Recombinant Human Growth/differentiation factor 7 (GDF7)(Active)

Shipped with Ice Packs
In Stock

Description

Molecular Structure and Production

Production and Formulation

  • Recombinant GDF7 is synthesized as a full-length mature protein without glycosylation.

  • Lyophilized or liquid formulations are available, with reconstitution typically in 4 mM HCl .

  • Carrier-free versions are recommended for applications where BSA interference is a concern .

Tendon and Ligament Repair

GDF7 is well-documented for its tenogenic potential:

  • Stem Cell Differentiation: Induces adipose-derived stem cells and mesenchymal progenitors toward tenogenic lineages via Smad1/5/8 signaling .

  • Tissue Engineering: Enhances tendon matrix formation in equine mesenchymal stem cells and human tendon-derived progenitors .

Liver Regeneration

Emerging evidence highlights GDF7’s role in hepatic repair:

  • Fibrosis Modulation: Elevated in fibrotic liver tissues, primarily expressed in hepatocytes and cholangiocytes .

  • Progenitor Cell Activation: Promotes liver organoid growth and expression of progenitor markers (LGR5, AXIN2) .

  • Paracrine Effects: Activated hepatic stellate cells (HSCs) induce GDF7 expression in co-cultured liver organoids, suggesting cross-talk between stromal and parenchymal cells .

Bone and Neuronal Development

  • Osteoblast Regulation: Stimulates alkaline phosphatase production in ATDC5 chondrogenic cells (ED50: 0.25–1.25 μg/mL) .

  • Neuronal Guidance: Required for commissural axon growth in the central nervous system .

In Vitro Assays

Model SystemObserved EffectKey FindingsSource
ATDC5 CellsAlkaline Phosphatase InductionED50: 0.25–1.25 μg/mL
LX-2 Hepatic Stellate CellsNo Direct ActivationGDF7 fails to activate HSCs in isolation
Human Liver OrganoidsProgenitor Cell ExpansionUpregulation of LGR5 and AXIN2

Disease Models

  • Sepsis-Induced Acute Lung Injury (ALI):

    • Mechanism: Activates AMPK via STING pathway inhibition, reducing inflammation and oxidative stress .

    • Outcome: Prevents pulmonary injury in LPS-challenged mice .

Signaling Mechanisms

GDF7 exerts its effects through heterodimeric receptor complexes (BMPR-IB and BMPR-II/ActRII), leading to Smad1/5/8 phosphorylation . Recent studies identify secondary pathways:

  • STING/AMPK Pathway: Mediates anti-inflammatory and antioxidant effects in sepsis .

  • Smad1/5/8 Pathway: Drives tenogenic differentiation in mesenchymal stem cells .

Therapeutic Potential

ApplicationRationaleSupporting EvidenceSource
Tendon/Ligament RepairEnhances stem cell tenogenesisHuman and equine stem cell studies
Liver FibrosisPromotes progenitor cell expansionCo-culture models of HSCs and organoids
Acute Lung InjuryRegulates STING/AMPK pathwayLPS-induced ALI mouse models

Challenges and Limitations

  • Cell-Specific Responsiveness: HSCs (e.g., LX-2) show no direct response to GDF7, requiring paracrine signaling .

  • Retracted Studies: Some findings (e.g., sepsis-ALI) require validation due to retraction notices .

Product Specs

Buffer
Lyophilized from a 0.2 µm filtered concentrated solution in 30% Acetonitrile and 0.1% TFA.
Form
Liquid or Lyophilized powder
Note: We prioritize shipping the format currently in stock. However, if you have a specific format requirement, please indicate it when placing your order, and we will fulfill your request.
Lead Time
5-10 business days
Shelf Life
The shelf life is influenced by several factors including storage conditions, buffer components, temperature, and the protein's inherent stability.
Generally, liquid form has a shelf life of 6 months at -20°C/-80°C. The shelf life of lyophilized form is 12 months at -20°C/-80°C.
Storage Condition
Upon receipt, store at -20°C/-80°C. For multiple use, aliquoting is recommended. Avoid repeated freeze-thaw cycles.
Tag Info
Tag-Free
Synonyms
bmp12; bone morphogenetic protein 12; GDF-7; Gdf7; GDF7_HUMAN; growth differentiation factor 7; Growth/differentiation factor 7
Datasheet & Coa
Please contact us to get it.
Expression Region
322-450aa
Mol. Weight
14.0 kDa
Protein Length
Full Length of Mature Protein
Purity
> 95 % as determined by SDS-PAGE
Research Area
Signal Transduction
Source
E.Coli
Species
Homo sapiens (Human)
Target Names
Uniprot No.

