Fact-checked by Grok 2 weeks ago

Platelet-derived growth factor

Platelet-derived growth factor (PDGF) is a family of dimeric glycoproteins composed of four polypeptide chains (A, B, C, and D) that assemble into five bioactive isoforms, acting as potent mitogens, chemoattractants, and regulators of cell survival for mesenchymal-origin cells such as fibroblasts, cells, and . Discovered in 1974 by Russell Ross and colleagues at the while studying the of , PDGF was initially identified as a soluble factor released from the alpha granules of activated platelets that promotes the and of arterial cells, contributing to the formation of atherosclerotic lesions. Subsequent research revealed its broader production by various cell types, including endothelial cells, macrophages, and fibroblasts, particularly at sites of injury or . Structurally, all PDGF isoforms share a conserved C-terminal cystine-knot domain stabilized by bonds, with molecular weights around 25–30 for the mature dimers; the A and B chains, encoded by genes on chromosomes 7 and 22 respectively, can form both homodimers (PDGF-AA, PDGF-BB) and the heterodimer PDGF-AB, whereas the more recently identified C and D chains (discovered in the late and early ) primarily form homodimers (PDGF-CC, PDGF-DD) and include an N-terminal that requires proteolytic cleavage by proteases like or for activation and secretion. These structural variations influence ligand-receptor specificity and , with PDGF-A and -B showing about 50% sequence identity to each other, while PDGF-C and -D share roughly 25% identity with A/B but 50% with one another. PDGF signals through two homologous cell-surface receptor kinases, PDGFRα (encoded on 4q12) and PDGFRβ (encoded on 5q32), which undergo dimerization (αα, αβ, or ββ) upon binding, leading to autophosphorylation and activation of downstream pathways including PI3K/AKT for cell survival and MAPK/ERK for and . In normal physiology, PDGF is indispensable for embryonic development—such as , , and cell —and adult tissue , including where it recruits to stabilize new vessels and stimulates production. Dysregulated PDGF signaling, however, drives pathological conditions like (e.g., in kidney and ), via excessive smooth muscle , and cancers such as gliomas and sarcomas through autocrine loops that promote tumor growth and . Therapeutically, PDGF inhibitors like target overactive receptors in certain malignancies, highlighting its clinical significance.

Discovery and History

Initial Discovery

Platelet-derived growth factor (PDGF) was first identified in the early 1970s as a key mitogen released from platelets, stimulating the proliferation of arterial smooth muscle cells (SMCs) in vitro. In 1974, Russell Ross and colleagues at the University of Washington demonstrated that dialyzed serum derived from clotted primate blood promoted robust SMC growth, whereas serum from recalcified platelet-poor plasma exhibited significantly reduced mitogenic activity. Experiments revealed that adding platelets or a platelet-free supernatant from thrombin-activated platelets to the plasma-derived serum restored its proliferative effects to levels comparable to whole blood serum, pinpointing the platelets as the primary source of the growth-promoting factor. This discovery was motivated by investigations into the cellular basis of atherosclerosis, where SMC proliferation plays a central role. Subsequent cell culture studies using platelet extracts further characterized the factor's potency. Extracts from human or primate platelets were applied to quiescent cultures of SMCs and fibroblasts, where they induced and cell division, as measured by incorporation of tritiated . For instance, in BALB/c 3T3 fibroblasts, platelet-derived material at concentrations equivalent to 1% whole triggered a marked increase in within 24 hours, highlighting its role as a competence factor for initiating the . These assays established the term "platelet-derived growth factor" (PDGF) to describe this cationic protein, distinct from other components like plasma growth-promoting activity. Between 1974 and 1978, initial purification efforts isolated PDGF from outdated , confirming its localization within platelet alpha granules—the dense storage organelles released upon activation. Using techniques such as , ion-exchange , gel filtration, and sodium dodecyl sulfate-polyacrylamide , researchers achieved up to 800,000-fold purification. The native protein was identified as a disulfide-bonded dimer with a molecular weight of approximately 30,000 , comprising two distinct polypeptide chains of about 14,000 and 17,000 . Early biochemical assays on glial cells and fibroblasts showed that purified PDGF at nanogram concentrations (e.g., 4 ng/mL) stimulated equivalently to 1% serum, underscoring its high .

Key Milestones

In 1979, key purification and partial characterization work by Carl-Henrik Heldin's group confirmed PDGF as a distinct mitogen released from alpha granules in platelets, demonstrating its role in stimulating connective tissue cell growth. The cloning of PDGF genes marked a pivotal advancement in the 1980s. In 1984, the PDGF-B chain gene was cloned by Heldin and colleagues, revealing its sequence homology to the v-sis oncogene from simian sarcoma virus and confirming PDGF as a dimeric protein composed of A and B chains. This was followed in 1986 by the cloning of the PDGF-A chain by Christer Betsholtz et al., which established the existence of homodimeric and heterodimeric isoforms and localized the gene to chromosome 7. Concurrently, in 1986, Yarden et al. cloned the PDGF receptor (PDGFR), identifying it as a transmembrane tyrosine kinase and elucidating its dimerization upon ligand binding, which laid the foundation for understanding PDGF signaling specificity. By the late 1980s, PDGF was recognized as the founding member of the PDGF/VEGF family, following the 1989 cloning of (VEGF) by Leung et al., which highlighted shared structural features such as the cystine-knot and conserved receptor-binding domains across these dimeric proteins. In the early 1990s, structural elucidation advanced with the 1992 determination of the of human PDGF-BB by Olofsson et al., revealing a novel antiparallel disulfide-linked homodimer with two protruding receptor-binding regions, which provided insights into its mitogenic potency and informed subsequent isoform comparisons. Parallel animal model studies emerged around this time; the first PDGF mice, reported in 1994 by Levéen et al. and Soriano, demonstrated lethal perinatal phenotypes in PDGF-B and PDGFR-β null mutants, including renal glomerulogenesis failure and cardiovascular defects, underscoring PDGF's essential role in vascular development and recruitment. The PDGF family was completed with the discovery of two additional chains in the early 2000s. PDGF-C was identified in 2000 by three independent groups through homology-based searches and expression studies, revealing it as a novel primarily activating PDGFRα. PDGF-D was cloned in 2001, showing specificity for PDGFRβ and requiring proteolytic activation similar to PDGF-C. These findings expanded the understanding of PDGF diversity and signaling specificity.

