PDGFRB

Last updated
PDGFRB
PDGFR-beta 3MJG.png
Available structures
PDB Ortholog search: PDBe RCSB
Identifiers
Aliases PDGFRB , CD140B, IBGC4, IMF1, JTK12, PDGFR, PDGFR-1, PDGFR1, KOGS, PENTT, platelet derived growth factor receptor beta
External IDs OMIM: 173410 MGI: 97531 HomoloGene: 1960 GeneCards: PDGFRB
Orthologs
SpeciesHumanMouse
Entrez
Ensembl
UniProt
RefSeq (mRNA)

NM_002609
NM_001355016
NM_001355017

NM_001146268
NM_008809

RefSeq (protein)

NP_002600
NP_001341945
NP_001341946

NP_001139740
NP_032835

Location (UCSC) Chr 5: 150.11 – 150.16 Mb Chr 18: 61.18 – 61.22 Mb
PubMed search [3] [4]
Wikidata
View/Edit Human View/Edit Mouse

Platelet-derived growth factor receptor beta is a protein that in humans is encoded by the PDGFRB gene. Mutations in PDGFRB are mainly associated with the clonal eosinophilia class of malignancies.

Contents

Gene

The PDGFRB gene is located on human chromosome 5 at position q32 (designated as 5q32) and contains 25 exons. The gene is flanked by the genes for granulocyte-macrophage colony-stimulating factor and Colony stimulating factor 1 receptor (also termed macrophage-colony stimulating factor receptor), all three of which may be lost together by a single deletional mutation thereby causing development of the 5q-syndrome. [5] Other genetic abnormalities in PDGFRB lead to various forms of potentially malignant bone marrow disorders: small deletions in and chromosome translocations causing fusions between PDGFRB and any one of at least 30 genes can cause Myeloproliferative neoplasms that commonly involve eosinophilia, eosinophil-induced organ injury, and possible progression to aggressive leukemia (see blow). [6]

Structure

The PDGFRB gene encodes a typical receptor tyrosine kinase, which belongs to the type III tyrosine kinase receptor (RTK) family and is structurally characterized by five extracellular immunoglobulin-like domains, a single membrane-spanning helix domain, an intracellular juxtamembrane domain, a split tyrosine kinase domain and a carboxylic tail. [7] In the absence of ligand, PDGFRβ adopts an inactive conformation in which the activation loop folds over the catalytic site, the juxtamembrane region over a loop occluding the active site and the carboxy-terminal tail over the kinase domain. Upon PDGF binding the dimerization of receptor releases the inhibitory conformations due to auto-phosphorylation of regulatory tyrosine residues in trans fashion. Tyrosine residues 857 and 751 are major phosphorylation sites for the activation of PDGFRβ. [8]

The molecular mass of the mature, glycosylated PDGFRβ protein is approximately 180 kDa.

Modes of activation

Activation of PDGFRβ requires de-repression of the receptor's kinase activity. The ligand for PDGFRβ (PDGF) accomplishes this in the course of assembling a PDGFRβ dimer. Two of the five PDGF isoforms activate PDGFRβ (PDGF-B and PDGF-D). The activated receptor phosphorylates itself and other proteins, and thereby engages intracellular signaling pathways that trigger cellular responses such as migration and proliferation. There are also PDGF-independent modes of de-repressing the PDGFRβ's kinase activity and hence activating it. For instance, forcing PDGFRβ into close proximity of each other by overexpression or with antibodies directed against the extracellular domain. Alternatively, mutations in the kinase domain that stabilize a kinase active conformation result in constitutive activation.

Unlike PDGFRα, PDGFRβ cannot be indirectly activated. This is because PDGFRβ recruits RasGAP and thereby attenuates Ras/PI3K activity, which is required to engage a feed-forward loop that is responsible for this mode of activation. [9] [10]

Role in physiology/pathology

The phenotype of knock out mice demonstrates that PDGFRB is essential for vascular development, and that PDGFRB is responsible for activating PDGFRβ during embryogenesis. Eliminating either PDGFRB, or PDGF-B reduces the number of pericytes and vascular smooth muscle cells, and thereby compromises the integrity and/or functionality of the vasculature in multiple organs, including the brain, heart, kidney, skin and eye. [11] [12] [13] [14]

In vitro studies using cultured cells indicate that endothelial cells secrete PDGF, which recruits PDGFRβ-expressing pericytes that stabilize nascent blood vessels. [15] Mice harboring a single activated allele of PDGFRB show a number of postnatal phenotypes including reduced differentiation of aortic vascular smooth muscle cells and brain pericytes. Similarly, differentiation of adipose from pericytes and mesenchymal cells is suppressed. [16] Misregulation of the PDGFRβ's kinase activity (typically activation) contributes to endemic diseases such as cancer and cardiovascular disease. [17] [18] [19]

