Fact-checked by Grok 2 weeks ago

Growth factor

A growth factor is a secreted, biologically active protein that regulates cellular processes by binding to specific receptors on target cells, thereby influencing , , , , and . These molecules are naturally produced by various cells throughout the body and play essential roles in maintaining , embryonic , and adult physiology. Growth factors exert their effects through autocrine, paracrine, or endocrine signaling mechanisms, where they bind to transmembrane receptors that activate intracellular pathways such as the MAPK/ERK and PI3K/AKT cascades, leading to changes that drive cellular responses. In normal physiology, they are critical for processes like , where they promote , , and synthesis to facilitate tissue repair. Dysregulation of growth factor signaling, however, contributes to pathological conditions, including cancer—where overexpression can stimulate uncontrolled —and fibrotic diseases, as well as diabetic complications like . Prominent examples of growth factors include epidermal growth factor (EGF), which stimulates epithelial and is vital for and mucosal repair; platelet-derived growth factor (PDGF), involved in and proliferation of cells during ; and vascular endothelial growth factor (VEGF), which induces blood vessel formation and permeability. These belong to broader families, such as the EGF family, PDGF family, and VEGF family, each with distinct receptor interactions and tissue-specific functions. Additionally, neurotrophic growth factors like nerve growth factor (NGF) support neuronal survival and differentiation, underscoring their diverse applications in and targeted therapies.

Definition and Fundamentals

Definition and Characteristics

Growth factors are naturally occurring proteins or polypeptides that bind to specific cell surface receptors on target cells, thereby regulating essential cellular processes such as , , , , and . These molecules function as signaling agents in intercellular communication, typically exerting their effects at low concentrations to modulate cellular behavior in a precise manner. Key characteristics of growth factors include their high specificity for particular s, mediated by the expression of corresponding receptors on responsive s, which ensures targeted regulation of physiological responses. Their actions are often dose-dependent, where the magnitude and type of cellular response vary with concentration, allowing fine-tuned control over processes like . Growth factors predominantly operate through , where they are secreted by one cell type to influence neighboring s, or , in which a cell responds to its own secreted factors; these modes facilitate short-range interactions critical for coordinated responses. They play pivotal roles in embryonic development by guiding and , in by promoting repair and , and in adult homeostasis by maintaining cellular balance and preventing degeneration. Structurally, growth factors are generally small polypeptides ranging from 5 to 20 in molecular weight, enabling efficient and stability in extracellular environments. Many possess specific motifs, such as heparin-binding domains in certain families, which facilitate interactions with components like proteoglycans, thereby modulating their and localization. In terms of function, growth factors commonly stimulate and progression by activating receptor kinases upon binding, leading to downstream cascades that promote and division through activation of transcription factors that induce and protein synthesis.

Historical Development

The concept of growth factors emerged from early 20th-century observations in and research, where scientists noted that clotted blood serum stimulated more effectively than cell-free plasma, implying the release of bioactive substances during . These findings laid the groundwork for identifying specific mitogenic agents involved in repair. In , researchers proposed the existence of "wound hormones" to explain accelerated regeneration at sites, attributing it to diffusible factors from damaged s that promoted cellular growth and migration, though these remained uncharacterized at the time. A major milestone came in 1974 with the isolation of (PDGF) by Russell Ross and colleagues, who demonstrated that this component, absent in , specifically stimulated the proliferation of arterial cells , highlighting its role as a key released from platelets during clotting. This marked the first purification of a mammalian growth factor and shifted focus toward polypeptide mediators of cell growth. Building on this, the 1980s saw significant breakthroughs, including Stanley Cohen's identification of (EGF) in the 1960s, which earned him the 1986 in or Medicine for revealing its mitogenic effects on epithelial cells; Cohen's work, detailed in his Nobel lecture, underscored EGF's isolation from mouse submaxillary glands and its stimulation of eyelid opening and incisor eruption in newborns. Concurrently, genes for fibroblast growth factors (FGFs), such as basic FGF (FGF2), were cloned in 1986 by Abraham et al., enabling molecular studies of their angiogenic and proliferative properties across bovine tissues. The and 2000s advanced understanding through the elucidation of (VEGF) in , first cloned and characterized by Leung et al. in 1989 from bovine pituitary cells, with subsequent studies in the by Ferrara confirming its specific mitogenic activity on endothelial cells and central role in pathological vessel formation. Similarly, research on insulin-like growth factors (IGFs), particularly IGF-1 and IGF-2, intensified during this period, revealing their regulation of and ; historical reviews trace their discovery to the 1950s but highlight 1990s-2000s advancements in linking IGFs to insulin signaling and metabolic disorders, as synthesized in Daughaday's foundational work and later genetic analyses. These developments expanded the growth factor paradigm to include diverse physiological contexts. From the 2000s onward, growth factor research integrated with biology and technologies, such as CRISPR-Cas9, to dissect gene functions and enhance regenerative applications.

