YAP and TAZ Mediators at the Crossroad between Metabolic and Cellular Reprogramming
Abstract
:1. Introduction
2. The Mammalian Hippo Pathway
2.1. The Hippo Kinase Cascade
2.2. Numerous Initiating Signals May Regulate YAP and TAZ Activity
3. YAP and TAZ in Tissue Regeneration and Cell Reprogramming
3.1. YAP and TAZ Promote Tissue Growth and Cell Specification during Development
3.2. YAP and TAZ Promote Tissue Regeneration by Regulating Stemness, Dedifferentiation and Differentiation
3.3. YAP and TAZ Manipulation for Cellular Reprogramming
4. YAP and TAZ Are Modulators of Metabolic Reprogramming
4.1. Metabolic Status Influences YAP and TAZ Activity
4.2. YAP and TAZ Influences Cell Metabolism
5. Potential Role of YAP and TAZ in the Orchestration of a Metabolic Switch upon Cell Reprogramming
5.1. Mechanisms of the Metabolic Switch during Cell Reprogramming
5.2. YAP and TAZ in the Metabolic Remodeling of Cell Reprogramming
6. Conclusions
Author Contributions
Funding
Conflicts of Interest
References
- Aydin, B.; Mazzoni, E.O. Cell Reprogramming: The Many Roads to Success. Annu. Rev. Cell Dev. Biol. 2019, 35, 433–452. [Google Scholar] [CrossRef]
- Bitman-Lotan, E.; Orian, A. Nuclear organization and regulation of the differentiated state. Cell. Mol. Life Sci. 2021. [Google Scholar] [CrossRef]
- Friedmann-Morvinski, D.; Verma, I.M. Dedifferentiation and reprogramming: Origins of cancer stem cells. EMBO Rep. 2014, 15, 244–253. [Google Scholar] [CrossRef] [Green Version]
- Caiazza, C.; D’Agostino, M.; Passaro, F.; Faicchia, D.; Mallardo, M.; Paladino, S.; Pierantoni, G.M.; Tramontano, D. Effects of Long-Term Citrate Treatment in the PC3 Prostate Cancer Cell Line. Int. J. Mol. Sci. 2019, 20, 2613. [Google Scholar] [CrossRef] [Green Version]
- Mathieu, J.; Ruohola-Baker, H. Metabolic remodeling during the loss and acquisition of pluripotency. Development 2017, 144, 541–551. [Google Scholar] [CrossRef] [Green Version]
- Nishimura, K.; Fukuda, A.; Hisatake, K. Mechanisms of the Metabolic Shift during Somatic Cell Reprogramming. Int. J. Mol. Sci. 2019, 20, 2254. [Google Scholar] [CrossRef] [Green Version]
- Warburg, O. On the origin of cancer cells. Science 1956, 123, 309–314. [Google Scholar] [CrossRef]
- Lai, X.; Li, Q.; Wu, F.; Lin, J.; Chen, J.; Zheng, H.; Guo, L. Epithelial-Mesenchymal Transition and Metabolic Switching in Cancer: Lessons From Somatic Cell Reprogramming. Front. Cell Dev. Biol. 2020, 8, 760. [Google Scholar] [CrossRef]
- Wu, J.; Ocampo, A.; Belmonte, J. Cellular Metabolism and Induced Pluripotency. Cell 2016, 166, 1371–1385. [Google Scholar] [CrossRef] [Green Version]
- Ryall, J.G.; Cliff, T.; Dalton, S.; Sartorelli, V. Metabolic Reprogramming of Stem Cell Epigenetics. Cell Stem Cell 2015, 17, 651–662. [Google Scholar] [CrossRef] [Green Version]
- Sun, H.; Yang, X.; Liang, L.; Zhang, M.; Li, Y.; Chen, J.; Wang, F.; Yang, T.; Meng, F.; Lai, X.; et al. Metabolic switch and epithelial-mesenchymal transition cooperate to regulate pluripotency. EMBO J. 2020, 39, e102961. [Google Scholar] [CrossRef]
- Yu, F.X.; Zhao, B.; Guan, K.L. Hippo Pathway in Organ Size Control, Tissue Homeostasis, and Cancer. Cell 2015, 163, 811–828. [Google Scholar] [CrossRef] [Green Version]
- Heng, B.C.; Zhang, X.; Aubel, D.; Bai, Y.; Li, X.; Li, X.; Wei, Y.; Fussenegger, M.; Deng, X. Role of YAP/TAZ in Cell Lineage Fate Determination and Related Signaling Pathways. Front. Cell Dev. Biol. 2020, 8, 735. [Google Scholar] [CrossRef]
- Moya, I.M.; Halder, G. Hippo–YAP/TAZ signalling in organ regeneration and regenerative medicine. Nat. Rev. Mol. Cell Biol. 2019, 20, 211–226. [Google Scholar] [CrossRef]
- Davis, J.R.; Tapon, N. Hippo signalling during development. Development 2019, 146, dev167106. [Google Scholar] [CrossRef] [Green Version]
- Pocaterra, A.; Romani, P.; Dupont, S. YAP/TAZ functions and their regulation at a glance. J. Cell Sci. 2020, 133, jcs230425. [Google Scholar] [CrossRef]
- Justice, R.W.; Zilian, O.; Woods, D.F.; Noll, M.; Bryant, P.J. The Drosophila tumor suppressor gene warts encodes a homolog of human myotonic dystrophy kinase and is required for the control of cell shape and proliferation. Genes Dev. 1995, 9, 534–546. [Google Scholar] [CrossRef] [Green Version]
- Xu, T.; Wang, W.; Zhang, S.; Stewart, R.A.; Yu, W. Identifying tumor suppressors in genetic mosaics: The Drosophila lats gene encodes a putative protein kinase. Development 1995, 121, 1053–1063. [Google Scholar]
- Fulford, A.; Tapon, N.; Ribeiro, P.S. Upstairs, downstairs: Spatial regulation of Hippo signalling. Curr. Opin. Cell Biol. 2018, 51, 22–32. [Google Scholar] [CrossRef]
- Sudol, M. Yes-associated protein (YAP65) is a proline-rich phosphoprotein that binds to the SH3 domain of the Yes proto-oncogene product. Oncogene 1994, 9, 2145–2152. [Google Scholar] [PubMed]
- Morin-Kensicki, E.M.; Boone, B.N.; Howell, M.; Stonebraker, J.R.; Teed, J.; Alb, J.G.; Magnuson, T.R.; O’Neal, W.; Milgram, S.L. Defects in yolk sac vasculogenesis, chorioallantoic fusion, and embryonic axis elongation in mice with targeted disruption of Yap65. Mol. Cell. Biol. 2006, 26, 77–87. [Google Scholar] [CrossRef] [Green Version]
- Kanai, F.; Marignani, P.A.; Sarbassova, D.; Yagi, R.; Hall, R.A.; Donowitz, M.; Hisaminato, A.; Fujiwara, T.; Ito, Y.; Cantley, L.C.; et al. TAZ: A novel transcriptional co-activator regulated by interactions with 14-3-3 and PDZ domain proteins. EMBO J. 2000, 19, 6778–6791. [Google Scholar] [CrossRef]
- Yin, F.; Yu, J.; Zheng, Y.; Chen, Q.; Zhang, N.; Pan, D. Spatial organization of Hippo signaling at the plasma membrane mediated by the tumor suppressor Merlin/NF2. Cell 2013, 154, 1342–1355. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Glantschnig, H.; Rodan, G.A.; Reszka, A.A. Mapping of MST1 kinase sites of phosphorylation. Activation and autophosphorylation. J. Biol. Chem. 2002, 277, 42987–42996. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Genevet, A.; Wehr, M.C.; Brain, R.; Thompson, B.J.; Tapon, N. Kibra is a regulator of the Salvador/Warts/Hippo signaling network. Dev. Cell 2010, 18, 300–308. [Google Scholar] [CrossRef] [Green Version]
