GDF7 Human

Growth and Differentiation factor 7 Human Recombinant
Shipped with Ice Packs
In Stock

Description

GDF7 Human Recombinant (322-450) produced in E.Coli is a disulfide-linked homodimeric, non-glycosylated, polypeptide chain containing 129 amino acids and having a molecular mass of 28kDa.
The GDF-7 is purified by proprietary chromatographic techniques.

Product Specs

Introduction
Growth Differentiation Factor-7 (GDF-7), a protein belonging to the Bone Morphogenetic Protein (BMP) family within the Transforming Growth Factor-beta (TGF-b) superfamily, exerts its biological effects by binding to a heterodimeric receptor complex. This complex consists of a type 1 receptor (BMPR-IB) and a type II receptor (BMPR-II or Activin RII), both of which are serine/threonine kinase receptors. Upon GDF-7 binding, Smad proteins are phosphorylated and activated, initiating downstream signaling. GDF-7 is implicated in various physiological processes, including the formation and repair of tendons and ligaments, regulation of bone formation, differentiation of mesenchymal stem cells, differentiation of neuronal cells, and guidance of axon growth.
Description
Recombinant human GDF7, spanning amino acids 322 to 450, is produced in E. coli. This protein is a disulfide-linked homodimer, lacking glycosylation, and comprises a polypeptide chain of 129 amino acids. It has a molecular weight of 28 kDa. The purification of GDF-7 is achieved through proprietary chromatographic techniques.
Physical Appearance
The product appears as a white powder, obtained through sterile filtration and lyophilization (freeze-drying).
Formulation
Prior to lyophilization, the product is in a 0.2µm filtered solution of hydrochloric acid (HCl).
Solubility
To reconstitute the lyophilized GDF-7, it is recommended to dissolve it in sterile 18M-cm H2O at a concentration of at least 100µg/ml. This solution can be further diluted in other aqueous solutions as needed.
Stability
Lyophilized GDF7 remains stable at room temperature for up to 3 weeks, but for long-term storage, it should be kept desiccated at a temperature below -18°C. After reconstitution, GDF-7 should be stored at 4°C for a period of 2 to 7 days. For extended storage, freezing at -18°C is recommended. To enhance stability during long-term storage, it is advisable to add a carrier protein, such as 0.1% HSA or BSA. Repeated freeze-thaw cycles should be avoided.
Purity
The purity of the product, as determined by SDS-PAGE analysis, is greater than 95.0%.
Biological Activity
The biological activity of GDF-7 is measured by its ability to induce alkaline phosphatase production in mouse ATDC5 cells. The ED50, which represents the concentration required for half-maximal induction, is less than 1.25µg/ml.
Synonyms
Growth Differentiation Factor 7, GDF-7, Growth/Differentiation Factor 7, BMP12, GDF7.
Source
Escherichia Coli.
Amino Acid Sequence
TALAGTRTAQ GSGGGAGRGH GRRGRSRCSR KPLHVDFKEL GWDDWIIAPL DYEAYHCEGL CDFPLRSHLE PTNHAIIQTL LNSMAPDAAP ASCCVPARLS PISILYIDAA NNVVYKQYED MVVEACGCR.

Q&A

What is the basic structure of human GDF7 protein?

Human GDF7 (BMP-12) 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 spans from Thr322 to Arg450 . The functional protein operates as a homodimer and belongs to the BMP family within the TGF-beta superfamily. Structurally, human GDF7 shares approximately 85% amino acid sequence identity with mouse GDF7 and 88% with rat GDF7, though human GDF7 notably lacks a glycine repeat found in both mouse and rat orthologs .

What signaling pathways does GDF7 activate in human cells?

GDF7 initiates cellular signaling by promoting the formation of a heterodimeric receptor complex consisting of a type I receptor (primarily BMPR-IB) and a type II receptor (either BMPR-II or Activin RII). This interaction triggers a phosphorylation cascade that activates cytosolic Smad proteins, specifically Smad1, Smad5, and Smad8 . The activation of these Smad proteins leads to their nuclear translocation where they function as transcription factors regulating gene expression. Researchers investigating GDF7 signaling should consider performing phospho-Smad immunoblotting, reporter gene assays, or chromatin immunoprecipitation to characterize pathway activation in their specific cell systems.

