molecular basis of zinc-dependent regulation of erp44 for ...determined the crystal structure of the...

1
63 HIGHLIGHTS 62 HIGHLIGHTS 4 - Life Science 6 Molecular Basis of Zinc-Dependent Regulation of ERp44 for Protein Quality Control in the Early Secretory Pathway ERp44 is a PDI family member involved in the transport of various secretory proteins and some ER-resident proteins from the endoplasmic reticulum (ER) to the Golgi. Here, we found that Zn 2+ binds to ERp44 with high affinity, modulating its intracellular localization and ability to retrieve clients. Crystal structures of Zn 2+ -bound ERp44 reveal that Zn 2+ binds to a conserved histidine-cluster, inducing large conformational changes. Consequently, the substrate-binding region of ERp44 is fully exposed to solvent, enhancing interactions with clients. These observations suggest a new physiological role of Zn 2+ as a key regulator for protein quality control in the early secretory pathway. ERp44, a member of the protein disulfide isomer- ase (PDI) family, is involved in the transport of various secretory proteins and several ER-resident proteins from the endoplasmic reticulum (ER) to the Golgi. ERp44 traps its clients in the Golgi, retrieving them to the ER in a pH-dependent manner. High-resolution crystal structures of ERp44 at pH 7.2 and 6.5 revealed that protonation of key histidine and cysteine residues induces pH-dependent conformational changes in the protein, leading to exposure of the positively charged client-binding site including the essential cysteine Cys29 [1]. The structures also suggested that metal ions can bind to a histidine-clustered site (His-cluster) compris- ing three highly conserved histidines (His299, His328, His332). In this study, we found that ERp44 specifically binds Zn 2+ with submicromolar affinity to form a Zn 2+ -depen- dent homodimer [2]. Depletion of intracellular zinc by a Zn 2+ chelator altered subcellular localization of ERp44. Furthermore, silencing the Zn 2+ transporters localized at the Golgi resulted in dysfunction of this chaperone, and increased secretion of its clients. To elucidate the detailed mechanism of how Zn 2+ regulates ERp44, we determined the crystal structure of the Zn 2+ bound form of ERp44 at 2.45Å resolution by the SAD method us- ing the anomalous signals of Zn 2+ . X-ray diffraction data were collected on the BL-1A beamline. The overall structure of ERp44 consists of three thioredoxin (Trx)-like domains (a, b, and b’) and a C-terminal tail (C-tail). The crystal structure of the Zn 2+ -bound ERp44 reveals that ERp44 forms a Zn 2+ -bridged homodimer, where the b’ domains from two protomers are bridged by a Zn 2+ ion (Fig. 1). Three types of Zn 2+ -binding sites (sites 1, 2 and 3) are found at the interface of the ERp44 homodimer. Site 1 is formed by the His-cluster in each protomer, at which a Zn 2+ ion is coordinated by His299, His328, His332 and a water molecule in a tetrahedral geometry (Fig. 1, left lower inset). At the center of the dimer interface (site 2), two histidine pairs (His277 and His288) from the b’ domains coordinate a Zn 2+ , playing an essential role in homodi- merization of ERp44 (Fig. 1, middle lower inset). The Zn-bridged ERp44 homodimer possesses a third Zn 2+ - binding site (site 3), which is unexpected because our ITC analyses suggested the presence of only two Zn 2+ - binding sites in ERp44. At site 3, Zn 2+ is coordinated by the thiol group of Cys29, the main-chain carbonyl oxy- gen of Tyr74 of one protomer, the Nδ of His333 of an- other protomer, and two chloride ions (Fig. 1, right lower inset). The coordination mode at site 3 is rarely found in other metalloproteins of known structures. Based on the ITC and other biochemical results, Zn 2+ binding to site 3 does not seem necessary for dimerization. The present structure further reveals that Zn 2+ binding to site 1 induces large displacement of the C-tail, resulting in significant movements of the Trx-like domains (Fig. 2). In the metal-unbound state, Pro353 intervenes between His299, His328 and His332 in the His-cluster [Fig. 2(A), left]. In the Zn 2+ bound state, how- ever, Pro353 is largely moved out and these three histi- dines get closer to one another, adopting a configuration suitable for Zn 2+ binding (site 1) [Fig. 2(A), right]. The large movement of Pro353 also accompanies a striking C-tail movement toward the b’ domain, impairing the interactions between the C-tail and the a domain. As a result, the client binding site in the Zn 2+ -bound form becomes fully exposed to the solvent [Fig. 2(B), right], H333 C29 Y78 Site 3 Mol A Mol B Zn3 Cl - Cl - H277 H281 H277 H281 Site 2 Mol B Mol A Zn2 Zn-bridge Mol B Mol A a a b b b’ b’ 1’ 3 1 2 3’ H332 H328 H299 Site 1 Zn1 H2O His-cluster in contrast to the closed conformation in the metal- unbound state [Fig. 2(B), left]. These observations suggest that the Zn 2+ -bound form of ERp44 recognizes its client proteins with higher affinity than the metal-free form. Indeed, our ITC analysis revealed that interactions of ERp44 with its clients were further enhanced in the presence of Zn 2+ . Based on the present results, we propose a new model of Zn 2+ -dependent protein quality control in the early secretory pathway [2]. Relative to pH changes, Zn 2+ binding to ERp44 induces even larger conforma- tional changes, leading to the enhanced interactions between ERp44 and its clients. These findings shed light on a new physiological role of cellular Zn 2+ as a key regulator for protein quality control. Figure 1: Crystal structure of the Zn-bridged ERp44 homodimer. The a, b, b’ domains and C-tail of Mol A and Mol B are shown in blue, green, yellow, and magenta, respectively. The Zn 2+ ions are shown as orange spheres. The lower insets display close-up views of the three Zn 2+ -binding sites: site 1 (left), site 2 (middle) and site 3 (right). Figure 2 : Zn 2+ -dependent conformational changes of ERp44 (A) Structural comparison of the His-cluster region in the metal-unbound (left) and Zn 2+ -bound (right) ERp44. (B) Comparison of the overall domain arrangement in the metal-unbound (left) and Zn 2+ -bound (right) states of ERp44 monomer. The essential cysteine (Cys29) is represented as spheres. Red-dashed circles indicate a client binding site. REFERENCES [1] S. Watanabe, M. Harayama, S. Kanemura, R. Sitia and K. Inaba, Proc. Natl. Acad. Sci. U.S.A. 114, E3224 (2017). [2] S. Watanabe, Y. Amagai, S. Sannino, T. Tempio, T. Anelli, M. Harayama, S. Masui, I. Sorrentino, M. Yamada, R. Sitia and K. Inaba, Nat. Commun. 10, 603 (2019). BEAMLINE BL-1A S. Watanabe 1 and K. Inaba 1 ( 1 Tohoku Univ. -IMRAM) b a b’ C29 Zn 2+ bound C-tail client binding site C29 Unbound C-tail b a b’ H299 H328 H332 H333 H299 H328 H332 H333 P353 P353 F331 F331 Unbound Zn 2+ bound A B (A) (B)

