Advances in the Regulation of Receptor Tyrosine Kinase on Autophagy

DONG Xue-ying,LIANG Kai,YE Ke-ying,ZHOU Ce-fan,TANG Jing-feng

China Biotechnology ›› 2021, Vol. 41 ›› Issue (5) : 72-78.

PDF(445 KB)
PDF(445 KB)
China Biotechnology ›› 2021, Vol. 41 ›› Issue (5) : 72-78. DOI: 10.13523/j.cb.2012041

Advances in the Regulation of Receptor Tyrosine Kinase on Autophagy

Author information +
History +

Abstract

Autophagy is a very conservative biological process of lysosomal degradation in eukaryotic evolution, which plays an important role in maintaining cell homeostasis and eliminating harmful components. Receptor tyrosine kinases (RTKs) are a class of kinase proteins that play an important role in the movement and invasion of normal cells and cancer cells. RTKs protein can not only promote autophagy, but also inhibit autophagy. Studies have shown that RTKs can play a regulatory role in tumors and related diseases through autophagy. For example, epidermal growth factor receptor (EGFR) can inhibit autophagy, thereby promoting tumor growth and proliferation; it can also pass RTK/Ras/ERK signaling pathways to induce autophagy, which in turn participates in related diseases such as cellular immune responses. The regulatory effects of RTKs on autophagy and related research results, which provide a basis for the theoretical basis of target targeted therapy were reviewed.

Key words

Autophagy / Receptor tyrosine kinase family / Cancer / Degenerative diseases / Resistance

Cite this article

Download Citations
Xue-ying DONG, Kai LIANG, Ke-ying YE, et al. Advances in the Regulation of Receptor Tyrosine Kinase on Autophagy[J]. China Biotechnology, 2021, 41(5): 72-78 https://doi.org/10.13523/j.cb.2012041

