FGF5 Human

Fibroblast Growth Factor-5 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Key Features

CharacteristicDetails
Molecular Weight29.1 kDa
IsoformsFGF5 (268 aa) and FGF5s (123 aa)
ReceptorFibroblast Growth Factor Receptor 1 (FGFR1)
Expression SitesHair follicle outer root sheath, perifollicular macrophages

FGF5 binds to FGFR1 in dermal papilla cells, activating signaling pathways (e.g., PI3K-AKT, RAS-MAPK) that drive the catagen phase . Its expression peaks during the late anagen phase, serving as a key switch for terminating hair growth .

Role in Hair Cycling and Genetic Mutations

FGF5 is critical for regulating hair length across species. In humans, mutations in FGF5 lead to familial trichomegaly, characterized by excessively long eyelashes and body hair.

Genetic Evidence

StudyMutationPhenotypeHair Analysis
PNAS (2014) Homozygous FGF5 mutationsLong eyelashes, increased anagen-phase hairForearm hair: 65% anagen (vs. 17% controls)
Frontiers in PharmacologyFGF5 deficiencyExtended anagen, increased wool yield in goats-
Wikipedia Angora mutationExtended anagen in mice, cats, and dogs-

In organ culture models, exogenous FGF5 induces premature catagen entry, reducing hair growth duration . Conversely, FGF5 knockout or mutations extend the anagen phase, as seen in angora mice and humans with trichomegaly .

Therapeutic Applications and Inhibitors

Blocking FGF5 activity has emerged as a strategy to extend the anagen phase, promoting hair growth and reducing shedding.

FGF5 Inhibitors

Inhibitor TypeMechanismClinical Relevance
Sanguisorba officinalisBotanical extract, blocks FGF5-FGFR1Topical formulations for androgenetic alopecia
MonoterpenoidsCompetitive inhibition of FGF5 bindingEnhanced hair growth in preclinical models
RNA AptamersBind FGF5, prevent receptor interactionInhibit FGF5-induced cell proliferation

Clinical trials demonstrate that topical FGF5 inhibitors increase hair density and growth rate in both men and women . For example, a decapeptide antagonist of FGF5 restored hair growth in mice treated with exogenous FGF5 .

Research Findings and Future Directions

  • Hair Loss Treatment: CRISPR modification of goats to knockout FGF5 increased wool yield without adverse effects, suggesting similar applications in humans .

  • Genetic Insights: Genome-wide association studies identified FGF5 polymorphisms linked to early-onset androgenetic alopecia .

  • Mechanistic Studies: FGF5s isoform antagonizes FGF5 by binding FGFR1, offering a natural template for therapeutic design .

Product Specs

Introduction
Fibroblast Growth Factor-5 (FGF5), a member of the FGF family, is a mitogenic peptide. Recombinant human FGF5 (rhFGF5) exhibits mitogenic activity on Balb/3T3 fibroblasts and bovine heart endothelial cells in vitro. It also acts as a muscle-derived survival factor for cultured spinal motoneurons. In vivo, FGF5 is believed to have crucial roles in embryology and neurobiology. During development, FGF5 mRNA is initially observed in the embryoblast and later in the lateral somatic mesoderm and myotomes. In the mesoderm, it might contribute to angiogenesis. In the myotomes, particularly those cranial to the tail region, FGF5 may play a role in delaying the terminal differentiation of myoblasts during cell migration.
Description
Recombinant Human FGF5, produced in E. coli, is a single, non-glycosylated polypeptide chain with a molecular weight of 27.7 kDa. It comprises 252 amino acids. The purification of FGF-5 is carried out using proprietary chromatographic methods.
Physical Appearance
White, lyophilized powder, sterile-filtered.
Formulation
FGF-5 protein was lyophilized from a 0.2µm filtered solution containing 10mM sodium phosphate, 100mM sodium chloride, at pH 7.5.
Solubility
For reconstitution of lyophilized FGF5, sterile 18M-cm H₂O is recommended at a concentration not less than 100µg/ml. This solution can be further diluted into other aqueous solutions.
Stability
Lyophilized FGF5, while stable at room temperature for up to 3 weeks, should ideally be stored desiccated at a temperature below -18°C. After reconstitution, FGF-5 should be stored at 4°C for a period of 2-7 days. For long-term storage, it is recommended to store it below -18°C. The addition of a carrier protein (0.1% HSA or BSA) is recommended for long-term storage. Avoid repeated freeze-thaw cycles.
Purity
The purity of the protein is greater than 95.0% as determined by SDS-PAGE analysis.
Synonyms
Fibroblast Growth Factor 5, Heparin-Binding Growth Factor 5, Smag-82, HBGF-5, TCMGLY, FGF-5, FGF5.
Source
Escherichia Coli.
Amino Acid Sequence
MAWAHGEKRL APKGQPGPAA TDRNPRGSSS RQSSSSAMSS SSASSSPAAS LGSQGSGLEQ SSFQWSPSGR RTGSLYCRVG IGFHLQIYPD GKVNGSHEAN MLSVLEIFAV SQGIVGIRGV FSNKFLAMSK KGKLHASAKF TDDCKFRERF QENSYNTYAS AIHRTEKTGR EWYVALNKRG KAKRGCSPRV KPQHISTHFL PRFKQSEQPE LSFTVTVPEK KKPPSPIKSK IPLSAPRKNT NSVKYRLKFR FG.