Structure and Classification

Molecular Composition

Platelet-derived (PDGF) consists of -linked homo- or heterodimers formed by two of four possible polypeptide chains, designated A, B, C, or D, resulting in a mature protein with a molecular weight of approximately 28-31 . These dimers are stabilized by covalent bonds, which are essential for the structural integrity and bioactivity of the . The four PDGF chains are encoded by separate genes located at distinct chromosomal positions in the : PDGFA on 7p22.3, PDGFB on 22q13.1, PDGFC on 4q32.1, and PDGFD on 11q22.3. The primary structure of each PDGF chain precursor comprises an N-terminal of 18-22 , followed by a prodomain and a central domain of about 100-140 residues. For PDGF-C and PDGF-D, the prodomain includes an N-terminal that maintains latency until proteolytic cleavage. This central domain contains eight highly conserved cysteine residues that form a characteristic cystine-knot motif, including three intermolecular disulfide bonds linking the two chains and two intramolecular bonds within each chain to maintain the dimeric fold. The is cleaved co-translationally during translocation into the , yielding the pro-PDGF form. Post-translational modifications are critical for PDGF maturation and include proteolytic to remove prodomains and, in some cases, N-linked . For PDGF-A and PDGF-B chains, intracellular by furin-like proprotein convertases at dibasic motifs (e.g., RRKR for A-chain, RGRR for B-chain) generates active dimers stored in platelet alpha-granules. In contrast, PDGF-C and PDGF-D are secreted as latent complexes with their prodomains intact and require extracellular proteolytic activation by serine proteases such as , tissue plasminogen activator (tPA), or plasminogen activator (uPA). occurs at specific residues in certain chains, such as three N-linked sites (Asn25, Asn55, Asn254) in PDGF-C, which may influence stability and , though it is absent or minimal in PDGF-A. Dimerization is represented simply as the covalent linkage of two monomeric chains, such as A + B → PDGF-AB, facilitated by the conserved cysteines in the domains. These structural elements form the foundational building blocks for the various PDGF isoforms.

Isoforms and Family Members

Platelet-derived (PDGF) exists in five principal dimeric isoforms, formed by disulfide-linked combinations of four distinct polypeptide chains: PDGF-A, PDGF-B, PDGF-C, and PDGF-D. These isoforms include the homodimers PDGF-AA, PDGF-BB, PDGF-CC, and PDGF-DD, as well as the heterodimer PDGF-AB. The chains share structural homology, particularly in their conserved C-terminal domains, but exhibit varied tissue-specific expression patterns that contribute to their functional diversity. For instance, PDGF-BB is the predominant isoform stored in platelet alpha-granules and released upon , while PDGF-AA is widely expressed in epithelial and mesenchymal cells during and repair processes. The isoforms display distinct ligand specificities for the two PDGF receptor tyrosine kinases, PDGFRα and PDGFRβ, which can form homodimers (αα, ββ) or heterodimers (αβ). PDGF-AA binds exclusively to PDGFRαα, PDGF-AB binds to both PDGFRαα and αβ, PDGF-BB binds to all three receptor dimers (αα, αβ, ββ), PDGF-CC binds to PDGFRαα and αβ, and PDGF-DD binds solely to PDGFRββ. This differential binding enables isoform-specific activation of signaling pathways tailored to cellular contexts. PDGF belongs to the PDGF/VEGF superfamily, sharing a characteristic cystine-knot fold in its domain—a structural motif involving three intramolecular bonds that stabilizes the dimeric for receptor . This evolutionary relationship with vascular endothelial growth factors (VEGFs), including VEGF-A, VEGF-B, VEGF-C, VEGF-D, VEGF-E, and (PlGF), underscores their common ancestry, despite VEGFs primarily promoting and through distinct receptor s. Homologs of PDGF are conserved across the animal kingdom, with PDGF/VEGF-like factors identified in such as (e.g., PVF1, PVF2, PVF3) and , where they regulate and patterning during development. These non-mammalian variants retain the cystine-knot structure and similar receptor-binding properties, highlighting the ancient origins of the superfamily.

Biological Mechanisms

Receptor Interactions

Platelet-derived growth factor (PDGF) ligands exert their effects by binding to two closely related cell-surface receptor kinases: platelet-derived growth factor receptor alpha (PDGFRα) and platelet-derived growth factor receptor beta (PDGFRβ). These receptors belong to the III of receptor kinases and possess an extracellular -binding domain composed of five immunoglobulin-like domains, a single transmembrane helix, and an intracellular kinase domain. Upon binding, PDGFRα and PDGFRβ can assemble into three possible dimeric complexes—homodimers αα and ββ, or the heterodimer αβ—each capable of transducing distinct signals depending on the involved. The specificities of the five dimeric PDGF isoforms (, , , , ) to these receptor dimers vary, with PDGF- exhibiting the broadest and highest affinity for all three combinations, while other isoforms show more restricted preferences. For instance, PDGF- and PDGF- bind exclusively to PDGFRα homodimers and αβ heterodimers, PDGF- binds to αα and αβ, and PDGF- binds only to αβ and ββ. These affinities have been quantified through assays, with dissociation constants (K_d) typically in the low nanomolar range for high-affinity interactions, such as K_d ≈ 0.1–1 for PDGF- to PDGFRββ. The following table summarizes the key specificities:
PDGF IsoformPDGFRααPDGFRαβPDGFRββ
AAHighHighNone
BBHighHighHigh
ABHighHighLow/None
CCHighHighNone
DDNoneHighHigh
This differential binding allows for isoform-specific activation of receptor dimers in various cellular contexts. Ligand binding to the extracellular domains of PDGFRs induces receptor dimerization, a critical step in activation. The dimeric PDGF ligands bridge two receptor molecules, primarily involving immunoglobulin-like domains 2 and 3 (D2 and D3) in the binding interface, which stabilizes the dimer and positions the intracellular domains in close proximity. This conformational change triggers trans-autophosphorylation of specific residues in the kinase domains and juxtamembrane regions, initiating the receptor's catalytic activity without requiring additional cofactors. Structural studies, including cryo-electron of the full-length PDGFRβ dimer bound to PDGF-BB, have revealed that ligand-induced dimerization involves a 2:2 (two ligands to two receptors) and highlights key hydrophobic and electrostatic interactions at the , such as the burial of a large hydrophobic surface area upon complex formation. Earlier crystallographic analyses of the extracellular domains, such as the 2010 structure of PDGF-B with the D1-D3 domains of PDGFRβ (PDB: 3MJG), further illustrate the dynamic rearrangements in the ligand's L1 loop upon receptor engagement. In terms of cellular distribution, PDGFRα is predominantly expressed on mesenchymal progenitor cells, including fibroblasts and osteoblasts during development, while PDGFRβ is primarily found on and associated with blood vessels. These expression patterns contribute to the tissue-specific responses elicited by PDGF signaling, with PDGFRα mediating broader proliferative effects in mesenchymal lineages and PDGFRβ supporting vascular stability.

