GDF11 Human, His

Growth and Differentiation factor 11 Human Recombinant, His Tag
Shipped with Ice Packs
In Stock

Description

GDF11 Human Recombinant produced in E.Coli is a single, non-glycosylated polypeptide chain containing 132 amino acids (299-407a.a) and having a molecular mass of 14.8kDa.
GDF11 is fused to a 23 amino acid His-tag at N-terminus & purified by proprietary chromatographic techniques.

Product Specs

Introduction
Growth Differentiation Factor 11 (GDF-11) is a protein belonging to the bone morphogenetic protein (BMP) family and the transforming growth factor beta (TGF-β) superfamily. It plays a crucial role in regulating muscle and nerve development. In adults, GDF-11 may contribute to heart health by promoting the regeneration of heart muscle cells.
Description
Recombinant human GDF11 protein, expressed in E. coli bacteria, is a single polypeptide chain without any sugar modifications. It consists of 132 amino acids (corresponding to positions 299-407a.a in the natural sequence) and has a molecular weight of 14.8 kilodaltons (kDa). For purification and detection purposes, a 23-amino acid His-tag is attached to the protein's N-terminus. The protein is purified using specialized chromatography techniques.
Physical Appearance
Clear and colorless liquid, free from any particles or contamination.
Formulation
The GDF11 protein is supplied as a solution with a concentration of 1 milligram per milliliter (mg/ml). The solution also contains 20 millimolar (mM) Tris-HCl buffer at pH 8.0 and 10% glycerol for stability.
Stability
For short-term storage (up to 2-4 weeks), keep the protein solution refrigerated at 4°C. For long-term storage, freeze the solution at -20°C. To prevent protein degradation during long-term storage, consider adding a carrier protein like albumin (HSA or BSA) at a concentration of 0.1%. Repeated freezing and thawing of the solution should be avoided.
Purity
The purity of the GDF11 protein is greater than 85%, as assessed by SDS-polyacrylamide gel electrophoresis (SDS-PAGE).
Synonyms
Growth Differentiation Factor 11, BMP11, Bone Morphogenetic Protein 11, BMP-11, GDF-11, Growth/Differentiation Factor 11, Growth/differentiation factor 11.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSNLGLDCD EHSSESRCCR YPLTVDFEAF GWDWIIAPKR YKANYCSGQC EYMFMQKYPH THLVQQANPR GSAGPCCTPT KMSPINMLYF NDKQQIIYGK IPGMVVDRCG CS.

Q&A

What is GDF11 and what are its primary biological functions?

GDF11 is a member of the TGF-β superfamily that controls anterior-posterior patterning by regulating the expression of Hox genes during development. It shares 89% amino acid sequence homology with GDF8 (myostatin), though their biological effects can be quite different . GDF11 functions include:

  • Determination of Hox gene expression domains and rostrocaudal identity in the caudal spinal cord

  • Regulation of mesodermal formation and neurogenesis during embryonic development

  • Inhibition of olfactory receptor neural progenitor proliferation

  • Control of progenitor cell competence to regulate retinal ganglionic cell development

  • Age-related tissue regeneration effects in multiple organs

  • Potential metabolic regulation in glucose homeostasis

GDF11's functions can vary significantly depending on cell lineage, tissue type, and developmental stage, making it a versatile but complex growth factor to study .

How is recombinant human GDF11 protein typically produced for research applications?

Recombinant human GDF11 is typically produced using E. coli expression systems. The active form contains the C-terminal domain spanning amino acids Asn299 to Ser407 of the full-length protein and is often tagged with a histidine (His) tag at the C-terminus to facilitate purification . The production process involves:

  • Cloning the DNA sequence encoding the mature domain (Asn299-Ser407) into an appropriate expression vector

  • Transforming E. coli with the recombinant plasmid

  • Inducing protein expression

  • Purifying the protein using metal affinity chromatography (taking advantage of the His-tag)

  • Quality control testing for purity (typically >98% by SDS-PAGE) and endotoxin levels

  • Lyophilization in buffer containing 20 mM sodium citrate and 0.2 M NaCl (pH 3.5)

How should recombinant GDF11 protein be reconstituted and stored for maximum stability?

For optimal stability and activity of recombinant GDF11 protein, follow these methodological recommendations:

  • Reconstitution: Dissolve the lyophilized protein in 4 mM HCl to a concentration not less than 200 μg/mL

  • Incubation: Allow the solution to stand at room temperature for at least 20 minutes to ensure complete dissolution

  • Aliquoting: Divide the reconstituted protein into small working aliquots to minimize freeze-thaw cycles

  • Storage: Store aliquots at -20°C or preferably -80°C for up to one month

  • Usage precautions: Avoid repeated freeze-thaw cycles and storage in frost-free freezers

  • Pre-use preparation: Gently spin the vial prior to opening

These methods help preserve the activity of the protein for downstream experimental applications.

