RET Receptor Tyrosine Kinase: Comparison
Please note this is a comparison between Version 2 by Peter Tang and Version 1 by Arun Kumar Mahato.

Rearranged during transfection (RET) is the tyrosine kinase receptor that under normal circumstances binds ligand at the cell surface and mediates various essential roles in a variety of cellular processes such as proliferation, differentiation, survival, migration, and metabolism. RET plays a pivotal role in the development of both peripheral and central nervous systems. RET is expressed from early stages of embryogenesis and remains expressed throughout all life stages. Mutations either activating or inhibiting RET result in several aggressive diseases, namely cancer and Hirschsprung disease. However, the physiological ligand-dependent activation of RET receptor is important for the survival and maintenance of several neuronal populations, appetite, and weight gain control, thus providing an opportunity for the development of disease-modifying therapeutics against neurodegeneration and obesity. In this review, we describe the structure of RET, its signaling, and its role in both normal development as well as in developmental disorders. We highlight the differences in the signaling and outcomes of constitutive and ligand-induced RET activation. Finally, we review the data on recently developed small molecular weight RET agonists and their potential for the treatment of various diseases.

  • rearranged in transfection (RET)
  • neurodegeneration
  • obesity
  • cancer
  • RET agonist
  • neurorestoration
  • retinitis pigmentosa
  • hirschsprung disease
  • glial cell line-derived neurotrophic factor family
  • growth differentiation factor 15
Please wait, diff process is still running!

References

  1. Du, Z.; Lovly, C.M. Mechanisms of receptor tyrosine kinase activation in cancer. Molecular Cancer 2018, 17, 58, doi:10.1186/s12943-018-0782-4.
  2. Plaza-Menacho, I.; Barnouin, K.; Barry, R.; Borg, A.; Orme, M.; Chauhan, R.; Mouilleron, S.; Martínez-Torres, R.J.; Meier, P.; McDonald, N.Q. RET Functions as a Dual-Specificity Kinase that Requires Allosteric Inputs from Juxtamembrane Elements. Cell Rep 2016, 17, 3319–3332, doi:10.1016/j.celrep.2016.11.061.
  3. Jmaeff, S.; Sidorova, Y.; Nedev, H.; Saarma, M.; Saragovi, H.U. Small-molecule agonists of the RET receptor tyrosine kinase activate biased trophic signals that are influenced by the presence of GFRa1 co-receptors. J. Biol. Chem. 2020, jbc.RA119.011802, doi:10.1074/jbc.RA119.011802.
  4. Esseghir, S.; Todd, S.K.; Hunt, T.; Poulsom, R.; Plaza-Menacho, I.; Reis-Filho, J.S.; Isacke, C.M. A Role for Glial Cell–Derived Neurotrophic Factor–Induced Expression by Inflammatory Cytokines and RET/GFRα1 Receptor Up-regulation in Breast Cancer. Cancer Res 2007, 67, 11732–11741, doi:10.1158/0008-5472.CAN-07-2343.
  5. Knowles, P.P.; Murray-Rust, J.; Kjær, S.; Scott, R.P.; Hanrahan, S.; Santoro, M.; Ibáñez, C.F.; McDonald, N.Q. Structure and Chemical Inhibition of the RET Tyrosine Kinase Domain. J. Biol. Chem. 2006, 281, 33577–33587, doi:10.1074/jbc.M605604200.
  6. Takahashi, M.; Ritz, J.; Cooper, G.M. Activation of a novel human transforming gene, ret, by DNA rearrangement. Cell 1985, 42, 581–588, doi:10.1016/0092-8674(85)90115-1.
  7. Takahashi, M.; Cooper, G.M. ret transforming gene encodes a fusion protein homologous to tyrosine kinases. Mol. Cell. Biol. 1987, 7, 1378–1385, doi:10.1128/mcb.7.4.1378.
  8. Takahashi, M.; Asai, N.; Iwashita, T.; Isomura, T.; Miyazaki, K.; Matsuyama, M. Characterization of the ret proto-oncogene products expressed in mouse L cells. Oncogene 1993, 8, 2925–2929.
  9. Runeberg‐Roos, P.; Saarma, P.M. Neurotrophic factor receptor RET: structure, cell biology, and inherited diseases. Annals of Medicine 2007, 39, 572–580, doi:10.1080/07853890701646256.
  10. Anders, J.; Kjar, S.; Ibáñez, C.F. Molecular modeling of the extracellular domain of the RET receptor tyrosine kinase reveals multiple cadherin-like domains and a calcium-binding site. J. Biol. Chem. 2001, 276, 35808–35817, doi:10.1074/jbc.M104968200.
  11. Nozaki, C.; Asai, N.; Murakami, H.; Iwashita, T.; Iwata, Y.; Horibe, K.; Klein, R.D.; Rosenthal, A.; Takahashi, M. Calcium-dependent Ret activation by GDNF and neurturin. Oncogene 1998, 16, 293–299, doi:10.1038/sj.onc.1201548.
  12. van Weering, D.H.J.; Moen, T.C.; Braakman, I.; Baas, P.D.; Bos, J.L. Expression of the Receptor Tyrosine Kinase Ret on the Plasma Membrane Is Dependent on Calcium. J. Biol. Chem. 1998, 273, 12077–12081, doi:10.1074/jbc.273.20.12077.
  13. Tahira, T.; Ishizaka, Y.; Itoh, F.; Sugimura, T.; Nagao, M. Characterization of ret proto-oncogene mRNAs encoding two isoforms of the protein product in a human neuroblastoma cell line. Oncogene 1990.
  14. Carter, M.T.; Yome, J.L.; Marcil, M.N.; Martin, C.A.; Vanhorne, J.B.; Mulligan, L.M. Conservation of RET proto-oncogene splicing variants and implications for RET isoform function. CGR 2001, 95, 169–176, doi:10.1159/000059341.
  15. de Graaff, E.; Srinivas, S.; Kilkenny, C.; D’Agati, V.; Mankoo, B.S.; Costantini, F.; Pachnis, V. Differential activities of the RET tyrosine kinase receptor isoforms during mammalian embryogenesis. Genes Dev 2001, 15, 2433–2444, doi:10.1101/gad.205001.
  16. Tsai, V.W.W.; Husaini, Y.; Sainsbury, A.; Brown, D.A.; Breit, S.N. The MIC-1/GDF15-GFRAL Pathway in Energy Homeostasis: Implications for Obesity, Cachexia, and Other Associated Diseases. Cell Metab. 2018, 28, 353–368, doi:10.1016/j.cmet.2018.07.018.
