Fact-checked by Grok 2 weeks ago

PDGFRB

PDGFRB is a located on chromosome 5q32 that encodes receptor beta (PDGFRβ), a transmembrane that binds members of the (PDGF) family to regulate critical cellular processes including proliferation, migration, differentiation, and survival, particularly in mesenchymal-derived cells and vascular structures. The PDGFRβ protein consists of an extracellular domain with five immunoglobulin-like domains for ligand binding, a single transmembrane helix, and an intracellular domain that, upon PDGF-induced dimerization (primarily PDGF-BB homodimers), autophosphorylates and activates downstream signaling pathways such as PI3K/AKT and MAPK/ERK, which control cytoskeletal reorganization and . Expressed widely in , fibroblasts, and cells, PDGFRβ plays a pivotal role in embryonic development of the cardiovascular system, including formation and blood-brain barrier integrity, as evidenced by pericyte-deficient models exhibiting microaneurysms and barrier defects. Dysregulation of PDGFRB through somatic mutations, fusions (e.g., ETV6-PDGFRB in chronic eosinophilic leukemia), or variants is implicated in several disorders; for instance, loss-of-function mutations such as L658P cause (IBGC4) through impaired signaling that disrupts vascular integrity and leads to calcium deposits in the , while activating variants such as R561C underlie infantile myofibromatosis (IMF1), characterized by multifocal tumors in infancy. Overgrowth syndromes like Kosaki syndrome (e.g., P584R mutation) and Penttinen-type premature aging (e.g., V665A or N666S) result from gain-of-function changes that enhance signaling, often with temperature-sensitive effects leading to phenotypes including , , and connective tissue abnormalities. In oncology, PDGFRB fusions drive and Philadelphia-like , rendering cells sensitive to tyrosine kinase inhibitors like . These associations underscore PDGFRβ's therapeutic potential, with inhibitors targeting it in cancers and fibrotic conditions such as .

Genetics

Gene Location and Organization

The PDGFRB gene is located on the long arm of human chromosome 5 at cytogenetic band 5q32, specifically spanning positions 150,113,839 to 150,155,845 (GRCh38.p14 assembly). It encompasses approximately 42 kb of genomic DNA and comprises 24 exons, with the majority encoding the functional protein domains. This gene is flanked by CSF1R (encoding the macrophage colony-stimulating factor receptor) on one side and CSF2 (encoding granulocyte-macrophage colony-stimulating factor) on the other; interstitial deletions in this 5q32 region, often involving these neighboring loci, are associated with myelodysplastic syndromes such as the 5q- syndrome. The official symbol PDGFRB was assigned by the (HGNC:8804), with historical aliases including PDGFR, PDGFR1, and PDGFR-1, reflecting its identification as a receptor isoform. PDGFRB exhibits strong evolutionary conservation across mammals, with orthologs present in over 240 species including , , and ; the human protein sequence shares 86% identity overall with the ortholog (Pdgfrb), rising to 98% in the cytoplasmic domain, underscoring its essential role in and signaling.

Expression and Regulation

The PDGFRB gene exhibits tissue-specific expression primarily in mesenchymal-derived types, including , vascular cells, fibroblasts, and other mesenchymal cells. In adult tissues, PDGFRB is prominently expressed in surrounding capillaries and small vessels, as well as in vascular cells of larger arteries, where it supports vascular stability and remodeling. Fibroblasts in connective tissues, such as those in the interstitium and , also show robust PDGFRB expression, contributing to production and processes. During embryonic , this expression pattern is conserved and expanded to broader mesenchymal populations, essential for . Expression of PDGFRB peaks during embryonic vascular formation, particularly between embryonic days 9.5 and 12.5 in mice, coinciding with the recruitment and proliferation of and vascular cells to nascent endothelial tubes. This temporal regulation ensures proper investment of blood vessels, preventing hemorrhage and supporting organ vascularization; disruptions lead to embryonic lethality due to vascular defects. Postnatally, expression levels decline but persist in perivascular niches to maintain vascular . Transcriptional regulation of PDGFRB is mediated by key factors binding to its promoter region. The transcription factor Sp1 binds to GC-rich motifs in the proximal promoter, driving basal expression and responding to growth signals in mesenchymal cells. In hypoxic conditions relevant to developmental vascular niches, hypoxia-inducible factors (HIFs), particularly HIF-1α, indirectly influence PDGFRB transcription through enhancement of related pathways, though direct binding is limited. Other regulators, such as TGFβ signaling via Smad proteins, activate the PDGFRB promoter by cooperating with Sp1 sites, fine-tuning expression in response to extracellular cues. Evidence for AP-2 involvement remains context-specific and less direct in mesenchymal regulation. Epigenetic mechanisms further control PDGFRB expression, with promoter playing a critical role in silencing or modulating activity. Hypermethylation of CpG islands in the PDGFRB promoter, observed in certain populations, correlates with reduced expression, while hypomethylation permits tissue-specific activation in and cells. modifications, including of H3K9 and of H3K4, facilitate open states at the promoter, enhancing accessibility for transcriptional machinery during developmental peaks. These epigenetic marks are dynamically altered in response to environmental signals, ensuring precise spatiotemporal control.

Protein Structure

Domain Architecture

The platelet-derived growth factor receptor beta (PDGFRB) is a transmembrane composed of 1,106 , with the mature, glycosylated form exhibiting a of approximately 190 kDa. This III features a modular architecture typical of its family, enabling recognition, across the membrane, and intracellular events. The extracellular region consists of five immunoglobulin-like (Ig-like) domains, designated Ig-loops 1 through 5, which mediate interactions with PDGF ligands. These domains are followed by a single α-helical transmembrane segment spanning approximately 25 . The intracellular domain encompasses a juxtamembrane , a bilobed domain interrupted by a flexible kinase insert of about 100 , and a C-terminal regulatory tail. Structural insights into PDGFRB have been derived from several (PDB) entries, including the unphosphorylated kinase domain homologs and ligand-bound extracellular fragments. For instance, PDB entry 3MJG reveals the complex of PDGF-BB with Ig-domains 1-3, highlighting the ligand-binding interface, while PDB entry 2L6W provides details on the transmembrane helix dimerization. These structures underscore the receptor's ability to undergo conformational changes upon activation. In comparison to PDGFRα, which shares overall architectural similarity, PDGFRB exhibits beta-specific features in its extracellular region, particularly in Ig-loop 4, that confer preferential binding specificity for PDGF-BB and PDGF-DD homodimers over PDGF-AA. This distinction arises from variations in the dimerization-influencing Ig-loop 4, contributing to the receptors' differential affinities.

Key Functional Sites

The receptor beta (PDGFRB) features several critical autophosphorylation sites within its intracellular domain that regulate its activity and downstream signaling. The residue at position 857 (Tyr857), located in the activation loop of the domain, undergoes to enhance the receptor's catalytic activity upon binding. Additionally, Tyr740 and Tyr751, situated in the kinase insert region, serve as docking sites for the p85 regulatory subunit of 3-kinase (PI3K), facilitating its recruitment and activation of the PI3K/AKT pathway. Additionally, Tyr1009 and Tyr1021 in the C-terminal tail serve as docking sites for gamma (PLCγ), initiating PLCγ-mediated signaling. These sites are essential for the receptor's activation and have been extensively characterized in studies demonstrating their role in efficiency. Glycosylation in the extracellular domains of PDGFRB modulates its stability, binding , and trafficking. The receptor contains three experimentally verified N-linked glycosylation sites at residues (Asn104, Asn193, and Asn373), which contribute to proper folding and maturation in the . These modifications are crucial for maintaining the structural integrity of the five immunoglobulin-like domains, preventing aggregation, and ensuring cell surface expression. has also been reported at a single site (Thr180), further influencing receptor dynamics. The intracellular kinase domain of PDGFRB is interrupted by a 100-amino-acid insert (residues 833-933), a characteristic feature of class III receptor s that separates the ATP-binding and substrate-binding lobes. This insert harbors regulatory motifs, including the aforementioned autophosphorylation sites, and influences kinase autoinhibition. The ATP-binding pocket, conserved among tyrosine kinases, is formed by residues in the N-lobe (e.g., Gly loop at 595-600 and β-sheet structures), enabling nucleotide binding and phosphate transfer; structural analyses reveal its accessibility to competitive inhibitors like . Ubiquitination motifs, primarily involving residues in the juxtamembrane region (e.g., Lys608) and C-terminal tail, are targeted by ligases such as c-Cbl and Cbl-b following activation, promoting receptor and lysosomal degradation to attenuate signaling. Disease-associated variants in PDGFRB can disrupt these functional sites, leading to pathological dysregulation. For instance, the variant p.Arg987Trp, located in the C-terminal autoinhibitory , results in reduced steady-state protein levels and impaired activity, contributing to conditions like through diminished receptor signaling and altered function. Functional assays show this substitution does not directly abolish autophosphorylation but indirectly affects output via protein instability.

