Abstract
The Concise Guide to PHARMACOLOGY 2013/14 provides concise overviews of the key properties of over 2000 human drug targets with their pharmacology, plus links to an open access knowledgebase of drug targets and their ligands (www.guidetopharmacology.org), which provides more detailed views of target and ligand properties. The full contents can be found at http://onlinelibrary.wiley.com/doi/10.1111/bph.12444/full.
Catalytic receptors are one of the seven major pharmacological targets into which the Guide is divided, with the others being G protein-coupled receptors, ligand-gated ion channels, ion channels, nuclear hormone receptors, transporters and enzymes. These are presented with nomenclature guidance and summary information on the best available pharmacological tools, alongside key references and suggestions for further reading. A new landscape format has easy to use tables comparing related targets.
It is a condensed version of material contemporary to late 2013, which is presented in greater detail and constantly updated on the website www.guidetopharmacology.org, superseding data presented in previous Guides to Receptors and Channels. It is produced in conjunction with NC-IUPHAR and provides the official IUPHAR classification and nomenclature for human drug targets, where appropriate. It consolidates information previously curated and displayed separately in IUPHAR-DB and the Guide to Receptors and Channels, providing a permanent, citable, point-in-time record that will survive database updates.
An Introduction to Catalytic Receptors
Catalytic receptors are cell-surface proteins, usually dimeric in nature, which typically encompass ligand binding and functional domains in one polypeptide chain. The ligand binding domain is placed on the extracellular surface of the plasma membrane and separated from the functional domain by a single transmembrane-spanning domain of 20-25 hydrophobic amino acids. The functional domain on the intracellular face of the plasma membrane has catalytic activity, or interacts with particular enzymes, giving the superfamily of receptors its name. Endogenous agonists of the catalytic receptor superfamily are peptides or proteins, the binding of which may induce dimerization of the receptor, which is the functional version of the receptor.
Amongst the catalytic receptors, particular subfamilies may be readily identified dependent on the function of the enzymatic portion of the receptor. The smallest group is the particulate guanylyl cyclases of the natriuretic peptide receptor family. The most widely recognized group is probably the receptor tyrosine kinase (RTK) family, epitomized by the neurotrophin receptor family, where a crucial initial step is the activation of a signalling cascade by autophosphorylation of the receptor on intracellular tyrosine residue(s) catalyzed by enzyme activity intrinsic to the receptor. A third group is the extrinsic protein tyrosine kinase receptors, where the catalytic activity resides in a separate protein from the binding site. Examples of this group include the GDNF receptor families, where one, catalytically silent, member of the heterodimer is activated upon binding the ligand, causing the second member of the heterodimer, lacking ligand binding capacity, to initiate signaling through tyrosine phosphorylation. A fourth group, the receptor threonine/serine kinase (RTSK) family, exemplified by TGF-β and BMP receptors, has intrinsic serine/threonine protein kinase activity in the heterodimeric functional unit. A fifth group is the receptor tyrosine phosphatases (RTP), which generally appear to lack cognate ligands, but may be triggered by events such as cell:cell contact and have identified roles in the skeletal, hematopoietic and immune systems.
A new group of catalytic receptors for the Guide is the integrins, which have roles in cell : cell communication, often associated with signalling in the blood.
Acknowledgments
We wish to acknowledge the tremendous help provided by the Consultants to the Guides past and present (see list in the Overview, p. 1452). We are extremely grateful for the financial contributions from the British Pharmacological Society, the International Union of Basic and Clinical Pharmacology, the Wellcome Trust (099156/Z/12/Z]), which support the website and the University of Edinburgh, who host the guidetopharmacology.org website.
Conflict of interest
The authors state that there is no conflict of interest to disclose.
List of records presented
1678 Cytokine receptor family
1684 GDNF receptor family
1685 Integrins
1688 Natriuretic peptide receptor family
1689 Pattern Recognition receptors
1692 Receptor serine/threonine kinase (RSTK) family
1695 Receptor tyrosine kinases
1702 Receptor tyrosine phosphatases (RTP)
1703 Tumour necrosis factor (TNF) receptor family
Cytokine receptor family
Overview
Cytokines are not a clearly defined group of agents, other than having an impact on immune signalling pathways, although many cytokines have effects on other systems, such as in development. A feature of some cytokines, which allows them to be distinguished from hormones, is that they may be produced by “non-secretory” cells, for example, endothelial cells. Within the cytokine receptor family, some subfamilies may be identified, which are described elsewhere in the Guide to PHARMACOLOGY, receptors for the TNF family, the TGF-β family and the chemokines. Within this group of records are described Type I cytokine receptors, typified by interleukin receptors, and Type II cytokine receptors, exemplified by interferon receptors. These receptors possess a conserved extracellular region, known as the cytokine receptor homology domain (CHD), along with a range of other structural modules, including extracellular immunoglobulin (Ig)-like and fibronectin type III (FBNIII)-like domains, a transmembrane domain, and intracellular homology domains. An unusual feature of this group of agents is the existence of soluble and decoy receptors. These bind cytokines without allowing signalling to occur. A further attribute is the production of endogenous antagonist molecules, which bind to the receptors selectively and prevent signalling. A commonality of these families of receptors is the ligand-induced homo- or hetero-oligomerisation, which results in the recruitment of intracellular protein partners to evoke cellular responses, particularly in inflammatory or haematopoietic signalling. Although not an exclusive signalling pathway, a common feature of the majority of cytokine receptors is activation of the JAK/STAT pathway. This cascade is based around the protein tyrosine kinase activity of the Janus kinases (JAK), which phosphorylate the receptor and thereby facilitate the recruitment of signal transducers and activators of transcription (STATs). The activated homo- or heterodimeric STATs function principally as transcription factors in the nucleus.
Type I cytokine receptors are characterized by two pairs of conserved cysteines linked via disulfide bonds and a C-terminal WSXWS motif within their CHD. Type I receptors are commonly classified into five groups, based on sequence and structual homology of the receptor and its cytokine ligand, which is potentially more reflective of evolutionary relationships than an earlier scheme based on the use of common signal transducing chains within a receptor complex.
IL-2 receptor family
Overview
The IL-2 receptor family consists of one or more ligand-selective subunits, and a common γ chain (γc): IL2RG, P31785), though IL-4 and IL-7 receptors can form complexes with other receptor chains. Receptors of this family associate with Jak1 and Jak3, primarily activating Stat5, although certain family members can also activate Stat1, Stat3, or Stat6. Ro264550 has been described as a selective IL-2 receptor antagonist, which binds to IL-2 3.
Nomenclature | Interleukin-2 receptor | Interleukin-4 receptor type I | Interleukin-4 receptor type II | Interleukin-7 receptor | Interleukin-9 receptor |
Subunits | Interleukin-2 receptor α subunit (Ligand-binding subunit), Interleukin-2 receptor β subunit (Ligand-binding subunit), Interleukin-2 receptor γ subunit (Other subunit) | Interleukin 4 receptor (Ligand-binding subunit), Interleukin-2 receptor γ subunit (Other subunit) | Interleukin 4 receptor (Ligand-binding subunit), Interleukin 13 receptor, α1 (Other subunit) | Interleukin 7 receptor (Ligand-binding subunit), Interleukin-2 receptor γ subunit (Other subunit) | Interleukin 9 receptor (Ligand-binding subunit), Interleukin-2 receptor γ subunit (Other subunit) |
Endogenous agonists | IL-2 (IL2, P60568) | IL-4 (IL4, P05112) | IL-13 (IL13, P35225), IL-4 (IL4, P05112) | IL-7 (IL7, P13232) | IL-9 (IL9, P15248) |
Endogenous antagonists | IL-1 receptor antagonist (IL1RN, P18510) | – | – | – | – |
Selective antagonists | AF12198 1, Ro264550 3 | – | – | – | – |
Nomenclature | Interleukin 13 receptor, α2 | Interleukin-15 receptor | Interleukin-21 receptor | Thymic stromal lymphopoietin receptor |
HGNC, UniProt | IL13RA2, Q14627 | – | – | – |
Subunits | – | Interleukin-2 receptor β subunit (Ligand-binding subunit), Interleukin 15 receptor, α subunit (Ligand-binding subunit), Interleukin-2 receptor γ subunit (Other subunit) | Interleukin 21 receptor (Ligand-binding subunit), Interleukin-2 receptor γ subunit (Other subunit) | Interleukin 7 receptor (Ligand-binding subunit), Cytokine receptor-like factor 2 (Other subunit) |
Endogenous agonists | – | IL-15 (IL15, P40933) | IL-21 (IL21, Q9HBE4) | TSLP (TSLP, Q969D9) |
Comment | Decoy receptor that binds IL-13 (IL13, P35225) as a monomer. | – | – | – |
IL-3 receptor family
Overview
The IL-3 receptor family signal through a receptor complex comprising of a ligand-specific α subunit and a common β chain (CSF2RB, P32927), which is associated with Jak2 and signals primarily through Stat5.
Nomenclature | Interleukin-3 receptor | Interleukin-5 receptor | Granulocyte macrophage colony-stimulating factor receptor |
Subunits | Interleukin 3 receptor, α subunit (Ligand-binding subunit), Cytokine receptor common β subunit (Other subunit) | Interleukin 5 receptor, α subunit (Ligand-binding subunit), Cytokine receptor common β subunit (Other subunit) | GM-CSF receptor, α subunit (Ligand-binding subunit), Cytokine receptor common β subunit (Other subunit) |
Endogenous agonists | IL-3 (IL3, P08700) | IL-5 (IL5, P05113) | G-CSF (CSF3, P09919), GM-CSF (CSF2, P04141) |
Selective antagonists | – | YM90709 2 | – |
IL-6 receptor family
Overview
The IL-6 receptor family signal through a ternary receptor complex consisting of the cognate receptor and either the IL-6 signal transducer gp130 (IL6ST, P40189) or the oncostatin M-specific receptor, β subunit (OSMR, Q99650), which then activates the JAK/STAT, Ras/Raf/MAPK and PI 3-kinase/PKB signalling modules. Unusually amongst the cytokine receptors, the CNTF receptor is a glycerophosphatidylinositol-linked protein.
Nomenclature | Interleukin-6 receptor | Interleukin-11 receptor | Interleukin-31 receptor | Ciliary neutrophic factor receptor |
Subunits | Interleukin-6 receptor, α subunit (Ligand-binding subunit), Interleukin-6 receptor, β subunit (Other subunit) | Interleukin-11 receptor, α subunit (Ligand-binding subunit), Interleukin-6 receptor, β subunit (Other subunit) | Interleukin-31 receptor, α subunit (Ligand-binding subunit), Oncostatin M-specific receptor, β subunit (Other subunit) | Ciliary neurotrophic factor receptor α subunit (Ligand-binding subunit), Leukemia inhibitory factor receptor (Other subunit), Interleukin-6 receptor, β subunit |
Endogenous agonists | IL-6 (IL6, P05231) | IL-11 (IL11, P20809) | IL-31 (IL31, Q6EBC2) | CNTF (CNTF, P26441), CRCF1/CLCF1 heterodimer (CRLF1, CLCF1, O75462, Q9UBD9) |
Nomenclature | Leptin receptor | Leukemia inhibitory factor receptor | Oncostatin-M receptor | Interleukin-27 receptor |
HGNC, UniProt | LEPR, P48357 | – | – | – |
Subunits | – | Leukemia inhibitory factor receptor (Ligand-binding subunit), Interleukin-6 receptor, β subunit (Other subunit) | Oncostatin M-specific receptor, β subunit (Ligand-binding subunit), Interleukin-6 receptor, β subunit (Other subunit) | Interleukin 27 receptor, alpha (Ligand-binding subunit), Interleukin-6 receptor, β subunit (Other subunit) |
Endogenous agonists | leptin (LEP, P41159) | CTF1 (CTF1, Q16619), LIF (LIF, P15018), OSM (OSM, P13725) | OSM (OSM, P13725) | IL-27 (IL27, EBI3, Q14213, Q8NEV9) |
IL-12 receptor family
Overview
IL-12 receptors are a subfamily of the IL-6 receptor family. IL12RB1 is shared between receptors for IL-12 and IL-23; the functional agonist at IL-12 receptors is a heterodimer of IL-12A/IL-12B, while that for IL-23 receptors is a heterodimer of IL-12B/IL-23A.
Subunits
Prolactin receptor family
Overview
Prolactin family receptors form homodimers in the presence of their respective ligands, associate exclusively with Jak2 and signal via Stat5.
Nomenclature | Eythropoietin receptor | Granulocyte colony-stimulating factor receptor | Growth hormone receptor | Prolactin receptor | Thrombopoietin receptor |
HGNC, UniProt | EPOR, P19235 | CSF3R, Q99062 | GHR, P10912 | PRLR, P16471 | MPL, P40238 |
Endogenous agonists | erythropoietin (EPO, P01588) | G-CSF (CSF3, P09919) | growth hormone 1 (GH1, P01241), growth hormone 2 (GH2, P01242) | choriomammotropin (CSH1, CSH2, P01243), chorionic somatomammotropin hormone-like 1 (CSHL1, Q14406), prolactin (PRL, P01236) | thrombopoietin (THPO, P40225) |
Type II cytokine receptors also have two pairs of conserved cysteines but with a different arrangement to Type I and also lack the WSXWS motif.
Interferon receptor family
Overview
The interferon receptor family includes receptors for type I (α, β κ and ω) and type II (γ) interferons. There are at least 13 different genesencoding IFN-Α subunits in a cluster on human chromosome 9p22: α1 (IFNA1, P01562), α2 (IFNA2, P01563), α4 (IFNA4, P05014), α5 (IFNA5, P01569), α6 (IFNA6, P05013), α7 (IFNA7, P01567), α8 (IFNA8, P32881), α10 (IFNA10, P01566), α13 (IFNA13, P01562), α14 (IFNA14, P01570), α16 (IFNA16, P05015), α17 (IFNA17, P01571) and α21 (IFNA21, P01568).
Nomenclature | Interferon-α/β receptor | Interferon-γ receptor |
Subunits | interferon α/β receptor 1 (Ligand-binding subunit), Interferon α/β receptor 2 (Other subunit) | Interferon γ receptor 1 (Ligand-binding subunit), Interferon γ receptor 2 (Other subunit) |
Endogenous agonists | IFN-α10 (IFNA10, P01566), IFN-α1/13 (IFNA1, IFNA13, P01562), IFN-α14 (IFNA14, P01570), IFN-α16 (IFNA16, P05015), IFN-α17 (IFNA17, P01571), IFN-α2 (IFNA2, P01563), IFN-α21 (IFNA21, P01568), IFN-α4 (IFNA4, P05014), IFN-α5 (IFNA5, P01569), IFN-α6 (IFNA6, P05013), IFN-α7 (IFNA7, P01567), IFN-α8 (IFNA8, P32881), IFN-β (IFNB1, P01574), IFN-κ (IFNK, Q9P0W0), IFN-ω (IFNW1, P05000) | IFN-γ (IFNG, P01579) |
IL-10 receptor family
Overview
The IL-10 family of receptors are heterodimeric combinations of family members: IL10RA/IL10RB responds to IL-10; IL20RA/IL20RB responds to IL-19, IL-20 and IL-24; IL22RA1/IL20RB responds to IL-20 and IL-24; IL22RA1/IL10RB responds to IL-22; IL28RA/IL10RB responds to IL-28A, IL28B and IL-29.