  17. Unsicker, K.; Spittau, B.; Krieglstein, K. The multiple facets of the TGF-β family cytokine growth/differentiation factor-15/macrophage inhibitory cytokine-1. Cytokine Growth Factor Rev. 2013, 24, 373–384, doi:10.1016/j.cytogfr.2013.05.003.
  18. Airaksinen, M.S.; Saarma, M. The GDNF family: Signalling, biological functions and therapeutic value. Nature Reviews Neuroscience 2002, 3, 383–394, doi:10.1038/nrn812.
  19. Manié, S.; Santoro, M.; Fusco, A.; Billaud, M. The RET receptor: function in development and dysfunction in congenital malformation. Trends in Genetics 2001, 17, 580–589, doi:10.1016/S0168-9525(01)02420-9.
  20. Bespalov, M.M.; Saarma, M. GDNF family receptor complexes are emerging drug targets. Trends Pharmacol. Sci. 2007, 28, 68–74, doi:10.1016/j.tips.2006.12.005.
  21. Sidorova, Y.A.; Mätlik, K.; Paveliev, M.; Lindahl, M.; Piranen, E.; Milbrandt, J.; Arumäe, U.; Saarma, M.; Bespalov, M.M. Persephin signaling through GFRalpha1: the potential for the treatment of Parkinson’s disease. Mol. Cell. Neurosci. 2010, 44, 223–232, doi:10.1016/j.mcn.2010.03.009.
  22. Baloh, R.H.; Tansey, M.G.; Lampe, P.A.; Fahrner, T.J.; Enomoto, H.; Simburger, K.S.; Leitner, M.L.; Araki, T.; Johnson, E.M.; Milbrandt, J. Artemin, a novel member of the GDNF ligand family, supports peripheral and central neurons and signals through the GFRalpha3-RET receptor complex. Neuron 1998, 21, 1291–1302, doi:10.1016/s0896-6273(00)80649-2.
  23. Yang, L.; Chang, C.-C.; Sun, Z.; Madsen, D.; Zhu, H.; Padkjær, S.B.; Wu, X.; Huang, T.; Hultman, K.; Paulsen, S.J.; et al. GFRAL is the receptor for GDF15 and is required for the anti-obesity effects of the ligand. Nature Medicine 2017, 23, 1158–1166, doi:10.1038/nm.4394.
  24. Mullican, S.E.; Lin-Schmidt, X.; Chin, C.-N.; Chavez, J.A.; Furman, J.L.; Armstrong, A.A.; Beck, S.C.; South, V.J.; Dinh, T.Q.; Cash-Mason, T.D.; et al. GFRAL is the receptor for GDF15 and the ligand promotes weight loss in mice and nonhuman primates. Nature Medicine 2017, 23, 1150–1157, doi:10.1038/nm.4392.
  25. Emmerson, P.J.; Wang, F.; Du, Y.; Liu, Q.; Pickard, R.T.; Gonciarz, M.D.; Coskun, T.; Hamang, M.J.; Sindelar, D.K.; Ballman, K.K.; et al. The metabolic effects of GDF15 are mediated by the orphan receptor GFRAL. Nat. Med. 2017, 23, 1215–1219, doi:10.1038/nm.4393.
  26. Hsu, J.-Y.; Crawley, S.; Chen, M.; Ayupova, D.A.; Lindhout, D.A.; Higbee, J.; Kutach, A.; Joo, W.; Gao, Z.; Fu, D.; et al. Non-homeostatic body weight regulation through a brainstem-restricted receptor for GDF15. Nature 2017, 550, 255–259, doi:10.1038/nature24042.
  27. Pierchala, B.A.; Milbrandt, J.; Johnson, E.M.; Jr Glial Cell Line-Derived Neurotrophic Factor-Dependent Recruitment of Ret into Lipid Rafts Enhances Signaling by Partitioning Ret from Proteasome-Dependent Degradation. The Journal of Neuroscience 2006, 26, 2777, doi:10.1523/JNEUROSCI.3420-05.2006.
  28. Tsui, C.C.; Gabreski, N.A.; Hein, S.J.; Pierchala, B.A. Lipid Rafts Are Physiologic Membrane Microdomains Necessary for the Morphogenic and Developmental Functions of Glial Cell Line-Derived Neurotrophic Factor In Vivo. The Journal of Neuroscience 2015, 35, 13233, doi:10.1523/JNEUROSCI.2935-14.2015.
  29. Murakumo, Y.; Jijiwa, M.; Asai, N.; Ichihara, M.; Takahashi, M. RET and neuroendocrine tumors. Pituitary 2006, 9, 179–192, doi:10.1007/s11102-006-0263-4.
  30. Santoro, M.; Melillo, R.M.; Carlomagno, F.; Vecchio, G.; Fusco, A. Minireview: RET: Normal and Abnormal Functions. Endocrinology 2004, 145, 5448–5451, doi:10.1210/en.2004-0922.
  31. Avantaggiato, V.; Dathan, N.A.; Grieco, M.; Fabien, N.; Lazzaro, D.; Fusco, A.; Simeone, A.; Santoro, M. Developmental expression of the RET protooncogene. Cell Growth & Differentiation 1994, 5, 305.
  32. Pachnis, V.; Mankoo, B.; Costantini, F. Expression of the c-ret proto-oncogene during mouse embryogenesis. Development 1993, 119, 1005–1017.
  33. Attié‐Bitach, T.; Abitbol, M.; Gérard, M.; Delezoide, A.-L.; Augé, J.; Pelet, A.; Amiel, J.; Pachnis, V.; Munnich, A.; Lyonnet, S.; et al. Expression of the RET proto-oncogene in human Embryos. American Journal of Medical Genetics 1998, 80, 481–486, doi:10.1002/(SICI)1096-8628(19981228)80:5<481::AID-AJMG8>3.0.CO;2-6.
  34. Kramer, E.R.; Liss, B. GDNF–Ret signaling in midbrain dopaminergic neurons and its implication for Parkinson disease. FEBS Letters 2015, 589, 3760–3772, doi:10.1016/j.febslet.2015.11.006.
  35. Mijatovic, J.; Airavaara, M.; Planken, A.; Auvinen, P.; Raasmaja, A.; Piepponen, T.P.; Costantini, F.; Ahtee, L.; Saarma, M. Constitutive Ret Activity in Knock-In Multiple Endocrine Neoplasia Type B Mice Induces Profound Elevation of Brain Dopamine Concentration via Enhanced Synthesis and Increases the Number of TH-Positive Cells in the Substantia Nigra. J. Neurosci. 2007, 27, 4799–4809, doi:10.1523/JNEUROSCI.5647-06.2007.
  36. Jiang, Q.; Liu, F.; Miao, C.; Li, Q.; Zhang, Z.; Xiao, P.; Su, L.; Yu, K.; Chen, X.; Zhang, F.; et al. RET somatic mutations are underrecognized in Hirschsprung disease. Genetics in Medicine 2018, 20, 770–777, doi:10.1038/gim.2017.178.
