MFN2 suppresses clear cell renal cell carcinoma progression by modulating mitochondria-dependent dephosphorylation of EGFR
Li Luo
State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, P. R. China
These authors contributed equally.
Search for more papers by this authorDenghui Wei
State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, P. R. China
These authors contributed equally.
Search for more papers by this authorYihui Pan
Department of Urology, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, P. R. China
These authors contributed equally.
Search for more papers by this authorQiu-Xia Wang
State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, P. R. China
Search for more papers by this authorJian-Xiong Feng
State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, P. R. China
Search for more papers by this authorBing Yu
State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, P. R. China
Search for more papers by this authorTiebang Kang
State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, P. R. China
Search for more papers by this authorCorresponding Author
Junhang Luo
Department of Urology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P. R. China
Correspondence
Song Gao, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, Guangdong, P. R. China.
Email: [email protected]
Jiefeng Yang, Department of Urology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, Guangdong, P. R. China.
Email: [email protected]
Junhang Luo, Department of Urology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, Guangdong, P. R. China
Email: [email protected]
Search for more papers by this authorCorresponding Author
Jiefeng Yang
Department of Urology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P. R. China
Correspondence
Song Gao, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, Guangdong, P. R. China.
Email: [email protected]
Jiefeng Yang, Department of Urology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, Guangdong, P. R. China.
Email: [email protected]
Junhang Luo, Department of Urology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, Guangdong, P. R. China
Email: [email protected]
Search for more papers by this authorCorresponding Author
Song Gao
State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, P. R. China
Correspondence
Song Gao, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, Guangdong, P. R. China.
Email: [email protected]
Jiefeng Yang, Department of Urology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, Guangdong, P. R. China.
Email: [email protected]
Junhang Luo, Department of Urology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, Guangdong, P. R. China
Email: [email protected]
Search for more papers by this authorLi Luo
State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, P. R. China
These authors contributed equally.
Search for more papers by this authorDenghui Wei
State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, P. R. China
These authors contributed equally.
Search for more papers by this authorYihui Pan
Department of Urology, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, P. R. China
These authors contributed equally.
Search for more papers by this authorQiu-Xia Wang
State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, P. R. China
Search for more papers by this authorJian-Xiong Feng
State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, P. R. China
Search for more papers by this authorBing Yu
State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, P. R. China
Search for more papers by this authorTiebang Kang
State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, P. R. China
Search for more papers by this authorCorresponding Author
Junhang Luo
Department of Urology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P. R. China
Correspondence
Song Gao, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, Guangdong, P. R. China.
Email: [email protected]
Jiefeng Yang, Department of Urology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, Guangdong, P. R. China.
Email: [email protected]
Junhang Luo, Department of Urology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, Guangdong, P. R. China
Email: [email protected]
Search for more papers by this authorCorresponding Author
Jiefeng Yang
Department of Urology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P. R. China
Correspondence
Song Gao, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, Guangdong, P. R. China.
Email: [email protected]
Jiefeng Yang, Department of Urology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, Guangdong, P. R. China.
Email: [email protected]
Junhang Luo, Department of Urology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, Guangdong, P. R. China
Email: [email protected]
Search for more papers by this authorCorresponding Author
Song Gao
State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, P. R. China
Correspondence
Song Gao, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, Guangdong, P. R. China.
Email: [email protected]
Jiefeng Yang, Department of Urology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, Guangdong, P. R. China.
Email: [email protected]
Junhang Luo, Department of Urology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, Guangdong, P. R. China
Email: [email protected]
Search for more papers by this authorAbstract
Background
Clear cell renal cell carcinoma (ccRCC) is the most lethal renal cancer. An overwhelming increase of patients experience tumor progression and unfavorable prognosis. However, the molecular events underlying ccRCC tumorigenesis and metastasis remain unclear. Therefore, uncovering the underlying mechanisms will pave the way for developing novel therapeutic targets for ccRCC. In this study, we sought to investigate the role of mitofusin-2 (MFN2) in supressing ccRCC tumorigenesis and metastasis.