Activation and Ligand Binding

Ligand Interactions

The receptor beta (PDGFRB), also known as PDGFR-β, primarily interacts with specific isoforms of (PDGF), including the homodimers PDGF-BB and PDGF-DD, as well as the heterodimer PDGF-AB. These ligands bind with high affinity, typically in the range of 0.1-1 nM (Kd), enabling potent activation of PDGFRB homodimers (ββ) or heterodimers (αβ). For instance, PDGF-BB exhibits a Kd of approximately 0.2-0.5 nM for PDGFRB, while PDGF-DD shows similar high-affinity binding primarily to ββ complexes. In contrast, PDGFRB does not respond to PDGF-AA, which selectively binds PDGFRα (αα homodimers), thereby distinguishing the ligand specificity of PDGFRB from its alpha counterpart and contributing to isoform-specific signaling in cellular contexts. binding occurs primarily within the immunoglobulin-like (Ig-like) domains 2 and 3 (D2 and D3) of the extracellular region of PDGFRB, where the bivalent nature of PDGF isoforms facilitates simultaneous engagement of two receptor molecules. This bivalent interaction is essential for stabilizing receptor dimerization and subsequent activation, as monovalent binding fails to induce the conformational changes necessary for signaling. In physiological settings, PDGF ligands can be presented to PDGFRB in either soluble or matrix-bound forms, influencing the spatial and temporal dynamics of receptor activation. Soluble PDGF-BB, released from platelets or other cells, allows for rapid, diffusible signaling, whereas matrix-bound PDGF (e.g., via retention motifs interacting with proteoglycans in the ) promotes localized, sustained presentation that may enhance recruitment or tissue remodeling. This dual presentation mode underscores the versatility of PDGFRB in responding to varying microenvironments without altering core binding affinity.

Dimerization and Phosphorylation

Upon binding of dimeric PDGF ligands such as PDGF-BB, which induces homodimerization of PDGFRB, or PDGF-AB, which promotes heterodimerization with PDGFRα, the extracellular domains of the receptors assemble into a stable complex, juxtaposing their intracellular domains. This ligand-induced dimerization is essential for receptor activation and occurs through an extensive interface spanning the length of the receptor ectodomains, as revealed by cryo-electron microscopy structures of the PDGFRB/PDGF-BB complex. The process is triggered by PDGF ligands (detailed in the Ligand Interactions section) and results in conformational changes that enable trans-interactions between the kinase domains. Dimerization facilitates asymmetric kinase activation, wherein one PDGFRB molecule allosterically stimulates the kinase activity of its partner, initiating a sequential autophosphorylation cascade. The cascade commences with trans-autophosphorylation at Tyr857 within the activation loop of the kinase domain, which induces a conformational shift to unlock the and enhance catalytic efficiency. This initial event enables subsequent cis- and trans-autophosphorylation of additional residues, including Tyr740, Tyr751 in the juxtamembrane and kinase insert regions, and Tyr1009, Tyr1021 in the C-terminal tail, thereby generating high-affinity docking sites for downstream signaling molecules without directly detailing their recruitment. Constitutive activation of PDGFRB can bypass dependence through mechanisms such as point that disrupt autoinhibitory interactions or juxtamembrane leading to ligand-independent dimerization and autophosphorylation. Overexpression of PDGFRB, often observed in malignancies, similarly promotes spontaneous dimerization and sustained activity. Negative regulation occurs primarily through protein tyrosine phosphatases, such as PTPRB, which dephosphorylates PDGFRB to terminate signaling and prevent excessive activation. Other phosphatases like PTP1B and DEP-1 (PTPRJ) contribute to this dephosphorylation, ensuring tight control over receptor responsiveness.

Downstream Signaling Pathways

Core Signaling Cascades

Upon activation, PDGFRB initiates several core intracellular signaling cascades through autophosphorylation of specific residues, which serve as docking sites for downstream effectors. These pathways primarily regulate , survival, , and , with distinct molecular mechanisms for each. The key cascades include the PI3K/AKT/mTOR, MAPK/ERK, PLCγ, and STAT5 pathways, each recruited via unique phosphotyrosine motifs on the receptor. The PI3K/AKT/ pathway is activated when PDGF-BB binding to PDGFRB induces autophosphorylation at residues 740 and 751 (Tyr740 and Tyr751) in the kinase insert domain. These sites recruit the p85 regulatory subunit of PI3K via its SH2 domains, leading to activation of the p110 catalytic subunit and production of PIP3. PIP3 then recruits and activates AKT (also known as PKB) at the plasma membrane through PDK1-mediated phosphorylation, with subsequent complex activation promoting protein synthesis and inhibiting . This cascade is critical for enhancing cell survival, growth, and metabolic reprogramming in mesenchymal and vascular cells. The MAPK/ERK cascade is triggered through recruitment of the adaptor protein to phosphorylated Tyr716 in the kinase insert region of PDGFRB. Grb2 binds via its and associates with , a that activates by promoting GDP-to-GTP exchange. Activated Ras then stimulates the Raf-MEK-ERK module: Raf phosphorylates and activates MEK1/2, which in turn phosphorylates ERK1/2. Nuclear translocation of phosphorylated ERK1/2 induces transcription factors such as Elk-1 and c-Fos, driving programs that promote and differentiation. This pathway is essential for mitogenic responses in fibroblasts and cells. PLCγ activation occurs via binding to phosphorylated Tyr1009 and Tyr1021 in the C-terminal tail of PDGFRB. Upon recruitment through its SH2 domains, PLCγ hydrolyzes (PIP2) into inositol 1,4,5-trisphosphate (IP3) and diacylglycerol (DAG). IP3 diffuses to the to trigger calcium release via IP3 receptors, elevating cytosolic Ca²⁺ levels, while DAG remains membrane-bound and activates conventional and novel isoforms of (PKC). The resulting Ca²⁺/PKC signaling modulates cytoskeletal dynamics, enzyme activation, and short-term cellular responses such as contraction and secretion. In hematopoietic contexts, PDGFRB directly phosphorylates and activates STAT5, particularly through fusion proteins like TEL-PDGFRB that constitutively signal in myeloid cells. Phosphorylation occurs at tyrosine residues such as Tyr1009, allowing STAT5 SH2 domain-mediated dimerization, nuclear translocation, and binding to specific DNA motifs. This induces transcription of genes involved in cell survival and proliferation, such as Bcl-xL and cyclin D1, supporting megakaryocyte and erythroid lineage expansion. STAT5 activation is prominent in PDGFRB-driven myeloproliferative disorders.

Pathway Crosstalk and Feedback

PDGFRB signaling exhibits significant crosstalk with the VEGF and pathways, particularly in the context of , where shared activation of the PI3K/Akt/ cascade amplifies vascular responses. In vascular development and tumor , PDGF-BB binding to PDGFRB promotes recruitment and vessel stabilization through PI3K-dependent mechanisms, which overlap with VEGF-induced endothelial via VEGFR2. Similarly, PDGF/PDGFRB can bypass EGFR inhibition by upregulating VEGFR1 and VEGFR2 expression, sustaining PI3K/Akt signaling to support angiogenic processes in models. This integration allows PDGFRB to compensate for VEGFR or EGFR blockade, enhancing endothelial cell survival and migration through common downstream effectors like Akt. A key self-regulatory mechanism in PDGFRB signaling involves negative feedback through RasGAP binding to phosphorylated Tyr771, which attenuates activation and thereby inhibits the MAPK/ERK pathway. This phosphorylation event, more pronounced in confluent cells, limits excessive mitogenic signaling by promoting GTP on , ensuring controlled cellular responses to PDGF-BB . In dense cultures, elevated Tyr771 enhances RasGAP recruitment, reducing MAPK activity and shifting emphasis from to other outcomes like . PDGFRB also integrates with integrin signaling to facilitate cytoskeletal rearrangements essential for cell motility and adhesion. At focal adhesions, PDGFRB colocalizes with β1 , where Src family kinases mediate cooperative signaling that reorganizes actin filaments and promotes lamellipodia formation in response to PDGF-BB. This synergy enhances focal adhesion kinase (FAK) activation, linking cues to PDGFRB-driven cytoskeletal dynamics without requiring direct PDGFRB- binding. The dominance of PDGFRB-activated pathways varies by , reflecting context-dependent regulation. In fibroblasts, PI3K/Akt signaling predominates, driving and through PDGF-BB-induced Akt and synthesis. In contrast, exhibit preferential pathway activation upon PDGFRB stimulation, supporting and survival via STAT5 , as observed in hematopoietic models. These differences arise from cell-specific adaptor expression and underscore PDGFRB's role in tailored physiological responses.

Physiological Roles

Developmental Functions

PDGFRB plays a critical role in embryonic vascular development by mediating the recruitment of to endothelial tubes, primarily through signaling induced by its PDGF-BB. This process is essential for stabilizing nascent blood vessels, promoting pericyte , , and , and preventing microvascular instability. Genetic disruption of PDGFRB leads to reduced pericyte coverage, resulting in hyperbranched, dilated vessels prone to microaneurysms. Mice lacking PDGFRB exhibit perinatal lethality due to widespread microvascular defects, including pericyte deficiency and hemorrhage, particularly in the , , and other organs. This underscores PDGFRB's indispensable function in forming mature microvasculature during embryogenesis. In the , PDGFRB is vital for glomerulogenesis, where PDGF-B secreted by endothelial cells recruits PDGFRB-expressing mesenchymal progenitors to develop into , ensuring proper glomerular tuft formation and structural integrity. PDGFRB also contributes to pulmonary vascular maturation during development, supporting pericyte investment in alveolar capillaries essential for infrastructure. In craniofacial development, PDGFRB regulates the migration and proliferation of neural crest-derived mesenchymal cells, forming functional homodimers and heterodimers with PDGFRα to coordinate signaling for proper . Ablation of PDGFRB in the neural crest lineage results in subtle defects such as widened nasal septa and delayed palatal shelf elevation, while combined loss with PDGFRα exacerbates malformations including clefting. These interactions highlight PDGFRB's role in maintaining mesenchymal proliferation beyond initial neural crest migration, ensuring accurate and structure formation.

