GDF10 Human

Growth differentiation factor 10 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Structure and Classification

GDF10 belongs to the TGF-β superfamily, specifically within the BMP subfamily. It shares structural homology with BMP3 but is distinct in its regulatory functions. Key characteristics include:

  • Gene location: Chromosome 10 in humans.

  • Tissue expression: Brain, adipose tissue, prostate, retina, pineal gland, and bone marrow .

  • Protein interactions: Binds to TGF-β receptors (TβRI and TβRII) and Smad proteins, activating downstream signaling pathways .

Table 1: Tissue Expression of GDF10

TissueExpression LevelSource
Subcutaneous fatHigh
BrainModerate
ProstateModerate
Nasopharyngeal tissueLow (in cancer)
RetinaModerate

Developmental and Skeletal Roles

GDF10 is involved in:

  • Head formation: Critical for embryonic development .

  • Skeletal morphogenesis: Regulates bone tissue formation and differentiation .

Metabolic Regulation

GDF10 is an adipokine with implications in obesity and insulin resistance:

  • SAT vs. VAT expression: Higher in subcutaneous adipose tissue (SAT) than visceral adipose tissue (VAT) .

  • Serum levels: Positively correlated with BMI (r = 0.308, P = 0.019) .

  • Pathway suppression: High GDF10 expression in SAT downregulates genes involved in insulin response, glucose/lipid metabolism, and fatty acid oxidation .

Table 2: Impact of GDF10 on Metabolic Pathways

PathwayEffect of High GDF10 ExpressionSource
Insulin signaling↓ Suppression
Oxidative phosphorylation↓ Suppression
PPAR signaling↓ Suppression
AMPK signaling↓ Suppression

Neurological Functions

GDF10 promotes axonal sprouting and functional recovery after stroke:

  • Mechanism: Activates TGFβRI/II signaling, downregulates PTEN, and upregulates PI3 kinase .

  • Clinical relevance: Improves motor recovery in murine models, with tumors shrinking by 85% in GDF10-treated mice .

Table 3: GDF10 in Stroke Recovery

ParameterEffect of GDF10 AdministrationSource
Axonal sprouting
Tumor weight↓ (from 128.9 mg to 19.86 mg)
Motor recovery timeReduced to 5 weeks post-stroke

Cancer Involvement

GDF10 exhibits tumor-suppressive properties:

  • NPC models: Overexpression inhibits proliferation, induces apoptosis, and suppresses epithelial-to-mesenchymal transition (EMT) .

  • Signaling: Activates Smad3 via TβRI/TβRII, reducing tumor growth .

Table 4: GDF10 in Nasopharyngeal Carcinoma (NPC)

ParameterEffect of GDF10 OverexpressionSource
Cell proliferation
Apoptosis
E-cadherin (adhesion)
Vimentin (mesenchymal)

Obesity-Related Discrepancies

  • Adults: Higher serum GDF10 correlates with obesity (BMI ≥25, 2674 ± 441 pg/mL vs. 2339 ± 639 pg/mL) .

  • Children: Lower plasma GDF10 linked to obesity and elevated cholesterol .
    Possible explanations: Age-dependent regulatory mechanisms or assay sensitivity differences.

Therapeutic Potential

  • Obesity: Targeting GDF10 to improve insulin sensitivity (e.g., via PPARγ modulation) .

  • Stroke: Recombinant GDF10 administration for axonal recovery .

  • Cancer: Exploiting GDF10’s Smad3-dependent tumor suppression .

Research Gaps and Future Directions

  1. Mechanistic clarity: Resolve conflicting obesity-related GDF10 levels in adults vs. children.

  2. Signaling specificity: Elucidate GDF10’s interaction with BMP vs. TGF-β receptors.

  3. Therapeutic translation: Develop GDF10-based therapies for stroke and metabolic disorders, balancing its dual roles in adipose tissue and cancer.

Product Specs

Introduction
Growth differentiation factor 10 (GDF10), a member of the bone morphogenetic protein (BMP) family within the transforming growth factor-beta (TGF-β) superfamily, plays a crucial role in skeletal development. Expressed in various tissues including the femur, brain, lung, skeletal muscle, pancreas, and testis, GDF10 contributes to head formation and potentially multiple aspects of skeletal morphogenesis. Human GDF10 mRNA is detected in the fetal cochlea and lung, as well as in adult tissues such as the testis, retina, pineal gland, and other neural tissues. GDF10, like other BMP family members, regulates cell growth and differentiation in both embryonic and adult tissues. These proteins share a characteristic polybasic proteolytic processing site, which upon cleavage, yields a mature protein containing seven conserved cysteine residues.
Description
Recombinant human GDF10, produced in E. coli, is a single, non-glycosylated polypeptide chain comprising 111 amino acids (residues 369-478). With a molecular mass of 12.5 kDa, it encompasses the bioactive region of GDF10. The protein is purified using proprietary chromatographic techniques to ensure high purity.
Physical Appearance
Clear, colorless solution, sterile-filtered.
Formulation
The GDF10 solution is provided at a concentration of 1 mg/ml in a buffer containing 10 mM sodium citrate (pH 3.5), 1 mM dithiothreitol (DTT), 40% glycerol, and 0.1 M sodium chloride.
Stability
For short-term storage (up to 2-4 weeks), the GDF10 solution can be stored at 4°C. For extended storage, it is recommended to store the protein frozen at -20°C. To further enhance long-term stability, the addition of a carrier protein such as 0.1% human serum albumin (HSA) or bovine serum albumin (BSA) is advised. Repeated freeze-thaw cycles should be avoided to maintain protein integrity.
Purity
The purity of GDF10 is determined to be greater than 95% by sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) analysis.
Synonyms
Bone morphogenetic protein 3b, BMP-3b, Growth/differentiation factor 10, GDF-10, Bone-inducing protein, BIP, GDF10, BMP3B.
Source
Escherichia Coli.
Amino Acid Sequence
MQWDEPRVCS RRYLKVDFAD IGWNEWIISP KSFDAYYCAG ACEFPMPKIV RPSNHATIQS IVRAVGIIPG IPEPCCVPDK MNSLGVLFLD ENRNVVLKVY PNMSVDTCAC R.

