Browse > Article
http://dx.doi.org/10.14348/molcells.2018.0438

The Interface Between ER and Mitochondria: Molecular Compositions and Functions  

Lee, Soyeon (Department of Biological Sciences, School of Life Sciences, Ulsan National Institute of Science and Technology)
Min, Kyung-Tai (Department of Biological Sciences, School of Life Sciences, Ulsan National Institute of Science and Technology)
Abstract
Mitochondria and endoplasmic reticulum (ER) are essential organelles in eukaryotic cells, which play key roles in various biological pathways. Mitochondria are responsible for ATP production, maintenance of $Ca^{2+}$ homeostasis and regulation of apoptosis, while ER is involved in protein folding, lipid metabolism as well as $Ca^{2+}$ homeostasis. These organelles have their own functions, but they also communicate via mitochondrial-associated ER membrane (MAM) to provide another level of regulations in energy production, lipid process, $Ca^{2+}$ buffering, and apoptosis. Hence, defects in MAM alter cell survival and death. Here, we review components forming the molecular junctions of MAM and how MAM regulates cellular functions. Furthermore, we discuss the effects of impaired ER-mitochondrial communication in various neurodegenerative diseases.
Keywords
ER-mitochondria tethering; mitochondrial-associated ER membrane (MAM); neurodegenerative disease;
Citations & Related Records
연도 인용수 순위
  • Reference
1 Bernhard, W., and Rouiller, C. (1956). Close topographical relationship between mitochondria and ergastoplasm of liver cells in a definite phase of cellular activity. J. Biophys. Biochem. Cytol. 2, 73-78.   DOI
2 Cali, T., Ottolini, D., Negro, A., and Brini, M. (2012). alpha-Synuclein controls mitochondrial calcium homeostasis by enhancing endoplasmic reticulum-mitochondria interactions. J. Biol. Chem. 287, 17914-17929.   DOI
3 Cheung, K.H., Shineman, D., Muller, M., Cardenas, C., Mei, L., Yang, J., Tomita, T., Iwatsubo, T., Lee, V.M., and Foskett, J.K. (2008). Mechanism of $Ca^{2+}$ disruption in Alzheimer's disease by presenilin regulation of InsP3 receptor channel gating. Neuron 58, 871-883.   DOI
4 Matsuzaki, H., Fujimoto, T., Ota, T., Ogawa, M., Tsunoda, T., Doi, K., Hamabashiri, M., Tanaka, M., and Shirasawa, S. (2012). Tespa1 is a novel inositol 1,4,5-trisphosphate receptor binding protein in T and B lymphocytes. FEBS Open Bio. 2, 255-259.   DOI
5 Matsuzaki, H., Fujimoto, T., Tanaka, M., and Shirasawa, S. (2013). Tespa1 is a novel component of mitochondria-associated endoplasmic reticulum membranes and affects mitochondrial calcium flux. Biochem. Biophys. Res. Commun. 433, 322-326.   DOI
6 McCormack, J.G., and Denton, R.M. (1993). Mitochondrial $Ca^{2+}$ transport and the role of intramitochondrial $Ca^{2+}$ in the regulation of energy metabolism. Dev. Neurosci. 15, 165-173.   DOI
7 McLelland, G.L., Goiran, T., Yi, W., Dorval, G., Chen, C.X., Lauinger, N.D., Krahn, A.I., Valimehr, S., Rakovic, A., Rouiller, I., et al. (2018). Mfn2 ubiquitination by PINK1/parkin gates the p97-dependent release of ER from mitochondria to drive mitophagy. Elife 7.
8 Merkwirth, C., and Langer, T. (2008). Mitofusin 2 builds a bridge between ER and mitochondria. Cell 135, 1165-1167.   DOI
9 Wu, H., Carvalho, P., and Voeltz, G.K. (2018). Here, there, and everywhere: the importance of ER membrane contact sites. Science 361.