References

[1]
Ho C J, Gorski S M. Molecular mechanisms underlying autophagy-mediated treatment resistance in cancer. Cancers, 2019,11(11):1775.
[2]
Noda N N, Fujioka Y. Atg1 family kinases in autophagy initiation. Cellular and Molecular Life Sciences, 2015,72(16):3083-3096.
[3]
Amaya C, Fader C M, Colombo M I. Autophagy and proteins involved in vesicular trafficking. FEBS Letters, 2015,589(22):3343-3353.
[4]
Itakura E, Mizushima N. Characterization of autophagosome formation site by a hierarchical analysis of mammalian Atg proteins. Autophagy, 2010,6(6):764-776.
Autophagy is an intracellular degradation process, through which cytosolic materials are delivered to the lysosome.Despite recent identification of many autophagy-related genes, how autophagosomes are generated remains unclear.Here, we examined the hierarchical relationships among mammalian Atg proteins. Under starvation conditions, ULK1,Atg14, WIPI-1, LC3 and Atg16L1 target to the same compartment, whereas DFCP1 localizes adjacently to these Atgproteins. In terms of puncta formation, the protein complex including ULK1 and FIP200 is the most upstream unit and is required for puncta formation of the Atg14-containing PI3-kinase complex. Puncta formation of both DFCP1 and WIPI-1 requires FIP200 and Atg14. The Atg12-Atg5-Atg16L1 complex and LC3 are downstream units among these factors. The punctate structures containing upstream Atg proteins such as ULK1 and Atg14 tightly associate with the ER, where the ER protein vacuole membrane protein 1 (VMP1) also transiently localizes. These structures are formed even when cells are treated with wortmannin to suppress autophagosome formation. These hierarchical analyses suggest that ULK1, Atg14 and VMP1 localize to the ER-associated autophagosome formation sites in a PI3-kinase activity-independent manner.
[5]
Xie Z P, Nair U, Klionsky D J. Atg8 controls phagophore expansion during autophagosome formation. Molecular Biology of the Cell, 2008,19(8):3290-3298.
[6]
Reggiori F, Tooze S A. Autophagy regulation through Atg9 traffic. Journal of Cell Biology, 2012,198(2):151-153.
[7]
Lemmon M A, Schlessinger J. Cell signaling by receptor tyrosine kinases. Cell, 2010,141(7):1117-1134.
Recent structural studies of receptor tyrosine kinases (RTKs) have revealed unexpected diversity in the mechanisms of their activation by growth factor ligands. Strategies for inducing dimerization by ligand binding are surprisingly diverse, as are mechanisms that couple this event to activation of the intracellular tyrosine kinase domains. As our understanding of these details becomes increasingly sophisticated, it provides an important context for therapeutically countering the effects of pathogenic RTK mutations in cancer and other diseases. Much remains to be learned, however, about the complex signaling networks downstream from RTKs and how alterations in these networks are translated into cellular responses.
[8]
Trenker R, Jura N. Receptor tyrosine kinase activation: From the ligand perspective. Current Opinion in Cell Biology, 2020,63:174-185.
[9]
Fraser J, Cabodevilla A G, Simpson J, et al. Interplay of autophagy, receptor tyrosine kinase signalling and endocytic trafficking. Essays in Biochemistry, 2017,61(6):597-607.
Vesicular trafficking events play key roles in the compartmentalization and proper sorting of cellular components. These events have crucial roles in sensing external signals, regulating protein activities and stimulating cell growth or death decisions. Although mutations in vesicle trafficking players are not direct drivers of cellular transformation, their activities are important in facilitating oncogenic pathways. One such pathway is the sensing of external stimuli and signalling through receptor tyrosine kinases (RTKs). The regulation of RTK activity by the endocytic pathway has been extensively studied. Compelling recent studies have begun to highlight the association between autophagy and RTK signalling. The influence of this interplay on cellular status and its relevance in disease settings will be discussed here.© 2017 The Author(s).
[10]
Lampada A, Hochhauser D, Salomoni P. Autophagy and receptor tyrosine kinase signalling: a mTORC2 matter. Cell Cycle, 2017,16(20):1855-1856.
[11]
Chukkapalli S, Amessou M, Dilly A K, et al. Role of the EphB2 receptor in autophagy, apoptosis and invasion in human breast cancer cells. Experimental Cell Research, 2014,320(2):233-246.