  37. Lin, L.F.; Doherty, D.H.; Lile, J.D.; Bektesh, S.; Collins, F. GDNF: a glial cell line-derived neurotrophic factor for midbrain dopaminergic neurons. Science 1993, 260, 1130–1132, doi:10.1126/science.8493557.
  38. Durbec, P.; Marcos-Gutierrez, C.V.; Kilkenny, C.; Grigoriou, M.; Wartiowaara, K.; Suvanto, P.; Smith, D.; Ponder, B.; Costantini, F.; Saarma, M.; et al. GDNF signalling through the Ret receptor tyrosine kinase. Nature 1996, 381, 789–793, doi:10.1038/381789a0.
  39. Schuchardt, A.; D’Agati, V.; Larsson-Blomberg, L.; Costantini, F.; Pachnis, V. Defects in the kidney and enteric nervous system of mice lacking the tyrosine kinase receptor Ret. Nature 1994, 367, 380–383, doi:10.1038/367380a0.
  40. Marcos, C.; Pachnis, V. The effect of the ret- mutation on the normal development of the central and parasympathetic nervous systems. Int. J. Dev. Biol. 1996, 40(Suppl 1), 137S-138S.Marcos, C.; Pachnis, V. The effect of the ret- mutation on the normal development of the central and parasympathetic nervous systems. Int. J. Dev. Biol. 1996, Suppl 1, 137S-138S.
  41. Jain, S.; Golden, J.P.; Wozniak, D.; Pehek, E.; Johnson, E.M.; Milbrandt, J. RET Is Dispensable for Maintenance of Midbrain Dopaminergic Neurons in Adult Mice. J Neurosci 2006, 26, 11230–11238, doi:10.1523/JNEUROSCI.1876-06.2006.
  42. Kramer, E.R.; Aron, L.; Ramakers, G.M.J.; Seitz, S.; Zhuang, X.; Beyer, K.; Smidt, M.P.; Klein, R. Absence of Ret Signaling in Mice Causes Progressive and Late Degeneration of the Nigrostriatal System. PLOS Biology 2007, 5, e39, doi:10.1371/journal.pbio.0050039.
  43. Mijatovic, J.; Patrikainen, O.; Yavich, L.; Airavaara, M.; Ahtee, L.; Saarma, M.; Piepponen, T.P. Characterization of the striatal dopaminergic neurotransmission in MEN2B mice with elevated cerebral tissue dopamine. Journal of Neurochemistry 2008, 105, 1716–1725, doi:10.1111/j.1471-4159.2008.05265.x.
  44. Mahato, A.K.; Kopra, J.; Renko, J.-M.; Visnapuu, T.; Korhonen, I.; Pulkkinen, N.; Bespalov, M.M.; Domanskyi, A.; Ronken, E.; Piepponen, T.P.; et al. Glial cell line-derived neurotrophic factor receptor Rearranged during transfection agonist supports dopamine neurons in Vitro and enhances dopamine release In Vivo. Mov. Disord. 2020, 35, 245–255, doi:10.1002/mds.27943.
  45. Mahato, A.K.; Renko, J.-M.; Kopra, J.; Visnapuu, T.; Korhonen, I.; Pulkkinen, N.; Bespalov, M.; Ronken, E.; Piepponen, T.P.; Voutilainen, M.; et al. GDNF receptor agonist supports dopamine neurons in vitro and protects their function in animal model of Parkinson’s; Neuroscience, 2019;
  46. Chmielarz, P.; Er, Ş.; Konovalova, J.; Bandrés, L.; Hlushchuk, I.; Albert, K.; Panhelainen, A.; Luk, K.; Airavaara, M.; Domanskyi, A. GDNF/RET signaling pathway activation eliminates Lewy Body pathology in midbrain dopamine neurons. bioRxiv 2019, 752899, doi:10.1101/752899.
  47. Dauer, W.; Przedborski, S. Parkinson’s Disease: Mechanisms and Models. Neuron 2003, 39, 889–909, doi:10.1016/S0896-6273(03)00568-3.
  48. Horger, B.A.; Nishimura, M.C.; Armanini, M.P.; Wang, L.-C.; Poulsen, K.T.; Rosenblad, C.; Kirik, D.; Moffat, B.; Simmons, L.; Johnson, E.; et al. Neurturin Exerts Potent Actions on Survival and Function of Midbrain Dopaminergic Neurons. J. Neurosci. 1998, 18, 4929–4937, doi:10.1523/JNEUROSCI.18-13-04929.1998.
  49. Zihlmann, K.B.; Ducray, A.D.; Schaller, B.; Huber, A.W.; Krebs, S.H.; Andres, R.H.; Seiler, R.W.; Meyer, M.; Widmer, H.R. The GDNF family members neurturin, artemin and persephin promote the morphological differentiation of cultured ventral mesencephalic dopaminergic neurons. Brain Res. Bull. 2005, 68, 42–53, doi:10.1016/j.brainresbull.2004.10.012.
  50. Hoffer, B.J.; Hoffman, A.; Bowenkamp, K.; Huettl, P.; Hudson, J.; Martin, D.; Lin, L.F.; Gerhardt, G.A. Glial cell line-derived neurotrophic factor reverses toxin-induced injury to midbrain dopaminergic neurons in vivo. Neurosci. Lett. 1994, 182, 107–111, doi:10.1016/0304-3940(94)90218-6.
  51. Bowenkamp, K.E.; Hoffman, A.F.; Gerhardt, G.A.; Henry, M.A.; Biddle, P.T.; Hoffer, B.J.; Granholm, A.-C.E. Glial cell line-derived neurotrophic factor supports survival of injured midbrain dopaminergic neurons. Journal of Comparative Neurology 1995, 355, 479–489, doi:10.1002/cne.903550402.
  52. Beck, K.D.; Valverde, J.; Alexi, T.; Poulsen, K.; Moffat, B.; Vandlen, R.A.; Rosenthal, A.; Hefti, F. Mesencephalic dopaminergic neurons protected by GDNF from axotomy-induced degeneration in the adult brain. Nature 1995, 373, 339–341, doi:10.1038/373339a0.
  53. Gash, D.M.; Zhang, Z.; Ovadia, A.; Cass, W.A.; Yi, A.; Simmerman, L.; Russell, D.; Martin, D.; Lapchak, P.A.; Collins, F.; et al. Functional recovery in parkinsonian monkeys treated with GDNF. Nature 1996, 380, 252–255, doi:10.1038/380252a0.