Major Classes and Examples

Growth factors are classified into families primarily based on their structural features and the types of receptors they bind to, which are often receptor kinases (RTKs) or serine/ kinase receptors. This helps in understanding their specificity and roles in cellular communication. Major families include the (PDGF) family, (EGF) family, (FGF) family, (IGF) family, (VEGF) family, and transforming growth factor-β (TGF-β) superfamily, among others. The PDGF family consists of four ligand isoforms—PDGF-A, PDGF-B, PDGF-C, and PDGF-D—that dimerize to form various combinations such as PDGF-AA, PDGF-BB, and PDGF-AB, binding to PDGFRα and PDGFRβ receptors, which are class III RTKs characterized by five immunoglobulin-like domains and a insert. These primarily target fibroblasts and cells. The EGF family includes ligands like EGF and transforming growth factor-α (TGF-α), which bind to receptors (/ErbB1, ErbB2–4), cysteine-rich RTKs, targeting epithelial cells. The FGF family encompasses 23 members (FGF1 through FGF23), with acidic FGF (FGF1) and basic FGF (FGF2) as prototypical examples, binding to FGFR1–4 receptors, which are RTKs with two to three immunoglobulin-like domains. They target a broad range of and . The IGF family comprises IGF-1 and IGF-2, which bind to the IGF-1 receptor (IGF-1R, a class II RTK) and IGF-2 receptor (IGF-2R/M6P receptor), influencing widespread cellular proliferation. The VEGF family includes VEGF-A through VEGF-D and (PlGF), binding to VEGFR1–3 receptors, which are RTKs with seven immunoglobulin-like domains, primarily targeting endothelial cells. The TGF-β superfamily consists of over 30 members, including TGF-β1–3, activins, and bone morphogenetic proteins (BMPs), which bind to heteromeric complexes of type I and type II serine/threonine receptors (e.g., TGFBR1 and TGFBR2), regulating , , , and extracellular matrix production across diverse tissues. Other notable growth factors include growth factor (HGF), which binds to the c-MET receptor (an RTK with a kinase domain), targeting epithelial and endothelial cells, and (NGF), a member of the family that binds to TrkA and p75NTR receptors, targeting neurons. Emerging classifications recognize , such as (BDNF), as a subset of growth factors acting on TrkB receptors to support neuronal survival, with research highlighting their roles in and therapeutic applications. morphogenetic proteins (BMPs), part of the TGF-β superfamily, function as growth factor subsets binding to BMP receptors (serine/threonine kinases), influencing and . Recent studies have explored cytokine-growth factor hybrids, such as BMPs exhibiting immunoregulatory cytokine-like properties alongside growth promotion.

Comparison to Cytokines

Growth factors and cytokines, while both serving as extracellular signaling proteins, exhibit notable structural differences. Growth factors, such as (EGF) and (PDGF), act as specific ligands for receptor tyrosine kinases (RTKs), enabling precise dimerization and activation upon binding. In contrast, cytokines like (e.g., IL-2) and interferons (e.g., IFN-α) bind to multi-subunit cytokine receptors lacking intrinsic kinase activity, often relying on associated kinases (JAKs) for signaling initiation. This structural specificity in growth factors supports targeted cellular responses, whereas the modular architecture of cytokine receptors allows for broader, context-dependent interactions across immune cell types. Functionally, growth factors primarily drive , , and survival during embryonic development, tissue repair, and , with examples like (VEGF) promoting in healing processes. Cytokines, however, predominantly orchestrate immune responses, , and hematopoiesis, often exerting both stimulatory and inhibitory effects; for instance, (TNF) can induce in infected cells while amplifying inflammatory cascades. These distinctions arise from divergent downstream pathways: growth factors typically activate RTK-mediated mitogen-activated protein kinase (MAPK) and phosphoinositide 3-kinase (PI3K) routes for mitogenic effects, whereas cytokines engage JAK/STAT signaling to modulate immune cell activation and recruitment. Despite these differences, overlaps exist, particularly with molecules like transforming growth factor-β (TGF-β), which functions as both a growth factor—promoting production and in and —and a , suppressing immune responses by inducing regulatory T cells (Tregs) and inhibiting pro-inflammatory T helper 1 (Th1) cells. In certain contexts, such as , TGF-β inhibits , blurring lines between growth promotion and immune modulation. Regulatory expression further differentiates them: growth factors are often constitutively produced by stromal cells to maintain , while cytokines are inducibly expressed by activated immune cells in response to pathogens or injury, ensuring rapid but transient signaling. Recent 2020s research highlights hybrid signaling in , where growth factors like EGF and VEGF contribute to elevated pro-inflammatory production (e.g., IL-6, TNF) in the , fostering and metastatic progression. For example, TGF-β's dual role exacerbates -driven inflammation in advanced tumors, promoting and immune evasion, which has informed combination therapies targeting both pathways to mitigate tumor-promoting inflammation.

Biological Sources and Roles

Growth factors are produced by a variety of cells, including endothelial cells, fibroblasts, and macrophages, in addition to platelets, which serve as a key storage and rapid-release depot.

Production in Platelets

Platelets function as a major storage depot for growth factors, primarily within their alpha-granules, which serve as specialized reservoirs enabling rapid release during physiological responses. These granules contain high concentrations of key growth factors, including (PDGF), transforming growth factor-β (TGF-β), (VEGF), (EGF), and (FGF). For instance, PDGF is stored at levels of approximately 80–100 ng per 10^9 platelets, while TGF-β reaches about 48 ng per 10^9 platelets, ensuring a potent local supply upon demand. The production of these growth factors begins in megakaryocytes during , where they are synthesized and packaged into nascent alpha-granules that are subsequently inherited by maturing platelets. Megakaryocytes actively produce PDGF and TGF-β through regulated biosynthetic pathways, contributing to the majority of the stored content. In addition, circulating platelets exhibit selective uptake of certain growth factors, such as IGF-I, from via , further enriching alpha-granule contents and allowing adaptation to systemic levels. Release from alpha-granules is triggered by platelet during , primarily by agonists such as , , or (ADP), which bind to surface receptors and initiate degranulation cascades. This process delivers growth factors in a localized manner at sites, with rapid kinetics: 70–80% of stored PDGF is released within the first 10 minutes of , facilitating immediate . Such efficient discharge supports early wound repair and vascular by promoting endothelial and remodeling. Emerging research underscores the involvement of platelet-derived extracellular vesicles in augmenting growth factor delivery. A 2023 first-in-human demonstrated that allogeneic platelet-derived extracellular vesicles are safe and well-tolerated, with potential for promoting in regenerative contexts.