The Eph and Ephrin proteins, which constitute the largest family of receptor tyrosine kinases, are involved in normal tissue development and cancer progression. Here, we examined the expression and role of the B-type Eph receptor EphB2 in breast cancers. By immunohistochemistry using a progression tissue microarray of human clinical samples, we found EphB2 to be expressed in benign tissues, but strongly increased in cancers particularly in invasive and metastatic carcinomas. Subsequently, we found evidence that EphB2, whose expression varies in established cell breast lines, possesses multiple functions. First, the use of a DOX-inducible system to restore EphB2 function to low expressers resulted in decreased tumor growth in vitro and in vivo, while its siRNA-mediated silencing in high expressers increased growth. This function involves the onset of apoptotic death paralleled by caspases 3 and 9 activation. Second, EphB2 was also found to induce autophagy, as assessed by immunofluorescence and/or immunoblotting examination of the LC3, ATG5 and ATG12 markers. Third, EphB2 also has a pro-invasive function in breast cancer cells that involves the regulation of MMP2 and MMP9 metalloproteases and can be blocked by treatment with respective neutralizing antibodies. Furthermore, EphB2-induced invasion is kinase-dependent and is impeded in cells expressing a kinase-dead mutant EphB2. In summary, we identified a mechanism involving a triple role for EphB2 in breast cancer progression, whereby it regulates apoptosis, autophagy, and invasion. © 2013 Published by Elsevier Inc.
[12]
Kandouz M, Haidara K, Zhao J, et al. The EphB2 tumor suppressor induces autophagic cell death via concomitant activation of the ERK1/2 and PI3K pathways. Cell Cycle, 2010,9(2):398-407.
[13]
Tanabe H, Kuribayashi K, Tsuji N, et al. Sesamin induces autophagy in colon cancer cells by reducing tyrosine phosphorylation of EphA1 and EphB2. International Journal of Oncology, 2011,39(1):33-40.
[14]
Zhong S Z, Pei D, Shi L, et al. Ephrin-B2 inhibits Aβ25-35-induced apoptosis by alleviating endoplasmic reticulum stress and promoting autophagy in HT22 cells. Neuroscience Letters, 2019,704:50-56.
[15]
Singh R, Karri D, Shen H, et al. TRAF4-mediated ubiquitination of NGF receptor TrkA regulates prostate cancer metastasis. The Journal of Clinical Investigation, 2018,128(7):3129-3143.
[16]
Molloy N H, Read D E, Gorman A M. Nerve growth factor in cancer cell death and survival. Cancers, 2011,3(1):510-530.
One of the major challenges for cancer therapeutics is the resistance of many tumor cells to induction of cell death due to pro-survival signaling in the cancer cells. Here we review the growing literature which shows that neurotrophins contribute to pro-survival signaling in many different types of cancer. In particular, nerve growth factor, the archetypal neurotrophin, has been shown to play a role in tumorigenesis over the past decade. Nerve growth factor mediates its effects through its two cognate receptors, TrkA, a receptor tyrosine kinase and p75NTR, a member of the death receptor superfamily. Depending on the tumor origin, pro-survival signaling can be mediated by TrkA receptors or by p75NTR. For example, in breast cancer the aberrant expression of nerve growth factor stimulates proliferative signaling through TrkA and pro-survival signaling through p75NTR. This latter signaling through p75NTR promotes increased resistance to the induction of cell death by chemotherapeutic treatments. In contrast, in prostate cells the p75NTR mediates cell death and prevents metastasis. In prostate cancer, expression of this receptor is lost, which contributes to tumor progression by allowing cells to survive, proliferate and metastasize. This review focuses on our current knowledge of neurotrophin signaling in cancer, with a particular emphasis on nerve growth factor regulation of cell death and survival in cancer.
[17]
Dadakhujaev S, Jung E J, Noh H S, et al. Interplay between autophagy and apoptosis in TrkA-induced cell death. Autophagy, 2009,5(1):103-105.
[18]
Dadakhujaev S, Noh H S, Jung E J, et al. The reduced catalase expression in TrkA-induced cells leads to autophagic cell death via ROS accumulation. Experimental Cell Research, 2008,314(17):3094-3106.
[19]
Koorstra J B M, Karikari C, Feldmann G, et al. The Axl receptor tyrosine kinase confers an adverse prognostic influence in pancreatic cancer and represents a new therapeutic target. Cancer Biology & Therapy, 2009,8(7):618-626.
[20]
Colavito S A. AXL as a target in breast cancer therapy. Journal of Oncology, 2020,2020:5291952.
[21]
Neubauer A, O’Bryan J P, Fiebeler A, et al. Axl, a novel receptor tyrosine kinase isolated from chronic myelogenous leukemia. Seminars in Hematology, 1993,30(3 Suppl 3):34.
[22]
Paccez J D, Vogelsang M, Parker M I, et al. The receptor tyrosine kinase Axl in cancer: biological functions and therapeutic implications. International Journal of Cancer, 2014,134(5):1024-1033.
[23]
Holland S J, Pan A, Franci C, et al. R428, a selective small molecule inhibitor of Axl kinase, blocks tumor spread and prolongs survival in models of metastatic breast cancer. Cancer Research, 2010,70(4):1544-1554.