  54. Kirik, D.; Rosenblad, C.; Bjorklund, A.; Mandel, R.J. Long-term rAAV-mediated gene transfer of GDNF in the rat Parkinson’s model: intrastriatal but not intranigral transduction promotes functional regeneration in the lesioned nigrostriatal system. J. Neurosci. 2000, 20, 4686–4700.
  55. Kordower, J.H.; Emborg, M.E.; Bloch, J.; Ma, S.Y.; Chu, Y.; Leventhal, L.; McBride, J.; Chen, E.Y.; Palfi, S.; Roitberg, B.Z.; et al. Neurodegeneration prevented by lentiviral vector delivery of GDNF in primate models of Parkinson’s disease. Science 2000, 290, 767–773, doi:10.1126/science.290.5492.767.
  56. Oiwa, Y.; Yoshimura, R.; Nakai, K.; Itakura, T. Dopaminergic neuroprotection and regeneration by neurturin assessed by using behavioral, biochemical and histochemical measurements in a model of progressive Parkinson’s disease. Brain Res. 2002, 947, 271–283, doi:10.1016/s0006-8993(02)02934-7.
  57. Runeberg-Roos, P.; Piccinini, E.; Penttinen, A.-M.; Mätlik, K.; Heikkinen, H.; Kuure, S.; Bespalov, M.M.; Peränen, J.; Garea-Rodríguez, E.; Fuchs, E.; et al. Developing therapeutically more efficient Neurturin variants for treatment of Parkinson’s disease. Neurobiol. Dis. 2016, 96, 335–345, doi:10.1016/j.nbd.2016.07.008.
  58. Gill, S.S.; Patel, N.K.; Hotton, G.R.; O’Sullivan, K.; McCarter, R.; Bunnage, M.; Brooks, D.J.; Svendsen, C.N.; Heywood, P. Direct brain infusion of glial cell line-derived neurotrophic factor in Parkinson disease. Nat. Med. 2003, 9, 589–595, doi:10.1038/nm850.
  59. Slevin, J.T.; Gerhardt, G.A.; Smith, C.D.; Gash, D.M.; Kryscio, R.; Young, B. Improvement of bilateral motor functions in patients with Parkinson disease through the unilateral intraputaminal infusion of glial cell line-derived neurotrophic factor. J. Neurosurg. 2005, 102, 216–222, doi:10.3171/jns.2005.102.2.0216.
  60. Marks, W.J.; Ostrem, J.L.; Verhagen, L.; Starr, P.A.; Larson, P.S.; Bakay, R.A.; Taylor, R.; Cahn-Weiner, D.A.; Stoessl, A.J.; Olanow, C.W.; et al. Safety and tolerability of intraputaminal delivery of CERE-120 (adeno-associated virus serotype 2-neurturin) to patients with idiopathic Parkinson’s disease: an open-label, phase I trial. Lancet Neurol 2008, 7, 400–408, doi:10.1016/S1474-4422(08)70065-6.
  61. Lang, A.E.; Gill, S.; Patel, N.K.; Lozano, A.; Nutt, J.G.; Penn, R.; Brooks, D.J.; Hotton, G.; Moro, E.; Heywood, P.; et al. Randomized controlled trial of intraputamenal glial cell line-derived neurotrophic factor infusion in Parkinson disease. Ann. Neurol. 2006, 59, 459–466, doi:10.1002/ana.20737.
  62. Marks, W.J.; Bartus, R.T.; Siffert, J.; Davis, C.S.; Lozano, A.; Boulis, N.; Vitek, J.; Stacy, M.; Turner, D.; Verhagen, L.; et al. Gene delivery of AAV2-neurturin for Parkinson’s disease: a double-blind, randomised, controlled trial. Lancet Neurol 2010, 9, 1164–1172, doi:10.1016/S1474-4422(10)70254-4.
  63. Whone, A.L.; Boca, M.; Luz, M.; Woolley, M.; Mooney, L.; Dharia, S.; Broadfoot, J.; Cronin, D.; Schroers, C.; Barua, N.U.; et al. Extended Treatment with Glial Cell Line-Derived Neurotrophic Factor in Parkinson’s Disease. J Parkinsons Dis 2019, 9, 301–313, doi:10.3233/JPD-191576.
  64. Whone, A.; Luz, M.; Boca, M.; Woolley, M.; Mooney, L.; Dharia, S.; Broadfoot, J.; Cronin, D.; Schroers, C.; Barua, N.U.; et al. Randomized trial of intermittent intraputamenal glial cell line-derived neurotrophic factor in Parkinson’s disease. Brain 2019, 142, 512–525, doi:10.1093/brain/awz023.
  65. Bartus, R.T.; Johnson, E.M. Clinical tests of neurotrophic factors for human neurodegenerative diseases, part 1: Where have we been and what have we learned? Neurobiol. Dis. 2017, 97, 156–168, doi:10.1016/j.nbd.2016.03.027.
  66. Mahato, A.K.; Sidorova, Y.A. Glial cell line-derived neurotrophic factors (GFLs) and small molecules targeting RET receptor for the treatment of pain and Parkinson’s disease. Cell Tissue Res 2020, doi:10.1007/s00441-020-03227-4.
  67. Datta, R.; Waheed, A.; Bonapace, G.; Shah, G.N.; Sly, W.S. Pathogenesis of retinitis pigmentosa associated with apoptosis-inducing mutations in carbonic anhydrase IV. Proc Natl Acad Sci U S A 2009, 106, 3437–3442, doi:10.1073/pnas.0813178106.
  68. Schwartz, S.G.; Wang, X.; Chavis, P.; Kuriyan, A.E.; Abariga, S.A. Vitamin A and fish oils for preventing the progression of retinitis pigmentosa. Cochrane Database Syst Rev 2020, 6, CD008428, doi:10.1002/14651858.CD008428.pub3.
  69. Muhammad Imran Qadir, S. Retinitis Pigmentosa: Pathophysiology and its Management. Fortune Journal of Health Sciences 2018, 1, 19–25.
  70. Ferrari, S.; Di Iorio, E.; Barbaro, V.; Ponzin, D.; Sorrentino, F.S.; Parmeggiani, F. Retinitis Pigmentosa: Genes and Disease Mechanisms. Curr Genomics 2011, 12, 238–249, doi:10.2174/138920211795860107.
  71. Dalkara, D.; Kolstad, K.D.; Guerin, K.I.; Hoffmann, N.V.; Visel, M.; Klimczak, R.R.; Schaffer, D.V.; Flannery, J.G. AAV Mediated GDNF Secretion From Retinal Glia Slows Down Retinal Degeneration in a Rat Model of Retinitis Pigmentosa. Mol Ther 2011, 19, 1602–1608, doi:10.1038/mt.2011.62.