Tissue Homeostasis and Cellular Processes

PDGFRB signaling is essential for regulating vascular cell (VSMC) contractility, thereby maintaining vascular and tissue in adult tissues. Activation of PDGFRB by PDGF-BB ligands modulates VSMC , allowing cells to balance contractile functions—characterized by expression of markers like heavy chain—with adaptive responses to hemodynamic demands. This regulation occurs via pathways such as p38 MAPK, which support VSMC adaptation without disrupting steady-state vessel tone. In fibroblasts, PDGFRB promotes the controlled production of () components, including and , which are vital for preserving tissue architecture and mechanical integrity during . High PDGFRB expression on fibroblast surfaces enables responsiveness to PDGF-BB, driving synthesis and deposition in a manner that supports structural maintenance rather than excessive accumulation. Knockdown studies demonstrate that PDGFRB is required for up to 90% of production in these cells, underscoring its role in balanced matrix . PDGFRB facilitates by enhancing , , and , particularly in and , to ensure timely tissue repair. PDGF-BB binding to PDGFRB stimulates fibroblast migration into wound beds, with in vitro assays showing increased closure rates, while models reveal reduced recruitment and delayed healing upon PDGFRB inhibition. is similarly promoted, as evidenced by significant decreases in BrdU-positive cells when signaling is blocked, and is supported through anti-apoptotic effects via PI3K/Akt activation. These functions are enabled by core downstream signaling cascades, including MAPK pathways. Pericytes expressing PDGFRB provide critical coverage to cerebral endothelial cells, thereby upholding blood-brain barrier () integrity and restricting paracellular permeability in adult brains. PDGF-BB from endothelial sources recruits PDGFRB-positive , which stabilize vessels by promoting tight junction proteins like claudin-5 and , while suppressing markers such as Plvap. Reduced coverage in PDGFRB-deficient models leads to increased BBB leakage, highlighting its necessity for maintaining barrier function under physiological conditions. PDGFRB contributes to tissue remodeling after injury by orchestrating fibroblast and pericyte activities that promote resolution and prevent dysregulated ECM buildup, such as fibrosis. In normal repair responses, PDGFRB-driven recruitment enhances fibroplasia and angiogenesis, facilitating connective tissue reorganization without scarring, as seen in rodent wound models where balanced signaling supports efficient healing. This remodeling ensures restoration of tissue function while limiting excessive matrix deposition.

Pathological Roles

Involvement in Cancer

PDGFRB overexpression has been implicated in promoting in several malignancies, including gliomas, sarcomas, and gastrointestinal stromal tumors (GIST). In gliomas, elevated PDGFRB signaling, often driven by PDGF-B ligand, enhances (VEGF) expression in tumor endothelial cells and recruits to neovessels, thereby supporting tumor vascularization and growth. Similarly, in sarcomas, PDGFRB overexpression correlates with increased tumor aggressiveness and poor , contributing to angiogenic processes through stromal interactions. In GIST, while primary oncogenic drivers are often PDGFRA mutations, PDGFRB is overexpressed in the tumor stroma, where it facilitates by stabilizing and improving vessel perfusion to support tumor progression. Autocrine and loops involving PDGF-B and PDGFRB play a in the , particularly within the . In various solid tumors, cancer cells secrete PDGF-B to activate PDGFRB on stromal fibroblasts and , promoting remodeling and immune evasion, which fosters a supportive niche for tumor expansion. Autocrine loops occur when tumor cells co-express PDGF-B and PDGFRB, driving self-sustained and , as observed in and gastric cancers where this signaling maintains an invasive . These loops enhance stromal , increasing interstitial and to the tumor core. PDGFRB activation contributes to cancer by enhancing cellular and inducing epithelial-mesenchymal transition (). In gastric , PDGF-B/PDGFRB signaling upregulates EMT markers such as N-cadherin and , enabling tumor cells to acquire migratory properties and breach basement membranes. In mammary cancers, autocrine PDGFRB loops sustain metastatic potential by promoting anoikis resistance and dissemination to distant sites. that amplify PDGFRB signaling, such as those detailed in genetic variants sections, can further exacerbate these metastatic traits. Recent studies from 2023 to 2025 have linked PDGFRB expression to resistance in cancers. In gastric cancer, high PDGFRB levels serve as a prognostic immune-related , associating with immunosuppressive microenvironments and reduced response to inhibitors by modulating T-cell infiltration. 2025 preclinical models in pancreatic ductal suggest that targeting PDGFRB-positive cancer-associated fibroblasts in stromal compartments, in combination with anti-PD-L1 therapy, can enhance tumor suppression and reduce . These findings underscore PDGFRB's emerging role in shaping immune evasion.

Contribution to Non-Cancer Diseases

PDGFRB, encoding the receptor beta, contributes to by promoting the proliferation and migration of vascular cells (SMCs) in response to PDGF-BB binding, leading to neointimal thickening and plaque formation. This receptor activation enhances SMC phenotypic modulation from a contractile to a synthetic state, exacerbating intimal in arterial walls. In fibrotic diseases such as systemic sclerosis (scleroderma), PDGFRB signaling drives excessive (ECM) deposition by activating fibroblasts and myofibroblasts, resulting in tissue stiffening and organ dysfunction. Stimulatory autoantibodies targeting PDGFRB have been implicated in this process, amplifying profibrotic pathways independent of canonical PDGF ligands and contributing to dermal and vascular . PDGFRB is expressed on perivascular cells in vascular anomalies like infantile hemangiomas, where it supports endothelial proliferation and vessel remodeling during the proliferative phase of these benign tumors. These PDGFRB-positive contribute to the angiogenic switch, facilitating rapid lesion growth in infancy before spontaneous involution. Recent 2025 studies highlight PDGFRB's role in craniofacial dysmorphisms associated with Penttinen premature aging syndrome, where gain-of-function mutations are linked to manifestations including and calvarial thinning. In fibrotic lung conditions, such as , elevated PDGFRB expression on fibroblasts correlates with disease progression; imaging has visualized PDGFRB activity in fibrotic regions, underscoring its potential as a therapeutic target. Dysregulated PDGFRB signaling in these non-cancer contexts often involves with TGF-β pathways, promoting differentiation.

Genetic Variants

Deletions and Haploinsufficiency

Deletions encompassing the PDGFRB locus at 5q32 contribute to the 5q- syndrome, a distinct subtype of (MDS) characterized by of multiple genes in the commonly deleted region (CDR), including PDGFRB alongside CSF1R and the gene containing CSF2 (encoding GM-CSF). of PDGFRB leads to reduced expression levels, approximately 73% lower in CD34+ cells from affected patients compared to controls, potentially contributing to dysregulated signaling in hematopoietic and stromal cells. Similarly, CSF1R haploinsufficiency impairs monocyte and differentiation, explaining the characteristic monocytic observed in biopsies. Loss of CSF2 further disrupts granulocyte-macrophage progenitor function, exacerbating ineffective hematopoiesis. The 5q- syndrome accounts for about 10% of all MDS cases and typically presents in older adults with isolated del(5q) as the sole cytogenetic abnormality. Key clinical features include severe with hypolobated micromegakaryocytes in the , normal or elevated platelet counts (thrombocytosis in up to 50% of cases), and a low risk of progression to (median survival exceeding 5 years). Diagnosis relies on cytogenetic analysis, with (FISH) probes targeting the 5q32 region (e.g., spanning EGR1 to PDGFRB) confirming the deletion in over 90% of suspected cases when combined with standard karyotyping. Lenalidomide, an immunomodulatory agent, is the frontline therapy for transfusion-dependent patients with 5q- syndrome, achieving complete cytogenetic remission in approximately 25-30% of cases and partial hematologic responses in over 60%, often by modulating and alleviating effects. In models, heterozygous deletion of Pdgfrb results in partial vascular defects, such as reduced coverage and mild glomerular abnormalities, underscoring PDGFRB's dosage-sensitive role in vascular stability without the perinatal lethality seen in homozygous knockouts.

