GDF5 Human, His

Growth differentiation factor 5 Human Recombinant, His Tag
Shipped with Ice Packs
In Stock

Description

GDF5 Human Recombinant produced in E.Coli is a single, non-glycosylated, polypeptide chain containing 141 amino acids (382-501 a.a.) and having a total molecular mass of 15.8 kDa.
GDF5 is fused to a 20 amino acid His Tag at N-terminus and purified by proprietary chromatographic techniques.

Product Specs

Introduction
Growth Differentiation Factor 5 (GDF-5), a protein belonging to the bone morphogenetic protein (BMP) family within the TGF-beta superfamily, plays a crucial role in regulating cell growth and differentiation during both embryonic and adult stages. Characterized by a polybasic proteolytic processing site that undergoes cleavage to produce a mature protein with seven conserved cysteine residues, GDF-5 is essential for skeletal development, as evidenced by the association of mutations in its gene with skeletal dysplasias like acromesomelic dysplasia, Hunter-Thompson type; brachydactyly, type C; and chondrodysplasia, Grebe type.
Description
Recombinant Human GDF5, expressed in E. coli, is a single, non-glycosylated polypeptide chain comprising 141 amino acids (382-501 a.a.). With a molecular weight of 15.8 kDa, this protein is fused to a 20 amino acid His Tag at its N-terminus and undergoes purification using proprietary chromatographic techniques.
Physical Appearance
Clear, colorless solution that has been sterilized by filtration.
Formulation
The GDF5 solution is formulated in a buffer containing 10mM sodium citrate (pH 3.5) and 10% glycerol.
Stability
For short-term storage (up to 2-4 weeks), the GDF5 solution can be stored at 4°C. For extended storage, it is recommended to freeze the solution at -20°C. To ensure optimal stability during long-term storage, the addition of a carrier protein (0.1% HSA or BSA) is advised. Repeated freezing and thawing of the solution should be avoided.
Purity
The purity of GDF5 is determined to be greater than 95.0% using SDS-PAGE analysis.
Synonyms
Cartilage-derived morphogenetic protein-1, CDMP-1, LAP4, SYNS2, GDF-5, Radotermin, CDMP1, GDF5, Growth differentiation factor 5, BMP-14.
Source
Escherichia Coli.
Amino Acid Sequence

MGSSHHHHHH SSGLVPRGSH MAPLATRQGK RPSKNLKARC SRKALHVNFK DMGWDDWIIA PLEYEAFHCE GLCEFPLRSH LEPTNHAVIQ TLMNSMDPES TPPTCCVPTR LSPISILFID SANNVVYKQY EDMVVESCGC R.

Q&A

What is GDF5 and what is its fundamental role in human joint development?

GDF5 (Growth and Differentiation Factor 5) is one of the earliest markers of joint formation and plays a crucial role in the normal development and maintenance of synovial joints in both humans and mice. The gene encodes a secreted growth factor that belongs to the TGF-β superfamily and is essential for multiple aspects of skeletogenesis . GDF5 functions in the formation of joints throughout the skeleton, including those in the digits, wrists, ankles, knees, and spine .

In humans, mutations in GDF5 can cause a spectrum of skeletal abnormalities including short stature, shortened digits, joint dislocations or fusions, and hip and knee joint dysplasia often presenting with osteoarthritis . Functional studies have demonstrated that GDF5 null mutations in mice result in shorter feet and limbs, missing joints in digits, wrists, and ankles, altered tendons, missing knee ligaments, and increased risk of osteoarthritis . These findings collectively establish GDF5 as a critical factor for proper joint formation and function.

How is the GDF5 gene structurally organized in the human genome?

The human GDF5 gene has a complex structural organization with regulatory elements distributed over more than a hundred kilobases of DNA. The gene contains both coding exons that directly encode the GDF5 protein and extensive regulatory regions both upstream (5') and downstream (3') of these coding sequences .

Research has identified multiple evolutionarily conserved non-coding regions flanking GDF5 that serve as enhancers, controlling expression in different subsets of developing joints . These regulatory elements are crucial for the precise spatial and temporal expression patterns of GDF5. The 5' untranslated region (5'UTR) contains important regulatory SNPs (rs143383 and rs143384) that have been associated with joint disorders, while the downstream region harbors a novel growth enhancer called GROW1, which is required for normal GDF5 expression at the ends of developing bones .