Q&A

What is GDF10 and what are its basic structural characteristics?

GDF10 (Growth Differentiation Factor 10) is a member of the bone morphogenetic protein (BMP) family and belongs to the transforming growth factor-β (TGF-β) superfamily. Also known as BMP-3B due to its close relationship with bone morphogenetic protein-3 (BMP3), GDF10 contains a characteristic polybasic proteolytic processing site that undergoes cleavage to produce a mature protein . This mature protein contains seven conserved cysteine residues that are essential for its structural integrity and function .

GDF10 functions as a secreted TGF-β receptor ligand with growth factor activity that plays roles in cell growth, differentiation, and proliferation in both embryonic and adult tissues . The protein is encoded by the GDF10 gene (NCBI Gene ID: 2662) and has been identified by several synonyms including BMP3B and BMP-3B .

Through what signaling mechanisms does GDF10 function in human cells?

GDF10 primarily signals through the TGF-β receptor pathway, involving three cell surface receptors: TGFBR1, TGFBR2, and TGFBR3 . The signaling cascade is initiated when GDF10 binds to these receptors, particularly TGFBR2, which then activates TGFBR1 . This activation leads to phosphorylation of Smad2/3 proteins, which subsequently combine with Smad4 to form a complex . This complex translocates to the nucleus where it regulates the transcription of target genes .

Experimental evidence confirms this pathway's importance in GDF10 function. In human-induced pluripotent stem cell-derived neurons (hiPS-neurons), GDF10-induced axonal outgrowth is blocked by either pharmacological inhibition of TGFβRI or knockdown of TGFβRII . Similarly, knockdown of Smad2/3 prevents GDF10-mediated axonal outgrowth, confirming the relevance of this canonical signaling pathway .

How is GDF10 expression regulated in normal tissues versus pathological conditions?

GDF10 expression varies significantly across different tissue types and is dysregulated in several pathological conditions. In normal tissues, GDF10 shows differential expression patterns that can be observed through various gene expression databases including GTEx, HPA, and BioGPS .

In pathological conditions, particularly cancer, GDF10 is frequently downregulated. In triple-negative breast cancer (TNBC), RNA-Seq analysis of clinical specimens has shown significant downregulation of GDF10 compared to tumor-matched controls . This downregulation correlates with parameters of disease severity. Quantitative PCR and western blotting confirmed significantly lower GDF10 expression in TNBC cell lines (BT-20, MDA-MB-157, and HS598T) compared to non-tumorigenic human breast epithelial MCF10A cells . Importantly, immunohistochemistry analysis revealed significantly decreased GDF10 expression in stage III/IV TNBC specimens compared with stage I/II, suggesting progressive loss during cancer advancement .

In neurological contexts, GDF10 upregulation after stroke has been observed consistently across mice, non-human primates, and humans, indicating a conserved response to neurological injury .

What experimental models are commonly used to study GDF10 function?

Several experimental models have been developed to study GDF10 function across different biological contexts:

Cell Culture Models:

  • Human TNBC cell lines: MDA-MB-231, BT-20, MDA-MB-453, MDA-MB-157, and HS598T

  • Human breast epithelial MCF10A cells (non-tumorigenic control)

  • Human induced pluripotent stem cell-derived neurons (hiPS-neurons)

  • Mouse and rat neuronal models

Genetic Manipulation Approaches:

  • RNA interference using short hairpin RNAs (shRNAs) targeting GDF10 (e.g., GDF10-shRNA1 and GDF10-shRNA2)

  • Overexpression systems using plasmid vectors containing GDF10 cDNA

  • Knockout mouse models (through the IMPC Knockout Mouse Phenotypes dataset)

Functional Assays:

  • Cell viability assays

  • Proliferation assays (including Ki67 immunofluorescence staining)

  • Transwell invasion assays

  • Cell cycle analysis

  • Immunofluorescence for protein localization

  • RNA-seq for transcriptome analysis

  • Mouse xenograft models for tumorigenicity assessment

What are the key tissue expression patterns of GDF10 in humans?

GDF10 exhibits tissue-specific expression patterns, with data compiled from multiple high-throughput expression databases. Analysis of these patterns provides valuable context for researchers designing tissue-specific experiments.

Table 1: GDF10 Expression Patterns Across Human Tissues

DatabaseTissues with Significant GDF10 ExpressionMethodology
GTExVariable expression across multiple tissuesRNA-Seq
HPA (Human Protein Atlas)Tissue-specific expression patternsAntibody-based proteomics
Allen Brain AtlasExpression in specific brain regionsMicroarray and RNA-Seq
BioGPSCell type and tissue expression profilesMicroarray
Roadmap EpigenomicsDNA methylation profiles across tissuesMethylation sequencing

Researchers should note that GDF10 expression varies significantly between tissues, making it important to validate expression in the specific experimental context before proceeding with functional studies .

What methodologies are most effective for studying GDF10 expression changes in cancer tissues?