Functions in Development and Homeostasis

Growth factors play essential roles in embryonic by regulating spatial and temporal patterns of , , and . For instance, fibroblast growth factors (FGFs) are critical for limb bud formation, where FGF8 and establish signaling centers that direct outgrowth and patterning of the limb axis through gradients that influence mesenchymal condensation and . Similarly, bone morphogenetic proteins (BMPs), particularly BMP4 and BMP7, mediate dorsoventral patterning in the by forming ventral-to-dorsal gradients that specify induction, somite formation, and tissue identity via differential activation of Smad-dependent transcription. In adult homeostasis, growth factors maintain organ size, vascular integrity, and epithelial integrity. Insulin-like growth factor 1 (IGF-1) sustains muscle and bone mass by promoting satellite cell proliferation in and osteoblast activity in , thereby counterbalancing age-related and ensuring structural stability. (VEGF) regulates and vascular tone, supporting endothelial cell survival and permeability to preserve tissue under physiological conditions. (EGF) drives epithelial renewal in the skin by stimulating keratinocyte migration and proliferation, facilitating continuous barrier maintenance against environmental stressors. During injury response, growth factors orchestrate phases from to resolution. (PDGF), released from platelets at injury sites, promotes the proliferative phase by recruiting fibroblasts and stimulating synthesis, accelerating formation. (TGF-β), particularly TGF-β1, guides the remodeling phase by inducing differentiation and crosslinking, which contracts and strengthens the wound bed for maturation. Dysregulated TGF-β signaling can lead to excessive matrix deposition and , transforming normal repair into pathological scarring with persistent and tissue stiffening. Growth factors integrate systemically through feedback loops with hormones, exemplified by IGF-1's interaction with insulin, where IGF-1 enhances insulin sensitivity in peripheral tissues while insulin reciprocally stimulates hepatic IGF-1 production to fine-tune glucose and anabolic homeostasis. These mechanisms exhibit evolutionary conservation across species, with core families like FGFs, BMPs, and IGFs retaining structural and functional homology from invertebrates to mammals, underscoring their ancient origins in metazoan development and tissue maintenance. Recent 2024 research highlights growth factors' involvement in microbiome-host interactions for gut homeostasis, where fibroblast growth factor 1 (FGF1) modulates microbial composition to reduce inflammation and improve epithelial barrier function in a high-fat diet model in rainbow trout, analogous to diabetic conditions.

Molecular Mechanisms

Receptor Binding and Activation

Growth factors exert their effects primarily through interaction with specific cell surface receptors, which transduce extracellular signals into intracellular responses. The majority of growth factors, such as those in the (PDGF) and (EGF) families, bind to receptor kinases (RTKs), a of transmembrane proteins characterized by an extracellular ligand-binding , a single transmembrane , and an intracellular . In contrast, transforming growth factor-β (TGF-β) family members engage serine/ receptors, while fibroblast growth factors (FGFs) and vascular endothelial growth factors (VEGFs) also utilize RTKs but often require co-receptors for efficient signaling. While the majority signal through RTKs or serine/ receptors, some growth factor receptors, including those for , can be transactivated by G-protein-coupled receptors (GPCRs), though RTKs predominate in canonical pathways. The binding process typically involves high-affinity interactions that induce receptor oligomerization, often dimerization, leading to activation. For instance, EGF binds to the () with a (Kd) of approximately 10^{-9} M, promoting homodimerization through conformational changes in the extracellular domain; this exposes a dimerization arm in subdomain II, stabilizing an asymmetric arrangement of the intracellular domains. Similarly, PDGF induces dimerization of PDGF receptors (PDGFRα and PDGFRβ), either as homodimers or heterodimers, via ligand crosslinking that reorients extracellular domains and activates the . In the case of TGF-β, the ligand sequentially binds the type II receptor (TβRII), which then recruits and phosphorylates the type I receptor (TβRI), forming a heterotetrameric complex of two type I and two type II subunits. These events trigger conformational shifts that expose domains, initiating transphosphorylation. Receptor specificity is maintained through structural features of the extracellular domains, yet allows multiple receptors to respond to a single growth factor, enabling nuanced cellular responses. VEGFs, for example, exhibit this through three related RTKs: VEGFR1 binds VEGF-A with high affinity (Kd ~2–10 pM) but signals weakly, acting partly as a ; VEGFR2, the primary mediator of , binds VEGF-A with moderate affinity (Kd ~100–400 pM) and drives robust signaling; and VEGFR3 preferentially interacts with VEGF-C and VEGF-D to promote lymphangiogenesis. For FGFs, specificity is enhanced by co-receptors such as proteoglycans (HSPGs), which stabilize the FGF-FGFR ternary complex by bridging the ligand and receptor, increasing binding affinity and facilitating dimerization essential for activation. This and co-receptor dependence allow growth factors to elicit context-specific effects across tissues. Upon dimerization or oligomerization, receptor activation culminates in autophosphorylation of intracellular (for RTKs) or serine/ (for TGF-β receptors) residues, primarily in the C-terminal tails or juxtamembrane regions. These phosphosites serve as docking platforms for adaptor proteins and effectors, such as , Shc, and PLCγ, via SH2 or PTB domains, thereby initiating downstream signaling without delving into those pathways here. For , key sites include tyrosines 1068, 1086, 1148, and 1173, which recruit multiple adapters to diversify responses like and . Recent structural studies using cryo-electron microscopy (cryo-EM) have provided high-resolution insights into these complexes, revealing dynamic interfaces and allosteric mechanisms. For example, the 2023 cryo-EM structure of the heterodimer at 3.3 Å resolution elucidates how ligand-induced asymmetry propagates from the extracellular to domains, refining models of activation and highlighting therapeutic targets. Similar advances for FGF-receptor-HS complexes confirm the coreceptor's role in ternary assembly, addressing prior gaps in understanding ligand specificity.