Target Background

Function
GDF7 may play an active role in the motor area of the primate neocortex.
Gene References Into Functions
  1. BMP-12 activates the tenogenic pathway in human adipose stem cells. PMID: 28214472
  2. Variants at GDF7 and TBX5 associated with Barrett's esophagus also increase the risk of esophageal adenocarcinoma. PMID: 26783083
  3. The closest protein-coding genes were respectively GDF7 (rs3072), encoding a ligand in the bone morphogenetic protein pathway, and TBX5 (rs2701108), encoding a transcription factor regulating esophageal and cardiac development. PMID: 25447851
  4. BMP12 induces tenogenic differentiation of adipose-derived stromal cells through the Smad1/5/8 pathway. PMID: 24155967
  5. Studies demonstrate that while tenogenic (BMP 12 and BMP 13) and osteogenic (BMP2) BMPs bind the same receptors with high affinity, they signal differently, resulting in distinct activation of osteogenic and tenogenic markers. PMID: 21702718
  6. GDF7 induces ligamentogenic differentiation in mesenchymal progenitors. PMID: 20334610
  7. GDF7 stimulates the expression of both chondrogenic and osteoblastic markers in pluripotent mesenchymal C3H10T1/2 cells. PMID: 20102312
  8. In mice, Gdf7 expression in roof plate cells is essential for accurate commissural axon growth. PMID: 12741987

Show More

Hide All

Database Links

HGNC: 4222

OMIM: 604651

KEGG: hsa:151449

STRING: 9606.ENSP00000272224

UniGene: Hs.447688

Protein Families
TGF-beta family
Subcellular Location
Secreted.

Q&A

What is the molecular structure of human GDF7?

Human GDF7 is synthesized as a large precursor protein consisting of an N-terminal 19 amino acid signal sequence, a 302 amino acid pro region, and a 129 amino acid C-terminal mature peptide. The mature human GDF7 protein corresponds to amino acids Thr322-Arg450 of the full-length precursor. Unlike mouse and rat GDF7, the mature human version lacks a glycine repeat sequence. The protein functions as a homodimer to elicit its biological activities . Sequence comparisons show that mature human GDF7 shares 85% and 88% amino acid sequence identity with mature GDF7 in mouse and rat, respectively .

What are the key differences between carrier-free and standard recombinant GDF7 preparations?

Carrier-free (CF) recombinant GDF7 lacks Bovine Serum Albumin (BSA), which is typically added as a carrier protein to enhance stability, increase shelf-life, and allow storage at more dilute concentrations. The CF version is formulated as a lyophilized preparation from a 0.2 μm filtered solution in HCl and should be reconstituted at 500 μg/mL in 4 mM HCl . CF formulations are specifically recommended for applications where BSA might interfere with experimental results, while BSA-containing preparations are better suited for cell/tissue culture applications or as ELISA standards .

How should recombinant human GDF7 be stored and handled to maintain optimal activity?

For optimal stability, recombinant human GDF7 should be:

  • Shipped at ambient temperature

  • Immediately stored according to manufacturer recommendations upon receipt

  • Stored in a manual defrost freezer

  • Protected from repeated freeze-thaw cycles which can compromise protein integrity

  • Reconstituted at 500 μg/mL in 4 mM HCl for the carrier-free version

What are the primary signaling mechanisms of GDF7?

GDF7 initiates signaling by mediating the formation of a heterodimeric receptor complex consisting of:

  • Type I receptor (BMPR-IB)

  • Type II receptor (BMPR-II or Activin RII)

These receptors function as serine/threonine kinases. Upon receptor activation, the signaling pathway results in the phosphorylation and activation of cytosolic Smad proteins, specifically Smad1, Smad5, and Smad8 . This creates a signaling cascade that ultimately regulates various cellular responses including differentiation, proliferation, and extracellular matrix production in target tissues.