When investigating GDF10 expression in cancer tissues, a multi-modal approach yields the most comprehensive results. Based on successful methodologies from recent studies, the following integrated approach is recommended:

  • Transcriptomic Analysis:

    • RNA-Seq provides genome-wide expression profiling to identify GDF10 downregulation in tumor samples

    • Quantitative PCR (qPCR) for validation of expression changes in larger sample cohorts

    • Consideration of RNA quality is critical, particularly in clinical samples with variable preservation conditions

  • Protein-Level Analysis:

    • Western blotting with validated antibodies to confirm protein-level changes

    • Immunohistochemistry (IHC) to visualize spatial distribution in tissue sections

    • Densitometric analysis of IHC staining to quantify expression differences between stages (e.g., stage I/II vs. stage III/IV)

  • Clinicopathological Correlation:

    • Association of GDF10 expression with clinical parameters (TNM staging, Ki67 proliferation index)

    • Patient outcome data correlation for prognostic significance assessment

    • Multivariate analysis to control for confounding factors

  • Epigenetic Regulation Assessment:

    • DNA methylation analysis using the Roadmap Epigenomics Cell and Tissue DNA Methylation Profiles

    • Histone modification characterization using ChIP-seq approaches

Methodological Consideration: When analyzing GDF10 expression in mixed cell populations within tumor samples, cell type deconvolution algorithms should be applied to distinguish tumor cell expression from stromal contribution. Additionally, laser capture microdissection may be employed for isolating specific cellular populations before expression analysis.

How do experimental approaches differ when investigating GDF10's role in neural tissues versus epithelial cells?

GDF10 exhibits context-dependent functions requiring tailored experimental approaches across different tissue types:

Neural Tissue Investigations:

  • Model Systems: Human induced pluripotent stem cell-derived neurons (hiPS-neurons), primary neuronal cultures from rodents, and in vivo stroke models

  • Functional Assays: Axonal outgrowth measurements, axonal sprouting quantification, behavioral recovery assessments following stroke

  • Molecular Readouts: RNA-seq analysis focused on axonal guidance molecules and PI3 kinase signaling pathway components

  • Pharmacogenetic Approaches: Gain and loss of function studies to determine effects on axonal sprouting and functional recovery after stroke

Epithelial Cell Investigations:

  • Model Systems: Breast epithelial cell lines (e.g., MCF10A), TNBC cell lines (e.g., MDA-MB-231, BT-20)

  • Functional Assays: Proliferation assays, invasion assays, cell cycle analysis, EMT marker assessment

  • Molecular Readouts: Expression of EMT markers (E-cadherin, N-cadherin, vimentin), cell cycle regulators (cyclin D1, CDK4, CDK6), and apoptosis markers

  • In Vivo Models: Mouse xenograft models to assess tumorigenicity

Comparative Methodology Table:

AspectNeural Tissue ApproachEpithelial Cell Approach
Primary EndpointAxonal sprouting, functional recoveryCell proliferation, invasion, EMT
Signaling FocusPI3K pathway, axonal guidance moleculesEMT pathway, cell cycle regulators
In Vivo ModelsStroke modelsXenograft tumor models
TimeframeOften longer (weeks) for recovery assessmentShorter (days) for proliferation/invasion
Technical ChallengesComplex neural network analysisDistinguishing direct vs. indirect effects

When designing experiments across these tissue types, researchers should be aware that GDF10's downstream effectors and biological outcomes differ substantially, necessitating tissue-specific validation of findings rather than direct extrapolation between neural and epithelial contexts.

What are the challenges in elucidating GDF10's role in the TGF-β signaling network?

Investigating GDF10's precise role within the complex TGF-β signaling network presents several methodological challenges:

  • Receptor Promiscuity and Redundancy:

    • GDF10 signals through TGFβRI/II receptors which are utilized by multiple ligands

    • Experimental challenge: Distinguishing GDF10-specific effects from those of other TGF-β family members requires careful experimental design with appropriate controls

    • Recommended approach: Use of specific receptor knockdown or knockout models alongside ligand manipulation

  • Context-Dependent Signaling:

    • GDF10 effects vary significantly between tissue types and disease states

    • The pathway exhibits biphasic or contradictory effects depending on cellular context

    • Methodological solution: Systematic comparison across multiple cell types under identical experimental conditions

  • Complex Downstream Effector Networks:

    • GDF10 signaling involves both Smad-dependent and Smad-independent pathways

    • Cross-talk with other signaling cascades complicates interpretation

    • Analytical approach: Systems biology approaches including pathway enrichment analysis and network modeling

  • Technical Limitations in Measuring Pathway Activation:

    • Transient nature of Smad phosphorylation events

    • Nuclear translocation dynamics are difficult to capture

    • Advanced solution: Live-cell imaging with fluorescently tagged Smad proteins or phospho-specific antibodies with temporal sampling

  • Genetic Compensation in Knockout Models:

    • Complete GDF10 knockout may trigger compensatory upregulation of related factors

    • Strategy to overcome: Inducible or conditional knockout systems combined with acute manipulation approaches

Recommended Integrated Approach:
Researchers should employ multiplexed assays that simultaneously monitor multiple nodes in the signaling network, combined with computational modeling to interpret the complex interplay between pathway components. Time-course experiments are essential to capture the dynamic nature of these signaling events.

How can researchers effectively measure the impact of GDF10 on epithelial-mesenchymal transition?