Intracellular Signaling

Upon ligand-induced dimerization and activation, platelet-derived growth factor receptors (PDGFRs) undergo autophosphorylation on multiple intracellular residues, generating high-affinity docking sites for Src homology 2 (SH2) domain-containing adaptor and effector proteins. These phosphotyrosine (pY) motifs recruit key signaling molecules such as 3-kinase (PI3K), Cγ (PLCγ), and Src family kinases, initiating diverse downstream cascades that mediate cellular responses like , , and . The specific pY sites vary between PDGFRα and PDGFRβ isoforms; for instance, pY740 and pY751 on PDGFRβ bind the p85 regulatory subunit of PI3K, while pY1009 and pY1021 recruit PLCγ and , respectively. The recruited proteins activate several major intracellular pathways. The /extracellular signal-regulated kinase (MAPK/ERK) pathway, stimulated via Grb2-Sos-Ras activation often involving , promotes by driving transcription of genes like c-fos and cyclin D1. The PI3K/Akt pathway, triggered by PI3K lipid kinase activity, enhances cell survival and motility by phosphorylating targets such as Bad and glycogen synthase -3β (GSK-3β), inhibiting . Additionally, the /signal transducer and activator of transcription (JAK/STAT) pathway contributes to gene expression regulation; PDGF induces tyrosine phosphorylation of and via JAK2 and , leading to their dimerization, nuclear translocation, and activation of mitogenic genes like c-myc. This simplified sequence of receptor autophosphorylation on (Y) residues → pY-SH2 binding underscores the initiation of these pathways. PDGF signaling integrates with other pathways through crosstalk, amplifying or modulating responses. For example, PDGFRs interact with signaling via direct association or shared effectors like and FAK, enhancing ERK activation and in adhesive contexts. Similarly, overlap with pathways occurs through the shared JAK/ axis, where PDGF can potentiate STAT activation alongside cytokine receptors, influencing inflammatory and proliferative outcomes. Negative regulation ensures signaling fidelity and termination. Protein phosphatases (PTPs), such as PTP1B and TC-PTP, dephosphorylate PDGFR phosphotyrosines, attenuating activity and downstream effects; for instance, TC-PTP promotes receptor recycling and limits prolonged ERK signaling. loops further constrain pathways, including ERK-mediated inhibition of the Grb2-Sos complex to dampen activation, and ubiquitin ligase Cbl-induced PDGFR ubiquitination leading to lysosomal degradation. These mechanisms prevent excessive signaling and maintain cellular .

Physiological Roles

Developmental Functions

Platelet-derived growth factor (PDGF) plays essential roles in embryonic development by regulating , , and through . During embryogenesis, PDGF ligands, particularly PDGF-A and PDGF-B, interact with their cognate receptors PDGFRα and PDGFRβ to orchestrate key morphogenetic processes, including vascular stabilization and organ formation. These functions are most prominent in mid-gestation stages, such as embryonic day 10.5 (E10.5) in mice, when PDGF expression peaks in specific tissues to guide patterning and tissue assembly. In vascular development, PDGF-B expressed by endothelial cells binds to PDGFRβ on pericyte progenitors, promoting their , , and investment around nascent endothelial tubes to stabilize blood vessels. This paracrine mechanism is critical for , particularly in the , where PDGF-B expression is strongest in angiogenic sprout tip cells around E10. This process ensures proper vessel maturation and prevents structural weaknesses. In PDGF-B mice, pericyte recruitment fails, leading to a deficiency in , endothelial , and the formation of lethal microaneurysms in organs like the and ; embryos exhibit dilated vessels and hemorrhages, resulting in perinatal lethality around E16–E19. PDGF-A contributes to by directing the migration and patterning of cells and supporting alveolar formation in the . In development, PDGF-A, produced by epithelia and , acts as a chemoattractant to guide cranial and cardiac cell via PDGFRα signaling, which regulates epithelial-mesenchymal transition and interactions, including and modulation. Although PDGF-A null mice do not show overt defects, PDGFRα knockouts result in cranial malformations, , and incomplete cardiac contribution leading to ventricular septal defects. In development, PDGF-A drives the and distal spreading of alveolar smooth muscle cell progenitors from the midline, essential for secondary septation and alveolarization; its absence causes an emphysema-like phenotype with enlarged airspaces due to failed myofibroblast recruitment. PDGF-A null mice also exhibit skeletal abnormalities, including rib fusion and sternal defects, highlighting its role in mesenchymal patterning. PDGF signaling coordinates with (FGF) pathways to refine patterning during . In chick and models, PDGF-A enhances mesodermal cell toward FGF4 gradients, regulating N-cadherin expression via PI3K to facilitate directed migration from the without altering initial mesoderm specification. This interaction ensures proper mediolateral patterning and intercalation of mesodermal cells, integrating proliferative cues from PDGF with differentiative signals from FGF.

Tissue Repair and Homeostasis

Platelet-derived growth factor (PDGF) is released from activated platelets and macrophages at sites of tissue injury, initiating key reparative processes in adult . Upon vascular damage, platelets degranulate to discharge PDGF isoforms, particularly PDGF-BB, which acts as a potent and chemoattractant for and cells. Macrophages, recruited to the injury site, further amplify this response by secreting PDGF-BB, stimulating fibroblast proliferation and the synthesis of (ECM) components such as and , thereby promoting formation and wound closure. In angiogenesis during tissue repair, contributes to stabilizing and maturing nascent blood vessels within healing wounds. Expressed by and , PDGF-DD binds to PDGF receptor-β (PDGFRβ) on , recruiting to endothelial tubes and enhancing vascular integrity through deposition and coverage, which prevents leakage and supports nutrient delivery to regenerating tissues. This maturation process is essential for transitioning from proliferative to remodeling phases of , ensuring functional vascular networks. Beyond injury response, PDGF contributes to tissue homeostasis by regulating fluid balance through PDGFRα signaling in fibroblasts. In steady-state conditions, PDGF ligands activate PDGFRα on fibroblasts, modulating cellular tension and remodeling via phosphatidylinositol-3-kinase (PI3K) pathways, which maintain appropriate fluid pressure and prevent . This homeostatic function supports overall tissue and structural integrity in organs like and lungs. Studies in animal models demonstrate PDGF's impact on wound repair efficiency; targeted PDGF-B overexpression via adenoviral delivery in diabetic mouse models enhances formation and , leading to faster rates compared to controls. Hypoxic conditions in injured tissues further regulate PDGF expression through hypoxia-inducible factor-1α (HIF-1α), which transcriptionally upregulates PDGF genes to coordinate repair. Under low oxygen, stabilized HIF-1α binds to hypoxia response elements in PDGF promoters, particularly for PDGF-B, enhancing its production in macrophages and to drive and fibroblast activation during the proliferative phase of .