  72. McGee Sanftner, L.H.; Abel, H.; Hauswirth, W.W.; Flannery, J.G. Glial cell line derived neurotrophic factor delays photoreceptor degeneration in a transgenic rat model of retinitis pigmentosa. Mol. Ther. 2001, 4, 622–629, doi:10.1006/mthe.2001.0498.
  73. Allocca, M.; Vicino, U.D.; Petrillo, M.; Carlomagno, F.; Domenici, L.; Auricchio, A. Constitutive and AP20187-Induced Ret Activation in Photoreceptors Does Not Protect from Light-Induced Damage. Invest. Ophthalmol. Vis. Sci. 2007, 48, 5199–5206, doi:10.1167/iovs.07-0140.Flachsbarth, K.; Jankowiak, W.; Kruszewski, K.; Helbing, S.; Bartsch, S.; Bartsch, U. Pronounced synergistic neuroprotective effect of GDNF and CNTF on axotomized retinal ganglion cells in the adult mouse. Exp. Eye Res. 2018, 176, 258–265, doi:10.1016/j.exer.2018.09.006.
  74. Jmaeff, S.; Sidorova, Y.; Lippiatt, H.; Barcelona, P.F.; Nedev, H.; Saragovi, L.M.; Hancock, M.A.; Saarma, M.; Saragovi, H.U. Small-Molecule Ligands that Bind the RET Receptor Activate Neuroprotective Signals Independent of but Modulated by Coreceptor GFRα1. Mol. Pharmacol. 2020, 98, 1–12, doi:10.1124/mol.119.118950.Allocca, M.; Vicino, U.D.; Petrillo, M.; Carlomagno, F.; Domenici, L.; Auricchio, A. Constitutive and AP20187-Induced Ret Activation in Photoreceptors Does Not Protect from Light-Induced Damage. Invest. Ophthalmol. Vis. Sci. 2007, 48, 5199–5206, doi:10.1167/iovs.07-0140.
  75. Flachsbarth, K.; Jankowiak, W.; Kruszewski, K.; Helbing, S.; Bartsch, S.; Bartsch, U. Pronounced synergistic neuroprotective effect of GDNF and CNTF on axotomized retinal ganglion cells in the adult mouse. Exp. Eye Res. 2018, 176, 258–265, doi:10.1016/j.exer.2018.09.006.Jmaeff, S.; Sidorova, Y.; Lippiatt, H.; Barcelona, P.F.; Nedev, H.; Saragovi, L.M.; Hancock, M.A.; Saarma, M.; Saragovi, H.U. Small-Molecule Ligands that Bind the RET Receptor Activate Neuroprotective Signals Independent of but Modulated by Coreceptor GFRα1. Mol. Pharmacol. 2020, 98, 1–12, doi:10.1124/mol.119.118950.
  76. Fu, S.; Dong, S.; Zhu, M.; Sherry, D.M.; Wang, C.; You, Z.; Haigh, J.J.; Le, Y.-Z. Müller Glia Are a Major Cellular Source of Survival Signals for Retinal Neurons in Diabetes. Diabetes 2015, 64, 3554–3563, doi:10.2337/db15-0180.
  77. Xu, B.; Zhang, H.; Zhu, M.; Le, Y.-Z. Critical Role of Trophic Factors in Protecting Müller Glia: Implications to Neuroprotection in Age-Related Macular Degeneration, Diabetic Retinopathy, and Anti-VEGF Therapies. Adv. Exp. Med. Biol. 2019, 1185, 469–473, doi:10.1007/978-3-030-27378-1_77.
  78. Wu, L.; Xiao, P.; Li, Q.; Zhang, Z.; Wang, H.; Jiang, Q.; Li, L. Altered expression of AKT1 and P38A in the colons of patients with Hirschsprung’s disease. Pediatr Surg Int 2020, 36, 719–725, doi:10.1007/s00383-020-04653-9.
  79. De Falco, V.; Carlomagno, F.; Li, H.; Santoro, M. The molecular basis for RET tyrosine-kinase inhibitors in thyroid cancer. Best Practice & Research Clinical Endocrinology & Metabolism 2017, 31, 307–318, doi:10.1016/j.beem.2017.04.013.
  80. Tomuschat, C.; Puri, P. RET gene is a major risk factor for Hirschsprung’s disease: a meta-analysis. Pediatr. Surg. Int. 2015, 31, 701–710, doi:10.1007/s00383-015-3731-y.
  81. Porokuokka, L.L.; Virtanen, H.T.; Lindén, J.; Sidorova, Y.; Danilova, T.; Lindahl, M.; Saarma, M.; Andressoo, J.-O. Gfra1 Underexpression Causes Hirschsprung’s Disease and Associated Enterocolitis in Mice. Cellular and Molecular Gastroenterology and Hepatology 2019, 7, 655–678, doi:10.1016/j.jcmgh.2018.12.007.
  82. Lui, V.C.H.; Samy, E.T.; Sham, M.H.; Mulligan, L.M.; Tam, P.K.H. Glial Cell Line–Derived Neurotrophic Factor Family Receptors Are Abnormally Expressed in Aganglionic Bowel of a Subpopulation of Patients with Hirschsprung’s Disease. Laboratory Investigation 2002, 82, 703–712, doi:10.1097/01.LAB.0000017364.13014.AE.
  83. - IASP Available online: https://www.iasp-pain.org/Advocacy/GYAP.aspx?ItemNumber=5054 (accessed on Aug 12, 2020).
  84. Yawn, B.P.; Wollan, P.C.; Weingarten, T.N.; Watson, J.C.; Hooten, W.M.; Melton, L.J. The prevalence of neuropathic pain: Clinical evaluation compared with screening tools in a community population. Pain Med 2009, 10, 586–593, doi:10.1111/j.1526-4637.2009.00588.x.
  85. Schaefer, F.; van de Walle, J.; Zurowska, A.; Gimpel, C.; van Hoeck, K.; Drozdz, D.; Montini, G.; Bagdasorova, I.V.; Sorof, J.; Sugg, J.; et al. Efficacy, safety and pharmacokinetics of candesartan cilexetil in hypertensive children from 1 to less than 6 years of age. J. Hypertens. 2010, 28, 1083–1090, doi:10.1097/HJH.0b013e328336b86b.Finnerup, N.B.; Attal, N.; Haroutounian, S.; McNicol, E.; Baron, R.; Dworkin, R.H.; Gilron, I.; Haanpaa, M.; Hansson, P.; Jensen, T.S.; et al. Pharmacotherapy for neuropathic pain in adults: systematic review, meta-analysis and updated NeuPSIG recommendations. Lancet Neurol 2015, 14, 162–173, doi:10.1016/S1474-4422(14)70251-0.