What are the optimal conditions for working with recombinant human GDF7 in cell culture experiments?

When designing experiments with recombinant human GDF7, researchers should consider several critical parameters. The protein is typically provided as a lyophilized formulation that should be reconstituted at a concentration of 500 μg/mL in 4 mM HCl to maintain stability . For cellular assays, the effective concentration range typically falls between 0.25-1.25 μg/mL for inducing biological responses . Carrier-free formulations are recommended for applications where BSA might interfere with experimental outcomes. For storage, use a manual defrost freezer and avoid repeated freeze-thaw cycles to maintain protein activity. When establishing tenogenic differentiation protocols for mesenchymal stem cells, GDF7 treatment should be optimized based on cell type, as different progenitor populations may exhibit varied response thresholds to the protein.

How can researchers effectively assess GDF7-mediated tenogenic differentiation in vitro?

To evaluate GDF7-induced tenogenic differentiation, researchers should implement a multi-parameter assessment approach. This should include:

  • Gene expression analysis: Quantify expression of tendon-specific markers such as scleraxis, tenomodulin, and collagen type I using qRT-PCR

  • Protein expression verification: Perform immunoblotting or immunofluorescence for tendon-specific proteins

  • Extracellular matrix evaluation: Assess collagen deposition and alignment using techniques such as Sirius Red staining or second harmonic generation microscopy

  • Mechanical testing: For 3D culture systems, measure tensile properties of engineered constructs

Research has demonstrated that GDF7 effectively induces tenogenic differentiation in multiple cell types, including equine fetal mesenchymal stem cells and human injured tendon-derived progenitors . For optimal results, experiment with concentration gradients and temporal administration patterns, as cellular response can vary significantly based on these parameters.

How does GDF7 contribute to tendon and ligament development and repair?

GDF7 plays a critical role in tendon and ligament biology through multiple mechanisms. In development, GDF7 guides the differentiation of mesenchymal progenitors toward the tenogenic lineage by activating specific transcription factors essential for tendon formation. In regenerative contexts, GDF7 enhances the tenogenic potential of various stem cell populations, as demonstrated in studies using both equine and human mesenchymal stem cells .

Research has shown that GDF7's effectiveness in promoting tendon repair is influenced by the microenvironment, particularly the composition of the tendon extracellular matrix. A 2018 study revealed that the tenogenic induction properties of GDF7 are significantly altered when cells are cultured on tendon matrix compared to standard culture conditions . This context-dependency highlights the importance of considering substrate properties and three-dimensional culture conditions when designing GDF7-based therapeutic approaches for tendon injuries.

What is known about GDF7's role in neuronal development and central nervous system function?

GDF7 exhibits significant roles in neuronal development and central nervous system function that extend beyond its better-known involvement in connective tissues. GDF7 mRNA is expressed within the roof plate during critical developmental periods when commissural axons initiate ventrally-directed growth . This expression pattern indicates its involvement in axon guidance mechanisms.

Studies in GDF7-null mutant mice have demonstrated the critical importance of this protein in neurodevelopment, as these animals develop hydrocephalus with considerable variation in the location of the dilated ventricles . Furthermore, a recent genetic study in cats identified a 7 bp deletion in GDF7 (c.221_227delGCCGCGC) resulting in a frameshift mutation (p.Arg74Profs) that causes a truncated protein of only 89 amino acids compared to the wildtype 455 amino acids . This mutation was associated with forebrain commissural malformation, ventriculomegaly, and interhemispheric cysts, further confirming GDF7's critical role in mammalian brain development. Researchers interested in neurodevelopmental processes should consider GDF7 as an important regulatory factor in their experimental designs.

What genetic variations in human GDF7 have been associated with disease phenotypes?

Genetic variations in and near GDF7 have been associated with several clinical conditions. A significant association has been identified between polymorphisms near GDF7 and Barrett's esophagus, a condition that increases risk for esophageal adenocarcinoma . While the exact mechanism of how these variants influence disease susceptibility remains under investigation, the association suggests GDF7 may play a role in esophageal epithelial homeostasis or response to injury.