- Hirate, Y.; Hirahara, S.; Inoue, K.; Suzuki, A.; Alarcon, V.B.; Akimoto, K.; Hirai, T.; Hara, T.; Adachi, M.; Chida, K.; et al. Polarity-dependent distribution of angiomotin localizes Hippo signaling in preimplantation embryos. Curr. Biol. 2013, 23, 1181–1194. [Google Scholar] [CrossRef] [Green Version]
- Li, Y.; Zhou, H.; Li, F.; Chan, S.W.; Lin, Z.; Wei, Z.; Yang, Z.; Guo, F.; Lim, W.J.; Xing, W.; et al. Angiomotin binding-induced activation of Merlin/NF2 in the Hippo pathway. Cell Res. 2015, 25, 801–817. [Google Scholar] [CrossRef] [Green Version]
- Chan, E.H.; Nousiainen, M.; Chalamalasetty, R.B.; Schäfer, A.; Nigg, E.A.; Sillje, H.H.W. The Ste20-like kinase Mst2 activates the human large tumor suppressor kinase Lats1. Oncogene 2005, 24, 2076–2086. [Google Scholar] [CrossRef]
- Meng, Z.; Moroishi, T.; Mottier-Pavie, V.; Plouffe, S.W.; Hansen, C.G.; Hong, A.W.; Park, H.W.; Mo, J.S.; Lu, W.; Lu, S.; et al. MAP4K family kinases act in parallel to MST1/2 to activate LATS1/2 in the Hippo pathway. Nat. Commun. 2015, 6, 8357. [Google Scholar] [CrossRef]
- Plouffe, S.W.; Meng, Z.; Lin, K.C.; Lin, B.; Hong, A.W.; Chun, J.V.; Guan, K.L. Characterization of Hippo Pathway Components by Gene Inactivation. Mol. Cell 2016, 64, 993–1008. [Google Scholar] [CrossRef] [Green Version]
- Boggiano, J.C.; Vanderzalm, P.J.; Fehon, R.G. Tao-1 phosphorylates Hippo/MST kinases to regulate the Hippo-Salvador-Warts tumor suppressor pathway. Dev. Cell 2011, 21, 888–895. [Google Scholar] [CrossRef] [Green Version]
- Poon, C.L.; Lin, J.I.; Zhang, X.; Harvey, K.F. The sterile 20-like kinase Tao-1 controls tissue growth by regulating the Salvador-Warts-Hippo pathway. Dev. Cell 2011, 21, 896–906. [Google Scholar] [CrossRef] [Green Version]
- Zhao, B.; Li, L.; Tumaneng, K.; Wang, C.Y.; Guan, K.L. A coordinated phosphorylation by Lats and CK1 regulates YAP stability through SCF(beta-TRCP). Genes Dev. 2010, 24, 72–85. [Google Scholar] [CrossRef] [Green Version]
- Liu, C.Y.; Zha, Z.Y.; Zhou, X.; Zhang, H.; Huang, W.; Zhao, D.; Li, T.; Chan, S.W.; Lim, C.J.; Hong, W.; et al. The hippo tumor pathway promotes TAZ degradation by phosphorylating a phosphodegron and recruiting the SCFβ-TrCP E3 ligase. J. Biol. Chem. 2010, 285, 37159–37169. [Google Scholar] [CrossRef] [Green Version]
- Nguyen, T.H.; Kugler, J.M. Ubiquitin-Dependent Regulation of the Mammalian Hippo Pathway: Therapeutic Implications for Cancer. Cancers 2018, 10, 121. [Google Scholar] [CrossRef] [Green Version]
- Liang, N.; Zhang, C.; Dill, P.; Panasyuk, G.; Pion, D.; Koka, V.; Gallazzini, M.; Olson, E.N.; Lam, H.; Henske, E.P.; et al. Regulation of YAP by mTOR and autophagy reveals a therapeutic target of tuberous sclerosis complex. J. Exp. Med. 2014, 211, 2249–2263. [Google Scholar] [CrossRef]
- Liu, C.Y.; Lv, X.; Li, T.; Xu, Y.; Zhou, X.; Zhao, S.; Xiong, Y.; Lei, Q.Y.; Guan, K.L. PP1 cooperates with ASPP2 to dephosphorylate and activate TAZ. J. Biol. Chem. 2011, 286, 5558–5566. [Google Scholar] [CrossRef] [Green Version]
- Wang, P.; Bai, Y.; Song, B.; Wang, Y.; Liu, D.; Lai, Y.; Bi, X.; Yuan, Z. PP1A-mediated dephosphorylation positively regulates YAP2 activity. PLoS ONE 2011, 6, e24288. [Google Scholar] [CrossRef] [Green Version]
- Zhang, W.; Gao, Y.; Li, P.; Shi, Z.; Guo, T.; Li, F.; Han, X.; Feng, Y.; Zheng, C.; Wang, Z.; et al. VGLL4 functions as a new tumor suppressor in lung cancer by negatively regulating the YAP-TEAD transcriptional complex. Cell Res. 2014, 24, 331–343. [Google Scholar] [CrossRef] [Green Version]
- Ota, M.; Sasaki, H. Mammalian Tead proteins regulate cell proliferation and contact inhibition as transcriptional mediators of Hippo signaling. Development 2008, 135, 4059–4069. [Google Scholar] [CrossRef] [Green Version]
- Zaidi, S.K.; Sullivan, A.J.; Medina, R.; Ito, Y.; van Wijnen, A.J. Tyrosine phosphorylation controls Runx2-mediated subnuclear targeting of YAP to repress transcription. EMBO J. 2004, 23, 790–799. [Google Scholar] [CrossRef]
- Grannas, K.; Arngården, L.; Lönn, P.; Mazurkiewicz, M.; Blokzijl, A.; Zieba, A.; Soderberg, O. Crosstalk between Hippo and TGFβ: Subcellular Localization of YAP/TAZ/Smad Complexes. J. Mol. Biol. 2015, 427, 3407–3415. [Google Scholar] [CrossRef]
- Zhao, B.; Wei, X.; Li, W.; Udan, R.S.; Yang, Q.; Kim, J.; Xie, J.; Ikenoue, T.; Yu, J.; Li, L.; et al. Inactivation of YAP oncoprotein by the Hippo pathway is involved in cell contact inhibition and tissue growth control. Genes Dev. 2007, 21, 2747–2761. [Google Scholar] [CrossRef] [Green Version]
- Sun, S.; Irvine, K.D. Cellular Organization and Cytoskeletal Regulation of the Hippo Signaling Network. Trends Cell Biol. 2016, 26, 694–704. [Google Scholar] [CrossRef] [Green Version]
- Yu, F.X.; Zhang, Y.; Park, H.W.; Jewell, J.L.; Chen, Q.; Deng, Y.; Pan, D.; Taylor, S.S.; Lai, Z.C.; Guan, K.L. Protein kinase A activates the Hippo pathway to modulate cell proliferation and differentiation. Genes Dev. 2013, 27, 1223–1232. [Google Scholar] [CrossRef] [Green Version]
- Wu, Y.; Griffin, E.E. Regulation of Cell Polarity by PAR-1/MARK Kinase. Curr. Top. Dev. Biol. 2017, 123, 365–397. [Google Scholar] [CrossRef] [Green Version]
- Fan, R.; Kim, N.G.; Gumbiner, B.M. Regulation of Hippo pathway by mitogenic growth factors via phosphoinositide 3-kinase and phosphoinositide-dependent kinase-1. Proc. Natl. Acad. Sci. USA 2013, 110, 2569–2574. [Google Scholar] [CrossRef] [Green Version]
- Reddy, B.V.; Irvine, K.D. Regulation of Hippo signaling by EGFR-MAPK signaling through Ajuba family proteins. Dev. Cell 2013, 24, 459–471. [Google Scholar] [CrossRef] [Green Version]
