, sterile-filtered.
Formulation
OSR2 protein solution at a concentration of 1 mg/ml. The solution is buffered with 20 mM Tris-HCl at pH 8.0 and contains 10% glycerol and 0.4 M urea.
Stability
For short-term storage (2-4 weeks), the protein should be kept at 4°C. For longer storage, it is recommended to freeze the protein at -20°C. Adding a carrier protein like HSA or BSA to a final concentration of 0.1% is advised for long-term storage. Avoid repeated freeze-thaw cycles.
Purity
Purity is determined to be greater than 80% via SDS-PAGE analysis.
Synonyms
Odd-Skipped Related Transcription Factor 2 , Odd-Skipped Related 2 (Drosophila), Protein odd-skipped-related 2.
Source
E.coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSMGSKALP APIPLHPSLQ LTNYSFLQAV NTFPATVDHL QGLYGLSAVQ TMHMNHWTLG YPNVHEITRS TITEMAAAQG LVDARFPFPA LPFTTHLFHP KQGAIAHVLP ALHKDRPRFD FANLAVAATQ EDPPKMGDLS KLSPGLGSPI SGLSKLTPDR KPSRGRLPSK TKKEFICKFC GRHFTKSYNL LIHERTHTDE RPYTCDICHK AFRRQDHLRD HRYIHSKEKP FKCQECGKGF CQSRTLAVHK TLHMQTSSPT AASSAAKCSG ETVICGGTA.

Q&A

What is the OSR2 gene and what is its basic function in humans?

OSR2 (odd-skipped related transcription factor 2) is a member of the zinc finger family of transcription factors that regulates gene expression during development. It functions as a transcriptional regulator that controls cell fate determination, tissue morphogenesis, and organ development. OSR2 is the mammalian homolog of the Drosophila odd-skipped family of transcription factors, with conserved functional domains across species . In humans, OSR2 plays critical roles in craniofacial development, particularly in palatogenesis, where it regulates the growth and morphogenesis of palatal shelves . The protein contains zinc finger domains that facilitate DNA binding and transcriptional regulation of target genes involved in developmental processes.

What are the key molecular characteristics of the human OSR2 protein?

The human OSR2 protein (UniProt ID: Q8N2R0) is characterized by multiple zinc finger domains that enable DNA binding and transcriptional regulation. Its sequence shows high conservation among mammals, with 96% identity to mouse and 99% identity to rat orthologs in certain regions . The protein contains specific functional domains including:

Domain/RegionFunction
Zinc finger domainsDNA binding and target gene recognition
Transcriptional regulatory domainsRepression or activation of target gene expression
Protein-protein interaction motifsRecruitment of cofactors and chromatin remodeling complexes

The protein exhibits sequence-specific DNA binding activity and functions primarily as a transcriptional repressor in developmental contexts, though it may act as an activator in certain cellular environments . OSR2 contains conserved sequences that are immunogenic and can be targeted by antibodies for research applications, including the sequence "TMHMNHWTLGYPNVHEITRSTITEMAAAQGLVDARFPFPALPFTTHLFHPKQGAIAHVLPALHKDRPRFDFANL" .

What are the primary tissue expression patterns of OSR2 in humans?

OSR2 exhibits specific expression patterns during development, with particularly important roles in craniofacial tissues. Based on expression profiling data, OSR2 shows differential expression across various tissues and developmental stages:

Tissue/Cell TypeExpression LevelDevelopmental Stage
Palatal mesenchymeHighEmbryonic (critical during E12.5-E13.5 in mouse models)
Craniofacial mesenchymeModerate to highEmbryonic
Mesenchymal cells undergoing EMTVariableEmbryonic and in certain cellular contexts
Adult brain tissuesVariable (region-specific)Adult

The gene expression profile of OSR2 can be analyzed using resources such as the Allen Brain Atlas and BioGPS datasets, which reveal tissue-specific expression patterns both in development and adult tissues . Understanding these expression patterns is crucial for researchers studying developmental processes or disease states involving OSR2 dysregulation.

What are the optimal experimental models for studying OSR2 function in craniofacial development?

When investigating OSR2 function in craniofacial development, researchers should consider multiple complementary experimental approaches:

  • Mouse genetic models: Osr2 knockout and conditional knockout mice provide valuable insights into palatal development. The Osr2^RFP/+ reporter mouse model allows visualization of OSR2-expressing cells during development . These models enable the study of phenotypic consequences of OSR2 loss or mutation in vivo.