Epithelial-mesenchymal transition (EMT) is a key process influenced by GDF10, particularly in cancer contexts. Based on successful approaches in the literature, the following comprehensive methodology is recommended:

  • Morphological Assessment:

    • Bright-field microscopy to document changes in cell shape and organization

    • Quantitative morphometric analysis using image processing software

    • Time-lapse imaging to capture dynamic morphological transitions

  • Molecular Marker Panel Analysis:

    • Epithelial Markers: E-cadherin, ZO-1, claudins, occludin

    • Mesenchymal Markers: N-cadherin, vimentin, fibronectin

    • EMT Transcription Factors: Snail, Slug, ZEB1, ZEB2, Twist

    • Methodology: Combined qPCR, western blotting, and immunofluorescence approaches

  • Functional EMT Assays:

    • Migration Assays: Wound healing/scratch assay with time-lapse imaging

    • Invasion Assays: Transwell chambers with Matrigel coating as demonstrated in GDF10 knockdown experiments

    • Cell Adhesion Assays: Measuring attachment to various substrates

  • Mechanistic Dissection:

    • Reporter assays for EMT-associated promoters

    • ChIP-seq to identify direct transcriptional targets

    • Analysis of subcellular localization of Smad proteins which correlate with changes in GDF10 expression

  • In Vivo Validation:

    • Orthotopic xenograft models with GDF10 manipulation

    • Immunohistochemical analysis of tumor sections for EMT markers

    • Circulating tumor cell analysis for EMT characteristics

Data Integration Approach:
Evidence from published studies indicates that GDF10 suppresses EMT in breast cancer cells . To comprehensively assess this function, researchers should establish a quantitative EMT score based on multiple parameters rather than relying on individual markers. This approach allows for detection of partial or intermediate EMT states that may have important biological significance.

What methodologies provide the best insights into GDF10's interaction with Smad proteins?

Understanding GDF10's interaction with Smad proteins requires a multi-faceted approach:

  • Protein-Protein Interaction Analysis:

    • Co-immunoprecipitation (Co-IP): To detect physical association between GDF10-activated receptors and Smad proteins

    • Proximity Ligation Assay (PLA): For visualizing interactions in situ with spatial resolution

    • FRET/BRET: To measure real-time interactions and conformational changes

    • Mammalian two-hybrid assays: For quantitative assessment of interaction strength

  • Smad Phosphorylation Dynamics:

    • Phospho-specific western blotting: Temporal profiling of Smad2/3 phosphorylation following GDF10

    • Mass spectrometry: Site-specific phosphorylation analysis

    • Kinetic analysis: Time-course experiments to determine activation/deactivation rates

    • Single-cell analysis: To address cell-to-cell variability in response

  • Nuclear Translocation and Chromatin Interaction:

    • Subcellular fractionation: Biochemical separation of cytoplasmic and nuclear fractions

    • Immunofluorescence: Visualization of Smad nuclear translocation

    • ChIP-seq: Genome-wide mapping of Smad binding sites after GDF10 stimulation

    • ATAC-seq: Assessment of chromatin accessibility changes

  • Transcriptional Output Analysis:

    • Reporter assays: Using Smad-responsive elements

    • RNA-seq: Global transcriptional changes following GDF10 treatment

    • GRO-seq or NET-seq: To measure nascent transcription

    • Single-cell RNA-seq: To capture cellular heterogeneity in response

  • Competitive Binding Studies:

    • Analysis of how GDF10-induced Smad signaling interacts with or is influenced by other TGF-β family members

    • Investigation of inhibitory Smad7 recruitment, which prevents TGF-β-dependent formation of Smad2/Smad4 complexes

Analytical Considerations:
Research has shown that changes in GDF10 expression correlate with changes in both expression and subcellular localization of Smad proteins . Therefore, experimental designs should incorporate both quantitative (expression level) and qualitative (localization, phosphorylation state) assessments. Additionally, since Smad signaling is highly dynamic, temporal resolution is crucial for accurate interpretation.

How can contradictory findings about GDF10 function across different tissue types be reconciled?

GDF10 exhibits context-dependent functions that sometimes appear contradictory across different tissues and experimental systems. Methodological approaches to reconcile these apparent contradictions include:

  • Systematic Tissue Comparison Studies:

    • Parallel experiments in multiple tissue types under identical conditions

    • Standardized readouts and analysis pipelines

    • Meta-analysis of published findings with careful attention to methodological differences

  • Cell Type-Specific Receptor and Co-factor Profiling:

    • Comprehensive analysis of TGF-β receptor expression patterns across tissues

    • Identification of tissue-specific co-receptors or adaptor proteins

    • Analysis of competing ligands in different microenvironments

  • Pathway Cross-talk Mapping:

    • Analysis of tissue-specific interaction with other signaling pathways

    • Identification of cell type-specific downstream effectors

    • Network analysis to identify divergent signaling nodes

  • Developmental and Microenvironmental Context:

    • Temporal analysis across developmental stages

    • Microenvironmental manipulation experiments

    • Artificial recreation of tissue-specific niches in vitro

Concrete Example from Literature:
RNA-seq analysis from peri-infarct cortical neurons indicates that GDF10 downregulates PTEN and upregulates PI3 kinase signaling while inducing specific axonal guidance molecules . In contrast, in epithelial cancer contexts, GDF10 influences EMT pathways and cell cycle regulators . These divergent effects likely reflect tissue-specific transcriptional landscapes and co-factor availability.

Interestingly, unsupervised genome-wide association analysis of the GDF10 transcriptome shows that it is not related to neurodevelopment but may partially overlap with other CNS injury patterns . This suggests that GDF10's function may be more related to contextual cellular states (e.g., injury response) than to inherent tissue identity.

To effectively reconcile contradictory findings, researchers should first establish whether differences are truly contradictory or simply reflect tissue-specific manifestations of a conserved underlying mechanism.

What are the most effective GDF10 gain and loss of function models for in vivo research?