The modular organization of GDF5 regulatory elements explains how this single gene can control development across different joint types throughout the skeleton, with each enhancer directing expression in specific subsets of joints .

What are the key GDF5 variants associated with joint disorders and their functional consequences?

Several single nucleotide polymorphisms (SNPs) in the GDF5 gene have been associated with joint disorders. The most studied variants include:

  • rs143383 and rs143384: These SNPs located in the 5'UTR of GDF5 have been consistently associated with osteoarthritis risk, conferring a 1.3 to 1.8-fold increased risk in various populations including Japan and China . Furthermore, these variants are associated with congenital dislocation of the hip (CDH) in Caucasians, with the most significant association observed for rs143384. Individuals homozygous for the T allele of rs143384 (corresponding to the A allele in NCBI and 1000 Genomes datasets) have a higher risk of developing CDH compared to carriers of the other genotypes .

  • rs4911178: This common variant (G to A substitution) maps within the highly conserved GROW1B enhancer sequence and appears to reduce enhancer activity. The lower activity A variant is rare in Africa but common in Eurasia and has been identified in ancient DNA from Neandertals and Denisovans . This variant shows strong signals of positive selection in non-African populations.

Fine-mapping studies of developmental dysplasia of the hip (DDH) have indicated that rs143384 has a >99% likelihood of being the causal variant at the GDF5 locus, assuming only one variant as causal . This variant has high epigenomic scores due to its location in the 5'UTR of GDF5, high nucleotide sequence conservation, and overlap with DNase hypersensitivity and activating histone marks .

How has natural selection shaped GDF5 variation across human populations?

The worldwide distribution of GDF5 variants shows interesting patterns that reflect human evolutionary history. For the rs143384 SNP, the distribution mirrors the spread of modern humans out of Africa according to the Single Origin Model. African populations appear almost monomorphic for the ancestral G allele, while the derived A allele is more frequent in South Asian regions that were reached by the early expansion of Homo sapiens. There is a progressive increase in A allele frequency in areas of more recent colonization such as Europe, East Asia, and the Americas .

Despite this striking geographic distribution, studies using Tajima's test and Population Branch Statistic (PBS) analysis have not found strong evidence of selective pressure on the whole GDF5 gene or specifically on the rs143384 polymorphism . This suggests that the observed distribution is more likely the result of gene flow, genetic drift, and possible founder effects due to subsequent waves of migrations, rather than direct selection on this variant.

In contrast, the rs4911178 variant in the GROW1B enhancer shows multiple molecular signatures of positive selection during recent human evolution . The Composite of Multiple Signals (CMS) method, which combines multiple selection signatures, shows a selection peak in the region where GROW1B has been mapped, with rs4911178 being one of the top three highest scoring SNPs in the entire 130 kb region . This suggests that this non-coding change provides the most likely molecular basis for recent strong selection in Eurasian populations.

What is the relationship between GDF5 variants and height variation?

The GDF5 gene has been linked to height variation in humans, with common variants associated with reduced height . The GROW1 enhancer downstream of GDF5 has been identified as an important regulator of bone growth. When this enhancer is deleted in mice, it results in reduced limb length and decreased GDF5 expression levels .

The rs4911178 variant in the GROW1B enhancer, which shows signals of positive selection in non-African populations, affects enhancer activity and likely influences bone growth. The derived A allele reduces enhancer activity and is associated with shorter stature . The selection on this variant may reflect adaptation to northern environments, potentially explaining the high frequency of a GDF5 haplotype that also increases arthritis susceptibility in many human populations .

This example illustrates how selection on one trait (possibly height or other growth-related phenotypes) can lead to the fixation of variants that have pleiotropic effects, including increased susceptibility to joint disorders like osteoarthritis.

What approaches are effective for identifying and characterizing GDF5 regulatory elements?

Researchers have employed multiple complementary approaches to identify and characterize the complex regulatory architecture of GDF5:

  • Comparative Genomics: Using tools like VISTA and PIP maker to identify evolutionarily conserved non-coding regions across species (human, mouse, and chicken). These analyses identified multiple non-coding regions with ≥70% nucleotide sequence identity over regions of 300 bp or more .