Nomenclature | Interleukin-10 receptor | Interleukin-20 receptor | Interleukin-22α1/20β heteromer | Interleukin-22α1/10β heteromer | Interleukin-22 receptor α2 | Interferon-λ receptor 1 |
HGNC, UniProt | – | – | – | – | IL22RA2, Q969J5 | – |
Subunits | Interleukin 10 receptor, α subunit (Ligand-binding subunit), Interleukin 10 receptor, β subunit (Other subunit) | Interleukin 20 receptor, α subunit (Ligand-binding subunit), Interleukin 20 receptor, β subunit (Other subunit) | Interleukin 20 receptor, β subunit (Ligand-binding subunit), Interleukin 22 receptor, α1 subunit (Ligand-binding subunit) | Interleukin 10 receptor, β subunit (Ligand-binding subunit), Interleukin 22 receptor, α1 subunit (Ligand-binding subunit) | – | Interferon-λ receptor 1 (Ligand-binding subunit), Interleukin 10 receptor, β subunit (Other subunit) |
Endogenous agonists | IL-10 (IL10, P22301) | IL-19 (IL19, Q9UHD0), IL-20 (IL20, Q9NYY1), IL-24 (IL24, Q13007) | IL-20 (IL20, Q9NYY1), IL-24 (IL24, Q13007) | IL-22 (IL22, Q9GZX6) | – | IFN-λ1 (IFNL1, Q8IU54), IFN-λ2 (IFNL2, Q8IZJ0), IFN-λ3 (IFNL3, Q8IZI9) |
Comment | – | – | – | – | Soluble decoy receptor that binds IL-22 (IL22, Q9GZX6) as a monomer | – |
Immunoglobulin-like family of IL-1 receptors
Overview
The immunoglobulin-like family of IL-1 receptors are heterodimeric receptors made up of a cognate receptor subunit and an IL-1 receptor accessory protein, IL1RAP (Q9NPH3, also known as C3orf13, IL-1RAcP, IL1R3). They are characterised by extracellular immunoglobulin-like domains and an intracellular Toll/Interleukin-1R (TIR) domain.
Nomenclature | Interleukin-1 receptor, type I | Interleukin-33 receptor | Interleukin-36 receptor | Interleukin-1 receptor, type II | Interleukin-18 receptor |
Subunits | Interleukin 1 receptor, type I (Ligand-binding subunit), IL-1 receptor accessory protein (Other subunit) | Interleukin-1 receptor-like 1 (Ligand-binding subunit), IL-1 receptor accessory protein (Other subunit) | Interleukin-1 receptor-like 2 (Ligand-binding subunit), IL-1 receptor accessory protein (Other subunit) | Interleukin 1 receptor, type II (Ligand-binding subunit), IL-1 receptor accessory protein (Other subunit) | Interleukin-18 1 (Ligand-binding subunit), IL-18 receptor accessory protein (Other subunit) |
Endogenous agonists | IL-1α (IL1A, P01583), IL-1β (IL1B, P01584) | IL-33 (IL33, O95760) | IL-36α (IL36A, Q9UHA7), IL-36β (IL36B, Q9NZH7), IL-36γ (IL36G, Q9NZH8), | – | IL-18 (IL18, Q14116), IL-37 (IL37, Q9NZH6) |
Endogenous antagonists | IL-1 receptor antagonist (IL1RN, P18510) | – | IL-36 receptor antagonist (IL36RN, Q9UBH0) | – | – |
Selective antagonists | AF12198 1 | – | – | – | – |
Comment | – | – | IL-36 receptor antagonist (IL36RN, Q9UBH0) is a highly specific antagonist of the response to IL-36γ (IL36G, Q9NZH8) | Decoy receptor that binds IL-1α (IL1A, P01583), IL-1β (IL1B, P01584) and IL-1 receptor antagonist (IL1RN, P18510) | – |
IL-17 receptor family
Overview
The IL17 cytokine family consists of six ligands (IL-17A-F), which signal through five receptors (IL-17RA-E).
Nomenclature | Interleukin-17 receptor | Interleukin-25 receptor | Interleukin-17C receptor | Interleukin-17 receptor D |
HGNC, UniProt | – | – | – | IL17RD, Q8NFM7 |
Subunits | Interleukin 17 receptor A (Ligand-binding subunit), interleukin 17 receptor C (Other subunit) | Interleukin 17 receptor B (Ligand-binding subunit), Interleukin 17 receptor A (Other subunit) | Interleukin 17 receptor E (Ligand-binding subunit), Interleukin 17 receptor A (Other subunit) | – |
Endogenous agonists | IL-17A (IL17A, Q16552), IL-17A/IL-17F (IL17F, IL17A, Q16552, Q96PD4), IL-17F (IL17F, Q96PD4) | IL-17B (IL17B, Q9UHF5), IL-25 (IL25, Q9H293) | IL-17C (IL17C, Q9P0M4) | The endogenous agonist for this receptor is unknown |
GDNF receptor family
Overview
GDNF family receptors (provisional nomenclature) are extrinsic tyrosine kinase receptors. Ligand binding to the extracellular domain of the glycosylphosphatidylinositol-linked cell-surface receptors (tabulated below) activates a transmembrane tyrosine kinase enzyme, RET (see Receptor Tyrosine Kinases). The endogenous ligands are typically dimeric, linked through disulphide bridges: glial cell-derived neurotrophic factor GDNF (GDNF, P39905) (211 aa); neurturin NRTN (NRTN, Q99748), 197 aa); artemin (ARTN (ARTN, Q5T4W7), 237 aa) and PSPN (PSPN, O60542) (PSPN, 156 aa).
Nomenclature | GDNF family receptor α1 | GDNF family receptor α2 | GDNF family receptor α3 | GDNF family receptor α4 |
Common abbreviation | GFRα1 | GFRα2 | GFRα3 | GFRα4 |
HGNC, UniProt | GFRA1, P56159 | GFRA2, O00451 | GFRA3, O60609 | GFRA4, Q9GZZ7 |
Potency order | GDNF (GDNF, P39905) > NRTN (NRTN, Q99748) > ARTN (ARTN, Q5T4W7) | NRTN (NRTN, Q99748) > GDNF (GDNF, P39905) | ARTN (ARTN, Q5T4W7) | PSPN (PSPN, O60542) |
Radioligands (Kd) | [125I]GDNF (rat) (3x10-12 – 6.3x10-11 M) 4,6 | – | – | – |
Comments
Inhibitors of other receptor tyrosine kinases, such as semaxinib, which inhibits VEGF receptor function, may also inhibit Ret function 5. Mutations of RET and GDNF genes may be involved in Hirschsprung's disease, which is characterized by the absence of intramural ganglion cells in the hindgut, often resulting in intestinal obstruction.
Integrins
Overview
Integrins (provisional nomenclature) are heterodimeric entities, composed of α and β subunits, each 1TM proteins, which bind components of the extracellular matrix or counter-receptors expressed on other cells. One class of integrin contains an inserted domain (I) in its α subunit, and if present (in α1, α2, α10, α11, αD, αE, αL, αM and αX), this I domain contains the ligand binding site. All β subunits possess a similar I-like domain, which has the capacity to bind ligand, often recognising the RGD motif. The presence of an α subunit I domain precludes ligand binding through the β subunit. Integrins provide a link between ligand and the actin cytoskeleton (through typically short intracellular domains). Integrins bind several divalent cations, including a Mg2+ atom in the I or I-like domain that is essential for ligand binding. Other cation binding sites may regulate integrin activity or stabilise the 3D structure. Integrins regulate the activity of particular protein kinases, including focal adhesion kinase and integrin-linked kinase. Cellular activation regulates integrin ligand affinity via inside-out signalling and ligand binding to integrins can regulate cellular activity via outside-in signalling.
Nomenclature | Subunits | Ligands | Selective inhibitors (pIC50) | Comment |
α1β1 | integrin, alpha 1 subunit, integrin, beta 1 subunit (fibronectin receptor, beta polypeptide, antigen CD29 includes MDF2, MSK12) | collagen, laminin | obtustatin (9.1) 11 | – |
α2β1 | integrin, alpha 2 subunit (CD49B, alpha 2 subunit of VLA-2 receptor), integrin, beta 1 subunit (fibronectin receptor, beta polypeptide, antigen CD29 includes MDF2, MSK12) | collagen, laminin, thrombospondin | TCI15 (7.9) 13 | – |
αIIbβ3 | integrin, alpha 2b subunit (platelet glycoprotein IIb of IIb/IIIa complex, antigen CD41), integrin, beta 3 subunit (platelet glycoprotein IIIa, antigen CD61) | fibrinogen, fibronectin, von Willebrand factor, vitronectin, thrombospondin | abciximab, eptifibatide, G4120 12, GR144053, Syk inhibitor III 14, tirofiban | – |
α4β1 | integrin, alpha 4 subunit (antigen CD49D, alpha 4 subunit of VLA-4 receptor), integrin, beta 1 subunit (fibronectin receptor, beta polypeptide, antigen CD29 includes MDF2, MSK12) | fibronectin, VCAM-1, osteopontin, thrombospondin | natalizumab, TCS2314, BIO1211 (8.3 – 9.0) 9 | LDV-FITC is used as a probe at this receptor |
αLβ2 | integrin, alpha L subunit (antigen CD11A (p180), lymphocyte function-associated antigen 1; alpha polypeptide), integrin, beta 2 subunit (complement component 3 receptor 3 and 4 subunit) | ICAM-1, ICAM-2 | efalizumab, A286982 (7.4 – 7.5) 10 | – |
αVβ3 | integrin, alpha V subunit, integrin, beta 3 subunit (platelet glycoprotein IIIa, antigen CD61) | vitronectin, fibronectin, fibrinogen, osteopontin, von Willebrand factor, thrombospondin, tenascin | etaracizumab, echistatin (11.7) 8, P11 (11.6) 8, cilengitide (8.5) 7 | – |
Subunits
Nomenclature | HGNC, UniProt |
integrin, alpha 1 subunit | ITGA1, P56199 |
integrin, alpha 2 subunit (CD49B, alpha 2 subunit of VLA-2 receptor) | ITGA2, P08514 |
integrin, alpha 2b subunit (platelet glycoprotein IIb of IIb/IIIa complex, antigen CD41) | ITGA2B, P17301 |
integrin, alpha 3 subunit (antigen CD49C, alpha 3 subunit of VLA-3 receptor) | ITGA3, P26006 |
integrin, alpha 4 subunit (antigen CD49D, alpha 4 subunit of VLA-4 receptor) | ITGA4, P13612 |
integrin, alpha 5 subunit (fibronectin receptor, alpha polypeptide) | ITGA5, P08648 |
integrin, alpha 6 subunit | ITGA6, P23229 |
integrin, alpha 7 subunit | ITGA7, Q13683 |
integrin, alpha 8 subunit | ITGA8, P53708 |
integrin, alpha 9 subunit | ITGA9, Q13797 |
integrin, alpha 10 subunit | ITGA10, O75578 |
integrin, alpha 11 subunit | ITGA11, Q9UKX5 |
integrin, alpha D subunit | ITGAD, Q13349 |
integrin, alpha E subunit (antigen CD103, human mucosal lymphocyte antigen 1; alpha polypeptide) | ITGAE, P38570 |
integrin, alpha L subunit (antigen CD11A (p180), lymphocyte function-associated antigen 1; alpha polypeptide) | ITGAL, P20701 |
integrin, alpha M subunit (complement component 3 receptor 3 subunit) | ITGAM, P11215 |
integrin, alpha V subunit | ITGAV, P06756 |
integrin, alpha X subunit (complement component 3 receptor 4 subunit) | ITGAX, P20702 |
Nomenclature | HGNC, UniProt |
integrin, beta 1 subunit (fibronectin receptor, beta polypeptide, antigen CD29 includes MDF2, MSK12) | ITGB1, P05556 |
integrin, beta 2 subunit (complement component 3 receptor 3 and 4 subunit) | ITGB2, P05107 |
integrin, beta 3 subunit (platelet glycoprotein IIIa, antigen CD61) | ITGB3, P05106 |
integrin, beta 4 subunit | ITGB4, P16144 |
integrin, beta 5 subunit | ITGB5, P18084 |
integrin, beta 6 subunit | ITGB6, P18564 |
integrin, beta 7 subunit | ITGB7, P26010 |
integrin, beta 8 subunit | ITGB8, P26012 |
Integrin ligands
Collagen is the most abundant protein in metazoa, rich in glycine and proline residues, made up of cross-linked triple helical structures, generated primarily by fibroblasts. Extensive post-translational processing is conducted by prolyl and lysyl hydroxylases, as well as transglutaminases. Over 40 genes for collagen-α subunits have been identified in the human genome. The collagen-binding integrins α1β1, α2β1, α10β1 and α11β1 recognise a range of triple-helical peptide motifs including GFOGER (O = hydroxyproline), a synthetic peptide.
Laminin is an extracellular glycoprotein composed of α, β and γ chains, for which five, four and three genes, respectively, are identified in the human genome. It binds to α1β1, α2β1, α3,β1, α7β1 and α6β4 integrins10.
Fibrinogen is a glycosylated hexamer composed of two α (FGA, P02671), two β (FGB, P02675) and two γ (FGG, P02679,) subunits, linked by disulphide bridges. It is found in plasma and alpha granules of platelets. It forms cross-links between activated platelets mediating aggregation by binding αIIbβ3; proteolysis by thrombin cleaves short peptides termed fibrinopeptides to generate fibrin, which polymerises as part of the blood coagulation cascade.
Fibronectin is a disulphide-linked homodimer found as two major forms; a soluble dimeric form found in the plasma and a tissue version that is polymeric, which is secreted into the extracellular matrix by fibroblasts. Splice variation of the gene product (FN1, P02751) generates multiple isoforms.
Vitronectin is a serum glycoprotein and extracellular matrix protein (VTN, P04004) which is found either as a monomer or, following proteolysis, a disulphide -linked dimer.
Osteopontin forms an integral part of the mineralized matrix in bone (SPP1, P10451), where it undergoes extensive post-translation processing, including proteolysis and phosphorylation.
Von Willebrand factor (VWF, P04275) is a glycoprotein synthesised in vascular endothelial cells as a disulphide-linked homodimer, but multimerises further in plasma and is deposited on vessel wall collagen as a high molecular weight multimer. It is responsible for capturing platelets under arterial shear flow (via GPIb) and in thrombus propagation (via integrin αIIbβ3).
Natriuretic peptide receptor family
Overview
Natriuretic peptide receptors (provisional nomenclature) are a family of homodimeric, catalytic receptors with a single TM domain and guanylyl cyclase (EC 4.6.1.2) activity on the intracellular domain of the protein sequence. Isoforms are activated by the peptide hormones atrial natriuretic peptide (ANP (NPPA, P01160)), brain natriuretic peptide (BNP (NPPB, P16860)) and C-type natriuretic peptide (CNP (NPPC, P23582)). Another family member is GC-C, the receptor for guanylin (GUCA2A, Q02747) and uroguanylin (GUCA2B, Q16661). Family members have conserved ligand-binding, catalytic (guanylyl cyclase) and regulatory domains with the exception of NPR-C which has an extracellular binding domain homologous to that of other NPRs, but with a truncated intracellular domain which appears to couple, via the Gi/o family of G-proteins, to activation of phospholipase C, inwardly-rectifying potassium channels and inhibition of adenylyl cyclase activity 25.
Nomenclature | NPR-A | NPR-B | NPR-C | guanylate cyclase 2C (heat stable enterotoxin receptor) |
HGNC, UniProt | NPR1, P16066 | NPR2, P20594 | NPR3, P17342 | GUCY2C, P25092 |
Potency order | ANP (NPPA, P01160) ≥ BNP (NPPB, P16860) >> CNP (NPPC, P23582) 27 | CNP (NPPC, P23582) >> ANP (NPPA, P01160) >> BNP (NPPB, P16860) 27 | ANP (NPPA, P01160) > CNP (NPPC, P23582) ≥ BNP (NPPB, P16860) 27 | uroguanylin (GUCA2B, Q16661) > guanylin (GUCA2A, Q02747) |
Endogenous agonists | ANP (NPPA, P01160) (Selective) 26, BNP (NPPB, P16860) (Selective) 26 | CNP (NPPC, P23582) (Selective) 27 | osteocrin (OSTN, P61366) (Selective) 23 | – |
Selective agonists | sANP 26 | – | cANF4-23 22 | E. coli heat-stable enterotoxin (STa), linaclotide 18 |
Selective antagonists | anantin 29, A-71915 (pKi 9.2 – 9.5) 15, [Asu7,23']β-ANP-(7-28) (pKi 7.5) 21 | monoclonal antibody 3G12 17, [Ser11](N-CNP,C-ANP)pBNP2-15 16 | M372049 19, AP811 (pKi 9.3) 28 | – |
Radioligands (Kd) | [125I]ANP | [125I]CNP (human) | [125I]ANP | [125I]Sta |
Comments
The polysaccharide obtained from fermentation of Aureobasidium species, HS142-1, acts as an antagonist at both NPR-A and NPR-B receptors 24. GUCY2D (RetGC1, GC-E, Q02846) and GUCY2F (RetGC2, GC-F, P51841) are predominantly retinal guanylyl cyclase activities, which are inhibited by calcium ions acting through the guanylyl cyclase activating peptides GCAP1 (GUCA1A, 43080), GCAP2 (GUCA1B, Q9UMX6) and GCAP3 (GUCA1C, O95843) 20.