10 Wakana, Y., Takai, S., Nakajima, K.i., Tani, K., Yamamoto, A., Watson, P., Stephens, D.J., Hauri, H.P., Tagaya, M., and Linstedt, A. (2008). Bap31 is an itinerant protein that moves between the peripheral endoplasmic reticulum (ER) and a juxtanuclear compartment related to ER-associated degradation. Mol. Biol. Cell 19, 1825-1836.   DOI
11 Wu, W., Lin, C., Wu, K., Jiang, L., Wang, X., Li, W., Zhuang, H., Zhang, X., Chen, H., Li, S., et al. (2016). FUNDC1 regulates mitochondrial dynamics at the ER-mitochondrial contact site under hypoxic conditions. EMBO J. 35, 1368-1384.   DOI
12 Wu, Z., and Bowen, W.D. (2008). Role of sigma-1 receptor Cterminal segment in inositol 1,4,5-trisphosphate receptor activation: constitutive enhancement of calcium signaling in MCF-7 tumor cells. J. Biol. Chem. 283, 28198-28215.   DOI
13 Xu, H., Guan, N., Ren, Y.L., Wei, Q.J., Tao, Y.H., Yang, G.S., Liu, X.Y., Bu, D.F., Zhang, Y., and Zhu, S.N. (2018). IP3R-Grp75-VDAC1-MCU calcium regulation axis antagonists protect podocytes from apoptosis and decrease proteinuria in an Adriamycin nephropathy rat model. BMC Nephrol. 19, 140.   DOI
14 Copeland, D.E., and Dalton, A. J. (1959). An association between mitochondria and the endoplasmic reticulum in cells of the pseudobranch gland of a teleost. J. Biophys. Biochem. Cytol. 5, 393-396.   DOI
15 Chung, J., Torta, F., Masai, K., Lucast, L., Czapla, H., Tanner, L.B., Narayanaswamy, P., Wenk, M.R., Nakatsu, F., and De Camilli, P. (2015). PI4P/phosphatidylserine countertransport at ORP5- and ORP8-mediated ER-plasma membrane contacts. Science 349, 428-432.
16 Cipolat, S., Martins de Brito, O., Dal Zilio, B., and Scorrano, L. (2004). OPA1 requires mitofusin 1 to promote mitochondrial fusion. Proc. Natl. Acad. Sci. USA 101, 15927-15932.   DOI
17 Colombini, M. (2012). VDAC structure, selectivity, and dynamics. Biochim. Biophys. Acta. 1818, 1457-1465.   DOI
18 Csordas, G., Varnai, P., Golenar, T., Roy, S., Purkins, G., Schneider, T.G., Balla, T., and Hajnoczky, G. (2010). Imaging interorganelle contacts and local calcium dynamics at the ER-mitochondrial interface. Mol. Cell 39, 121-132.   DOI
19 D'Angelo, G., Vicinanza, M., and De Matteis, M.A. (2008). Lipidtransfer proteins in biosynthetic pathways. Curr. Opin. Cell Biol. 20, 360-370.   DOI
20 Yu, C., Han, W., Shi, T., Lv, B., He, Q., Zhang, Y., Li, T., Zhang, Y., Song, Q., Wang, L., et al. (2008). PTPIP51, a novel 14-3-3 binding protein, regulates cell morphology and motility via Raf-ERK pathway. Cell Signal. 20, 2208-2220.   DOI
21 Zampese, E., Fasolato, C., Kipanyula, M.J., Bortolozzi, M., Pozzan, T., and Pizzo, P. (2011). Presenilin 2 modulates endoplasmic reticulum (ER)-mitochondria interactions and $Ca^{2+}$ cross-talk. Proc. Natl. Acad. Sci. USA 108, 2777-2782.   DOI
22 Wadhwa, R., Taira, K., and Kaul, S.C. (2002). An Hsp70 family chaperone, mortalin/mthsp70/PBP74/Grp75: what, when, and where? Cell Stress Chaperones. 7, 309.   DOI
23 Nguyen, M., Breckenridge, D.G., Ducret, A., and Shore, G.C. (2000). Caspase-resistant BAP31 inhibits fas-mediated apoptotic membrane fragmentation and release of cytochrome c from mitochondria. Mol. Biol. Cell 20, 6731-6740.   DOI