Q&A

What is the molecular structure of human FGF5 and how does it compare to other FGF family members?

Human FGF5 is a 268-amino-acid protein with a molecular weight of approximately 29.1 kDa. The protein contains conserved cysteine residues that are characteristic of the FGF family, sharing 30-50% sequence homology with other FGF family members at the amino acid level . The human FGF5 cDNA encodes a protein with a putative 22 amino acid residue signal peptide . The functional region of human FGF5 spans from Glu23-Gly268, with notable mutations at positions Lys238Asn and Pro245Ser that may affect its biological activity .

When comparing across species, murine FGF5 shows 84% homology to the human protein at the amino acid sequence level, and importantly, human and murine FGF5 exhibit cross-species activity . This conservation suggests evolutionarily preserved functional significance of this growth factor.

What are the primary receptors for FGF5 and their signaling mechanisms?

FGF5 binds with high affinity to the IIIc isoforms of FGFR1 and FGFR2, which are preferentially expressed in mesenchymal lineages . The binding of FGF5 to these receptors requires the cofactor heparin sulfate, which is critical for receptor activation . Upon binding, the intracellular tyrosine kinase domains of the receptors are activated, triggering downstream signaling cascades.

The primary signaling pathways activated by FGF5 include:

  • MAPK signaling pathway - documented in multiple cancer types including pancreatic cancer, hepatocellular carcinoma, and osteosarcoma

  • NFAT signaling axis - particularly relevant in melanoma

These signaling mechanisms mediate various cellular responses including proliferation, differentiation, and migration, contributing to FGF5's roles in both normal development and pathological conditions.

What is the normal tissue expression pattern of FGF5 in humans?

FGF5 exhibits a specific tissue expression pattern in humans, with significant expression observed in certain tissues and cell types. While the search results don't provide a comprehensive tissue expression profile, they indicate that FGF5 plays important roles in multiple systems:

  • In the nervous system, FGF5 has been identified in neurons associated with the limbic system, notably in neurons of the olfactory bulb and pyramidal cells of the hippocampus .

  • During embryonic development, FGF5 mRNA is initially found in the embryoblast, followed by expression in the lateral somatic mesoderm and myotomes cranial to the tail region .

  • In adult tissues, FGF5 expression has been documented in various cell types, including fibroblasts and certain endothelial cells .

The Human Protein Atlas contains more comprehensive data on FGF5 tissue expression patterns, showing the subcellular distribution of the protein and RNA sequencing data from various tissue culture cell lines .

How does FGF5 specifically contribute to cell proliferation compared to other FGF family members?

FGF5 exhibits distinct effects on cell proliferation compared to other FGF family members, particularly FGF2. Studies using RNA aptamers have demonstrated the specificity of FGF5-induced cell proliferation. When cells were cultured with human FGF5 and human FGF2, the FGF5-specific RNA aptamers inhibited only FGF5-induced cell proliferation but had no effect on FGF2-induced proliferation .