PDGFRB mutations

5q- Syndrome

Human chromosome 5 deletions that remove three adjacent genes, those for granulocyte-macrophage colony-stimulating factor, PDGFRB, and Colony stimulating factor 1 receptor, cause the Chromosome 5q deletion syndrome (5q- syndrome). This syndrome is a unique type of myelodysplastic syndrome characterized by a prolonged disease course, a low rate of transformation to an aggressive form of leukemia, and an anemia which in many patients is profound, refractory to traditional therapies (e.g. iron supplements, Erythropoietin), and requiring maintenance red blood cell transfusions. The disease is treated with a chemotherapy drug, lenalidomide. [5] [20]

PDGFRB Translocations

Human chromosome translocations between the PDGFRB gene and at least any one of 30 genes on other chromosomes lead to myeloid and/or lymphoid neoplasms that are many ways similar to the neoplasm caused by the fusion of the PDGFRA (i.e. platelet derived growth factor receptor A or alpha-type-platelet derived growth factor receptor) gene with the FIP1L1 gene (see FIP1L1-PDGFRA fusion gene. The most common of these rare mutations is the translocation of PDGFRB gene with the ETV6 gene (also termed ETS variant gene 6).

PDGFRB-ETV6 translocations

The ETV6 gene codes for a transcription factor protein that in mice appears to be required for hematopoiesis and maintenance of the developing vascular network. The gene is located on human chromosome 12 at the p13 position, consists of 14 exons, and is well-known to be involved in a large number of chromosomal rearrangements associated with leukemia and congenital fibrosarcoma. [21] Translocations between it and the PDGFRB gene, notated as t(5;12)(q33;p13), yield a PDGFRB-ETV6 fused gene that encodes a fusion protein, PDGFRB-ETV6. This chimeric protein, unlike the PDGFRB protein: a) has continuously active PDGFRB-mediated tyrosine kinase due to its forced dimerization by the PNT protein binding domain of the ETV6 protein; b) is highly stable due to its resistance to ubiquitin-Proteasome degradation; and c) therefore over-stimulates cell signaling pathways such as STAT5, NF-κB, and Extracellular signal-regulated kinases which promote cell growth and proliferation. This continuous signaling, it is presumed, leads to the development of myeloid and/or lymphoid neoplasms that commonly include increased numbers of blood born and tissue eosinophils, eosinophil-induced organ and tissue injury, and possible progression to aggressive form of leukemia. [22]

PDGFRB-ETV6 fusion protein-induced neoplasms often present with features that would classify them as Chronic myelomonocytic leukemias, juvenile myelomonocytic leukemia, Atypical or Philadelphia chromosome negative chronic myeloid leukemias, myelodysplastic syndromes, acute myelogenous leukemias, or acute lymphoblastic leukemias. The disease is now classified by the World Health Organization as one form of clonal eosinophilia. [23] It is critical that the PDGFRB-ETV6 fusion protein-driven disease be diagnostically distinguished from many of the just cited other diseases because of its very different treatment.

Patients with the PDGFRB-ETV6 fusion protein-driven disease are more often adult males but rarely children. They present with anemia, increases in blood eosinophils and monocytes, splenomegaly, and, less often, lymphadenopathy. Bone marrow examination may reveal cellular features similar to that seen in the aforementioned diseases. Diagnosis is may by conventional cytogenetic examination of blood or bone marrow cells to test for PDGFRB rearrangements using Fluorescence in situ hybridization or to test for the fused FDGFRB-ATV6 fluorescence in situ hybridization and/or Real-time polymerase chain reaction using appropriate nucleotide probes. [22] These patients, unlike many patients with similarly appearing neoplasms, respond well to the tyrosine kinase inhibitor, imatinib. The drug often causes long-term complete hematological and cytogenic remissions as doses well below those used to treat chronic myelogenous leukemia. Primary or acquired drug resistance to this drug is very rare. Additional adjuvant chemotherapy may be necessary if a patient's disease is unresponsive to tyrosine kinase inhibitor therapy and/or progresses to more aggressive disease phase similar to that seen in the blast crisis of chronic myelogenous leukemia. [22] [6]

Other PDGFRB translocations

The PDGFRB gene has been found to fuse with at least 36 other genes to form fusion genes that encode chimeric proteins that are known or presumed to possess: a) continuously active PDGFRB-derived tyrosine kinase activity; b) the ability to continuously stimulate the growth and proliferation of hematological stem cells; and c) the ability to cause myeloid and lymphoid neoplasms that commonly but not always are associated with eosinophilia. In all instances, these gene fusion diseases are considered types of clonal eosinophilia with recommended treatment regimens very different than those of similar hematological malignancies. The genes fusing to PDGFRB, their chromosomal location, and the notations describing their fused genes are given in the following table. [6] [22]