Pattern Recognition receptors
Overview
Pattern recognition receptors (PRR, 42) participate in the innate immune response to microbial agents, the stimulation of which leads to activation of intracellular enzymes and regulation of gene transcription. PRR include both cell-surface and intracellular proteins, including toll-like receptors (TLR), nucleotide-binding oligomerization domain-like receptors (NLR, also known as NOD-like receptors) and the mannose receptor family (ENSFM00250000004089). PRR may be divided into signalling-associated members, identified here, and endocytic members (such as the mannose receptor family), the function of which appears to be to recognise particular microbial motifs for subsequent cell attachment, internalisation and destruction.
PRRs express multiple leucine-rich regions to bind a range of microbially-derived ligands, termed PAMPs or pathogen-associated molecular patterns, which includes peptides, carbohydrates, peptidoglycans, lipoproteins, lipopolysaccharides, and nucleic acids.
Toll-like receptor family
Overview
Members of this family share significant homology with the interleukin-1 receptor family and appear to require dimerization either as homo- or heterodimers for functional activity. Heterodimerization appears to influence the potency of ligand binding substantially (e.g. TLR1/2 and TLR2/6, 43,44). TLR1, TLR2, TLR4, TLR5, TLR6 and TLR11 are cell-surface proteins, while other members are associated with intracellular organelles, signalling through the MyD88-dependent pathways (with the exception of TLR3). As well as responding to exogenous infectious agents, it has been suggested that selected members of the family may be activated by endogenous ligands, such as hsp60 (HSPD1, P10809) 38.
Nomenclature | HGNC, UniProt | Agonists | Comment |
TLR1 | TLR1, Q15399 | – | – |
TLR2 | TLR2, O60603 | peptidoglycan 41,45 | – |
TLR3 | TLR3, O15455 | polyIC 30 | – |
TLR4 | TLR4, O00206 | LPS 39, taxol 36 | eritoran (E5564) is a lipid A analogue, which has been described as a TLR4 antagonist 35 |
TLR5 | TLR5, O60602 | flagellin 31 | – |
TLR6 | TLR6, Q9Y2C9 | – | – |
TLR7 | TLR7, Q9NYK1 | imiquimod 33, loxoribine 32, R848 33 | – |
TLR8 | TLR8, Q9NR97 | imiquimod, R848 33 | – |
TLR9 | TLR9, Q9NR96 | CpG 34 | – |
TLR10 | TLR10, Q9BXR5 | – | – |
TLR11 | –, Q6R5P0 | – | Found in the mouse |
NOD-like receptor family
Overview
Structural analysis has identified a common motif of a mid-peptide located nucleotide-binding and oligomerization (NACHT) domain, which allows division of NOD-like receptors into three subfamilies, NLRC (or NODs), NLRP (or NALP) and IPAF 40. NLRC members are named on the basis of a sequence motif expressed at their N-termini, the caspase recruitment domain (CARD), while NLRP members have a pyrin domain. NLRs express C-terminal leucine-rich regions which have regulatory function and appear to recognize the microbial products to which the NLRs respond. NLRC family members recruit a serine/threonine kinase RIPK2 (receptor-interacting serine/threonine kinase 2, O43353, also known as CARD3, CARDIAK, RICK, RIP2) leading to signalling through NFκB and MAP kinase. NLRP family members, upon activation, recruit adaptor proteins (e.g. ASC, also known as PYCARD, CARD5, TMS-1, Q9ULZ3). Activated NLRs associate in multiprotein complexes, known as inflammasomes 40, allowing the recruitment of caspases.
Nomenclature | HGNC, UniProt | Agonists | Comment |
NLRC1 | NOD1, Q9Y239 | meso-DAP | – |
NLRC2 | NOD2, Q9HC29 | muramyl dipeptide | – |
NLRC3 | NLRC3, Q7RTR2 | – | – |
NLRC5 | NLRC5, Q86WI3 | – | – |
NLRX1 | NLRX1, Q86UT6 | – | – |
CIITA | CIITA, P33076 | – | – |
NLRP1 | NLRP1, Q9C000 | muramyl dipeptide | – |
NLRP2 | NLRP2, Q9NX02 | – | – |
NLRP3 | NLRP3, Q96P20 | – | Multiple virus particles have been shown to act as agonists, including Sendai and influenza |
NLRP4 | NLRP4, Q96MN2 | – | – |
NLRP5 | NLRP5, P59047 | – | – |
NLRP6 | NLRP6, P59044 | – | – |
NLRP7 | NLRP7, Q8WX94 | – | – |
NLRP8 | NLRP8, Q86W28 | – | – |
NLRP9 | NLRP9, Q7RTR0 | – | – |
NLRP10 | NLRP10, Q86W26 | – | – |
NLRP11 | NLRP11, P59045 | – | – |
NLRP12 | NLRP12, P59046 | – | – |
NLRP13 | NLRP13, Q86W25 | – | – |
NLRP14 | NLRP14, Q86W24 | – | – |
IPAF | NLRC4, Q9NPP4 | – | – |
NAIP | NAIP, Q13075 | – | – |
Comments
NLRP3 has also been reported to respond to host-derived products, known as danger-associated molecular patterns, or DAMPs, including uric acid 37, ATP, L-glucose, hyaluronan and amyloid β (APP, P05067) 40.
Loss-of-function mutations of NLRP3 are associated with cold autoinflammatory and Muckle-Wells syndromes.
Receptor serine/threonine kinase (RSTK) family
Overview
Receptor serine/threonine kinases (RTSK), EC 2.7.11.30, respond to particular cytokines, the transforming growth factor β (TGFβ) and bone morphogenetic protein (BMP) families, and may be divided into two subfamilies on the basis of structural similarities. Agonist binding initiates formation of a cell-surface complex of type I and type II RSTK, possibly heterotetrameric, where where both subunits express serine/threonine kinase activity. The type I receptor serine/threonine kinases (ENSFM00250000000213) are also known as activin receptors or activin receptor-like kinases, ALKs, for which a systematic nomenclature has been proposed (ALK1-7). The type II protein phosphorylates the kinase domain of the type I partner (sometimes referred to as the signal propagating subunit), causing displacement of the protein partners, such as the FKBP12 FK506-binding protein FKBP1A (P62942) and allowing the binding and phosphorylation of particular members of the Smad family. These migrate to the nucleus and act as complexes to regulate gene transcription. Type III receptors, sometimes called co-receptors or accessory proteins, regulate the signalling of the receptor complex, in either enhancing (for example, presenting the ligand to the receptor) or inhibitory manners. TGFβ family ligand signalling may be inhibited by endogenous proteins, such as follistatin (FST, P19883), which binds and neutralizes activins to prevent activation of the target receptors.
Endogenous agonists, approximately 30 in man, are often described as paracrine messengers acting close to the source of production. They are characterized by six conserved cysteine residues and are divided into two subfamilies on the basis of sequence comparison and signalling pathways activated, the TGFβ/activin/nodal subfamily and the BMP/GDF (growth/differentiation factor)/MIS (Müllerian inhibiting substance) subfamily. Ligands active at RSTKs appear to be generated as large precursors which undergo complex maturation processes 47. Some are known to form disulphide-linked homo- and/or heterodimeric complexes. Thus, inhibins are α subunits linked to a variety of β chains, while activins are combinations of β subunits.
Type I receptor serine/threonine kinases
Overview
The type I receptor serine/threonine kinases (ENSFM00250000000213) are also known as activin receptors or activin receptor-like kinases, ALKs, for which a systematic nomenclature has been proposed (ALK1-7).
Nomenclature | activin A receptor type II-like 1 | activin A receptor, type I | bone morphogenetic protein receptor, type IA | activin A receptor, type IB | transforming growth factor, beta receptor 1 | bone morphogenetic protein receptor, type IB | activin A receptor, type IC |
Common abbreviation | ALK1 | ALK2 | BMPR1A | ALK4 | TGFBR1 | BMPR1B | ALK7 |
HGNC, UniProt | ACVRL1, P37023 | ACVR1, Q04771 | BMPR1A, P36894 | ACVR1B, P36896 | TGFBR1, P36897 | BMPR1B, O00238 | ACVR1C, Q8NER5 |
Type II receptor serine/threonine kinases
Nomenclature | activin A receptor, type IIA | activin A receptor, type IIB | anti-Mullerian hormone receptor, type II | bone morphogenetic protein receptor, type II (serine/threonine kinase) | transforming growth factor, beta receptor II (70/80kDa) |
Common abbreviation | ActR2 | ActR2B | MISR2 | BMPR2 | TGFBR2 |
HGNC, UniProt | ACVR2A, P27037 | ACVR2B, Q13705 | AMHR2, Q16671 | BMPR2, Q13873 | TGFBR2, P37173 |
Type III receptor serine/threonine kinases
Nomenclature | transforming growth factor, beta receptor III |
Common abbreviation | TGFBR3 |
HGNC, UniProt | TGFBR3, Q03167 |
RSTK functional heteromers
Nomenclature | Transforming growth factor β receptor | Bone morphogenetic protein receptors | Growth/differentiation factor receptors | Activin receptors | Anti-Müllerian hormone receptors |
Subunits | transforming growth factor, beta receptor 1 (Type I), transforming growth factor, beta receptor II (70/80kDa) (Type II), transforming growth factor, beta receptor III (Type III) | activin A receptor type II-like 1 (Type I), activin A receptor, type I (Type I), bone morphogenetic protein receptor, type IA (Type I), bone morphogenetic protein receptor, type IB (Type I), activin A receptor, type IIA (Type II), activin A receptor, type IIB (Type II), bone morphogenetic protein receptor, type II (serine/threonine kinase) (Type II) | bone morphogenetic protein receptor, type IA (Type I), activin A receptor, type IB (Type I), transforming growth factor, beta receptor 1 (Type I), bone morphogenetic protein receptor, type IB (Type I), activin A receptor, type IC (Type I), activin A receptor, type IIA (Type II), activin A receptor, type IIB (Type II), bone morphogenetic protein receptor, type II (serine/threonine kinase) (Type II) | activin A receptor, type IB (Type I), activin A receptor, type IC (Type I), activin A receptor, type IIA (Type II), activin A receptor, type IIB (Type II) | activin A receptor, type I (Type I), bone morphogenetic protein receptor, type IA (Type I), bone morphogenetic protein receptor, type IB (Type I), anti-Mullerian hormone receptor, type II (Type II) |
Coupling | Smad2, Smad3 48,49 | Smad1, Smad5, Smad8 48,49 | Smad1, Smad5, Smad8 48,49 | Smad2, Smad3 49 | Smad1, Smad5, Smad8 48,49 |
Endogenous agonists | TGFβ1 (TGFB1, P01137), TGFβ2 (TGFB2, P61812), TGFβ3 (TGFB3, P10600) | BMP-10 (BMP10, O95393), BMP-2 (BMP2, P12643), BMP-4 (BMP4, P12644), BMP-5 (BMP5, P22003), BMP-6 (BMP6, P22004), BMP-7 (BMP7, P18075), BMP-8A (BMP8A, Q7Z5Y6), BMP-8B (BMP8B, P34820), BMP-9 (GDF2, Q9UK05) | GDF1 (GDF1, P27539), GDF10 (GDF10, P55107), GDF9 (GDF9, O60383), GDF3 (GDF3, Q9NR23) | inhibin βA (INHBA, P08476), inhibin βB (INHBB, P09529) | Müllerian inhibiting substance (AMH, P03971) |
Comments
A number of endogenous inhibitory ligands have been identified for RSTKs, including BMP3, inhibinα, inhibinβC and inhibinβE.
An appraisal of small molecule inhibitors of TGFβ and BMP signalling concluded that TGFβ pathway inhibitors were more selective than BMP signalling inhibitors 50. The authors confirmed the selectivity of SB505124 to inhibit TGFβ signalling through ALK4, ALK5, ALK7 46. dorsomorphin inhibits BMP signalling through ALK2 and ALK3, it also inhibits AMP kinase 51.
Smads were identified as mammalian orthologues of Drosophila genes termed “mothers against decapentaplegic” and may be divided into Receptor-regulated Smads (R-Smads, including Smad1, Smad2, Smad3, Smad5 and Smad8), Co-mediated Smad (Co-Smad, Smad4) and Inhibitory Smads (I-Smad, Smad6 and Smad7). R-Smads form heteromeric complexes with Co-Smad. I-Smads compete for binding of R-Smad with both receptors and Co-Smad.
Nomenclature | HGNC, UniProt | Other names |
Smad1 | SMAD1, Q15797 | JV4-1, MADH1, MADR1 |
Smad2 | SMAD2, Q15796 | JV18-1, MADH2, MADR2 |
Smad3 | SMAD3, P84022 | HsT17436, JV15-2, MADH3 |
Smad4 | SMAD4, Q13485 | DPC4, MADH4 |
Smad5 | SMAD5, Q99717 | Dwfc, JV5-1, MADH5 |
Smad6 | SMAD6, O43541 | HsT17432, MADH6, MADH7 |
Smad7 | SMAD7, O15105 | MADH7, MADH8 |
Smad8 | SMAD9, O15198 | MADH6, MADH9 |
Receptor tyrosine kinases
Overview
Receptor tyrosine kinases (RTKs, EC 2.7.10.1), a family of cell-surface receptors, which transduce signals to polypeptide and protein hormones, cytokines and growth factors are key regulators of critical cellular processes, such as proliferation and differentiation, cell survival and metabolism, cell migration and cell cycle control 55,65,82. In the human genome, 58 RTKs have been identified, which fall into 20 families 70.
All RTKs display an extracellular ligand binding domain, a single transmembrane helix, a cytoplasmic region containing the protein tyrosine kinase activity (occasionally split into two domains by an insertion, termed the kinase insertion), with juxta-membrane and C-terminal regulatory regions. Agonist binding to the extracellular domain evokes dimerization, and sometimes oligomerization, of RTKs (a small subset of RTKs forms multimers even in the absence of activating ligand). This leads to autophosphorylation in the tyrosine kinase domain in a trans orientation, serving as a site of assembly of protein complexes and stimulation of multiple signal transduction pathways, including phospholipase C-γ, mitogen-activated protein kinases and phosphatidylinositol 3-kinase 82.
RTKs are of widespread interest not only through physiological functions, but also as drug targets in many types of cancer and other disease states. Many diseases result from genetic changes or abnormalities that either alter the activity, abundance, cellular distribution and/or regulation of RTKs. Therefore, drugs that modify the dysregulated functions of these RTKs have been developed which fall into two categories. One group is often described as ‘biologicals', which block the activation of RTKs directly or by chelating the cognate ligands, while the second are small molecules designed to inhibit the tyrosine kinase activity directly.
Type I RTKs: ErbB (epidermal growth factor) receptor family
Overview
ErbB family receptors are Class I receptor tyrosine kinases 65. ERBB2 (also known as HER-2 or NEU; ERBB2, P04626) appears to act as an essential partner for the other members of the family without itself being activated by a cognate ligand 66. Ligands of the ErbB family of receptors are peptides, many of which are generated by proteolytic cleavage of cell-surface proteins. HER/ErbB is the viral counterpart to the receptor tyrosine kinase EGFR. All family members heterodimerize with each other to activate downstream signalling pathways and are aberrantly expressed in many cancers, particularly forms of breast cancer.