How can researchers distinguish between the effects of GDF11 and GDF8 (myostatin) in experimental systems?

Despite sharing 89% amino acid sequence homology in their mature domains, GDF11 and GDF8 (myostatin) can have distinct and sometimes opposing biological effects . To differentiate between their effects:

  • Expression pattern analysis: GDF8 expression in humans is restricted primarily to cardiac and skeletal muscle, while GDF11 is more broadly expressed across tissues

  • Domain-specific antibodies: Use antibodies targeting the pro-domains, which share only 54% homology between GDF11 and GDF8

  • Receptor binding assays: GDF11 has stronger affinity for its receptors compared to GDF8 and is more dependent on direct receptor contacts

  • Functional assays:

    • Hemoglobin induction in K562 cells (ED50 < 4 ng/mL for GDF11)

    • Alkaline phosphatase production in ATDC5 cells (ED50 < 11 ng/mL for GDF11)

  • Genetic approaches: Use specific siRNA knockdown of each factor followed by rescue experiments with purified recombinant proteins

When designing experiments, researchers should consider these distinguishing characteristics to avoid misattribution of observed effects.

What are the contradictory findings regarding GDF11's role in aging and how can researchers address these contradictions?

The literature contains significant contradictions regarding GDF11's role in aging:

To address these contradictions, researchers should:

  • Use standardized protein preparations: Ensure consistent quality and activity of GDF11 recombinant proteins across studies

  • Control for cross-reactivity: Use specific detection methods that can distinguish between GDF11 and the highly homologous GDF8

  • Consider context-dependent effects:

    • Cell type specificity (stemness capacity of target cells)

    • Age and physiological state of experimental subjects

    • Dose-dependent responses

    • Route of administration

  • Implement comprehensive experimental designs:

    • Include both in vitro and in vivo models

    • Assess multiple physiological parameters

    • Perform dose-response studies

    • Include appropriate time-course analyses

  • Examine the relationship with physical activity: Recent studies show that physical fitness correlates with GDF11 levels in serum, suggesting exercise may influence GDF11 expression and function

What is the mechanistic basis for GDF11's differential effects across various cancer types?

GDF11 exhibits context-dependent effects in cancer, functioning as either a tumor suppressor or promoter depending on the cancer type . The mechanistic basis includes:

Cancer TypeGDF11 EffectMolecular MechanismRelevant Models
Hepatocellular CarcinomaTumor suppressiveTranscriptional repression of cyclins D1 and A; Overexpression of p27; Downregulation of stemness markers (Epcam, CD133, CD24); Induction of mesenchymal-to-epithelial transitionHuh7, Hep3B, SNU-182, Hepa1-6, HepG2 cell lines
Other CancersVariable (both suppressive and promotional)Dependent on stemness capacity of target cells; Activation of canonical SMAD and non-canonical MAPK pathwaysMultiple cell line models (see Table 1 in reference)

To investigate these differential effects, researchers should:

  • Assess GDF11 expression in paired tumor/normal tissue samples

  • Characterize the stemness profile of target cancer cells

  • Evaluate the activation status of both canonical (SMAD) and non-canonical (MAPK) signaling pathways

  • Determine the receptor expression profile (ALK4/ALK5/ALK7) in target cells

  • Consider the influence of the tumor microenvironment on GDF11 signaling

What are the validated bioassays for determining the activity of recombinant GDF11 protein?

Researchers can use the following validated bioassays to determine the biological activity of recombinant GDF11 protein:

  • Alkaline phosphatase (ALP) induction:

    • Cell system: ATDC5 chondrogenic cells

    • Readout: Quantification of alkaline phosphatase activity

    • Expected potency: ED50 < 11 ng/mL

    • Controls: Include known active GDF11 standards and negative controls

  • Hemoglobin expression assay:

    • Cell system: K562 erythroleukemia cells

    • Readout: Hemoglobin expression (spectrophotometric or flow cytometry detection)

    • Expected potency: ED50 < 4 ng/mL

    • Incubation time: Typically 3-5 days

  • SMAD signaling activation:

    • Cell systems: Various responsive cell lines

    • Readouts: Phosphorylated SMAD2/3 detection by Western blot or ELISA

    • Timeframe: Acute response (minutes to hours)

  • Cell proliferation inhibition:

    • Cell systems: Hepatocellular carcinoma cell lines (Huh7, Hep3B)

    • Readout: Decreased proliferation, colony formation, and spheroid development

    • Concentration: Typically effective at 50 ng/mL

    • Treatment duration: 72 hours for observable effects

How can researchers effectively deliver GDF11 in different experimental models?