How does GDF7 contribute to neural development and function?

GDF7 plays multiple roles in neural development:

  • It regulates neuronal differentiation in the central nervous system

  • It guides axon development and pathfinding

  • GDF7-lineage cells contribute to diverse neuronal and glial cell types in the cerebellum, including:

    • Glutamatergic granule neurons

    • Unipolar brush cells

    • Purkinje neurons

    • GABAergic interneurons

    • Bergmann glial cells

    • White matter astrocytes

Recent research has established the hindbrain roof plate as a novel source of diverse neural cell types in the cerebellum that can undergo oncogenic transformation when Sonic hedgehog signaling is deregulated .

What is the role of GDF7 in connective tissue development and repair?

GDF7 is critically involved in:

  • Tendon and ligament formation during development

  • Tendon and ligament repair following injury

  • Bone formation processes

  • Differentiation of mesenchymal stem cells toward tenogenic lineages

Several studies have demonstrated the ability of GDF7 to promote tenogenic differentiation of mesenchymal stromal cells, as evidenced by its application in equine models and human injured tendons .

How can researchers assess GDF7 bioactivity in vitro?

The bioactivity of recombinant human GDF7 can be functionally assessed by measuring alkaline phosphatase production in ATDC5 mouse chondrogenic cells. The typical effective dose (ED50) for this effect ranges from 0.25-1.25 μg/mL . This standardized bioassay provides a reliable method to confirm protein activity before proceeding with more specialized experimental applications.

What are optimal cell culture conditions for GDF7-induced tenogenic differentiation of mesenchymal stem cells?

For effective tenogenic induction of mesenchymal stem cells using GDF7:

  • Cell preparation:

    • Use early passage (P2-P4) mesenchymal stem cells from appropriate sources (bone marrow, adipose tissue, or injured tendons)

    • Seed cells at 60-70% confluence in appropriate culture medium

  • GDF7 treatment:

    • Apply recombinant human GDF7 at concentrations of 0.25-1.25 μg/mL

    • For equine models, optimal concentrations may vary

    • Consider the microenvironment effects on differentiation outcomes

  • Culture duration:

    • Monitor tenogenic marker expression at intervals (days 3, 7, 14, 21)

    • Assess both early (SCX, TNMD) and mature (COL1, DCN) tenogenic markers

Research indicates that glucose metabolism control is important in tenogenic differentiation, so careful monitoring of metabolic parameters may enhance experimental outcomes .

What methods can be used to study GDF7 function in sepsis-induced acute lung injury models?

To investigate GDF7 function in sepsis-induced acute lung injury (ALI) models:

  • In vivo model setup:

    • Administer recombinant mouse GDF7 protein subcutaneously

    • Induce ALI through intratracheal lipopolysaccharide (LPS) injection

    • Include appropriate control groups

  • Assessment parameters:

    • Measure inflammatory markers in bronchoalveolar lavage fluid and lung tissue

    • Evaluate oxidative stress markers (MDA, SOD, GSH)

    • Assess pulmonary function parameters

    • Perform histopathological analysis

  • Mechanistic investigation:

    • Analyze AMPK activation status

    • Evaluate STING expression levels

    • Consider using AMPK inhibitors to confirm mechanistic pathways

These methods have revealed that GDF7 prevents LPS-induced inflammatory response, oxidative stress, and ALI by regulating the STING/AMPK pathway .

How does GDF7 interact with other members of the TGF-β superfamily in regulatory networks?

While GDF7 functions primarily as a homodimer, its interaction with the broader TGF-β superfamily creates complex regulatory networks. Based on sequence similarity, GDF7 is categorized with GDF-5 and GDF-6 as a subgroup within the BMP family . Advanced research should consider:

  • Potential heterodimer formation between GDF7 and other BMP family members

  • Competitive binding to shared receptors (BMPR-IB, BMPR-II)

  • Cross-talk between GDF7-initiated signaling and other TGF-β pathways

  • Tissue-specific co-expression patterns that may influence functional outcomes

Research approaches might include co-immunoprecipitation studies, proximity ligation assays, and comprehensive transcriptomic analysis following combinatorial treatments with multiple TGF-β superfamily members.