Fusion Genes from Translocations

Fusion genes involving PDGFRB arise primarily from chromosomal translocations that juxtapose the domain of PDGFRB on chromosome 5q32 with various partner genes, resulting in oncogenic activation through ligand-independent dimerization and constitutive kinase signaling. These rearrangements are characteristic of myeloid and lymphoid neoplasms with , classified under the as myeloid/lymphoid neoplasms with eosinophilia and gene fusions. A prototypical example is the t(5;12)(q33;p13) translocation, which generates the ETV6-PDGFRB fusion gene, most commonly associated with chronic eosinophilic leukemia or featuring prominent hypereosinophilia. ETV6, located on 12p13, provides oligomerization domains that drive the aberrant activation of PDGFRB. Over 30 distinct partner genes have been identified, including RABEP1 via t(5;17)(q33;p13) in and HIP1 via t(5;7)(q33;q11.2) in similar eosinophilia-associated disorders, each contributing to sustained activity that promotes uncontrolled . Clinically, these fusions manifest as with marked hypereosinophilia, often involving multiorgan infiltration and potential progression to if untreated. Notably, neoplasms harboring PDGFRB fusions exhibit high sensitivity to inhibitors like , with durable responses observed in the majority of cases, underscoring their therapeutic relevance. Detection typically relies on (RT-PCR) for known fusions or next-generation sequencing (NGS) for novel variants, enabling precise identification in or peripheral blood. As of 2025, emerging reports highlight rare PDGFRB rearrangements in solid tumors, such as a novel tandem duplication activating the juxtamembrane domain in infantile myofibromatosis, expanding the spectrum beyond hematologic malignancies and suggesting potential responsiveness in these pediatric neoplasms.

Point Mutations and Rare Variants

Point mutations and rare variants in the PDGFRB gene, which encodes the receptor beta (PDGFRβ), have been implicated in several rare disorders, primarily through loss-of-function or gain-of-function effects on receptor signaling. These variants often exhibit autosomal dominant inheritance with variable , leading to disruptions in function, vascular integrity, and cellular signaling pathways. In (PFBC), a neurodegenerative characterized by bilateral calcifications, heterozygous missense mutations in PDGFRB are a known cause, accounting for a subset of cases. For instance, the p.P154S variant, located in the D2 immunoglobulin-like domain of the extracellular region, abolishes PDGF-BB ligand and reduces cell surface expression of the receptor, resulting in impaired downstream signaling and pericyte that contributes to vascular . Similarly, the p.R226C mutation in the D3 domain decreases ligand affinity and cell surface trafficking, further compromising PDGF-B/PDGFRβ signaling essential for vascular . Functional studies of PDGFRB missense variants in PFBC reveal diverse impacts on activity and receptor localization. Mutations such as p.G612R, p.L658P, p.D826Y, and p.D844G, situated in the domain, lead to a complete loss of autophosphorylation and activity, severely attenuating . In contrast, the p.P596L in the juxtamembrane domain causes partial reduction in activity, correlating with milder phenotypes and incomplete observed in some families. A 2024 genomic study identified a novel p.R334Q missense in the D4 extracellular domain in a family with PFBC, predicted to disrupt binding and cause dysfunction, with autosomal dominant segregation confirmed by whole-exome sequencing; this highlights ongoing discoveries in variant-specific vascular instability. These loss-of-function effects underscore PDGFRB's role in maintaining the blood- barrier, where haploinsufficiency-like mechanisms promote calcium leakage into parenchyma. Beyond PFBC, rare activating PDGFRB variants are associated with vascular malformations and syndromes exhibiting autoinflammatory features. In infantile myofibromatosis, a vascular tumor disorder, the recurrent germline p.R561C missense mutation in the juxtamembrane domain confers constitutive receptor activation, promoting mesenchymal cell proliferation and tumor formation through enhanced kinase activity; this variant often requires a somatic second hit for full penetrance, following an autosomal dominant pattern with incomplete expressivity. The p.R561C substitution stabilizes the active receptor conformation, leading to ligand-independent signaling and sensitivity to tyrosine kinase inhibitors like imatinib. In Penttinen syndrome, a progeroid disorder with autoinflammatory manifestations such as chronic skin inflammation and interferon-like responses, de novo missense variants like p.V665A and p.N666S in the kinase domain hyperactivate PDGFRβ, triggering excessive STAT1/STAT3 signaling and cellular senescence-like changes in vascular smooth muscle cells. These gain-of-function variants illustrate how altered receptor trafficking and sustained kinase activity can drive dysregulated pericyte-vascular interactions, contributing to multifocal lesions and inflammatory phenotypes. Recent analyses, including a 2024 case series, emphasize the spectrum of these rare variants in overlapping neurovascular and inflammatory conditions, advocating for targeted sequencing in atypical presentations.

Molecular Interactions

Protein Binding Partners

PDGFRB, upon activation by (PDGF) ligands, undergoes autophosphorylation on multiple tyrosine residues in its intracellular domain, creating docking sites for Src homology 2 (SH2) domain-containing proteins that mediate downstream signaling. These interactions are highly specific, with binding affinities typically in the nanomolar to micromolar range as determined by co-immunoprecipitation and studies in analyses. Key SH2-domain adaptors include the p85 regulatory subunit of 3-kinase (PI3K), which binds directly to phosphotyrosines 740 and 751 (pY740 and pY751) on PDGFRB, recruiting the PI3K holoenzyme to the plasma membrane.39813-2/fulltext) , an adaptor linking to the SOS , primarily associates with pY716 on PDGFRB, though indirect binding via Shc can occur at additional sites. The Shc adaptor protein binds to several phosphotyrosines, including pY579, pY740, pY751, and pY771, facilitating recruitment of and contributing to mitogenic responses. SHP-2 (also known as ), a , interacts via its SH2 domains with pY720, pY754, and pY763, enabling both positive and negative regulation of receptor phosphorylation. Among cytoskeletal regulators, Crk, an SH2/SH3-domain adaptor, binds to pY762 on PDGFRB, promoting actin reorganization and cell migration through interactions with paxillin and other effectors. Nck1, another SH2/SH3 adaptor, associates with pY751, linking PDGFRB to the regulatory complex and facilitating lamellipodia formation during . Caveolin-1, a principal component of caveolae, directly interacts with the domain of PDGFRB, inhibiting its autophosphorylation and promoting receptor via clathrin-independent pathways; this association has been quantified with dissociation constants around 10-50 nM in fractionation studies.71963-2/fulltext) Negative regulators of PDGFRB include RasGAP (also called RASA1), which binds to pY771 through its SH2 domains, accelerating GTP hydrolysis on Ras and attenuating mitogenic signaling; this interaction was first identified in early affinity purification experiments showing stoichiometric binding upon PDGF stimulation. SHP-1 (PTPN6), a hematopoietic-specific , associates directly with autophosphorylated PDGFRB via its tandem SH2 domains, leading to of key tyrosines and dampening of receptor activity, as demonstrated in co-immunoprecipitation assays from PDGF-stimulated fibroblasts.93781-6/fulltext) approaches, such as mass spectrometry-based affinity capture, have confirmed these interactions with high confidence, revealing PDGFRB as a hub for over 20 SH2-domain proteins, though the listed partners represent the most prominent direct binders.

Regulatory Mechanisms

PDGFRB undergoes primarily through -mediated pathways involving the AP-2 adaptor complex, which facilitates receptor following binding. This process accounts for a significant portion of PDGFRB uptake, with inhibition of heavy chain, dynamin-2, or AP-2 μ2 subunit reducing by 77–93% at low concentrations (10 ng/ml PDGF-BB). Internalized PDGFRB is then sorted in early endosomes, directing it toward lysosomal degradation to terminate signaling or recycling back to the plasma membrane to sustain activity. -mediated is crucial for proper signaling, as its disruption impairs activation by approximately 50% without affecting other pathways like AKT or PLCγ. Ubiquitination of PDGFRB is mediated by the Cbl family of ligases, including c-Cbl and Cbl-b, which promote signal termination by enhancing receptor . Upon PDGF-BB stimulation, both ligases are required for efficient ubiquitination, with their simultaneous depletion reducing PDGFRB ubiquitination and slowing rates. Cbl-b binds directly to phosphorylated residues (Tyr-1009 and Tyr-1021) on PDGFRB, while c-Cbl acts in concert, potentially indirectly; this modification increases of the receptor and amplifies downstream signals such as , , and PLCγ, thereby enhancing . Although ubiquitination facilitates , it does not primarily drive proteasomal degradation of PDGFRB. Lipid rafts provide an allosteric regulatory environment for PDGFRB by influencing receptor localization and downstream signaling in a cell-type-specific manner. Cholesterol depletion disrupts rafts and abolishes ERK1/2 activation in fibroblasts while modestly reducing AKT , but in cells, it impairs PDGFRB tyrosine and AKT/ signaling without affecting ERK1/2. In endothelial contexts, raft association modulates PDGFRB activation, with /RhoA-mediated tension inhibiting signaling specifically within rafts but not non-raft regions. in further influences raft dynamics, altering lipid order and fluidity to regulate mechanotransduction, though direct effects on PDGFRB remain linked to broader raft-dependent signaling. Post-activation feedback loops attenuate PDGFRB expression through miRNA-mediated suppression, establishing to prevent prolonged signaling. with PDGF-BB induces a rapid decline in PDGFRB protein and mRNA levels, starting at 1 and 6 hours, respectively, which persists for up to 48 hours in cardiomyocytes. MicroRNA-9 (miR-9) expression increases significantly by 6 hours post-stimulation, directly targeting the 3' untranslated region of PDGFRB mRNA to suppress and reduce angiogenic paracrine effects. This miR-9 induction forms a loop, as anti-miR-9 treatment prevents ligand-induced downregulation of PDGFRB.