Effective delivery of GDF11 varies based on the experimental model:

  • In vitro cell culture models:

    • Direct addition to culture medium (typically 10-100 ng/mL)

    • Pre-treatment with 4 mM HCl during reconstitution to ensure solubility

    • Addition of carrier proteins (0.1-0.5% BSA) to prevent protein loss due to adsorption

    • Treatment duration: 24-72 hours for most cellular responses

  • Ex vivo tissue models:

    • Organ culture: Direct addition to culture medium

    • Tissue explants: Embedding in matrices (collagen, Matrigel) containing GDF11

  • In vivo animal models:

    • Systemic delivery: Intravenous or intraperitoneal injection (typically 0.1-1 mg/kg)

    • Local delivery: Direct injection into target tissues

    • Sustained release: Using osmotic pumps or encapsulation technologies

    • Gene therapy approaches: Adeno-associated virus (AAV) vectors for GDF11 gene transfer have been used to alleviate obesity and hyperglycemia in high-fat diet models

  • Genetic models:

    • Conditional knockout/knockin systems

    • Inducible expression systems for temporal control

    • CRISPR-Cas9 genome editing for endogenous gene modification

What experimental controls are essential when studying GDF11 function in complex biological systems?

When studying GDF11 function, researchers should implement these essential controls:

  • Specificity controls:

    • GDF8 (myostatin) treatments to distinguish GDF11-specific effects

    • Blocking antibodies against GDF11

    • Receptor antagonists (targeting ALK4/5/7)

    • RNA interference to knock down endogenous GDF11 or its receptors

  • Activity controls:

    • Heat-inactivated GDF11 protein

    • Biological activity verification using validated bioassays (ALP induction or hemoglobin expression)

    • Phospho-SMAD2/3 detection to confirm pathway activation

  • Contextual controls:

    • Age-matched subjects for aging studies

    • Multiple cell lines of varying differentiation states

    • Tissue-specific controls considering GDF11's varied effects across tissues

    • Time-course experiments to capture both acute and chronic responses

  • Technical controls:

    • Carrier protein only (BSA) treatments

    • Buffer-only treatments matching reconstitution conditions

    • Testing multiple protein lots to rule out batch-specific effects

    • Endotoxin testing to ensure bacterial contaminants don't confound results

How should researchers interpret conflicting data regarding GDF11 levels during aging?

Researchers face contradictory findings regarding GDF11 levels during aging:

To properly interpret these conflicting data, researchers should:

  • Critically evaluate detection methods:

    • Antibody specificity (cross-reactivity with GDF8)

    • Assay sensitivity and dynamic range

    • Sample preparation methods (serum vs. plasma)

  • Consider cohort characteristics:

    • Age distribution and stratification

    • Health status and comorbidities

    • Physical activity levels (as fitness correlates with GDF11 levels)

    • Sex differences in GDF11 regulation

  • Examine tissue-specific expression:

    • Circulating vs. tissue-specific levels

    • Local vs. systemic effects

    • Cell type-specific expression patterns

  • Account for post-translational regulation:

    • Pro-domain processing by proteases like PCSK5

    • Active vs. latent forms in circulation

    • Binding proteins that may sequester GDF11

  • Implement meta-analysis approaches:

    • Systematic review of methodologies

    • Statistical pooling of consistent datasets

    • Identification of factors contributing to variability

What are the key experimental challenges in studying GDF11 signaling pathways?

Researchers face several challenges when investigating GDF11 signaling pathways:

  • Receptor promiscuity:

    • GDF11 can bind multiple type I receptors: ALK4, ALK5, and ALK7

    • Predominant signaling occurs through ALK4 and ALK5

    • Different receptor usage may explain tissue-specific effects

  • Pathway redundancy:

    • Activation of canonical SMAD and non-canonical MAPK pathways

    • Context-dependent signaling outcomes

    • Potential crosstalk with other TGF-β family members

  • Assay limitations:

    • Difficulty distinguishing between GDF11 and GDF8 signaling

    • Limited availability of specific inhibitors for pathway components

    • Technical challenges in measuring pathway components in vivo

  • Methodological solutions:

    • Use receptor-specific knockdown/knockout approaches

    • Implement phospho-specific antibodies to detect activated signaling components

    • Develop reporter systems with pathway-specific response elements

    • Apply systems biology approaches to model complex signaling networks

    • Utilize advanced proteomics to identify novel interacting partners

How can researchers reconcile the seemingly contradictory effects of GDF11 in cancer biology?