- Straßburger, K.; Tiebe, M.; Pinna, F.; Breuhahn, K.; Teleman, A.A. Insulin/IGF signaling drives cell proliferation in part via Yorkie/YAP. Dev. Biol. 2012, 367, 187–196. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Azzolin, L.; Panciera, T.; Soligo, S.; Enzo, E.; Bicciato, S.; Dupont, S.; Bresolin, S.; Frasson, C.; Basso, G.; Guzzardo, V.; et al. YAP/TAZ incorporation in the β-catenin destruction complex orchestrates the Wnt response. Cell 2014, 158, 157–170. [Google Scholar] [CrossRef] [Green Version]
- Park, H.W.; Kim, Y.C.; Yu, B.; Moroishi, T.; Mo, J.S.; Plouffe, S.W.; Meng, Z.; Lin, K.C.; Yu, F.X.; Alexander, C.M.; et al. Alternative Wnt Signaling Activates YAP/TAZ. Cell 2015, 162, 780–794. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yu, F.X.; Zhao, B.; Panupinthu, N.; Jewell, J.L.; Lian, I.; Wang, L.H.; Zhao, J.; Yuan, H.; Tumaneng, K.; Li, H.; et al. Regulation of the Hippo-YAP pathway by G-protein-coupled receptor signaling. Cell 2012, 150, 780–791. [Google Scholar] [CrossRef] [Green Version]
- Baddour, J.A.; Sousounis, K.; Tsonis, P.A. Organ repair and regeneration: An overview. Birth Defects Res. C Embryo Today 2012, 96, 1–29. [Google Scholar] [CrossRef] [PubMed]
- Merrell, A.; Stanger, B. Adult cell plasticity in vivo: De-differentiation and transdifferentiation are back in style. Nat. Rev. Mol. Cell Biol. 2016, 17, 413–425. [Google Scholar] [CrossRef]
- Mao, A.S.; Mooney, D.J. Regenerative medicine: Current therapies and future directions. Proc. Natl. Acad. Sci. USA 2015, 112, 14452–14459. [Google Scholar] [CrossRef] [Green Version]
- Makita, R.; Uchijima, Y.; Nishiyama, K.; Amano, T.; Chen, Q.; Takeuchi, T.; Mitani, A.; Nagase, T.; Yatomi, Y.; Aburatani, H.; et al. Multiple renal cysts, urinary concentration defects, and pulmonary emphysematous changes in mice lacking TAZ. Am. J. Physiol. Renal Physiol. 2008, 294, F542–F553. [Google Scholar] [CrossRef]
- Mitani, A.; Nagase, T.; Fukuchi, K.; Aburatani, H.; Makita, R.; Kurihara, H. Transcriptional coactivator with PDZ-binding motif is essential for normal alveolarization in mice. Am. J. Respir. Crit. Care Med. 2009, 180, 326–338. [Google Scholar] [CrossRef] [PubMed]
- Nishioka, N.; Inoue, K.; Adachi, K.; Kiyonari, H.; Ota, M.; Ralston, A.; Yabuta, N.; Hirahara, S.; Stephenson, R.O.; Ogonuki, N.; et al. The Hippo signaling pathway components Lats and Yap pattern Tead4 activity to distinguish mouse trophectoderm from inner cell mass. Dev. Cell 2009, 16, 398–410. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- von Gise, A.; Lin, Z.; Schlegelmilch, K.; Honor, L.B.; Pan, G.M.; Buck, J.N.; Ma, Q.; Ishiwata, T.; Zhou, B.; Camargo, F.D.; et al. YAP1, the nuclear target of Hippo signaling, stimulates heart growth through cardiomyocyte proliferation but not hypertrophy. Proc. Natl. Acad. Sci. USA 2012, 109, 2394–2399. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ragni, C.V.; Diguet, N.; Le Garrec, J.F.; Novotova, M.; Resende, T.P.; Pop, S.; Charon, N.; Guillemot, L.; Kitasato, L.; Badouel, C.; et al. Amotl1 mediates sequestration of the Hippo effector Yap1 downstream of Fat4 to restrict heart growth. Nat. Commun. 2017, 8, 14582. [Google Scholar] [CrossRef] [PubMed]
- Wang, X.; Freire Valls, A.; Schermann, G.; Shen, Y.; Moya, I.M.; Castro, L.; Urban, S.; Solecki, G.M.; Winkler, F.; Riedemann, L.; et al. YAP/TAZ orchestrate VEGF signaling during developmental angiogenesis. Dev. Cell 2017, 42, 462–478.e7. [Google Scholar] [CrossRef] [Green Version]
- Kim, J.; Kim, Y.H.; Kim, J.; Park, D.Y.; Bae, H.; Lee, D.H.; Kim, K.H.; Hong, S.P.; Jang, S.P.; Kubota, Y.; et al. YAP/TAZ regulates sprouting angiogenesis and vascular barrier maturation. J. Clin. Investig. 2017, 127, 3441–3461. [Google Scholar] [CrossRef]
- Singh, A.; Ramesh, S.; Cibi, D.M.; Yun, L.S.; Li, J.; Manderfield, L.J.; Olson, E.N.; Epstein, J.A.; Singh, M.K. Hippo signaling mediators Yap and Taz are required in the epicardium for coronary vasculature development. Cell Rep. 2016, 15, 1384–1393. [Google Scholar] [CrossRef] [Green Version]
- Lavado, A.; Park, J.Y.; Paré, J.; Finkelstein, D.; Pan, H.; Xu, B.; Fan, Y.; Kumar, R.P.; Neale, G.; Kwak, Y.D.; et al. The hippo pathway prevents YAP/TAZ-driven hypertranscription and controls neural progenitor number. Dev. Cell 2018, 47, 576–591.e8. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Van Hateren, N.J.; Das, R.M.; Hautbergue, G.M.; Borycki, A.G.; Placzek, M.; Wilson, S.A. FatJ acts via the Hippo mediator Yap1 to restrict the size of neural progenitor cell pools. Development 2011, 138, 1893–1902. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cao, X.; Pfaff, S.L.; Gage, F.H. YAP regulates neural progenitor cell number via the TEA domain transcription factor. Genes Dev. 2008, 22, 3320–3334. [Google Scholar] [CrossRef] [Green Version]
- Xia, W.; Liu, Y.; Jiao, J. GRM7 regulates embryonic neurogenesis via CREB and YAP. Stem Cell Rep. 2015, 4, 795–810. [Google Scholar] [CrossRef] [Green Version]
- Lin, Y.T.; Ding, J.Y.; Li, M.Y.; Yeh, T.S.; Wang, T.W.; Yu, J.Y. YAP regulates neuronal differentiation through Sonic hedgehog signaling pathway. Exp. Cell Res. 2012, 318, 1877–1888. [Google Scholar] [CrossRef] [PubMed]
- Reginensi, A.; Scott, R.P.; Gregorieff, A.; Bagherie-Lachidan, M.; Chung, C.; Lim, D.S.; Pawson, T.; Wrana, J.; McNeill, H. Yap- and Cdc42-dependent nephrogenesis and morphogenesis during mouse kidney development. PLoS Genet. 2013, 9, e1003380. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- McNeill, H.; Reginensi, A. Lats1/2 regulate Yap/Taz to control nephron progenitor epithelialization and inhibit myofibroblast formation. J. Am. Soc. Nephrol. 2017, 28, 852–861. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Du, K.; Hyun, J.; Premont, R.T.; Choi, S.S.; Michelotti, G.A.; Swiderska-Syn, M.; Dalton, G.D.; Thelen, E.; Rizi, B.S.; Jung, Y.; et al. Hedgehog-YAP signaling pathway regulates glutaminolysis to control activation of hepatic stellate cells. Gastroenterology 2018, 154, 1465–1479.e13. [Google Scholar] [CrossRef] [Green Version]