  • Primary cell cultures: Isolation of palatal mesenchyme cells from embryonic day 12.5 (E12.5) and E13.5 mouse embryos allows for ex vivo manipulation and analysis of OSR2-dependent processes . These cultures can be subjected to RNA-seq, ChIP-seq, and functional assays to elucidate OSR2 target genes and regulatory networks.

  • 3D organoid models: Developing palatal organoids from human iPSCs or primary cells offers a more physiologically relevant system for studying human-specific aspects of OSR2 function. This approach is particularly valuable for translational research focused on craniofacial anomalies.

  • CRISPR/Cas9 genome editing: Generation of OSR2 mutations or tagged alleles in cellular models enables precise dissection of molecular functions and protein interactions.

For optimal results, researchers should combine multiple approaches to validate findings across different experimental systems, particularly when investigating complex developmental processes like palatogenesis.

What are the recommended methods for detecting OSR2 protein expression and activity?

Several complementary techniques can be employed for comprehensive analysis of OSR2 protein expression and activity:

MethodApplicationConsiderations
Immunohistochemistry/ImmunofluorescenceSpatial localization in tissuesRequires validated antibodies; consider using recombinant protein controls for blocking experiments
Western blottingProtein expression levelsUse appropriate controls; 100x molar excess of protein fragment control is recommended for blocking experiments
ChIP-seqGenome-wide binding sitesRequires high-quality ChIP-grade antibodies or tagged alleles
RNA-seqDownstream gene expression changesCan be combined with OSR2 manipulation (knockout/overexpression)
Reporter assaysTranscriptional activityUseful for testing direct regulation of target promoters

When using antibody-based detection methods, researchers should validate specificity using appropriate controls. For example, pre-incubation of the antibody with recombinant OSR2 protein fragment (100x molar excess) for 30 minutes at room temperature can be used in blocking experiments to confirm specificity . Additionally, using OSR2 knockout tissues or cells as negative controls enhances confidence in detection specificity.

How can researchers effectively manipulate OSR2 expression for functional studies?

For comprehensive functional analysis of OSR2, researchers can employ several complementary approaches to manipulate its expression:

  • CRISPR/Cas9-mediated gene editing:

    • For complete knockout: Target critical exons of OSR2

    • For conditional regulation: Use inducible Cas9 systems or floxed alleles with tissue-specific Cre recombinase

    • For tagging: Insert reporter genes (e.g., RFP) to track endogenous expression

  • RNA interference (RNAi):

    • siRNA or shRNA targeting OSR2 transcripts for transient or stable knockdown

    • Design multiple targeting sequences to control for off-target effects

    • Validate knockdown efficiency at both mRNA and protein levels

  • Overexpression systems:

    • For studying gain-of-function effects

    • Consider using expression vectors with uORF2 elements to modulate translation levels and minimize potential toxic effects of overexpression

    • Include appropriate tags (HA, FLAG) for detection while verifying tag doesn't interfere with function

  • Inducible expression systems:

    • Tet-On/Tet-Off systems for temporal control of OSR2 expression

    • Allows examination of stage-specific requirements and dose-dependent effects

When manipulating OSR2 expression, researchers should carefully consider the developmental timing and cell type specificity, as OSR2 functions may vary across different contexts. Additionally, rescue experiments with wild-type OSR2 should be performed to confirm phenotype specificity.

What is the role of OSR2 in palatogenesis and how is it implicated in cleft palate?

OSR2 functions as a key intrinsic regulator of palatal shelf growth and morphogenesis, with its dysfunction strongly linked to cleft palate formation. Mechanistically, OSR2 coordinates several critical aspects of palatogenesis:

  • Regulation of palatal mesenchyme proliferation: OSR2 maintains appropriate proliferative capacity of palatal mesenchyme cells during critical stages of palate development .

  • Suppression of osteogenic differentiation: RNA-seq analysis of OSR2-deficient palatal mesenchyme revealed upregulation of multiple osteogenic pathway genes, indicating that OSR2 normally prevents premature osteogenic differentiation that would impair palatal shelf growth and elevation .

  • Modulation of signaling pathways: OSR2 controls both BMP and Semaphorin 3 (Sema3) signaling pathways in the developing palate . Specifically, OSR2 directly represses expression of several Sema3 family members, which play diverse roles in cell proliferation, migration, and differentiation.

  • Spatial organization of developmental programs: OSR2 contributes to establishing proper medial-lateral patterning within the developing palatal shelves, which is essential for correct fusion.