Additionally, by extrapolation from animal models, alterations in human GDF7 may potentially contribute to neurodevelopmental disorders, particularly those involving commissural formation and ventricular development. The feline study demonstrating that a frameshift mutation in GDF7 leads to brain malformations resembling mild holoprosencephaly provides compelling evidence for investigating GDF7 variations in human neurodevelopmental disorders . Researchers should consider GDF7 as a candidate gene when studying human congenital brain malformations, particularly those involving forebrain commissural development.

How do GDF7 genetic variants affect protein function and downstream signaling?

The functional consequences of GDF7 genetic variants can be manifold, depending on the location and nature of the mutation. The 7 bp deletion in GDF7 identified in cats (c.221_227delGCCGCGC [p.Arg74Profs]) produces a severely truncated protein of only 89 amino acids compared to the 455 amino acids in the wildtype protein . This truncation likely eliminates the mature signaling domain, resulting in complete loss of function.

For human variants, functional studies should assess:

  • Protein expression and secretion (using Western blotting of cell lysates and conditioned media)

  • Dimerization capacity (through non-reducing SDS-PAGE)

  • Receptor binding (via surface plasmon resonance or co-immunoprecipitation)

  • Smad signaling activation (through phospho-Smad immunoblotting or reporter assays)

  • Target gene induction (via qRT-PCR or RNA-seq analysis)

Researchers investigating GDF7 variants should integrate computational prediction tools with experimental validation to thoroughly characterize their impact on protein function and downstream pathways.

How can GDF7 be utilized in tissue engineering approaches for tendon and ligament repair?

GDF7 presents significant potential for tissue engineering applications focused on tendon and ligament repair. Advanced approaches should consider:

  • Delivery systems: Controlled release platforms such as microspheres, hydrogels, or electrospun scaffolds can optimize GDF7 bioavailability

  • Combinatorial approaches: Co-delivery of GDF7 with other factors (e.g., PDGF or IGF-1) may enhance regenerative outcomes

  • Cell-based strategies: Pre-treatment of mesenchymal stem cells with GDF7 before implantation can improve their tenogenic potential

  • Biomechanical stimulation: Integration of mechanical loading regimens with GDF7 treatment often yields synergistic effects

Research has shown that GDF7's efficacy in promoting tenogenesis is influenced by the microenvironment, particularly the extracellular matrix composition. A study published in 2018 demonstrated that tenogenic induction of multipotent mesenchymal stromal cells by GDF7 is significantly altered when cells are cultured on tendon matrix versus standard conditions . This finding indicates that tissue engineering strategies should incorporate appropriate extracellular matrix cues alongside GDF7 delivery to maximize regenerative outcomes.

What are the challenges and contradictions in current GDF7 research literature?

Several challenges and contradictions exist in the current GDF7 research landscape:

  • Dosage discrepancies: Different studies report varying optimal concentrations for biological effects (0.25-1.25 μg/mL is commonly cited) , but concentration requirements appear to be cell type-dependent.

  • Context-dependent effects: GDF7's activity varies considerably based on cellular microenvironment, with evidence suggesting that tendon matrix components significantly modify GDF7-mediated differentiation responses . This contextual dependency complicates the translation of in vitro findings to in vivo applications.

  • Species differences: While human GDF7 shares high sequence identity with mouse (85%) and rat (88%) orthologs, human GDF7 lacks a glycine repeat found in rodent versions . The functional consequences of these differences remain incompletely characterized.

  • Pathway redundancy: The degree of functional overlap between GDF7 and related family members (particularly GDF5 and GDF6) remains unclear, with evidence suggesting both unique and redundant functions.

  • Neurodevelopmental roles: The recently identified association between GDF7 mutations and brain malformations in cats contrasts with the historical focus on GDF7's role in connective tissue. Reconciling these diverse biological functions presents a conceptual challenge that requires further investigation.

Researchers should approach these contradictions as opportunities for deeper investigation, considering factors such as experimental design differences, model system variations, and technical limitations when interpreting seemingly discrepant results.

What animal and cellular models are most appropriate for studying GDF7 function?