  86. Ossipov, M.H. Growth factors and neuropathic pain. Curr Pain Headache Rep 2011, 15, 185–192, doi:10.1007/s11916-011-0183-5.Schaefer, F.; van de Walle, J.; Zurowska, A.; Gimpel, C.; van Hoeck, K.; Drozdz, D.; Montini, G.; Bagdasorova, I.V.; Sorof, J.; Sugg, J.; et al. Efficacy, safety and pharmacokinetics of candesartan cilexetil in hypertensive children from 1 to less than 6 years of age. J. Hypertens. 2010, 28, 1083–1090, doi:10.1097/HJH.0b013e328336b86b.
  87. Blomqvist, K.J.; Viisanen, H.; Ahlström, F.H.G.; Jokinen, V.; Sidorova, Y.A.; Suleymanova, I.; Rauhala, P.V.; Kalso, E.A.; Lilius, T.O. Morphine-3-glucuronide causes antinociceptive cross-tolerance to morphine and increases spinal substance P expression. Eur. J. Pharmacol. 2020, 875, 173021, doi:10.1016/j.ejphar.2020.173021.Ossipov, M.H. Growth factors and neuropathic pain. Curr Pain Headache Rep 2011, 15, 185–192, doi:10.1007/s11916-011-0183-5.
  88. Finnerup, N.B.; Attal, N.; Haroutounian, S.; McNicol, E.; Baron, R.; Dworkin, R.H.; Gilron, I.; Haanpaa, M.; Hansson, P.; Jensen, T.S.; et al. Pharmacotherapy for neuropathic pain in adults: systematic review, meta-analysis and updated NeuPSIG recommendations. Lancet Neurol 2015, 14, 162–173, doi:10.1016/S1474-4422(14)70251-0.Blomqvist, K.J.; Viisanen, H.; Ahlström, F.H.G.; Jokinen, V.; Sidorova, Y.A.; Suleymanova, I.; Rauhala, P.V.; Kalso, E.A.; Lilius, T.O. Morphine-3-glucuronide causes antinociceptive cross-tolerance to morphine and increases spinal substance P expression. Eur. J. Pharmacol. 2020, 875, 173021, doi:10.1016/j.ejphar.2020.173021.
  89. Bennett, D.L.; Boucher, T.J.; Armanini, M.P.; Poulsen, K.T.; Michael, G.J.; Priestley, J.V.; Phillips, H.S.; McMahon, S.B.; Shelton, D.L. The glial cell line-derived neurotrophic factor family receptor components are differentially regulated within sensory neurons after nerve injury. J. Neurosci. 2000, 20, 427–437.
  90. Josephson, A.; Widenfalk, J.; Trifunovski, A.; Widmer, H.R.; Olson, L.; Spenger, C. GDNF and NGF family members and receptors in human fetal and adult spinal cord and dorsal root ganglia. J. Comp. Neurol. 2001, 440, 204–217, doi:10.1002/cne.1380.
  91. Gardell, L.R.; Wang, R.; Ehrenfels, C.; Ossipov, M.H.; Rossomando, A.J.; Miller, S.; Buckley, C.; Cai, A.K.; Tse, A.; Foley, S.F.; et al. Multiple actions of systemic artemin in experimental neuropathy. Nat. Med. 2003, 9, 1383–1389, doi:10.1038/nm944.
  92. Wang, R.; Rossomando, A.; Sah, D.W.Y.; Ossipov, M.H.; King, T.; Porreca, F. Artemin induced functional recovery and reinnervation after partial nerve injury. Pain 2014, 155, 476–484, doi:10.1016/j.pain.2013.11.007.
  93. Boucher, T.J.; Okuse, K.; Bennett, D.L.; Munson, J.B.; Wood, J.N.; McMahon, S.B. Potent analgesic effects of GDNF in neuropathic pain states. Science 2000, 290, 124–127, doi:10.1126/science.290.5489.124.
  94. Backonja, M.; Williams, L.; Miao, X.; Katz, N.; Chen, C. Safety and efficacy of neublastin in painful lumbosacral radiculopathy: a randomized, double-blinded, placebo-controlled phase 2 trial using Bayesian adaptive design (the SPRINT trial). Pain 2017, 158, 1802–1812, doi:10.1097/j.pain.0000000000000983.
  95. Rolan, P.E.; O’Neill, G.; Versage, E.; Rana, J.; Tang, Y.; Galluppi, G.; Aycardi, E. First-In-Human, Double-Blind, Placebo-Controlled, Randomized, Dose-Escalation Study of BG00010, a Glial Cell Line-Derived Neurotrophic Factor Family Member, in Subjects with Unilateral Sciatica. PLoS ONE 2015, 10, e0125034, doi:10.1371/journal.pone.0125034.
  96. Lippoldt, E.K.; Elmes, R.R.; McCoy, D.D.; Knowlton, W.M.; McKemy, D.D. Artemin, a glial cell line-derived neurotrophic factor family member, induces TRPM8-dependent cold pain. J. Neurosci. 2013, 33, 12543–12552, doi:10.1523/JNEUROSCI.5765-12.2013.
  97. Lippoldt, E.K.; Ongun, S.; Kusaka, G.K.; McKemy, D.D. Inflammatory and neuropathic cold allodynia are selectively mediated by the neurotrophic factor receptor GFRα3. Proc. Natl. Acad. Sci. U.S.A. 2016, 113, 4506–4511, doi:10.1073/pnas.1603294113.
  98. Usoskin, D.; Furlan, A.; Islam, S.; Abdo, H.; Lönnerberg, P.; Lou, D.; Hjerling-Leffler, J.; Haeggström, J.; Kharchenko, O.; Kharchenko, P.V.; et al. Unbiased classification of sensory neuron types by large-scale single-cell RNA sequencing. Nature Neuroscience 2015, 18, 145–153, doi:10.1038/nn.3881.
  99. Clemmensen, C.; Müller, T.D.; Woods, S.C.; Berthoud, H.-R.; Seeley, R.J.; Tschöp, M.H. Gut-Brain Cross-Talk in Metabolic Control. Cell 2017, 168, 758–774, doi:10.1016/j.cell.2017.01.025.
  100. Wang, A.; Huen, S.C.; Luan, H.H.; Yu, S.; Zhang, C.; Gallezot, J.-D.; Booth, C.J.; Medzhitov, R. Opposing Effects of Fasting Metabolism on Tissue Tolerance in Bacterial and Viral Inflammation. Cell 2016, 166, 1512-1525.e12, doi:10.1016/j.cell.2016.07.026.
  101. Quartu, M.; Serra, M.P.; Boi, M.; Ferretti, M.T.; Lai, M.L.; Del Fiacco, M. Tissue distribution of Ret, GFRalpha-1, GFRalpha-2 and GFRalpha-3 receptors in the human brainstem at fetal, neonatal and adult age. Brain Research 2007, 1173, 36–52, doi:10.1016/j.brainres.2007.07.064.