  • Transgenic Reporter Assays: Cloning different conserved non-coding regions upstream of a minimal promoter and lacZ reporter, then testing their ability to drive specific expression patterns in mouse embryos. This approach revealed that different regulatory elements control expression in distinct anatomical locations .

  • BAC Transgenesis: Using bacterial artificial chromosomes (BACs) containing large genomic regions to test the activity of extended regulatory landscapes. This approach demonstrated that regulatory elements both upstream and downstream of GDF5 coding exons are required for normal expression patterns .

  • Functional Rescue Tests: Performing rescue experiments in mice to confirm that large flanking regions are required to restore normal joint formation and patterning in GDF5 mutants .

  • Targeted Deletion and Mutation Studies: Deleting specific enhancers (e.g., GROW1) to assess their function in vivo, showing their requirement for normal GDF5 expression and bone length .

  • Transcription Factor Binding Site Analysis: Identifying and mutating predicted transcription factor binding sites within enhancers to determine their requirement for expression in particular joints .

These approaches collectively provide a comprehensive toolkit for dissecting the regulatory architecture of complex developmental genes like GDF5.

What are the challenges and best practices in fine-mapping causative variants in the GDF5 locus?

Fine-mapping causative variants in the GDF5 locus presents several challenges, as demonstrated by genome-wide association studies (GWAS) of developmental dysplasia of the hip (DDH) and osteoarthritis:

  • Complex Linkage Disequilibrium Patterns: The GDF5 locus contains many variants in high linkage disequilibrium, making it difficult to pinpoint the specific causal variants. For example, in DDH studies, while rs143384 showed a >99% likelihood of being causal at the GDF5 locus, other loci contained hundreds of potential causal variants in their credible sets (483 and 618 variants for chromosomes 10 and 9 loci, respectively) .

  • Regulatory Complexity: The GDF5 regulatory landscape spans more than 100 kb, with multiple enhancers controlling expression in different joints. This complexity necessitates comprehensive analysis beyond just the coding and promoter regions.

Best practices for fine-mapping in this context include:

  • Statistical Fine-mapping: Using statistical methods that incorporate prior functional information to calculate posterior probabilities of causality for each variant. This approach successfully narrowed down the causal variant at the GDF5 locus to rs143384 in DDH studies .

  • Functional Annotation: Incorporating epigenomic and functional genomic data to prioritize variants. For example, rs143384 and rs143383 have high epigenomic scores due to their location in the 5'UTR, sequence conservation, and overlap with regulatory marks .

  • Gene-based Analysis: Using tools like MAGMA to identify genes containing multiple variants contributing to the phenotype. This approach identified GDF5, UQCC1, MMP24, RETSAT, and PDRG1 as significantly associated with DDH susceptibility .

  • Expression Quantitative Trait Loci (eQTL) Analysis: Examining whether variants regulate gene expression. Studies identified 14 variants as eQTLs that regulate various nearby genes .

  • Cross-species Validation: Using animal models to validate the function of potential causal variants in conserved regulatory elements .

How can researchers effectively study the interaction between GDF5 regulatory elements and transcription factors?

Understanding the interaction between GDF5 regulatory elements and transcription factors is crucial for elucidating the mechanisms controlling joint-specific expression. Several effective approaches include:

  • Identification of Transcription Factor Binding Sites: Using computational prediction tools to identify potential binding sites within GDF5 enhancers. Studies have shown that predicted transcription factor binding sites within GDF5 regulatory enhancers are required for expression in particular joints .

  • Site-directed Mutagenesis: Introducing mutations in predicted binding sites followed by reporter assays to test their functional significance. This approach can determine which sites are necessary for enhancer activity in specific joints.

  • Chromatin Immunoprecipitation (ChIP): Using ChIP to identify transcription factors that bind to GDF5 enhancers in different joint tissues and developmental stages.

  • DNA-Protein Interaction Assays: Employing electrophoretic mobility shift assays (EMSA) or DNA pulldown experiments to verify direct interactions between transcription factors and GDF5 regulatory sequences.

  • CRISPR-Cas9 Genome Editing: Using CRISPR-Cas9 to delete or mutate specific enhancers or transcription factor binding sites in cell lines or animal models to assess their functional significance.