Intracellular Signaling Pathways

Upon binding to their receptors, growth factors initiate a cascade of intracellular signaling events that transduce extracellular cues into cellular responses such as , , and . These pathways are primarily activated through receptor kinases (RTKs) or other receptor families, leading to events that recruit and activate downstream effectors. The (MAPK)/extracellular signal-regulated kinase (ERK) pathway is a central mediator of growth factor-induced . In this cascade, ligand-bound RTKs phosphorylate adaptor proteins like and Shc, which recruit the Sos to activate by promoting GTP binding. Active then stimulates , which sequentially phosphorylates and activates MEK1/2, culminating in ERK1/2 activation; phosphorylated ERK translocates to the to phosphorylate transcription factors like Elk-1, driving expression of genes such as c-Fos and D1. This pathway is exemplified by (EGF) signaling through , where sustained ERK activation correlates with progression. The (PI3K)-Akt pathway promotes cell survival, growth, and metabolic adaptation in response to growth factors like (IGF-1). Upon RTK activation, PI3K is recruited via its regulatory subunit p85 and converts phosphatidylinositol-4,5-bisphosphate (PIP2) to phosphatidylinositol-3,4,5-trisphosphate (PIP3), which docks and activates Akt (also known as ) at the plasma membrane through PDK1-mediated . Active Akt inhibits pro-apoptotic proteins like FoxO and Bad while activating to enhance protein synthesis and . In certain contexts, particularly with growth factors overlapping cytokine functions such as interleukin-6 or granulocyte-macrophage colony-stimulating factor, the Janus kinase (JAK)-signal transducer and activator of transcription (STAT) pathway directly influences transcription. Receptor-associated JAKs autophosphorylate upon ligand binding, creating docking sites for STAT proteins, which are then phosphorylated, dimerize, and translocate to the nucleus to regulate genes involved in proliferation and immune responses. Phospholipase Cγ (PLCγ)-mediated signaling mobilizes intracellular calcium to support processes like and . Growth factors such as (PDGF) activate PLCγ via RTK , leading to of PIP2 into diacylglycerol (DAG) and inositol-1,4,5-trisphosphate (IP3); IP3 binds IP3 receptors on the , releasing Ca²⁺ stores that amplify downstream effectors like calmodulin-dependent kinases. These pathways exhibit extensive and integration to fine-tune cellular decisions. For instance, ERK and PI3K-Akt converge on shared transcription factors: ERK activates AP-1 (c-Jun/c-Fos heterodimers) for genes, while both pathways can synergize to activate , enhancing survival and inflammatory responses through IκB degradation. mechanisms, such as PTEN dephosphorylation of PIP3 to curb PI3K activity, prevent excessive signaling and maintain . Regulation of these cascades involves temporal and spatial dynamics. ERK phosphorylation typically peaks 10-15 minutes after growth factor stimulation before declining due to phosphatase activity like that of DUSP6, ensuring transient signaling for proliferation rather than differentiation. Spatial control is achieved through scaffold proteins, such as KSR1 for the Ras-Raf-MEK-ERK module, which localize components to specific cellular compartments like endosomes or the plasma membrane. Dysregulation of growth factor signaling pathways contributes to oncogenesis, often through hyperactivation via mutant RTKs. For example, mutations in lock the kinase in an active state, leading to constitutive MAPK/ERK and PI3K-Akt signaling that drives uncontrolled proliferation; similarly, mutations in gastrointestinal stromal tumors sustain JAK-STAT activity. Recent advances using single-cell sequencing (scRNA-seq) have revealed heterogeneity in growth factor signaling across tissues, highlighting cell-type-specific pathway activation. In muscle regeneration models, scRNA-seq demonstrated varied FGF7-mediated interactions between satellite cells and fibro-adipogenic progenitors, uncovering subpopulation-specific ERK and PI3K responses that influence repair outcomes. These findings, from 2024 studies, underscore the need for context-dependent models of signaling dynamics.

Medical and Therapeutic Applications

Clinical Uses and Therapies

Recombinant platelet-derived growth factor (PDGF-BB), marketed as becaplermin gel, was approved by the U.S. Food and Drug Administration (FDA) in 1997 for the treatment of lower extremity diabetic neuropathic ulcers that extend into the subcutaneous tissue or beyond, with adequate blood supply and debridement. Clinical trials supporting this approval demonstrated that becaplermin accelerates wound healing by promoting granulation tissue formation and epithelialization, with patients achieving complete wound closure in approximately 14-20% more cases compared to placebo controls after 20 weeks of treatment. Granulocyte colony-stimulating factor (G-CSF), in the form of filgrastim (Neupogen), received FDA approval in 1991 to reduce the duration of severe neutropenia and the incidence of febrile neutropenia in patients with nonmyeloid malignancies undergoing myelosuppressive chemotherapy. Pivotal trials showed filgrastim shortening neutropenia duration by 2-3 days, thereby decreasing infection risk. Erythropoietin (EPO), a hematopoietic growth factor, was approved by the FDA in 1989 as epoetin alfa (Epogen) for treating anemia associated with chronic kidney disease, including in dialysis and non-dialysis patients. This approval was based on studies where EPO increased hemoglobin levels by 2-4 g/dL, reducing the need for transfusions. Vascular endothelial growth factor (VEGF) inhibitors, such as (Avastin), have been approved by the FDA since 2004 for various oncology indications, including metastatic , non-small cell , and , by blocking VEGF-mediated to starve tumors. In combination therapies, has extended by 4-6 months in advanced patients. (FGF), particularly FGF-2, has been investigated for cardiac repair following (MI); early phase I/II trials demonstrated safety and improved myocardial via intracoronary administration in refractory patients, though larger phase II trials did not confirm significant enhancements in left ventricular function or clinical outcomes. Combinations of growth factors with stem cell therapies are being explored in regenerative medicine for tissue repair, where factors like PDGF and VEGF enhance mesenchymal engraftment and , promoting vascularization and reducing scar formation in preclinical models of ischemic injury. Growth factors are delivered through various methods tailored to therapeutic needs, including topical gels for localized application, as in becaplermin for diabetic ulcers, which provides sustained release over 12-24 hours per application. Systemic or targeted injections, such as subcutaneous for or intravenous EPO for , achieve rapid with peak effects within hours to days. Investigational vectors, including viral plasmids encoding FGF or VEGF, enable prolonged endogenous production at injury sites; early preclinical and phase I/II trials have shown accelerated closure rates when combined with scaffolds for chronic ulcers. Recent advancements include 2024 FDA approvals of biosimilars for established growth factor therapies, such as additional variants, expanding access while maintaining efficacy equivalence to originators. Anti-NGF monoclonal antibodies like have demonstrated approximately 30-40% pain reduction in phase III trials for diabetic . These applications build on growth factors' roles in normal by amplifying cellular and at therapeutic sites.