Clinical and Therapeutic Aspects

Diagnostic Implications

Alterations in the PDGFRB gene, located on chromosome 5q32, are detected using (FISH) and (PCR) techniques to diagnose myelodysplastic syndromes (MDS) associated with 5q deletions. In MDS with isolated del(5q), FISH probes targeting the 5q region, including PDGFRB, confirm the loss of one allele, which is a defining cytogenetic feature linked to a relatively favorable compared to other MDS subtypes. For myeloid neoplasms with eosinophilia, such as chronic eosinophilic leukemia or hypereosinophilic syndrome (HES), FISH panels screen for PDGFRB rearrangements, while (RT-PCR) identifies fusion transcripts like ETV6-PDGFRB, aiding in precise classification and distinguishing these from other eosinophilia-associated disorders. Immunohistochemistry (IHC) serves as a valuable tool for assessing PDGFRB protein overexpression in certain solid tumors. Elevated PDGFRB expression detected via IHC has also been observed in some gastrointestinal stromal tumors (GISTs), sarcomas, and carcinomas. Next-generation sequencing (NGS) panels are increasingly employed to identify rare PDGFRB variants in non-hematologic disorders, such as primary familial brain calcification (PFBC) and vascular anomalies. In PFBC, heterozygous missense mutations in PDGFRB, accounting for approximately 5% of genetically confirmed cases, are detected through targeted NGS of calcification-associated genes, often revealing variants like p.Arg695Cys that correlate with mild phenotypes including basal ganglia calcifications visible on CT imaging. For vascular disorders, including infantile myofibromatosis and fusiform cerebral aneurysms, NGS identifies both germline and somatic gain-of-function PDGFRB variants, such as p.Arg561Cys or activating substitutions in the kinase domain, facilitating early diagnosis in familial or sporadic presentations. PDGFRB expression levels serve as prognostic biomarkers in various cancers, with high expression often correlating with increased risk and poorer outcomes. For instance, in , elevated PDGFRB in stromal fibroblasts independently predicts biochemical recurrence and , as determined by IHC scoring in large cohorts. Similarly, in , increased PDGFRB expression is associated with higher recurrence risk, while in gastric cancer, it links to advanced tumor stages and reduced survival through promotion of . These biomarkers are typically assessed via quantitative IHC or sequencing to stratify patients for risk.

Targeted Therapies and Inhibitors

Tyrosine kinase inhibitors (TKIs) targeting PDGFRB have demonstrated substantial efficacy in treating fusion-driven leukemias, particularly Philadelphia chromosome-like acute lymphoblastic leukemia (Ph-like ALL) and chronic myeloproliferative disorders. Imatinib, a first-generation TKI, induces high response rates in patients with PDGFRB fusions, achieving complete remission in approximately 96% of cases in myeloid malignancies associated with PDGFRB rearrangements, often with durable cytogenetic and molecular responses. Dasatinib and nilotinib, second-generation TKIs, also exhibit activity against PDGFRB fusions, though preclinical data indicate imatinib may be the optimal choice for PDGFRB-rearranged ALL due to superior sensitivity compared to dasatinib in cell line models. In pediatric Ph-like ALL, where PDGFRB fusions occur in about 3% of cases, combining TKIs with chemotherapy improves complete remission rates to around 80-90%; a 2024 analysis of 28 cases reported a 90% complete remission rate with this approach. Multi-kinase inhibitors such as , which potently inhibit PDGFRB alongside and VEGFRs, are employed in gastrointestinal stromal tumors (GIST) following failure, providing median of 6-8 months in advanced disease. Although primary activating mutations in PDGFRB are rare in GIST—unlike the more common and PDGFRA alterations—'s broad-spectrum inhibition of PDGFRB contributes to its antitumor effects in KIT-mutant contexts by disrupting downstream signaling and . This approach extends to other PDGFRB-driven pathologies, where serves as a second-line option after progression on selective TKIs. Emerging therapies as of 2025 focus on greater selectivity and novel degradation mechanisms to address limitations of existing TKIs. Selective small-molecule PDGFRB inhibitors like CP-673451 demonstrate potent blockade of wild-type and mutant PDGFRB in preclinical models of and solid tumors, with values in the nanomolar range and reduced off-target effects compared to multi-kinase agents. PROTACs based on have been developed to induce ubiquitin-mediated degradation of PDGFRB, achieving dose-dependent protein reduction in cells and overcoming resistance associated with kinase domain mutations. Additionally, fully human anti-PDGFRB monoclonal antibodies, such as those targeting the extracellular domain, show promise in preclinical studies for inhibiting binding and receptor dimerization in non-hematologic cancers, potentially synergizing with TKIs. Resistance to PDGFRB-targeted TKIs often arises from on-target mutations, including the T681I variant, which sterically hinders binding and confers resistance to and in Ph-like ALL models, though it remains sensitive to certain multi-target agents like CHZ868. Other mutations, such as C843G, similarly promote relapse by reducing TKI affinity across , , , and . Combination strategies, including sequential TKI switching (e.g., from to ) or pairing TKIs with , mitigate resistance and improve event-free survival in fusion-positive leukemias, with ongoing trials evaluating PROTACs and antibodies to target mutant forms.