- Lu, L.; Finegold, M.J.; Johnson, R.L. Hippo pathway coactivators Yap and Taz are required to coordinate mammalian liver regeneration. Exp. Mol. Med. 2018, 50, e423. [Google Scholar] [CrossRef] [PubMed]
- Qin, H.; Hejna, M.; Liu, Y.; Percharde, M.; Wossidlo, M.; Blouin, L.; Durruthy-Durruthy, J.; Wong, P.; Qi, Z.; Yu, J.; et al. YAP induces human naive pluripotency. Cell Rep. 2016, 14, 2301–2312. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lian, I.; Kim, J.; Okazawa, H.; Zhao, J.; Zhao, B.; Yu, J.; Chinnaiyan, A.; Israel, M.A.; Goldstein, L.S.B.; Abujarour, R.; et al. The role of YAP transcription coactivator in regulating stem cell self-renewal and differentiation. Genes Dev. 2010, 24, 1106–1118. [Google Scholar] [CrossRef] [Green Version]
- Tamm, C.; Böwer, N.; Annerén, C. Regulation of mouse embryonic stem cell self-renewal by a Yes-YAP-TEAD2 signaling pathway downstream of LIF. J. Cell Sci. 2011, 124, 1136–1144. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chung, H.; Lee, B.K.; Uprety, N.; Shen, W.; Lee, J.; Kim, J. Yap1 is dispensable for self-renewal but required for proper differentiation of mouse embryonic stem (ES) cells. EMBO Rep. 2016, 17, 519–529. [Google Scholar] [CrossRef] [Green Version]
- Papaspyropoulos, A.; Bradley, L.; Thapa, A.; Leung, C.Y.; Toskas, K.; Koennig, D.; Pefani, D.E.; Raso, C.; Grou, C.; Hamilton, G.; et al. RASSF1A uncouples Wnt from Hippo signalling and promotes YAP mediated differentiation via p73. Nat. Commun. 2018, 9, 424. [Google Scholar] [CrossRef]
- Passaro, F.; De Martino, I.; Zambelli, F.; Di Benedetto, G.; Barbato, M.; D’Erchia, A.M.; Manzari, C.; Pesole, G.; Mutarelli, M.; Cacchiarelli, D.; et al. YAP contributes to DNA methylation remodeling upon mouse embryonic stem cell differentiation. J. Biol. Chem. 2021, 296. [Google Scholar] [CrossRef]
- Xu, R.H.; Peck, R.M.; Li, D.S.; Feng, X.; Ludwig, T.; Thomson, J.A. Basic FGF and suppression of BMP signaling sustain undifferentiated proliferation of human ES cells. Nat. Methods 2005, 2, 185–190. [Google Scholar] [CrossRef]
- Varelas, X.; Sakuma, R.; Samavarchi-Tehrani, P.; Peerani, R.; Rao, B.M.; Dembowy, J.; Yaffe, M.B.; Zandstra, P.W.; Wrana, J.L. TAZ controls Smad nucleocytoplasmic shuttling and regulates human embryonic stem-cell self-renewal. Nat. Cell Biol. 2008, 10, 837–848. [Google Scholar] [CrossRef]
- Musah, S.; Wrighton, P.J.; Zaltsman, Y.; Zhong, X.; Zorn, S.; Parlato, M.B.; Hsiao, C.; Palecek, S.P.; Chang, Q.; Murphy, W.L.; et al. Substratum-induced differentiation of human pluripotent stem cells reveals the coactivator YAP is a potent regulator of neuronal specification. Proc. Natl. Acad. Sci. USA 2014, 111, 13805–13810. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ohgushi, M.; Minaguchi, M.; Sasai, Y. Rho-signaling-directed YAP/TAZ activity underlies the long-term survival and expansion of human embryonic Stem cells. Cell Stem Cell 2015, 17, 448–461. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hsiao, C.; Lampe, M.; Nillasithanukroh, S.; Han, W.; Lian, X.; Palecek, S.P. Human pluripotent stem cell culture density modulates YAP signaling. Biotechnol. J. 2016, 11, 662–675. [Google Scholar] [CrossRef] [Green Version]
- Han, D.; Byun, S.H.; Park, S.; Kim, J.; Kim, I.; Ha, S.; Kwon, M.; Yoon, K. YAP/TAZ enhance mammalian embryonic neural stem cell characteristics in a Tead-dependent manner. Biochem. Biophys. Res. Commun. 2015, 458, 110–116. [Google Scholar] [CrossRef]
- Bao, X.M.; He, Q.; Wang, Y.; Huang, Z.H.; Yuan, Z.Q. The roles and mechanisms of the Hippo/YAP signaling pathway in the nervous system. Yi Chuan 2017, 39, 630–641. [Google Scholar] [CrossRef] [PubMed]
- Judson, R.N.; Tremblay, A.M.; Knopp, P.; White, R.B.; Urcia, R.; De Bari, C.; Zammit, P.S.; Camargo, F.D.; Wackerhage, H. The Hippo pathway member Yap plays a key role in influencing fate decisions in muscle satellite cells. J. Cell Sci. 2012, 125, 6009–6019. [Google Scholar] [CrossRef] [Green Version]
- Imajo, M.; Ebisuya, M.; Nishida, E. Dual role of YAP and TAZ in renewal of the intestinal epithelium. Nat. Cell Biol. 2015, 17, 7–19. [Google Scholar] [CrossRef]
- Hu, J.K.; Du, W.; Shelton, S.J.; Oldham, M.C.; DiPersio, C.M.; Klein, O.D. An FAK-YAP-mTOR signaling axis regulates stem cell-based tissue renewal in Mice. Cell Stem Cell 2017, 21, 91–106.e6. [Google Scholar] [CrossRef] [Green Version]
- Tang, Y.; Weiss, S.J. Snail/Slug-YAP/TAZ complexes cooperatively regulate mesenchymal stem cell function and bone formation. Cell Cycle 2017, 16, 399–405. [Google Scholar] [CrossRef] [Green Version]
- Qian, W.; Gong, L.; Cui, X.; Zhang, Z.; Bajpai, A.; Liu, C.; Castillo, A.B.; Teo, J.C.M.; Chen, W. Nanotopographic regulation of human mesenchymal stem cell osteogenesis. ACS Appl. Mater. Interfaces 2017, 9, 41794–41806. [Google Scholar] [CrossRef]
- Loebel, C.; Mauck, R.L.; Burdick, J.A. Local nascent protein deposition and remodelling guide mesenchymal stromal cell mechanosensing and fate in three-dimensional hydrogels. Nat. Mater. 2019, 18, 883–891. [Google Scholar] [CrossRef]
- Song, L.; Wang, K.; Li, Y.; Yang, Y. Nanotopography promoted neuronal differentiation of human induced pluripotent stem cells. Colloids Surf. B Biointerfaces 2016, 148, 49–58. [Google Scholar] [CrossRef] [PubMed]
- Watt, K.I.; Judson, R.; Medlow, P.; Reid, K.; Kurth, T.B.; Burniston, J.G.; Ratkevicius, A.; De Bari, C.; Wackerhage, H. Yap is a novel regulator of C2C12 myogenesis. Biochem. Biophys. Res. Commun. 2010, 393, 619–624. [Google Scholar] [CrossRef] [PubMed]