Accumulating evidence suggests important roles for the receptor tyrosine kinase Axl in cancer progression, invasion, metastasis, drug resistance, and patient mortality, highlighting Axl as an attractive target for therapeutic development. We have generated and characterized a potent and selective small-molecule inhibitor, R428, that blocks the catalytic and procancerous activities of Axl. R428 inhibits Axl with low nanomolar activity and blocked Axl-dependent events, including Akt phosphorylation, breast cancer cell invasion, and proinflammatory cytokine production. Pharmacologic investigations revealed favorable exposure after oral administration such that R428-treated tumors displayed a dose-dependent reduction in expression of the cytokine granulocyte macrophage colony-stimulating factor and the epithelial-mesenchymal transition transcriptional regulator Snail. In support of an earlier study, R428 inhibited angiogenesis in corneal micropocket and tumor models. R428 administration reduced metastatic burden and extended survival in MDA-MB-231 intracardiac and 4T1 orthotopic (median survival, >80 days compared with 52 days; P < 0.05) mouse models of breast cancer metastasis. Additionally, R428 synergized with cisplatin to enhance suppression of liver micrometastasis. Our results show that Axl signaling regulates breast cancer metastasis at multiple levels in tumor cells and tumor stromal cells and that selective Axl blockade confers therapeutic value in prolonging survival of animals bearing metastatic tumors.
[24]
Vajkoczy P, Knyazev P, Kunkel A, et al. Dominant-negative inhibition of the Axl receptor tyrosine kinase suppresses brain tumor cell growth and invasion and prolongs survival. Proceedings of the National Academy of Sciences of the United States of America, 2006,103(15):5799-5804.
[25]
Han J, Bae J, Choi C Y, et al. Autophagy induced by AXL receptor tyrosine kinase alleviates acute liver injury via inhibition of NLRP3 inflammasome activation in mice. Autophagy, 2016,12(12):2326-2343.
[26]
Lee C H, Chun T. Anti-inflammatory role of TAM family of receptor tyrosine kinases via modulating macrophage function. Molecules and Cells, 2019,42(1):1-7.
[27]
Han W D, Pan H M, Chen Y, et al. EGFR tyrosine kinase inhibitors activate autophagy as a cytoprotective response in human lung cancer cells. PLoS One, 2011,6(6):e18691.
[28]
Wei Y J, Zou Z J, Becker N, et al. EGFR-mediated beclin 1 phosphorylation in autophagy suppression, tumor progression, and tumor chemoresistance. Cell, 2013,154(6):1269-1284.
[29]
So K S, Kim C H, Rho J K, et al. Autophagosome-mediated EGFR down-regulation induced by the CK2 inhibitor enhances the efficacy of EGFR-TKI on EGFR-mutant lung cancer cells with resistance by T790M. PLoS One, 2014,9(12):e114000.
[30]
Wang R C, Wei Y, An Z, et al. Akt-mediated regulation of autophagy and tumorigenesis through beclin 1 phosphorylation. Science, 2012,338(6109):956-959.
[31]
Kwon Y, Kim M, Jung H S, et al. Targeting autophagy for overcoming resistance to anti-EGFR treatments. Cancers, 2019,11(9):1374.
[32]
Domigan C K, Warren C M, Antanesian V, et al. Autocrine VEGF maintains endothelial survival through regulation of metabolism and autophagy. Journal of Cell Science, 2015,128(12):2236-2248.
[33]
Lee S, Chen T T, Barber C L, et al. Autocrine VEGF signaling is required for vascular homeostasis. Cell, 2007,130(4):691-703.
[34]
Warr M R, Binnewies M, Flach J, et al. FOXO3A directs a protective autophagy program in haematopoietic stem cells. Nature, 2013,494(7437):323-327.
[35]
Spengler K, Kryeziu N, Groβe S, et al. VEGF triggers transient induction of autophagy in endothelial cells via AMPKα1. Cells, 2020,9(3):687.
[36]
Chen C H, Liu Y M, Pan S L, et al. Trichlorobenzene-substituted azaaryl compounds as novel FGFR inhibitors exhibiting potent antitumor activity in bladder cancer cells in vitro and in vivo. Oncotarget, 2016,7(18):26374-26387.
[37]
Dieci M V, Arnedos M, Andre F, et al. Fibroblast growth factor receptor inhibitors as a cancer treatment: from a biologic rationale to medical perspectives. Cancer Discovery, 2013,3(3):264-279.
[38]
Du Z F, Lovly C M. Mechanisms of receptor tyrosine kinase activation in cancer. Molecular Cancer, 2018,17(1):1-13.
[39]
Lampada A, O’Prey J, Szabadkai G, et al. mTORC1-independent autophagy regulates receptor tyrosine kinase phosphorylation in colorectal cancer cells via an mTORC2-mediated mechanism. Cell Death & Differentiation, 2017,24(6):1045-1062.
[40]
Wu M, Zhang P H. EGFR-mediated autophagy in tumourigenesis and therapeutic resistance. Cancer Letters, 2020,469:207-216.
[41]
Zhou C F, Qian X H, Hu M, et al. STYK1 promotes autophagy through enhancing the assembly of autophagy-specific class III phosphatidylinositol 3-kinase complex I. Autophagy, 2020,16(10):1786-1806.
[42]