GenelocusnotationgenelocusnotationGenelocusnotationgenelocusnotationgenelocusnotationgenelocusnotation
TPM3 1q21t(1;5)(q21;q32) PDE4DIP 1q22t(1;5)(q22;q32) SPTBN1 2p16t(2;5)(p16;q32) GOLGA4 3p21-25t(3;5)(p21-25;q31-35)WRD48 [24] 3p21-22t(1;3;5)(p36;p21;q32) PRKG2 [25] 4q21t(4;5)(p21;q32)
CEP85L [26] 6q22t(5;6)(q32;q22) HIP1 7q11t(5;7)(q32;q11) KANK1 9q24t(5;9)(q32;q24) BCR 9q34t(5;9)(q32;q34) CCDC6 10q21t(5;10)(q32;q21H4(D10S170) [27] 10q21.2t(5;10)(q32;q21.2)
GPIAP1 [28] 11p13multiple ETV6 12p13t(5;12)q32;p13) ERC1 12p13.3t(5;12)(q32;p13.3) GIT2 12q24t(5;12)(q31-33;q24)NIN [29] 14q24t(5;14)(q32;q24 TRIP11 14q32t(5;14)(q32;q32)
CCDC88C [30] 14q32t(5;14)(q33;q32) TP53BP1 15q22t(5;15)q33;22) NDE1 16p13t(5;16)(q33;p13) SPECC1 17p11t(5;17)(q32;p11.2) NDEL1 17p13t(5;17)(q32;p13) MYO18A 17q11.2t(5;17)(q32;q11.2)
BIN2 [31] 12q13t(5;12)(q32;q13) COL1A1 17q22t(5;17)q32;q22)DTD1 [32] 20p11t(5;20)(q32;p11) CPSF6 12q15t(5;12)(q32;q15) RABEP1 17p13t(5;17)(q32;p13) MPRIP 17p11t(5;17)(q32;p11)
SPTBN1 2p16t(5;2)(q32;p16)WDR48 [24] 3p22t(5;3)q32;p22) GOLGB1 3q12t(3;5)(q12;q32) DIAPH1 5q31t(5;5)(q32;q31) TNIP1 5q33t(5;5)(q32;q33) SART3 12q23t(5;12)(q32;q23)

Similar to PDGFRB-ETV6 translocations, these translocations are generally in-frame and encode for fusion proteins with their PDGFRB-derived tyrosine kinase being continuously active and responsible for causing the potentially malignant growth of its myeloid and/or lymphoid harboring cells. Patients are usually middle-aged men. They commonly present with anemia, eosinophilia, monocytosis, and splenomegaly and have their disease classified as chronic myelomonocytic leukemia, atypical chronic myelomonocytic leukemia, juvenile myelomonocytic leukemia, myelodysplastic syndrome, acute myelogenous leukemia, acute lymphoblastic leukemia, or T lymphoblastic lymphoma. Diagnosis relies on cytogenetic analyses to detect breakpoints in the long arm of chromosome 5 by Fluorescence in situ hybridization. These patients usually respond well to imatinib therapy. [6] [22] [33]

Primary familial brain calcification

Primary familial brain calcification (see Fahr's syndrome) is a rare disease involving bilateral calcifications in the brain, predominantly in basal ganglia but also cerebellum, thalamus, and brainstem in patients presenting with diverse neurologic (e.g. movement disorders, parkinsonism, seizures, headache) features and psychiatric (e.g. cognitive impairment, mood disorders, psychotic symptoms, and obsessive-compulsive) disturbances. In a minority of cases, the disease is associated with apparent autosomal dominant loss of function mutations in PDGFRB or the gene which encodes a ligand that simulates PDGFRB, Platelet-derived growth factor, PDGFB. PDGFRB is extensively expressed in the neurons, chorioid plexus, vascular smooth muscle cells, and pericytes of the human brain, particularly the basal ganglia and the dentate nucleus. It is proposed that signal transduction through PDGFRB maintains blood–brain barrier integrity and that loss of the PDGFRB receptor or its ligand, PDGFB, disrupts the blood–brain barrier, subsequently promoting (peri)vascular calcium deposition and thereby causing the dysfunction and death of neurons. [34] [35]

Interactions

PDGFRB has been shown to interact with:

Notes

See also

Related Research Articles

<span class="mw-page-title-main">Tyrosine kinase</span> Class hi residues

A tyrosine kinase is an enzyme that can transfer a phosphate group from ATP to the tyrosine residues of specific proteins inside a cell. It functions as an "on" or "off" switch in many cellular functions.

<span class="mw-page-title-main">Philadelphia chromosome</span> Genetic abnormality in leukemia cancer cells

The Philadelphia chromosome or Philadelphia translocation (Ph) is a specific genetic abnormality in chromosome 22 of leukemia cancer cells. This chromosome is defective and unusually short because of reciprocal translocation, t(9;22)(q34;q11), of genetic material between chromosome 9 and chromosome 22, and contains a fusion gene called BCR-ABL1. This gene is the ABL1 gene of chromosome 9 juxtaposed onto the breakpoint cluster region BCR gene of chromosome 22, coding for a hybrid protein: a tyrosine kinase signaling protein that is "always on", causing the cell to divide uncontrollably by interrupting the stability of the genome and impairing various signaling pathways governing the cell cycle.