References

  1. [1]
    PDGFRB gene: MedlinePlus Genetics
    ### Summary of PDGFRB Gene
  2. [2]
    Entry - *173410 - PLATELET-DERIVED GROWTH FACTOR ... - OMIM
    The Treacher Collins Syndrome Collaborative Group (1996) determined that the PDGFRB gene is located within approximately 900 kb proximal of the TCOF1 gene ( ...Missing: position | Show results with:position
  3. [3]
    Structural and Functional Properties of Platelet-Derived Growth ...
    PDGF and SCF receptors are type III PTK receptors with five Ig-like domains, activated by dimerization, and involved in cell growth, survival, and migration. ...
  4. [4]
    Structure of full-length human PDGFRβ bound to its activating ligand ...
    The architecture of the entire PDGF-B/PDGFRβFL complex comprises three layers: extracellular domain (ECD), transmembrane domain (TMD), and tyrosine kinase ...
  5. [5]
  6. [6]
    PDGFRB platelet derived growth factor receptor beta [ (human)] - NCBI
    Sep 14, 2025 · This gene is essential for normal development of the cardiovascular system and aids in rearrangement of the actin cytoskeleton.
  7. [7]
  8. [8]
  9. [9]
  10. [10]
    Cloning and expression of a cDNA coding for the human platelet ...
    Comparison to the mouse PDGF receptor reveals an overall amino acid sequence identity of 86%. This sequence identity rises to 98% in the cytoplasmic split ...
  11. [11]
    Platelet-derived growth factor receptor beta identifies mesenchymal ...
    Platelet-derived growth factor (PDGF) receptor beta (PDGFR-β), also known as CD140b, is expressed on perivascular mesenchymal cells including pericytes and ...
  12. [12]
    Dysregulated mesenchymal PDGFR‐β drives kidney fibrosis
    Jan 14, 2020 · PDGFR‐β was expressed in renal mesangial cells, interstitial fibroblasts and pericytes, and vascular smooth muscle cells (Fig 1B). In ...
  13. [13]
    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 ...
  14. [14]
    Role of PDGF-B and PDGFR-β in recruitment of vascular smooth ...
    Jul 15, 1999 · Vascular PDGF-B and PDGFR-β expression. PDGF-B is expressed in capillary and arterial or arteriolar EC during embryonic and early postnatal ...
  15. [15]
    Conserved and context-dependent roles for pdgfrb signaling during ...
    Platelet derived growth factor beta and its receptor, Pdgfrb, play essential roles in the development of vascular mural cells, including pericytes and vascular ...
  16. [16]
    Sp1 is a key regulator of the PDGF beta-receptor transcription
    In this report, we show that Sp1 plays an essential role in the PDGF beta-receptor transcription. Within the upstream GC rich area there are two Sp1 binding ...
  17. [17]
    Sp1 phosphorylation regulates inducible expression of platelet ... - NIH
    Sp1 was the first endogenous nuclear factor shown to bind the PDGF-B promoter ∼30 bp upstream of the TATA box. This specific interaction mediates basal PDGF-B ...
  18. [18]
    Pdgfrb is a direct regulatory target of TGFβ signaling in ...
    The type II receptor phosphorylates (activates) the type I receptor, which subsequently phosphorylates SMAD2 and SMAD3, which are also known as TGFβ Receptor- ...
  19. [19]
    HIF1α-induced PDGFRβ signaling promotes developmental HSC ...
    To identify which cell types may be responding to Hif1α regulation through PDGFRβ signaling, we compared the expression levels of pdgfba and pdgfrb in sorted ...
  20. [20]
    PDGFRβ expression and function in fibroblasts derived from ...
    We have studied changes in DNA methylation of the PDGFRB promoter upon ... S2), revealed that 20% of CpG sites in the PDGFRB promoter were methylated (Fig.
  21. [21]
    PDGFRβ translocates to the nucleus and regulates chromatin ... - NIH
    Papadopoulos et al. show that PDGFRβ interacts with TMF-1 and Fer kinase in the nucleus, regulating chromatin remodeling by the SWI–SNF complex and controlling ...
  22. [22]
    p09619 · pgfrb_human - UniProt
    Gene name, Length. E5RJ14, E5RJ14_HUMAN, PDGFRB, 146. E5RII0, E5RII0_HUMAN, PDGFRB, 174. E5RH16, E5RH16_HUMAN, PDGFRB, 239. Features. Showing features for ...
  23. [23]
    Genetic screening and functional characterization of PDGFRB ... - NIH
    An early PDGFRβ precursor of ~130 kDa probably results from core glycosylation. ... L658P mutations affected the levels of the mature 180 kDa PDGFRβ form. While ...
  24. [24]
    Platelet-derived growth factors and their receptors: structural ... - NIH
    All PDGFs have a conserved growth factor domain in the cystine-knot fold that is related to vascular endothelial growth factors (VEGFs), which is the only part ...
  25. [25]
    3MJG: The structure of a platelet derived growth factor receptor ...
    Jun 16, 2010 · We show that PDGFs share a conserved region in their prodomain sequences which can remain noncovalently associated with the mature cystine-knot growth factor ...Missing: architecture | Show results with:architecture
  26. [26]
  27. [27]
    Regulation of Platelet-derived Growth Factor Receptor Function by ...
    Phosphorylation of the conserved tyrosine residue in the kinase domain (Tyr-849 of PDGFRα and Tyr-857 of PDGFRβ) increases catalytic activity of the kinases, ...
  28. [28]
    Tyrosines-740/751 of PDGFRβ contribute to the activation of Akt ...
    Sep 23, 2017 · The PDGFRβ autophosphorylation sites Tyr-740 and Tyr-751 are necessary for the expression of Hif1α. Furthermore, we show that tyrosine ...Missing: PDGFRB functional
  29. [29]
    The role of small molecule platelet-derived growth factor receptor ...
    The PDGF receptors contain an extracellular domain that is made up of five immunoglobulin-like domains (Ig-d1/2/3/4/5), a transmembrane segment, a juxtamembrane ...<|control11|><|separator|>
  30. [30]
    The Ubiquitin Ligases c-Cbl and Cbl-b Negatively Regulate Platelet ...
    In this report, we demonstrate that, after ligand stimulation, the PDGF β receptor (PDGFRβ) becomes ubiquitinated in a manner requiring both the c-Cbl and Cbl-b ...Immunofluorescence Staining · Cell Migration Assay · C-Cbl And Cbl-B Interact...
  31. [31]
    Genetic Screening and Functional Characterization of PDGFRB ...
    May 5, 2014 · ... Genetic Screening and Functional Characterization of PDGFRB Mutations Associated with Basal Ganglia Calcification of Unknown Etiology. Monica ...
  32. [32]
    Functional importance of platelet-derived growth factor (PDGF ...
    Dec 26, 1997 · ... affinity (Kd = 0.21-0.42 nM). Identical results were obtained for 125I-PDGF BB binding to beta-PDGFR mutants D1-D5 and D2-D3, establishing ...Missing: PDGFRB | Show results with:PDGFRB
  33. [33]
    Mechanism of platelet-derived growth factor (PDGF) AA, AB, and BB ...
    Scatchard analysis revealed high affinity binding (Kd = 0.5-1.0 nM) of radiolabeled PDGF AA and AB to alpha receptor and of PDGF BB to both receptor subtypes.Missing: PDGFRB | Show results with:PDGFRB
  34. [34]
    PDGF receptor-β modulates metanephric mesenchyme chemotaxis ...
    The α-receptor has greater affinity for PDGF AA than BB. PDGFR-β binds PDGF BB and DD with high affinity (5, 11, 26, 39, 52). Activation of PDGFR-β mediates ...Missing: PDGFRB | Show results with:PDGFRB
  35. [35]
    Platelet-Derived Growth Factor Signaling in the Lung ... - ATS Journals
    Jul 25, 2014 · Later in embryogenesis, lung pdgfr-a–positive mesenchymal cells undergo PDGF-A–mediated migration to the site of alveolar septation, which does ...
  36. [36]
    Platelet Derived Growth Factor C - an overview | ScienceDirect Topics
    In addition, the matrix-bound forms of PDGF-A and PDGF-B, which contain a retention motif, may require proteolytic cleavage to be released from the ...
  37. [37]
    Role of platelet-derived growth factors in physiology and medicine
    Available genetic data suggest that PDGF-A is a critical ligand for PDGFR-α in oligodendrocyte development. PDGF-A is expressed by neurons and astrocytes ...
  38. [38]
    Structure of Full-Length Human PDGFRβ Bound to Its Activating ...
    These Class III RTKs feature five immunoglobulin-like domains (Ig) in the extracellular segment (D1–D5), a single-pass transmembrane domain (TMD) and a split ...Missing: PDGFRB | Show results with:PDGFRB
  39. [39]
    New insights about the PDGF/PDGFR signaling pathway as a ...
    This study will concentrate on the regulation of the PDGF/PDGFR signaling system, in particular the current methods and inhibitors used in cancer treatment.Missing: immunoglobulin specificity
  40. [40]
    PDGFRα/β heterodimer activation negatively affects downstream ...
    May 22, 2025 · PDGFRα and PDGFRβ share the highest amino acid homology in the kinase domains, with significantly reduced homology in the extracellular and ...
  41. [41]
    Functions of the major tyrosine phosphorylation site of the PDGF ...
    Our data suggest that phosphorylation of Y857 may be important for stimulation of kinase activity of the receptors and for downstream actions.
  42. [42]
    Unusual PDGFRB fusion reveals novel mechanism of kinase ...
    Feb 21, 2023 · Constitutive kinase activation of oncogenic chimeric-fusions is driven by retained oligomerization domains of N-terminal partner proteins, which ...
  43. [43]
    Phosphoproteomic analysis of lung tissue from patients with ...
    Aug 1, 2021 · PDGFR beta hyperphosphorylation and PTP down-regulation were also present in vivo in mice with chronic hypoxia-induced pulmonary hypertension.
  44. [44]
    Effect of Oxidative Stress on Protein Tyrosine Phosphatase-1B ... - NIH
    PDGFR is phosphorylated upon PDGF stimulation, and dephosphorylated by protein tyrosine phosphatases (PTPs), including PTP1B. In this study we determine whether ...
  45. [45]
    Site-selective Dephosphorylation of the Platelet-derived Growth ...
    We have investigated PDGF β-receptor dephosphorylation by receptor-like protein-tyrosine phosphatase DEP-1 using a cell line with inducible DEP-1 expression and ...
  46. [46]
    Role of Phosphoinositide 3-Kinase Regulatory Isoforms in ... - NIH
    PDGF binding to the receptor activates autophosphorylation at sites (Tyr 740 and Tyr 751) ... Targeting the PI3K-Akt pathway in human cancer: rationale and promise ...Pi3k Assay · Mefs Lacking P85α, P55α... · Fig. 6Missing: PDGFRB | Show results with:PDGFRB