Risks, Challenges, and Research Directions

Growth factor therapies pose significant risks, primarily due to their inherent pro-proliferative properties that can inadvertently promote pathological processes. For example, (VEGF) enhances , which supports tumor growth, invasion, and metastasis in various cancers, raising concerns about exacerbating malignancies in patients with undetected tumors. Similarly, excessive administration of transforming growth factor-β (TGF-β) can induce by stimulating deposition and activation, leading to tissue scarring in organs such as the liver or lungs. Additionally, recombinant growth factors often trigger , resulting in the development of neutralizing antibodies that reduce therapeutic efficacy and potentially cause reactions. Key challenges in clinical application include the short of factors, necessitating frequent dosing to maintain therapeutic levels, which complicates patient and increases burden. Off-target effects arise from receptor across tissues, where factors may activate unintended pathways, leading to systemic side effects like or aberrant . Furthermore, the high production costs of these biologics limit accessibility, particularly in resource-constrained settings, and contribute to economic barriers in widespread adoption. Emerging research directions aim to mitigate these issues through engineered variants, such as PEGylated growth factors, which enhance and extend half-life while preserving bioactivity, as demonstrated in (FGF21) conjugates. Nanotechnology-based delivery systems, including nanoparticles and scaffolds, offer controlled release and targeted localization to minimize off-target actions and improve . modeling is being applied to predict pathway dysregulation, enabling simulation of growth factor interactions in like the pathway to identify safer intervention points. Preclinical studies, such as those evaluating mimetics like BNN27 for neurodegenerative conditions, explore multi-factor cocktails combining growth factors with other agents to address multifactorial diseases like Alzheimer's. Ethical concerns surrounding gene-edited growth factors, particularly heritable modifications to enhance factor expression, include risks of unintended genetic changes passed to offspring and inequities in access to such technologies. Long-term safety data from over 20 years of recombinant growth hormone therapy indicate no confirmed increased risk of or other neoplasias, though monitoring for rare events like persists.