Methods
The expression pattern and clinical significance of MFN2 in ccRCC were analyzed by using the Cancer Genome Atlas datasets and samples from our independent ccRCC cohort. Both in vitro and in vivo experiments, including cell proliferation, xenograft mouse models and transgenic mouse model, were used to determine the role of MFN2 in regulating the malignant behaviors of ccRCC. RNA-sequencing, mass spectrum analysis, co-immunoprecipitation, bio-layer interferometry and immunofluorescence were employed to elucidate the molecular mechanisms for the tumor-supressing role of MFN2.
Results
we reported a tumor-suppressing pathway in ccRCC, characterized by mitochondria-dependent inactivation of epidermal growth factor receptor (EGFR) signaling. This process was mediated by the outer mitochondrial membrane (OMM) protein MFN2. MFN2 was down-regulated in ccRCC and associated with favorable prognosis of ccRCC patients. in vivo and in vitro assays demonstrated that MFN2 inhibited ccRCC tumor growth and metastasis by suppressing the EGFR signaling pathway. In a kidney-specific knockout mouse model, loss of MFN2 led to EGFR pathway activation and malignant lesions in kidney. Mechanistically, MFN2 preferably binded small GTPase Rab21 in its GTP-loading form, which was colocalized with endocytosed EGFR in ccRCC cells. Through this EGFR-Rab21-MFN2 interaction, endocytosed EGFR was docked to mitochondria and subsequently dephosphorylated by the OMM-residing tyrosine-protein phosphatase receptor type J (PTPRJ).
Conclusions
Our findings uncover an important non-canonical mitochondria-dependent pathway regulating EGFR signaling by the Rab21-MFN2-PTPRJ axis, which contributes to the development of novel therapeutic strategies for ccRCC.
CONFLICTS OF INTEREST STATEMENT
The authors declare that they have no competing interests.
Open Research
DATA AVAILABILITY STATEMENT
The raw RNA sequencing data are available at the Genome Sequence Archive (GSA) database (accession number HRA002229). The mass spectrometry proteomics data have been deposited to the ProteomeXchange Consortium via the PRIDE33 partner repository with the dataset identifiers PXD033112 for the interactome of MFN2, PXD033104 for the interactome of Rab21, and PXD033099 for the mitochondrial proteome of 786-O cells. The key raw data have been deposited into the Research Data Deposit (www.researchdata.org.cn), with the approval number of 2302230001.
Supporting Information
Filename | Description |
---|---|
cac212428-sup-0001-FigureS1.tif21 MB | Supporting Information |
cac212428-sup-0002-FigureS2.tif24.5 MB | Supporting Information |
cac212428-sup-0003-FigureS3.tif13.7 MB | Supporting Information |
cac212428-sup-0004-FigureS4.tif20.9 MB | Supporting Information |
cac212428-sup-0005-FigureS5.tif26.5 MB | Supporting Information |
cac212428-sup-0006-FigureS6.tif16.1 MB | Supporting Information |
cac212428-sup-0007-FigureS7.tif13.4 MB | Supporting Information |
cac212428-sup-0008-FigureS8.tif25.3 MB | Supporting Information |
cac212428-sup-0009-FigureS9.tif25.7 MB | Supporting Information |
cac212428-sup-0010-FigureS10.tif23.2 MB | Supporting Information |
cac212428-sup-0011-FiguresCaption.docx45 KB | Supporting Information |
Please note: The publisher is not responsible for the content or functionality of any supporting information supplied by the authors. Any queries (other than missing content) should be directed to the corresponding author for the article.
REFERENCES
- 1Siegel RL, Miller KD, Fuchs HE, Jemal A. Cancer Statistics, 2021. CA Cancer J Clin. 2021; 71(1): 7–33.
- 2Qiu H, Cao S, Xu R. Cancer incidence, mortality, and burden in China: a time-trend analysis and comparison with the United States and United Kingdom based on the global epidemiological data released in 2020. Cancer Commun (Lond). 2021; 41(10): 1037–48.
- 3Capitanio U, Montorsi F. Renal cancer. Lancet. 2016; 387(10021): 894–906.