<span class="mw-page-title-main">Platelet-derived growth factor</span> Signaling glycoprotein regulating cell proliferation

Platelet-derived growth factor (PDGF) is one among numerous growth factors that regulate cell growth and division. In particular, PDGF plays a significant role in blood vessel formation, the growth of blood vessels from already-existing blood vessel tissue, mitogenesis, i.e. proliferation, of mesenchymal cells such as fibroblasts, osteoblasts, tenocytes, vascular smooth muscle cells and mesenchymal stem cells as well as chemotaxis, the directed migration, of mesenchymal cells. Platelet-derived growth factor is a dimeric glycoprotein that can be composed of two A subunits (PDGF-AA), two B subunits (PDGF-BB), or one of each (PDGF-AB).

<span class="mw-page-title-main">Chromosome 3</span> Human chromosome

Chromosome 3 is one of the 23 pairs of chromosomes in humans. People normally have two copies of this chromosome. Chromosome 3 spans 201 million base pairs and represents about 6.5 percent of the total DNA in cells.

<span class="mw-page-title-main">Platelet-derived growth factor receptor</span> Protein family

Platelet-derived growth factor receptors (PDGF-R) are cell surface tyrosine kinase receptors for members of the platelet-derived growth factor (PDGF) family. PDGF subunits -A and -B are important factors regulating cell proliferation, cellular differentiation, cell growth, development and many diseases including cancer. There are two forms of the PDGF-R, alpha and beta each encoded by a different gene. Depending on which growth factor is bound, PDGF-R homo- or heterodimerizes.

<span class="mw-page-title-main">Fibroblast growth factor receptor 1</span> Protein-coding gene in the species Homo sapiens

Fibroblast growth factor receptor 1 (FGFR1), also known as basic fibroblast growth factor receptor 1, fms-related tyrosine kinase-2 / Pfeiffer syndrome, and CD331, is a receptor tyrosine kinase whose ligands are specific members of the fibroblast growth factor family. FGFR1 has been shown to be associated with Pfeiffer syndrome, and clonal eosinophilias.

<span class="mw-page-title-main">ETV6</span> Protein-coding gene in the species Homo sapiens

ETV6 protein is a transcription factor that in humans is encoded by the ETV6 gene. The ETV6 protein regulates the development and growth of diverse cell types, particularly those of hematological tissues. However, its gene, ETV6 frequently suffers various mutations that lead to an array of potentially lethal cancers, i.e., ETV6 is a clinically significant proto-oncogene in that it can fuse with other genes to drive the development and/or progression of certain cancers. However, ETV6 is also an anti-oncogene or tumor suppressor gene in that mutations in it that encode for a truncated and therefore inactive protein are also associated with certain types of cancers.

<span class="mw-page-title-main">PDGFA</span> Mammalian protein found in Homo sapiens

Platelet-derived growth factor subunit A is a protein that in humans is encoded by the PDGFA gene.

<span class="mw-page-title-main">PDGFB</span> Protein-coding gene in the species Homo sapiens

Platelet-derived growth factor subunit B is a protein that in humans is encoded by the PDGFB gene.

<span class="mw-page-title-main">TEC (gene)</span> Human gene

Tyrosine-protein kinase Tec is a tyrosine kinase that in humans is encoded by the TEC gene. Tec kinase is expressed in hematopoietic, liver, and kidney cells and plays an important role in T-helper cell processes. Tec kinase is the name-giving member of the Tec kinase family, a family of non-receptor protein-tyrosine kinases.

<span class="mw-page-title-main">FIP1L1</span> Protein-coding gene in the species Homo sapiens

Factor interacting with PAPOLA and CPSF1 is a protein that in humans is encoded by the FIP1L1 gene. A medically important aspect of the FIP1L1 gene is its fusion with other genes to form fusion genes which cause clonal hypereosinophilia and leukemic diseases in humans.

<span class="mw-page-title-main">PDGFC</span> Protein-coding gene in the species Homo sapiens

Platelet-derived growth factor C, also known as PDGF-C, is a 345-amino acid protein that in humans is encoded by the PDGFC gene. Platelet-derived growth factors are important in connective tissue growth, survival and function, and consist of disulphide-linked dimers involving two polypeptide chains, PDGF-A and PDGF-B. PDGF-C is a member of the PDGF/VEGF family of growth factors with a unique two-domain structure and expression pattern. PDGF-C was not previously identified with PDGF-A and PDGF-B, possibly because it may be that it is synthesized and secreted as a latent growth factor, requiring proteolytic removal of the N-terminal CUB domain for receptor binding and activation.