Clinical Applications

Therapeutic Uses

Becaplermin, a recombinant form of platelet-derived growth factor BB (PDGF-BB), was approved by the U.S. (FDA) in 1997 as a topical for the treatment of lower extremity diabetic neuropathic ulcers that extend into the or beyond and have an adequate blood supply. The mechanism involves stimulating and of cells critical for wound repair, such as fibroblasts, endothelial cells, and monocytes, thereby accelerating formation and epithelialization. In a combined analysis of four randomized, double-blind, -controlled phase III trials involving 478 patients with nonhealing diabetic foot ulcers, becaplermin (100 μg/g) applied daily increased the incidence of complete to 50% compared to 36% with (p=0.007) for ulcers ≤10 cm², with the time to complete healing reduced by approximately 30% (median 14.1 weeks vs. 20.1 weeks, p=0.01). In regenerative medicine, PDGF-BB has been incorporated into scaffolds for bone and periodontal tissue regeneration, particularly in dental applications. For instance, GEM 21S, an FDA-approved (2005) growth-factor enhanced matrix combining recombinant human PDGF-BB with beta-tricalcium phosphate, promotes bone fill, periodontal ligament regeneration, and cementum formation in intrabony periodontal defects and localized alveolar ridge augmentation around dental implants. Augment Bone Graft, approved by the FDA in 2015, incorporates rhPDGF-BB with beta-tricalcium phosphate for use in hindfoot and ankle fusion procedures as an alternative to autograft, supported by a randomized controlled trial showing non-inferior fusion rates (86.4% vs. 89.6% at 12 months). Clinical studies demonstrate that PDGF-BB enhances osteogenesis and vascularization in these matrices, leading to improved implant stability and bone volume compared to bone grafts alone, with bone fill rates up to 4.5 mm in periodontal defects after 6-12 months. Investigational combination therapies pairing PDGF with (VEGF) aim to enhance in ischemic conditions by promoting both endothelial cell (via VEGF) and pericyte recruitment for vessel maturation (via PDGF). Preclinical and early-phase human trials using vectors delivering PDGF-B and VEGF-A have shown prolonged and improved in models of and myocardial ischemia, though no combined formulation is currently approved for clinical use. Delivery methods for therapeutic PDGF include topical formulations like the becaplermin gel for direct application and sustained-release matrices for orthopedic and dental sites, as well as investigational approaches using adenoviral or vectors to express PDGF isoforms locally in ischemic tissues. A phase II randomized, double-blind, -controlled trial in the late 1990s evaluated becaplermin for pressure ulcers, reporting higher healing rates (45% [14/31] complete closure at 16 weeks) versus (13% [4/31]), supporting its potential extension to other chronic wounds beyond diabetic ulcers, though not yet FDA-approved for venous leg ulcers.

Diagnostic and Prognostic Roles

Platelet-derived growth factor (PDGF) and its receptors (PDGFRs) serve as valuable biomarkers in clinical diagnostics and prognostics, particularly in fibrotic and neoplastic conditions. In fibrosis, serum levels of PDGF isoforms, such as PDGF-AA and PDGF-BB, are elevated in systemic sclerosis (scleroderma), reflecting disease activity and fibroblast activation; for instance, bronchoalveolar lavage fluid from scleroderma patients shows significantly higher PDGF-AA and PDGF-BB concentrations compared to controls, correlating with pulmonary involvement. Similarly, in liver fibrosis associated with chronic hepatitis B, serum PDGF-BB levels increase with advancing fibrosis stages and can serve as a non-invasive marker for assessing disease progression. These elevations highlight PDGF's role in monitoring fibrotic burden without invasive procedures. In , (IHC) for PDGFR expression in tumor biopsies aids in grading and . High PDGFR-α and PDGFR-β protein expression is significantly associated with malignant in both adult and pediatric gliomas, with IHC revealing upregulated receptors in higher-grade tumors compared to low-grade ones, supporting its use in histopathological assessment. For prognosis, elevated PDGF-B or PDGFR-β in stroma correlates with increased risk and poorer outcomes; studies from large cohorts demonstrate that high stromal PDGFR-β expression, often driven by PDGF-B ligands, predicts biochemical recurrence and metastatic progression post-prostatectomy. Emerging liquid biopsy approaches leverage circulating PDGF isoforms for non-invasive monitoring of treatment response in cancer. Extracellular vesicles expressing PDGF-β have been identified as potential biomarkers in liquid biopsies from cancer patients, enabling serial assessment of disease dynamics and therapeutic efficacy without tissue sampling. Quantification of PDGF in or typically employs enzyme-linked immunosorbent assay () kits, which provide sensitive and reproducible measurements of isoforms like PDGF-BB, facilitating their integration into routine clinical workflows for and cancer surveillance.

Pathological Implications

Involvement in Diseases

Dysregulated platelet-derived growth factor (PDGF) signaling contributes to the of various cancers through overexpression and autocrine stimulation. In , PDGF-BB forms an autocrine loop that drives tumor and survival by activating PDGF receptor beta (PDGFR-β), promoting the transformed of malignant cells. Similarly, in dermatofibrosarcoma protuberans, a fuses the COL1A1 gene with PDGFB, leading to ectopic production of PDGF-BB that stimulates autocrine and via PDGFR-β, thereby enhancing tumor growth and invasion. In fibrotic diseases, PDGF isoforms play a key role in activation and deposition. Specifically, PDGF-C is upregulated in models, where it activates PDGFR-α on s, inducing their into myofibroblasts and exacerbating fibrotic remodeling through and . In (IPF), PDGF levels in fluid and lung tissue are elevated, correlating with disease severity and hyperactivity. PDGF also promotes vascular pathology in atherosclerosis by stimulating smooth muscle cell proliferation within plaques. Endothelial cells, particularly under disturbed shear stress conditions, secrete PDGF-BB, which acts on PDGFR-β in vascular smooth muscle cells to induce , , and phenotypic , contributing to intimal thickening and plaque instability. Mutations in the PDGF receptor gene PDGFRA underlie certain rare genetic disorders, notably gastrointestinal stromal tumors (GIST). Activating mutations in PDGFRA, occurring in approximately 5-10% of GIST cases, lead to ligand-independent receptor autophosphorylation and downstream signaling that drives uncontrolled of interstitial cells of Cajal, resulting in tumor formation predominantly in the .