Mutations or deletion of OSR2 result in cleft palate phenotypes in mouse models, characterized by reduced palatal shelf growth, abnormal shelf morphology, and failure of palatal fusion. These developmental defects arise from dysregulation of the molecular programs that OSR2 normally controls, including aberrant activation of osteogenic differentiation pathways and disrupted Sema3 signaling .

How does OSR2 regulate epithelial-mesenchymal transition (EMT) and what are the implications for cellular reprogramming?

OSR2 functions as a novel regulator of epithelial-mesenchymal transition (EMT), with significant implications for cellular reprogramming processes. Recent research has revealed that:

  • OSR2 promotes mesenchymal phenotypes: OSR2 acts as a positive regulator of EMT, maintaining mesenchymal characteristics in cells where it is expressed .

  • TGF-β signaling mediation: OSR2 induces EMT through activation of the TGF-β signaling pathway, a master regulator of mesenchymal transition .

  • Wnt signaling suppression: OSR2 expression appears to inhibit Wnt signaling, which is often associated with epithelial states .

  • Reprogramming barrier function: During somatic cell reprogramming to induced pluripotent stem cells (iPSCs), OSR2 downregulation is crucial for efficient reprogramming progress . This is because:

    • Reprogramming requires mesenchymal-to-epithelial transition (MET), the reverse of EMT

    • Continued OSR2 expression maintains mesenchymal characteristics that impede reprogramming

    • OSR2 downregulation allows diminished TGF-β signaling and activation of Wnt signaling, promoting MET and acquisition of pluripotency

These findings illuminate the functional significance of OSR2 in cellular plasticity and lineage specification. For researchers working on cellular reprogramming, targeting OSR2 expression could potentially enhance reprogramming efficiency by facilitating the critical MET process. Similarly, in developmental contexts or disease states involving abnormal EMT (such as fibrosis or cancer progression), OSR2 may represent a novel therapeutic target for modulating cellular phenotypes.

What are the currently known downstream targets and pathways regulated by OSR2?

OSR2 regulates a complex network of target genes and signaling pathways that collectively mediate its effects on development and cellular phenotypes. Whole transcriptome RNA sequencing analyses of OSR2-deficient tissues have identified several key targets and pathways:

Pathway/ProcessEffect of OSR2Target GenesBiological Consequence
BMP signalingSuppressionMultiple BMP pathway componentsPrevents premature osteogenic differentiation in palatal mesenchyme
Semaphorin 3 signalingDirect repressionMultiple Sema3 family membersControls cell proliferation, migration, and differentiation in palatal mesenchyme
TGF-β signalingActivationTGF-β pathway componentsPromotes and maintains mesenchymal phenotypes during EMT
Wnt signalingSuppressionWnt pathway componentsInhibits epithelial characteristics; downregulation of OSR2 allows Wnt activation during MET
Osteogenic differentiationSuppressionGenes involved in skeletal development and osteoblast differentiationMaintains appropriate timing of differentiation in developing tissues

Differential expression analysis from RNA-seq studies has identified that 70 genes were upregulated and 61 genes were downregulated by >1.5-fold in OSR2-deficient palatal mesenchyme cells compared to controls . Gene ontology analysis revealed enrichment of signaling molecules and transcription factors critical for skeletal development and osteoblast differentiation among those significantly upregulated in OSR2 mutant tissue .

OSR2 can directly bind to the regulatory regions of target genes, functioning primarily as a transcriptional repressor. For instance, it has been demonstrated to directly repress the expression of several Sema3 family members in the developing palatal mesenchyme by binding to their promoters .

How can single-cell transcriptomics advance our understanding of OSR2 function in heterogeneous tissues?

Single-cell transcriptomics represents a powerful approach to dissect OSR2 function within heterogeneous developmental contexts where bulk RNA-seq may mask critical cell type-specific effects:

  • Cell population identification: Single-cell RNA-seq (scRNA-seq) can identify distinct subpopulations of OSR2-expressing cells within complex tissues, revealing previously unrecognized cellular heterogeneity. This is particularly valuable in the developing palate and other craniofacial tissues where multiple cell types interact during morphogenesis.

  • Trajectory analysis: Computational reconstruction of developmental trajectories from scRNA-seq data can reveal how OSR2 expression changes during cell fate transitions, providing insights into its temporal regulation during development.

  • Cell-type specific targets: By comparing transcriptomes of OSR2-expressing versus non-expressing cells within the same tissue microenvironment, researchers can identify cell type-specific targets and functions of OSR2. This approach may uncover context-dependent roles that are obscured in bulk analyses.