The ED50 for FGF5's mitogenic effects on cells such as Balb/3T3 fibroblasts and bovine heart endothelial cells is approximately 2-10 ng/mL in the presence of 1 μg/mL of heparin . This defined dosage response helps researchers distinguish FGF5's specific proliferative effects from those of other growth factors.

The mechanistic differences between FGF5 and other family members likely stem from:

  • Unique receptor binding affinities and preferences

  • Differential activation of downstream signaling cascades

  • Tissue-specific expression patterns and microenvironmental contexts

What are the known interactions between FGF5 and other signaling pathways in human diseases?

FGF5 intersects with multiple signaling pathways that contribute to human diseases, particularly cancer. Three key pathway interactions have been identified:

  • Hedgehog (Hh) signaling pathway: In prostate cancer, increased FGF5 levels were observed when normal prostate fibroblasts (NPFs) were transduced with a constitutively active Smo:RFP construct to mimic aberrant Hh signaling . This interaction was also observed in triple-negative breast cancer, where a 60-fold upregulation of FGF5 mRNA in the stroma was linked to the Hh signaling pathway, contributing to cell plasticity, proliferation, and stemness .

  • Androgen receptor (AR) signaling: Evidence suggests potential associations between FGF5 expression and AR signaling, which may be particularly relevant in hormone-responsive cancers like prostate cancer .

  • Sex Determining Region Y-box 2 (SOX2) signaling: FGF5 expression has been linked to SOX2 signaling pathways, which regulate stem cell properties and may contribute to cancer progression .

These pathway interactions represent critical nodes for potential therapeutic intervention and highlight the complex role of FGF5 in disease pathogenesis.

What is the relationship between FGF5 genetic variants and hypertension?

The genetic variant rs1458038 near the FGF5 gene has been significantly associated with both systolic (SBP) and diastolic blood pressure (DBP) in Chinese adults from the China Health and Nutrition Survey (CHNS) . This association exhibits a notable genotype-by-BMI interaction, with the strongest variant effects observed in individuals with the highest BMI (P interaction = 0.0018 for SBP; P interaction = 0.049 for DBP) .

This finding was replicated in the Fangchenggang Area Male Health and Examination Survey (FAMHES), which confirmed that rs1458038 showed stronger effects on SBP and DBP among men with higher BMI . These results suggest that high BMI increases the effect of the blood pressure-increasing allele at rs1458038 near FGF5, highlighting the importance of considering obesity as a modifying factor in genetic studies of hypertension.

The mechanism by which FGF5 affects blood pressure may be related to its involvement in the metabolic syndrome. The study notes that "In addition to ANTXR2, FGF5 is also possibly involved in the metabolic syndrome" , suggesting complex interactions between FGF5, metabolism, and cardiovascular regulation.

What are the most effective methods for studying FGF5-receptor interactions in vitro?

Several methodological approaches have proven effective for studying FGF5-receptor interactions:

  • Surface plasmon resonance (SPR): This technique allows for precise measurement of binding affinities between FGF5 and its receptors. A study on RNA aptamers demonstrated that SPR could accurately determine the dissociation constant (Kd) of FGF5 binding. For example, the aptamer F5f1 was shown to bind to FGF5 with a Kd of 0.7 ± 0.2 nM, while the truncated aptamer F5f1_56 exhibited even higher affinity with a Kd of 0.118 ± 0.003 nM .

  • Cell proliferation assays: These assays can demonstrate functional receptor activation by measuring FGF5-induced cell proliferation. Researchers typically culture cells with human FGF5 (and comparators like FGF2) to assess proliferative responses . The ED50 for FGF5's mitogenic effects on cells such as Balb/3T3 fibroblasts is approximately 2-10 ng/mL in the presence of 1 μg/mL of heparin .

  • Competitive binding assays: These can be used to assess specificity of FGF5 binding to different FGFRs by comparing binding affinities to FGF1, FGF2, FGF4, FGF6, and FGFR1 .

When designing these experiments, researchers should consider using recombinant human FGF5 protein, specifically the E. coli-derived version spanning residues Glu23-Gly268 (with Lys238Asn and Pro245Ser mutations) or Leu26-Gly268 (with the same mutations) .

What experimental models are most appropriate for studying FGF5 functions in cancer progression?