  • Single-cell Approaches: Applying single-cell RNA-seq and ATAC-seq to identify cell-type-specific transcription factors that may regulate GDF5 expression in different joint compartments.

These approaches can help elucidate the transcriptional regulatory network controlling joint-specific GDF5 expression and potentially identify new therapeutic targets for joint disorders.

How does GDF5 variation in ancient hominins compare with modern human populations?

The study of GDF5 variation in ancient hominins provides fascinating insights into human evolution. The derived A allele of rs4911178 in the GROW1B enhancer, which reduces enhancer activity, is rare in Africa but common in Eurasia and has been found in ancient DNA from Neandertals and Denisovans . This suggests that this variant arose before the divergence of modern humans and Neandertals, approximately 500,000 years ago.

The presence of this variant in both Neandertals and modern Eurasians suggests several possible scenarios:

  • Ancient Shared Polymorphism: The variant may have been present in the common ancestor of Neandertals and modern humans, and subsequently maintained in both lineages.

  • Introgression: The variant could have been introduced into modern human populations through interbreeding with Neandertals after modern humans left Africa.

  • Convergent Evolution: The variant might have evolved independently in both lineages due to similar selective pressures, although this is less likely given the specific nature of the mutation.

The high frequency of this variant in non-African populations, combined with signatures of positive selection, suggests that it conferred some adaptive advantage in environments outside of Africa, possibly related to changes in body proportions or other skeletal features .

This ancient variant in the GROW1B enhancer is part of a GDF5 haplotype that also increases arthritis susceptibility in many human populations, illustrating how past selection on growth phenotypes can influence disease risk in contemporary populations .

What do population-specific patterns of GDF5 variation tell us about human adaptation?

Population-specific patterns of GDF5 variation reveal insights into human adaptation to different environments. The worldwide distribution of the rs143384 SNP shows a clinal pattern that follows human migration out of Africa, with African populations being almost monomorphic for the ancestral G allele, while the derived A allele increases in frequency along migration routes through South Asia, Europe, East Asia, and the Americas .

While Tajima's test and PBS analysis did not find strong evidence of selective pressure on rs143384, the distribution pattern suggests genetic drift and founder effects during human migrations played important roles in shaping its current distribution .

In contrast, the rs4911178 variant in the GROW1B enhancer shows clear signatures of positive selection in non-African populations. The derived A allele, which reduces enhancer activity and potentially affects bone growth, has been repeatedly selected in northern environments . This suggests adaptation to specific environmental conditions, possibly related to changes in body proportions that might be advantageous in colder climates.

These population-specific patterns highlight how different evolutionary forces—genetic drift, migration, and natural selection—have shaped human genetic variation at the GDF5 locus. Understanding these patterns provides context for interpreting the association between GDF5 variants and disease risk in different populations and may inform population-specific approaches to treating and preventing joint disorders.

How can understanding GDF5 regulatory architecture inform therapeutic strategies for joint disorders?

The detailed characterization of GDF5's complex regulatory architecture offers several promising avenues for developing therapeutic strategies for joint disorders:

  • Enhancer-targeted Therapies: The identification of joint-specific enhancers opens possibilities for targeted interventions that modulate GDF5 expression in specific joints affected by disorders. For instance, technologies like CRISPR-Cas9 could potentially be used to correct disease-associated variants in specific enhancers while preserving normal function in other tissues .

  • Allele-specific Modulation: For common variants like rs143383 and rs143384 that affect expression levels rather than protein structure, therapies that selectively upregulate expression from the low-expressing allele could help restore normal GDF5 levels in individuals carrying risk alleles .

  • Developmental Timing Considerations: The findings that different enhancers control GDF5 expression at different developmental stages suggests that therapeutic interventions might need to be timed appropriately to target the most relevant developmental windows .

  • Joint-specific Delivery Systems: Knowledge that different joints rely on distinct regulatory elements could inform the development of joint-specific delivery systems for GDF5-based biologics or gene therapies, potentially reducing off-target effects in other tissues.

  • Transcription Factor Modulation: Understanding the transcription factors that interact with GDF5 enhancers in specific joints could provide additional therapeutic targets, allowing indirect modulation of GDF5 expression through their regulatory networks .

Understanding the evolutionary history of GDF5 variants also has implications for therapeutic development, suggesting that approaches may need to be tailored to specific populations based on their GDF5 haplotype frequencies .