Several model systems have proven valuable for investigating GDF7 function:

Animal Models:

  • Mice: GDF7-null mice exhibit hydrocephalus with variable ventricle dilation , making them valuable for neurodevelopmental studies

  • Cats: A naturally occurring GDF7 mutation in cats causes brain malformations , providing an important large animal model

  • Equine: Horse models have been used to evaluate GDF7's effects on tendon healing and regeneration

Cellular Models:

  • Mesenchymal stem cells: Both human and equine MSCs respond to GDF7 treatment with tenogenic differentiation

  • Neural progenitor cells: Useful for studying GDF7's effects on neuronal differentiation and axon guidance

  • Tendon-derived progenitors: Human injured tendon-derived progenitors show glucose metabolism changes during GDF7-induced tenogenic differentiation

When selecting a model system, researchers should consider the specific aspect of GDF7 biology under investigation. For tendon/ligament studies, mesenchymal stem cells and tendon progenitors provide accessible in vitro models, while for neurodevelopmental research, neural progenitors and transgenic animal models are more appropriate.

How do research findings on GDF7 translate between model organisms and human applications?

The translation of GDF7 research findings between species requires careful consideration of both conserved and divergent aspects:

Conserved Elements:

  • The core BMP signaling pathway components (receptors and Smad proteins) are highly conserved across mammals

  • The tenogenic and osteogenic effects of GDF7 appear consistent across multiple species

  • The neurodevelopmental functions of GDF7 show similarities between cats and mice

Divergent Elements:

  • Human GDF7 lacks the glycine repeat present in rodent GDF7

  • The feline 7 bp deletion in GDF7 causes brain malformations , but equivalent human mutations have not yet been extensively characterized

  • Response thresholds to GDF7 treatment may vary between species and cell types

When translating animal model findings to human applications, researchers should validate key mechanisms in human cells whenever possible. Additionally, comparative analyses of GDF7 protein sequence, expression patterns, and signaling dynamics across species can help identify potential translational challenges. The 86.2% identity between human and horse GDF7 and 77.2% identity with mouse suggest generally conserved function but potential species-specific differences in detailed mechanisms or regulatory interactions.

What are the critical quality control parameters when working with recombinant GDF7 proteins?

Ensuring consistent quality of recombinant GDF7 is essential for reliable research outcomes. Critical quality control parameters include:

  • Purity assessment: >95% purity by SDS-PAGE is recommended for research applications

  • Endotoxin testing: Low endotoxin levels (<1.0 EU/μg) are crucial for cell culture applications

  • Activity verification: Bioactivity should be confirmed using established assays (e.g., induction of tenogenic markers in responsive cells)

  • Proper reconstitution: GDF7 should be reconstituted in 4 mM HCl at 500 μg/mL to maintain stability

  • Storage conditions: Use a manual defrost freezer and avoid repeated freeze-thaw cycles

For carrier-free formulations, which lack BSA as a stabilizing protein, special attention should be paid to protein concentration and storage conditions, as these preparations may have reduced stability. The carrier-free version is particularly recommended for applications where BSA might interfere with experimental outcomes .

How can researchers address inconsistent results when studying GDF7-mediated cellular responses?

Inconsistent results in GDF7 research often stem from several factors that can be systematically addressed:

  • Protein quality variability: Source recombinant GDF7 from reliable suppliers and maintain consistent handling protocols; always verify bioactivity before use

  • Cell heterogeneity: Establish well-characterized cell populations; consider clonal isolation for highly purified populations when appropriate

  • Context-dependent effects: Thoroughly document and standardize culture conditions, including:

    • Substrate composition (as GDF7 effects are modified by extracellular matrix)

    • Serum concentration and source

    • Cell density at treatment initiation

    • Duration of GDF7 exposure

  • Experimental readout sensitivity: Employ multiple complementary assays to evaluate GDF7 effects:

    • Use both gene and protein expression analyses

    • Include early (Smad phosphorylation), intermediate (gene expression), and late (protein expression, matrix deposition) readouts

  • Receptor expression verification: Confirm expression of appropriate GDF7 receptors (BMPR-IB, BMPR-II or Activin RII) in your cell system, as receptor levels can affect responsiveness

By systematically addressing these potential sources of variability, researchers can improve the consistency and reproducibility of their GDF7-related experiments.