  • Regulatory network inference: Integration of scRNA-seq with other single-cell modalities (e.g., ATAC-seq, ChIP-seq) enables construction of cell type-specific gene regulatory networks centered on OSR2, revealing cooperative and antagonistic interactions with other transcription factors.

For optimal implementation, researchers should:

  • Use fluorescent reporters (e.g., OSR2-RFP) or antibody-based methods to enrich for OSR2-expressing cells

  • Include multiple developmental timepoints to capture dynamic processes

  • Apply trajectory analysis tools (e.g., RNA velocity) to infer directionality of cellular transitions

  • Validate key findings with spatial transcriptomics or in situ methods to preserve tissue context

This approach could reveal previously unrecognized roles of OSR2 in specific cell populations within developing tissues, potentially identifying new therapeutic targets for craniofacial abnormalities.

What are the challenges and solutions in studying OSR2 protein-protein interactions and transcriptional complexes?

Investigating OSR2 protein-protein interactions and transcriptional complexes presents several challenges due to its context-dependent function as a transcription factor. Here are the major challenges and methodological solutions:

ChallengeMethodological Solutions
Low endogenous expression levels1. Use tagged OSR2 constructs (ensuring tag doesn't interfere with function)
2. Implement tandem affinity purification (TAP) for enhanced sensitivity
3. Develop knock-in cell lines with endogenous tags via CRISPR/Cas9
Context-dependent interactions1. Study interactions in relevant developmental contexts (e.g., palatal mesenchyme)
2. Compare interaction profiles across different cell types and developmental stages
3. Use inducible systems to capture dynamic interaction changes
Technical artifacts in overexpression systems1. Validate with reciprocal immunoprecipitation of endogenous proteins
2. Use proximity labeling methods (BioID, APEX) at near-physiological expression levels
3. Implement the expression vector with uORF2 to reduce translation and minimize artifacts
Distinguishing direct from indirect interactions1. Combine co-IP with crosslinking methods
2. Use protein fragment complementation assays
3. Perform in vitro binding assays with purified components
Identifying DNA-bound complexes1. ChIP-seq with sequential ChIP for co-occupancy
2. CUT&RUN or CUT&Tag for higher resolution and sensitivity
3. HiChIP to connect interactions with chromatin organization

For comprehensive characterization, researchers should implement a multi-method approach:

  • Initial screening using mass spectrometry-based interactome analysis

  • Validation of key interactions using orthogonal methods (co-IP, proximity labeling)

  • Functional validation through mutagenesis of interaction domains

  • Integration with genomic binding data to identify context-specific transcriptional complexes

This systematic approach can uncover how OSR2 assembles different protein complexes to regulate distinct target genes in various developmental contexts, providing mechanistic insights into its diverse functions.

How might OSR2 function be harnessed for regenerative medicine and tissue engineering applications?

The unique properties of OSR2 in regulating developmental processes and cellular transitions present several promising applications for regenerative medicine and tissue engineering:

  • Craniofacial tissue engineering:

    • Modulating OSR2 expression in mesenchymal stem cells could guide their differentiation trajectory for palatal and other craniofacial tissue reconstruction

    • Temporal control of OSR2 activity could recapitulate developmental sequences for more naturalistic tissue formation

    • Engineering scaffolds with OSR2-expressing cells may improve integration and patterning of engineered craniofacial tissues

  • Enhanced cellular reprogramming:

    • Targeted inhibition of OSR2 during somatic cell reprogramming could significantly improve iPSC generation efficiency by facilitating mesenchymal-epithelial transition (MET)

    • Sequential modulation of OSR2 (initial suppression followed by controlled expression) might enable more precise direction of cell fate during reprogramming

    • Integration of OSR2 regulation with other reprogramming factors could yield more complete epigenetic reprogramming

  • Controlling cell plasticity in tissue regeneration:

    • Temporal regulation of OSR2 could help control the balance between EMT and MET during wound healing to reduce scarring and fibrosis

    • In tissues that heal poorly, transient OSR2 manipulation might promote regenerative versus scarring outcomes

    • Combining OSR2 modulation with extracellular matrix modifications could enhance tissue integration

  • Disease modeling and drug discovery:

    • OSR2-reporter systems in organoids could serve as biosensors for developmental toxicity testing

    • Patient-derived cells with OSR2 mutations could be used to create disease models of craniofacial anomalies

    • High-throughput screening for compounds that modulate OSR2 activity could identify potential therapeutics for conditions involving aberrant EMT (fibrosis, cancer progression)

Implementing these applications requires addressing several challenges:

  • Developing methods for precise temporal and spatial control of OSR2 activity

  • Creating delivery systems for OSR2-modulating agents that target specific tissues

  • Understanding the interplay between OSR2 and tissue-specific factors in different regenerative contexts

Research in these directions could transform our approach to craniofacial reconstruction, cellular reprogramming, and regenerative therapies for conditions involving aberrant development or tissue repair.