<span class="mw-page-title-main">SH2B3</span> Protein-coding gene in the species Homo sapiens

SH2B adapter protein 3 (SH2B3), also known as lymphocyte adapter protein (LNK), is a protein that in humans is encoded by the SH2B3 gene on chromosome 12.

<span class="mw-page-title-main">Platelet-derived growth factor receptor A</span>

Platelet-derived growth factor receptor A, also termed CD140a, is a receptor located on the surface of a wide range of cell types. The protein is encoded in the human by the PDGFRA gene. This receptor binds to certain isoforms of platelet-derived growth factors (PDGFs) and thereby becomes active in stimulating cell signaling pathways that elicit responses such as cellular growth and differentiation. The receptor is critical for the embryonic development of certain tissues and organs, and for their maintenance, particularly hematologic tissues, throughout life. Mutations in PDGFRA, are associated with an array of clinically significant neoplasms, notably ones of the clonal hypereosinophilia class of malignancies, as well as gastrointestinal stromal tumors (GISTs).

Infantile myofibromatosis (IMF) is a rare tumor found in 1 in 150,000 to 1 in 400,000 live births. It is nonetheless the most common tumor derived from fibrous connective tissue that occurs primarily in infants and young children. IMF tumors are benign in the sense that they do not metastasize to distant tissues although when occurring in the viscera, i.e. internal organs, carry guarded to poor prognoses and can be life-threatening, particularly in newborns and young infants. The condition was first described by Arthur Purdy Stout as congenital generalized fibromatosis – in which he coined the word fibromatosis – in 1954.

ETV6-NTRK3 gene fusion is the translocation of genetic material between the ETV6 gene located on the short arm of chromosome 12 at position p13.2 and the NTRK3 gene located on the long arm of chromosome 15 at position q25.3 to create the (12;15)(p13;q25) fusion gene, ETV6-NTRK3. This new gene consists of the 5' end of ETV6 fused to the 3' end of NTRK3. ETV6-NTRK3 therefore codes for a chimeric oncoprotein consisting of the helix-loop-helix (HLH) protein dimerization domain of the ETV6 protein fused to the tyrosine kinase domain of the NTRK3 protein. The ETV6 gene codes for the transcription factor protein, ETV6, which suppresses the expression of, and thereby regulates, various genes that in mice are required for normal hematopoiesis as well as the development and maintenance of the vascular network. NTRK3 codes for Tropomyosin receptor kinase C a NT-3 growth factor receptor cell surface protein that when bound to its growth factor ligand, neurotrophin-3, becomes an active tyrosine kinase that phosphorylates tyrosine residues on, and thereby stimulates, signaling proteins that promote the growth, survival, and proliferation of their parent cells. The tyrosine kinase of the ETV6-NTRK3 fusion protein is dysfunctional in that it is continuously active in phosphorylating tyrosine residues on, and thereby continuously stimulating, proteins that promote the growth, survival, and proliferation of their parent cells. In consequence, these cells take on malignant characteristics and are on the pathway of becoming cancerous. Indeed, the ETV6-NTRK3 fusion gene appears to be a critical driver of several types of cancers. It was originally identified in congenital fibrosarcoma and subsequently found in mammary secretory carcinoma, mammary analogue secretory carcinoma of salivary glands, salivary gland–type carcinoma of the thyroid, secretory carcinoma of the skin, congenital fibrosarcoma, congenital mesoblastic nephroma, rare cases of acute myelogenous leukemia, ALK-negative Inflammatory myofibroblastic tumour, cholangiocarcinoma, and radiation-induced papillary thyroid carcinoma.

<span class="mw-page-title-main">Crenolanib</span> Chemical compound

Crenolanib besylate is an investigational inhibitor being developed by AROG Pharmaceuticals, LLC. The compound is currently being evaluated for safety and efficacy in clinical trials for various types of cancer, including acute myeloid leukemia (AML), gastrointestinal stromal tumor (GIST), and glioma. Crenolanib is an orally bioavailable benzimidazole that selectively and potently inhibits signaling of wild-type and mutant isoforms of class III receptor tyrosine kinases (RTK) FLT3, PDGFR α, and PDGFR β. Unlike most RTK inhibitors, crenolanib is a type I mutant-specific inhibitor that preferentially binds to phosphorylated active kinases with the ‘DFG in’ conformation motif.

Fibroblast growth factor receptor oncogene partner 2 (FGFR1OP2) was identified in a study on myeloproliferative syndrome (EMS). The study aimed to identify the partner genes to the fibroblast growth factor receptor 1 (FGFR1) involved in the syndrome. Using the 5'-RACE PCR technique, FGFR1OP2 was identified as a novel gene with no known function.

Mammary analogue secretory carcinoma (MASC), also termed MASCSG, is a salivary gland neoplasm. It is a secretory carcinoma which shares the microscopic pathologic features with other types of secretory carcinomas including mammary secretory carcinoma, secretory carcinoma of the skin, and salivary gland–type carcinoma of the thyroid. MASCSG was first described by Skálová et al. in 2010. The authors of this report found a chromosome translocation in certain salivary gland tumors, i.e. a (12;15)(p13;q25) fusion gene mutation. The other secretory carcinoma types carry this fusion gene.