  102. Mulligan, L.M. GDNF and the RET Receptor in Cancer: New Insights and Therapeutic Potential. Front. Physiol. 2019, 9, doi:10.3389/fphys.2018.01873.
  103. Santoro, M.; Carlomagno, F. Central Role of RET in Thyroid Cancer. Cold Spring Harb Perspect Biol 2013, 5, doi:10.1101/cshperspect.a009233.
  104. Eng, C. Multiple Endocrine Neoplasia Type 2. In GeneReviews®; Adam, M.P., Ardinger, H.H., Pagon, R.A., Wallace, S.E., Bean, L.J., Stephens, K., Amemiya, A., Eds.; University of Washington, Seattle: Seattle (WA), 1993.
  105. Runeberg-Roos, P.; Virtanen, H.; Saarma, M. RET(MEN 2B) is active in the endoplasmic reticulum before reaching the cell surface. Oncogene 2007, 26, 7909–7915, doi:10.1038/sj.onc.1210591.
  106. Regad, T. Targeting RTK Signaling Pathways in Cancer. Cancers (Basel) 2015, 7, 1758–1784, doi:10.3390/cancers7030860.
  107. Sangwan, V.; Park, M. Receptor tyrosine kinases: role in cancer progression. Curr Oncol 2006, 13, 191–193.
  108. Nikiforov, Y.E. RET/PTC rearrangement in thyroid tumors. Endocr. Pathol. 2002, 13, 3–16, doi:10.1385/ep:13:1:03.
  109. Thomas, G.A.; Bunnell, H.; Cook, H.A.; Williams, E.D.; Nerovnya, A.; Cherstvoy, E.D.; Tronko, N.D.; Bogdanova, T.I.; Chiappetta, G.; Viglietto, G.; et al. High Prevalence of RET/PTC Rearrangements in Ukrainian and Belarussian Post-Chernobyl Thyroid Papillary Carcinomas: A Strong Correlation between RET/PTC3 and the Solid-Follicular Variant. J Clin Endocrinol Metab 1999, 84, 4232–4238, doi:10.1210/jcem.84.11.6129.
  110. Kohno, T.; Ichikawa, H.; Totoki, Y.; Yasuda, K.; Hiramoto, M.; Nammo, T.; Sakamoto, H.; Tsuta, K.; Furuta, K.; Shimada, Y.; et al. KIF5B-RET fusions in lung adenocarcinoma. Nat Med 2012, 18, 375–377, doi:10.1038/nm.2644.
  111. Lipson, D.; Capelletti, M.; Yelensky, R.; Otto, G.; Parker, A.; Jarosz, M.; Curran, J.A.; Balasubramanian, S.; Bloom, T.; Brennan, K.W.; et al. Identification of new ALK and RET gene fusions from colorectal and lung cancer biopsies. Nat Med 2012, 18, 382–384, doi:10.1038/nm.2673.
  112. Ju, Y.S.; Lee, W.-C.; Shin, J.-Y.; Lee, S.; Bleazard, T.; Won, J.-K.; Kim, Y.T.; Kim, J.-I.; Kang, J.-H.; Seo, J.-S. A transforming KIF5B and RET gene fusion in lung adenocarcinoma revealed from whole-genome and transcriptome sequencing. Genome Res 2012, 22, 436–445, doi:10.1101/gr.133645.111.
  113. Meka, D.P.; Müller-Rischart, A.K.; Nidadavolu, P.; Mohammadi, B.; Motori, E.; Ponna, S.K.; Aboutalebi, H.; Bassal, M.; Annamneedi, A.; Finckh, B.; et al. Parkin cooperates with GDNF/RET signaling to prevent dopaminergic neuron degeneration. J Clin Invest 2015, 125, 1873–1885, doi:10.1172/JCI79300.
  114. Wang, C.; Mayer, J.A.; Mazumdar, A.; Brown, P.H. The rearranged during transfection/papillary thyroid carcinoma tyrosine kinase is an estrogen-dependent gene required for the growth of estrogen receptor positive breast cancer cells. Breast Cancer Res Treat 2012, 133, 487–500, doi:10.1007/s10549-011-1775-9.
  115. Yadav, L.; Pietilä, E.; Öhman, T.; Liu, X.; Mahato, A.K.; Sidorova, Y.; Lehti, K.; Saarma, M.; Varjosalo, M. PTPRA Phosphatase Regulates GDNF-Dependent RET Signaling and Inhibits the RET Mutant MEN2A Oncogenic Potential. iScience 2020, 23, doi:10.1016/j.isci.2020.100871.
  116. Fritsche-Guenther, R.; Witzel, F.; Sieber, A.; Herr, R.; Schmidt, N.; Braun, S.; Brummer, T.; Sers, C.; Blüthgen, N. Strong negative feedback from Erk to Raf confers robustness to MAPK signalling. Mol Syst Biol 2011, 7, 489, doi:10.1038/msb.2011.27.
  117. Lake, D.; Corrêa, S.A.L.; Müller, J. Negative feedback regulation of the ERK1/2 MAPK pathway. Cell Mol Life Sci 2016, 73, 4397–4413, doi:10.1007/s00018-016-2297-8.
  118. Arkun, Y.; Yasemi, M. Dynamics and control of the ERK signaling pathway: Sensitivity, bistability, and oscillations. PLOS ONE 2018, 13, e0195513, doi:10.1371/journal.pone.0195513.
  119. Tansey, M.G.; Baloh, R.H.; Milbrandt, J.; Johnson, E.M. GFRα-Mediated Localization of RET to Lipid Rafts Is Required for Effective Downstream Signaling, Differentiation, and Neuronal Survival. Neuron 2000, 25, 611–623, doi:10.1016/S0896-6273(00)81064-8.
  120. Gil, Z.; Cavel, O.; Kelly, K.; Brader, P.; Rein, A.; Gao, S.P.; Carlson, D.L.; Shah, J.P.; Fong, Y.; Wong, R.J. Paracrine Regulation of Pancreatic Cancer Cell Invasion by Peripheral Nerves. J Natl Cancer Inst 2010, 102, 107–118, doi:10.1093/jnci/djp456.
  121. 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. Biochimica et Biophysica Acta (BBA) - Reviews on Cancer 2012, 1826, 112–120, doi:10.1016/j.bbcan.2012.03.010.
  122. Ban, K.; Feng, S.; Shao, L.; Ittmann, M. RET signaling in prostate cancer. Clin Cancer Res 2017, 23, 4885–4896, doi:10.1158/1078-0432.CCR-17-0528.