What are the implications of OSR2 dysregulation in human pathological conditions beyond craniofacial abnormalities?

While OSR2's role in craniofacial development is well-established, emerging evidence suggests its dysregulation may contribute to various pathological conditions through its effects on cellular plasticity and differentiation:

  • Fibrotic disorders:

    • As a regulator of EMT and TGF-β signaling, OSR2 may contribute to fibrotic processes in multiple organs

    • Sustained OSR2 activation could potentially maintain mesenchymal phenotypes in fibroblasts, promoting fibrosis

    • Tissues showing high OSR2 expression may exhibit different fibrotic responses, suggesting tissue-specific pathogenic mechanisms

  • Cancer progression and metastasis:

    • OSR2's ability to regulate EMT suggests potential roles in cancer cell invasion and metastasis

    • Examining OSR2 expression across cancer databases might reveal correlations with invasive phenotypes or patient outcomes

    • Cancer-specific mutations or epigenetic alterations in OSR2 could contribute to disease progression in certain malignancies

  • Wound healing and tissue regeneration disorders:

    • Aberrant OSR2 expression might disrupt the balance between regeneration and scarring

    • The temporal dynamics of OSR2 expression during wound healing could influence repair outcomes

    • Genetic variations in OSR2 might contribute to individual differences in scarring propensity

  • Stem cell dysfunction and aging:

    • OSR2's role in cellular plasticity suggests potential involvement in stem cell maintenance and function

    • Age-related changes in OSR2 expression or activity could contribute to declining regenerative capacity

    • Epigenetic regulation of OSR2 might link developmental programming to adult tissue homeostasis

  • Connective tissue disorders:

    • Given its role in mesenchymal cell biology, OSR2 dysfunction might contribute to certain connective tissue abnormalities

    • Studying OSR2 in models of connective tissue disorders could reveal previously unrecognized pathogenic mechanisms

Research strategies to explore these connections include:

  • Gene association studies in patient cohorts with relevant pathologies

  • Analysis of OSR2 expression patterns in disease tissue banks

  • Functional studies in disease-specific cellular and animal models

  • Integration of OSR2 regulatory networks with disease-associated pathways

These investigations may reveal OSR2 as a potential therapeutic target in multiple pathological contexts beyond craniofacial development, particularly in conditions involving dysregulated cellular plasticity, inappropriate EMT, or aberrant tissue remodeling.

What are common technical challenges when working with OSR2 and how can they be addressed?

Researchers working with OSR2 frequently encounter several technical challenges that can impede progress. Here are the most common issues and effective solutions:

ChallengeCausesSolutions
Antibody specificity issuesCross-reactivity with related zinc finger proteins; non-specific binding1. Validate using OSR2 knockout samples as negative controls
2. Perform blocking experiments with recombinant OSR2 protein fragments (100x molar excess)
3. Use multiple antibodies targeting different epitopes for confirmation
Inconsistent knockdown efficiencySecondary structure of OSR2 mRNA; cell type-specific factors1. Design multiple siRNA/shRNA sequences targeting different regions
2. Optimize transfection conditions for each cell type
3. Consider using CRISPR/Cas9 for complete knockout instead
Functional redundancy with other factorsCompensation by related transcription factors (e.g., OSR1)1. Perform double knockdown/knockout experiments
2. Use domain-specific mutations rather than complete elimination
3. Implement acute depletion systems (e.g., degron tags) to minimize compensation
Developmental stage-specific effectsTemporal dynamics of OSR2 function1. Use inducible expression/knockdown systems
2. Sample multiple developmental timepoints
3. Implement lineage tracing to follow cellular trajectories
Difficulties in ChIP experimentsLow abundance; context-specific binding1. Use epitope-tagged OSR2 constructs to enhance immunoprecipitation
2. Implement CUT&RUN or CUT&Tag for higher sensitivity
3. Carefully select relevant cell types that recapitulate in vivo binding contexts

Additional considerations for experimental design:

  • When using reporter constructs, include multiple biological replicates and appropriate controls

  • For developmental studies, carefully stage embryos and consider strain-specific differences in developmental timing

  • When comparing wild-type and OSR2-deficient samples, use littermate controls to minimize genetic background effects

How should researchers interpret seemingly contradictory data on OSR2 function across different experimental systems?