Nomenclature | Common abbreviation | HGNC, UniProt | Endogenous ligands |
epidermal growth factor receptor | EGFR | EGFR, P00533 | amphiregulin (AREG, AREGB, P15514), betacellulin (BTC, P35070), EGF (EGF, P01133), epigen (EPGN, Q6UW88), epiregulin (EREG, O14944), HB-EGF (HBEGF, Q99075), TGFα (TGFA, P01135) |
v-erb-b2 avian erythroblastic leukemia viral oncogene homolog 3 | HER3 | ERBB3, P21860 | NRG-1 (NRG1, Q02297), NRG-2 (NRG2, O14511) |
v-erb-b2 avian erythroblastic leukemia viral oncogene homolog 4 | HER4 | ERBB4, Q15303 | betacellulin (BTC, P35070), epiregulin (EREG, O14944), HB-EGF (HBEGF, Q99075), NRG-1 (NRG1, Q02297), NRG-2 (NRG2, O14511), NRG-3 (NRG3, P56975), NRG-4 (NRG4, Q8WWG1) |
Comments
[125I]EGF (human) has been used to label the ErbB1 EGF receptor. The extracellular domain of ErbB2 can be targetted by the antibodies trastuzumab and pertuzumab to inhibit ErbB family action. The intracellular ATP-binding site of the tyrosine kinase domain can be inhibited by GW583340 (7.9–8.0, 63), gefitinib, erlotinib and tyrphostins AG879 and AG1478.
Type II RTKs: Insulin receptor family
Overview
The circulating peptide hormones insulin (INS, P01308) and the related insulin-like growth factors (IGF) activate Class II receptor tyrosine kinases 65, to evoke cellular responses, mediated through multiple intracellular adaptor proteins. Exceptionally amongst the catalytic receptors, the functional receptor in the insulin receptor family is derived from a single gene product, cleaved post-translationally into two peptides, which then cross-link via disulphide bridges to form a heterotetramer. Intriguingly, the endogenous peptide ligands are formed in a parallel fashion with post-translational processing producing a heterodimer linked by disulphide bridges. Signalling through the receptors is mediated through a rapid autophosphorylation event at intracellular tyrosine residues, followed by recruitment of multiple adaptor proteins, notably IRS1 (P35568), IRS2 (Q9Y4H2), SHC1 (P29353), GRB2 (P62993) and SOS1 (Q07889).
Serum levels of free IGFs are kept low by the action of IGF binding proteins (IGFBP1-5, P08833, P18065, P17936, P22692, P24593), which sequester the IGFs; overexpression of IGFBPs may induce apoptosis, while IGFBP levels are also altered in some cancers.
Comments
There is evidence for low potency binding and activation of insulin receptors by IGF1. IGF2 also binds and activates the cation-independent mannose 6-phosphate receptor (also known as the insulin-like growth factor II receptor), which lacks classical signalling capacity and appears to subserve a trafficking role 72. INSRR, which has a much more discrete localization, being predominant in the kidney 69, currently lacks a cognate ligand or evidence for functional impact.
Antibodies targetting IGF1, IGF2 and the extracellular portion of the IGF1 receptor are in clinical trials.
PQ401 inhibits the insulin-like growth factor receptor 56, while BMS-536924 inhibits both the insulin receptor and the insulin-like growth factor receptor 85.
Type III RTKs: PDGFR, CSFR, Kit, FLT3 receptor family
Overview
Type III RTKs include PDGFR, CSF-1R (Ems), Kit and FLT3, which function as homo- or heterodimers. Endogenous ligands of PDGF receptors are homo- or heterodimeric: PDGFA, PDGFB, VEGFE and PDGFD combine as homo- or heterodimers to activate homo- or heterodimeric PDGF receptors. SCF is a dimeric ligand for KIT. Ligands for CSF1R are either monomeric or dimeric glycoproteins, while the endogenous agonist for FLT3 is a homodimer.
Nomenclature | platelet-derived growth factor receptor, alpha polypeptide | platelet-derived growth factor receptor, beta polypeptide | v-kit Hardy-Zuckerman 4 feline sarcoma viral oncogene homolog | colony stimulating factor 1 receptor | fms-related tyrosine kinase 3 |
Common abbreviation | PDGFRα | PDGFRβ | Kit | CSFR | FLT3 |
HGNC, UniProt | PDGFRA, P16234 | PDGFRB, P09619 | KIT, P10721 | CSF1R, P07333 | FLT3, P36888 |
Endogenous ligands | PDGF | PDGF | SCF (KITLG, P21583) | G-CSF (CSF3, P09919), GM-CSF (CSF2, P04141), M-CSF (CSF1, P09603) | FLT3L (FLT3LG, P49771) |
Comment | – | – | – | – | 5'-fluoroindirubinoxime has been described as a selective FLT3 inhibitor 57 |
Comments
Various small molecular inhibitors of type III RTKs have been described, including imatinib and nilotinib (targetting PDGFR, KIT and CSF1R); midostaurin and AC220 (quizartinib; FLT3), as well as pan-type III RTK inhibitors such as sunitinib and sorafenib 78; 5'-fluoroindirubinoxime has been described as a selective FLT3 inhibitor 53.
Type IV RTKs: VEGF (vascular endothelial growth factor) receptor family
Overview
VEGF receptors are homo- and heterodimeric proteins, which are characterized by seven Ig-like loops in their extracellular domains and a split kinase domain in the cytoplasmic region. They are key regulators of angiogenesis and lymphangiogenesis; as such, they have been the focus of drug discovery for conditions such as metastatic cancer. Splice variants of VEGFR1 and VEGFR2 generate truncated proteins limited to the extracellular domains, capable of homodimerisation and binding VEGF ligands as a soluble, non-signalling entity. Ligands at VEGF receptors are typically homodimeric. VEGFA (VEGFA, P15692) is able to activate VEGFR1 homodimers, VEGFR1/2 heterodimers and VEGFR2/3 heterodimers. VEGFB (VEGFB, P49765) and placental growth factor activate VEGFR1 homodimers, while VEGFC (VEGFC, P49767) and VEGFD (FIGF, O43915) activate VEGFR2/3 heterodimers and VEGFR3 homodimers, and, following proteolysis, VEGFR2 homodimers.
Nomenclature | fms-related tyrosine kinase 1 | kinase insert domain receptor (a type III receptor tyrosine kinase) | fms-related tyrosine kinase 4 |
Common abbreviation | VEGFR-1 | VEGFR-2 | VEGFR-3 |
HGNC, UniProt | FLT1, P17948 | KDR, P35968 | FLT4, P35916 |
Endogenous ligands | VEGFA (VEGFA, P15692), VEGFB (VEGFB, P49765) | VEGFA (VEGFA, P15692), VEGFC (VEGFC, P49767), VEGFE (PDGFC, Q9NRA1) | VEGFC (VEGFC, P49767), VEGFD (FIGF, O43915), VEGFE (PDGFC, Q9NRA1) |
Comments
The VEGFR, as well as VEGF ligands, have been targeted by antibodies and tyrosine kinase inhibitors. DMH4 62, Ki8751 68 and ZM323881, a novel inhibitor of vascular endothelial growth factor-receptor-2 tyrosine kinase activity 84 are described as VEGFR2-selective tyrosine kinase inhibitors. Bevacizumab is a monoclonal antibody directed against VEGF-A, used clinically for the treatment of certain metastatic cancers; an antibody fragment has been used for wet age-related macular degeneration.
Type V RTKs: FGF (fibroblast growth factor) receptor family
Overview
Fibroblast growth factor (FGF) family receptors act as homo- and heterodimers, and are characterized by Ig-like loops in the extracellular domain, in which disulphide bridges may form across protein partners to allow the formation of covalent dimers which may be constitutively active. FGF receptors have been implicated in achondroplasia, angiogenesis and numerous congenital disorders. At least 22 members of the FGF gene family have been identified in the human genome 61. Within this group, subfamilies of FGF may be divided into canonical, intracellular and hormone-like FGFs. FGF1-FGF10 have been identified to act through FGF receptors, while FGF11-14 appear to signal through intracellular targets. Other family members are less well characterized 83.
Nomenclature | fibroblast growth factor receptor 1 | fibroblast growth factor receptor 2 | fibroblast growth factor receptor 3 | fibroblast growth factor receptor 4 |
Common abbreviation | FGFR1 | FGFR2 | FGFR3 | FGFR4 |
HGNC, UniProt | FGFR1, P11362 | FGFR2, P21802 | FGFR3, P22607 | FGFR4, P22455 |
Endogenous ligands | FGF-1 (FGF1, P05230), FGF-2 (FGF2, P09038), FGF-4 (FGF4, P08620) > FGF-5 (FGF5, P12034), FGF-6 (FGF6, P10767) 77 | FGF-1 (FGF1, P05230) > FGF-4 (FGF4, P08620), FGF-7 (FGF7, P21781), FGF-9 (FGF9, P31371) > FGF-2 (FGF2, P09038), FGF-6 (FGF6, P10767) 77 | FGF-1 (FGF1, P05230), FGF-2 (FGF2, P09038), FGF-9 (FGF9, P31371) > FGF-4 (FGF4, P08620), FGF-8 (FGF8, P55075) 77 | FGF-1 (FGF1, P05230), FGF-2 (FGF2, P09038), FGF-4 (FGF4, P08620), FGF-9 (FGF9, P31371) > FGF-6 (FGF6, P10767), FGF-8 (FGF8, P55075) 77 |
Comments
Splice variation of the receptors can influence agonist responses. FGFRL1 (Q8N441) is a truncated kinase-null analogue.
Various antibodies and tyrosine kinase inhibitors have been developed against FGF receptors 71,87. PD161570 is an FGFR tyrosine kinase inhibitor 54, while PD173074 has been described to inhibit FGFR1 and FGFR3 80.
Type VII RTKs: Neurotrophin receptor/Trk family
Overview
The neurotrophin receptor family of RTKs include trkA, trkB and trkC (tropomyosin-related kinase) receptors, which respond to NGF, BDNF and neurotrophin-3, respectively. They are associated primarily with proliferative and migration effects in neural systems. Various isoforms of neurotrophin receptors exist, including truncated forms of trkB and trkC, which lack catalytic domains. p75(TNFRSF16, also known as nerve growth factor receptor), which has homologies with tumour necrosis factor receptors, lacks a tyrosine kinase domain, but can signal via ceramide release and nuclear factor κB (NF-κB) activation. Both trkA and trkB contain two leucine-rich regions and can exist in monomeric or dimeric forms.
Nomenclature | neurotrophic tyrosine kinase, receptor, type 1 | neurotrophic tyrosine kinase, receptor, type 2 | neurotrophic tyrosine kinase, receptor, type 3 |
Common abbreviation | trkA | trkB | trkC |
HGNC, UniProt | NTRK1, P04629 | NTRK2, Q16620 | NTRK3, Q16288 |
Endogenous ligands | NGF (NGF, P01138) > NT-3 (NTF3, P20783) | BDNF (BDNF, P23560), NT-4 (NTF4, P34130) > NT-3 (NTF3, P20783) | NT-3 (NTF3, P20783) |
Comments
[125I]NGF (human) and [125I]BDNF have been used to label the trkA and trkB receptor, respectively. p75 influences the binding of NGF (NGF, P01138) and NT-3 (NTF3, P20783) to trkA. The ligand selectivity of p75 appears to be dependent on the cell type; for example, in sympathetic neurones, it binds NT-3 (NTF3, P20783) with comparable affinity to trkC 60.
Small molecule agonists of trkB have been described, including LM22A4 73, while ANA12 has been described as a non- competitive antagonist of BDNF binding to trkB 56. GNF5837 is a family-selective tyrosine kinase inhibitor 52, while the tyrosine kinase activity of the trkA receptor can be inhibited by GW441756 (pIC50= 8.7, 86) and tyrphostin AG879 76.
Type VIII RTKs: ROR family
Overview
Members of the ROR family (ENSFM00510000502747) appear to be activated by ligands complexing with other cell-surface proteins. Thus, ROR1 and ROR2 appear to be activated by Wnt-5a (WNT5A, P41221) binding to a Frizzled receptor thereby forming a cell-surface multiprotein complex 67.
Type X RTKs: HGF (hepatocyte growth factor) receptor family
Overview
HGF receptors regulate maturation of the liver in the embryo, as well as having roles in the adult, for example, in the innate immune system. HGF is synthesized as a single gene product, which is post-translationally processed to yield a heterodimer linked by a disulphide bridge. The maturation of HGF is enhanced by a serine protease, HGF activating complex, and inhibited by HGF-inhibitor 1, a serine protease inhibitor. MST1, the ligand of RON, is two disulphide-linked peptide chains generated by proteolysis of a single gene product.
Comments
PF04217903 is a selective Met tyrosine kinase inhibitor 58. SU11274 is an inhibitor of the HGF receptor 79, with the possibility of further targets 53.
Type XI RTKs: TAM (TYRO3-, AXL- and MER-TK) receptor family
Overview
Members of this RTK family (ENSFM00500000269872) represented a novel structural motif, when sequenced. The ligands for this family, Gas6 (GAS6, Q14393) and protein S (PROS1, P07225), are secreted plasma proteins which undergo vitamin K-dependent post-translational modifications generating carboxyglutamate-rich domains which are able to bind to negatively-charged surfaces of apoptotic cells.
Nomenclature | AXL receptor tyrosine kinase | TYRO3 protein tyrosine kinase | c-mer proto-oncogene tyrosine kinase |
Common abbreviation | Axl | Tyro3 | Mer |
HGNC, UniProt | AXL, P30530 | TYRO3, Q06418 | MERTK, Q12866 |
Endogenous ligands | Gas6 (GAS6, Q14393) 75, protein S (PROS1, P07225) 81 | Gas6 (GAS6, Q14393) 75, protein S (PROS1, P07225) 81 | Gas6 (GAS6, Q14393) 75 |
Comments
AXL tyrosine kinase inhibitors have been described 74.
Type XII RTKs: TIE family of angiopoietin receptors
Overview
The TIE family were initially associated with formation of blood vessels. Endogenous ligands are angiopoietin-1 (ANGPT1, Q15389), angiopoietin-2 (ANGPT2, O15123), and angiopoietin-4 (ANGPT4, Q9Y264). angiopoietin-2 (ANGPT2, O15123) appears to act as an endogenous antagonist of angiopoietin-1 function.
Type XIII RTKs: Ephrin receptor family
Overview
Ephrin receptors (ENSFM00250000000121) are a family of 15 RTKs (the largest family of RTKs) with two identified subfamilies (EphA and EphB), which have a role in the regulation of neuronal development, cell migration, patterning and angiogenesis. Their ligands are membrane-associated proteins, thought to be glycosylphosphatidylinositol-linked for EphA (EFNA1 (EFNA1, P20827), EFNA2 (EFNA2, O43921), EFNA3 (EFNA3, P52797), EFNA4 (EFNA4, P52798) and EFNA5 (EFNA5, P52803)) and 1TM proteins for Ephrin B (ENSFM00250000002014: EFNB1 (EFNB1, P98172), EFNB2 (EFNB2, P52799) and EFNB3 (EFNB3, Q15768)), although the relationship between ligands and receptors has been incompletely defined.