What methodological approaches can researchers use to study GDF5 function in human joint development and disease?

Researchers can employ a variety of methodological approaches to study GDF5 function in human joint development and disease:

  • Human iPSC-derived Models: Generating induced pluripotent stem cells (iPSCs) from individuals with different GDF5 genotypes and differentiating them into chondrocytes, osteoblasts, or joint organoids to study the effects of genetic variants on cellular phenotypes.

  • CRISPR-Cas9 Genome Editing: Creating isogenic cell lines that differ only in specific GDF5 variants to isolate their effects on gene expression and cellular function. This approach can also be used to introduce or correct specific mutations in model organisms.

  • Single-cell Genomics: Applying single-cell RNA-seq, ATAC-seq, and spatial transcriptomics to characterize cell type-specific effects of GDF5 variants in developing joints and in osteoarthritic tissues.

  • Functional Genomics Screens: Using CRISPR activation/interference screens to identify factors that modulate GDF5 expression in joint cells, potentially revealing new therapeutic targets.

  • Animal Models: Developing knock-in mouse models carrying human GDF5 variants to study their effects on joint development and disease susceptibility in vivo.

  • Biobank Integration: Leveraging large biobanks with genotype and phenotype data to perform phenome-wide association studies (PheWAS) of GDF5 variants, potentially uncovering new associations beyond known joint disorders.

  • 3D Chromatin Analysis: Using chromosome conformation capture techniques (Hi-C, 4C, etc.) to characterize long-range interactions between GDF5 enhancers and promoters in joint tissues, providing insights into the 3D regulatory architecture.

  • Longitudinal Imaging Studies: Conducting longitudinal imaging studies in individuals with different GDF5 genotypes to track joint development and disease progression over time.

These methodological approaches, used in combination, can provide comprehensive insights into how GDF5 variants influence joint development and contribute to disease, ultimately informing the development of precision medicine approaches for joint disorders.

Product Science Overview

Structure and Production

The human recombinant GDF5 with a His Tag is produced in E. coli as a single, non-glycosylated polypeptide chain containing 141 amino acids (residues 382-501) and has a total molecular mass of approximately 15.8 kDa . The His Tag, consisting of 20 amino acids, is fused to the N-terminus of the protein to facilitate purification using chromatographic techniques .

Function and Mechanism

GDF5 is primarily involved in the development and maintenance of bone and cartilage. It regulates the differentiation of chondrogenic tissue through two main pathways:

  1. Binding to BMPR1B: GDF5 binds with high affinity to the Bone Morphogenetic Protein Receptor Type 1B (BMPR1B), leading to the induction of SMAD1-SMAD5-SMAD8 complex phosphorylation and subsequent SMAD protein signaling transduction .
  2. Binding to BMPR1A: GDF5 also binds to BMPR1A with less affinity, contributing to the regulation of chondrogenic differentiation .
Clinical Significance

Mutations in the GDF5 gene are associated with several skeletal disorders, including:

  • Acromesomelic Dysplasia: A condition characterized by short stature and limb abnormalities.
  • Brachydactyly: A condition involving shortened fingers and toes.
  • Chondrodysplasia: A disorder affecting cartilage development.
  • Multiple Synostoses Syndrome: A condition involving the fusion of bones.
  • Proximal Symphalangism: A condition characterized by the fusion of the proximal interphalangeal joints.
  • Osteoarthritis Susceptibility: Variants in the GDF5 gene have been linked to an increased risk of developing osteoarthritis .
Applications

Recombinant GDF5 is used in various research and therapeutic applications, including:

  • Bone and Cartilage Repair: Due to its role in bone and cartilage formation, GDF5 is investigated for its potential in treating bone fractures and cartilage injuries.
  • Tissue Engineering: GDF5 is used in the development of tissue-engineered constructs for regenerative medicine.
  • Drug Development: GDF5 serves as a target for developing new drugs aimed at treating skeletal disorders and enhancing bone and cartilage regeneration.
Storage and Stability

The recombinant GDF5 protein is typically stored at -20°C for long-term storage and at 4°C if used within 2-4 weeks. It is recommended to add a carrier protein, such as 0.1% HSA or BSA, to prevent multiple freeze-thaw cycles .

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.