Interpreting contradictory findings about OSR2 requires systematic analysis of experimental context and methodological differences. Here's a framework for reconciling apparently conflicting data:

  • Analyze context-dependent functions:

    • OSR2 may act differently in various cellular contexts due to:

      • Presence/absence of specific cofactors or interacting proteins

      • Cell type-specific chromatin landscapes affecting DNA binding

      • Different signaling environments modulating OSR2 activity

    • Solution: Directly compare OSR2 function across standardized cell systems with defined variables

  • Evaluate developmental timing effects:

    • OSR2 may have stage-specific functions during development

    • Early vs. late effects may appear contradictory but represent temporal specificity

    • Solution: Use time-course experiments with precise developmental staging

  • Consider dose-dependent effects:

    • Different OSR2 expression levels may yield opposite phenotypes

    • Partial vs. complete loss-of-function may reveal different aspects of OSR2 biology

    • Solution: Implement graduated expression systems to test dose-response relationships

  • Assess methodological variables:

    • Different knockout/knockdown strategies may have distinct off-target effects

    • Acute vs. chronic manipulation may allow different compensatory mechanisms

    • Solution: Use multiple complementary approaches to validate key findings

  • Examine species-specific differences:

    • OSR2 function may vary between mouse models and human systems

    • Regulatory networks may be differently wired across species

    • Solution: Compare findings in multiple species when possible and acknowledge limitations of model systems

When encountering contradictory data, researchers should:

  • Systematically compare experimental conditions across studies

  • Test whether apparent contradictions reflect different aspects of a more complex biological role

  • Design experiments that directly address the source of contradiction

  • Consider that seemingly opposite effects might reflect a common underlying mechanism with context-dependent outcomes

This approach transforms contradictory findings from obstacles into opportunities to develop more nuanced understanding of OSR2's multifaceted roles in development and disease.

What are the best practices for designing experiments to study OSR2 in the context of human diseases and developmental disorders?

When investigating OSR2 in human disease contexts, researchers should implement these best practices to ensure clinical relevance and translational potential:

  • Patient-derived cellular models:

    • Utilize patient-specific iPSCs from individuals with relevant craniofacial abnormalities

    • Differentiate iPSCs into disease-relevant cell types (palatal mesenchyme, neural crest derivatives)

    • Compare OSR2 expression, localization, and function between patient and control cells

    • Perform rescue experiments with wild-type OSR2 to confirm causality

  • Genetic association analyses:

    • Screen for OSR2 variants in patient cohorts with craniofacial abnormalities

    • Analyze both coding and regulatory regions, including enhancer elements

    • Perform functional validation of identified variants using reporter assays and CRISPR knock-in models

    • Consider gene-environment interactions that may modify OSR2-associated phenotypes

  • Disease-relevant 3D models:

    • Develop organoid systems that recapitulate key aspects of human craniofacial development

    • Implement OSR2 mutations or manipulations in organoid systems

    • Evaluate phenotypic outcomes using both morphological and molecular analyses

    • Test potential therapeutic interventions in established disease models

  • Translational research design:

    • Focus on reversible aspects of OSR2-associated phenotypes

    • Establish clear endpoints that have human disease relevance

    • Design experiments with clinically feasible interventions in mind

    • Consider age and sex as biological variables in experimental design

  • Multi-omic approaches:

    • Integrate transcriptomic, epigenomic, and proteomic analyses

    • Compare multi-omic profiles between normal and disease states

    • Identify disease-specific alterations in OSR2 regulatory networks

    • Use systems biology approaches to identify potential therapeutic targets

  • Rigorous controls and validation:

    • Include multiple control cell lines or samples to account for genetic background

    • Validate key findings across different experimental systems

    • Implement blinded analysis where appropriate

    • Consider statistical power when designing experiments with patient samples

  • Ethical considerations:

    • Ensure appropriate informed consent for patient samples

    • Consider privacy implications of genetic data

    • Design experiments to maximize scientific value from limited patient resources

    • Include patient perspectives in research prioritization when possible

By following these best practices, researchers can enhance the clinical relevance of their findings on OSR2 function in human diseases, potentially accelerating the translation of basic insights into therapeutic strategies for related developmental disorders.

What emerging technologies could enhance our understanding of OSR2 regulation and function?