Targeted Therapies

Targeted therapies for platelet-derived growth factor (PDGF) signaling primarily involve inhibitors that block PDGF receptors (PDGFRs), aiming to disrupt aberrant activation in diseases such as cancers and . inhibitors (TKIs) represent a cornerstone of these approaches, with (Gleevec) being the first approved TKI targeting PDGFRs. inhibits PDGFRα and PDGFRβ, in addition to other kinases like BCR-ABL and , and was initially approved by the FDA in 2001 for chronic myeloid leukemia (CML) due to its activity against BCR-ABL. Its application expanded to gastrointestinal stromal tumors (GIST) in 2002, where it targets and PDGFRα mutations driving oncogenesis. In PDGFRA-mutant GIST (excluding resistant D842V mutations), at 400 mg daily yields partial response rates of approximately 60-70% and clinical benefit in over 80% of patients, significantly improving compared to historical controls. For PDGFRA exon 18 mutations including D842V, which confer primary resistance to , (Ayvakit) was approved by the FDA in 2020, showing objective response rates of about 84% in clinical trials. Monoclonal antibodies targeting PDGFRs have also been developed, with olaratumab (Lartruvo) as a notable example. Olaratumab is a IgG1 that binds PDGFRα, preventing ligand-induced receptor dimerization and downstream signaling. It received accelerated FDA approval in 2016 for use in combination with in adults with advanced (STS) not amenable to surgery or radiation, based on phase II data showing improved overall survival (median 26.5 months vs. 14.7 months with doxorubicin alone). However, confirmatory phase III trials failed to replicate these benefits, leading to its voluntary withdrawal from the market by the manufacturer in 2019, with FDA concurrence in 2020. Other small molecule multi-kinase inhibitors, such as sunitinib (Sutent), incorporate PDGFR inhibition within broader anti-angiogenic profiles. Sunitinib potently inhibits PDGFRα and PDGFRβ, alongside VEGFRs and KIT, and was approved by the FDA in 2006 for advanced renal cell carcinoma (RCC) following cytokine-refractory disease, based on phase III data demonstrating a median progression-free survival of 11 months versus 5 months with interferon alfa. In metastatic RCC, where PDGF signaling contributes to tumor angiogenesis and stromal support, sunitinib's PDGFR blockade reduces vascular permeability and tumor growth, establishing it as a first-line option until newer immunotherapies supplanted it. Despite these advances, resistance to PDGF-targeted therapies poses significant challenges, often arising from pathway bypass mechanisms. For instance, activating in the PI3K/AKT/ axis, such as PIK3CA gain-of-function variants, can sustain cell survival and independently of PDGFR inhibition, as seen in resistant GIST and RCC models. These hyperactivate downstream effectors, circumventing TKI blockade and leading to disease progression, with preclinical studies highlighting the need for combination strategies targeting PI3K to restore sensitivity. Ongoing clinical efforts focus on refining anti-PDGF strategies for fibrotic conditions, where PDGF drives excessive deposition. has shown promise in early pilot studies for systemic sclerosis ()-associated , with reports of lung function stabilization in over 50% of patients and a trend toward improvement in forced . More recently, phase II trials of novel anti-PDGF agents, including PDGFR-specific antibodies and small molecules, are investigating efficacy in , aiming to halt progression; for example, a multicenter phase II study evaluates PDGFR modulation in early diffuse , with interim data suggesting improved modified Rodnan skin scores. These trials underscore the potential of targeted PDGF inhibition to address unmet needs in antifibrotic therapy.

Current Research

Emerging Findings

Recent studies utilizing single-cell sequencing (scRNA-seq) have highlighted the heterogeneity in PDGF-driven gliomas, revealing distinct subpopulations of cells that contribute to tumor progression and resistance. For instance, in , scRNA-seq analyses have identified PDGF-driven cell states that exhibit varying levels of proliferative and invasive potential, underscoring the role of PDGF signaling in shaping intratumor diversity. Investigations into the role of PDGF in COVID-19-associated lung have demonstrated elevated PDGF levels in patients with severe outcomes, particularly in the development of post-infection pulmonary sequelae. Data from 2021-2023 cohorts indicate that high serum PDGF concentrations, alongside other profibrotic markers like TGF-β, are associated with activation and persistent fibrotic changes in the lungs, contributing to long-term respiratory impairment in survivors. These findings suggest PDGF as a key mediator in the transition from acute to chronic following SARS-CoV-2 infection. Non-canonical functions of PDGF in immune modulation have emerged, particularly its influence on T-cell activation. Recent 2024 papers demonstrate that PDGF-DD isoform binding to NKp44 on plasmacytoid dendritic cells costimulates TLR9 signaling, indirectly enhancing T-cell proinflammatory responses and production in antiviral immunity. This reveals PDGF's broader role beyond activity, extending to of adaptive immune dynamics in inflammatory contexts.

Future Directions

In precision medicine, genotyping for mutations in PDGF receptors offers substantial potential for tailoring inhibitor therapies to individual cancer patients. Activating in PDGFRA, such as the exon 18 D842V variant found in 5-10% of gastrointestinal stromal tumors, enable selection of targeted inhibitors like , which effectively address resistance by binding the inactive receptor conformation. Similarly, rearrangements in myeloid neoplasms with respond to following confirmation, improving remission rates in responsive subtypes. Preclinical investigations in the 2020s have highlighted nanoparticle-based systems as a promising avenue for administering PDGF to promote healing. nanoparticles integrated into electrospun fibers provide sustained PDGF release, enhancing migration and in vitro while accelerating closure in diabetic models. nanospheres co-encapsulating PDGF and VEGF genes further demonstrate improved , formation, and deposition in preclinical diabetic ulcers, underscoring the potential for controlled, localized to overcome degradation challenges in impaired healing environments. The role of PDGF in neurodegeneration remains an area of significant unresolved inquiry, with emerging links to illustrating its complex, context-dependent effects. PDGF-BB exerts neuroprotective actions by safeguarding hippocampal neurons against and energy deprivation through antioxidant pathway activation and mitochondrial preservation, yet reduced PDGFB expression in AD-affected brains correlates with degeneration and blood-brain barrier breakdown, potentially exacerbating amyloid-beta accumulation and vascular dysfunction. These contradictory findings—protective in some models but contributory to pathology in others—necessitate further studies to delineate PDGF's mechanistic contributions to Alzheimer's progression and evaluate its therapeutic modulation. Advancements in are facilitating sophisticated modeling of PDGF signaling networks to accelerate . Hybrid frameworks, incorporating and algorithms like K DEEP, have screened vast compound libraries to identify novel PDGFR inhibitors for non-small cell , predicting binding affinities through molecular fingerprint analysis and dynamics simulations that capture pathway perturbations. By simulating interactions such as hydrogen bonding in the PDGFRA domain, these AI-driven approaches enable precise targeting of oncogenic signaling, reducing experimental timelines and enhancing hit rates for inhibitors. Gene editing of PDGF-related loci in heritable diseases, exemplified by gain-of-function PDGFRB mutations driving familial infantile myofibromatosis, presents profound ethical challenges. CRISPR-Cas9-based germline corrections could mitigate multigenerational transmission of such variants, which cause aggressive vascular tumors, but raise risks of off-target mutations, genetic mosaicism, and unintended phenotypic alterations across generations. Key concerns include obtaining for edited embryos, preventing non-therapeutic enhancements that evoke , and navigating international prohibitions on heritable editing until safety benchmarks are met.