Huang Z, Ma N, Xiong Y L, et al. Aberrantly high expression of NOK/STYK1 is tightly associated with the activation of the AKT/GSK3β/N-cadherin pathway in non-small cell lung cancer. OncoTargets and Therapy, 2019,12:10299-10309.
High metastasis is a leading risk factor for the survival of non-small cell lung cancer (NSCLC) and epithelial-mesenchymal transition (EMT) is a vital step of metastasis. The expression of novel oncogene with kinase domain (NOK) has been observed in some human malignancies, including non-small cell lung cancer (NSCLC); however, the biological function of NOK in NSCLC remains unclear. In the study, we explored the function of NOK in NSCLC, with an aim to elucidate the relevant underlying mechanisms.We investigate the expression of NOK, p-Akt, p-GSK-3β, E-cadherin and N-cadherin expression by immunohistochemical analysis using tissue microarrays of 72 paired NSCLC samples of cancerous and adjacent normal tissues. The associations between NOK expression and clinicopathological factors, overall survival, other proteins were assessed. Immunofluorescence analysis of NSCLC tissues was performed to study the location of NOK, Akt and GSK-3β. Up or down-regulated of NOK were conducted in two NSCLC cell lines to analyze its impact on AKT/GSK3β pathway.Statistical analysis revealed NOK expression increased in NSCLC tissues compared with normal tissues (<0.05). It also showed that low NOK expression were associated with a higher possibility of non-lymphatic metastasis, an early pN stage and clinical stage (<0.05). Moreover, NOK expression was positively correlated with the expression of oncogene p-Akt (Thr308), p-GSK-3β (Ser9) and N-cadherin (<0.05). Immunofluorescence analysis of NSCLC tissues revealed that NOK is co-located with Akt and GSK-3β. Further study in NSCLC cell lines revealed that NOK overexpression can activate the AKT/GSK3β pathway. Conversely, knockdown of NOK can suppress the AKT/GSK3β pathway.Our results suggest that NOK overexpression correlated significantly with lymphatic metastasis, advanced pN and clinical stage in NSCLC. And NOK may promote EMT by activating the AKT/GSK3β/N-cadherin pathway in NSCLC.© 2019 Huang et al.
[43]
Meng X R, Wang H J, Zhao J Z, et al. Apatinib inhibits cell proliferation and induces autophagy in human papillary thyroid carcinoma via the PI3K/Akt/mTOR signaling pathway. Frontiers in Oncology, 2020,10:217.
[44]
Tao J Y, Sun D T, Hou H L. Role of YES1 amplification in EGFR mutation-positive non-small cell lung cancer: Primary resistance to afatinib in a patient. Thoracic Cancer, 2020,11(9):2736-2739.
[45]
Curry D W, Stutz B, Andrews Z B, et al. Targeting AMPK signaling as a neuroprotective strategy in Parkinson’s disease. Journal of Parkinson’s Disease, 2018,8(2):161-181.
[46]
Vernizzi L, Paiardi C, Licata G, et al. Glutamine synthetase 1 increases autophagy lysosomal degradation of mutant huntingtin aggregates in neurons, ameliorating motility in a Drosophila model for Huntington’s disease. Cells, 2020,9(1):196.
[47]
Forloni G, Balducci C. Alzheimer’s disease, oligomers, and inflammation. Journal of Alzheimer’s Disease, 2018,62(3):1261-1276.
[48]
Sun B L, Li W W, Zhu C, et al. Clinical research on Alzheimer’s disease: progress and perspectives. Neuroscience Bulletin, 2018,34(6):1111-1118.
[49]
Zou L, Wang Z, Shen L, et al. Receptor tyrosine kinases positively regulate BACE activity and Amyloid-beta production through enhancing BACE internalization. Cell Research, 2007,17(5):389-401.
[50]
Frisardi V, Santamato A, Cheeran B. Parkinson’s disease: new insights into pathophysiology and rehabilitative approaches. Parkinson’s Disease, 2016,2016:1-2.
[51]
Chittoor-Vinod V G, Villalobos-Cantor S, Roshak H, et al. Dietary amino acids impact LRRK2-induced neurodegeneration in Parkinson’s disease models. The Journal of Neuroscience, 2020,40(32):6234-6249.
[52]
Valionyte E, Yang Y, Roberts S L, et al. Lowering mutant huntingtin levels and toxicity: autophagy-endolysosome pathways in Huntington’s disease. Journal of Molecular Biology, 2020,432(8):2673-2691.
Huntington's disease (HD) is a monogenetic neurodegenerative disease, which serves as a model of neurodegeneration with protein aggregation. Autophagy has been suggested to possess a great value to tackle protein aggregation toxicity and neurodegenerative diseases. Current studies suggest that autophagy-endolysosomal pathways are critical for HD pathology. Here we review recent advancement in the studies of autophagy and selective autophagy relating HD. Restoration of autophagy flux and enhancement of selective removal of mutant huntingtin/disease-causing protein would be effective approaches towards tackling HD as well as other similar neurodegenerative disorders.Copyright © 2019 Elsevier Ltd. All rights reserved.

RIGHTS & PERMISSIONS

Copyright reserved © 2021. Office of China Biotechnology.
PDF(445 KB)

Accesses

Citation

Detail

Sections
Recommended

/