Selecting appropriate gain and loss of function models is critical for robust GDF10 research. Based on successful approaches in the literature, the following models are recommended:

Loss of Function Models:

  • RNA Interference Approaches:

    • Short hairpin RNA (shRNA) targeting GDF10 has been effectively used in human cell lines

    • Both transient and stable knockdown systems using lentiviral vectors

    • Multiple shRNA constructs (e.g., GDF10-shRNA1 and GDF10-shRNA2) should be employed to control for off-target effects

  • CRISPR/Cas9 Genome Editing:

    • Complete knockout via indel formation

    • Knockin of inactivating mutations

    • CRISPRi for transcriptional repression without altering the genomic sequence

  • Conditional/Inducible Systems:

    • Cre-loxP systems for tissue-specific deletion

    • Tetracycline-inducible shRNA expression

    • Temporal control using tamoxifen-inducible Cre recombinase

Gain of Function Models:

  • Viral Vector Overexpression:

    • Adeno-associated virus (AAV) or lentiviral vectors carrying GDF10 cDNA

    • Tissue-specific promoters for targeted expression

    • Demonstrated efficacy in promoting axonal sprouting and functional recovery after stroke

  • Recombinant Protein Administration:

    • Direct application of purified GDF10 protein

    • Controlled release systems (e.g., hydrogels, microspheres)

    • Demonstrated to promote axonal outgrowth in human iPSC-derived neurons

  • Transgenic Overexpression:

    • Constitutive or inducible transgenic models

    • Tissue-specific promoters for targeted expression

    • Knock-in of additional gene copies

Experimental Design Recommendations:

  • Control Selection:

    • For knockdown: non-targeting shRNA controls

    • For protein treatments: heat-inactivated protein or irrelevant protein of similar size

    • Vehicle controls for all delivery methods

  • Validation Approaches:

    • qPCR and western blotting to confirm expression changes

    • Functional readouts appropriate to tissue context

    • Dose-response characterization for gain of function studies

  • Combinatorial Approaches:

    • Rescue experiments (knockdown followed by reintroduction)

    • Epistasis studies with downstream effectors

    • Combinatorial manipulation with pathway components

Pharmacogenetic gain and loss of function studies have demonstrated that GDF10 produces axonal sprouting and enhanced functional recovery after stroke, while knocking down GDF10 blocks axonal sprouting and reduces recovery . These findings validate the effectiveness of these models in studying GDF10 function in vivo.

What considerations are important when designing experiments to study GDF10's role in cell cycle regulation?

GDF10 has been implicated in cell cycle regulation, particularly in cancer contexts. The following experimental design considerations are critical:

  • Cell Synchronization Strategies:

    • Serum starvation for G0/G1 synchronization

    • Thymidine block or nocodazole treatment for specific phase enrichment

    • Important because GDF10 has been shown to induce G0/G1 arrest

  • Cell Cycle Analysis Methods:

    • Flow Cytometry: Propidium iodide staining for DNA content

    • EdU/BrdU Incorporation: For S-phase detection

    • Immunofluorescence: Cell cycle marker detection (e.g., Ki67 for proliferative state)

    • Time-lapse Imaging: For direct observation of division kinetics

  • Molecular Target Assessment:

    • Analysis of cyclins (particularly cyclin D1) and CDKs (CDK4, CDK6) which are regulated by GDF10

    • CDK inhibitors (p21, p27) expression analysis

    • Assessment of pocket protein (Rb) phosphorylation status

    • DNA damage response markers

  • Experimental Timeline Considerations:

    • Acute versus chronic GDF10 manipulation

    • Temporal profiling of cell cycle markers

    • Recovery experiments following GDF10 withdrawal

  • Context-Dependent Variables:

    • Growth factor availability in culture conditions

    • Cell density and contact inhibition effects

    • Extracellular matrix composition

Methodological Table for Cell Cycle Analysis:

PhaseMarkersTechniqueConsideration with GDF10
G0/G1Cyclin D1, CDK4/6Western blot, IFPrimary target of GDF10 regulation
SEdU incorporation, PCNAFlow cytometry, IFSecondary effect of G1/S transition block
G2/MCyclin B1, pH3Flow cytometry, IFLess affected by GDF10 directly
Quiescencep27, p21Western blot, IFIncreased with GDF10 overexpression

Critical Controls:
When studying GDF10's effects on cell cycle, researchers should include:

  • Positive controls (known cell cycle inhibitors like palbociclib)

  • Cell type-matched controls (e.g., comparing effects in cancer vs. non-transformed cells)

  • Time-matched vehicle controls

  • Dose-response experiments

Research has demonstrated that GDF10 overexpression in TNBC cells inhibits proliferation by inducing G0 arrest, highlighting the importance of appropriate cell cycle analysis methods when studying this protein .

What are the optimal experimental conditions for studying GDF10's role in axonal sprouting?

Research has established GDF10 as a signal for axonal sprouting and functional recovery after stroke . Designing rigorous experiments to study this function requires careful consideration of the following factors:

  • Neuronal Model Selection:

    • Primary Neurons: Rat or mouse primary cortical neurons

    • Human Models: Human induced pluripotent stem cell-derived neurons (hiPS-neurons)

    • Organotypic Slice Cultures: For preserved neural circuit architecture

    • In Vivo Models: Stroke models (e.g., middle cerebral artery occlusion)

  • GDF10 Delivery Methods:

    • Recombinant Protein Application: Purified GDF10 at physiologically relevant concentrations

    • Viral Vector Expression: AAV or lentiviral vectors for sustained expression

    • Conditional Expression Systems: For temporal control of GDF10 expression

    • Local Delivery Systems: Hydrogels or controlled release formulations

  • Axonal Sprouting Quantification Methods:

    • Neurite Outgrowth Assays: Measurement of neurite length and branching complexity

    • Axonal Tracing Techniques: Anterograde tracers for in vivo sprouting assessment

    • High-Content Imaging: Automated image acquisition and analysis

    • Live Imaging: Time-lapse microscopy for dynamic growth assessment

  • Receptor and Signaling Inhibition Controls:

    • TGFβRI pharmacological inhibitors to block GDF10 signaling

    • TGFβRII knockdown using siRNA/shRNA approaches

    • Smad2/3 knockdown to disrupt downstream signaling

    • PI3 kinase inhibitors to block upregulated pathway components

  • Functional Assessment in Stroke Models:

    • Behavioral tests for motor recovery (e.g., grid walking, cylinder test)

    • Electrophysiological recordings to assess functional connectivity

    • Correlation between axonal sprouting and behavioral improvement

Experimental Design Table:

ParameterRecommended ApproachValidation Method
GDF10 Concentration5-50 ng/mL for in vitro studiesDose-response curve
Treatment Duration24-72 hours for acute, 1-4 weeks for chronicTime-course analysis
Culture SubstrateLaminin or poly-D-lysine coatingComparison of growth on different substrates
Growth MediumNeurobasal with B27 supplementSerum vs. serum-free comparison
Analysis Timepoints24h, 72h, 7d post-treatmentMultiple timepoint sampling

Research has demonstrated that GDF10 promotes significant axonal outgrowth in human iPSC-derived neurons through TGFβRI/II signaling and Smad2/3 activation . This effect is conserved across mouse, rat, and human neurons, indicating robust cross-species validity of experimental findings .

How should researchers interpret contradictory data on GDF10 expression across different cancer types?

GDF10 expression patterns show significant variability across cancer types, requiring careful interpretation:

  • Subtype Stratification:

    • Different molecular subtypes within a cancer type may show divergent GDF10 expression

    • In breast cancer, GDF10 downregulation is particularly associated with triple-negative subtype

    • Stratification by molecular subtype, histological grade, and stage is essential for meaningful interpretation

  • Technical Considerations in Expression Analysis:

    • Platform Bias: Differences between microarray, RNA-seq, and qPCR methodologies

    • Probe/Primer Specificity: Ensuring detection of all relevant GDF10 isoforms

    • Reference Gene Selection: Critical for accurate normalization

    • Tumor Purity: Stromal contamination affecting bulk tumor expression profiles

  • Contextual Factors Affecting Expression:

    • Epigenetic Regulation: Differential methylation across cancer types

    • Genetic Alterations: Copy number variations, mutations affecting expression

    • Microenvironmental Influences: Hypoxia, inflammation, stromal interactions

    • Treatment Effects: Prior therapy potentially altering expression

  • Statistical Approaches for Meta-Analysis:

    • Random-effects models to account for interstudy heterogeneity

    • Publication bias assessment using funnel plots

    • Sensitivity analysis excluding outlier studies

    • Subgroup analysis based on methodology and patient characteristics

Analytical Framework for Resolving Contradictions:

When faced with contradictory findings regarding GDF10 expression:

  • Evaluate methodological differences (sample processing, detection methods)

  • Compare patient cohort characteristics (demographics, disease stage, treatment history)

  • Assess tumor microenvironment factors (stromal content, immune infiltration)

  • Consider cancer evolutionary context (primary vs. metastatic, treatment-naïve vs. resistant)

Research has shown that genetic variations in GDF10 are associated with different breast cancer subtypes (ER-PR+ and ER-PR-), suggesting that GDF10's role may fundamentally differ across cancer subtypes . This underscores the importance of careful subtype stratification when interpreting expression data.

What analytical approaches best capture the dynamics of GDF10-induced signaling?

GDF10 signaling involves complex, dynamic interactions that require sophisticated analytical approaches:

  • Temporal Profiling Methods:

    • Time-Course Experiments: Multiple sampling points from minutes to hours

    • Pulse-Chase Approaches: For receptor turnover and signal duration

    • Mathematical Modeling: Ordinary differential equations (ODEs) to capture pathway dynamics

    • Single-Cell Time-Lapse Imaging: For heterogeneity in response timing

  • Multiparametric Analysis:

    • Multiplexed Phospho-Flow Cytometry: Simultaneous measurement of multiple phosphorylation events

    • Mass Cytometry (CyTOF): High-dimensional signaling analysis

    • Multiplex Western Blotting: Simultaneous detection of multiple pathway components

    • Phosphoproteomics: Global phosphorylation changes following GDF10 stimulation

  • Spatial Signaling Analysis:

    • Subcellular Fractionation: Biochemical separation of signaling compartments

    • High-Resolution Microscopy: Tracking protein translocation events

    • FRET Biosensors: Real-time visualization of protein-protein interactions

    • Proximity Ligation Assay: Detection of protein complexes in situ

  • Computational Analysis Approaches:

    • Principal Component Analysis: Dimension reduction for complex signaling datasets

    • Clustering Algorithms: Identification of signaling patterns

    • Network Inference: Reconstruction of signaling networks from experimental data

    • Partial Least Squares Regression: Relating signaling activities to biological outcomes

Key Parameters to Measure:

Signaling EventMeasurement TechniqueTemporal Window
Receptor ActivationPhospho-specific antibodies5-30 minutes
Smad2/3 PhosphorylationWestern blot, ELISA15-60 minutes
Smad Nuclear TranslocationImmunofluorescence, nuclear fractionation30-120 minutes
Transcriptional ChangesRNA-seq, qPCR1-24 hours
Protein Expression ChangesWestern blot, proteomics4-48 hours
Phenotypic EffectsCell-type specific functional assays24-72 hours

Research has shown that GDF10 signals through TGFβRI/II and activates Smad2/3, which then regulates transcription of target genes . This signaling cascade exhibits temporal dynamics that must be captured through appropriate time-course experiments and analytical methods.

How can researchers effectively analyze GDF10's differential effects across diverse cell populations?