Based on the search results, several experimental models have proven valuable for studying FGF5's role in cancer progression across different tumor types:

Cell Line Models:

  • Breast cancer: Triple-negative breast cancer (TNBC) patient-derived xenografts

  • Pancreatic cancer: COLO-357 cell line

  • Esophageal squamous cell carcinoma (ESCC): 11 ESCC cell lines

  • Hepatocellular carcinoma (HCC): 5 HCC cell lines

  • Glioblastoma multiforme (GBM): T98G, U373, MGC cell lines

  • Osteosarcoma: U2OS, SAOS, MG63 cell lines

  • Melanoma: 28 human melanoma cell lines

Animal Models:

  • HCC mouse model

  • Nude mouse orthotopic model for osteosarcoma

  • Murine xenograft models for melanoma

  • Biclonal xenograft tumors for prostate cancer

Patient Samples:

  • 12 pancreatic cancer patient samples

  • 117 ESCC samples

  • 192 HCC patient samples

  • Astrocytic glioma patient samples (grades I-III) and GBM

  • 15 OS patient samples

The choice of model should align with the specific research question. For instance, studies focusing on stromal-epithelial interactions in prostate cancer have utilized a model where stromal cells (SmoM2-NPFs) were recombined with non-tumorigenic human prostate epithelial cells (BPH-1) and grafted in vivo , demonstrating the importance of considering cellular microenvironment.

What techniques are available for specific inhibition of FGF5 in experimental settings?

Several approaches for specifically inhibiting FGF5 have been developed:

  • RNA aptamers: Systematic Evolution of Ligands by EXponential enrichment (SELEX) has been used to generate novel RNA aptamers with high affinity to human FGF5. These aptamers specifically inhibit FGF5-induced cell proliferation without affecting FGF2-induced proliferation . The aptamer F5f1 binds to FGF5 with high affinity (Kd = 0.7 ± 0.2 nM) and shows minimal binding to FGF1, FGF2, FGF4, FGF6, or FGFR1 . A truncated version, F5f1_56, exhibits even higher affinity (Kd = 0.118 ± 0.003 nM) .

  • Small molecule inhibitors: While not specifically detailed in the search results, the development of FGF5 inhibitors is mentioned as a potential therapeutic approach for FGF5-related diseases including prostate cancer and benign prostatic hyperplasia .

  • Genetic approaches: Techniques such as RNA interference (siRNA or shRNA) or CRISPR-Cas9 gene editing could be employed to downregulate FGF5 expression in experimental models, though these are not explicitly discussed in the search results.

When designing experiments to inhibit FGF5, researchers should consider the specificity of the inhibition method, as many FGF family members share structural similarities. The RNA aptamers described in the search results demonstrate high specificity to FGF5, making them promising tools for experimental studies .

What is the evidence for FGF5 as a therapeutic target in human cancers?

FGF5 has emerged as a promising therapeutic target in multiple cancer types, with evidence supporting its oncogenic roles across various malignancies:

Cancer typeFGF5 expressionMechanism of actionProposed role in tumorigenesisModel used
Breast Cancer60-fold upregulation of FGF5 mRNA in stromaHh signaling pathwayCell plasticity, proliferation, and stemnessTriple negative breast cancer (TNBC) patient derived xenograft
Pancreatic cancerOverexpression of FGF5 mRNAMAPK signaling pathwayProliferation12 patient samples; COLO-357 cell line
Esophageal squamous cell carcinoma (ESCC)Reduced FGF5 expressionUndefinedConfers resistance to definitive chemoradiotherapy117 ESCC samples; 11 ESCC cell lines
Hepatocellular carcinoma (HCC)Overexpression of FGF5MAPK signaling pathwayProliferation, metastasis192 HCC patient samples; 5 HCC cell lines; HCC mouse model
Glioblastoma multiforme (GBM)Overexpression of FGF5 mRNA and proteinUndefinedCell proliferation, viability, migratory capacity, angiogenesis, and malignancyAstrocytic glioma patient samples (grades I-III) and GBM; GBM cell lines
Osteosarcoma (OS)Overexpression of FGF5 mRNA and proteinMAPK signaling pathwayDedifferentiation, metastasis, increased tumor size and stage15 OS patient samples; cell lines; nude mouse model
Melanoma>50-fold upregulation of mRNAMAPK NFAT signaling axisClonogenicity, invasion, tumor growth, angiogenesis, and proliferation28 human melanoma cell lines; murine xenograft model

This evidence suggests that FGF5 inhibition could be a viable therapeutic strategy in multiple cancer types . The development of specific FGF5 inhibitors, such as the RNA aptamers described in the search results, represents a promising approach for targeted cancer therapy .