What is the relationship between GDF7 expression and tumorigenesis in the cerebellum?

Recent research has uncovered that GDF7-lineage cells are susceptible to oncogenic transformation by deregulated Sonic hedgehog (Shh) signaling in the cerebellum . The specific findings include:

  • Tumorigenic potential:

    • GDF7-lineage cells expressing constitutively active SmoM2 develop tumors marked by YFP

    • These tumors show high levels of Shh signaling pathway activation

    • Target genes of the pathway are robustly expressed

  • Tumor characterization:

    • GDF7-mediated medulloblastomas display cerebellar granule neuron precursor features

    • They show similar molecular phenotypes to medulloblastomas in Patched1 LacZ/+ mice

    • The progenitors appear to be multipotent

These findings establish a novel connection between hindbrain roof plate-derived GDF7-lineage cells and susceptibility to medulloblastoma formation when Shh signaling is aberrantly activated.

What techniques are recommended for studying GDF7 autocrine signaling in liver fibrosis models?

Recent research has identified GDF7 autocrine signaling as important in hepatic progenitor cell expansion during liver fibrosis . To effectively study this phenomenon:

  • Isolation and culture methods:

    • Use lineage tracing techniques to identify GDF7-expressing cells

    • Establish primary cultures of hepatic progenitor cells

    • Implement conditional knockout or overexpression systems for GDF7

  • Signaling pathway analysis:

    • Evaluate canonical (Smad-dependent) and non-canonical pathways

    • Perform phospho-proteomics to identify downstream targets

    • Use specific inhibitors to dissect pathway components

  • In vivo verification:

    • Develop hepatic-specific GDF7 knockout or overexpression mouse models

    • Induce liver fibrosis using established protocols (CCl4, bile duct ligation)

    • Assess progenitor cell expansion and fibrosis progression

These approaches can help elucidate the precise mechanisms by which GDF7 autocrine signaling promotes hepatic progenitor cell expansion in the context of liver fibrosis .

What are common issues when working with recombinant GDF7 and how can they be addressed?

ChallengePotential CauseSolution
Loss of activityImproper storage or reconstitutionStrictly follow manufacturer guidelines; avoid repeated freeze-thaw cycles
Inconsistent resultsVariability in protein preparationUse the same lot number for complete experimental series; include appropriate positive controls
Poor solubilityIncorrect reconstitution bufferEnsure use of 4 mM HCl for carrier-free preparations; verify pH before use
Cellular toxicityExcessive concentrationsPerform dose-response assays (0.1-10 μg/mL) to determine optimal concentrations for your cell type
Lack of responseReceptor expression issuesVerify target cells express appropriate receptors (BMPR-IB, BMPR-II or Activin RII)

How should researchers interpret contradictory results between in vitro and in vivo GDF7 studies?

When facing contradictory results between in vitro and in vivo GDF7 studies, consider:

  • Contextual differences:

    • In vivo microenvironments contain multiple cell types and extracellular matrix components that may modify GDF7 activity

    • The presence of endogenous inhibitors or enhancers may alter responses in vivo

  • Concentration variations:

    • Local effective concentrations in tissues may differ significantly from in vitro applications

    • Pharmacokinetics and biodistribution affect actual exposure in various tissues

  • Methodological approaches:

    • Validate findings using multiple complementary techniques

    • Consider using ex vivo organ cultures as an intermediate model

    • Employ tissue-specific conditional knockout models for definitive answers

  • Receptor and co-receptor considerations:

    • Verify the presence and distribution of GDF7 receptors in target tissues

    • Assess potential competition with other ligands in the in vivo environment

What are the best practices for validating GDF7-initiated signaling pathways?