Upload: others

Post on 30-Jan-2021

0 views

Category:

Documents


0 download

TRANSCRIPT

  • 63HIGHLIGHTS62 HIGHLIGHTS

    4 - Life Science 6

    Molecular Basis of Zinc-Dependent Regulation of ERp44 for Protein Quality Control in the Early Secretory Pathway

    ERp44 is a PDI family member involved in the transport of various secretory proteins and some ER-resident proteins from the endoplasmic reticulum (ER) to the Golgi. Here, we found that Zn2+ binds to ERp44 with high affinity, modulating its intracellular localization and ability to retrieve clients. Crystal structures of Zn2+-bound ERp44 reveal that Zn2+ binds to a conserved histidine-cluster, inducing large conformational changes. Consequently, the substrate-binding region of ERp44 is fully exposed to solvent, enhancing interactions with clients. These observations suggest a new physiological role of Zn2+ as a key regulator for protein quality control in the early secretory pathway.

    ERp44, a member of the protein disulfide isomer-ase (PDI) family, is involved in the transport of various secretory proteins and several ER-resident proteins from the endoplasmic reticulum (ER) to the Golgi. ERp44 traps its clients in the Golgi, retrieving them to the ER in a pH-dependent manner. High-resolution crystal structures of ERp44 at pH 7.2 and 6.5 revealed that protonation of key histidine and cysteine residues induces pH-dependent conformational changes in the protein, leading to exposure of the positively charged client-binding site including the essential cysteine Cys29 [1]. The structures also suggested that metal ions can bind to a histidine-clustered site (His-cluster) compris-ing three highly conserved histidines (His299, His328, His332).

    In this study, we found that ERp44 specifically binds Zn2+ with submicromolar affinity to form a Zn2+-depen-dent homodimer [2]. Depletion of intracellular zinc by a Zn2+ chelator altered subcellular localization of ERp44. Furthermore, silencing the Zn2+ transporters localized

    at the Golgi resulted in dysfunction of this chaperone, and increased secretion of its clients. To elucidate the detailed mechanism of how Zn2+ regulates ERp44, we determined the crystal structure of the Zn2+ bound form of ERp44 at 2.45Å resolution by the SAD method us-ing the anomalous signals of Zn2+. X-ray diffraction data were collected on the BL-1A beamline.