Clonal hypereosinophilia, also termed primary hypereosinophilia or clonal eosinophilia, is a grouping of hematological disorders all of which are characterized by the development and growth of a pre-malignant or malignant population of eosinophils, a type of white blood cell that occupies the bone marrow, blood, and other tissues. This population consists of a clone of eosinophils, i.e. a group of genetically identical eosinophils derived from a sufficiently mutated ancestor cell.

References

  1. 1 2 3 GRCh38: Ensembl release 89: ENSG00000113721 - Ensembl, May 2017
  2. 1 2 3 GRCm38: Ensembl release 89: ENSMUSG00000024620 - Ensembl, May 2017
  3. "Human PubMed Reference:". National Center for Biotechnology Information, U.S. National Library of Medicine.
  4. "Mouse PubMed Reference:". National Center for Biotechnology Information, U.S. National Library of Medicine.
  5. 1 2 "PDGFRB platelet derived growth factor receptor beta [Homo sapiens (human)] - Gene - NCBI".
  6. 1 2 3 4 Reiter A, Gotlib J (2017). "Myeloid neoplasms with eosinophilia". Blood. 129 (6): 704–714. doi: 10.1182/blood-2016-10-695973 . PMID   28028030.
  7. Heldin CH, Lennartsson J (August 2013). "Structural and functional properties of platelet-derived growth factor and stem cell factor receptors". Cold Spring Harbor Perspectives in Biology. 5 (8): a009100. doi:10.1101/cshperspect.a009100. PMC   3721287 . PMID   23906712.
  8. Kelly JD, Haldeman BA, Grant FJ, Murray MJ, Seifert RA, Bowen-Pope DF, et al. (May 1991). "Platelet-derived growth factor (PDGF) stimulates PDGF receptor subunit dimerization and intersubunit trans-phosphorylation". The Journal of Biological Chemistry. 266 (14): 8987–92. doi: 10.1016/S0021-9258(18)31541-2 . PMID   1709159.
  9. Lei H, Kazlauskas A (2014). "A reactive oxygen species-mediated, self-perpetuating loop persistently activates platelet-derived growth factor receptor α". Molecular and Cellular Biology. 34 (1): 110–22. doi:10.1128/MCB.00839-13. PMC   3911282 . PMID   24190966.
  10. Lei H, Qian CX, Lei J, Haddock LJ, Mukai S, Kazlauskas A (2015). "RasGAP Promotes Autophagy and Thereby Suppresses Platelet-Derived Growth Factor Receptor-Mediated Signaling Events, Cellular Responses, and Pathology". Molecular and Cellular Biology. 35 (10): 1673–85. doi:10.1128/MCB.01248-14. PMC   4405646 . PMID   25733681.
  11. Soriano P (1994). "Abnormal kidney development and hematological disorders in PDGF beta-receptor mutant mice". Genes & Development. 8 (16): 1888–96. doi: 10.1101/gad.8.16.1888 . PMID   7958864.
  12. Lindahl P, Johansson BR, Levéen P, Betsholtz C (1997). "Pericyte loss and microaneurysm formation in PDGF-B-deficient mice". Science. 277 (5323): 242–5. doi:10.1126/science.277.5323.242. PMID   9211853.
  13. Lindahl P, Hellström M, Kalén M, Karlsson L, Pekny M, Pekna M, Soriano P, Betsholtz C (1998). "Paracrine PDGF-B/PDGF-Rbeta signaling controls mesangial cell development in kidney glomeruli". Development. 125 (17): 3313–22. doi:10.1242/dev.125.17.3313. PMID   9693135.
  14. Levéen P, Pekny M, Gebre-Medhin S, Swolin B, Larsson E, Betsholtz C (1994). "Mice deficient for PDGF B show renal, cardiovascular, and hematological abnormalities". Genes & Development. 8 (16): 1875–87. doi: 10.1101/gad.8.16.1875 . PMID   7958863.
  15. Darland DC, D'Amore PA (1999). "Blood vessel maturation: vascular development comes of age". The Journal of Clinical Investigation. 103 (2): 157–8. doi:10.1172/JCI6127. PMC   407889 . PMID   9916126.
  16. Olson LE, Soriano P (2011). "PDGFRβ signaling regulates mural cell plasticity and inhibits fat development". Developmental Cell. 20 (6): 815–26. doi:10.1016/j.devcel.2011.04.019. PMC   3121186 . PMID   21664579.
  17. Andrae J, Gallini R, Betsholtz C (2008). "Role of platelet-derived growth factors in physiology and medicine". Genes & Development. 22 (10): 1276–312. doi:10.1101/gad.1653708. PMC   2732412 . PMID   18483217.