GDF10 exerts different, sometimes opposing effects across cell types. Effective analysis of these differential effects requires:

  • Single-Cell Analysis Approaches:

    • Single-Cell RNA-Seq: Transcriptomic profiling at cellular resolution

    • Single-Cell Proteomics: Protein-level response heterogeneity

    • Mass Cytometry: High-parameter protein analysis in heterogeneous populations

    • Imaging Mass Cytometry: Spatial context of cellular responses

  • Cell Type-Specific Isolation Methods:

    • FACS Sorting: Based on cell surface markers

    • Laser Capture Microdissection: From tissue sections

    • Magnetic-Activated Cell Sorting: For bulk isolation of specific populations

    • Single-Cell Picking: For highly pure population analysis

  • Co-Culture Experimental Designs:

    • Direct Co-Culture Systems: For cell-cell contact effects

    • Transwell Systems: For paracrine signaling analysis

    • Conditioned Media Experiments: For secreted factor effects

    • Microfluidic Co-Culture: For precise microenvironmental control

  • Computational Deconvolution Methods:

    • Cell Type Deconvolution Algorithms: For bulk tissue analysis

    • Trajectory Inference: To map cellular state transitions

    • Network Analysis: To identify cell type-specific signaling modules

    • Differential Correlation Analysis: To identify context-specific interactions

Analytical Framework for Multi-Cell Type Analysis:

  • Establish GDF10 response in pure populations of each cell type

  • Investigate how response changes in co-culture conditions

  • Determine if effects are direct (cell-autonomous) or indirect (paracrine)

  • Identify cell type-specific receptors and signaling components

  • Map microenvironmental factors that modify cell type-specific responses

Research has demonstrated distinct effects of GDF10 in different contexts: in TNBC cells, it inhibits proliferation and invasion , while in neurons it promotes axonal sprouting and functional recovery . These differential effects likely reflect cell type-specific receptor expression patterns, downstream signaling components, and transcriptional landscapes.

What are the most promising therapeutic applications of GDF10 research?

GDF10 research has revealed several potential therapeutic applications that merit further investigation:

  • Cancer Therapeutics:

    • Rationale: GDF10 functions as a tumor suppressor in TNBC and other epithelial cancers

    • Approach: Restoration of GDF10 expression or function in tumors where it is downregulated

    • Methods: Gene therapy, recombinant protein delivery, or small molecules that mimic GDF10 activity

    • Target Populations: Patients with TNBC and other cancers showing GDF10 downregulation

    • Research Priority: Developing delivery systems that can effectively restore GDF10 function in tumor cells

  • Neurological Recovery:

    • Rationale: GDF10 promotes axonal sprouting and functional recovery after stroke

    • Approach: Administration of GDF10 or activators of its pathway during recovery phase

    • Methods: Direct protein delivery, viral vector expression, or small molecule agonists

    • Target Populations: Stroke patients in recovery phase

    • Research Priority: Optimizing delivery timing and method for maximum functional recovery

  • Diagnostic and Prognostic Markers:

    • Rationale: GDF10 expression correlates with disease stage and severity in TNBC

    • Approach: Development of GDF10-based biomarkers for cancer diagnosis and prognosis

    • Methods: Tissue or liquid biopsy assays measuring GDF10 expression or its regulated genes

    • Target Populations: Cancer patients for risk stratification

    • Research Priority: Validation in large, diverse patient cohorts

  • Tissue Engineering and Regenerative Medicine:

    • Rationale: GDF10's role in cell differentiation and axonal growth

    • Approach: Incorporation into tissue engineering scaffolds or regenerative protocols

    • Methods: Controlled release systems, functionalized biomaterials

    • Target Applications: Neural tissue engineering, wound healing

    • Research Priority: Optimizing dosage and spatiotemporal presentation

Research Challenges and Opportunities:

Therapeutic AreaCurrent StatusKey ChallengesEnabling Technologies
Cancer TherapyPreclinicalTumor-specific deliveryNanoparticle delivery, CAR-T cells
Stroke RecoveryAnimal modelsBlood-brain barrier penetrationAAV vectors, focused ultrasound
DiagnosticsExploratoryStandardization, specificityDigital PCR, multiplexed assays
Regenerative MedicineEarly researchControlled presentationHydrogels, 3D bioprinting

Research suggests that for TNBC, "restoring GDF10 expression arises as an exciting and novel potential intervention to treat TNBC" , highlighting the therapeutic potential in this area. Similarly, the finding that GDF10 is "a stroke-induced signal for axonal sprouting and functional recovery" opens promising avenues for neurological rehabilitation.

What unresolved questions about GDF10 regulation should future research address?

Despite significant advances, several key questions about GDF10 regulation remain unresolved:

  • Transcriptional and Epigenetic Regulation:

    • What transcription factors directly regulate GDF10 expression?

    • How does DNA methylation status affect GDF10 expression across different tissues?

    • Are there tissue-specific enhancers or silencers that control GDF10 expression?

    • How do chromatin modifications influence GDF10 accessibility?

  • Post-Transcriptional Regulation:

    • What microRNAs regulate GDF10 mRNA stability and translation?

    • Are there RNA-binding proteins that affect GDF10 mRNA processing or localization?

    • How is GDF10 mRNA stability regulated in different cellular contexts?

    • What alternative splicing events affect GDF10 function?

  • Post-Translational Modifications:

    • What proteolytic processing is required for GDF10 maturation?

    • How do glycosylation patterns affect GDF10 secretion and function?

    • Are there other post-translational modifications that regulate GDF10 activity?

    • How is GDF10 protein stability and turnover regulated?

  • Receptor Interaction Specificity:

    • What determines the specificity of GDF10 for different TGF-β receptor combinations?

    • Are there co-receptors that modify GDF10 signaling in a tissue-specific manner?

    • How does receptor availability affect GDF10 signaling outcomes?

    • What is the structural basis for GDF10-receptor interactions?

  • Feedback and Cross-talk Mechanisms:

    • How does GDF10 signaling interact with other TGF-β family members?

    • What feedback mechanisms regulate GDF10 signaling intensity and duration?

    • How does cross-talk with other pathways (e.g., Wnt, Notch) modify GDF10 effects?