24 Mesmin, B., Bigay, J., Moser von Filseck, J., Lacas-Gervais, S., Drin, G., and Antonny, B. (2013). A four-step cycle driven by PI(4)P hydrolysis directs sterol/PI(4)P exchange by the ER-Golgi tether OSBP. Cell 155, 830-843.   DOI
25 Mikoshiba, K. (2007). IP3 receptor/$Ca^{2+}$ channel: from discovery to new signaling concepts. J. Neurochem. 102, 1426-1446.   DOI
26 Myhill, N., Lynes, E.M., Nanji, J.A., Blagoveshchenskaya, A.D., Fei, H., Simmen, K.C., Cooper, T.J., Thomas, G., Simmen, T., and Linstedt, A. (2008). The subcellular distribution of calnexin is mediated by PACS-2. Mol. Biol. Cell 19, 2777-2788.   DOI
27 Patergnani, S., Suski, J.M., Agnoletto, C., Bononi, A., Bonora, M., De Marchi, E., Giorgi, C., Marchi, S., Missiroli, S., Poletti, F., et al. (2011). Calcium signaling around mitochondria associated membranes (MAMs). Cell Commun. Signal. 9, 19.   DOI
28 Nishimura, A.L., Mitne-Neto, M., Silva, H.C., Richieri-Costa, A., Middleton, S., Cascio, D., Kok, F., Oliveira, J.R., Gillingwater, T., Webb, J., et al. (2004). A mutation in the vesicle-trafficking protein VAPB causes late-onset spinal muscular atrophy and amyotrophic lateral sclerosis. Am. J. Hum. Genet. 75, 822-831.   DOI
29 O'Brien, R.J., and Wong, P.C. (2011). Amyloid precursor protein processing and Alzheimer's disease. Annu. Rev. Neurosci. 34, 185-204.   DOI
30 Ottolini, D., Cali, T., Negro, A., and Brini, M. (2013). The Parkinson disease-related protein DJ-1 counteracts mitochondrial impairment induced by the tumour suppressor protein p53 by enhancing endoplasmic reticulum-mitochondria tethering. Hum. Mol. Genet. 22, 2152-2168.   DOI
31 Detmer, S.A., and Chan, D.C. (2007). Functions and dysfunctions of mitochondrial dynamics. Nat. Rev. Mol. Cell Biol. 8, 870-879.   DOI
32 Murphy, S.E., and Levine, T.P. (2016). VAP, a versatile access point for the endoplasmic reticulum: review and analysis of FFAT-like motifs in the VAPome. Biochim. Biophys. Acta. 1861, 952-961.
33 Das, A.M., and Harris, D.A. (1990). Control of mitochondrial ATP synthase in heart cells: inactive to active transitions caused by beating or positive inotropic agents. Cardiovasc. Res. 24, 411-417.   DOI
34 Davison, E.J., Pennington, K., Hung, C.-C., Peng, J., Rafiq, R., Ostareck-Lederer, A., Ostareck, D.H., Ardley, H.C., Banks, R.E., and Robinson, P.A. (2009). Proteomic analysis of increased Parkin expression and its interactants provides evidence for a role in modulation of mitochondrial function. PROTEOMICS 9, 4284-4297.   DOI
35 de Brito, O.M., and Scorrano, L. (2008). Mitofusin 2 tethers endoplasmic reticulum to mitochondria. Nature 456, 605-610.   DOI
36 de Brito, O.M., and Scorrano, L. (2010). An intimate liaison: spatial organization of the endoplasmic reticulum-mitochondria relationship. EMBO J. 29, 2715-2723.   DOI
37 De Strooper, B. (2007). Loss-of-function presenilin mutations in Alzheimer disease. Talking Point on the role of presenilin mutations in Alzheimer disease. EMBO Rep. 8, 141-146.   DOI