- Beverdam, A.; Claxton, C.; Zhang, X.; James, G.; Harvey, K.F.; Key, B. Yap controls stem/progenitor cell proliferation in the mouse postnatal epidermis. J. Investig. Dermatol. 2013, 133, 1497–1505. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lee, M.J.; Byun, M.R.; Furutani-Seiki, M.; Hong, J.H.; Jung, H.S. YAP and TAZ regulate skin wound healing. J. Investig. Dermatol. 2014, 134, 518–525. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Totaro, A.; Castellan, M.; Battilana, G.; Zanconato, F.; Azzolin, L.; Giulitti, S.; Cordenonsi, M.; Piccolo, S. YAP/TAZ link cell mechanics to Notch signalling to control epidermal stem cell fate. Nat. Commun. 2017, 8, 15206. [Google Scholar] [CrossRef] [PubMed]
- Le Bouteiller, M.; Jensen, K.B. Hippo signalling directs intestinal fate. Nat. Cell Biol. 2015, 17, 5–6. [Google Scholar] [CrossRef] [PubMed]
- Hou, N.; Wen, Y.; Yuan, X.; Xu, H.; Wang, X.; Li, F.; Ye, B. Activation of Yap1/Taz signaling in ischemic heart disease and dilated cardiomyopathy. Exp. Mol. Pathol. 2017, 103, 267–275. [Google Scholar] [CrossRef]
- Del Re, D.P.; Yang, Y.; Nakano, N.; Cho, J.; Zhai, P.; Yamamoto, T.; Zhang, N.; Yabuta, N.; Nojima, H.; Pan, D.; et al. Yes-associated protein isoform 1 (Yap1) promotes cardiomyocyte survival and growth to protect against myocardial ischemic injury. J. Biol. Chem. 2013, 288, 3977–3988. [Google Scholar] [CrossRef] [Green Version]
- Konishi, T.; Schuster, R.M.; Lentsch, A.B. Proliferation of hepatic stellate cells, mediated by YAP and TAZ, contributes to liver repair and regeneration after liver ischemia-reperfusion injury. Am. J. Physiol. Gastrointest. Liver Physiol. 2018, 314, G471–G482. [Google Scholar] [CrossRef]
- Zhao, Y.; Fei, X.; Guo, J.; Zou, G.; Pan, W.; Zhang, J.; Huang, Y.; Liu, T.; Cheng, W. Induction of reprogramming of human amniotic epithelial cells into iPS cells by overexpression of Yap, Oct4, and Sox2 through the activation of the Hippo-Yap pathway. Exp. Ther. Med. 2017, 14, 199–206. [Google Scholar] [CrossRef]
- Qin, H.; Blaschke, K.; Wei, G.; Ohi, Y.; Blouin, L.; Qi, Z.; Yu, J.; Yeh, R.F.; Hebrok, M.; Ramalho-Santos, M. Transcriptional analysis of pluripotency reveals the Hippo pathway as a barrier to reprogramming. Hum. Mol. Genet. 2012, 21, 2054–2067. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hartman, A.A.; Scalf, S.M.; Zhang, J.; Hu, X.; Chen, X.; Eastman, A.E.; Yang, C.; Guo, S. YAP Non-cell-autonomously Promotes Pluripotency Induction in Mouse Cells. Stem Cell Rep. 2020, 14, 730–743. [Google Scholar] [CrossRef] [PubMed]
- Jun, J.I.; Lau, L.F. The matricellular protein CCN1 induces fibroblast senescence and restricts fibrosis in cutaneous wound healing. Nat. Cell Biol. 2010, 12, 676–685. [Google Scholar] [CrossRef]
- Passaro, F.; Testa, G. Implications of Cellular Aging in Cardiac Reprogramming. Front. Cardiovasc. Med. 2018, 5, 43. [Google Scholar] [CrossRef] [Green Version]
- Panciera, T.; Azzolin, L.; Fujimura, A.; Di Biagio, D.; Frasson, C.; Bresolin, S.; Soligo, S.; Basso, G.; Bicciato, S.; Rosato, A.; et al. Induction of expandable tissue-specific stem/progenitor cells through transient expression of YAP/TAZ. Cell Stem Cell 2016, 19, 725–737. [Google Scholar] [CrossRef] [Green Version]
- Yui, S.; Azzolin, L.; Maimets, M.; Pedersen, M.T.; Fordham, R.P.; Hansen, S.L.; Larsen, H.L.; Guiu, J.; Alves, M.R.P.; Rundsten, C.F.; et al. YAP/TAZ-dependent reprogramming of colonic epithelium links ECM remodeling to tissue regeneration. Cell Stem Cell 2018, 22, 35–49.e7. [Google Scholar] [CrossRef] [Green Version]
- Fitamant, J.; Kottakis, F.; Benhamouche, S.; Tian, H.S.; Chuvin, N.; Parachoniak, C.A.; Nagle, J.M.; Perera, R.M.; Lapouge, M.; Deshpande, V.; et al. YAP inhibition restores hepatocyte differentiation in advanced HCC, leading to tumor regression. Cell Rep. 2015, 10, 1692–1707. [Google Scholar] [CrossRef] [Green Version]
- Passaro, F.; Testa, G.; Ambrosone, L.; Costagliola, C.; Tocchetti, C.G.; di Nezza, F.; Russo, M.; Pirozzi, F.; Abete, P.; Russo, T.; et al. Nanotechnology-Based Cardiac Targeting and Direct Cardiac Reprogramming: The Betrothed. Stem Cells Int. 2017, 2017, 4940397. [Google Scholar] [CrossRef] [Green Version]
- Testa, G.; Russo, M.; Di Benedetto, G.; Barbato, M.; Parisi, S.; Pirozzi, F.; Tocchetti, C.G.; Abete, P.; Bonaduce, D.; Russo, T.; et al. Bmi1 inhibitor PTC-209 promotes Chemically-induced Direct Cardiac Reprogramming of cardiac fibroblasts into cardiomyocytes. Sci. Rep. 2020, 10, 7129. [Google Scholar] [CrossRef] [PubMed]
- Kurotsu, S.; Sadahiro, T.; Fujita, R.; Tani, H.; Yamakawa, H.; Tamura, F.; Isomi, M.; Kojima, H.; Yamada, Y.; Abe, Y.; et al. Soft Matrix Promotes Cardiac Reprogramming via Inhibition of YAP/TAZ and Suppression of Fibroblast Signatures. Stem Cell Rep. 2020, 15, 612–628. [Google Scholar] [CrossRef]
- Mills, R.J.; Titmarsh, D.M.; Koenig, X.; Parker, B.L.; Ryall, J.G.; Quaife-Ryan, G.A.; Voges, H.K.; Hodson, M.P.; Ferguson, C.; Drowley, L.; et al. Functional screening in human cardiac organoids reveals a metabolic mechanism for cardiomyocyte cell cycle arrest. Proc. Natl. Acad. Sci. USA 2017, 114, E8372–E8381. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mochizuki, M.; Lorenz, V.; Ivanek, R.; Della Verde, G.; Gaudiello, E.; Marsano, A.; Pfister, O.; Kuster, G.M. Polo-like Kinase 2 is dynamically regulated to coordinate proliferation and early lineage specification downstream of yes-associated protein 1 in Cardiac progenitor cells. J. Am. Heart Assoc. 2017, 6, e005920. [Google Scholar] [CrossRef] [Green Version]
- Folmes, C.D.; Nelson, T.J.; Martinez-Fernandez, A.; Arrell, D.K.; Lindor, J.Z.; Dzeja, P.P.; Ikeda, Y.; Perez-Terzic, C.; Terzic, A. Somatic oxidative bioenergetics transitions into pluripotency-dependent glycolysis to facilitate nuclear reprogramming. Cell Metab. 2011, 14, 264–271. [Google Scholar] [CrossRef] [Green Version]
- Park, S.J.; Lee, S.A.; Prasain, N.; Bae, D.; Kang, H.; Ha, T.; Kim, J.S.; Hong, K.H.; Mantel, C.; Moon, S.H.; et al. Metabolome Profiling of Partial and Fully Reprogrammed Induced Pluripotent Stem Cells. Stem Cells Dev. 2017, 26, 734–742. [Google Scholar] [CrossRef]