    The overall structure of ERp44 consists of three thioredoxin (Trx)-like domains (a, b, and b’) and a C-terminal tail (C-tail). The crystal structure of the Zn2+-bound ERp44 reveals that ERp44 forms a Zn2+-bridged homodimer, where the b’ domains from two protomers are bridged by a Zn2+ ion (Fig. 1). Three types of Zn2+-binding sites (sites 1, 2 and 3) are found at the interface of the ERp44 homodimer. Site 1 is formed by the His-cluster in each protomer, at which a Zn2+ ion is coordinated by His299, His328, His332 and a water molecule in a tetrahedral geometry (Fig. 1, left lower inset). At the center of the dimer interface (site 2), two histidine pairs (His277 and His288) from the b’ domains

    coordinate a Zn2+, playing an essential role in homodi-merization of ERp44 (Fig. 1, middle lower inset). The Zn-bridged ERp44 homodimer possesses a third Zn2+-binding site (site 3), which is unexpected because our ITC analyses suggested the presence of only two Zn2+-binding sites in ERp44. At site 3, Zn2+ is coordinated by the thiol group of Cys29, the main-chain carbonyl oxy-gen of Tyr74 of one protomer, the Nδ of His333 of an-other protomer, and two chloride ions (Fig. 1, right lower inset). The coordination mode at site 3 is rarely found in other metalloproteins of known structures. Based on the ITC and other biochemical results, Zn2+ binding to site 3 does not seem necessary for dimerization.

    The present structure further reveals that Zn2+ binding to site 1 induces large displacement of the C-tail, resulting in significant movements of the Trx-like domains (Fig. 2). In the metal-unbound state, Pro353 intervenes between His299, His328 and His332 in the His-cluster [Fig. 2(A), left]. In the Zn2+ bound state, how-ever, Pro353 is largely moved out and these three histi-dines get closer to one another, adopting a configuration suitable for Zn2+ binding (site 1) [Fig. 2(A), right]. The large movement of Pro353 also accompanies a striking C-tail movement toward the b’ domain, impairing the interactions between the C-tail and the a domain. As a result, the client binding site in the Zn2+-bound form becomes fully exposed to the solvent [Fig. 2(B), right],

    H333

    C29

    Y78

    Site 3

    Mol A

    Mol B

    Zn3

    Cl-

    Cl-

    H277

    H281 H277

    H281

    Site 2

    Mol B

    Mol A

    Zn2

    Zn-bridge

    Mol BMol A

    a

    a

    b b

    b’

    b’

    1’

    3

    1

    2

    3’

    H332

    H328

    H299

    Site 1

    Zn1

    H2OHis-cluster

    in contrast to the closed conformation in the metal-unbound state [Fig. 2(B), left]. These observations suggest that the Zn2+-bound form of ERp44 recognizes its client proteins with higher affinity than the metal-free form. Indeed, our ITC analysis revealed that interactions of ERp44 with its clients were further enhanced in the presence of Zn2+.

    Based on the present results, we propose a new model of Zn2+-dependent protein quality control in the early secretory pathway [2]. Relative to pH changes, Zn2+ binding to ERp44 induces even larger conforma-tional changes, leading to the enhanced interactions between ERp44 and its clients. These findings shed light on a new physiological role of cellular Zn2+ as a key regulator for protein quality control.

    Figure 1: Crystal structure of the Zn-bridged ERp44 homodimer. The a, b, b’ domains and C-tail of Mol A and Mol B are shown in blue, green, yellow, and magenta, respectively. The Zn2+ ions are shown as orange spheres. The lower insets display close-up views of the three Zn2+-binding sites: site 1 (left), site 2 (middle) and site 3 (right).

    Figure 2 : Zn2+-dependent conformational changes of ERp44 (A) Structural comparison of the His-cluster region in the metal-unbound (left) and Zn2+-bound (right) ERp44. (B) Comparison of the overall domain arrangement in the metal-unbound (left) and Zn2+-bound (right) states of ERp44 monomer. The essential cysteine (Cys29) is represented as spheres. Red-dashed circles indicate a client binding site.

    REFERENCES[1] S. Watanabe, M. Harayama, S. Kanemura, R. Sitia and

    K. Inaba, Proc. Natl. Acad. Sci. U.S.A. 114, E3224 (2017).[2] S. Watanabe, Y. Amagai, S. Sannino, T. Tempio, T. Anelli,

    M. Harayama, S. Masui, I. Sorrentino, M. Yamada, R. Sitia and K. Inaba, Nat. Commun. 10, 603 (2019).

    BEAMLINEBL-1A

    S. Watanabe1 and K. Inaba1 (1Tohoku Univ. -IMRAM)

    b

    a

    b’

    C29

    Zn2+ bound

    C-tailclient binding site

    C29

    Unbound

    C-tail

    b

    a

    b’

    H299

    H328

    H332

    H333

    H299

    H328

    H332

    H333

    P353

    P353

    F331

    F331

    Unbound Zn2+ bound

    A

    B

    (A)

    (B)