References

  1. [1]
    Platelet-Derived Growth Factor - an overview | ScienceDirect Topics
    PDGF was originally discovered as a constituent of platelets, which are released into serum in conjunction with blood coagulation. Although the α-granules ...
  2. [2]
    Platelet-derived Growth Factor (PDGF) Family - Sino Biological
    Platelet-Derived Growth Factor (PDGF) is a growth factor regulating connective tissue cell growth, including fibroblasts and smooth muscle cells.
  3. [3]
    History of Discovery: Platelet-derived Growth Factor - PubMed Central
    PDGF was discovered while studying atherosclerosis, initially as a growth-promoting activity from activated platelets, and later identified as a factor driving ...
  4. [4]
    (PDF) Platelet-derived growth factor. Structure, function, and roles in ...
    Aug 6, 2025 · PDGF is generally believed to be the principal mitogen that stimulates cell division when vessel integrity has been compromised and platelet ...
  5. [5]
    Structural and functional specificities of PDGF‐C and PDGF‐D, the ...
    Nov 8, 2005 · This review focuses on the structure and function of the two novel members, PDGF-C and -D, of the PDGF subfamily of the cystine knot superfamily.
  6. [6]
  7. [7]
    Platelet alpha granules contain a growth factor for fibroblasts
    The data demonstrate that the alpha granules of platelets provide a unique delivery system for PDGF, a polypeptide hormone with growth-promoting activity for ...
  8. [8]
    Platelet-derived growth factor: purification and partial characterization.
    Platelet-derived growth factor: purification and partial characterization. C H Heldin, B Westermark, and A WastesonAuthors Info & Affiliations. August 15, 1979.
  9. [9]
    Platelet-derived growth factor up-regulates the expression of ...
    It was isolated from human platelets as a protein with a molecular mass of 28–31 kDa [2], [3]. PDGF consists of two polypeptide chains, designated A and B, held ...
  10. [10]
    The Role of Individual Cysteine Residues in the Structure ... - Science
    PDGF, as well as processed forms of the v-sis gene product, is a disulfide-linked dimer with eight conserved cysteine residues in the minimum region necessary ...
  11. [11]
    PDGFA platelet derived growth factor subunit A [ (human)] - NCBI
    Sep 9, 2025 · There are four platelet-derived growth factor (PDGF) genes (PDGFA, PDGFB, PDGFC and PDGFD) that reside on chromosomes 7, 22, 4 and 11. TLR ...
  12. [12]
    PDGFB platelet derived growth factor subunit B [ (human)] - NCBI
    Sep 13, 2025 · The gene that encodes for PDGFB gene is localized on chromosome 22q12.3-q13.1, regions that have reported linkage to schizophrenia. Read ...
  13. [13]
    Platelet-derived growth factors and their receptors: structural ... - NIH
    The PDGF-BB crystal structure has been determined [6] two decades ago. Although the free PDGF-AA structure has not yet been determined, a propeptide-bound ...
  14. [14]
  15. [15]
    Role of platelet-derived growth factors in physiology and medicine
    Platelet-derived growth factor (PDGF) was identified more than three decades ago as a serum growth factor for fibroblasts, smooth muscle cells (SMCs), and glia ...
  16. [16]
    Platelet-Derived Growth Factor (PDGF)/PDGF Receptors (PDGFR ...
    All four PDGF chains contain a highly conserved growth factor domain of approximately 100 amino acids that is also found in the VEGF family. Until now, five ...
  17. [17]
    Two classes of PDGF receptor recognize different isoforms of PDGF
    The B receptor binds only the BB dimer, whereas the A/B receptor binds AA, BB, and AB dimers. Human dermal fibroblasts appear to express seven times as much B ...
  18. [18]
    A PDGF/VEGF homologue provides new insights into the nucleus ...
    Dec 30, 2017 · In this study, a PVF gene, belonging to the PDGF/VEGF family, was cloned and characterized from Pinctada fucata. It contained an ORF of 1110bp ...
  19. [19]
    The vascular endothelial growth factor (VEGF) family
    Phylogenetic analysis of these sequences reveals that the VEGF/PDGF family tree is essentially composed of two branches evolved from a putative common ancestor, ...
  20. [20]
  21. [21]
    Structure of full-length human PDGFRβ bound to its activating ligand ...
    In this regard, this paper provides an initial structural characterization of a complex consisting of PDGF-B bound to its cognate receptor PDGFRβ in its ...
  22. [22]
    Structures of a platelet-derived growth factor/propeptide ... - PNAS
    Jun 2, 2010 · PDGFRs belong to the class III receptor tyrosine kinases (RTKs), a five-member clan that includes PDGFRα, PDGFRβ, KIT, FMS, and FLT3, which are ...
  23. [23]
    Pericytes of multiple organs do not behave as mesenchymal stem ...
    Mar 2, 2018 · Here we show that, although in adulthood PDGFRβ expression is largely restricted to pericytes and vascular smooth muscle, during embryogenesis ...
  24. [24]
    Pericyte-specific expression of PDGF beta receptor in mouse ...
    Aug 25, 2010 · We show for the first time that PDGFRβ is exclusively expressed in pericytes, and not in neurons, astrocytes or endothelial cells, in the adult brain of ...
  25. [25]
  26. [26]
  27. [27]
  28. [28]
    Role of platelet-derived growth factors in physiology and medicine
    Platelet-derived growth factor (PDGF) was identified more than three decades ago as a serum growth factor for fibroblasts, smooth muscle cells (SMCs), and glia ...
  29. [29]
    Mice deficient for PDGF B show renal, cardiovascular, and ...
    We show here that mice deficient for PDGF B die perinatally and display several anatomical and histological abnormalities. Kidney glomerular tufts do not form, ...
  30. [30]
    PDGF function in diverse neural crest cell populations - PMC - NIH
    In this review, we examine the current knowledge of PDGF function during neural crest cell (NCC) development, focusing on its role in the formation of different ...
  31. [31]
    PDGF signalling controls the migration of mesoderm cells during ...
    Nov 1, 2008 · PDGF signalling controls N-cadherin expression on mesoderm cells, which is required for efficient migration.Knockdown Of Pdgfa Impairs... · Pdgfa Can Act As An... · Pdgf Signalling Controls...
  32. [32]
    Role of platelet-derived growth factor in wound healing - PubMed
    PDGF stimulates chemotaxis, proliferation, and new gene expression in monocytes-macrophages and fibroblasts in vitro, cell types considered essential for tissue ...
  33. [33]