- 4Choueiri TK, Kaelin WG, Jr. Targeting the HIF2-VEGF axis in renal cell carcinoma. Nat Med. 2020; 26(10): 1519–30.
- 5Sequist LV, Han JY, Ahn MJ, Cho BC, Yu H, Kim SW, et al. Osimertinib plus savolitinib in patients with EGFR mutation-positive, MET-amplified, non-small-cell lung cancer after progression on EGFR tyrosine kinase inhibitors: interim results from a multicentre, open-label, phase 1b study. Lancet Oncol. 2020; 21(3): 373–86.
- 6Motzer RJ, Rini BI, Mcdermott DF, Aren Frontera O, Hammers HJ, Carducci MA, et al. Nivolumab plus ipilimumab versus sunitinib in first-line treatment for advanced renal cell carcinoma: extended follow-up of efficacy and safety results from a randomised, controlled, phase 3 trial. Lancet Oncol. 2019; 20(10): 1370–85.
- 7Mcdermott DF, Huseni MA, Atkins MB, Motzer RJ, Rini BI, Escudier B, et al. Clinical activity and molecular correlates of response to atezolizumab alone or in combination with bevacizumab versus sunitinib in renal cell carcinoma. Nat Med. 2018; 24(6): 749–57.
- 8Cella D, Grunwald V, Escudier B, Hammers HJ, George S, Nathan P, et al. Patient-reported outcomes of patients with advanced renal cell carcinoma treated with nivolumab plus ipilimumab versus sunitinib (CheckMate 214): a randomised, phase 3 trial. Lancet Oncol. 2019; 20(2): 297–310.
- 9Ravaud A, Hawkins R, Gardner JP, Von Der Maase H, Zantl N, Harper P, et al. Lapatinib versus hormone therapy in patients with advanced renal cell carcinoma: a randomized phase III clinical trial. J Clin Oncol. 2008; 26(14): 2285–91.
- 10Hsieh JJ, Purdue MP, Signoretti S, Swanton C, Albiges L, Schmidinger M, et al. Renal cell carcinoma. Nat Rev Dis Primers. 2017; 3:17009.
- 11Franovic A, Gunaratnam L, Smith K, Robert I, Patten D, Lee S. Translational up-regulation of the EGFR by tumor hypoxia provides a nonmutational explanation for its overexpression in human cancer. Proc Natl Acad Sci U S A. 2007; 104(32): 13092–7.
- 12Kassouf W. Editorial comment on: Von Hippel-Lindau tumor suppressor gene loss in renal cell carcinoma promotes oncogenic epidermal growth factor receptor signaling via Akt-1 and MEK-1. Eur Urol. 2008; 54(4): 853–4.
- 13Wang A, Bao Y, Wu Z, Zhao T, Wang D, Shi J, et al. Long noncoding RNA EGFR-AS1 promotes cell growth and metastasis via affecting HuR mediated mRNA stability of EGFR in renal cancer. Cell Death Dis. 2019; 10(3): 154.
- 14Feng ZH, Fang Y, Zhao LY, Lu J, Wang YQ, Chen ZH, et al. RIN1 promotes renal cell carcinoma malignancy by activating EGFR signaling through Rab25. Cancer Sci. 2017; 108(8): 1620–7.
- 15Massari F, Ciccarese C, Santoni M, Brunelli M, Piva F, Modena A, et al. Metabolic alterations in renal cell carcinoma. Cancer Treat Rev. 2015; 41(9): 767–76.
- 16Schoffski P, Dumez H, Clement P, Hoeben A, Prenen H, Wolter P, et al. Emerging role of tyrosine kinase inhibitors in the treatment of advanced renal cell cancer: a review. Ann Oncol. 2006; 17(8): 1185–96.
- 17Tomas A, Futter CE, Eden ER. EGF receptor trafficking: consequences for signaling and cancer. Trends Cell Biol. 2014; 24(1): 26–34.
- 18Yarden Y, Shilo BZ. SnapShot: EGFR signaling pathway. Cell. 2007; 131(5): 1018.
- 19Ceresa BP, Schmid SL. Regulation of signal transduction by endocytosis. Curr Opin Cell Biol. 2000; 12(2): 204–10.