Nomenclature | EPH receptor A1 | EPH receptor A2 | EPH receptor A3 | EPH receptor A4 | EPH receptor A5 | EPH receptor A6 | EPH receptor A7 | EPH receptor A8 | EPH receptor A10 | EPH receptor B1 | EPH receptor B2 | EPH receptor B3 | EPH receptor B4 | EPH receptor B6 |
Common abbreviation | EphA1 | EphA2 | EphA3 | EphA4 | EphA5 | EphA6 | EphA7 | EphA8 | EphA10 | EphB1 | EphB2 | EphB3 | EphB4 | EphB6 |
HGNC, UniProt | EPHA1, P21709 | EPHA2, P29317 | EPHA3, P29320 | EPHA4, P54764 | EPHA5, P54756 | EPHA6, Q9UF33 | EPHA7, Q15375 | EPHA8, P29322 | EPHA10, Q5JZY3 | EPHB1, P54762 | EPHB2, P29323 | EPHB3, P54753 | EPHB4, P54760 | EPHB6, O15197 |
Type XVI RTKs: DDR (collagen receptor) family
Overview
Discoidin domain receptors 1 and 2 (DDR1 and DDR2) are structurally-related membrane protein tyrosine kinases activated by collagen. Collagen is probably the most abundant protein in man, with at least 29 families of genes encoding proteins, which undergo splice variation and post-translational processing, and may exist in monomeric or polymeric forms, producing a triple-stranded, twine-like structure. In man, principal family members include COL1A1 (COL1A1, P02452), COL2A1 (COL2A1, P02458), COL3A1 (COL3A1, P02461) and COL4A1 (COL4A1, P02462).
Comments
The tyrosine kinase inhibitors of DDR, imatinib and nilotinib, were identified from proteomic analysis 59.
Type XIX RTKs: Leukocyte tyrosine kinase (LTK) receptor family
Overview
The LTK family (ENSFM00500000270379) appear to lack endogenous ligands. LTK is subject to tissue-specific splice variation, which appears to generate products in distinct subcellular locations. Alk fusions derived from gene translocations are associated with large cell lymphomas and inflammatory myofibrilastic tumours.
Receptor tyrosine phosphatases (RTP)
Overview
Receptor tyrosine phosphatases (RTP) are cell-surface proteins with a single TM region and intracellular phosphotyrosine phosphatase activity. Many family members exhibit constitutive activity in heterologous expression, dephosphorylating intracellular targets such as Src tyrosine kinase (SRC) to activate signalling cascades. Family members bind components of the extracellular matrix or cell-surface proteins indicating a role in intercellular communication.
Nomenclature | HGNC, UniProt | Putative endogenous ligands |
RTP Type A | PTPRA, P18433 | – |
RTP Type B | PTPRB, P23467 | – |
RTP Type C | PTPRC, P08575 | galectin-1 (LGALS1, P09382) 93 |
RTP Type D | PTPRD, P23468 | netrin-G3 ligand (LRRC4B, Q9NT99) 90 |
RTP Type E | PTPRE, P23469 | – |
RTP Type F | PTPRF, P10586 | netrin-G3 ligand (LRRC4B, Q9NT99) 90 |
RTP Type G | PTPRG, P23470 | contactin-3 (CNTN3, Q9P232), contactin-4 (CNTN4, Q8IWV2), contactin-5 (CNTN5, O94779), contactin-6 (CNTN6, Q9UQ52) 88 |
RTP Type H | PTPRH, Q9HD43 | – |
RTP Type J | PTPRJ, Q12913 | – |
RTP Type K | PTPRK, Q15262 | galectin-3 (LGALS3, P17931), galectin-3 binding protein (LGALS3BP, Q08380) 89 |
RTP Type M | PTPRM, P28827 | – |
RTP Type N | PTPRN, Q16849 | – |
RTP Type N2 | PTPRN2, Q92932 | – |
RTP Type O | PTPRO, Q16827 | – |
RTP Type Q | PTPRQ, Q9UMZ3 | – |
RTP Type R | PTPRR, Q15256 | – |
RTP Type S | PTPRS, Q13332 | chondroitin sulphate proteoglycan 3 (NCAN, O14594), netrin-G3 ligand (LRRC4B, Q9NT99) 90,92 |
RTP Type T | PTPRT, O14522 | – |
RTP Type U | PTPRU, Q92729 | – |
RTP Type Z1 | PTPRZ1, P23471 | contactin-1 (CNTN1, Q12860), pleiotrophin (PTN, C9JR52) (acts as a negative regulator) 88,91 |
Tumour necrosis factor (TNF) receptor family
Overview
The TNF receptor superfamily (TNFRSF, provisional nomenclature) displays limited homology beyond an extracellular domain rich in cysteine residues and is activated by at least 18 different human homologues of TNF referred to as the TNF superfamily (TNFSF). Some homologues lacking transmembrane and cytoplasmic domains function as decoy receptors binding ligand without inducing cell signalling. Many of these receptors and ligands function as multimeric entities. Signalling through these receptors is complex and involves interaction with cytoplasmic adaptor proteins (such as TRADD and TRAF1). Several of these receptors contain cytoplasmic motifs known as ‘death domains’, which upon activation serve to recruit death domain- and death effector domain-containing proteins crucial for the initiation of an apoptotic response. Additional signalling pathways include the regulation of the nuclear factor κB or mitogen-activated protein kinase pathways. Pharmacological manipulation of these receptors is mainly enacted through chelating the endogenous agonists with humanised monoclonal antibodies (e.g. infliximab or adalimumab) or recombinant fusion proteins of IgG and soluble receptors (e.g. etanercept). Some mutated forms of TNF ligands are capable of selecting for different receptor subtypes.
Receptors
Nomenclature | Systematic nomenclature | Common abbreviation | HGNC, UniProt | Adaptor proteins | Endogenous ligands | Comment |
tumor necrosis factor receptor 1 | TNFRSF1A | TNFR1 | TNFRSF1A, P19438 | TRADD | TNFSF1 (LTA, P01374), TNF membrane form (TNF, P01375), TNF shed form (TNF, P01375) | – |
tumor necrosis factor receptor 2 | TNFRSF1B | TNFR2 | TNFRSF1B, P20333 | TRAF1, TRAF2, TRAF5 | TNFSF1 (LTA, P01374), TNF membrane form (TNF, P01375) | – |
lymphotoxin β receptor | TNFRSF3 | – | LTBR, P36941 | TRAF3, TRAF4, TRAF5 | LIGHT (TNFSF14, O43557), lymphotoxin β2α1 heterotrimer (LTA, LTB, Q06643, P01374) | – |
OX40 | TNFRSF4 | – | TNFRSF4, P43489 | TRAF1, TRAF2, TRAF3, TRAF5 | OX-40 ligand (TNFSF4, P23510) | – |
CD40 | TNFRSF5 | – | CD40, P25942 | TRAF1, TRAF2, TRAF3, TRAF5, TRAF6 | CD40 ligand (CD40LG, P29965) | – |
Fas | TNFRSF6 | – | FAS, P25445 | FADD | Fas ligand (FASLG, P48023) | – |
CD27 | TNFRSF7 | – | CD27, P26842 | TRAF2, SIVA | CD70 (CD70, P32970) | – |
CD30 | TNFRSF8 | – | TNFRSF8, P28908 | TRAF1, TRAF2, TRAF3, TRAF5 | CD30 ligand (TNFSF8, P32971) | – |
4-1BB | TNFRSF9 | – | TNFRSF9, Q07011 | TRAF1, TRAF2, TRAF3 | 4-1BB ligand (TNFSF9, P41273) | – |
death receptor 4 | TNFRSF10A | DR4 | TNFRSF10A, O00220 | FADD | TRAIL (TNFSF10, P50591) | – |
death receptor 5 | TNFRSF10B | DR5 | TNFRSF10B, O14763 | FADD | TRAIL (TNFSF10, P50591) | – |
receptor activator of NF-kappa B | TNFRSF11A | RANK | TNFRSF11A, Q9Y6Q6 | TRAF1, TRAF2, TRAF3, TRAF5, TRAF6 | RANK ligand (TNFSF11, O14788) | – |
osteoprotegerin | TNFRSF11B | OPG | TNFRSF11B, O00300 | – | – | Acts as a decoy receptor for RANK ligand (TNFSF11, O14788) and possibly for TRAIL (TNFSF10, P50591) |
death receptor 3 | TNFRSF25 | DR3 | TNFRSF25, Q93038 | TRADD | TL1A (TNFSF15, O95150) | – |
TWEAK receptor | TNFRSF12A | – | TNFRSF12A, Q9NP84 | TRAF1, TRAF2, TRAF3 | TWEAK (TNFSF12, O43508) | – |
TACI | TNFRSF13B | – | TNFRSF13B, O14836 | TRAF2, TRAF5, TRAF6 | APRIL (TNFSF13, O75888), BAFF (TNFSF13B, Q9Y275) | – |
BAFF receptor | TNFRSF13C | BAFF-R | TNFRSF13C, Q96RJ3 | TRAF3 | BAFF (TNFSF13B, Q9Y275) | – |
herpes virus entry mediator | TNFRSF14 | HVEM | TNFRSF14, Q92956 | TRAF2, TRAF3, TRAF5 | BTLA (BTLA, Q7Z6A9), LIGHT (TNFSF14, O43557), TNFSF1 (LTA, P01374) | – |
nerve growth factor receptor | TNFRSF16 | – | NGFR, P08138 | TRAF2, TRAF4, TRAF6 | BDNF (BDNF, P23560), NT-3 (NTF3, P20783), NT-4 (NTF4, P34130), NGF (NGF, P01138) | – |
B cell maturation antigen | TNFRSF17 | BCMA | TNFRSF17, Q02223 | TRAF1, TRAF2, TRAF3, TRAF5, TRAF6 | APRIL (TNFSF13, O75888), BAFF (TNFSF13B, Q9Y275) | – |
glucocorticoid-induced TNF receptor | TNFRSF18 | GITR | TNFRSF18, Q9Y5U5 | TRAF1, TRAF2, TRAF3, SIVA | TL6 (TNFSF18, Q9UNG2) | – |
toxicity and JNK inducer | TNFRSF19 | TAJ | TNFRSF19, Q9NS68 | TRAF1, TRAF2, TRAF3, TRAF5 | TNFSF1 (LTA, P01374) | – |
RELT | TNFRSF19L | – | RELT, Q969Z4 | TRAF1 | – | – |
death receptor 6 | TNFRSF21 | DR6 | TNFRSF21, O75509 | TRADD | – | – |
ectodysplasin A2 isoform receptor | TNFRS27 | – | EDA2R, Q9HAV5 | TRAF1, TRAF3, TRAF6 | ectodysplasin A2 (EDA, Q92838) 94 | – |
Comments
TNFRSF1A is preferentially activated by the shed form of TNF ligand, whereas the membrane-bound form of TNF serves to activate TNFRSF1A and TNFRSF1B equally. The neurotrophins nerve growth factor (NGF (NGF, P01138), P01138), brain-derived neurotrophic factor (BDNF (BDNF, P23560), P23560), NT-3 (NTF3, P20783) (NTF3, P20783) and NT-4 (NTF4, P34130) (NTF4, P34130) are structurally unrelated to the TNF ligand superfamily but exert some of their actions through the “low affinity nerve growth factor receptor” (NGFR (TNFRSF16)) as well as through the TRK family of receptor tyrosine kinases. The endogenous ligands for EDAR and EDA2R are, respectively, the membrane (Q92838[1-391]) and secreted (Q92838[160-391]) isoforms of Ectodysplasin-A (EDA, Q92838).
Further reading
- Broughton SE. Dhagat U. Hercus TR. Nero TL. Grimbaldeston MA. Bonder CS. Lopez AF. Parker MW. The GM-CSF/IL-3/IL-5 cytokine receptor family: from ligand recognition to initiation of signaling. Immunol Rev. 2013;250:277–302. doi: 10.1111/j.1600-065X.2012.01164.x. [PMID:23046136] [DOI] [PubMed] [Google Scholar]
- Chang SH. Dong C. Signaling of interleukin-17 family cytokines in immunity and inflammation. Cell Signal. 2011;23:1069–1075. doi: 10.1016/j.cellsig.2010.11.022. [PMID:21130872] [DOI] [PMC free article] [PubMed] [Google Scholar]
- Donnelly RP. Dickensheets H. O'Brien TR. Interferon-lambda and therapy for chronic hepatitis C virus infection. Trends Immunol. 2011;32:443–450. doi: 10.1016/j.it.2011.07.002. [PMID:21820962] [DOI] [PMC free article] [PubMed] [Google Scholar]
- George PM. Badiger R. Alazawi W. Foster GR. Mitchell JA. Pharmacology and therapeutic potential of interferons. Pharmacol Ther. 2012;135:44–53. doi: 10.1016/j.pharmthera.2012.03.006. [PMID:22484806] [DOI] [PubMed] [Google Scholar]
- Gibbert K. Schlaak JF. Yang D. Dittmer U. IFN-α subtypes: distinct biological activities in anti-viral therapy. Br J Pharmacol. 2013;168:1048–1058. doi: 10.1111/bph.12010. [PMID:23072338] [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mackall CL. Fry TJ. Gress RE. Harnessing the biology of IL-7 for therapeutic application. Nat Rev Immunol. 2011;11:330–342. doi: 10.1038/nri2970. [PMID:21508983] [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mihara M. Hashizume M. Yoshida H. Suzuki M. Shiina M. IL-6/IL-6 receptor system and its role in physiological and pathological conditions. Clin Sci. 2012;122:143–159. doi: 10.1042/CS20110340. [PMID:22029668] [DOI] [PubMed] [Google Scholar]
- Miller AM. Liew FY. The IL-33/ST2 pathway–A new therapeutic target in cardiovascular disease. Pharmacol Ther. 2011;131:179–186. doi: 10.1016/j.pharmthera.2011.02.005. [PMID:21356240] [DOI] [PubMed] [Google Scholar]
- Miossec P. Kolls JK. Targeting IL-17 and TH17 cells in chronic inflammation. Nat Rev Drug Discov. 2012;11:763–776. doi: 10.1038/nrd3794. [PMID:23023676] [DOI] [PubMed] [Google Scholar]
- Murugaiyan G. Saha B. IL-27 in tumor immunity and immunotherapy. Trends Mol Med. 2013;19:108–116. doi: 10.1016/j.molmed.2012.12.002. [PMID:23306374] [DOI] [PubMed] [Google Scholar]
- Palmer G. Gabay C. Interleukin-33 biology with potential insights into human diseases. Nat Rev Rheumatol. 2011;7:321–329. doi: 10.1038/nrrheum.2011.53. [PMID:21519352] [DOI] [PubMed] [Google Scholar]
- Pappu R. Ramirez-Carrozzi V. Sambandam A. The interleukin-17 cytokine family: critical players in host defence and inflammatory diseases. Immunology. 2011;134:8–16. doi: 10.1111/j.1365-2567.2011.03465.x. [PMID:21726218] [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pappu R. Rutz S. Ouyang W. Regulation of epithelial immunity by IL-17 family cytokines. Trends Immunol. 2012;33:343–349. doi: 10.1016/j.it.2012.02.008. [PMID:22476048] [DOI] [PubMed] [Google Scholar]
- Parker D. Prince A. Type I interferon response to extracellular bacteria in the airway epithelium. Trends Immunol. 2011;32:582–588. doi: 10.1016/j.it.2011.09.003. [PMID:21996313] [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pestka S. Krause CD. Sarkar D. Walter MR. Shi Y. Fisher PB. Interleukin-10 and related cytokines and receptors. Annu Rev Immunol. 2004;22:929–979. doi: 10.1146/annurev.immunol.22.012703.104622. [PMID:15032600] [DOI] [PubMed] [Google Scholar]
- Rincon M. Interleukin-6: from an inflammatory marker to a target for inflammatory diseases. Trends Immunol. 2012;33:571–577. doi: 10.1016/j.it.2012.07.003. [PMID:22883707] [DOI] [PubMed] [Google Scholar]
- Rubino SJ. Geddes K. Girardin SE. Innate IL-17 and IL-22 responses to enteric bacterial pathogens. Trends Immunol. 2012;33:112–118. doi: 10.1016/j.it.2012.01.003. [PMID:22342740] [DOI] [PubMed] [Google Scholar]