    Role of platelet-derived growth factor in wound healing - NIH
    PDGF, delivered by platelets to the site of injury in vivo, may play an important role in the initiation of the wound repair process.
  34. [34]
    Platelet isoforms of platelet-derived growth factor stimulate ...
    Platelets and monocyte/macrophages accumulate at these sites and release a variety of growth factors that are thought to initiate and sustain the repair.
  35. [35]
    From Growth Factors to Structure: PDGF and TGF-β in Granulation ...
    ... PDGF-BB stimulate fibroblast proliferation and extracellular matrix synthesis, which is crucial for tissue repair. TGFβ, with its three isoforms, influences ...
  36. [36]
    PDGF-D induces macrophage recruitment, increased interstitial ...
    PDGF-D induces macrophage recruitment, increased interstitial pressure, and blood vessel maturation during angiogenesis.
  37. [37]
    Stromal PDGFR-α Activation Enhances Matrix Stiffness, Impedes ...
    Mammary fibroblasts with PDGFRα hyperactivation also decreased hydraulic ... interstitial fluid pressure (IFP). Our study suggests that this increase ...
  38. [38]
    Pdgfrα+ stromal cells, a key regulator for tissue homeostasis and ...
    These findings reveal the muscle-resident Pdgfrα+ stromal cells play a central role in maintaining muscle homeostasis. Besides muscles, Pdgfrα+ stromal cells ...<|separator|>
  39. [39]
    Adenoviral Mediated Gene Transfer of PDGF-B Enhances Wound ...
    The vulnerary effect of adenoviral mediated gene therapy with PDGF-B significantly enhanced wound healing and neovascularization in diabetic wounds.
  40. [40]
    Stabilization of HIF-1α is critical to improve wound healing in ...
    Hypoxia has been shown to induce major cytokines (VEGF, TGF-β, PDGF), to stimulate proliferation and migration of fibroblasts and keratinocytes (recently ...
  41. [41]
    Hypoxia Enhances Platelet-derived Growth Factor Signaling in the ...
    Nov 4, 2009 · Hypoxia-inducible factor (HIF)-1α is involved in this regulation of gene expression, because hypoxia-induced βPDGFR hyperphosphorylation and PTP ...<|separator|>
  42. [42]
    Efficacy and safety of becaplermin (recombinant human ... - PubMed
    Becaplermin gel-100 microg/g significantly decreased (p = 0.01) the time to complete healing compared with placebo gel, with the 35th percentile of time to ...
  43. [43]
    Elevated levels of platelet derived growth factor and transforming ...
    Results: SSc BAL fluid contains significantly elevated levels of PDGF-AA and PDGF-BB. Where TGF-beta 1 was significantly elevated in SSc lavage fluid, the ...
  44. [44]
    a potential biomarker for the assessment of liver fibrosis in patients ...
    Serum PDGF-BB decreased remarkably as fibrosis progressed, and this could be used as a non-invasive biomarker for the assessment of fibrosis stage in ...
  45. [45]
    Protein expression of platelet-derived growth factor receptor ...
    Jun 1, 2008 · High PDGFR protein expression is significantly associated with malignant histology in pediatric gliomas, but it does not represent an ...
  46. [46]
    Platelet-derived growth factor and its receptors in human glioma tissue
    Jun 1, 1992 · The expression of platelet-derived growth factor (PDGF) and its receptors was analyzed in 14 gliomas of various degrees of malignancy and ...
  47. [47]
    High Expression of PDGFR-β in Prostate Cancer Stroma ... - PubMed
    Feb 24, 2017 · Herein, we aimed to investigate the prognostic impact of PDGFR-β and its ligands (PDGF-B and PDGF-D) in a multicenter prostatectomy cohort of ...
  48. [48]
    PDGF receptors in tumor biology: prognostic and predictive potential
    High stromal PDGFRβ expression or activation is associated with poor prognosis in breast and prostate cancer. ... β in malignant cells of common epithelial tumor ...
  49. [49]
    Identification and validation of the surface proteins FIBG, PDGF-β ...
    Aug 1, 2024 · Extracellular vesicles (EVs) have emerged as promising candidates for cancer monitoring using liquid biopsy. However, the complexity of EVs ...
  50. [50]
    Normal ranges of angiogenesis regulatory proteins in human platelets
    We introduce a new ELISA-based method for accurate and reproducible measurement of vascular endothelial growth factor (VEGF), platelet-derived growth factor ( ...
  51. [51]
    Activated platelet-derived growth factor autocrine pathway drives the ...
    The interruption of the autocrine loop by the PDGF antibodies reversed the transformed phenotype of the glioblastoma cell, as determined by (1) diminished DNA ...
  52. [52]
    The dermatofibrosarcoma protuberans-associated ... - PubMed
    We conclude that the COLIA1/PDGFB fusion associated with DFSP contributes to tumor development through ectopic production of PDGF-BB and the formation of an ...
  53. [53]
  54. [54]
    Targeting Progression in Pulmonary Fibrosis: An Overview of ...
    Concentrations of PDGF have also been found to be increased in BALF and lung samples from patients with IPF [37]. PDGF, secreted by activated platelets and ...
  55. [55]
    Shear stress-stimulated endothelial cells induce smooth ... - PubMed
    Conclusion: Shear stress stimulates endothelial cells to secrete several SMC chemoattractants, including PDGF-BB and IL-1alpha; both PDGF-BB and IL-1alpha ...
  56. [56]
    KIT and PDGFRA mutations in gastrointestinal stromal tumors (GISTs)
    PDGFRA mutations usually occur in gastric GISTs, especially in the epithelioid variants; their overall frequency is approximately 30% to 40% of KIT mutation ...
  57. [57]
    Approval summary: imatinib mesylate in the treatment of metastatic ...
    On February 1, 2001, imatinib mesylate was approved by the United States Food and Drug Administration for the treatment of malignant metastatic and/or ...Missing: PDGFR | Show results with:PDGFR
  58. [58]
    [PDF] 2927153 This label may not be the latest approved by FDA. For ...
    The pharmacokinetics of Gleevec are similar in CML and GIST patients. Imatinib is well absorbed after oral administration with Cmax achieved within 2-4 hours ...
  59. [59]
    Efficacy of Imatinib in Patients with Platelet-Derived Growth Factor ...
    Significantly, poorer PFS was observed for patients with D842V-mutant GISTs than those with non-D842V PDGFRA-mutant GISTs: median 3.8 months (95% CI, 1.4 to 6.3) ...
  60. [60]
    Efficacy of Imatinib in Patients with Platelet-Derived Growth Factor ...
    Significantly, poorer PFS was observed for patients with D842V-mutant GISTs than those with non-D842V PDGFRA-mutant GISTs: median 3.8 months (95% CI, 1.4 to 6.3) ...