The contradictory effects of GDF11 in cancer biology (both tumor suppressive and tumor promoting) require careful experimental approaches:

  • Systematic characterization across cancer types:

    • Establish a database of GDF11 effects across multiple cancer types

    • Correlate effects with molecular subtypes and genetic backgrounds

    • Consider evidence from Table 1 in reference showing divergent effects

  • Cell state dependency analysis:

    • Determine if effects depend on differentiation state

    • Assess stemness characteristics of responsive cells

    • Evaluate epithelial-mesenchymal transition (EMT) status

  • Molecular mechanism investigations:

    • Compare transcriptional programs activated in different contexts

    • Assess pathway utilization (SMAD vs. non-SMAD)

    • Examine epigenetic regulation (including HDAC-mediated effects)

  • Microenvironmental factors:

    • Study GDF11 in the context of tumor-stroma interactions

    • Evaluate immune cell responses to GDF11 signaling

    • Consider hypoxia and nutrient availability as modulating factors

  • Translational approaches:

    • Correlate GDF11 expression with patient outcomes in different cancer types

    • Develop predictive biomarkers for GDF11 response

    • Consider GDF11 pathway components as therapeutic targets in specific contexts

What are the emerging applications of GDF11 in metabolic disease research?

Recent findings suggest promising applications for GDF11 in metabolic disease research:

  • Diabetes management:

    • GDF11 has been shown to improve the survival and morphology of β-cells

    • It improves glucose metabolism in both non-genetic and genetic mouse models of type 2 diabetes

    • Systematic replenishment of GDF11 shows therapeutic potential

  • Obesity interventions:

    • GDF11 gene transfer alleviates high-fat diet (HFD)-induced obesity

    • It can reduce hyperglycemia, insulin resistance, and fatty liver development

    • It triggers a calorie restriction-like phenotype without affecting appetite

  • Adipose tissue modulation:

    • GDF11 stimulates adiponectin secretion from white adipose tissue

    • Direct action on adipocytes has been demonstrated

    • It may restore insulin/IGF-1 signaling pathways

  • Methodological approaches for metabolic research:

    • Gene therapy delivery systems for GDF11

    • Metabolic phenotyping of GDF11-treated models

    • Combined interventions (GDF11 + exercise or dietary modifications)

How might the structural biology of GDF11 inform the development of more specific research tools?

Understanding GDF11's structure can guide the development of specific research tools:

  • Structure-guided antibody development:

    • The human GDF11 structure has been resolved in recent years

    • Antibodies targeting unique epitopes not shared with GDF8 can be designed

    • Conformation-specific antibodies distinguishing active vs. latent forms

  • Receptor binding interface modifications:

    • GDF11 depends more on direct receptor contacts than GDF8

    • Engineered variants with altered receptor specificity could help dissect signaling pathways

    • Structure-based design of receptor-specific GDF11 variants

  • Pro-domain engineering:

    • The pro-domains of GDF11 and GDF8 share only 54% homology

    • Modified pro-domains could alter activation kinetics for experimental purposes

    • Tools to study PCSK5-mediated processing specifically

  • Imaging probes:

    • Structure-guided design of fluorescent or radioactive probes

    • Development of activity-based probes for active GDF11 detection

    • FRET-based biosensors for real-time GDF11 activity monitoring

Product Science Overview

Structure and Production

GDF11 is a recombinant protein produced in Escherichia coli (E. coli). The recombinant form is a single, non-glycosylated polypeptide chain containing 132 amino acids, with a molecular mass of approximately 14.8 kDa. The protein is fused to a 23 amino acid His-tag at the N-terminus, which facilitates purification through chromatographic techniques .

Biological Function

GDF11 plays a crucial role in embryonic development and tissue homeostasis. It is involved in the regulation of:

  • Skeletal system development
  • Neural development
  • Cardiac hypertrophy reversal by promoting the rejuvenation of cardiomyocytes .

In adults, GDF11 has been shown to have a rejuvenating effect on various tissues, including skeletal muscle, pancreas, kidney, nervous system, and retina .

Mechanism of Action

GDF11 functions as a cytokine and binds to the Transforming Growth Factor Beta receptors. This binding initiates a signaling cascade that regulates gene expression and cellular responses. The protein’s sequence is highly conserved across species, including humans, mice, and rats .

Applications

The recombinant form of GDF11 is widely used in:

  • Cell culture studies
  • Differentiation studies
  • Functional assays

It is optimized for use in various experimental setups and has cross-reactivity with mouse and rat cells .

Storage and Stability

For optimal stability, GDF11 should be stored at 4°C if used within 2-4 weeks. For longer storage, it is recommended to keep the protein frozen at -20°C, with the addition of a carrier protein to prevent degradation. Avoiding multiple freeze-thaw cycles is crucial to maintain the protein’s integrity .

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.