- Yamaguchi, H.; Taouk, G.M. A potential role of YAP/TAZ in the interplay between metastasis and metabolic alterations. Front. Oncol. 2020, 10, 928. [Google Scholar] [CrossRef] [PubMed]
- Enzo, E.; Santinon, G.; Pocaterra, A.; Aragona, M.; Bresolin, S.; Forcato, M.; Grifoni, D.; Pession, A.; Zanconato, F.; Guzzo, G.; et al. Aerobic glycolysis tunes YAP/TAZ transcriptional activity. EMBO J. 2015, 34, 1349–1370. [Google Scholar] [CrossRef]
- DeRan, M.; Yang, J.; Shen, C.H.; Peters, E.C.; Fitamant, J.; Chan, P.; Hsieh, M.; Zhu, S.; Asara, J.M.; Zheng, B.; et al. Energy stress regulates hippo-YAP signaling involving AMPK-mediated regulation of angiomotin-like 1 protein. Cell Rep. 2014, 9, 495–503. [Google Scholar] [CrossRef] [Green Version]
- Mo, J.S.; Meng, Z.; Kim, Y.C.; Park, H.W.; Hansen, C.G.; Kim, S.; Lim, D.S.; Guan, K.L. Cellular energy stress induces AMPK-mediated regulation of YAP and the Hippo pathway. Nat. Cell Biol. 2015, 17, 500–510. [Google Scholar] [CrossRef]
- Wang, W.; Xiao, Z.D.; Li, X.; Aziz, K.E.; Gan, B.; Johnson, R.; Chen, J. AMPK modulates Hippo pathway activity to regulate energy homeostasis. Nat Cell Biol. 2015, 17, 490–499. [Google Scholar] [CrossRef] [Green Version]
- Peng, C.; Zhu, Y.; Zhang, W.; Liao, Q.; Chen, Y.; Zhao, X.; Guo, Q.; Shen, P.; Zhen, B.; Quian, X.; et al. Regulation of the Hippo-YAP Pathway by Glucose Sensor O-GlcNAcylation. Mol. Cell 2017, 68, 591–604.e5. [Google Scholar] [CrossRef]
- Zhang, X.; Qiao, Y.; Wu, Q.; Chen, Y.; Zou, S.; Zhao, Y.; Chen, Y.; Yu, Y.; Pan, Q.; Wang, J.; et al. The essential role of YAP O-GlcNAcylation in high-glucose-stimulated liver tumorigenesis. Nat. Commun. 2017, 8, 15280. [Google Scholar] [CrossRef]
- Röhrig, F.; Schulze, A. The multifaceted roles of fatty acid synthesis in cancer. Nat. Rev. Cancer 2016, 16, 732–749. [Google Scholar] [CrossRef]
- Noto, A.; De Vitis, C.; Pisanu, M.E.; Roscilli, G.; Ricci, G.; Catizone, A.; Sorrentino, G.; Chianese, G.; Taglialatela-Scafati, O.; Trisciuoglio, D.; et al. Stearoyl-CoA-desaturase 1 regulates lung cancer stemness via stabilization and nuclear localization of YAP/TAZ. Oncogene 2017, 36, 4573–4584. [Google Scholar] [CrossRef]
- Sorrentino, G.; Ruggeri, N.; Specchia, V.; Cordenonsi, M.; Mano, M.; Dupont, S.; Manfrin, A.; Ingallina, E.; Sommaggio, R.; Piazza, S.; et al. Metabolic control of YAP and TAZ by the mevalonate pathway. Nat. Cell Biol. 2014, 16, 357–366. [Google Scholar] [CrossRef]
- Koo, J.H.; Guan, K.L. Interplay between YAP/TAZ and Metabolism. Cell Metab. 2018, 28, 196–206. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- White, S.M.; Avantaggiati, M.L.; Nemazanyy, I.; Di Poto, C.; Yang, Y.; Pende, M.; Gibney, G.T.; Ressom, H.W.; Field, J.; Atkins, M.B.; et al. YAP/TAZ Inhibition Induces Metabolic and Signaling Rewiring Resulting in Targetable Vulnerabilities in NF2-Deficient Tumor Cells. Dev. Cell 2019, 49, 425–443.e9. [Google Scholar] [CrossRef] [PubMed]
- Chen, R.; Zhu, S.; Fan, X.G.; Wang, H.; Lotze, M.T.; Zeh, H.J., 3rd; Billiar, T.R.; Kang, R.; Tang, D. High mobility group protein B1 controls liver cancer initiation through yes-associated protein-dependent aerobic glycolysis. Hepatology 2018, 67, 1823–1841. [Google Scholar] [CrossRef] [Green Version]
- Jia, Y.; Li, H.Y.; Wang, J.; Wang, Y.; Zhang, P.; Ma, N.; Mo, S.J. Phosphorylation of 14-3-3ζ links YAP transcriptional activation to hypoxic glycolysis for tumorigenesis. Oncogenesis 2019, 8, 31. [Google Scholar] [CrossRef]
- Kuo, C.C.; Ling, H.H.; Chiang, M.C.; Chung, C.H.; Lee, W.Y.; Chu, C.Y.; Wu, Y.C.; Chen, C.H.; Lai, Y.W.; Tsai, I.L.; et al. Metastatic Colorectal Cancer Rewrites Metabolic Program Through a Glut3-YAP-dependent Signaling Circuit. Theranostics 2019, 9, 2526–2540. [Google Scholar] [CrossRef]
- Song, L.; Tang, H.; Liao, W.; Luo, X.; Li, Y.; Chen, T.; Zhang, X. FOXC2 positively regulates YAP signaling and promotes the glycolysis of nasopharyngeal carcinoma. Exp. Cell Res. 2017, 357, 17–24. [Google Scholar] [CrossRef]
- Gao, Y.; Yang, Y.; Yuan, F.; Huang, J.; Xu, W.; Mao, B.; Yuan, Z.; Bi, W. TNFα-YAP/p65-HK2 axis mediates breast cancer cell migration. Oncogenesis 2017, 6, e383. [Google Scholar] [CrossRef] [Green Version]
- Zheng, X.; Han, H.; Liu, G.P.; Ma, Y.X.; Pan, R.L.; Sang, L.J.; Li, R.H.; Yang, L.J.; Marks, J.R.; Wang, W.; et al. LncRNA wires up Hippo and Hedgehog signaling to reprogramme glucose metabolism. EMBO J. 2017, 36, 3325–3335. [Google Scholar] [CrossRef]
- Jeong, S.H.; Kim, H.B.; Kim, M.C.; Lee, J.M.; Lee, J.H.; Kim, J.H.; Kim, J.W.; Park, W.Y.; Kim, S.Y.; Kim, J.B.; et al. Hippo-mediated suppression of IRS2/AKT signaling prevents hepatic steatosis and liver cancer. J. Clin. Investig. 2018, 128, 1010–1025. [Google Scholar] [CrossRef] [Green Version]
- Tharp, K.M.; Kang, M.S.; Timblin, G.A.; Dempersmier, J.; Dempsey, G.E.; Zushin, P.J.H.; Benavides, J.; Choi, C.; Li, C.X.; Jha, A.K.; et al. Actomyosin-Mediated Tension Orchestrates Uncoupled Respiration in Adipose Tissues. Cell Metab. 2018, 27, 602–615.e4. [Google Scholar] [CrossRef] [Green Version]
- Shu, Z.; Gao, Y.; Zhang, G.; Zhou, Y.; Cao, J.; Wan, D.; Zhu, X.; Xiong, W. A functional interaction between Hippo-YAP signalling and SREBPs mediates hepatic steatosis in diabetic mice. J. Cell. Mol. Med. 2019, 23, 3616–3628. [Google Scholar] [CrossRef] [Green Version]
- Lee, C.K.; Jeong, S.H.; Jang, C.; Bae, H.; Kim, Y.H.; Park, I.; Kim, S.K.; Koh, G.Y. Tumor metastasis to lymph nodes requires YAP-dependent metabolic adaptation. Science 2019, 363, 644–649. [Google Scholar] [CrossRef] [Green Version]
- Cox, A.G.; Hwang, K.L.; Brown, K.K.; Evason, K.; Beltz, S.; Tsomides, A.; O’Connor, K.; Galli, G.G.; Yimlamai, D.; Chhangawala, S.; et al. Yap reprograms glutamine metabolism to increase nucleotide biosynthesis and enable liver growth. Nat. Cell Biol. 2016, 18, 886–896. [Google Scholar] [CrossRef] [Green Version]