What are the emerging applications of GDF7 in regenerative medicine beyond tendon and ligament repair?

While GDF7's role in tendon and ligament biology has been the primary focus of research, several emerging applications show promise:

  • Neurological applications: The identification of GDF7's role in brain development and axon guidance opens possibilities for neural tissue engineering and potential applications in neurodegenerative conditions

  • Induced pluripotent stem cell (iPSC) differentiation: GDF7 could be incorporated into differentiation protocols for generating specific cell lineages from iPSCs, particularly for connective tissue engineering

  • Combination therapies: Synergistic effects between GDF7 and other growth factors or mechanical stimulation could enhance tissue regeneration in multiple systems

  • Gene therapy approaches: Targeted delivery of GDF7 gene constructs could provide sustained, localized expression for chronic conditions

The discovery of GDF7's involvement in brain development through studies of the feline 7 bp deletion particularly highlights the need to explore its potential in neurological applications. This represents a significant expansion beyond its traditional association with musculoskeletal tissues.

What are the key unanswered questions in GDF7 research that warrant investigation?

Several critical knowledge gaps remain in our understanding of GDF7 biology:

  • Receptor specificity: The precise binding affinities of GDF7 for different type I and type II receptor combinations across various cell types need clarification

  • Transcriptional targets: Comprehensive identification of GDF7-regulated genes in different cellular contexts through techniques like RNA-seq and ChIP-seq would enhance our understanding of its diverse functions

  • Interaction with other signaling pathways: How GDF7 signaling integrates with other pathways (Wnt, Notch, etc.) remains incompletely characterized

  • Genetic variations in human populations: A systematic analysis of human GDF7 variants and their potential association with developmental disorders, particularly those affecting brain development, is warranted given the findings in animal models

  • Metabolic regulation: Recent findings suggest GDF7 affects glucose metabolism during tenogenic differentiation , but the mechanisms and broader metabolic impacts remain unclear

Addressing these questions will require integrative approaches combining structural biology, genomics, proteomics, and in vivo models to fully elucidate GDF7's multifaceted roles in development, homeostasis, and disease.

Product Science Overview

Structure and Synthesis

GDF7 is synthesized as a large precursor protein that consists of:

  • An N-terminal 19 amino acid signal sequence
  • A 302 amino acid pro-region
  • A 129 amino acid C-terminal mature peptide

The mature form of GDF7 is a disulfide-linked homodimer, which is essential for its biological activity .

Biological Functions

GDF7 plays a crucial role in several biological processes, including:

  • Tendon and Ligament Formation: GDF7 is involved in the formation and repair of tendons and ligaments .
  • Hepatic Progenitor Cell Expansion: Recent studies have shown that GDF7 promotes the expansion of hepatic progenitor cells in liver fibrosis, suggesting a potential pro-regenerative function .
  • Chondrogenesis: GDF7 has been shown to induce alkaline phosphatase production in chondrogenic cells, which is a marker of cartilage formation .
Mechanism of Action

GDF7 exerts its effects through a heterodimeric receptor complex composed of:

  • A type I serine/threonine kinase receptor (BMPR-IB)
  • A type II serine/threonine kinase receptor (BMPR-II or Activin RII)

Upon binding to its receptors, GDF7 activates the Smad signaling pathway, leading to the phosphorylation and activation of Smad proteins . These activated Smad proteins then translocate to the nucleus, where they regulate the expression of target genes involved in cell growth and differentiation.

Clinical Implications

The role of GDF7 in tissue repair and regeneration has significant clinical implications. For instance:

  • Liver Fibrosis: GDF7 has been shown to promote liver organoid growth and enhance the expression of progenitor cell markers in fibrotic liver tissue . This suggests that GDF7 could be utilized for the treatment of hepatic fibrosis caused by chronic liver diseases.
  • Tendon and Ligament Injuries: Due to its role in tendon and ligament formation, GDF7 could be a potential therapeutic agent for the treatment of tendon and ligament injuries .
Recombinant Human GDF7

Recombinant human GDF7 is produced using Escherichia coli expression systems and is available in carrier-free formulations . It is typically lyophilized and reconstituted in hydrochloric acid for use in various research and clinical applications .

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.