- Sato N. Miyajima A. Multimeric cytokine receptors: common versus specific functions. Curr Opin Cell Biol. 1994;6:174–179. doi: 10.1016/0955-0674(94)90133-3. [PMID:8024807] [DOI] [PubMed] [Google Scholar]
- Schindler C. Levy DE. Decker T. JAK-STAT signaling: from interferons to cytokines. J Biol Chem. 2007;282:20059–20063. doi: 10.1074/jbc.R700016200. [PMID:17502367] [DOI] [PubMed] [Google Scholar]
- Shevach EM. Application of IL-2 therapy to target T regulatory cell function. Trends Immunol. 2012;33:626–632. doi: 10.1016/j.it.2012.07.007. [PMID:22951308] [DOI] [PMC free article] [PubMed] [Google Scholar]
- Steel JC. Waldmann TA. Morris JC. Interleukin-15 biology and its therapeutic implications in cancer. Trends Pharmacol Sci. 2012;33:35–41. doi: 10.1016/j.tips.2011.09.004. [PMID:22032984] [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tanaka T. Narazaki M. Kishimoto T. Therapeutic targeting of the interleukin-6 receptor. Annu Rev Pharmacol Toxicol. 2012;52:199–219. doi: 10.1146/annurev-pharmtox-010611-134715. [PMID:21910626] [DOI] [PubMed] [Google Scholar]
- Lely AJ. Kopchick JJ. Growth hormone receptor antagonists. Neuroendocrinology. 2006;83:264–268. doi: 10.1159/000095537. [PMID:17047392] [DOI] [PubMed] [Google Scholar]
- Wojno ED. Hunter CA. New directions in the basic and translational biology of interleukin-27. Trends Immunol. 2012;33:91–97. doi: 10.1016/j.it.2011.11.003. [PMID:22177689] [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zepp J. Wu L. Li X. IL-17 receptor signaling and T helper 17-mediated autoimmune demyelinating disease. Trends Immunol. 2011;32:232–239. doi: 10.1016/j.it.2011.02.007. [PMID:21493143] [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhu S. Qian Y. IL-17/IL-17 receptor system in autoimmune disease: mechanisms and therapeutic potential. Clin Sci. 2012;122:487–511. doi: 10.1042/CS20110496. [PMID:22324470] [DOI] [PubMed] [Google Scholar]
Further reading
- Allen SJ. Watson JJ. Shoemark DK. Barua NU. Patel NK. GDNF, NGF and BDNF as therapeutic options for neurodegeneration. Pharmacol Ther. 2013;138:155–175. doi: 10.1016/j.pharmthera.2013.01.004. [PMID:23348013] [DOI] [PubMed] [Google Scholar]
- Carnicella S. Ron D. GDNF–a potential target to treat addiction. Pharmacol Ther. 2009;122:9–18. doi: 10.1016/j.pharmthera.2008.12.001. [PMID:19136027] [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liu H. Li X. Xu Q. Lv S. Li J. Ma Q. Role of glial cell line-derived neurotrophic factor in perineural invasion of pancreatic cancer. Biochim Biophys Acta. 2012;1826:112–120. doi: 10.1016/j.bbcan.2012.03.010. [PMID:22503821] [DOI] [PubMed] [Google Scholar]
- Mickiewicz AL. Kordower JH. GDNF family ligands: a potential future for Parkinson's disease therapy. CNS Neurol Disord Drug Targets. 2011;10:703–711. doi: 10.2174/187152711797247876. [PMID:21838676] [DOI] [PubMed] [Google Scholar]
- Pascual A. Hidalgo-Figueroa M. Gómez-Díaz R. López-Barneo J. GDNF and protection of adult central catecholaminergic neurons. J Mol Endocrinol. 2011;46:R83–R92. doi: 10.1530/JME-10-0125. [PMID:21357726] [DOI] [PubMed] [Google Scholar]
- Rangasamy SB. Soderstrom K. Bakay RA. Kordower JH. Neurotrophic factor therapy for Parkinson's disease. Prog Brain Res. 2010;184:237–264. doi: 10.1016/S0079-6123(10)84013-0. [PMID:20887879] [DOI] [PubMed] [Google Scholar]
Further reading
- Anthis NJ. Campbell ID. The tail of integrin activation. Trends Biochem Sci. 2011;36:191–198. doi: 10.1016/j.tibs.2010.11.002. [PMID:21216149] [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bledzka K. Smyth SS. Plow EF. Integrin αIIbβ3: from discovery to efficacious therapeutic target. Circ Res. 2013;112:1189–1200. doi: 10.1161/CIRCRESAHA.112.300570. [PMID:23580774] [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cavallaro U. Dejana E. Adhesion molecule signalling: not always a sticky business. Nat Rev Mol Cell Biol. 2011;12:189–197. doi: 10.1038/nrm3068. [PMID:21346732] [DOI] [PubMed] [Google Scholar]
- Cox D. Brennan M. Moran N. Integrins as therapeutic targets: lessons and opportunities. Nat Rev Drug Discov. 2010;9:804–820. doi: 10.1038/nrd3266. [PMID:20885411] [DOI] [PubMed] [Google Scholar]
- Hogg N. Patzak I. Willenbrock F. The insider's guide to leukocyte integrin signalling and function. Nat Rev Immunol. 2011;11:416–426. doi: 10.1038/nri2986. [PMID:21597477] [DOI] [PubMed] [Google Scholar]
- Hu P. Luo BH. Integrin bi-directional signaling across the plasma membrane. J Cell Physiol. 2013;228:306–312. doi: 10.1002/jcp.24154. [PMID:22767296] [DOI] [PubMed] [Google Scholar]
- Humphries JD. Byron A. Humphries MJ. Integrin ligands at a glance. J Cell Sci. 2006;119:3901–3903. doi: 10.1242/jcs.03098. (Pt 19): [PMID:16988024] [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ivaska J. Heino J. Cooperation between integrins and growth factor receptors in signaling and endocytosis. Annu Rev Cell Dev Biol. 2011;27:291–320. doi: 10.1146/annurev-cellbio-092910-154017. [PMID:21663443] [DOI] [PubMed] [Google Scholar]
- Kim C. Ye F. Ginsberg MH. Regulation of integrin activation. Annu Rev Cell Dev Biol. 2011;27:321–345. doi: 10.1146/annurev-cellbio-100109-104104. [PMID:21663444] [DOI] [PubMed] [Google Scholar]
- Roca-Cusachs P. Iskratsch T. Sheetz MP. Finding the weakest link: exploring integrin-mediated mechanical molecular pathways. J Cell Sci. 2012;125:3025–3038. doi: 10.1242/jcs.095794. (Pt 13): [PMID:22797926] [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shattil SJ. Kim C. Ginsberg MH. The final steps of integrin activation: the end game. Nat Rev Mol Cell Biol. 2010;11:288–300. doi: 10.1038/nrm2871. [PMID:20308986] [DOI] [PMC free article] [PubMed] [Google Scholar]
- Weber GF. Bjerke MA. DeSimone DW. Integrins and cadherins join forces to form adhesive networks. J Cell Sci. 2011;124:1183–1193. doi: 10.1242/jcs.064618. (Pt 8): [PMID:21444749] [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wickström SA. Fässler R. Regulation of membrane traffic by integrin signaling. Trends Cell Biol. 2011;21:266–273. doi: 10.1016/j.tcb.2011.02.003. [PMID:21440440] [DOI] [PubMed] [Google Scholar]
- Wu X. Reddy DS. Integrins as receptor targets for neurological disorders. Pharmacol Ther. 2012;134:68–81. doi: 10.1016/j.pharmthera.2011.12.008. [PMID:22233753] [DOI] [PMC free article] [PubMed] [Google Scholar]
Further reading
- Kuhn M. Endothelial actions of atrial and B-type natriuretic peptides. Br J Pharmacol. 2012;166:522–531. doi: 10.1111/j.1476-5381.2012.01827.x. [PMID:22220582] [DOI] [PMC free article] [PubMed] [Google Scholar]
- Misono KS. Philo JS. Arakawa T. Ogata CM. Qiu Y. Ogawa H. Young HS. Structure, signaling mechanism and regulation of the natriuretic peptide receptor guanylate cyclase. FEBS J. 2011;278:1818–1829. doi: 10.1111/j.1742-4658.2011.08083.x. [PMID:21375693] [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pandey KN. The functional genomics of guanylyl cyclase/natriuretic peptide receptor-A: perspectives and paradigms. FEBS J. 2011;278:1792–1807. doi: 10.1111/j.1742-4658.2011.08081.x. [PMID:21375691] [DOI] [PMC free article] [PubMed] [Google Scholar]
- Potter LR. Guanylyl cyclase structure, function and regulation. Cell Signal. 2011;23:1921–1926. doi: 10.1016/j.cellsig.2011.09.001. [PMID:21914472] [DOI] [PMC free article] [PubMed] [Google Scholar]
- Potter LR. Natriuretic peptide metabolism, clearance and degradation. FEBS J. 2011;278:1808–1817. doi: 10.1111/j.1742-4658.2011.08082.x. [PMID:21375692] [DOI] [PMC free article] [PubMed] [Google Scholar]
- Potter LR. Regulation and therapeutic targeting of peptide-activated receptor guanylyl cyclases. Pharmacol Ther. 2011;130:71–82. doi: 10.1016/j.pharmthera.2010.12.005. [PMID:21185863] [DOI] [PMC free article] [PubMed] [Google Scholar]
Further reading
- Barton GM. Kagan JC. A cell biological view of Toll-like receptor function: regulation through compartmentalization. Nat Rev Immunol. 2009;9:535–542. doi: 10.1038/nri2587. [PMID:19556980] [DOI] [PMC free article] [PubMed] [Google Scholar]
- Buchanan MM. Hutchinson M. Watkins LR. Yin H. Toll-like receptor 4 in CNS pathologies. J Neurochem. 2010;114:13–27. doi: 10.1111/j.1471-4159.2010.06736.x. [PMID:20402965] [DOI] [PMC free article] [PubMed] [Google Scholar]
- Celis E. Toll-like receptor ligands energize peptide vaccines through multiple paths. Cancer Res. 2007;67:7945–7947. doi: 10.1158/0008-5472.CAN-07-1652. [PMID:17804699] [DOI] [PubMed] [Google Scholar]
- Chao W. Toll-like receptor signaling: a critical modulator of cell survival and ischemic injury in the heart. Am J Physiol Heart Circ Physiol. 2009;296:H1–12. doi: 10.1152/ajpheart.00995.2008. [PMID:19011041] [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chiron D. Jego G. Pellat-Deuceunynck C. Toll-like receptors: expression and involvement in multiple myeloma. Leuk Res. 2010;34:1545–1550. doi: 10.1016/j.leukres.2010.06.002. [PMID:20594595] [DOI] [PubMed] [Google Scholar]
- Downes CE. Crack PJ. Neural injury following stroke: are Toll-like receptors the link between the immune system and the CNS? Br J Pharmacol. 2010;160:1872–1888. doi: 10.1111/j.1476-5381.2010.00864.x. [PMID:20649586] [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ehlers M. Ravetch JV. Opposing effects of Toll-like receptor stimulation induce autoimmunity or tolerance. Trends Immunol. 2007;28:74–79. doi: 10.1016/j.it.2006.12.006. [PMID:17197239] [DOI] [PubMed] [Google Scholar]
- Garantziotis S. Hollingsworth JW. Zaas AK. Schwartz DA. The effect of toll-like receptors and toll-like receptor genetics in human disease. Annu Rev Med. 2008;59:343–359. doi: 10.1146/annurev.med.59.061206.112455. [PMID:17845139] [DOI] [PubMed] [Google Scholar]
- Hennessy EJ. Parker AE. O'Neill LA. Targeting Toll-like receptors: emerging therapeutics? Nat Rev Drug Discov. 2010;9:293–307. doi: 10.1038/nrd3203. [PMID:20380038] [DOI] [PubMed] [Google Scholar]
- Hirsch I. Caux C. Hasan U. Bendriss-Vermare N. Olive D. Impaired Toll-like receptor 7 and 9 signaling: from chronic viral infections to cancer. Trends Immunol. 2010;31:391–397. doi: 10.1016/j.it.2010.07.004. [PMID:20832362] [DOI] [PubMed] [Google Scholar]
- Hori M. Nishida K. Toll-like receptor signaling: defensive or offensive for the heart? Circ Res. 2008;102:137–139. doi: 10.1161/CIRCRESAHA.107.170225. [PMID:18239139] [DOI] [PubMed] [Google Scholar]
- Kanzler H. Barrat FJ. Hessel EM. Coffman RL. Therapeutic targeting of innate immunity with Toll-like receptor agonists and antagonists. Nat Med. 2007;13:552–559. doi: 10.1038/nm1589. [PMID:17479101] [DOI] [PubMed] [Google Scholar]
- Könner AC. Brüning JC. Toll-like receptors: linking inflammation to metabolism. Trends Endocrinol Metab. 2011;22:16–23. doi: 10.1016/j.tem.2010.08.007. [PMID:20888253] [DOI] [PubMed] [Google Scholar]
- Lecat A. Piette J. Legrand-Poels S. The protein Nod2: an innate receptor more complex than previously assumed. Biochem Pharmacol. 2010;80:2021–2031. doi: 10.1016/j.bcp.2010.07.016. [PMID:20643110] [DOI] [PubMed] [Google Scholar]
- Li H. Sun B. Toll-like receptor 4 in atherosclerosis. J Cell Mol Med. 2007;11:88–95. doi: 10.1111/j.1582-4934.2007.00011.x. [PMID:17367503] [DOI] [PMC free article] [PubMed] [Google Scholar]
- Marsh BJ. Williams-Karnesky RL. Stenzel-Poore MP. Toll-like receptor signaling in endogenous neuroprotection and stroke. Neuroscience. 2009;158:1007–1020. doi: 10.1016/j.neuroscience.2008.07.067. [PMID:18809468] [DOI] [PMC free article] [PubMed] [Google Scholar]
- Marshak-Rothstein A. Rifkin IR. Immunologically active autoantigens: the role of toll-like receptors in the development of chronic inflammatory disease. Annu Rev Immunol. 2007;25:419–441. doi: 10.1146/annurev.immunol.22.012703.104514. [PMID:17378763] [DOI] [PubMed] [Google Scholar]
- Monie TP. Bryant CE. Gay NJ. Activating immunity: lessons from the TLRs and NLRs. Trends Biochem Sci. 2009;34:553–561. doi: 10.1016/j.tibs.2009.06.011. [PMID:19818630] [DOI] [PubMed] [Google Scholar]
- O'Neill LA. Bowie AG. The family of five: TIR-domain-containing adaptors in Toll-like receptor signalling. Nat Rev Immunol. 2007;7:353–364. doi: 10.1038/nri2079. [PMID:17457343] [DOI] [PubMed] [Google Scholar]
- O'Neill LA. Sheedy FJ. McCoy CE. MicroRNAs: the fine-tuners of Toll-like receptor signalling. Nat Rev Immunol. 2011;11:163–175. doi: 10.1038/nri2957. [PMID:21331081] [DOI] [PubMed] [Google Scholar]