To robustly validate GDF7-initiated signaling pathways:

  • Receptor activation analysis:

    • Confirm receptor complex formation using co-immunoprecipitation

    • Employ proximity ligation assays to visualize receptor interactions

    • Use phospho-specific antibodies to detect activated receptors

  • Downstream signaling validation:

    • Monitor Smad1/5/8 phosphorylation with temporal resolution (5-60 minutes post-treatment)

    • Assess nuclear translocation of activated Smads using cellular fractionation or imaging

    • Perform chromatin immunoprecipitation to identify direct Smad target genes

  • Pathway specificity controls:

    • Use receptor kinase inhibitors as negative controls

    • Include receptor-neutralizing antibodies to confirm specificity

    • Employ siRNA knockdown of pathway components as validation

  • Cross-pathway analysis:

    • Investigate potential activation of non-Smad pathways (MAPK, PI3K/Akt)

    • Examine cross-talk with other signaling systems (Wnt, Notch, Hedgehog)

    • Consider genome-wide approaches (RNA-seq, phospho-proteomics) for comprehensive pathway mapping

What emerging technologies might enhance our understanding of GDF7 biology?

Several cutting-edge technologies hold promise for advancing GDF7 research:

  • CRISPR-based approaches:

    • Precise genome editing to create reporter lines for visualizing GDF7 expression

    • Base editing for studying specific mutations in GDF7 or its receptors

    • CRISPRa/CRISPRi systems for temporal control of GDF7 expression

  • Single-cell technologies:

    • Single-cell RNA-seq to identify cell populations responsive to GDF7

    • Single-cell ATAC-seq to understand chromatin accessibility changes

    • Spatial transcriptomics to map GDF7 signaling in intact tissues

  • Advanced protein engineering:

    • Development of GDF7 variants with enhanced stability or receptor specificity

    • Creation of optogenetic tools for spatiotemporal control of GDF7 signaling

    • Engineering bifunctional GDF7 molecules to target specific tissues

  • Organoid technologies:

    • Development of cerebellum organoids to study GDF7 in neural development

    • Tendon organoids for investigating GDF7's role in tenogenesis

    • Multi-cellular organoid systems to examine cell-cell interactions in GDF7 signaling

What are potential therapeutic applications of GDF7 that warrant further investigation?

Based on current knowledge, several therapeutic applications of GDF7 merit further exploration:

  • Regenerative medicine:

    • Tendon and ligament repair enhancement

    • Cartilage regeneration approaches

    • Bone healing acceleration in difficult fractures

  • Inflammatory conditions:

    • Sepsis-induced acute lung injury prevention and treatment

    • Exploration of anti-inflammatory effects in other organs

    • Investigation of STING/AMPK regulation in diverse inflammatory contexts

  • Neurological applications:

    • Neural stem cell manipulation for regenerative approaches

    • Axon regeneration following spinal cord injury

    • Potential neuroprotective effects in neurodegenerative conditions

  • Oncology considerations:

    • Better understanding of the dual nature of GDF7 in tumor suppression versus promotion

    • Development of targeted approaches based on GDF7-lineage tumors

    • Exploration of GDF7 as a biomarker for specific tumor subtypes

How might systems biology approaches contribute to understanding GDF7's role in developmental and disease contexts?

Systems biology approaches offer powerful frameworks for integrating multiple levels of GDF7 biology:

  • Network modeling:

    • Integration of transcriptomic, proteomic, and metabolomic data to build comprehensive GDF7 signaling networks

    • Identification of network motifs and feedback loops regulating GDF7 function

    • Prediction of system behaviors under perturbation conditions

  • Multi-scale modeling:

    • Linking molecular interactions to cellular behaviors and tissue-level outcomes

    • Incorporating temporal dynamics of GDF7 signaling during development

    • Modeling how altered GDF7 signaling contributes to disease states

  • Comparative systems approaches:

    • Cross-species analysis of GDF7 networks to identify conserved modules

    • Evolutionary analysis of GDF7 signaling across diverse tissue contexts

    • Identification of context-dependent vs. context-independent GDF7 functions

  • Clinical data integration:

    • Correlation of GDF7 pathway alterations with patient outcomes

    • Identification of biomarkers related to GDF7 pathway activation

    • Development of personalized medicine approaches based on GDF7 pathway status

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.