  61. [61]
    [PDF] LARTRUVO (olaratumab) injection, for intravenous use
    LARTRUVO™ is indicated, in combination with doxorubicin, for the treatment of adult patients with soft tissue sarcoma (STS) with a histologic subtype for which ...
  62. [62]
    New Treatment Approved for Soft Tissue Sarcoma - NCI
    Nov 2, 2016 · UPDATE: On October 2, 2019, olaratumab (Lartruvo) was officially withdrawn from the market, according to a posting on the Food and Drug ...Missing: anti- PDGFRα
  63. [63]
    Prices and Trends in FDA-Approved Medications for Sarcomas - MDPI
    Apr 18, 2024 · The asterisk represents olaratumab (indicated for advanced soft tissue sarcoma), which was FDA-approved in 2016 and withdrawn in 2020. Table ...
  64. [64]
    Sunitinib versus Interferon Alfa in Metastatic Renal-Cell Carcinoma
    Jan 11, 2007 · We report on the results of a randomized, phase 3 trial of sunitinib, as compared with interferon alfa, as first-line treatment of metastatic renal-cell ...
  65. [65]
    Sunitinib in the treatment of metastatic renal cell carcinoma - PMC
    Sunitinib received approval in 2006 and became a standard treatment option in the first-line treatment of metastatic renal cell cancer (mRCC)
  66. [66]
    Mechanisms of sunitinib resistance in renal cell carcinoma and ...
    Sep 23, 2023 · Sunitinib could inhibit the angiogenesis or tumour cell proliferation in advanced renal cell carcinoma. ... VEGF and PDGF then promote ...
  67. [67]
    Sunitinib Acts Primarily on Tumor Endothelium rather than Tumor ...
    Jan 31, 2010 · Sunitinib acts primarily on tumor endothelium rather than tumor cells to inhibit the growth of renal cell carcinoma.
  68. [68]
    Protein tyrosine kinase inhibitor resistance in malignant tumors
    Sep 17, 2022 · PTK can develop secondary mutations during treatment with TKI, leading to acquired drug resistance. The gatekeeper mutation is one of the first ...
  69. [69]
    Mechanisms of Resistance to PI3K Inhibitors in Cancer
    Mutations causing the loss or inactivation of PTEN have been shown to lead to the development of cancer due to the hyperactivation of the PI3K pathway [19].
  70. [70]
    Targeting PI3K/Akt signal transduction for cancer therapy - Nature
    Dec 16, 2021 · Members of the PI3K/Akt/mTOR pathway are often mutated and activated in cancer. The study of PI3K/Akt networks has led to the discovery of ...
  71. [71]
    PDGFRs are critical for PI3K/Akt activation and negatively regulated ...
    To examine the potential role of PDGFR in EGF-mediated Akt activation, we used AG1478, an EGFR inhibitor, and AG1295, a PDGFR inhibitor, to treat WT MEFs.
  72. [72]
    Low-Dose Oral Imatinib for Scleroderma Pulmonary Involvement
    The purpose of this study is to verify effect and tolerability of imatinib on pulmonary and skin fibrosis in patients affected by systemic sclerosis. Detailed ...
  73. [73]
    Drugs in Phase I and Phase II Clinical Trials for Systemic Sclerosis
    This review summarizes investigational agents in recent Phase I and II clinical trials evaluated for the treatment of SSc, with a focus on skin in patients ...Table 2 · 2.1. Biologics · 2.2. Anti-Fibrotic Therapy<|control11|><|separator|>
  74. [74]
    PDGF/PDGFR: A Possible Molecular Target in Scleroderma Fibrosis
    Mar 31, 2022 · In this review: (i) we focused on the PDGF/PDGFR pathway as key signaling molecules in the development of tissue fibrosis;3. Pdgf Receptor (pdgfr) · 6. Anti-Pdgfrα... · 8. Pdgf/pdgfr Targeting...
  75. [75]
    An update on targeted therapies in systemic sclerosis based on a ...
    Jun 1, 2021 · We aimed to perform a systemic literature review of phase 1, phase 2 and phase 3 clinical trials and large observational studies about targeted therapies in ...
  76. [76]
    Single-Cell RNA Sequencing Reveals Heterogeneity in the Tumor ...
    Single-cell RNA sequencing reveals heterogeneity in the tumor microenvironment between young-onset and old-onset colorectal cancer.Missing: PDGF | Show results with:PDGF
  77. [77]
    The Interaction Between Pulmonary Fibrosis and COVID-19 ... - NIH
    The risk factors of idiopathic pulmonary fibrosis and COVID-19 have a lot in common, such as age, sex, smoking history, hypertension, diabetes, obesity, and so ...
  78. [78]
    Recent progress on the role of miR-140 in cartilage matrix ...
    Aug 18, 2020 · miR-140 can act on different targets to regulate chondrocyte differentiation and OA development at different stages. The effect of miR-140 on ...
  79. [79]
    [PDF] Analysis of PDGF receptor internalization and signaling using ...
    For PDGFRβ, crystal structures exist for domains D1-2-3 in complex with ligand [75], for D4-D5. [76], and cryo-EM of the full length receptor [77]. Much ...
  80. [80]
    PDGF-DD Binding to NKp44 Costimulates TLR9 Signaling and ...
    Feb 1, 2024 · Cutting Edge: PDGF-DD Binding to NKp44 Costimulates TLR9 Signaling and Proinflammatory Cytokine Secretion in Human Plasmacytoid Dendritic Cells.
  81. [81]
    PDGF receptor mutations in human diseases - PMC - PubMed Central
    Jan 15, 2021 · Early studies suggested that PDGF ligands and receptors may play a role in cancer development, and genetic alterations in both receptor genes ...
  82. [82]
    Growth factors and growth factor gene therapies for treating chronic ...
    Dec 28, 2023 · This review provides an overview of chronic wound healing, emphasizing the need and potential for growth factor therapies.
  83. [83]
    Insights Into the Role of Platelet-Derived Growth Factors - Frontiers
    Platelet-derived growth factor regulates the functional activities of neurons by regenerating, stabilizing, and stimulating the synapses of neuronal axons, ...
  84. [84]
    Discovery of novel PDGFR inhibitors targeting non-small cell lung ...
    Jan 10, 2025 · In this research article, we focus on the application of Machine Learning, alongside the RDKit toolkit, to identify potential anti-cancer drug candidates ...
  85. [85]
    PDGFRB gain-of-function mutations in sporadic infantile ...
    In conclusion, we identified gain-of-function PDGFRB mutations in the majority of multifocal infantile myofibromatosis cases, shedding light on the mechanism of ...
  86. [86]
    Bioethical issues in genome editing by CRISPR-Cas9 technology
    Some of the ethical dilemmas of genome editing in the germline arise from the fact that changes in the genome can be transferred to the next generations.Missing: PDGF | Show results with:PDGF