  47. [47]
    Tyr-716 in the platelet-derived growth factor beta-receptor ... - PubMed
    Here we show that Tyr-716, a novel autophosphorylation site in the PDGF beta-receptor kinase insert, mediates direct binding of GRB2 in vitro and in vivo.
  48. [48]
    Platelet-derived growth factor activates mitogen-activated protein ...
    1A shows by immunoblotting that caveolae fractions from the quiescent human fibroblasts used in this study contain PDGF receptor, Ras, Raf-1, Mek-1, and erk2.
  49. [49]
    Platelet-derived growth factor receptor β activation and regulation in ...
    These phosphorylated residues serve as binding sites for downstream signaling components and activate, among others, phospholipase C γ (PLCγ) ... Pdgfrb.
  50. [50]
    Platelet-derived growth factors and their receptors in normal ... - NIH
    PDGF binds to receptors, and aberrant activation occurs in myeloid malignancies. PDGF may regulate some hematopoietic cells, but its role in normal ...
  51. [51]
    Myeloproliferative disease induced by TEL-PDGFRB displays ...
    Stat5 is required in primary murine hematopoietic cells for TEL-PDGFRB–mediated growth. Mice harboring targeted deletions of both Stat5a and Stat5b genes ...
  52. [52]
  53. [53]
  54. [54]
    Src family kinases are required for integrin but not PDGFR signal ...
    These initial signaling events may play an important role in mediating cytoskeletal rearrangements required for cell motility (Lauffenburger and Horwitz, 1996).Missing: PDGFRB | Show results with:PDGFRB
  55. [55]
    Integrins, Oncogenes, and Anchorage Independence
    Nov 3, 1997 · The finding that integrins transduce signals that influence intracellular growth regulatory pathways provided some insight into anchorage ...
  56. [56]
    Role of PDGFR-β/PI3K/AKT signaling pathway in PDGF-BB induced ...
    PDGF-BB may induce CF proliferation and their transformation into myofibroblasts, which leads to increased synthesis of collagen, resulting in myocardial ...Missing: eosinophils | Show results with:eosinophils
  57. [57]
    ETV6-PDGFRB and FIP1L1-PDGFRA Stimulate Human ... - PubMed
    The aim of this work was to gain insight into the mechanisms whereby fusion genes affect human hematopoietic cells and in particular the eosinophil lineage.
  58. [58]
    Defining Endothelial Cell-Derived Factors That Promote Pericyte ...
    Aug 20, 2020 · Genetic disruption of either PDGF-BB or its receptor, PDGFR-β in mice leads to a strong decrease in the accumulation of pericytes within ...
  59. [59]
    Endothelial PDGF-B retention is required for proper investment of ...
    We conclude that retention of PDGF-B in microvessels is essential for proper recruitment and organization of pericytes and for renal and retinal function in ...
  60. [60]
    Retention of PDGFR-β function in mice in the absence of ... - NIH
    In both the PDGFR-β−/− and the PDGF-B−/− mice, the loss of pericytes results in perinatal lethality caused by microaneurisms, especially in the brain and the ...
  61. [61]
    Roles of PDGF in animal development
    Oct 15, 2003 · In both mouse and human, the PDGF signaling network consists of four ligands, PDGFA-D, and two receptors, PDGFRα and PDGFRβ(Fig. 1). All ...The Pdgf Signaling Family · Pdgf Roles In The... · Pdgf Roles In Organogenesis
  62. [62]
    PDGFRβ regulates craniofacial development through homodimers ...
    Nov 17, 2016 · Targeted disruption of Pdgfra in mice results in embryonic lethality ... Pdgfrb conditional knockout mice (Fig. 2G–G′′) are ...
  63. [63]
    Pdgfra and Pdgfrb Genetically Interact in the Murine Neural Crest ...
    We found that PDGFRα plays a predominant role in cNCC migration whereas PDGFRβ primarily contributes to proliferation of the facial mesenchyme past mid- ...
  64. [64]
    Platelet-derived growth factor receptor-β regulates vascular smooth ...
    The main function of VSMCs is to regulate tissue perfusion through graded contraction. Importantly, VSMC display a remarkable plasticity by converting among ...Missing: PDGFRB | Show results with:PDGFRB
  65. [65]
    Platelet-Derived Growth Factor-β Receptor Activation Is Essential for ...
    Platelet-derived growth factor-β receptor activation is essential for fibroblast and pericyte recruitment during cutaneous wound healing.
  66. [66]
    Deletion of the PDGFR-β Gene Affects Key Fibroblast Functions ...
    PDGF binding to the receptors activates a variety of intracellular signaling molecules (12). One of these is phosphatidylinositol 3-kinase (PI3K), which results ...
  67. [67]
    Pericytes are required for blood–brain barrier integrity during ...
    Oct 13, 2010 · We demonstrate that pericytes are necessary for the formation of the BBB, and that absolute pericyte coverage determines relative vascular permeability.
  68. [68]
    PDGFR-β restores blood-brain barrier functions in a mouse model of ...
    We have recently demonstrated that PDGFR-β signalling plays a vital role in BBB integrity and functional recovery after cerebral ischemia through the induction ...
  69. [69]
    PDGFRα plays a crucial role in connective tissue remodeling - Nature
    Dec 7, 2015 · The role of the PDGF-B/PDGFRβ interaction in promoting the recruitment of fibroblasts, pericytes and endothelial cells to enhance wound healing ...
  70. [70]
    Platelet-derived growth factor-B enhances glioma angiogenesis by ...
    PDGF-B enhances glioma angiogenesis, at least in part, by stimulating VEGF expression in tumor endothelia and by recruiting pericytes to neovessels.
  71. [71]
    Expression and prognostic significance of PDGF ligands and ...
    Jan 29, 2021 · The results presented here show that the overexpression of PDGFA might correlate with tumor aggressiveness in sarcoma: overexpression of PDGFA, ...
  72. [72]
    Targeting the PDGF signaling pathway in tumor treatment
    Dec 20, 2013 · Inhibition of PDGF receptor signaling has proven to useful for the treatment of patients with certain rare tumors. Whether treatment with PDGF/ ...
  73. [73]
    Autocrine PDGF stimulation in malignancies - PMC - NIH
    Apr 19, 2012 · In tumors, PDGF isoforms are often over-expressed and contribute to the growth of both normal and malignant cells. This review focuses on tumors ...
  74. [74]
    Autocrine PDGFR signaling promotes mammary cancer metastasis
    Jun 1, 2006 · Here we show that metastatic potential of oncogenic mammary epithelial cells requires an autocrine PDGF/PDGFR loop.
  75. [75]
    Platelet-derived growth factor-B signalling might promote epithelial ...
    We found that PDGF-B signalling can induce EMT in gastric carcinoma cells. The tumour microenvironment is imperative in the process of PDGF-B signalling ...
  76. [76]
    Down‐regulation of PDGFRβ suppresses invasion and migration in ...
    Jun 24, 2020 · Here we assessed the effects of PDGFRβ on invasive and migratory abilities, such as the epithelial–mesenchymal transition and ...
  77. [77]
    Identification of PDGFRB as a prognostic immune-related biomarker ...
    The immune-related gene PDGFRB demonstrated significant prognostic value in GC and may serve as a potential biomarker for immune response modulation. Our ...
  78. [78]
    Unraveling the role of cancer-associated fibroblasts - ScienceDirect
    In this review, we discuss the role of CAFs in the development of tumor resistance to chemotherapy, radiotherapy, targeted therapy, and immunotherapy.
  79. [79]
    Depletion of Adipose Stroma-Like Cancer-Associated Fibroblasts ...
    Jan 2, 2025 · The effects of subpopulations of cancer-associated fibroblasts (CAF) on cancer progression and therapy resistance are incompletely understood.
  80. [80]
    PDGF and cardiovascular disease - PubMed - NIH
    PDGF playing a prominent role in the migration of SMC into the neointima following acute injury and in atherosclerosis.
  81. [81]
    Desensitization of platelet-derived growth factor receptor-beta by ...
    The platelet-derived growth factor receptor-beta (PDGFRbeta) signaling pathway regulates smooth muscle cell (SMC) migration and proliferation and plays a role
  82. [82]
    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;
  83. [83]
    Stimulatory autoantibodies to the PDGF receptor in systemic sclerosis
    Jun 22, 2006 · Background: Systemic sclerosis (scleroderma) is characterized by immunologic abnormalities, injury of endothelial cells, and tissue fibrosis.
  84. [84]
    PDGFR-β (+) perivascular cells from infantile hemangioma display ...
    May 15, 2014 · Infantile hemangioma, a common benign tumor of infancy, grows quickly in the first year of life, and then regresses slowly to fibrofatty ...
  85. [85]
    Management of Pediatric Patient with Multiple Cranial, Intracranial ...
    Aug 12, 2024 · Penttinen premature aging syndrome is caused by mutations in the PDGFRB gene and presents with varying previously described cranial ...
  86. [86]
    PET imaging of platelet derived growth factor receptor β in lung fibrosis
    Jul 15, 2025 · PET imaging with radiotracers targeting fibroblast activation protein (FAP) has demonstrated strong uptake in fibrotic lung regions (Bergmann et ...
  87. [87]
    Regulation of PDGF and its receptors in fibrotic diseases - PubMed
    This review summarizes the literature on the role of PDGF and its receptors in the development of fibrosis in a variety of organ systems.Missing: sclerosis | Show results with:sclerosis
  88. [88]
    Common deleted genes in the 5q- syndrome: thrombocytopenia and ...
    Jul 12, 2007 · We conclude that many of the genes within the CDR associated with the 5q- syndrome exhibit significantly decreased expression.
  89. [89]
    Important Genes in the Pathogenesis of 5q- Syndrome and ... - NIH
    These abnormalities include interstitial deletion of a segment of the long arm of chromosome 5q [del(5q), 5q-syndrome], monosomy, and unbalanced translocations.Missing: PDGFRB flanking
  90. [90]
    What lies beyond del(5q) in myelodysplastic syndrome? - PMC - NIH
    Just under 50% of patients with de novo MDS will be found to have cytogenetic abnormalities, of which del(5q) is the most common. In the 10% of cases with the ...Missing: PDGFRB | Show results with:PDGFRB