References

  1. [1]
    Physiology, Growth Factor - StatPearls - NCBI Bookshelf
    A growth factor, as initially defined, is a secreted biologically active molecule that can affect the growth of cells.
  2. [2]
    Definition of growth factor - NCI Dictionary of Cancer Terms
    A substance made by the body that functions to regulate cell division and cell survival. Some growth factors are also produced in the laboratory.
  3. [3]
    Role of platelet-derived growth factors in physiology and medicine
    PDGFs may directly (thick arrows) stimulate tumor cell growth and EMT. PDGFs may further be involved in the recruitment of tumor fibroblasts and pericytes.
  4. [4]
    The role of growth factors in wound healing - PubMed
    Growth factors attract cells into the wound, stimulate their proliferation, and have profound influence on extracellular matrix deposition.
  5. [5]
    Growth factors and cancer - PubMed - NIH
    Growth factors, defined as polypeptides that stimulate cell proliferation, are major growth-regulatory molecules for cells in culture and probably also for ...<|control11|><|separator|>
  6. [6]
    Classification of Growth Factors and Their Receptors - NCBI - NIH
    Stem cell factor (SCF), also designated kit ligand, mast cell growth factor, or steel factor (SLF), is a hematopoietic and tissue growth factor that binds to ...Epidermal Growth Factor Family · The Insulin Family · Hepatocyte Growth Factor
  7. [7]
    Classes of Growth Factors Acting in the Nervous System - NCBI - NIH
    Some of these, such as the neurotrophins, act almost exclusively in the nervous system, whereas others, for example, fibroblast growth factors and insulin-like ...
  8. [8]
    Growth Factor - an overview | ScienceDirect Topics
    Growth factors are defined as a group of proteins capable of stimulating cellular growth, migration, proliferation, and differentiation.
  9. [9]
    Growth Factors and Cancer1 | Cancer Research - AACR Journals
    Growth factors, defined as polypeptides that stimulate cell proliferation, are major growth-regulatory molecules for cells in culture and probably also for ...
  10. [10]
    (PDF) Growth Factors - ResearchGate
    May 5, 2021 · Growth factors are relatively small and stable soluble polypeptides that mediate short-range cell-to-cell interactions.
  11. [11]
    Growth Factors in Regeneration and Regenerative Medicine
    Jul 6, 2020 · In this mini review, we focus on the ambiguous role of growth factors in regeneration, discuss their evolutionary importance, and highlight them ...Introduction · The Growth Factor/RTK Axis in... · The Epigenetic Landscape...
  12. [12]
    Fibroblast growth factors | Genome Biology | Full Text - BioMed Central
    Mar 9, 2001 · Fibroblast growth factors (FGFs) make up a large family of polypeptide growth factors that are found in organisms ranging from nematodes to humans.<|control11|><|separator|>
  13. [13]
    [PDF] Stanley Cohen - EPIDERMAL GROWTH FACTOR - Nobel Prize
    The results demonstrated that EGF directly stimulated the proliferation of epidermal cells in organ cultures of chick embryo skin; this mitogenic action of EGF, ...
  14. [14]
    The History of the Insulin-Like Growth Factor System - Available
    Nov 29, 2022 · This article focuses on the history of the components of the IGF system, with an emphasis on the peptide hormones, IGF-I and -II, their cell surface receptors,
  15. [15]
    CRISPR/Cas system: An emerging technology in stem cell research
    In this review, we describe recent applications of CRISPR/Cas9 to stem cells for counteracting degenerative diseases.Crispr/cas System: An... · The Crispr/cas9 Technology · Crispr/cas9 Applications In...Missing: 2000-2025 | Show results with:2000-2025
  16. [16]
    Stem cell and CRISPR/Cas9 gene editing technology in Alzheimer's ...
    Researchers have found that CRISPR/Cas9 has a very high accuracy rate in correcting disease-causing genes, and it has been found that converting APOE4 to APOE3 ...Missing: 2000-2025 | Show results with:2000-2025
  17. [17]
    Insulin-like growth factors: Ligands, binding proteins, and receptors
    The insulin-like growth factor family of ligands (IGF-I, IGF-II, and insulin), receptors (IGF-IR, M6P/IGF-IIR, and insulin receptor [IR]), and IGF-binding ...Missing: classification | Show results with:classification
  18. [18]
    Mesenchymal stem cells in treating human diseases - Nature
    Aug 22, 2025 · MSCs secrete neurotrophic factors, such as brain-derived neurotrophic factor (BDNF) and β-nerve growth factor (β-NGF), which are vital for ...
  19. [19]
    Bone Morphogenetic Proteins - PMC - PubMed Central - NIH
    BMPs and the related “growth and differentiation factors” (GDFs) are members of the transforming growth factor β (TGF-β) family, and transduce their signals ...
  20. [20]
    Article Bone Morphogenic Proteins Are Immunoregulatory Cytokines ...
    Oct 6, 2020 · Bone morphogenic proteins (BMPs) are members of the transforming growth factor β (TGF-β) cytokine family promoting differentiation, ...
  21. [21]
  22. [22]
    Cytokines, chemokines and growth factors - Autoimmunity - NCBI - NIH
    Cytokines are important molecules for cell communication. The term cytokine (Greek –cyto, cell, and –kinos, movement) was proposed by Stanley Cohen in 1974.Introduction · Cytokines, cytokine receptors... · Cytokines families signaling...
  23. [23]
    Platelet CD62 expression and PDGFAB secretion in patients ... - NIH
    PDGF secretion from thrombin-stimulated platelets was slightly reduced from 99 ± 33 ng/109 platelets in sample I to 82 ± 31 ng/109 platelets under abciximab ...Platelet Aggregation · Flow Cytometry · Figure 1<|separator|>
  24. [24]
    Platelet TGF-β1 contributions to plasma TGF-β1, cardiac fibrosis ...
    Similarly, total TGF-β1 levels in platelet releasates were approximately 94% lower in Tgfb1flox mice (3.0 ± 1.0 ng/109 platelets; n = 4) than control mice (48.0 ...
  25. [25]
    Synthesis of Transforming Growth Factor-β1 by Megakaryocytes and ...
    In this study, we clearly demonstrated localization of TGF/β1 to platelet and megakaryocyte α-granules by several techniques based on immunologic recognition.
  26. [26]
    A Novel Mechanism Directing Them into α Granules of Platelets ...
    Lysis of platelets releases the contents of the α-granules, which contain growth factors, including insulin-like growth factor I (IGF-I) and IGF-binding protein ...
  27. [27]
    Signaling During Platelet Adhesion and Activation
    Platelets are activated by adhesion to adhesive proteins, such as von Willebrand factor and collagen, or by soluble platelet agonists, such as ADP, thrombin, ...
  28. [28]