38 De Vos, K.J., Morotz, G.M., Stoica, R., Tudor, E.L., Lau, K.F., Ackerley, S., Warley, A., Shaw, C.E., and Miller, C.C. (2012). VAPB interacts with the mitochondrial protein PTPIP51 to regulate calcium homeostasis. Hum. Mol. Genet. 21, 1299-1311.   DOI
39 Eisenberg-Bord, M., Shai, N., Schuldiner, M., and Bohnert, M. (2016). A tether is a tether is a tether: tethering at membrane contact sites. Dev. Cell 39, 395-409.   DOI
40 Filadi, R., Greotti, E., Turacchio, G., Luini, A., Pozzan, T., and Pizzo, P. (2016). Presenilin 2 modulates endoplasmic reticulum-mitochodria coupling by tuning the antagonistic effect of mitofusin 2. Cell Rep. 15, 2226-2238.   DOI
41 Rostovtseva, T.K., Tan, W., and Colombini, M. (2005). On the role of VDAC in apoptosis: fact and fiction. J. Bioenerg. Biomembr. 37, 129-142.   DOI
42 Peretti, D., Dahan, N., Shimoni, E., Hirschberg, K., Lev, S., and Malhotra, V. (2008). Coordinated lipid transfer between the endoplasmic reticulum and the golgi complex requires the VAP proteins and is essential for golgi-mediated transport. Mol. Biol. Cell 19, 3871-3884.   DOI
43 Petronilli, V., Penzo, D., Scorrano, L., Bernardi, P., and Di Lisa, F. (2001). The mitochondrial permeability transition, release of cytochrome c and cell death. Correlation with the duration of pore openings in situ. J. Biol. Chem. 276, 12030-12034.   DOI
44 Prinz, W.A. (2014). Bridging the gap: membrane contact sites in signaling, metabolism, and organelle dynamics. J. Cell Biol. 205, 759-769.   DOI
45 Raiborg, C., Wenzel, E.M., Pedersen, N.M., Olsvik, H., Schink, K.O., Schultz, S.W., Vietri, M., Nisi, V., Bucci, C., Brech, A., et al. (2015). Repeated ER-endosome contacts promote endosome translocation and neurite outgrowth. Nature 520, 234-238.   DOI
46 Rizzuto, R., Marchi, S., Bonora, M., Aguiari, P., Bononi, A., De Stefani, D., Giorgi, C., Leo, S., Rimessi, A., Siviero, R., et al. (2009). $Ca(^{2+})$ transfer from the ER to mitochondria: when, how and why. Biochim. Biophys. Acta. 1787, 1342-1351.   DOI
47 Rowland, A.A., Chitwood, P.J., Phillips, M.J., and Voeltz, G.K. (2014). ER contact sites define the position and timing of endosome fission. Cell 159, 1027-1041.   DOI
48 Rowland, A.A., and Voeltz, G.K. (2012). Endoplasmic reticulummitochondria contacts: function of the junction. Nat. Rev. Mol. Cell Biol. 13, 607-625.   DOI
49 Simmen, T., Aslan, J.E., Blagoveshchenskaya, A.D., Thomas, L., Wan, L., Xiang, Y., Feliciangeli, S.F., Hung, C.H., Crump, C.M., and Thomas, G. (2005). PACS‐2 controls endoplasmic reticulum–mitochondria communication and Bid‐mediated apoptosis. EMBO J. 24, 717-729.   DOI
50 Galmes, R., Houcine, A., van Vliet, A.R., Agostinis, P., Jackson, C.L., and Giordano, F. (2016). ORP5/ORP8 localize to endoplasmic reticulum-mitochondria contacts and are involved in mitochondrial function. EMBO Rep. 17, 800-810.   DOI
51 Stoica, R., De Vos, K.J., Paillusson, S., Mueller, S., Sancho, R.M., Lau, K.F., Vizcay-Barrena, G., Lin, W.L., Xu, Y.F., Lewis, J., et al. (2014). ER-mitochondria associations are regulated by the VAPB-PTPIP51 interaction and are disrupted by ALS/FTD-associated TDP-43. Nat. Commun. 5, 3996.   DOI