Several cutting-edge technologies show particular promise for advancing OSR2 research:

  • Spatial transcriptomics and proteomics:

    • Technologies like Visium, MERFISH, or Slide-seq can map OSR2 expression and its targets with spatial resolution in developing tissues

    • This approach would reveal microenvironmental influences on OSR2 function and identify region-specific roles in complex tissues like the developing palate

    • Integration of spatial data with temporal dynamics would provide 4D understanding of OSR2 activity during morphogenesis

  • CRISPR screening technologies:

    • CRISPR activation/interference (CRISPRa/CRISPRi) screens targeting OSR2 regulatory elements could identify distal enhancers controlling context-specific expression

    • Combinatorial CRISPR screens could uncover genetic interactions between OSR2 and other developmental regulators

    • Base editing and prime editing enable precise introduction of disease-associated variants for functional characterization

  • Single-molecule imaging:

    • Live-cell imaging of fluorescently tagged OSR2 would reveal dynamics of DNA binding and transcriptional complex assembly

    • Single-molecule tracking could determine residence times at target loci and correlate with transcriptional outcomes

    • Super-resolution microscopy could visualize OSR2-containing complexes at subnuclear resolution

  • Organoid technologies and microphysiological systems:

    • Advanced organoid systems modeling palate development would enable study of human-specific aspects of OSR2 function

    • Organoid-on-chip approaches could incorporate fluid flow and mechanical forces relevant to craniofacial morphogenesis

    • Patient-derived organoids would facilitate personalized study of OSR2 variants in disease contexts

  • Multi-modal single-cell technologies:

    • Methods combining transcriptomic, epigenomic, and proteomic profiling from the same cells would provide comprehensive view of OSR2 regulatory networks

    • CITE-seq approaches could link surface markers with OSR2-dependent transcriptional programs

    • Trajectory inference from multi-modal data would reveal mechanisms of cell fate decisions controlled by OSR2

  • In situ genome and proteome editing:

    • Spatially resolved genome editing in developing tissues could reveal region-specific OSR2 functions

    • Optogenetic or chemogenetic control of OSR2 activity would enable precise temporal manipulation

    • Targeted protein degradation approaches would allow acute depletion of OSR2 to distinguish direct from secondary effects

Implementation of these technologies will require interdisciplinary collaboration and integration of computational approaches for data analysis, but promises to transform our understanding of OSR2 biology across developmental, cellular, and disease contexts.

What are the most promising therapeutic approaches for addressing OSR2-related developmental disorders?

Several innovative therapeutic strategies show potential for addressing OSR2-related developmental disorders:

  • Gene therapy approaches:

    • AAV-mediated delivery of functional OSR2 could potentially rescue loss-of-function phenotypes

    • CRISPR-based approaches might correct specific OSR2 mutations in affected tissues

    • Targeted enhancement of endogenous OSR2 expression via CRISPRa could compensate for haploinsufficiency

    • Challenges include achieving appropriate developmental timing and cell type specificity of intervention

  • Small molecule modulators:

    • High-throughput screening could identify compounds that modulate OSR2 activity or stabilize mutant proteins

    • Targeting downstream pathways (e.g., BMP, TGF-β, or Wnt signaling) might bypass OSR2 dysfunction

    • Chemical chaperones could potentially rescue misfolding of certain OSR2 mutant proteins

    • Advantages include temporal control and potential for dosage adjustment

  • Cellular therapies:

    • Neural crest-derived stem cells engineered with corrected OSR2 could potentially contribute to craniofacial repair

    • iPSC-derived mesenchymal cells with optimized OSR2 expression might enhance tissue engineering approaches

    • Ex vivo gene correction followed by autologous transplantation could reduce immunological complications

    • Integration with bioengineered scaffolds could improve functional outcomes

  • RNA therapeutics:

    • Antisense oligonucleotides could modulate OSR2 splicing or expression

    • mRNA delivery could provide transient OSR2 expression at critical developmental windows

    • RNA editing approaches might correct specific mutations without permanent genome modification

    • Potential for repeated administration if needed for maintenance therapy

  • Combinatorial approaches:

    • Sequential modulation of multiple pathways (e.g., initial OSR2 rescue followed by targeted growth factor delivery)

    • Integration of pharmacological and cellular approaches for synergistic effects

    • Personalized therapy design based on specific genetic variants and phenotypic manifestations

    • Multi-stage interventions aligned with developmental windows

Critical considerations for translational development include:

  • Prenatal diagnosis would be essential for early intervention in developmental disorders

  • Postnatal interventions might still address secondary complications or progressive aspects

  • Delivery methods must consider the blood-brain barrier for neurological manifestations

  • Safety profiles must be extensively evaluated given potential off-target effects in developing tissues

While significant challenges remain, these approaches represent promising avenues for therapeutic development, potentially transforming currently untreatable OSR2-related developmental disorders into manageable or preventable conditions.