- Sabroe I. Parker LC. Dower SK. Whyte MK. The role of TLR activation in inflammation. J Pathol. 2008;214:126–135. doi: 10.1002/path.2264. [PMID:18161748] [DOI] [PubMed] [Google Scholar]
- Saitoh S. Miyake K. Regulatory molecules required for nucleotide-sensing Toll-like receptors. Immunol Rev. 2009;227:32–43. doi: 10.1111/j.1600-065X.2008.00729.x. [PMID:19120473] [DOI] [PubMed] [Google Scholar]
- Sanjuan MA. Milasta S. Green DR. Toll-like receptor signaling in the lysosomal pathways. Immunol Rev. 2009;227:203–220. doi: 10.1111/j.1600-065X.2008.00732.x. [PMID:19120486] [DOI] [PubMed] [Google Scholar]
- Schroder K. Tschopp J. The inflammasomes. Cell. 2010;140:821–832. doi: 10.1016/j.cell.2010.01.040. [PMID:20303873] [DOI] [PubMed] [Google Scholar]
- Shaw PJ. Lamkanfi M. Kanneganti TD. NOD-like receptor (NLR) signaling beyond the inflammasome. Eur J Immunol. 2010;40:624–627. doi: 10.1002/eji.200940211. [PMID:20201016] [DOI] [PMC free article] [PubMed] [Google Scholar]
- Takeuchi O. Akira S. Pattern recognition receptors and inflammation. Cell. 2010;140:805–820. doi: 10.1016/j.cell.2010.01.022. [PMID:20303872] [DOI] [PubMed] [Google Scholar]
- Trinchieri G. Sher A. Cooperation of Toll-like receptor signals in innate immune defence. Nat Rev Immunol. 2007;7:179–190. doi: 10.1038/nri2038. [PMID:17318230] [DOI] [PubMed] [Google Scholar]
- Wenzel J. Tormo D. Tüting T. Toll-like receptor-agonists in the treatment of skin cancer: history, current developments and future prospects. Handb Exp Pharmacol. 2008;183:201–220. doi: 10.1007/978-3-540-72167-3_10. [PMID:18071661] [DOI] [PubMed] [Google Scholar]
- Werling D. Jann OC. Offord V. Glass EJ. Coffey TJ. Variation matters: TLR structure and species-specific pathogen recognition. Trends Immunol. 2009;30:124–130. doi: 10.1016/j.it.2008.12.001. [PMID:19211304] [DOI] [PubMed] [Google Scholar]
Further reading
- Ehrlich M. Horbelt D. Marom B. Knaus P. Henis YI. Homomeric and heteromeric complexes among TGF-β and BMP receptors and their roles in signaling. Cell Signal. 2011;23:1424–1432. doi: 10.1016/j.cellsig.2011.04.004. [PMID:21515362] [DOI] [PubMed] [Google Scholar]
- Hinck AP. Structural studies of the TGF-βs and their receptors - insights into evolution of the TGF-β superfamily. FEBS Lett. 2012;586:1860–1870. doi: 10.1016/j.febslet.2012.05.028. [PMID:22651914] [DOI] [PubMed] [Google Scholar]
- Massagué J. TGFβ signalling in context. Nat Rev Mol Cell Biol. 2012;13:616–630. doi: 10.1038/nrm3434. [PMID:22992590] [DOI] [PMC free article] [PubMed] [Google Scholar]
- Moustakas A. Heldin CH. The regulation of TGFbeta signal transduction. Development. 2009;136:3699–3714. doi: 10.1242/dev.030338. [PMID:19855013] [DOI] [PubMed] [Google Scholar]
- Rider CC. Mulloy B. Bone morphogenetic protein and growth differentiation factor cytokine families and their protein antagonists. Biochem J. 2010;429:1–12. doi: 10.1042/BJ20100305. [PMID:20545624] [DOI] [PubMed] [Google Scholar]
- Santibañez JF. Quintanilla M. Bernabeu C. TGF-β/TGF-β receptor system and its role in physiological and pathological conditions. Clin Sci. 2011;121:233–251. doi: 10.1042/CS20110086. [PMID:21615335] [DOI] [PubMed] [Google Scholar]
- Xu P. Liu J. Derynck R. Post-translational regulation of TGF-β receptor and Smad signaling. FEBS Lett. 2012;586:1871–1884. doi: 10.1016/j.febslet.2012.05.010. [PMID:22617150] [DOI] [PMC free article] [PubMed] [Google Scholar]
Further reading
- Alsina FC. Ledda F. Paratcha G. New insights into the control of neurotrophic growth factor receptor signaling: implications for nervous system development and repair. J Neurochem. 2012;123:652–661. doi: 10.1111/jnc.12021. [PMID:22994539] [DOI] [PubMed] [Google Scholar]
- Arteaga CL. Sliwkowski MX. Osborne CK. Perez EA. Puglisi F. Gianni L. Treatment of HER2-positive breast cancer: current status and future perspectives. Nat Rev Clin Oncol. 2012;9:16–32. doi: 10.1038/nrclinonc.2011.177. [PMID:22124364] [DOI] [PubMed] [Google Scholar]
- Camidge DR. Doebele RC. Treating ALK-positive lung cancer–early successes and future challenges. Nat Rev Clin Oncol. 2012;9:268–277. doi: 10.1038/nrclinonc.2012.43. [PMID:22473102] [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chen Y. Fu AK. Ip NY. Eph receptors at synapses: implications in neurodegenerative diseases. Cell Signal. 2012;24:606–611. doi: 10.1016/j.cellsig.2011.11.016. [PMID:22120527] [DOI] [PubMed] [Google Scholar]
- Fu HL. Valiathan RR. Arkwright R. Sohail A. Mihai C. Kumarasiri M. Mahasenan KV. Mobashery S. Huang P. Agarwal G, et al. Discoidin domain receptors: unique receptor tyrosine kinases in collagen-mediated signaling. J Biol Chem. 2013;288:7430–7437. doi: 10.1074/jbc.R112.444158. [PMID:23335507] [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gherardi E. Birchmeier W. Birchmeier C. Vande Woude G. Targeting MET in cancer: rationale and progress. Nat Rev Cancer. 2012;12:89–103. doi: 10.1038/nrc3205. [PMID:22270953] [DOI] [PubMed] [Google Scholar]
- Goetz R. Mohammadi M. Exploring mechanisms of FGF signalling through the lens of structural biology. Nat Rev Mol Cell Biol. 2013;14:166–180. doi: 10.1038/nrm3528. [PMID:23403721] [DOI] [PMC free article] [PubMed] [Google Scholar]
- Guillemot F. Zimmer C. From cradle to grave: the multiple roles of fibroblast growth factors in neural development. Neuron. 2011;71:574–588. doi: 10.1016/j.neuron.2011.08.002. [PMID:21867876] [DOI] [PubMed] [Google Scholar]
- Higashiyama S. Nanba D. Nakayama H. Inoue H. Fukuda S. Ectodomain shedding and remnant peptide signalling of EGFRs and their ligands. J Biochem. 2011;150:15–22. doi: 10.1093/jb/mvr068. [PMID:21610047] [DOI] [PubMed] [Google Scholar]
- Ibáñez CF. Simi A. p75 neurotrophin receptor signaling in nervous system injury and degeneration: paradox and opportunity. Trends Neurosci. 2012;35:431–440. doi: 10.1016/j.tins.2012.03.007. [PMID:22503537] [DOI] [PubMed] [Google Scholar]
- Koh GY. Orchestral actions of angiopoietin-1 in vascular regeneration. Trends Mol Med. 2013;19:31–39. doi: 10.1016/j.molmed.2012.10.010. [PMID:23182855] [DOI] [PubMed] [Google Scholar]
- Larsen AK. Ouaret D. El Ouadrani K. Petitprez A. Targeting EGFR and VEGF(R) pathway cross-talk in tumor survival and angiogenesis. Pharmacol Ther. 2011;131:80–90. doi: 10.1016/j.pharmthera.2011.03.012. [PMID:21439312] [DOI] [PubMed] [Google Scholar]
- Lefebvre J. Ancot F. Leroy C. Muharram G. Lemière A. Tulasne D. Met degradation: more than one stone to shoot a receptor down. FASEB J. 2012;26:1387–1399. doi: 10.1096/fj.11-197723. [PMID:22223753] [DOI] [PubMed] [Google Scholar]
- Leitinger B. Transmembrane collagen receptors. Annu Rev Cell Dev Biol. 2011;27:265–290. doi: 10.1146/annurev-cellbio-092910-154013. [PMID:21568710] [DOI] [PubMed] [Google Scholar]
- Lennartsson J. Rönnstrand L. Stem cell factor receptor/c-Kit: from basic science to clinical implications. Physiol Rev. 2012;92:1619–1649. doi: 10.1152/physrev.00046.2011. [PMID:23073628] [DOI] [PubMed] [Google Scholar]
- Liang G. Liu Z. Wu J. Cai Y. Li X. Anticancer molecules targeting fibroblast growth factor receptors. Trends Pharmacol Sci. 2012;33:531–541. doi: 10.1016/j.tips.2012.07.001. [PMID:22884522] [DOI] [PubMed] [Google Scholar]
- Lisle JE. Mertens-Walker I. Rutkowski R. Herington AC. Stephenson SA. Eph receptors and their ligands: promising molecular biomarkers and therapeutic targets in prostate cancer. Biochim Biophys Acta. 2013;1835:243–257. doi: 10.1016/j.bbcan.2013.01.003. [PMID:23396052] [DOI] [PubMed] [Google Scholar]
- Lu B. Nagappan G. Guan X. Nathan PJ. Wren P. BDNF-based synaptic repair as a disease-modifying strategy for neurodegenerative diseases. Nat Rev Neurosci. 2013;14:401–416. doi: 10.1038/nrn3505. [PMID:23674053] [DOI] [PubMed] [Google Scholar]
- Morandi A. Plaza-Menacho I. Isacke CM. RET in breast cancer: functional and therapeutic implications. Trends Mol Med. 2011;17:149–157. doi: 10.1016/j.molmed.2010.12.007. [PMID:21251878] [DOI] [PubMed] [Google Scholar]
- Peters S. Adjei AA. MET: a promising anticancer therapeutic target. Nat Rev Clin Oncol. 2012;9:314–326. doi: 10.1038/nrclinonc.2012.71. [PMID:22566105] [DOI] [PubMed] [Google Scholar]
- Roskoski R., Jr Anaplastic lymphoma kinase (ALK): structure, oncogenic activation, and pharmacological inhibition. Pharmacol Res. 2013;68:68–94. doi: 10.1016/j.phrs.2012.11.007. [PMID:23201355] [DOI] [PubMed] [Google Scholar]
- Sheffler-Collins SI. Dalva MB. EphBs: an integral link between synaptic function and synaptopathies. Trends Neurosci. 2012;35:293–304. doi: 10.1016/j.tins.2012.03.003. [PMID:22516618] [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shibuya M. Vascular endothelial growth factor and its receptor system: physiological functions in angiogenesis and pathological roles in various diseases. J Biochem. 2013;153:13–19. doi: 10.1093/jb/mvs136. [PMID:23172303] [DOI] [PMC free article] [PubMed] [Google Scholar]
- Turner CA. Watson SJ. Akil H. The fibroblast growth factor family: neuromodulation of affective behavior. Neuron. 2012;76:160–174. doi: 10.1016/j.neuron.2012.08.037. [PMID:23040813] [DOI] [PMC free article] [PubMed] [Google Scholar]
- Woo KV. Baldwin HS. Role of Tie1 in shear stress and atherosclerosis. Trends Cardiovasc Med. 2011;21:118–123. doi: 10.1016/j.tcm.2012.03.009. [PMID:22681967] [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yamanashi Y. Tezuka T. Yokoyama K. Activation of receptor protein-tyrosine kinases from the cytoplasmic compartment. J Biochem. 2012;151:353–359. doi: 10.1093/jb/mvs013. [PMID:22343747] [DOI] [PubMed] [Google Scholar]
Further reading
- Böhmer F. Szedlacsek S. Tabernero L. Ostman A. Hertog J. Protein tyrosine phosphatase structure-function relationships in regulation and pathogenesis. FEBS J. 2013;280:413–431. doi: 10.1111/j.1742-4658.2012.08655.x. [PMID:22682070] [DOI] [PubMed] [Google Scholar]
- Dushek O. Goyette J. Merwe PA. Non-catalytic tyrosine-phosphorylated receptors. Immunol Rev. 2012;250:258–276. doi: 10.1111/imr.12008. [PMID:23046135] [DOI] [PubMed] [Google Scholar]
- He R. Zeng LF. He Y. Zhang S. Zhang ZY. Small molecule tools for functional interrogation of protein tyrosine phosphatases. FEBS J. 2013;280:731–750. doi: 10.1111/j.1742-4658.2012.08718.x. [PMID:22816879] [DOI] [PMC free article] [PubMed] [Google Scholar]
- Julien SG. Dubé N. Hardy S. Tremblay ML. Inside the human cancer tyrosine phosphatome. Nat Rev Cancer. 2011;11:35–49. doi: 10.1038/nrc2980. [PMID:21179176] [DOI] [PubMed] [Google Scholar]
- Mohebiany AN. Nikolaienko RM. Bouyain S. Harroch S. Receptor-type tyrosine phosphatase ligands: looking for the needle in the haystack. FEBS J. 2013;280:388–400. doi: 10.1111/j.1742-4658.2012.08653.x. [PMID:22682003] [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sastry SK. Elferink LA. Checks and balances: interplay of RTKs and PTPs in cancer progression. Biochem Pharmacol. 2011;82:435–440. doi: 10.1016/j.bcp.2011.06.016. [PMID:21704606] [DOI] [PubMed] [Google Scholar]
Further reading
- Aggarwal BB. Signalling pathways of the TNF superfamily: a double-edged sword. Nat Rev Immunol. 2003;3:745–756. doi: 10.1038/nri1184. [PMID:12949498] [DOI] [PubMed] [Google Scholar]
- Ashkenazi A. Targeting death and decoy receptors of the tumour-necrosis factor superfamily. Nat Rev Cancer. 2002;2:420–430. doi: 10.1038/nrc821. [PMID:12189384] [DOI] [PubMed] [Google Scholar]
- Huang EJ. Reichardt LF. Neurotrophins: roles in neuronal development and function. Annu Rev Neurosci. 2001;24:677–736. doi: 10.1146/annurev.neuro.24.1.677. [PMID:11520916] [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mahmood Z. Shukla Y. Death receptors: targets for cancer therapy. Exp Cell Res. 2010;316:887–899. doi: 10.1016/j.yexcr.2009.12.011. [PMID:20026107] [DOI] [PubMed] [Google Scholar]
- Rickert RC. Jellusova J. Miletic AV. Signaling by the tumor necrosis factor receptor superfamily in B-cell biology and disease. Immunol Rev. 2011;244:115–133. doi: 10.1111/j.1600-065X.2011.01067.x. [PMID:22017435] [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tansey MG. Szymkowski DE. The TNF superfamily in 2009: new pathways, new indications, and new drugs. Drug Discov Today. 2009;14:1082–1088. doi: 10.1016/j.drudis.2009.10.002. [PMID:19837186] [DOI] [PubMed] [Google Scholar]
References
- 1.Akeson AL, et al. J Biol Chem. 1996;271:30517–30523. doi: 10.1074/jbc.271.48.30517. [PMID: 8940020 ] [DOI] [PubMed] [Google Scholar]
- 2.Morokata, et al. Int Immunopharmacol. 2002;2:1693–1702. doi: 10.1016/s1567-5769(02)00191-1. [PMID: 12469943 ] [DOI] [PubMed] [Google Scholar]