52 Harmon, M., Larkman, P., Hardingham, G., Jackson, M., and Skehel, P. (2017). A Bi-fluorescence complementation system to detect associations between the Endoplasmic reticulum and mitochondria. Sci. Rep. 7, 17467.   DOI
53 Gomez-Suaga, P., Paillusson, S., Stoica, R., Noble, W., Hanger, D.P., and Miller, C.C.J. (2017). The ER-mitochondria tethering complex VAPB-PTPIP51 regulates autophagy. Curr. Biol. 27, 371-385.   DOI
54 Guardia-Laguarta, C., Area-Gomez, E., Rub, C., Liu, Y., Magrane, J., Becker, D., Voos, W., Schon, E.A., and Przedborski, S. (2014). alpha-Synuclein is localized to mitochondria-associated ER membranes. J. Neurosci. 34, 249-259.   DOI
55 Hansford, R.G., and Zorov, D. (1998). Role of mitochondrial calcium transport in the control of substrate oxidation. Mol. Cell Biochem. 184, 359-369.   DOI
56 Haworth, R.A., and Hunter, D.R. (1979). The $Ca^{2+}$-induced membrane transition in mitochondria: II. Nature of the $Ca^{2+}$ trigger site. Arch. Biochem. Biophys. 195, 460-467.   DOI
57 Hayashi, T., Rizzuto, R., Hajnoczky, G., and Su, T.P. (2009). MAM: more than just a housekeeper. Trends Cell Biol. 19, 81-88.   DOI
58 Hayashi, T., and Su, T.P. (2007). Sigma-1 receptor chaperones at the ER-mitochondrion interface regulate $Ca(^{2+})$ signaling and cell survival. Cell 131, 596-610.   DOI
59 Hedskog, L., Pinho, C.M., Filadi, R., Ronnback, A., Hertwig, L., Wiehager, B., Larssen, P., Gellhaar, S., Sandebring, A., Westerlund, M., et al. (2013). Modulation of the endoplasmic reticulummitochondria interface in Alzheimer's disease and related models. Proc. Natl. Acad. Sci. USA 110, 7916-7921.   DOI
60 Stojanovski, D., Koutsopoulos, O.S., Okamoto, K., and Ryan, M.T. (2004). Levels of human Fis1 at the mitochondrial outer membrane regulate mitochondrial morphology. J. Cell Sci. 117, 1201-1210.   DOI
61 Stone, S.J., and Vance, J.E. (2000). Phosphatidylserine synthase-1 and -2 are localized to mitochondria-associated membranes. J. Biol. Chem. 275, 34534-34540.   DOI
62 Su, T.P., Su, T.C., Nakamura, Y., and Tsai, S.Y. (2016). The sigma-1 receptor as a pluripotent modulator in living systems. Trends Pharmacol. Sci. 37, 262-278.   DOI
63 Szabadkai, G., Bianchi, K., Varnai, P., De Stefani, D., Wieckowski, M.R., Cavagna, D., Nagy, A.I., Balla, T., and Rizzuto, R. (2006). Chaperone-mediated coupling of endoplasmic reticulum and mitochondrial $Ca^{2+}$ channels. J. Cell Biol. 175, 901-911.   DOI
64 Van Laar, V.S., Roy, N., Liu, A., Rajprohat, S., Arnold, B., Dukes, A.A., Holbein, C.D., and Berman, S.B. (2015). Glutamate excitotoxicity in neurons triggers mitochondrial and endoplasmic reticulum accumulation of Parkin, and, in the presence of N-acetyl cysteine, mitophagy. Neurobiol. Dis. 74, 180-193.   DOI
65 Vance, J.E. (2014). MAM (mitochondria-associated membranes) in mammalian cells: lipids and beyond. Biochim. Biophys. Acta. 1841, 595-609.   DOI
66 Joseph, S.K., and Hajnoczky, G. (2007). IP3 receptors in cell survival and apoptosis: $Ca^{2+}$ release and beyond. Apoptosis 12, 951-968.   DOI
67 Honrath, B., Metz, I., Bendridi, N., Rieusset, J., Culmsee, C., and Dolga, A.M. (2017). Glucose-regulated protein 75 determines ERmitochondrial coupling and sensitivity to oxidative stress in neuronal cells. Cell Death Discov. 3, 17076.   DOI