- Hansen, C.G.; Ng, Y.L.; Lam, W.L.; Plouffe, S.W.; Guan, K.L. The hippo pathway effectors YAP and TAZ promote cell growth by modulating amino acid signaling to mTORC1. Cell Res. 2015, 25, 1299–1313. [Google Scholar] [CrossRef]
- Bertero, T.; Oldham, W.M.; Cottrill, K.A.; Pisano, S.; Vanderpool, R.R.; Yu, Q.; Zhao, J.; Tai, Y.; Tang, Y.; Zhang, Y.Y.; et al. Vascular stiffness mechanoactivates YAP/TAZ-dependent glutaminolysis to drive pulmonary hypertension. J. Clin. Investig. 2016, 126, 3313–3335. [Google Scholar] [CrossRef] [Green Version]
- Yang, C.S.; Stampouloglou, E.; Kingston, N.M.; Zhang, L.; Monti, S.; Varelas, X. Glutamine-utilizing transaminases are a metabolic vulnerability of TAZ/YAP-activated cancer cells. EMBO Rep. 2018, 19, e43577. [Google Scholar] [CrossRef]
- Bertero, T.; Oldham, W.M.; Grasset, E.M.; Bourget, I.; Boulter, E.; Pisano, S.; Hofman, P.; Bellvert, F.; Meneguzzi, G.; Bulavin, D.V.; et al. Tumor-Stroma Mechanics Coordinate Amino Acid Availability to Sustain Tumor Growth and Malignancy. Cell Metab. 2019, 29, 124–140.e10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tumaneng, K.; Schlegelmilch, K.; Russell, R.C.; Yimlamai, D.; Basnet, H.; Mahadevan, N.; Fitamant, J.; Bardeesy, N.; Camargo, F.D.; Guan, K.L. YAP mediates crosstalk between the Hippo and PI(3)K–TOR pathways by suppressing PTEN via miR-29. Nat. Cell Biol. 2012, 14, 1322–1329. [Google Scholar] [CrossRef]
- Artinian, N.; Cloninger, C.; Holmes, B.; Benavides-Serrato, A.; Bashir, T.; Gera, J. Phosphorylation of the Hippo Pathway Component AMOTL2 by the mTORC2 Kinase Promotes YAP Signaling, Resulting in Enhanced Glioblastoma Growth and Invasiveness. J. Biol. Chem. 2015, 290, 19387–19401. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sciarretta, S.; Zhai, P.; Maejima, Y.; Del Re, D.P.; Nagarajan, N.; Yee, D.; Liu, T.; Magnuson, M.A.; Volpe, M.; Frati, G.; et al. mTORC2 regulates cardiac response to stress by inhibiting MST1. Cell Rep. 2015, 11, 125–136. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Nagaraj, R.; Gururaja-Rao, S.; Jones, K.T.; Slattery, M.; Negre, N.; Braas, D.; Christofk, H.; White, K.P.; Mann, R.; Banerjee, U. Control of mitochondrial structure and function by the Yorkie/YAP oncogenic pathway. Genes Dev. 2012, 26, 2027–2037. [Google Scholar] [CrossRef] [Green Version]
- Von Eyss, B.; Jaenicke, L.A.; Kortlever, R.M.; Royla, N.; Wiese, K.E.; Letschert, S.; McDuffus, L.A.; Sauer, M.; Rosenwald, A.; Evan, G.I.; et al. A MYC-Driven Change in Mitochondrial Dynamics Limits YAP/TAZ Function in Mammary Epithelial Cells and Breast Cancer. Cancer Cell 2015, 28, 743–757. [Google Scholar] [CrossRef] [Green Version]
- Kim, H.; Jang, H.; Kim, T.W.; Kang, B.H.; Lee, S.E.; Jeon, Y.K.; Chung, D.H.; Choi, J.; Shin, J.; Cho, E.J.; et al. Core Pluripotency Factors Directly Regulate Metabolism in Embryonic Stem Cell to Maintain Pluripotency. Stem Cells 2015, 33, 2699–2711. [Google Scholar] [CrossRef]
- Kida, Y.S.; Kawamura, T.; Wei, Z.; Sogo, T.; Jacinto, S.; Shigeno, A.; Kushige, H.; Yoshihara, E.; Liddle, C.; Ecker, J.R.; et al. ERRs Mediate a Metabolic Switch Required for Somatic Cell Reprogramming to Pluripotency. Cell Stem Cell 2015, 16, 547–555. [Google Scholar] [CrossRef] [Green Version]
- Hawkins, K.E.; Joy, S.; Delhove, J.M.; Kotiadis, V.N.; Fernandez, E.; Fitzpatrick, L.M.; Whiteford, J.R.; King, P.J.; Bolanos, J.P.; Duchen, M.R.; et al. NRF2 Orchestrates the Metabolic Shift during Induced Pluripotent Stem Cell Reprogramming. Cell Rep. 2016, 14, 1883–1891. [Google Scholar] [CrossRef] [Green Version]
- Mathieu, J.; Zhou, W.; Xing, Y.; Sperber, H.; Ferreccio, A.; Agoston, Z.; Kuppusamy, K.T.; Moon, R.T.; Ruohola-Baker, H. Hypoxia-inducible factors have distinct and stage-specific roles during reprogramming of human cells to pluripotency. Cell Stem Cell 2014, 14, 592–605. [Google Scholar] [CrossRef] [Green Version]
- Prigione, A.; Rohwer, N.; Hoffmann, S.; Mlody, B.; Drews, K.; Bukowiecki, R.; Blumlein, K.; Wanker, E.E.; Ralser, M.; Cramer, T.; et al. HIF1alpha modulates cell fate reprogramming through early glycolytic shift and upregulation of PDK1-3 and PKM2. Stem Cells 2014, 32, 364–376. [Google Scholar] [CrossRef] [Green Version]
- Yoshida, Y.; Takahashi, K.; Okita, K.; Ichisaka, T.; Yamanaka, S. Hypoxia enhances the generation of induced pluripotent stem cells. Cell Stem Cell 2009, 5, 237–241. [Google Scholar] [CrossRef] [Green Version]
- Prieto, J.; Seo, A.Y.; Leon, M.; Santacatterina, F.; Torresano, L.; Palomino-Schatzlein, M.; Gimenez, K.; Vallet-sanchez, A.; Ponsoda, X.; Pineda-Lucena, A.; et al. MYC Induces a Hybrid Energetics Program Early in Cell Reprogramming. Stem Cell Rep. 2018, 11, 1479–1492. [Google Scholar] [CrossRef] [Green Version]
- Khaw, S.L.; Min-Wen, C.; Koh, C.G.; Lim, B.; Shyh-Chang, N. Oocyte factors suppress mitochondrial polynucleotide phosphorylase to remodel the metabolome and enhance reprogramming. Cell Rep. 2015, 12, 1080–1088. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mathieu, J.; Ruohola-Baker, H. Metabolic RemodeLIN of Pluripotency. Cell Stem Cell 2016, 19, 3–4. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Parisi, S.; Passaro, F.; Russo, L.; Musto, A.; Navarra, A.; Romano, S.; Petrosino, G.; Russo, T. Lin28 is induced in primed embryonic stem cells and regulates let-7-independent events. FASEB J. 2017, 31, 1046–1058. [Google Scholar] [CrossRef] [Green Version]
- Parisi, S.; Castaldo, D.; Piscitelli, S.; D’Ambrosio, C.; Divisato, G.; Passaro, F.; Avolio, R.; Castellucci, A.; Gianfico, P.; Masullo, M.; et al. Identification of RNA-binding proteins that partner with Lin28a to regulate Dnmt3a expression. Sci. Rep. 2021, 11, 2345. [Google Scholar] [CrossRef] [PubMed]