    Applications of the regenerative capacity of platelets in modern ...
    In activated PRP, 70% of the GFs are released within the first 10 min and almost 100% within one hour [1], [3]. Many varieties of PRP preparations exist: pure ...
  29. [29]
    First‐in‐human clinical trial of allogeneic, platelet‐derived ...
    Jun 23, 2023 · First-in-human clinical trial of allogeneic, platelet-derived extracellular vesicles as a potential therapeutic for delayed wound healing.
  30. [30]
    Functional Roles of FGF Signaling in Early Development of ... - NIH
    Aug 20, 2021 · Fibroblast growth factors (FGFs) comprise a large family of growth factors, regulating diverse biological processes including cell proliferation, migration, ...
  31. [31]
    BMP Signaling: Lighting up the Way for Embryonic Dorsoventral ...
    In this review, we summarize the current knowledge of the function and regulation of BMP signaling in DV patterning.
  32. [32]
    The Insulin-Like Growth Factor System in Bone - NIH
    This review discusses the roles of the IGF system in the regulation of skeletal homeostasis. ... Circulating levels of IGF-1 directly regulate bone growth and ...
  33. [33]
    The use of epidermal growth factor in dermatological practice - PMC
    Dec 30, 2022 · Epidermal growth factor (EGF) is a growth factor that plays a pivotal role in wound healing and maintaining tissue homeostasis.
  34. [34]
    Growth factors and cytokines in wound healing - PubMed - NIH
    This review will focus on the specific roles of these growth factors and cytokines during the wound healing process.
  35. [35]
    Critical Role of Transforming Growth Factor Beta in Different Phases ...
    This review highlights the critical role of transforming growth factor beta (TGF-β)1–3 within different phases of wound healing, in particular, late-stage wound ...
  36. [36]
    Inflammation, wound repair, and fibrosis - PubMed Central - NIH
    This 2013 Annual Review Issue of The Journal of Pathology offers an up-to-date glimpse of ongoing research in the fields of inflammation, wound healing, and ...
  37. [37]
    The role of insulin-like growth factor-I and its binding proteins in ...
    This review addresses the possible role of the insulin-like growth factor (IGF)-axis in normal glucose homoeostasis and in the etiopathogenesis of type 2 ...
  38. [38]
    TGF-β/BMP signaling and other molecular events: regulation of ...
    The evolutionary significance of the BMP family is highlighted by the conserved nature of the canonical TGF-β/BMP signaling over at least 700 million years ...
  39. [39]
    Understanding Cytokine and Growth Factor Receptor Activation ...
    In this article we review the current state of knowledge on the activation mechanisms of cytokine and growth factor receptors.
  40. [40]
    Review cell signaling by receptor tyrosine kinases - ScienceDirect.com
    Jun 25, 2010 · In this Review, we discuss insights into the mechanism of RTK regulation that have emerged from recent structural and functional studies.
  41. [41]
    Mechanisms of TGF-β Signaling from Cell Membrane to the Nucleus
    A TGF-β ligand initiates signaling by binding to and bringing together type I and type II receptor serine/threonine kinases on the cell surface. This allows ...
  42. [42]
    Activation of the EGF Receptor by Ligand Binding and Oncogenic ...
    In this review, we discuss the mechanisms underlying the ligand-induced activation of the preformed EGFR dimer, as well as how oncogenic mutations ...
  43. [43]
    Mechanism of activation of the TGF-β receptor - Nature
    Aug 4, 1994 · Transforming growth factor-β (TGF-β) signals by contacting two distantly related transmem-brane serine/threonine kinases called receptors I ...
  44. [44]
    Structural Basis for Vascular Endothelial Growth Factor Receptor ...
    Dec 15, 2020 · VEGFR1 and VEGFR2 play a major role in angiogenesis, whereas VEGFR3 is mainly involved in lymphangiogenesis. (B) Isoforms of VEGF ligands: ...
  45. [45]
    Structural basis for FGF hormone signalling - Nature
    Jun 7, 2023 · Paracrine FGFs depend on heparan sulfate (HS) glycosaminoglycans as a mandatory coreceptor to stably bind and dimerize their cognate FGFRs. HS ...<|separator|>
  46. [46]
    Specific epidermal growth factor receptor autophosphorylation sites ...
    The six autophosphorylation sites in the COOH-terminal tail of EGFR are tyrosines 992, 1045, 1068, 1086, 1148, and 1173.
  47. [47]
    Structure and dynamics of the EGFR/HER2 heterodimer - PMC - NIH
    Feb 13, 2023 · Here, we report cryo-EM structures of the EGF- and epiregulin-bound EGFR/HER2 ectodomain complexes at resolutions of 3.3 Å and 4.5 Å, respectively.
  48. [48]
    Regulation of MAPKs by growth factors and receptor tyrosine kinases
    Hyperactivation of MAPKs, primarily the Erk pathway, is a landmark of cancer. We describe the many links of MAPKs to tumor biology and review studies that ...
  49. [49]
    MAPK signal pathways in the regulation of cell proliferation ... - Nature
    Mar 1, 2002 · ERK pathway. ERK has been the best characteried MAPK and the Raf-MEK-ERK pathway represents one of the best characteried MAPK signaling pathway ...
  50. [50]
    Targeting the RAS/RAF/MAPK pathway for cancer therapy - Nature
    Dec 18, 2023 · In this review, we delve deeper into the mechanistic insights surrounding RAF kinase signaling in tumorigenesis and RAFi-resistance.
  51. [51]
    PI3K/AKT/mTOR signaling transduction pathway and targeted ...
    Aug 18, 2023 · The PI3K/AKT/mTOR (PAM) signaling pathway is a highly conserved signal transduction network in eukaryotic cells that promotes cell survival, cell growth, and ...
  52. [52]
    The JAK/STAT signaling pathway: from bench to clinic - Nature
    Nov 26, 2021 · More than 50 cytokines and growth factors have been identified in the JAK/STAT signaling pathway, such as hormones, interferons (IFN) ...
  53. [53]
    The Inositol Trisphosphate/Calcium Signaling Pathway in Health ...
    Aug 10, 2016 · External stimuli, such as neurotransmitters, hormones, and growth factors, stimulate the formation of InsP3 by activating either the G protein- ...
  54. [54]
    Crosstalk in NF-κB signaling pathways | Nature Immunology
    Jul 19, 2011 · TRAFs represent a central point of divergence for activation of the NF-κB and AP-1 transcription factor pathways (Fig. 2a). Activation of AP-1 ...
  55. [55]
    A guide to ERK dynamics, part 1: mechanisms and models - PMC
    Outside the nucleus, ERK regulates cytoplasmic proteins involved in cell growth, metabolism, and differentiation. Pathway termination is regulated by numerous ...
  56. [56]
    Mechanisms of receptor tyrosine kinase activation in cancer