68 Hoppe, U.C. (2010). Mitochondrial calcium channels. FEBS Lett. 584, 1975-1981.   DOI
69 Iwasawa, R., Mahul-Mellier, A.L., Datler, C., Pazarentzos, E., and Grimm, S. (2011). Fis1 and Bap31 bridge the mitochondria-ER interface to establish a platform for apoptosis induction. EMBO J. 30, 556-568.   DOI
70 Kanekura, K., Nishimoto, I., Aiso, S., and Matsuoka, M. (2006). Characterization of amyotrophic lateral sclerosis-linked P56S mutation of vesicle-associated membrane protein-associated protein B (VAPB/ALS8). J. Biol. Chem. 281, 30223-30233.   DOI
71 Bernard-Marissal, N., Medard, J.J., Azzedine, H., and Chrast, R. (2015). Dysfunction in endoplasmic reticulum-mitochondria crosstalk underlies SIGMAR1 loss of function mediated motor neuron degeneration. Brain 138, 875-890.   DOI
72 Al-Saif, A., Al-Mohanna, F., and Bohlega, S. (2011). A mutation in sigma-1 receptor causes juvenile amyotrophic lateral sclerosis. Ann. Neurol. 70, 913-919.   DOI
73 Area-Gomez, E., de Groof, A.J., Boldogh, I., Bird, T.D., Gibson, G.E., Koehler, C.M., Yu, W.H., Duff, K.E., Yaffe, M.P., Pon, L.A., et al. (2009). Presenilins are enriched in endoplasmic reticulum membranes associated with mitochondria. Am. J. Pathol. 175, 1810-1816.   DOI
74 Area-Gomez, E., Del Carmen Lara Castillo, M., Tambini, M.D., Guardia-Laguarta, C., de Groof, A.J., Madra, M., Ikenouchi, J., Umeda, M., Bird, T.D., Sturley, S.L., et al. (2012). Upregulated function of mitochondria-associated ER membranes in Alzheimer disease. EMBO J. 31, 4106-4123.   DOI
75 Liou, J., Fivaz, M., Inoue, T., and Meyer, T. (2007). Live-cell imaging reveals sequential oligomerization and local plasma membrane targeting of stromal interaction molecule 1 after $Ca^{2+}$ store depletion. Proc. Natl. Acad. Sci. USA 104, 9301-9306.   DOI
76 Kornmann, B., Currie, E., Collins, S.R., Schuldiner, M., Nunnari, J., Weissman, J.S., and Walter, P. (2009). An ER-mitochondria tethering complex revealed by a synthetic biology screen. Science 325, 477-481.   DOI
77 Kroemer, G., Galluzzi, L., and Brenner, C. (2007). Mitochondrial membrane permeabilization in cell death. Physiol. Rev. 87, 99-163.   DOI
78 Liang, J., Lyu, J., Zhao, M., Li, D., Zheng, M., Fang, Y., Zhao, F., Lou, J., Guo, C., Wang, L., et al. (2017). Tespa1 regulates T cell receptorinduced calcium signals by recruiting inositol 1,4,5-trisphosphate receptors. Nat. Commun. 8, 15732.   DOI
79 Maries, E., Dass, B., Collier, T.J., Kordower, J.H., and Steece-Collier, K. (2003). The role of alpha-synuclein in Parkinson's disease: insights from animal models. Nat. Rev. Neurosci. 4, 727-738.   DOI
80 Marchi, S., and Pinton, P. (2014). The mitochondrial calcium uniporter complex: molecular components, structure and physiopathological implications. J. Physiol. 592, 829-839.   DOI
81 Matsuda, N., Sato, S., Shiba, K., Okatsu, K., Saisho, K., Gautier, C.A., Sou, Y.S., Saiki, S., Kawajiri, S., Sato, F., et al. (2010). PINK1 stabilized by mitochondrial depolarization recruits Parkin to damaged mitochondria and activates latent Parkin for mitophagy. J. Cell Biol. 189, 211-221.   DOI