  18. Heldin CH (2013). "Targeting the PDGF signaling pathway in tumor treatment". Cell Communication and Signaling. 11: 97. doi: 10.1186/1478-811X-11-97 . PMC   3878225 . PMID   24359404.
  19. Heldin CH (2014). "Targeting the PDGF signaling pathway in the treatment of non-malignant diseases". Journal of Neuroimmune Pharmacology. 9 (2): 69–79. doi:10.1007/s11481-013-9484-2. PMID   23793451. S2CID   17343813.
  20. Nimer SD (2006). "Clinical management of myelodysplastic syndromes with interstitial deletion of chromosome 5q". Journal of Clinical Oncology. 24 (16): 2576–82. doi:10.1200/JCO.2005.03.6715. PMID   16735711.
  21. "ETV6 ETS variant 6 [Homo sapiens (human)] - Gene - NCBI".
  22. 1 2 3 4 5 Vega F, Medeiros LJ, Bueso-Ramos CE, Arboleda P, Miranda RN (2015). "Hematolymphoid neoplasms associated with rearrangements of PDGFRA, PDGFRB, and FGFR1". American Journal of Clinical Pathology. 144 (3): 377–92. doi: 10.1309/AJCPMORR5Z2IKCEM . PMID   26276769.
  23. Arber DA, Orazi A, Hasserjian R, Thiele J, Borowitz MJ, Le Beau MM, Bloomfield CD, Cazzola M, Vardiman JW (2016). "The 2016 revision to the World Health Organization classification of myeloid neoplasms and acute leukemia". Blood. 127 (20): 2391–405. doi: 10.1182/blood-2016-03-643544 . PMID   27069254.
  24. 1 2 "WDR48 WD repeat domain 48 [Homo sapiens (human)] - Gene - NCBI".
  25. "PRKG2 protein kinase cGMP-dependent 2 [Homo sapiens (human)] - Gene - NCBI".
  26. "CEP85L centrosomal protein 85 like [Homo sapiens (human)] - Gene - NCBI".
  27. "CCDC6 coiled-coil domain containing 6 [Homo sapiens (human)] - Gene - NCBI".
  28. "CAPRIN1 cell cycle associated protein 1 [Homo sapiens (human)] - Gene - NCBI".
  29. "NIN ninein [Homo sapiens (human)] - Gene - NCBI".
  30. "CCDC88C coiled-coil domain containing 88C [Homo sapiens (human)] - Gene - NCBI".
  31. "BIN2 bridging integrator 2 [Homo sapiens (human)] - Gene - NCBI".
  32. "DTD1 D-tyrosyl-tRNA deacylase 1 [Homo sapiens (human)] - Gene - NCBI".
  33. Gotlib J (2015). "World Health Organization-defined eosinophilic disorders: 2015 update on diagnosis, risk stratification, and management". American Journal of Hematology. 90 (11): 1077–89. doi: 10.1002/ajh.24196 . PMID   26486351. S2CID   42668440.
  34. Tadic V, Westenberger A, Domingo A, Alvarez-Fischer D, Klein C, Kasten M (2015). "Primary familial brain calcification with known gene mutations: a systematic review and challenges of phenotypic characterization". JAMA Neurology. 72 (4): 460–7. doi:10.1001/jamaneurol.2014.3889. PMID   25686319.
  35. Batla A, Tai XY, Schottlaender L, Erro R, Balint B, Bhatia KP (2017). "Deconstructing Fahr's disease/syndrome of brain calcification in the era of new genes" (PDF). Parkinsonism & Related Disorders (Submitted manuscript). 37: 1–10. doi:10.1016/j.parkreldis.2016.12.024. PMID   28162874.
  36. Matsumoto T, Yokote K, Take A, Takemoto M, Asaumi S, Hashimoto Y, Matsuda M, Saito Y, Mori S (April 2000). "Differential interaction of CrkII adaptor protein with platelet-derived growth factor alpha- and beta-receptors is determined by its internal tyrosine phosphorylation". Biochem. Biophys. Res. Commun. 270 (1): 28–33. doi:10.1006/bbrc.2000.2374. PMID   10733900.
  37. Yamamoto M, Toya Y, Jensen RA, Ishikawa Y (March 1999). "Caveolin is an inhibitor of platelet-derived growth factor receptor signaling". Exp. Cell Res. 247 (2): 380–8. doi:10.1006/excr.1998.4379. PMID   10066366.
  38. 1 2 3 Braverman LE, Quilliam LA (February 1999). "Identification of Grb4/Nckbeta, a src homology 2 and 3 domain-containing adapter protein having similar binding and biological properties to Nck". J. Biol. Chem. 274 (9): 5542–9. doi: 10.1074/jbc.274.9.5542 . PMID   10026169.