- 3.Tilley JW, et al. Journal of the American Chemical Society. 1997;119:7589–7590. [Google Scholar]
- 4.Klein RD, et al. Nature. 1997;387:717–721. doi: 10.1038/42722. [PMID: 9192898 ] [DOI] [PubMed] [Google Scholar]
- 5.Mologni L, et al. J Mol Endocrinol. 2006;37:199–212. doi: 10.1677/jme.1.01999. [PMID: 17032739 ] [DOI] [PubMed] [Google Scholar]
- 6.Treanor JJ, et al. Nature. 1996;382:80–83. doi: 10.1038/382080a0. [PMID: 8657309 ] [DOI] [PubMed] [Google Scholar]
- 7.Goodman SL, et al. J Med Chem. 2002;45:1045–1051. doi: 10.1021/jm0102598. [PMID: 11855984 ] [DOI] [PubMed] [Google Scholar]
- 8.Lee Y, et al. J Biomol Screen. 2004;9:687–694. doi: 10.1177/1087057104268125. [PMID: 15634795 ] [DOI] [PubMed] [Google Scholar]
- 9.Lin Kc, et al. J Med Chem. 1999;42:920–934. doi: 10.1021/jm980673g. [PMID: 10072689 ] [DOI] [PubMed] [Google Scholar]
- 10.Liu G, et al. J Med Chem. 2000;43:4025–4040. doi: 10.1021/jm0002782. [PMID: 11052808 ] [DOI] [PubMed] [Google Scholar]
- 11.Marcinkiewicz C, et al. Cancer Res. 2003;63:2020–2023. [PMID: 12727812 ] [PubMed] [Google Scholar]
- 12.Matsuno H, et al. Circulation. 1994;90:2203–2206. doi: 10.1161/01.cir.90.5.2203. [PMID: 7955174 ] [DOI] [PubMed] [Google Scholar]
- 13.Miller MW, et al. Proc Natl Acad Sci U S A. 2009;106:719–724. doi: 10.1073/pnas.0811622106. [PMID: 19141632 ] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Wang WY, et al. Mol Pharmacol. 2006;70:1380–1389. doi: 10.1124/mol.106.023986. [PMID: 16837624 ] [DOI] [PubMed] [Google Scholar]
- 15.Delporte C, et al. Eur J Pharmacol. 1991;207:81–88. doi: 10.1016/s0922-4106(05)80041-0. [PMID: 1680722 ] [DOI] [PubMed] [Google Scholar]
- 16.Deschênes J, et al. Peptides. 2005;26:517–524. doi: 10.1016/j.peptides.2004.10.017. [PMID: 15652659 ] [DOI] [PubMed] [Google Scholar]
- 17.Drewett JG, et al. J Biol Chem. 1995;270:4668–4674. doi: 10.1074/jbc.270.9.4668. [PMID: 7876238 ] [DOI] [PubMed] [Google Scholar]
- 18.Harris LA, Crowell MD. Curr Opin Mol Ther. 2007;9:403–410. [PMID: 17694454 ] [PubMed] [Google Scholar]
- 19.Hobbs A, et al. Circulation. 2004;110:1231–1235. doi: 10.1161/01.CIR.0000141802.29945.34. [PMID: 15337698 ] [DOI] [PubMed] [Google Scholar]
- 20.Hunt DM, et al. Mol Cell Biochem. 2010;334:157–168. doi: 10.1007/s11010-009-0331-y. [PMID: 19941038 ] [DOI] [PubMed] [Google Scholar]
- 21.Kambayashi Y, et al. FEBS Lett. 1989;248:28–34. doi: 10.1016/0014-5793(89)80425-9. [PMID: 2542088 ] [DOI] [PubMed] [Google Scholar]
- 22.Maack T, et al. Science. 1987;238:675–678. doi: 10.1126/science.2823385. [PMID: 2823385 ] [DOI] [PubMed] [Google Scholar]
- 23.Moffatt P, et al. J Biol Chem. 2007;282:36454–36462. doi: 10.1074/jbc.M708596200. [PMID: 17951249 ] [DOI] [PubMed] [Google Scholar]
- 24.Morishita Y, et al. Biochem Biophys Res Commun. 1991;176:949–957. doi: 10.1016/0006-291x(91)90374-g. [PMID: 1674870 ] [DOI] [PubMed] [Google Scholar]
- 25.Murthy KS, Makhlouf GM. J Biol Chem. 1999;274:17587–17592. doi: 10.1074/jbc.274.25.17587. [PMID: 10364194 ] [DOI] [PubMed] [Google Scholar]
- 26.Olson LJ, et al. Mol Pharmacol. 1996;50:430–435. [PMID: 8700153 ] [PubMed] [Google Scholar]
- 27.Suga S, et al. Endocrinology. 1992;130:229–239. doi: 10.1210/endo.130.1.1309330. [PMID: 1309330 ] [DOI] [PubMed] [Google Scholar]
- 28.Veale CA, et al. Bioorg Med Chem Lett. 2000;10:1949–1952. doi: 10.1016/s0960-894x(00)00387-5. [PMID: 10987424 ] [DOI] [PubMed] [Google Scholar]
- 29.Wyss DF, et al. J Antibiot. 1991;44:172–180. doi: 10.7164/antibiotics.44.172. [PMID: 1826288 ] [DOI] [PubMed] [Google Scholar]
- 30.Alexopoulou L, et al. Nature. 2001;413:732–738. doi: 10.1038/35099560. [PMID: 11607032 ] [DOI] [PubMed] [Google Scholar]
- 31.Hayashi F, et al. Nature. 2001;410:1099–1103. doi: 10.1038/35074106. [PMID: 11323673 ] [DOI] [PubMed] [Google Scholar]
- 32.Heil F, et al. Eur J Immunol. 2003;33:2987–2997. doi: 10.1002/eji.200324238. [PMID: 14579267 ] [DOI] [PubMed] [Google Scholar]
- 33.Hemmi H, et al. Nat Immunol. 2002;3:196–200. doi: 10.1038/ni758. [PMID: 11812998 ] [DOI] [PubMed] [Google Scholar]
- 34.Hemmi H, et al. Nature. 2000;408:740–745. doi: 10.1038/35047123. [PMID: 11130078 ] [DOI] [PubMed] [Google Scholar]
- 35.Ingalls RR, et al. J Immunol. 1998;161:5413–5420. [PMID: 9820516 ] [PubMed] [Google Scholar]
- 36.Kawasaki K, et al. J Biol Chem. 2000;275:2251–2254. doi: 10.1074/jbc.275.4.2251. [PMID: 10644670 ] [DOI] [PubMed] [Google Scholar]
- 37.Martinon F, et al. Nature. 2006;440:237–241. doi: 10.1038/nature04516. [PMID: 16407889 ] [DOI] [PubMed] [Google Scholar]
- 38.Ohashi K, et al. J Immunol. 2000;164:558–561. doi: 10.4049/jimmunol.164.2.558. [PMID: 10623794 ] [DOI] [PubMed] [Google Scholar]
- 39.Poltorak A, et al. Science. 1998;282:2085–2088. doi: 10.1126/science.282.5396.2085. [PMID: 9851930 ] [DOI] [PubMed] [Google Scholar]
- 40.Schroder K, Tschopp J. Cell. 2010;140:821–832. doi: 10.1016/j.cell.2010.01.040. [PMID: 20303873 ] [DOI] [PubMed] [Google Scholar]
- 41.Schwandner R, et al. J Biol Chem. 1999;274:17406–17409. doi: 10.1074/jbc.274.25.17406. [PMID: 10364168 ] [DOI] [PubMed] [Google Scholar]
- 42.Takeuchi O, Akira S. Cell. 2010;140:805–820. doi: 10.1016/j.cell.2010.01.022. [PMID: 20303872 ] [DOI] [PubMed] [Google Scholar]
- 43.Takeuchi O, et al. Int Immunol. 2001;13:933–940. doi: 10.1093/intimm/13.7.933. [PMID: 11431423 ] [DOI] [PubMed] [Google Scholar]
- 44.Takeuchi O, et al. J Immunol. 2002;169:10–14. doi: 10.4049/jimmunol.169.1.10. [PMID: 12077222 ] [DOI] [PubMed] [Google Scholar]
- 45.Yoshimura A, et al. J Immunol. 1999;163:1–5. [PMID: 10384090 ] [PubMed] [Google Scholar]
- 46.DaCosta Byfield S, et al. Mol Pharmacol. 2004;65:744–752. doi: 10.1124/mol.65.3.744. [PMID: 14978253 ] [DOI] [PubMed] [Google Scholar]
- 47.Li MO, Flavell RA. Cell. 2008;134:392–404. doi: 10.1016/j.cell.2008.07.025. [PMID: 18692464 ] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Moustakas A, Heldin CH. Development. 2009;136:3699–3714. doi: 10.1242/dev.030338. [PMID: 19855013 ] [DOI] [PubMed] [Google Scholar]
- 49.Shi Y, Massagué J. Cell. 2003;113:685–700. doi: 10.1016/s0092-8674(03)00432-x. [PMID: 12809600 ] [DOI] [PubMed] [Google Scholar]
- 50.Vogt J, et al. Cell Signal. 2011;23:1831–1842. doi: 10.1016/j.cellsig.2011.06.019. [PMID: 21740966 ] [DOI] [PubMed] [Google Scholar]
- 51.Zhou G, et al. J Clin Invest. 2001;108:1167–1174. doi: 10.1172/JCI13505. [PMID: 11602624 ] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Albaugh P, et al. ACS Medicinal Chemistry Letters. 2012;3:140–145. doi: 10.1021/ml200261d. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Arena S, et al. Proc Natl Acad Sci USA. 2007;104:11412–11417. doi: 10.1073/pnas.0703205104. [PMID: 17595299 ] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Batley BL, et al. Life Sci. 1998;62:143–150. doi: 10.1016/s0024-3205(97)01060-6. [PMID: 9488112 ] [DOI] [PubMed] [Google Scholar]
- 55.Blume-Jensen P, Hunter T. Nature. 2001;411:355–365. doi: 10.1038/35077225. [PMID: 11357143 ] [DOI] [PubMed] [Google Scholar]
- 56.Cazorla M, et al. J Clin Invest. 2011;121:1846–1857. doi: 10.1172/JCI43992. [PMID: 21505263 ] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Choi SJ, et al. Bioorg Med Chem Lett. 2010;20:2033–2037. doi: 10.1016/j.bmcl.2010.01.039. [PMID: 20153646 ] [DOI] [PubMed] [Google Scholar]
- 58.Cui JJ, et al. J Med Chem. 2012;55:8091–8109. doi: 10.1021/jm300967g. [PMID: 22924734 ] [DOI] [PubMed] [Google Scholar]
- 59.Day E, et al. Eur J Pharmacol. 2008;599:44–53. doi: 10.1016/j.ejphar.2008.10.014. [PMID: 18938156 ] [DOI] [PubMed] [Google Scholar]
- 60.Dechant G, et al. J Neurosci. 1997;17:5281–5287. doi: 10.1523/JNEUROSCI.17-14-05281.1997. [PMID: 9204912 ] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Fabbro D, et al. Methods Mol Biol. 2012;795:1–34. doi: 10.1007/978-1-61779-337-0_1. [PMID: 21960212 ] [DOI] [PubMed] [Google Scholar]
- 62.Fraley ME, et al. Bioorg Med Chem Lett. 2002;12:3537–3541. doi: 10.1016/s0960-894x(02)00827-2. [PMID: 12443771 ] [DOI] [PubMed] [Google Scholar]
- 63.Gaul MD, et al. Bioorg Med Chem Lett. 2003;13:637–640. doi: 10.1016/s0960-894x(02)01047-8. [PMID: 12639547 ] [DOI] [PubMed] [Google Scholar]
- 64.Gerber DE, Minna JD. Cancer Cell. 2010;18:548–551. doi: 10.1016/j.ccr.2010.11.033. [PMID: 21156280 ] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Grassot J, et al. Nucleic Acids Res. 2003;31:353–358. doi: 10.1093/nar/gkg036. [PMID: 12520021 ] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Graus-Porta D, et al. EMBO J. 1997;16:1647–1655. doi: 10.1093/emboj/16.7.1647. [PMID: 9130710 ] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Grumolato L, et al. Genes Dev. 2010;24:2517–2530. doi: 10.1101/gad.1957710. [PMID: 21078818 ] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Kubo K, et al. J Med Chem. 2005;48:1359–1366. doi: 10.1021/jm030427r. [PMID: 15743179 ] [DOI] [PubMed] [Google Scholar]
- 69.Kurachi H, et al. Biochem Biophys Res Commun. 1992;187:934–939. doi: 10.1016/0006-291x(92)91287-z. [PMID: 1530648 ] [DOI] [PubMed] [Google Scholar]
- 70.Lemmon MA, Schlessinger J. Cell. 2010;141:1117–1134. doi: 10.1016/j.cell.2010.06.011. [PMID: 20602996 ] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Liang G, et al. Trends Pharmacol Sci. 2012;33:531–541. doi: 10.1016/j.tips.2012.07.001. [PMID: 22884522 ] [DOI] [PubMed] [Google Scholar]
- 72.MacDonald RG, et al. Science. 1988;239:1134–1137. doi: 10.1126/science.2964083. [PMID: 2964083 ] [DOI] [PubMed] [Google Scholar]
- 73.Massa SM, et al. J Clin Invest. 2010;120:1774–1785. doi: 10.1172/JCI41356. [PMID: 20407211 ] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Mollard A, et al. ACS Med Chem Lett. 2011;2:907–912. doi: 10.1021/ml200198x. [PMID: 22247788 ] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Nagata K, et al. J Biol Chem. 1996;271:30022–30027. doi: 10.1074/jbc.271.47.30022. [PMID: 8939948 ] [DOI] [PubMed] [Google Scholar]
- 76.Ohmichi M, et al. Biochemistry. 1993;32:4650–4658. doi: 10.1021/bi00068a024. [PMID: 7683492 ] [DOI] [PubMed] [Google Scholar]
- 77.Ornitz DM, et al. J Biol Chem. 1996;271:15292–15297. doi: 10.1074/jbc.271.25.15292. [PMID: 8663044 ] [DOI] [PubMed] [Google Scholar]
- 78.Pearson MA, Fabbro D. Expert Rev Anticancer Ther. 2004;4:1113–1124. doi: 10.1586/14737140.4.6.1113. [PMID: 15606337 ] [DOI] [PubMed] [Google Scholar]
- 79.Sattler M, et al. Cancer Res. 2003;63:5462–5469. [PMID: 14500382 ] [PubMed] [Google Scholar]
- 80.Skaper SD, et al. J Neurochem. 2000;75:1520–1527. doi: 10.1046/j.1471-4159.2000.0751520.x. [PMID: 10987832 ] [DOI] [PubMed] [Google Scholar]
- 81.Stitt TN, et al. Cell. 1995;80:661–670. doi: 10.1016/0092-8674(95)90520-0. [PMID: 7867073 ] [DOI] [PubMed] [Google Scholar]
- 82.Ullrich A, Schlessinger J. Cell. 1990;61:203–212. doi: 10.1016/0092-8674(90)90801-k. [PMID: 2158859 ] [DOI] [PubMed] [Google Scholar]
- 83.Wesche J, et al. Biochem J. 2011;437:199–213. doi: 10.1042/BJ20101603. [PMID: 21711248 ] [DOI] [PubMed] [Google Scholar]
- 84.Whittles CE, et al. Microcirculation. 2002;9:513–522. doi: 10.1038/sj.mn.7800164. [PMID: 12483548 ] [DOI] [PubMed] [Google Scholar]
- 85.Wittman M, et al. J Med Chem. 2005;48:5639–5643. doi: 10.1021/jm050392q. [PMID: 16134929 ] [DOI] [PubMed] [Google Scholar]
- 86.Wood ER, et al. Bioorg Med Chem Lett. 2004;14:953–957. doi: 10.1016/j.bmcl.2003.12.002. [PMID: 15013000 ] [DOI] [PubMed] [Google Scholar]
- 87.Zhou W, et al. Chem Biol. 2010;17:285–295. doi: 10.1016/j.chembiol.2010.02.007. [PMID: 20338520 ] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Bouyain S, Watkins DJ. Proc Natl Acad Sci U S A. 2010;107:2443–2448. doi: 10.1073/pnas.0911235107. [PMID: 20133774 ] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Kim YS, et al. Biochem Biophys Res Commun. 2011;404:96–102. doi: 10.1016/j.bbrc.2010.11.071. [PMID: 21094132 ] [DOI] [PubMed] [Google Scholar]
- 90.Kwon SK, et al. J Biol Chem. 2010;285:13966–13978. doi: 10.1074/jbc.M109.061127. [PMID: 20139422 ] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Meng K, et al. Proc Natl Acad Sci U S A. 2000;97:2603–2608. doi: 10.1073/pnas.020487997. [PMID: 10706604 ] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Shen Y, et al. Science. 2009;326:592–596. doi: 10.1126/science.1178310. [PMID: 19833921 ] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Walzel H, et al. Immunol Lett. 1999;67:193–202. doi: 10.1016/s0165-2478(99)00012-7. [PMID: 10369126 ] [DOI] [PubMed] [Google Scholar]
- 94.Yan M, et al. Science. 2000;290:523–527. doi: 10.1126/science.290.5491.523. [PMID: 11039935 ] [DOI] [PubMed] [Google Scholar]