- Zhang, J.; Ratanasirintrawoot, S.; Chandrasekaran, S.; Wu, Z.; Ficarro, S.B.; Yu, C.; Ross, C.A.; Cacchiarelli, D.; Xia, Q.; Seligson, M.; et al. LIN28 Regulates Stem Cell Metabolism and Conversion to Primed Pluripotency. Cell Stem Cell 2016, 19, 66–80. [Google Scholar] [CrossRef] [Green Version]
- Suhr, S.T.; Chang, E.A.; Tjong, J.; Alcasid, N.; Perkins, G.A.; Goissis, M.D.; Ellisman, M.H.; Perez, G.I.; Cibelli, J.B. Mitochondrial rejuvenation after induced pluripotency. PLoS ONE 2010, 5, e14095. [Google Scholar] [CrossRef] [Green Version]
- Prigione, A.; Fauler, B.; Lurz, R.; Lehrach, H.; Adjaye, J. The senescence-related mitochondrial/oxidative stress pathway is repressed in human induced pluripotent stem cells. Stem Cells 2010, 28, 721–733. [Google Scholar] [CrossRef]
- Ma, T.; Li, J.; Xu, Y.; Yu, C.; Xu, T.; Wang, H.; Liu, K.; Cao, N.; Nie, B.M.; Zhu, S.Y.; et al. Atg5-independent autophagy regulates mitochondrial clearance and is essential for iPSC reprogramming. Nat. Cell Biol. 2015, 17, 1379–1387. [Google Scholar] [CrossRef]
- Prigione, A.; Hossini, A.M.; Lichtner, B.; Serin, A.; Fauler, B.; Megges, M.; Lurz, R.; Lehrach, H.; Makrantonaki, E.; Zouboulis, C.C.; et al. Mitochondrial-associated cell death mechanisms are reset to an embryonic-like state in aged donor-derived iPS cells harboring chromosomal aberrations. PLoS ONE 2011, 6, e27352. [Google Scholar] [CrossRef] [Green Version]
- Li, D.; Liu, J.; Yang, X.; Zhou, C.; Guo, J. Chromatin Accessibility Dynamics during iPSC Reprogramming. Cell Stem Cell 2017, 21, 819–833.e6. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Parisi, S.; Piscitelli, S.; Passaro, F.; Russo, T. HMGA Proteins in Stemness and Differentiation of Embryonic and Adult Stem Cells. Int. J. Mol. Sci. 2020, 21, 362. [Google Scholar] [CrossRef] [Green Version]
- Harvey, A.; Caretti, G.; Moresi, V.; Renzini, A.; Adamo, S. Interplay between Metabolites and the Epigenome in Regulating Embryonic and Adult Stem Cell Potency and Maintenance. Stem Cell Rep. 2019, 13, 573–589. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Shyh-Chang, N.; Locasale, J.W.; Lyssiotis, C.A.; Zheng, Y.; Teo, R.Y.; Ratanasirintrawoot, S.; Zhang, J.; Onder, T.; Uternaehrer, J.J.; Zhu, H.; et al. Influence of threonine metabolism on S-adenosylmethionine and histone methylation. Science 2013, 339, 222–226. [Google Scholar] [CrossRef] [Green Version]
- Panopoulos, A.D.; Yanes, O.; Ruiz, S.; Kida, Y.S.; Diep, D.; Tautenhahn, R.; Herrerias, A.; Batchelder, E.M.; Plongthongkum, N.; Lutz, M.; et al. The metabolome of induced pluripotent stem cells reveals metabolic changes occurring in somatic cell reprogramming. Cell Res. 2012, 22, 168–177. [Google Scholar] [CrossRef] [Green Version]
- Shiraki, N.; Shiraki, Y.; Tsuyama, T.; Obata, F.; Miura, M.; Nagae, G.; Aburatani, H.; Kume, K.; Rndo, F.; Kume, S. Methionine metabolism regulates maintenance and differentiation of human pluripotent stem cells. Cell Metab. 2014, 19, 780–794. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Moussaieff, A.; Rouleau, M.; Kitsberg, D.; Cohen, M.; Levy, G.; Barasch, D.; Nemirovski, A.; Shen.Orr, S.; Laevsky, I.; Amit, M.; et al. Glycolysis-mediated changes in acetyl-CoA and histone acetylation control the early differentiation of embryonic stem cells. Cell Metab. 2015, 21, 392–402. [Google Scholar] [CrossRef] [Green Version]
- Wellen, K.E.; Hatzivassiliou, G.; Sachdeva, U.M.; Bui, T.V.; Cross, J.R.; Thompson, C.B. ATP-citrate lyase links cellular metabolism to histone acetylation. Science 2009, 324, 1076–1080. [Google Scholar] [CrossRef] [Green Version]
- Totaro, A.; Zhuang, Q.; Panciera, T.; Battilana, G.; Azzolin, L.; Brumana, G.; Gandin, A.; Brusatin, G.; Cordenonsi, M.; Piccolo, S. Cell phenotypic plasticity requires autophagic flux driven by YAP/TAZ mechanotransduction. Proc. Natl. Acad. Sci. USA 2019, 116, 17848–17857. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hillmer, R.E.; Link, B.A. The roles of Hippo signaling transducers Yap and Taz in chromatin remodeling. Cells 2019, 8, 502. [Google Scholar] [CrossRef] [Green Version]
- Chaulk, S.G.; Lattanzi, V.J.; Hiemer, S.E.; Fahlman, R.P.; Varelas, X. The Hippo pathway effectors TAZ/YAP regulate dicer expression and microRNA biogenesis through Let-7. J. Biol. Chem. 2014, 289, 1886–1891. [Google Scholar] [CrossRef] [Green Version]
- Divisato, G.; Passaro, F.; Russo, T.; Parisi, S. The Key Role of MicroRNAs in Self-Renewal and Differentiation of Embryonic Stem Cells. Int. J. Mol. Sci. 2020, 21, 6285. [Google Scholar] [CrossRef]
- Mori, M.; Triboulet, R.; Mohseni, M.; Schlegelmilch, K.; Shrestha, K.; Camargo, F.D.; Gregory, R.I. Hippo signaling regulates microprocessor and links cell-density-dependent miRNA biogenesis to cancer. Cell 2014, 156, 893–906. [Google Scholar] [CrossRef] [Green Version]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2021 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (http://creativecommons.org/licenses/by/4.0/).
Share and Cite
Di Benedetto, G.; Parisi, S.; Russo, T.; Passaro, F. YAP and TAZ Mediators at the Crossroad between Metabolic and Cellular Reprogramming. Metabolites 2021, 11, 154. https://doi.org/10.3390/metabo11030154
Di Benedetto G, Parisi S, Russo T, Passaro F. YAP and TAZ Mediators at the Crossroad between Metabolic and Cellular Reprogramming. Metabolites. 2021; 11(3):154. https://doi.org/10.3390/metabo11030154
Chicago/Turabian StyleDi Benedetto, Giorgia, Silvia Parisi, Tommaso Russo, and Fabiana Passaro. 2021. "YAP and TAZ Mediators at the Crossroad between Metabolic and Cellular Reprogramming" Metabolites 11, no. 3: 154. https://doi.org/10.3390/metabo11030154
APA StyleDi Benedetto, G., Parisi, S., Russo, T., & Passaro, F. (2021). YAP and TAZ Mediators at the Crossroad between Metabolic and Cellular Reprogramming. Metabolites, 11(3), 154. https://doi.org/10.3390/metabo11030154