  39. Arvidsson AK, Rupp E, Nånberg E, Downward J, Rönnstrand L, Wennström S, Schlessinger J, Heldin CH, Claesson-Welsh L (October 1994). "Tyr-716 in the platelet-derived growth factor beta-receptor kinase insert is involved in GRB2 binding and Ras activation". Mol. Cell. Biol. 14 (10): 6715–26. doi:10.1128/mcb.14.10.6715. PMC   359202 . PMID   7935391.
  40. Tang J, Feng GS, Li W (October 1997). "Induced direct binding of the adapter protein Nck to the GTPase-activating protein-associated protein p62 by epidermal growth factor". Oncogene. 15 (15): 1823–32. doi: 10.1038/sj.onc.1201351 . PMID   9362449.
  41. Li W, Hu P, Skolnik EY, Ullrich A, Schlessinger J (December 1992). "The SH2 and SH3 domain-containing Nck protein is oncogenic and a common target for phosphorylation by different surface receptors". Mol. Cell. Biol. 12 (12): 5824–33. doi:10.1128/MCB.12.12.5824. PMC   360522 . PMID   1333047.
  42. Chen M, She H, Davis EM, Spicer CM, Kim L, Ren R, Le Beau MM, Li W (September 1998). "Identification of Nck family genes, chromosomal localization, expression, and signaling specificity". J. Biol. Chem. 273 (39): 25171–8. doi: 10.1074/jbc.273.39.25171 . PMID   9737977.
  43. Chen M, She H, Kim A, Woodley DT, Li W (November 2000). "Nckbeta adapter regulates actin polymerization in NIH 3T3 fibroblasts in response to platelet-derived growth factor bb". Mol. Cell. Biol. 20 (21): 7867–80. doi:10.1128/mcb.20.21.7867-7880.2000. PMC   86398 . PMID   11027258.
  44. Rupp E, Siegbahn A, Rönnstrand L, Wernstedt C, Claesson-Welsh L, Heldin CH (October 1994). "A unique autophosphorylation site in the platelet-derived growth factor alpha receptor from a heterodimeric receptor complex". Eur. J. Biochem. 225 (1): 29–41. doi: 10.1111/j.1432-1033.1994.00029.x . PMID   7523122.
  45. Seifert RA, Hart CE, Phillips PE, Forstrom JW, Ross R, Murray MJ, Bowen-Pope DF (May 1989). "Two different subunits associate to create isoform-specific platelet-derived growth factor receptors". J. Biol. Chem. 264 (15): 8771–8. doi: 10.1016/S0021-9258(18)81860-9 . PMID   2542288.
  46. Keilhack H, Müller M, Böhmer SA, Frank C, Weidner KM, Birchmeier W, Ligensa T, Berndt A, Kosmehl H, Günther B, Müller T, Birchmeier C, Böhmer FD (January 2001). "Negative regulation of Ros receptor tyrosine kinase signaling. An epithelial function of the SH2 domain protein tyrosine phosphatase SHP-1". J. Cell Biol. 152 (2): 325–34. doi:10.1083/jcb.152.2.325. PMC   2199605 . PMID   11266449.
  47. Lechleider RJ, Sugimoto S, Bennett AM, Kashishian AS, Cooper JA, Shoelson SE, Walsh CT, Neel BG (October 1993). "Activation of the SH2-containing phosphotyrosine phosphatase SH-PTP2 by its binding site, phosphotyrosine 1009, on the human platelet-derived growth factor receptor". J. Biol. Chem. 268 (29): 21478–81. doi: 10.1016/S0021-9258(20)80562-6 . PMID   7691811.
  48. Farooqui T, Kelley T, Coggeshall KM, Rampersaud AA, Yates AJ (1999). "GM1 inhibits early signaling events mediated by PDGF receptor in cultured human glioma cells". Anticancer Res. 19 (6B): 5007–13. PMID   10697503.
  49. Ekman S, Kallin A, Engström U, Heldin CH, Rönnstrand L (March 2002). "SHP-2 is involved in heterodimer specific loss of phosphorylation of Tyr771 in the PDGF beta-receptor". Oncogene. 21 (12): 1870–5. doi: 10.1038/sj.onc.1205210 . PMID   11896619.
  50. Yokote K, Mori S, Hansen K, McGlade J, Pawson T, Heldin CH, Claesson-Welsh L (May 1994). "Direct interaction between Shc and the platelet-derived growth factor beta-receptor". J. Biol. Chem. 269 (21): 15337–43. doi: 10.1016/S0021-9258(17)36611-5 . PMID   8195171.
  51. Maudsley S, Zamah AM, Rahman N, Blitzer JT, Luttrell LM, Lefkowitz RJ, Hall RA (November 2000). "Platelet-derived growth factor receptor association with Na(+)/H(+) exchanger regulatory factor potentiates receptor activity". Mol. Cell. Biol. 20 (22): 8352–63. doi:10.1128/mcb.20.22.8352-8363.2000. PMC   102142 . PMID   11046132.

Further reading