How might systems biology approaches reveal new insights into OSR2's role in development and disease?

Systems biology approaches offer powerful frameworks for understanding OSR2's complex roles within broader developmental and disease networks:

  • Network-based analysis:

    • Construction of OSR2-centered gene regulatory networks across developmental timepoints

    • Identification of key network motifs (feedback loops, feed-forward circuits) involving OSR2

    • Network perturbation analysis to predict system-wide effects of OSR2 dysfunction

    • Comparison of network architecture across normal and disease states

  • Multi-scale modeling:

    • Integration of molecular interactions, cellular behaviors, and tissue morphogenesis into cohesive models

    • Agent-based modeling to simulate how OSR2-regulated cellular behaviors emerge as tissue-level phenotypes

    • Mechanistic modeling of how physical forces interact with OSR2-dependent gene expression during morphogenesis

    • Prediction of critical developmental thresholds where OSR2 perturbation leads to pathological outcomes

  • Comparative systems approaches:

    • Cross-species analysis of OSR2 regulatory networks to identify evolutionarily conserved vs. species-specific components

    • Comparison of OSR2 networks across different developmental contexts (craniofacial vs. other tissues)

    • Analysis of network rewiring in disease states compared to normal development

    • Identification of compensatory mechanisms that buffer against OSR2 perturbation

  • Multi-omic data integration:

    • Bayesian approaches to integrate diverse data types (genomic, transcriptomic, proteomic, metabolomic)

    • Machine learning methods to identify patterns in complex multi-omic datasets related to OSR2 function

    • Time-series analysis to capture dynamic aspects of OSR2-dependent processes

    • Causal inference methods to distinguish drivers from passengers in OSR2-associated phenotypes

  • Translational systems biology:

    • In silico prediction of drug targets within OSR2-dependent networks

    • Computational repositioning of existing drugs that may modulate OSR2-related pathways

    • Simulation of potential therapeutic interventions before experimental testing

    • Personalized modeling based on patient-specific genetic variants

Implementation of these approaches requires:

  • Development of computational tools specifically designed for developmental biology contexts

  • Standardized data collection across multiple model systems and human samples

  • Interdisciplinary collaboration between developmental biologists, computational scientists, and clinicians

  • Integration of qualitative biological knowledge with quantitative modeling approaches

Systems biology approaches can transform our understanding of OSR2 from isolated molecular mechanisms to integrated networks, potentially revealing emergent properties and non-intuitive relationships that would remain hidden to reductionist approaches. This systems-level understanding could ultimately guide more effective therapeutic strategies for developmental disorders involving OSR2 dysfunction.

Product Science Overview

Gene and Protein Structure

The OSR2 gene is located on chromosome 8 in humans and encodes a protein that consists of 276 amino acids . This protein contains DNA-binding C2H2-type zinc finger domains in its C-terminal half, which are essential for its function as a transcription factor . The gene is highly conserved across different species, indicating its importance in developmental processes.

Function and Role in Development

OSR2 is primarily involved in the development of the mandibular molar tooth germ at the bud stage . It also plays a significant role in the growth and morphogenesis of the secondary palate . The secondary palate is a crucial structure that separates the nasal cavity from the oral cavity, and its proper development is essential for normal facial structure and function.

In an expression screen to identify new transcription factors regulating palate development, OSR2 was found to be specifically activated in the nascent palatal mesenchyme at the onset of palatal outgrowth . A targeted null mutation in OSR2 impairs palatal shelf growth and causes a delay in palatal shelf elevation, resulting in cleft palate . This highlights the gene’s critical role in palatal development and its potential involvement in congenital defects such as cleft palate.

Related Pathways and Disorders

OSR2 is associated with several developmental pathways, including those related to primary ovarian insufficiency . Mutations or dysregulation of OSR2 can lead to developmental disorders. For instance, intellectual developmental disorder, autosomal recessive 77, and intellectual developmental disorder, autosomal recessive 76, are associated with mutations in the OSR2 gene .

Research and Clinical Implications

Understanding the function and regulation of OSR2 is essential for developing therapeutic strategies for congenital defects such as cleft palate. Research on OSR2 can also provide insights into the molecular mechanisms underlying other developmental disorders. The human recombinant form of OSR2 is used in various research applications to study its function and role in development.

In conclusion, Odd-Skipped Related Transcription Factor 2 (Human Recombinant) is a vital protein involved in human development, particularly in the formation of the secondary palate. Its role in developmental pathways and associated disorders makes it a significant focus of research in developmental biology and genetics.

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.