- 20Dobrowolski R, De Robertis EM. Endocytic control of growth factor signalling: multivesicular bodies as signalling organelles. Nat Rev Mol Cell Biol. 2011; 13(1): 53–60.
- 21Goh LK, Sorkin A. Endocytosis of receptor tyrosine kinases. Cold Spring Harb Perspect Biol. 2013; 5(5):a017459.
- 22Stenmark H. Rab GTPases as coordinators of vesicle traffic. Nat Rev Mol Cell Biol. 2009; 10(8): 513–25.
- 23Wei D, Zhan W, Gao Y, Huang L, Gong R, Wang W, et al. RAB31 marks and controls an ESCRT-independent exosome pathway. Cell Res. 2021; 31(2): 157–77.
- 24Boerner JL, Demory ML, Silva C, Parsons SJ. Phosphorylation of Y845 on the epidermal growth factor receptor mediates binding to the mitochondrial protein cytochrome c oxidase subunit II. Mol Cell Biol. 2004; 24(16): 7059–71.
- 25Demory ML, Boerner JL, Davidson R, Faust W, Miyake T, Lee I, et al. Epidermal growth factor receptor translocation to the mitochondria: regulation and effect. J Biol Chem. 2009; 284(52): 36592–604.
- 26Yue X, Song W, Zhang W, Chen L, Xi Z, Xin Z, et al. Mitochondrially localized EGFR is subjected to autophagic regulation and implicated in cell survival. Autophagy. 2008; 4(5): 641–9.
- 27Youle RJ, Van Der Bliek AM. Mitochondrial fission, fusion, and stress. Science. 2012; 337(6098): 1062–5.
- 28Archer SL. Mitochondrial dynamics–mitochondrial fission and fusion in human diseases. N Engl J Med. 2013; 369(23): 2236–51.
- 29Gao S, Hu J. Mitochondrial Fusion: The Machineries In and Out. Trends Cell Biol. 2021; 31(1): 62–74.
- 30Qi M, Dai D, Liu J, Li Z, Liang P, Wang Y, et al. AIM2 promotes the development of non-small cell lung cancer by modulating mitochondrial dynamics. Oncogene. 2020; 39(13): 2707–23.
- 31Hernandez-Alvarez MI, Sebastian D, Vives S, Ivanova S, Bartoccioni P, Kakimoto P, et al. Deficient Endoplasmic Reticulum-Mitochondrial Phosphatidylserine Transfer Causes Liver Disease. Cell. 2019; 177(4): 881-95 e17.
- 32Schrepfer E, Scorrano L. Mitofusins, from Mitochondria to Metabolism. Mol Cell. 2016; 61(5): 683–94.
- 33Cao YL, Meng S, Chen Y, Feng JX, Gu DD, Yu B, et al. MFN1 structures reveal nucleotide-triggered dimerization critical for mitochondrial fusion. Nature. 2017; 542(7641): 372–6.
- 34Li YJ, Cao YL, Feng JX, Qi Y, Meng S, Yang JF, et al. Structural insights of human mitofusin-2 into mitochondrial fusion and CMT2A onset. Nat Commun. 2019; 10(1): 4914.
- 35Zhang J, Fonovic M, Suyama K, Bogyo M, Scott MP. Rab35 controls actin bundling by recruiting fascin as an effector protein. Science. 2009; 325(5945): 1250–4.
- 36Martens M, Ammar A, Riutta A, Waagmeester A, Slenter DN, Hanspers K, et al. WikiPathways: connecting communities. Nucleic Acids Res. 2021; 49(D1): D613-D21.
- 37Kim KT, Lee HW, Lee HO, Song HJ, Jeong Da E, Shin S, et al. Application of single-cell RNA sequencing in optimizing a combinatorial therapeutic strategy in metastatic renal cell carcinoma. Genome Biol. 2016; 17: 80.
- 38Brugarolas J. Renal-cell carcinoma–molecular pathways and therapies. N Engl J Med. 2007; 356(2): 185–7.