    Feb 19, 2018 · Abnormal RTK activation in human cancers is mediated by four principal mechanisms: gain-of-function mutations, genomic amplification, chromosomal ...
  57. [57]
    Single-cell RNA-seq reveals novel interaction between muscle ...
    May 16, 2024 · Single-cell RNA-seq reveals novel interaction between muscle satellite cells and fibro-adipogenic progenitors mediated with FGF7 signalling.
  58. [58]
    Becaplermin - Product Approval Information - Licensing Action
    Becaplermin is indicated for treatment of lower extremity diabetic neuropathic ulcers that extend into the subcutaneous tissue or beyond, and have adequate ...
  59. [59]
    A review of becaplermin gel in the treatment of diabetic neuropathic ...
    When it was approved by the FDA in 1997 for the treatment of chronic lower extremity diabetic neuropathic ulcers, rhPDGF-BB became the first and remains the ...
  60. [60]
    Filgrastim - StatPearls - NCBI Bookshelf - NIH
    Jul 3, 2023 · Filgrastim is a medication used to manage and treat neutropenia in patients with myelosuppression from chemotherapy or radiation
  61. [61]
    [PDF] NEUPOGEN (filgrastim) - accessdata.fda.gov
    There is a theoretical possibility that an antibody directed against filgrastim may cross-react with endogenous G-CSF, resulting in immune-mediated neutropenia;.
  62. [62]
    Information for Epogen/Procrit (Epoetin alfa) - FDA
    Apr 13, 2017 · Epogen/Procrit was approved on June 1, 1989 for the treatment of anemia associated with chronic renal failure, including patients on dialysis and patients not ...
  63. [63]
    Erythropoietin Stimulating Agents - StatPearls - NCBI Bookshelf - NIH
    The FDA has also approved ESAs for the treatment of anemia secondary to zidovudine therapy ... treatment, and the presence of EPO-neutralizing antibodies ...
  64. [64]
    FDA Approves Atezolizumab Plus Bevacizumab for Liver Cancer - NCI
    Jun 4, 2020 · FDA has approved the immunotherapy drug atezolizumab, used with bevacizumab, to treat some patients with advanced liver cancer.Missing: oncology | Show results with:oncology
  65. [65]
    Bevacizumab Granted Full FDA Approval for Glioblastoma
    Dec 7, 2017 · Bevacizumab (Avastin) has been granted a full approval by the FDA for the treatment of adult patients with glioblastoma that progressed following prior therapy.<|separator|>
  66. [66]
    Growth Factor Engineering Strategies for Regenerative Medicine ...
    Jan 21, 2020 · BMP, bone morphogenetic protein; EGF, epidermal growth factor; FGF, fibroblast growth factor; HGF, hepatocyte growth factor; IGF, insulin-like ...
  67. [67]
    Safety and efficacy of Becaplermin gel in the treatment of diabetic ...
    Jul 29, 2014 · Becaplermin gel remains the only US Food and Drug Administration (FDA)-approved growth factor for the treatment of diabetic neuropathic foot ulcers.
  68. [68]
    NEUPOGEN® (filgrastim) | Granulocyte Colony-Stimulating Factor
    NEUPOGEN is indicated to decrease the incidence of infection, as manifested by febrile neutropenia, in patients with nonmyeloid malignancies receiving ...Febrile Neutropenia (FN) · G-CSF Injection · Important Safety Information · Support
  69. [69]
    Growth factors & gene therapies for chronic wounds
    Dec 28, 2023 · Gene therapy offers an alternative approach to deliver growth factors to the chronic wound environment, but safety concerns surrounding gene ...
  70. [70]
    A Banner Year for Biosimilars: The 19 FDA Approvals From 2024
    Jan 21, 2025 · In 2024, the FDA approved 19 biosimilars across various therapeutic areas, including the first biosimilars for ustekinumab and denosumab.
  71. [71]
    Reinvigorating drug development around NGF signaling for pain - JCI
    Feb 17, 2025 · Nerve growth factor (NGF) signaling is a clinically validated target for the treatment of several prevalent types of chronic pain.
  72. [72]
    The Multifunctional Contribution of FGF Signaling to Cardiac ...
    FGFs are involved in the adaptive responses after myocardial infarction (MI) and induced hypertrophy. Moreover, FGFs are known to protect the heart against ...
  73. [73]
    Angiogenic signaling pathways and anti-angiogenic therapy for cancer
    May 11, 2023 · The vascular endothelial growth factor (VEGF) is the most typical regulator in tumor angiogenesis, which can mediate vascular permeability and ...
  74. [74]
    Anti-TGF-beta strategies for the treatment of chronic liver disease
    Permanent alcohol abuse may lead to chronic liver injury with deleterious sequelae such as liver cirrhosis and hepatocellular carcinoma.
  75. [75]
    Immunogenicity of recombinant human proteins: causes ... - PubMed
    Antibodies develop to varying degrees during treatment with human proteins, including insulin, growth hormone, granulocyte-macrophage colony-stimulating factor ...
  76. [76]
    Engineering growth factors for regenerative medicine applications
    Protein stability and half-life. A major limitation of natural growth factors is their short effective half-life due to poor stability or fast blood clearance.
  77. [77]
    Emerging Therapies in Chronic Wound Healing: Advances in Stem ...
    Oct 11, 2025 · Nonetheless, the translation of growth factor therapies remains limited by issues such as short half-lives, high production costs, and potential ...
  78. [78]
    Polyethylene Glycol Modified FGF21 Engineered to Maximize ...
    Apr 18, 2013 · We engineered site-directed PEGylated variants of FGF21 with sustained potency and minimized vacuole formation.
  79. [79]
    The Delivery and Activation of Growth Factors Using Nanomaterials ...
    Mar 22, 2023 · Nanomaterials are found to activate endogenous GFs via immune system stimulation, the stimulation of signal pathways, or gene therapy. The ...
  80. [80]
    Mathematical Modeling and Inference of Epidermal Growth Factor ...
    Dysregulation of the MAPK pathway is associated with various diseases ... Modeling cellular noise underlying heterogeneous cell responses in the epidermal growth ...
  81. [81]
    Multimodal beneficial effects of BNN27, a nerve growth factor ...
    Nov 25, 2024 · BNN27 is a non-toxic, newly developed 17-spiro-steroid analog, penetrating the blood-brain-barrier (BBB) and mimicking the neuroprotective effects of NGF.
  82. [82]
    What are the Ethical Concerns of Genome Editing?
    Aug 3, 2017 · Most ethical discussions about genome editing center on human germline editing because changes are passed down to future generations.
  83. [83]
    Long-term safety of recombinant human growth hormone in children
    Nov 11, 2009 · Conclusion: After more than 20 yr, leukemia, a major safety issue initially believed associated with GH, has not been confirmed, but other ...