  123. Zeng, Q.; Cheng, Y.; Zhu, Q.; Yu, Z.; Wu, X.; Huang, K.; Zhou, M.; Han, S.; Zhang, Q. The Relationship between Over-expression of Glial Cell-derived Neurotrophic Factor and Its RET Receptor with Progression and Prognosis of Human Pancreatic Cancer: Journal of International Medical Research 2008, doi:10.1177/147323000803600406.
  124. Plaza-Menacho, I.; Morandi, A.; Robertson, D.; Pancholi, S.; Drury, S.; Dowsett, M.; Martin, L.-A.; Isacke, C.M. Targeting the receptor tyrosine kinase RET sensitizes breast cancer cells to tamoxifen treatment and reveals a role for RET in endocrine resistance. Oncogene 2010, 29, 4648–4657, doi:10.1038/onc.2010.209.
  125. Morandi, A.; Martin, L.-A.; Gao, Q.; Pancholi, S.; Mackay, A.; Robertson, D.; Zvelebil, M.; Dowsett, M.; Plaza-Menacho, I.; Isacke, C.M. GDNF–RET Signaling in ER-Positive Breast Cancers Is a Key Determinant of Response and Resistance to Aromatase Inhibitors. Cancer Res 2013, 73, 3783–3795, doi:10.1158/0008-5472.CAN-12-4265.
  126. Gattelli, A.; Nalvarte, I.; Boulay, A.; Roloff, T.C.; Schreiber, M.; Carragher, N.; Macleod, K.K.; Schlederer, M.; Lienhard, S.; Kenner, L.; et al. Ret inhibition decreases growth and metastatic potential of estrogen receptor positive breast cancer cells. EMBO Mol Med 2013, 5, 1335–1350, doi:10.1002/emmm.201302625.
  127. White, U.A.; Stephens, J.M. The gp130 Receptor Cytokine Family: Regulators of Adipocyte Development and Function. Curr Pharm Des 2011, 17, 340–346.
  128. Boulay, A.; Breuleux, M.; Stephan, C.; Fux, C.; Brisken, C.; Fiche, M.; Wartmann, M.; Stumm, M.; Lane, H.A.; Hynes, N.E. The Ret Receptor Tyrosine Kinase Pathway Functionally Interacts with the ERα Pathway in Breast Cancer. Cancer Res 2008, 68, 3743–3751, doi:10.1158/0008-5472.CAN-07-5100.
  129. Hidalgo-Figueroa, M.; Bonilla, S.; Gutiérrez, F.; Pascual, A.; López-Barneo, J. GDNF Is Predominantly Expressed in the PV+ Neostriatal Interneuronal Ensemble in Normal Mouse and after Injury of the Nigrostriatal Pathway. J. Neurosci. 2012, 32, 864–872, doi:10.1523/JNEUROSCI.2693-11.2012.
  130. Ibáñez, C.F.; Paratcha, G.; Ledda, F. RET-independent signaling by GDNF ligands and GFRα receptors. Cell Tissue Res 2020, doi:10.1007/s00441-020-03261-2.
  131. Paratcha, G.; Ledda, F.; Ibáñez, C.F. The neural cell adhesion molecule NCAM is an alternative signaling receptor for GDNF family ligands. Cell 2003, 113, 867–879, doi:10.1016/s0092-8674(03)00435-5.
  132. Turconi, G.; Kopra, J.; Võikar, V.; Kulesskaya, N.; Vilenius, C.; Piepponen, T.P.; Andressoo, J.-O. Chronic 2-Fold Elevation of Endogenous GDNF Levels Is Safe and Enhances Motor and Dopaminergic Function in Aged Mice. Mol Ther Methods Clin Dev 2020, 17, 831–842, doi:10.1016/j.omtm.2020.04.003.
  133. Villanueva, M.T. GDF15 tells the brain to lose weight. Nat Rev Drug Discov 2017, 16, 827–827, doi:10.1038/nrd.2017.241.
  134. Bespalov, M.M.; Sidorova, Y.A.; Tumova, S.; Ahonen-Bishopp, A.; Magalhães, A.C.; Kulesskiy, E.; Paveliev, M.; Rivera, C.; Rauvala, H.; Saarma, M. Heparan sulfate proteoglycan syndecan-3 is a novel receptor for GDNF, neurturin, and artemin. J. Cell Biol. 2011, 192, 153–169, doi:10.1083/jcb.201009136.
  135. Sidorova, Y.A.; Bespalov, M.M.; Wong, A.W.; Kambur, O.; Jokinen, V.; Lilius, T.O.; Suleymanova, I.; Karelson, G.; Rauhala, P.V.; Karelson, M.; et al. A Novel Small Molecule GDNF Receptor RET Agonist, BT13, Promotes Neurite Growth from Sensory Neurons in Vitro and Attenuates Experimental Neuropathy in the Rat. Front. Pharmacol. 2017, 8, doi:10.3389/fphar.2017.00365.
  136. Viisanen, H.; Nuotio, U.; Kambur, O.; Mahato, A.K.; Jokinen, V.; Lilius, T.; Li, W.; Santos, H.A.; Karelson, M.; Rauhala, P.; et al. Novel RET agonist for the treatment of experimental neuropathies. Mol Pain 2020, 16, 1744806920950866, doi:10.1177/1744806920950866.
  137. Ivanova, L.; Tammiku-Taul, J.; García-Sosa, A.T.; Sidorova, Y.; Saarma, M.; Karelson, M. Molecular Dynamics Simulations of the Interactions between Glial Cell Line-Derived Neurotrophic Factor Family Receptor GFRα1 and Small-Molecule Ligands. ACS Omega 2018, 3, 11407–11414, doi:10.1021/acsomega.8b01524.
  138. Amirouchene-Angelozzi, N.; Swanton, C.; Bardelli, A. Tumor Evolution as a Therapeutic Target. Cancer Discov 2017, 7, 805–817, doi:10.1158/2159-8290.CD-17-0343.
  139. Trupp, M.; Arenas, E.; Fainzilber, M.; Nilsson, A.-S.; Sieber, B.-A.; Grigoriou, M.; Kilkenny, C.; Salazar-Grueso, E.; Pachnis, V.; Arumäe, U.; et al. Functional receptor for GDNF encoded by the c- ret proto-oncogene. Nature 1996, 381, 785–789, doi:10.1038/381785a0.
  140. Cintrón-Colón, A.F.; Almeida-Alves, G.; Boynton, A.M.; Spitsbergen, J.M. GDNF synthesis, signaling, and retrograde transport in motor neurons. Cell Tissue Res 2020, doi:10.1007/s00441-020-03287-6.
  141. Ron, D.; Janak, P.H. GDNF and addiction. Rev Neurosci 2005, 16, 277–285, doi:10.1515/revneuro.2005.16.4.277.
More