- 39Herbst RS, Fukuoka M, Baselga J. Gefitinib–a novel targeted approach to treating cancer. Nat Rev Cancer. 2004; 4(12): 956–65.
- 40Qiu C, Huang S, Park J, Park Y, Ko YA, Seasock MJ, et al. Renal compartment-specific genetic variation analyses identify new pathways in chronic kidney disease. Nat Med. 2018; 24(11): 1721–31.
- 41Carter P, Schnell U, Chaney C, Tong B, Pan X, Ye J, et al. Deletion of Lats1/2 in adult kidney epithelia leads to renal cell carcinoma. J Clin Invest. 2021; 131(11): e144108.
- 42Tostain J, Li G, Gentil-Perret A, Gigante M. Carbonic anhydrase 9 in clear cell renal cell carcinoma: a marker for diagnosis, prognosis and treatment. Eur J Cancer. 2010; 46(18): 3141–8.
- 43Eden ER, White IJ, Tsapara A, Futter CE. Membrane contacts between endosomes and ER provide sites for PTP1B-epidermal growth factor receptor interaction. Nat Cell Biol. 2010; 12(3): 267–72.
- 44Yang X, Zhang Y, Li S, Liu C, Jin Z, Wang Y, et al. Rab21 attenuates EGF-mediated MAPK signaling through enhancing EGFR internalization and degradation. Biochem Biophys Res Commun. 2012; 421(4): 651–7.
- 45Tarcic G, Boguslavsky SK, Wakim J, Kiuchi T, Liu A, Reinitz F, et al. An unbiased screen identifies DEP-1 tumor suppressor as a phosphatase controlling EGFR endocytosis. Curr Biol. 2009; 19(21): 1788–98.
- 46Morgenstern M, Peikert CD, Lubbert P, Suppanz I, Klemm C, Alka O, et al. Quantitative high-confidence human mitochondrial proteome and its dynamics in cellular context. Cell Metab. 2021; 33(12): 2464-83 e18.
- 47Mattila E, Pellinen T, Nevo J, Vuoriluoto K, Arjonen A, Ivaska J. Negative regulation of EGFR signalling through integrin-alpha1beta1-mediated activation of protein tyrosine phosphatase TCPTP. Nat Cell Biol. 2005; 7(1): 78–85.
- 48De Cubas AA, Rathmell WK. Epigenetic modifiers: activities in renal cell carcinoma. Nat Rev Urol. 2018; 15(10): 599–614.
- 49Yang J, Luo L, Zhao C, Li X, Wang Z, Zeng Z, et al. A Positive Feedback Loop between Inactive VHL-Triggered Histone Lactylation and PDGFRbeta Signaling Drives Clear Cell Renal Cell Carcinoma Progression. Int J Biol Sci. 2022; 18(8): 3470–83.
- 50Kaelin WG, Jr. The von Hippel-Lindau tumour suppressor protein: O2 sensing and cancer. Nat Rev Cancer. 2008; 8(11): 865–73.
- 51Zuchner S, Mersiyanova IV, Muglia M, Bissar-Tadmouri N, Rochelle J, Dadali EL, et al. Mutations in the mitochondrial GTPase mitofusin 2 cause Charcot-Marie-Tooth neuropathy type 2A. Nat Genet. 2004; 36(5): 449–51.
- 52Lim KL, Ng XH, Grace LG, Yao TP. Mitochondrial dynamics and Parkinson's disease: focus on parkin. Antioxid Redox Signal. 2012; 16(9): 935–49.
- 53Su B, Wang X, Bonda D, Perry G, Smith M, Zhu X. Abnormal mitochondrial dynamics–a novel therapeutic target for Alzheimer's disease? Mol Neurobiol. 2010; 41(2-3): 87–96.
- 54Tang J, Liu N, Zhuang S. Role of epidermal growth factor receptor in acute and chronic kidney injury. Kidney Int. 2013; 83(5): 804–10.
- 55Liu C, Liu L, Wang K, Li XF, Ge LY, Ma RZ, et al. VHL-HIF-2alpha axis-induced SMYD3 upregulation drives renal cell carcinoma progression via direct trans-activation of EGFR. Oncogene. 2020; 39(21): 4286–98.