How can researchers evaluate the efficacy of FGF5 inhibitors in preclinical studies?

Researchers evaluating the efficacy of FGF5 inhibitors in preclinical studies should employ a multi-faceted approach incorporating various experimental models and endpoints:

  • In vitro efficacy assessment:

    • Cell proliferation assays: Measure the ability of FGF5 inhibitors to block FGF5-induced cell proliferation, as demonstrated with RNA aptamers .

    • Receptor binding assays: Use surface plasmon resonance to determine binding affinities and specificity of inhibitors to FGF5 versus other FGF family members .

    • Signaling pathway analysis: Assess the effects of FGF5 inhibitors on downstream signaling molecules in the MAPK and other relevant pathways .

  • Specificity evaluation:

    • Compare effects on FGF5-induced versus FGF2-induced (or other FGF members) cellular responses to confirm target specificity .

    • Test for cross-reactivity with other FGF family members (FGF1, FGF2, FGF4, FGF6) and receptors (FGFR1) .

  • In vivo efficacy models:

    • Select appropriate animal models based on the cancer type being studied (see table in section 4.1).

    • Evaluate tumor growth, metastasis, angiogenesis, and survival endpoints.

    • Consider using patient-derived xenograft models for higher clinical relevance .

  • Combination therapy assessment:

    • Evaluate FGF5 inhibitors in combination with standard-of-care therapies relevant to the specific cancer type.

    • For esophageal squamous cell carcinoma, consider combining with chemoradiotherapy given FGF5's role in therapy resistance .

When designing these studies, researchers should carefully consider dosing, administration routes, pharmacokinetics, and pharmacodynamics of the FGF5 inhibitors to ensure optimal efficacy assessment.

What is the relationship between FGF5 and cardiovascular disease risk?

The relationship between FGF5 and cardiovascular disease risk, particularly hypertension, has been investigated through genetic association studies. Key findings include:

  • The genetic variant rs1458038 near the FGF5 gene is significantly associated with both systolic blood pressure (SBP) and diastolic blood pressure (DBP) in Chinese adults .

  • This genetic association exhibits a notable genotype-by-BMI interaction, with the strongest effects of the variant observed in individuals with the highest BMI (P interaction = 0.0018 for SBP; P interaction = 0.049 for DBP) .

  • This finding was replicated in an independent cohort of Chinese men from the Fangchenggang Area Male Health and Examination Survey (FAMHES), confirming that rs1458038 showed stronger effects on blood pressure among men with higher BMI .

  • FGF5 is potentially involved in metabolic syndrome, which is a cluster of conditions that increase the risk of heart disease, stroke, and type 2 diabetes .

These findings highlight the complex interaction between genetic factors (FGF5 variants) and environmental factors (obesity as measured by BMI) in determining cardiovascular disease risk. This research suggests that prevention strategies targeting obesity may be particularly important for individuals carrying the blood pressure-increasing allele at rs1458038 near FGF5 .

What are the potential applications of FGF5 research beyond cancer and hair growth regulation?

While FGF5 is well-established as a regulator of hair growth and as an oncogenic factor in several cancers, emerging research suggests broader applications:

  • Cardiovascular disease management: The association between FGF5 genetic variants and blood pressure, particularly in individuals with high BMI, suggests potential applications in personalized cardiovascular disease prevention and management . Further research could explore whether FGF5 inhibition might have beneficial effects on blood pressure regulation in specific genetic backgrounds.

  • Neurobiology and neuroprotection: FGF5 has been identified in neurons associated with the limbic system, notably in neurons of the olfactory bulb and pyramidal cells of the hippocampus . In the nervous system, FGF5 is suggested to serve as a neurotrophic and differentiative factor for cholinergic and serotonergic neurons projecting to the hippocampus . This suggests potential applications in neurodegenerative disorders or brain injury.