  91. [91]
    5q minus syndrome: MedlinePlus Genetics
    Nov 1, 2015 · 5q minus (5q-) syndrome is a type of bone marrow disorder called myelodysplastic syndrome (MDS). MDS comprises a group of conditions in which immature blood ...Missing: PDGFRB | Show results with:PDGFRB
  92. [92]
    CytoCell Del(5q) Plus Tri-Colour Deletion FISH Probe
    The probe will also detect deletions of RPS14 (ribosomal protein S14) at 5q33.1, patients with MDS with del(5q) are haploinsufficient for RPS14 which leads ...
  93. [93]
    Functional Characterization of Germline Mutations in PDGFB and ...
    ... Genetic Screening and Functional Characterization of PDGFRB Mutations Associated with Basal Ganglia Calcification of Unknown Etiology. Hum Mutat. 2014. doi ...
  94. [94]
    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, ...
  95. [95]
    Patients with myeloid malignancies bearing PDGFRB fusion genes ...
    Reciprocal translocations involving PDGFRB result in fusion genes with constitutively activated receptor tyrosine kinase sensitive to inhibition with imatinib.
  96. [96]
    Available and emerging therapies for bona fide advanced systemic ...
    Dec 9, 2022 · At the time of writing, there have been 8 PDGFRA partner fusion genes characterized, over 30 for PDGFRB, 16 for FGFR1, 3 for JAK2, and 7 for ...
  97. [97]
    PDGFRB-rearranged T-lymphoblastic leukemia/lymphoma occurring ...
    This report documents the occurrence of MLNs with PDGFRB rearrangements and justifies the inclusion of PDGFRB in the list of genes associated with the WHO ...
  98. [98]
    t(5;7)(q33;q11) HIP1/PDGFRB
    Aug 1, 1998 · Fusion of Huntingtin interacting protein 1 to platelet-derived growth factor beta receptor (PDGFbetaR) in chronic myelomonocytic leukemia with t ...
  99. [99]
    Novel ETV6::RAPGEF6 fusion gene in chronic eosinophilic leukemia
    Feb 9, 2025 · Karyotyping showed a translocation t(5;12)(q31;p13). Targeted RNA Sequencing identified a novel ETV6::RAPGEF6 fusion gene, confirmed by RT-PCR.Methods And Results · Case Presentation · Fig. 1<|control11|><|separator|>
  100. [100]
    Novel PDGFRB Gene Fusions in Two Cases of Infantile ... - PubMed
    Although gene fusions are frequent and potent oncogenic drivers in soft-tissue neoplasia, fusion genes involving PDGFRB have not previously been linked to IM.<|control11|><|separator|>
  101. [101]
  102. [102]
  103. [103]
  104. [104]
    [PDF] Regulation of PDGFR signaling - Diva Portal
    rosine kinase domain split by a 100 amino acid residues long stretch, called kinase insert, and a C-terminal tail [13]. This domain structure is common for ...Missing: pocket | Show results with:pocket
  105. [105]
    Essential function of adaptor protein Nck1 in platelet-derived growth ...
    Jan 20, 2022 · We reported here a crucial role of Nck1 protein in regulating PDGFR-mediated LEC activation using LEC with a silenced expression of Nck1 protein.Missing: caveolin- | Show results with:caveolin-
  106. [106]
    PDGF induction of tyrosine phosphorylation of GTPase activating ...
    Dec 7, 1989 · Here we report that in intact quiescent fibroblasts, both AA and BB homodimers of PDGF rapidly induce tyrosine phosphorylation of GAP.
  107. [107]
    PDGFRB Result Summary | BioGRID
    caveolin 1, caveolae protein, 22kDa. 3. View. FOXK1. H. sapiens. FOXK1L, forkhead ... -, v-crk avian sarcoma virus CT10 oncogene homolog-like. 1. View. CSF1R. H ...
  108. [108]
  109. [109]
  110. [110]
  111. [111]
  112. [112]
    5q deletion FISH - Sonic Genetics
    In patients with MDS, deletion of 5q (as the sole cytogenetic abnormality) is associated with a favourable prognosis. Deletion of 5q is also associated with ...<|separator|>
  113. [113]
    International Consensus Classification of Myeloid Neoplasms and ...
    Sep 15, 2022 · We present the new International Consensus Classification (ICC) of myeloid and lymphoid neoplasms, introduced by a Perspective by Sehn and ...
  114. [114]
    PDGFRB rearrangement - Pathology Outlines
    Sep 22, 2023 · Myeloid / lymphoid neoplasm with PDGFRB rearrangement is a myeloid or lymphoid neoplasm with frequent eosinophilia, sometimes neutrophilia ...
  115. [115]
    Eosinophilia FISH Panel - NeoGenomics Laboratories
    The eosinophilia FISH panel is used to aid in the diagnosis of myeloid and lymphoid neoplasms with eosinophilia and prediction of therapeutic response. The ...
  116. [116]
    Genomics of PDGFR-rearranged hypereosinophilic syndrome - NIH
    4, 5, 6, 7 Patients with F/P or rearrangements of PDGFRB may present with hypereosinophilic syndrome (HES), defined as >1.5 × 106 eosinophils/mL blood and ...
  117. [117]
    PDGFR expression in differential diagnosis between KIT-negative ...
    Immunostaining with PDGFR-alpha is a helpful marker in discriminating between KIT-negative GISTs and other gastrointestinal mesenchymal lesions.
  118. [118]
    [PDF] The Relationship of Clinicopathological Findings and PDGFR-β ...
    Nov 26, 2021 · The relationship of clinicopathological findings and PDGFR-β expression with tumor recurrence in gastrointestinal stromal tumors. Turk J ...
  119. [119]
    A novel PDGFRB sequence variant in a family with a mild form of ...
    Apr 12, 2019 · Patients with a PDGFRB mutation seem to exhibit a milder phenotype and milder brain calcification on brain imaging than patients with SLC20A2 ...Missing: neurodevelopmental | Show results with:neurodevelopmental
  120. [120]
    Mutation of the PDGFRB gene as a cause of idiopathic basal ...
    Aug 6, 2025 · This gene encodes for platelet-derived growth factor receptor beta (PDGFR-β) which is a cell surface tyrosine kinase receptor, playing a vital ...
  121. [121]
    Report A Recurrent PDGFRB Mutation Causes Familial Infantile ...
    PDGFRB encodes the β polypeptide of the platelet-derived growth factor receptor (PDGFR-β), a RTK and a mitogen for mesenchyme-derived cells, including ...<|control11|><|separator|>
  122. [122]
    Somatic PDGFRB Activating Variants in Fusiform Cerebral Aneurysms
    Apr 25, 2019 · Somatic gain-of-function variants in PDGFRB are a novel mechanism in the pathophysiology of fusiform cerebral aneurysms and suggest a potential ...
  123. [123]
    Genomic profiling informs diagnoses and treatment in vascular ...
    Vascular anomalies are malformations or tumors of the blood or lymphatic vasculature and can be life-threatening. Although molecularly targeted therapies ...Missing: PFBC | Show results with:PFBC
  124. [124]
    High expression of PDGFR-β in prostate cancer stroma is ... - Nature
    Feb 24, 2017 · This large study highlights the prognostic importance of PDGFR-β expression, implicating its involvement in prostate cancer progression even in early stage ...<|control11|><|separator|>
  125. [125]
    Prognostic and predictive impact of stroma cells defined by PDGFRb ...
    Our study suggests that patients with higher expression of PDGFRb might have an increased risk of any breast cancer recurrence, but due to correlation with ...
  126. [126]
    PDGFRB is a potential prognostic biomarker and correlated with ...
    High PDGFRB expression can negatively affect GC prognosis by promoting angiogenesis and modulating the tumor immune microenvironment. These results strongly ...Missing: risk | Show results with:risk
  127. [127]
    Patients with myeloid malignancies bearing PDGFRB fusion genes ...
    Jun 5, 2014 · Of the patients, 96% responded; no patients who achieved a complete cytogenetic (n = 13) or molecular (n = 8) remission lost their response or ...<|control11|><|separator|>
  128. [128]
    Tyrosine kinase inhibitor response of ABL-class acute lymphoblastic ...
    May 23, 2024 · This finding suggests that imatinib may be the best choice for PDGFRB-fused ALL. Our data suggest that although ABL-class fusions are considered ...Missing: rates | Show results with:rates
  129. [129]
    Tyrosine Inhibitors Improve PDGFRB-Positive ALL Outcomes
    Sep 4, 2024 · An approximate 71% of patients with the fusion received combined treatment with chemotherapy and tyrosine kinase inhibitors, and the remainder ...
  130. [130]
    Sunitinib for the treatment of metastatic gastrointestinal stromal tumors
    Jun 3, 2024 · Sunitinib is an oral anticancer drug approved for the treatment of among others gastrointestinal stromal tumor (GIST).
  131. [131]
    Pharmacological management of gastrointestinal stromal tumours
    Sunitinib has demonstrated clinical benefit in patients with unresectable and/or metastatic GIST resistant or intolerant to imatinib. Use of sunitinib in a ...Missing: PDGFRB | Show results with:PDGFRB
  132. [132]
    CP-673451, a Selective Platelet-Derived Growth Factor Receptor ...
    PDGFR inhibitors have demonstrated effective antitumor activity in several solid tumors, including gastrointestinal stromal tumors, prostate cancer, non-small ...Missing: PDGFRB | Show results with:PDGFRB
  133. [133]
    Discovery of selective platelet-derived growth factor receptor-beta ...
    Jan 1, 2023 · We report the development of multi-targeted kinase inhibitor sorafenib-based PROTACs, they can selectively degrade platelet-derived growth factor receptor beta ...
  134. [134]
    Development of a Fully Human Anti-PDGFRβ Antibody That ... - NIH
    Treatment with imatinib in NSCLC is associated with decrease of phosphorylated PDGFR-beta and VEGF expression, decrease in interstitial fluid pressure and ...
  135. [135]
    TKI resistance in Ph-like ALL | Blood | American Society of Hematology
    May 17, 2018 · Although the T681I gatekeeper mutation in PDGFRB confers imatinib resistance in vitro, no patients have been identified with this mutation to ...
  136. [136]
    PDGFRB mutation and tyrosine kinase inhibitor resistance in Ph-like ...
    Imatinib was then given as monotherapy and induced rapid leukemia clearance with decrease of blast percentage from 51% to 3% within 8 weeks. Unfortunately, the ...
  137. [137]
    Protein tyrosine kinase inhibitor resistance in malignant tumors
    Sep 17, 2022 · Combination of Gatekeeper Mutations and Cysteine 481 Replacement Causes Super Resistance to the Irreversible BTK Inhibitors Ibrutinib ...