- 56Morris ZS, Mcclatchey AI. Aberrant epithelial morphology and persistent epidermal growth factor receptor signaling in a mouse model of renal carcinoma. Proc Natl Acad Sci U S A. 2009; 106(24): 9767–72.
- 57Carpenter G. The EGF receptor: a nexus for trafficking and signaling. BioEssays: news and reviews in molecular, cellular and developmental biology. 2000; 22(8): 697–707.
- 58Miaczynska M. Effects of membrane trafficking on signaling by receptor tyrosine kinases. Cold Spring Harb Perspect Biol. 2013; 5(11):a009035.
- 59Jean S, Kiger AA. Coordination between RAB GTPase and phosphoinositide regulation and functions. Nat Rev Mol Cell Biol. 2012; 13(7): 463–70.
- 60Simpson JC, Griffiths G, Wessling-Resnick M, Fransen JA, Bennett H, Jones AT. A role for the small GTPase Rab21 in the early endocytic pathway. J Cell Sci. 2004; 117(Pt 26): 6297–311.
- 61Alanko J, Mai A, Jacquemet G, Schauer K, Kaukonen R, Saari M, et al. Integrin endosomal signalling suppresses anoikis. Nat Cell Biol. 2015; 17(11): 1412–21.
- 62Moreno-Layseca P, Jantti NZ, Godbole R, Sommer C, Jacquemet G, Al-Akhrass H, et al. Cargo-specific recruitment in clathrin- and dynamin-independent endocytosis. Nat Cell Biol. 2021; 23(10): 1073–84.
- 63Burte F, Carelli V, Chinnery PF, Yu-Wai-Man P. Disturbed mitochondrial dynamics and neurodegenerative disorders. Nat Rev Neurol. 2015; 11(1): 11–24.
- 64Mizushima N, Levine B. Autophagy in mammalian development and differentiation. Nat Cell Biol. 2010; 12(9): 823–30.
- 65Das A, Nag S, Mason AB, Barroso MM. Endosome-mitochondria interactions are modulated by iron release from transferrin. J Cell Biol. 2016; 214(7): 831–45.
- 66Kashatus JA, Nascimento A, Myers LJ, Sher A, Byrne FL, Hoehn KL, et al. Erk2 phosphorylation of Drp1 promotes mitochondrial fission and MAPK-driven tumor growth. Mol Cell. 2015; 57(3): 537–51.
- 67Li T, Han J, Jia L, Hu X, Chen L, Wang Y. PKM2 coordinates glycolysis with mitochondrial fusion and oxidative phosphorylation. Protein Cell. 2019; 10(8): 583–94.
- 68Yu Y, Peng XD, Qian XJ, Zhang KM, Huang X, Chen YH, et al. Fis1 phosphorylation by Met promotes mitochondrial fission and hepatocellular carcinoma metastasis. Signal Transduct Target Ther. 2021; 6(1): 401.
- 69Sun T, Aceto N, Meerbrey KL, Kessler JD, Zhou C, Migliaccio I, et al. Activation of multiple proto-oncogenic tyrosine kinases in breast cancer via loss of the PTPN12 phosphatase. Cell. 2011; 144(5): 703–18.
- 70Julien SG, Dube N, Read M, Penney J, Paquet M, Han Y, et al. Protein tyrosine phosphatase 1B deficiency or inhibition delays ErbB2-induced mammary tumorigenesis and protects from lung metastasis. Nat Genet. 2007; 39(3): 338–46.
- 71Ruivenkamp C, Hermsen M, Postma C, Klous A, Baak J, Meijer G, et al. LOH of PTPRJ occurs early in colorectal cancer and is associated with chromosomal loss of 18q12-21. Oncogene. 2003; 22(22): 3472–4.
- 72Balavenkatraman KK, Jandt E, Friedrich K, Kautenburger T, Pool-Zobel BL, Ostman A, et al. DEP-1 protein tyrosine phosphatase inhibits proliferation and migration of colon carcinoma cells and is upregulated by protective nutrients. Oncogene. 2006; 25(47): 6319–24.