  • Developmental biology: During embryonic development, FGF5 mRNA is found in the embryoblast, lateral somatic mesoderm, and myotomes cranial to the tail region . FGF5 may delay terminal myoblast differentiation during cell migration and continue to impact muscle post-natally where it functions as a target-derived neurotrophic factor of skeletal muscle . This suggests applications in understanding developmental disorders and potentially in regenerative medicine.

  • Angiogenesis regulation: FGF5 promotes angiogenesis of human aortic endothelial cells , suggesting potential applications in wound healing, tissue engineering, and vascular disorders.

These diverse functions of FGF5 highlight the need for continued research into its regulatory mechanisms and potential therapeutic applications across multiple biological systems.

How might integrating genomic, transcriptomic, and proteomic approaches advance FGF5 research?

Integrating multi-omics approaches offers several advantages for advancing FGF5 research:

  • Identifying novel regulatory mechanisms: Combining genomic analysis of FGF5 variants (such as rs1458038) with transcriptomic and proteomic data could reveal how genetic variation influences FGF5 expression and function across different tissues and disease states . This could explain the underlying mechanisms behind associations with blood pressure and other phenotypes.

  • Characterizing tissue-specific functions: The Human Protein Atlas resource contains information on FGF5 expression profiles at both mRNA and protein levels across 44 normal tissue types . Integrating this data with cell-type specific transcriptomics and proteomics could reveal tissue-specific functions and interacting partners of FGF5.

  • Understanding pathway interactions: The interactions between FGF5 and other signaling pathways (Hedgehog, androgen receptor, and SOX2 signaling) could be better characterized through integrated analyses of transcriptomic and proteomic changes following FGF5 modulation .

  • Biomarker discovery: In cancer research, integrated multi-omics approaches could identify signature patterns of FGF5 expression and associated molecular changes that might serve as biomarkers for diagnosis, prognosis, or treatment response .

  • Drug target validation: Proteomic and transcriptomic analyses following FGF5 inhibition (e.g., with RNA aptamers) could validate on-target effects and identify potential off-target effects, helping to refine therapeutic strategies .

By leveraging these integrated approaches, researchers can develop a more comprehensive understanding of FGF5 biology and accelerate the translation of basic research findings into clinical applications.

Product Science Overview

Structure and Expression

FGF-5 is a secreted, heparin-binding protein with a molecular weight of approximately 27.7 kDa . It is expressed in various tissues, including the mesenchyme, skeletal muscles, central nervous system, and hair follicles . The human recombinant form of FGF-5 is typically produced in Escherichia coli and is available as a lyophilized powder suitable for cell culture .

Biological Functions

FGF-5 plays a crucial role in several physiological processes:

  1. Hair Growth Regulation: FGF-5 is known to regulate the hair growth cycle. Its inactivation in mice leads to the angora phenotype, characterized by long hair . In humans, FGF-5 causes the regression of hair follicles, thereby controlling hair length .

  2. Neurotrophic Activity: In the peripheral nervous system, FGF-5 acts as a muscle-derived trophic factor for motor neurons. It demonstrates neurotrophic activity in vivo and influences astroglial characteristics in vitro .

  3. Oncogenic Potential: FGF-5 functions as an oncogene in glioblastoma due to its autocrine and paracrine functions. Its expression is elevated in human astrocytic brain tumors, which is linked to increased malignancy .

  4. Cell Differentiation and Proliferation: FGF-5 promotes cell differentiation and proliferation by binding to FGF receptor 1 and FGF receptor 2 (FGFR1 and FGFR2, respectively) . It plays an important regulatory role in skeletal muscle development and has been identified in neurons of the limbic system, especially those of the olfactory bulb and pyramidal cells of the hippocampus .

Applications

Human recombinant FGF-5 is widely used in research and biotechnology. It has been utilized for the induction of differentiation in various embryonic stem cell lines and for studying the mechanisms of hair growth and neurotrophic activity . Additionally, FGF-5’s role in oncogenesis makes it a potential target for cancer research and therapy .

Physical Form and Storage

The human recombinant FGF-5 is typically lyophilized from a solution containing MOPS, sodium sulfate, EDTA, DTT, and trehalose, with bovine serum albumin as a stabilizer . It is recommended to store the lyophilized powder at -20°C to maintain its stability and avoid repeated 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.