    • Are there inhibitory proteins that specifically target GDF10 signaling?

Research Prioritization Framework:

Question CategoryExperimental ApproachesExpected Impact
Epigenetic RegulationATAC-seq, ChIP-seq, methylation analysisIdentify therapeutic targets for restoring expression
miRNA RegulationmiRNA screening, 3'UTR reporter assaysNew regulatory mechanisms and therapeutic targets
Receptor SpecificityProtein binding assays, structural studiesBetter targeting of signaling pathway
Pathway Cross-talkMultiplexed signaling analysis, genetic screensUnderstanding context-specific outcomes
Feedback MechanismsTime-course studies, mathematical modelingImproved pathway manipulation strategies

Research has identified that "EMT transcription factors are involved in the regulation of GDF10" , but the specific mechanisms require further investigation. Additionally, while GDF10 has been shown to signal through TGFβR1/2/3 receptors , the factors determining receptor specificity across different tissues remain largely unknown.

How might integrated multi-omics approaches advance our understanding of GDF10 biology?

Multi-omics integration offers powerful approaches to comprehensively understand GDF10 biology:

  • Genomics-Transcriptomics Integration:

    • Approach: Correlation of genetic variants (SNPs, CNVs) with GDF10 expression

    • Methodologies: eQTL analysis, allele-specific expression

    • Research Question: How do genetic variations impact GDF10 expression and function?

    • Example Application: Understanding why "genetic variations in GDF10 were associated with ER-PR+ and ER-PR- breast cancer subtypes"

  • Transcriptomics-Proteomics Correlation:

    • Approach: Parallel analysis of mRNA and protein expression across conditions

    • Methodologies: RNA-seq paired with mass spectrometry

    • Research Question: How does post-transcriptional regulation affect GDF10 protein levels?

    • Example Application: Identifying discrepancies between transcript and protein levels in cancer progression

  • Epigenomics-Transcriptomics Analysis:

    • Approach: Correlation of DNA methylation, histone modifications with expression

    • Methodologies: WGBS, ChIP-seq, ATAC-seq integrated with RNA-seq

    • Research Question: How do epigenetic mechanisms regulate GDF10 expression?

    • Example Application: Mapping the epigenetic landscape at the GDF10 locus across tissue types

  • Metabolomics-Signaling Pathway Integration:

    • Approach: Analysis of metabolic changes induced by GDF10 signaling

    • Methodologies: Mass spectrometry-based metabolomics with signaling assays

    • Research Question: How does GDF10 signaling alter cellular metabolism?

    • Example Application: Understanding metabolic changes during GDF10-induced G0/G1 arrest

  • Spatial Multi-omics:

    • Approach: Spatial resolution of gene expression, proteins, and signaling

    • Methodologies: Spatial transcriptomics, imaging mass cytometry

    • Research Question: How does GDF10 signaling vary across tissue microenvironments?

    • Example Application: Mapping GDF10 expression and receptor distribution in tumor microenvironments

Integrative Analysis Framework:

Data IntegrationComputational MethodsBiological Insights
Genome-TranscriptomeeQTL mapping, mediation analysisGenetic determinants of expression
Proteome-PhosphoproteomeKinase activity inference, network analysisSignaling pathway activation
Multi-omics ClusteringSimilarity network fusion, multi-view clusteringDisease subtypes with distinct GDF10 biology
Causal Network InferenceBayesian networks, structural equation modelingCausal mechanisms in GDF10 regulation
Time-course Multi-omicsDynamic network models, trajectory inferenceTemporal dynamics of GDF10 responses

Research using RNA-seq has already identified differentially expressed genes (DEGs) between clinical TNBC specimens and controls, leading to the discovery of GDF10 downregulation . Further RNA-seq analysis from peri-infarct cortical neurons has shown that GDF10 downregulates PTEN and upregulates PI3 kinase signaling . These successful applications demonstrate the power of omics approaches, which could be further enhanced through multi-omics integration.

Product Science Overview

Gene and Protein Structure

The GDF10 gene is located on chromosome 10 in humans . The protein encoded by this gene is a secreted ligand that binds to TGF-β receptors, leading to the recruitment and activation of SMAD family transcription factors . These transcription factors regulate gene expression and play crucial roles in various biological processes.

Biological Functions

GDF10 is closely related to Bone Morphogenetic Protein 3 (BMP3) and is involved in several key biological processes :

  • Skeletal Morphogenesis: GDF10 plays a significant role in the development and formation of bones and skeletal structures .
  • Head Formation: It is also implicated in the formation of head structures during embryonic development .
  • Regulation of Apoptosis: GDF10 is involved in the regulation of programmed cell death, which is essential for maintaining cellular homeostasis .
  • SMAD Protein Signal Transduction: It positively regulates the phosphorylation of SMAD proteins, which are critical mediators of TGF-β signaling pathways .
Expression Patterns

In humans, GDF10 mRNA is found in various tissues, including the cochlea and lung of fetuses, and in the testis, retina, pineal gland, and other neural tissues of adults . In mice, GDF10 mRNA is abundant in the brain, inner ear, uterus, prostate, neural tissues, blood vessels, and adipose tissue .

Recombinant Production

Recombinant GDF10 is produced using various expression systems, including bacterial and mammalian cells . The production of human recombinant growth factors, such as GDF10, involves the use of fusion partners to enhance the yield and solubility of the protein . These recombinant proteins are used in research and therapeutic applications due to their ability to regulate cellular processes such as growth, differentiation, and proliferation .

Applications

GDF10 has potential applications in regenerative medicine and tissue engineering due to its role in bone and skeletal development . It is also studied for its involvement in neural development and its potential therapeutic effects in neurodegenerative diseases .

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 2024 Thebiotek. All Rights Reserved.